1
|
Tang J, Cao Z, Lei M, Yu Q, Mai Y, Xu J, Liao W, Ruan Y, Shi L, Yang L, Liu J. Heterogeneity of cerebral atrophic rate in mild cognitive impairment and its interactive association with proteins related to microglia activity on longitudinal cognitive changes. Arch Gerontol Geriatr 2024; 127:105582. [PMID: 39079281 DOI: 10.1016/j.archger.2024.105582] [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: 04/21/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Heterogeneity of cerebral atrophic rate commonly exists in mild cognitive impairment (MCI), which may be associated with microglia-involved neuropathology and have an influence on cognitive outcomes. OBJECTIVE We aim to explore the heterogeneity of cerebral atrophic rate among MCI and its association with plasma proteins related to microglia activity, with further investigation of their interaction effects on long-term cognition. SUBJECTS A total of 630 MCI subjects in the ADNI database were included, of which 260 subjects were available with baseline data on plasma proteins. METHODS Group-based multi-trajectory modeling (GBMT) was used to identify the latent classes with heterogeneous cerebral atrophic rates. Associations between latent classes and plasma proteins related to microglia activity were investigated with generalized linear models. Linear mixed effect models (LME) were implemented to explore the interaction effects between proteins related to microglia activity and identified latent classes on longitudinal cognitive changes. RESULTS Two latent classes were identified and labeled as the slow-atrophy class and the fast-atrophy class. Associations were found between such heterogeneity of atrophic rates and plasma proteins related to microglia activity, especially AXL receptor tyrosine kinase (AXL), CD40 antigen (CD40), and tumor necrosis factor receptor-like 2 (TNF-R2). Interaction effects on longitudinal cognitive changes showed that higher CD40 was associated with faster cognitive decline in the slow-atrophy class and higher AXL or TNF-R2 was associated with slower cognitive decline in the fast-atrophy class. CONCLUSIONS Heterogeneity of atrophic rates at the MCI stage is associated with several plasma proteins related to microglia activity, which show either protective or adverse effects on long-term cognition depending on the variability of atrophic rates.
Collapse
Affiliation(s)
- Jingyi Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Zhiyu Cao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Ming Lei
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Qun Yu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yingren Mai
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Jiaxin Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Wang Liao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yuting Ruan
- Department of Rehabilitation, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen City, Guangdong Province, MN 518000, China; Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, MN 999077, China
| | - Lianhong Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China.
| | - Jun Liu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China.
| |
Collapse
|
2
|
Jao CW, Wu YT, Yeh JH, Tsai YF, Hsiao CY, Lau CI. Exploring cortical morphology biomarkers of amnesic mild cognitive impairment using novel fractal dimension-based structural MRI analysis. Eur J Neurosci 2024; 60:6254-6266. [PMID: 39353858 DOI: 10.1111/ejn.16557] [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: 03/03/2024] [Revised: 08/29/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
Amnestic mild cognitive impairment (aMCI) is considered as an intermediate stage of Alzheimer's disease, but no MRI biomarkers currently distinguish aMCI from healthy individuals effectively. Fractal dimension, a quantitative parameter, provides superior morphological information compared to conventional cortical thickness methods. Few studies have used cortical fractal dimension values to differentiate aMCI from healthy controls. In this study, we aim to build an automated discriminator for accurately distinguishing aMCI using fractal dimension measures of the cerebral cortex. Thirty aMCI patients and 30 health controls underwent structural MRI of the brain. First, the atrophy of participants' cortical sub-regions of Desikan-Killiany cortical atlas was assessed using fractal dimension and cortical thickness. The fractal dimension is more sensitive than cortical thickness in reducing dimensional effects and may accurately reflect morphological changes of the cortex in aMCI. The aMCI group had significantly lower fractal dimension values in the bilateral temporal lobes, right limbic lobe and right parietal lobe, whereas they showed significantly lower cortical thickness values only in the bilateral temporal lobes. Fractal dimension analysis was able to depict most of the significantly different focal regions detected by cortical thickness, but additionally with more regions. Second, applying the measured fractal dimensions (and cortical thickness) of both cerebral hemispheres, an unsupervised discriminator was built for the aMCI and healthy controls. The proposed fractal dimension-based method achieves 80.54% accuracy in discriminating aMCI from healthy controls. The fractal dimension appears to be a promising biomarker for cortical morphology changes that can discriminate patients with aMCI from healthy controls.
Collapse
Affiliation(s)
- Chi-Wen Jao
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yu-Te Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jiann-Horng Yeh
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yuh-Feng Tsai
- College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Diagnostic Radiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chen-Yu Hsiao
- Department of Diagnostic Radiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chi Ieong Lau
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Dementia Center, Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Applied Cognitive Neuroscience Group, Institute of Cognitive Neuroscience, University College London, London, UK
- University Hospital, Taipa, Macau
| |
Collapse
|
3
|
Sultana OF, Bandaru M, Islam MA, Reddy PH. Unraveling the complexity of human brain: Structure, function in healthy and disease states. Ageing Res Rev 2024; 100:102414. [PMID: 39002647 PMCID: PMC11384519 DOI: 10.1016/j.arr.2024.102414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
The human brain stands as an intricate organ, embodying a nexus of structure, function, development, and diversity. This review delves into the multifaceted landscape of the brain, spanning its anatomical intricacies, diverse functional capacities, dynamic developmental trajectories, and inherent variability across individuals. The dynamic process of brain development, from early embryonic stages to adulthood, highlights the nuanced changes that occur throughout the lifespan. The brain, a remarkably complex organ, is composed of various anatomical regions, each contributing uniquely to its overall functionality. Through an exploration of neuroanatomy, neurophysiology, and electrophysiology, this review elucidates how different brain structures interact to support a wide array of cognitive processes, sensory perception, motor control, and emotional regulation. Moreover, it addresses the impact of age, sex, and ethnic background on brain structure and function, and gender differences profoundly influence the onset, progression, and manifestation of brain disorders shaped by genetic, hormonal, environmental, and social factors. Delving into the complexities of the human brain, it investigates how variations in anatomical configuration correspond to diverse functional capacities across individuals. Furthermore, it examines the impact of neurodegenerative diseases on the structural and functional integrity of the brain. Specifically, our article explores the pathological processes underlying neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases, shedding light on the structural alterations and functional impairments that accompany these conditions. We will also explore the current research trends in neurodegenerative diseases and identify the existing gaps in the literature. Overall, this article deepens our understanding of the fundamental principles governing brain structure and function and paves the way for a deeper understanding of individual differences and tailored approaches in neuroscience and clinical practice-additionally, a comprehensive understanding of structural and functional changes that manifest in neurodegenerative diseases.
Collapse
Affiliation(s)
- Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Madhuri Bandaru
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA 5. Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
4
|
Li J, Jiang Z, Duan S, Zhu X. Multiple Early Biomarkers to Predict Cognitive Decline in Dementia-Free Older Adults. J Geriatr Psychiatry Neurol 2024; 37:395-402. [PMID: 38335267 DOI: 10.1177/08919887241232650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
INTRODUCTION Baseline olfactory impairment, poor performance on cognitive test, and medial temporal lobe atrophy are considered biomarkers for predicting future cognitive decline in dementia-free older adults. However, the combined effect of these predictors has not been fully investigated. METHODS A group of 110 participants without dementia were continuously recruited into this study, and underwent olfactory, cognitive tests and MRI scanning at baseline and 5-year follow-up. Olfactory function was assessed using the University of Pennsylvania Smell Identification Test (UPSIT). Participants were divided into the cognitive decliners and non-decliners. RESULTS Among 87 participants who completed the 5-year follow-up, cognitive decline was present in 32 cases and 55 remained stable. Compared with non-decliners, cognitive decliners presented lower scores on both the UPSIT and the Montreal Cognitive Assessment (MoCA), and smaller hippocampal volume at baseline (all P < .001). The logistic regression analysis revealed that lower scores on UPSIT and MoCA, and smaller hippocampal volume were strongly associated with subsequent cognitive decline, respectively (all P < .001). For the prediction of cognitive decline, lower score on UPSIT performed the sensitivity of 63.6% and specificity of 81.2%, lower score on MoCA with the sensitivity of 74.5% and specificity of 65.6%, smaller hippocampal volume with the sensitivity of 70.9% and specificity of 78.1%, respectively. Combining three predictors resulted in the sensitivity of 83.6% and specificity of 93.7%. CONCLUSIONS The combination of olfactory test, cognitive test with structural MRI may enhance the predictive ability for future cognitive decline for dementia-free older adults.
Collapse
Affiliation(s)
- Juan Li
- Department of Radiology, Heji Hospital Affiliated to Changzhi Medical University, Changzhi, China
| | - Zhiying Jiang
- Department of Radiology, Heji Hospital Affiliated to Changzhi Medical University, Changzhi, China
| | - Shengjie Duan
- Department of Neurology, Heji Hospital Affiliated to Changzhi Medical University, Changzhi, China
| | - Xingxing Zhu
- Department of Radiology, Honghe Hani and Yi Autonomous Prefecture Third People's Hospital, Gejiu, China
| |
Collapse
|
5
|
Murphy AJ, Wilton SD, Aung-Htut MT, McIntosh CS. Down syndrome and DYRK1A overexpression: relationships and future therapeutic directions. Front Mol Neurosci 2024; 17:1391564. [PMID: 39114642 PMCID: PMC11303307 DOI: 10.3389/fnmol.2024.1391564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Down syndrome is a genetic-based disorder that results from the triplication of chromosome 21, leading to an overexpression of many triplicated genes, including the gene encoding Dual-Specificity Tyrosine Phosphorylation-Regulated Kinase 1A (DYRK1A). This protein has been observed to regulate numerous cellular processes, including cell proliferation, cell functioning, differentiation, and apoptosis. Consequently, an overexpression of DYRK1A has been reported to result in cognitive impairment, a key phenotype of individuals with Down syndrome. Therefore, downregulating DYRK1A has been explored as a potential therapeutic strategy for Down syndrome, with promising results observed from in vivo mouse models and human clinical trials that administered epigallocatechin gallate. Current DYRK1A inhibitors target the protein function directly, which tends to exhibit low specificity and selectivity, making them unfeasible for clinical or research purposes. On the other hand, antisense oligonucleotides (ASOs) offer a more selective therapeutic strategy to downregulate DYRK1A expression at the gene transcript level. Advances in ASO research have led to the discovery of numerous chemical modifications that increase ASO potency, specificity, and stability. Recently, several ASOs have been approved by the U.S. Food and Drug Administration to address neuromuscular and neurological conditions, laying the foundation for future ASO therapeutics. The limitations of ASOs, including their high production cost and difficulty delivering to target tissues can be overcome by further advances in ASO design. DYRK1A targeted ASOs could be a viable therapeutic approach to improve the quality of life for individuals with Down syndrome and their families.
Collapse
Affiliation(s)
- Aidan J. Murphy
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - May T. Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Craig S. McIntosh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- Perron Institute for Neurological and Translational Science, Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
6
|
Wang M, Hua Y, Bai Y. A review of the application of exercise intervention on improving cognition in patients with Alzheimer's disease: mechanisms and clinical studies. Rev Neurosci 2024; 0:revneuro-2024-0046. [PMID: 39029521 DOI: 10.1515/revneuro-2024-0046] [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: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, leading to sustained cognitive decline. An increasing number of studies suggest that exercise is an effective strategy to promote the improvement of cognition in AD. Mechanisms of the benefits of exercise intervention on cognitive function may include modulation of vascular factors by affecting cardiovascular risk factors, regulating cardiorespiratory health, and enhancing cerebral blood flow. Exercise also promotes neurogenesis by stimulating neurotrophic factors, affecting neuroplasticity in the brain. Additionally, regular exercise improves the neuropathological characteristics of AD by improving mitochondrial function, and the brain redox status. More and more attention has been paid to the effect of Aβ and tau pathology as well as sleep disorders on cognitive function in persons diagnosed with AD. Besides, there are various forms of exercise intervention in cognitive improvement in patients with AD, including aerobic exercise, resistance exercise, and multi-component exercise. Consequently, the purpose of this review is to summarize the findings of the mechanisms of exercise intervention on cognitive function in patients with AD, and also discuss the application of different exercise interventions in cognitive impairment in AD to provide a theoretical basis and reference for the selection of exercise intervention in cognitive rehabilitation in AD.
Collapse
Affiliation(s)
- Man Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
7
|
By S, Kahl A, Cogswell PM. Alzheimer's Disease Clinical Trials: What Have We Learned From Magnetic Resonance Imaging. J Magn Reson Imaging 2024. [PMID: 39031716 DOI: 10.1002/jmri.29462] [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: 03/04/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 07/22/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of cognitive impairment and dementia worldwide with rising prevalence, incidence and mortality. Despite many decades of research, there remains an unmet need for disease-modifying treatment that can significantly alter the progression of disease. Recently, with United States Food and Drug Administration (FDA) drug approvals, there have been tremendous advances in this area, with agents demonstrating effects on cognition and biomarkers. Magnetic resonance imaging (MRI) plays an instrumental role in these trials. This review article aims to outline how MRI is used for screening eligibility, monitoring safety and measuring efficacy in clinical trials, leaning on the landscape of past and recent AD clinical trials that have used MRI as examples; further, insight on promising MRI biomarkers for future trials is provided. LEVEL OF EVIDENCE: 1. TECHNICAL EFFICACY: Stage 4.
Collapse
Affiliation(s)
- Samantha By
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Anja Kahl
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | | |
Collapse
|
8
|
Leonardsen EH, Persson K, Grødem E, Dinsdale N, Schellhorn T, Roe JM, Vidal-Piñeiro D, Sørensen Ø, Kaufmann T, Westman E, Marquand A, Selbæk G, Andreassen OA, Wolfers T, Westlye LT, Wang Y. Constructing personalized characterizations of structural brain aberrations in patients with dementia using explainable artificial intelligence. NPJ Digit Med 2024; 7:110. [PMID: 38698139 PMCID: PMC11066104 DOI: 10.1038/s41746-024-01123-7] [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: 10/10/2023] [Accepted: 04/23/2024] [Indexed: 05/05/2024] Open
Abstract
Deep learning approaches for clinical predictions based on magnetic resonance imaging data have shown great promise as a translational technology for diagnosis and prognosis in neurological disorders, but its clinical impact has been limited. This is partially attributed to the opaqueness of deep learning models, causing insufficient understanding of what underlies their decisions. To overcome this, we trained convolutional neural networks on structural brain scans to differentiate dementia patients from healthy controls, and applied layerwise relevance propagation to procure individual-level explanations of the model predictions. Through extensive validations we demonstrate that deviations recognized by the model corroborate existing knowledge of structural brain aberrations in dementia. By employing the explainable dementia classifier in a longitudinal dataset of patients with mild cognitive impairment, we show that the spatially rich explanations complement the model prediction when forecasting transition to dementia and help characterize the biological manifestation of disease in the individual brain. Overall, our work exemplifies the clinical potential of explainable artificial intelligence in precision medicine.
Collapse
Affiliation(s)
- Esten H Leonardsen
- Department of Psychology, University of Oslo, Oslo, Norway.
- Norwegian Centre for Mental Disorders Research (NORMENT), Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Karin Persson
- The Norwegian National Centre for Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Edvard Grødem
- Department of Psychology, University of Oslo, Oslo, Norway
- Computational Radiology & Artificial Intelligence (CRAI) Unit, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Nicola Dinsdale
- Oxford Machine Learning in NeuroImaging (OMNI) Lab, University of Oxford, Oxford, UK
| | - Till Schellhorn
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - James M Roe
- Department of Psychology, University of Oslo, Oslo, Norway
| | | | | | - Tobias Kaufmann
- Norwegian Centre for Mental Disorders Research (NORMENT), Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Munich, Germany
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Andre Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Geir Selbæk
- The Norwegian National Centre for Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Center for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Thomas Wolfers
- Department of Psychology, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Munich, Germany
| | - Lars T Westlye
- Department of Psychology, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Center for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Yunpeng Wang
- Department of Psychology, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Quek Y, Fung YL, Bourgeat P, Vogrin SJ, Collins SJ, Bowden SC. Combining neuropsychological assessment and structural neuroimaging to identify early Alzheimer's disease in a memory clinic cohort. Brain Behav 2024; 14:e3505. [PMID: 38688879 PMCID: PMC11061200 DOI: 10.1002/brb3.3505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 05/02/2024] Open
Abstract
INTRODUCTION The current study examined the contributions of comprehensive neuropsychological assessment and volumetric assessment of selected mesial temporal subregions on structural magnetic resonance imaging (MRI) to identify patients with amnestic mild cognitive impairment (aMCI) and mild probable Alzheimer's disease (AD) dementia in a memory clinic cohort. METHODS Comprehensive neuropsychological assessment and automated entorhinal, transentorhinal, and hippocampal volume measurements were conducted in 40 healthy controls, 38 patients with subjective memory symptoms, 16 patients with aMCI, 16 patients with mild probable AD dementia. Multinomial logistic regression was used to compare the neuropsychological and MRI measures. RESULTS Combining the neuropsychological and MRI measures improved group membership prediction over the MRI measures alone but did not improve group membership prediction over the neuropsychological measures alone. CONCLUSION Comprehensive neuropsychological assessment was an important tool to evaluate cognitive impairment. The mesial temporal volumetric MRI measures contributed no diagnostic value over and above the determinations made through neuropsychological assessment.
Collapse
Affiliation(s)
- Yi‐En Quek
- Melbourne School of Psychological SciencesThe University of MelbourneParkvilleVictoriaAustralia
| | - Yi Leng Fung
- Melbourne School of Psychological SciencesThe University of MelbourneParkvilleVictoriaAustralia
| | - Pierrick Bourgeat
- The Australian e‐Health Research CentreCSIRO Health and BiosecurityHerstonQueenslandAustralia
| | - Simon J. Vogrin
- Department of Clinical NeurosciencesSt. Vincent's Hospital MelbourneFitzroyVictoriaAustralia
| | - Steven J. Collins
- Department of Clinical NeurosciencesSt. Vincent's Hospital MelbourneFitzroyVictoriaAustralia
- Department of MedicineThe Royal Melbourne HospitalThe University of MelbourneParkvilleVictoriaAustralia
| | - Stephen C. Bowden
- Melbourne School of Psychological SciencesThe University of MelbourneParkvilleVictoriaAustralia
- Department of Clinical NeurosciencesSt. Vincent's Hospital MelbourneFitzroyVictoriaAustralia
| |
Collapse
|
10
|
Doering S, McCullough A, Gordon BA, Chen CD, McKay N, Hobbs D, Keefe S, Flores S, Scott J, Smith H, Jarman S, Jackson K, Hornbeck RC, Ances BM, Xiong C, Aschenbrenner AJ, Hassenstab J, Cruchaga C, Daniels A, Bateman RJ, Morris JC, Benzinger TLS. Deconstructing pathological tau by biological process in early stages of Alzheimer disease: a method for quantifying tau spatial spread in neuroimaging. EBioMedicine 2024; 103:105080. [PMID: 38552342 PMCID: PMC10995809 DOI: 10.1016/j.ebiom.2024.105080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Neuroimaging studies often quantify tau burden in standardized brain regions to assess Alzheimer disease (AD) progression. However, this method ignores another key biological process in which tau spreads to additional brain regions. We have developed a metric for calculating the extent tau pathology has spread throughout the brain and evaluate the relationship between this metric and tau burden across early stages of AD. METHODS 445 cross-sectional participants (aged ≥ 50) who had MRI, amyloid PET, tau PET, and clinical testing were separated into disease-stage groups based on amyloid positivity and cognitive status (older cognitively normal control, preclinical AD, and symptomatic AD). Tau burden and tau spatial spread were calculated for all participants. FINDINGS We found both tau metrics significantly elevated across increasing disease stages (p < 0.0001) and as a function of increasing amyloid burden for participants with preclinical (p < 0.0001, p = 0.0056) and symptomatic (p = 0.010, p = 0.0021) AD. An interaction was found between tau burden and tau spatial spread when predicting amyloid burden (p = 0.00013). Analyses of slope between tau metrics demonstrated more spread than burden in preclinical AD (β = 0.59), but then tau burden elevated relative to spread (β = 0.42) once participants had symptomatic AD, when the tau metrics became highly correlated (R = 0.83). INTERPRETATION Tau burden and tau spatial spread are both strong biomarkers for early AD but provide unique information, particularly at the preclinical stage. Tau spatial spread may demonstrate earlier changes than tau burden which could have broad impact in clinical trial design. FUNDING This research was supported by the Knight Alzheimer Disease Research Center (Knight ADRC, NIH grants P30AG066444, P01AG026276, P01AG003991), Dominantly Inherited Alzheimer Network (DIAN, NIH grants U01AG042791, U19AG03243808, R01AG052550-01A1, R01AG05255003), and the Barnes-Jewish Hospital Foundation Willman Scholar Fund.
Collapse
Affiliation(s)
- Stephanie Doering
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Nicole McKay
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Diana Hobbs
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Sarah Keefe
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Jalen Scott
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Hunter Smith
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Stephen Jarman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Kelley Jackson
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Beau M Ances
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Chengjie Xiong
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | | - Jason Hassenstab
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Alisha Daniels
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Randall J Bateman
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | | |
Collapse
|
11
|
Huber BN, Fulton EK. Meta-prospective memory accuracy in older adults with and without suspected Mild Cognitive Impairment (sMCI). APPLIED NEUROPSYCHOLOGY. ADULT 2024:1-16. [PMID: 38564814 DOI: 10.1080/23279095.2024.2334892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
This study examined if the relationship between generalized and task-specific appraisals of prospective memory (PM) and actual PM performance (i.e., meta-PM accuracy) differed between healthy and suspected Mild Cognitive Impairment (sMCI) older adults. Older adults recruited included 50 healthy and 31 sMCI participants from the community and an outpatient neuropsychology clinic. Data collected consisted of self-reported appraisals and task-specific predictions/postdictions of PM performance, objective PM performance, and executive functioning (EF). The sMCI group had significantly lower scores on objective PM and EF measures related to simple and complex task-switching. Moreover, sMCI participants displayed lower task-specific meta-PM accuracy in the direction of overconfidence, but they displayed relatively equivalent generalized meta-PM accuracy when compared to the healthy group. Notably, the sMCI group's task-specific inaccuracies became non-significant in relation to the healthy group on the final long-term PM tasks after exposure to metacognitive reflection on the first two PM tasks. Despite lower scores on EF measures, EF performance did not explain task-specific meta-PM differences between groups beyond neurocognitive status. Based on these data, sMCI patients may be better assisted by metacognitive calibration strategies, EF protocols, and the implementation of general compensatory memory strategies as targets for early intervention and prevention of neurocognitive decline.
Collapse
Affiliation(s)
- Becca N Huber
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Psychology, Idaho State University, Pocatello, ID, USA
| | - Erika K Fulton
- Department of Psychology, Idaho State University, Pocatello, ID, USA
| |
Collapse
|
12
|
Krämer C, Stumme J, da Costa Campos L, Dellani P, Rubbert C, Caspers J, Caspers S, Jockwitz C. Prediction of cognitive performance differences in older age from multimodal neuroimaging data. GeroScience 2024; 46:283-308. [PMID: 37308769 PMCID: PMC10828156 DOI: 10.1007/s11357-023-00831-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/17/2023] [Indexed: 06/14/2023] Open
Abstract
Differences in brain structure and functional and structural network architecture have been found to partly explain cognitive performance differences in older ages. Thus, they may serve as potential markers for these differences. Initial unimodal studies, however, have reported mixed prediction results of selective cognitive variables based on these brain features using machine learning (ML). Thus, the aim of the current study was to investigate the general validity of cognitive performance prediction from imaging data in healthy older adults. In particular, the focus was with examining whether (1) multimodal information, i.e., region-wise grey matter volume (GMV), resting-state functional connectivity (RSFC), and structural connectivity (SC) estimates, may improve predictability of cognitive targets, (2) predictability differences arise for global cognition and distinct cognitive profiles, and (3) results generalize across different ML approaches in 594 healthy older adults (age range: 55-85 years) from the 1000BRAINS study. Prediction potential was examined for each modality and all multimodal combinations, with and without confound (i.e., age, education, and sex) regression across different analytic options, i.e., variations in algorithms, feature sets, and multimodal approaches (i.e., concatenation vs. stacking). Results showed that prediction performance differed considerably between deconfounding strategies. In the absence of demographic confounder control, successful prediction of cognitive performance could be observed across analytic choices. Combination of different modalities tended to marginally improve predictability of cognitive performance compared to single modalities. Importantly, all previously described effects vanished in the strict confounder control condition. Despite a small trend for a multimodal benefit, developing a biomarker for cognitive aging remains challenging.
Collapse
Affiliation(s)
- Camilla Krämer
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Johanna Stumme
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lucas da Costa Campos
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Paulo Dellani
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Rubbert
- Department of Diagnostic and Interventional Radiology, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julian Caspers
- Department of Diagnostic and Interventional Radiology, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christiane Jockwitz
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany.
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
13
|
Kawade N, Yamanaka K. Novel insights into brain lipid metabolism in Alzheimer's disease: Oligodendrocytes and white matter abnormalities. FEBS Open Bio 2024; 14:194-216. [PMID: 37330425 PMCID: PMC10839347 DOI: 10.1002/2211-5463.13661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A genome-wide association study has shown that several AD risk genes are involved in lipid metabolism. Additionally, epidemiological studies have indicated that the levels of several lipid species are altered in the AD brain. Therefore, lipid metabolism is likely changed in the AD brain, and these alterations might be associated with an exacerbation of AD pathology. Oligodendrocytes are glial cells that produce the myelin sheath, which is a lipid-rich insulator. Dysfunctions of the myelin sheath have been linked to white matter abnormalities observed in the AD brain. Here, we review the lipid composition and metabolism in the brain and myelin and the association between lipidic alterations and AD pathology. We also present the abnormalities in oligodendrocyte lineage cells and white matter observed in AD. Additionally, we discuss metabolic disorders, including obesity, as AD risk factors and the effects of obesity and dietary intake of lipids on the brain.
Collapse
Affiliation(s)
- Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityJapan
- Center for One Medicine Innovative Translational Research (COMIT)Nagoya UniversityJapan
| |
Collapse
|
14
|
Shui L, Shibata D, Chan KCG, Zhang W, Sung J, Haynor DR. Longitudinal Relationship Between Brain Atrophy Patterns, Cognitive Decline, and Cerebrospinal Fluid Biomarkers in Alzheimer's Disease Explored by Orthonormal Projective Non-Negative Matrix Factorization. J Alzheimers Dis 2024; 98:969-986. [PMID: 38517788 DOI: 10.3233/jad-231149] [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] [Indexed: 03/24/2024]
Abstract
Background Longitudinal magnetic resonance imaging (MRI) has been proposed for tracking the progression of Alzheimer's disease (AD) through the assessment of brain atrophy. Objective Detection of brain atrophy patterns in patients with AD as the longitudinal disease tracker. Methods We used a refined version of orthonormal projective non-negative matrix factorization (OPNMF) to identify six distinct spatial components of voxel-wise volume loss in the brains of 83 subjects with AD from the ADNI3 cohort relative to healthy young controls from the ABIDE study. We extracted non-negative coefficients representing subject-specific quantitative measures of regional atrophy. Coefficients of brain atrophy were compared to subjects with mild cognitive impairment and controls, to investigate the cross-sectional and longitudinal associations between AD biomarkers and regional atrophy severity in different groups. We further validated our results in an independent dataset from ADNI2. Results The six non-overlapping atrophy components represent symmetric gray matter volume loss primarily in frontal, temporal, parietal and cerebellar regions. Atrophy in these regions was highly correlated with cognition both cross-sectionally and longitudinally, with medial temporal atrophy showing the strongest correlations. Subjects with elevated CSF levels of TAU and PTAU and lower baseline CSF Aβ42 values, demonstrated a tendency toward a more rapid increase of atrophy. Conclusions The present study has applied a transferable method to characterize the imaging changes associated with AD through six spatially distinct atrophy components and correlated these atrophy patterns with cognitive changes and CSF biomarkers cross-sectionally and longitudinally, which may help us better understand the underlying pathology of AD.
Collapse
Affiliation(s)
- Lan Shui
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- National Alzheimer's Coordinating Center, Seattle, WA, USA
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX, USA
| | - Dean Shibata
- Department of Radiology, University of Washington, Seattle, WA, USA
- National Alzheimer's Coordinating Center, Seattle, WA, USA
| | - Kwun Chuen Gary Chan
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- National Alzheimer's Coordinating Center, Seattle, WA, USA
| | - Wenbo Zhang
- Department of Statistics, University of California Irvine, CA, USA
| | - Junhyoun Sung
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - David R Haynor
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
15
|
Ten Kate M, Barkhof F, Schwarz AJ. Consistency between Treatment Effects on Clinical and Brain Atrophy Outcomes in Alzheimer's Disease Trials. J Prev Alzheimers Dis 2024; 11:38-47. [PMID: 38230715 PMCID: PMC10994869 DOI: 10.14283/jpad.2023.92] [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: 04/17/2023] [Accepted: 06/17/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND Longitudinal changes in volumetric MRI outcome measures have been shown to correlate well with longitudinal changes in clinical instruments and have been widely used as biomarker outcomes in clinical trials for Alzheimer's disease (AD). While instances of discordant findings have been noted in some trials, especially the recent amyloid-removing therapies, the overall relationship between treatment effects on brain atrophy and clinical outcomes, and how it might depend on treatment target or mechanism, clinical instrument or imaging variable is not yet clear. OBJECTIVE To systematically assess the consistency and therapeutic class-dependence of treatment effects on clinical outcomes and on brain atrophy in published reports of clinical trials conducted in mild cognitive impairment (MCI) and/or AD. DESIGN Quantitative review of the published literature. The consistency of treatment effects on clinical and brain atrophy outcomes was assessed in terms of statistical agreement with hypothesized equal magnitude effects (e.g., 30% slowing of both) and nominal directional concordance, as a function of therapeutic class. SETTING Interventional randomized clinical trials. PARTICIPANTS MCI or AD trial participants. INTERVENTION Treatments included were those that involved ingestion or injection of a putatively active substance into the body, encompassing both pharmacological and controlled dietary interventions. MEASUREMENTS Each trial included in the analysis reported at least one of the required clinical outcomes (ADAS-Cog, CDR-SB or MMSE) and at least one of the required imaging outcomes (whole brain, ventricular or hippocampal volume). RESULTS Data from 35 trials, comprising 185 pairwise comparisons, were included. Overall, the 95% confidence bounds overlapped with the line of identity for 150/185 (81%) of the imaging-clinical variable pairs. The greatest proportion of outliers was found in trials of anti-amyloid antibodies that have been shown to dramatically reduce the level of PET-detectable amyloid plaques, for which only 13/33 (39%) of observations overlapped the identity line. A Deming regression calculated using all data points yielded a slope of 0.54, whereas if data points from the amyloid remover class were excluded, the Deming regression line had a slope of 0.92. Directional discordance of treatment effects was also most pronounced for the amyloid-removing class, and for comparisons involving ventricular volume. CONCLUSION Our results provide a frame of reference for the interpretation of clinical and brain atrophy results from future clinical trials and highlight the importance of mechanism of action in the interpretation of imaging results.
Collapse
Affiliation(s)
- M Ten Kate
- Adam J Schwarz, PhD, Takeda Pharmaceuticals, Ltd., 40 Landsdowne St., Cambridge MA 02139, USA Tel: (+1) 317 282 3557,
| | | | | |
Collapse
|
16
|
Kim GW, Park K, Jeong GW. Early Detection of Alzheimer's Disease in Postmenopausal Women Using Thalamic Subnuclear Volumetry. J Clin Med 2023; 12:6844. [PMID: 37959308 PMCID: PMC10648434 DOI: 10.3390/jcm12216844] [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: 09/18/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) and aging are intrinsically interconnected with each other and are mediated by molecular, cellular, and biological systems. In particular, a specific pattern of brain volume atrophy is the most profound risk factor for cognitive impairment, including AD, that is directly linked to aging. Thus, this study aimed to investigate knowledge on the early detection of AD in postmenopausal women, focusing on the volume changes of the subcortical regions, including the thalamic subnuclei, in women with AD vs. postmenopausal women. Twenty-one women with AD and twenty-one postmenopausal women without AD underwent magnetic resonance imaging (MRI). Women with AD showed significantly reduced volumes in the hippocampus, thalamus, and amygdala compared with postmenopausal women (p < 0.05, FWE-corrected). After adjustments for age, the right hippocampal volume was found to be significantly lower in the women with AD, but the volumes of the thalamus and amygdala were relatively unaffected. The women with AD exhibited significantly reduced volume in the right laterodorsal nucleus of the thalamus compared with the postmenopausal women (p < 0.05, Bonferroni-corrected). Our findings suggest that the reduced volume of both the right laterodorsal thalamic nucleus and right hippocampus may serve as a potential biomarker for the early detection of AD in postmenopausal women.
Collapse
Affiliation(s)
- Gwang-Won Kim
- Advanced Institute of Aging Science, Chonnam National University, Gwangju 61186, Republic of Korea; (G.-W.K.); (K.P.)
| | - Kwangsung Park
- Advanced Institute of Aging Science, Chonnam National University, Gwangju 61186, Republic of Korea; (G.-W.K.); (K.P.)
- Department of Urology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Gwang-Woo Jeong
- Department of Radiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
17
|
Pan X, Cheng X, Zhang J, Xia Y, Zhong C, Fei G. A comparison of the five-minute cognitive test with the mini-mental state examination in the elderly for cognitive impairment screening. Front Neurosci 2023; 17:1146552. [PMID: 37378012 PMCID: PMC10292014 DOI: 10.3389/fnins.2023.1146552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
The five-minute cognitive test (FCT) is a novel cognitive screening method with the quick and reliable merit for detecting cognitive impairment at an early stage. The diagnostic power of FCT in differentiating subjects with cognitive impairment from people with cognition in a normal range was demonstrated effective as that of the Mini-Mental Status Evaluation (MMSE) in a previous cohort study. Here, we analyzed the effect of sociodemographic and health-related factors on FCT performance and further investigated the consistency of FCT. Then, we compared the correlation of subitem scores of FCT or MMSE with a comprehensive battery of neuropsychological tests that focus on specific domains of cognition. Finally, the association of the total FCT scores with the volumes of brain subregions was investigated. There were 360 subjects aged 60 years or above enrolled in this study, including 226 adults with cognitive abilities in normal range, 107 subjects with mild cognitive impairment (MCI) and 27 mild Alzheimer's disease (AD). The results showed that the total FCT scores was negatively associated with increasing age (β = -0.146, p < 0.001), and positively associated with education attainment (β = 0.318, p < 0.001), dwelling condition with family (β = 0.153, p < 0.001) and the Body Mass Index (β = 1.519, p < 0.01). The internal consistency of the FCT (Cronbach's α) was 0.644. The sub-scores of FCT showed a significant correlation with other specific neuropsychological tests. Impressively, the total FCT scores showed a significantly positive association with the volumes of hippocampus related subregions (r = 0.523, p < 0.001) and amygdala (r = 0.479, p < 0.001), but not with cerebellum (r = 0.158, p > 0.05) or subcortical subregions (r = 0.070, p > 0.05). Combining with previous data, FCT is a reliable and valid cognitive screening test for detecting cognitive impairment in a community setting.
Collapse
Affiliation(s)
- Xiaoli Pan
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Xiamen, Fujian, China
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoqin Cheng
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingfeng Xia
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guoqiang Fei
- Department of Neurology, Zhongshan Hospital (Xiamen Branch), Fudan University, Xiamen, Fujian, China
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Wright LM, De Marco M, Venneri A. Current Understanding of Verbal Fluency in Alzheimer's Disease: Evidence to Date. Psychol Res Behav Manag 2023; 16:1691-1705. [PMID: 37179686 PMCID: PMC10167999 DOI: 10.2147/prbm.s284645] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/05/2023] [Indexed: 05/15/2023] Open
Abstract
Since their development, verbal fluency tests (VFTs) have been used extensively throughout research and in clinical settings to assess a variety of cognitive functions in diverse populations. In Alzheimer's disease (AD), these tasks have proven particularly valuable in identifying the earliest forms of cognitive decline in semantic processing and have been shown to relate specifically to brain regions associated with the initial stages of pathological change. In recent years, researchers have developed more nuanced techniques to evaluate verbal fluency performance, extracting a wide range of cognitive metrics from these simple neuropsychological tests. Such novel techniques allow for a more detailed exploration of the cognitive processes underlying successful task performance beyond the raw test score. The versatility of VFTs and the richness of data they may provide, in light of their low cost and speed of administration, therefore, highlight their potential value both in future research as outcome measures for clinical trials and in a clinical setting as a screening measure for early detection of neurodegenerative diseases.
Collapse
Affiliation(s)
- Laura M Wright
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Matteo De Marco
- Department of Life Sciences, Brunel University London, London, UK
| | - Annalena Venneri
- Department of Life Sciences, Brunel University London, London, UK
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
19
|
Kaplan H, Hooper PL, Gatz M, Mack WJ, Law EM, Chui HC, Sutherland ML, Sutherland JD, Rowan CJ, Wann LS, Allam AH, Thompson RC, Michalik DE, Lombardi G, Miyamoto MI, Eid Rodriguez D, Copajira Adrian J, Quispe Gutierrez R, Beheim BA, Cummings DK, Seabright E, Alami S, R. Garcia A, Buetow K, Thomas GS, Finch CE, Stieglitz J, Trumble BC, Gurven MD, Irimia A. Brain volume, energy balance, and cardiovascular health in two nonindustrial South American populations. Proc Natl Acad Sci U S A 2023; 120:e2205448120. [PMID: 36940322 PMCID: PMC10068758 DOI: 10.1073/pnas.2205448120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 01/24/2023] [Indexed: 03/22/2023] Open
Abstract
Little is known about brain aging or dementia in nonindustrialized environments that are similar to how humans lived throughout evolutionary history. This paper examines brain volume (BV) in middle and old age among two indigenous South American populations, the Tsimane and Moseten, whose lifestyles and environments diverge from those in high-income nations. With a sample of 1,165 individuals aged 40 to 94, we analyze population differences in cross-sectional rates of decline in BV with age. We also assess the relationships of BV with energy biomarkers and arterial disease and compare them against findings in industrialized contexts. The analyses test three hypotheses derived from an evolutionary model of brain health, which we call the embarrassment of riches (EOR). The model hypothesizes that food energy was positively associated with late life BV in the physically active, food-limited past, but excess body mass and adiposity are now associated with reduced BV in industrialized societies in middle and older ages. We find that the relationship of BV with both non-HDL cholesterol and body mass index is curvilinear, positive from the lowest values to 1.4 to 1.6 SDs above the mean, and negative from that value to the highest values. The more acculturated Moseten exhibit a steeper decrease in BV with age than Tsimane, but still shallower than US and European populations. Lastly, aortic arteriosclerosis is associated with lower BV. Complemented by findings from the United States and Europe, our results are consistent with the EOR model, with implications for interventions to improve brain health.
Collapse
Affiliation(s)
- Hillard Kaplan
- Economic Science Institute, Chapman University, Orange, CA82866
| | - Paul L. Hooper
- Economic Science Institute, Chapman University, Orange, CA82866
- Department of Anthropology, University of New Mexico, Albuquerque, NM87131
| | - Margaret Gatz
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA90089
| | - Wendy J. Mack
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - E. Meng Law
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
- Department of Radiology, The Alfred Health Hospital, Melbourne, VIC3004, Australia
- iBRAIN Research Laboratory, Departments of Neuroscience, Computer Systems and Electrical Engineering, Monash University, Melbourne, VIC3800, Australia
| | - Helena C. Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
- Alzheimer’s Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | | | | | - Christopher J. Rowan
- Renown Institute for Heart and Vascular Health, Reno, NV89502
- School of Medicine, University of Nevada, Reno, NV89557
| | - L. Samuel Wann
- Division of Cardiology, University of New Mexico, Albuquerque, NM87131
| | - Adel H. Allam
- Department of Cardiology, School of Medicine, Al-Azhar University, Al Mikhaym Al Daem, Cairo4334003, Egypt
| | - Randall C. Thompson
- Saint Luke’s Mid America Heart Institute, University of Missouri - Kansas City, Kansas City, MO64111
| | - David E. Michalik
- Department of Pediatrics, School of Medicine, University of California at Irvine, Orange, CA92617
- MemorialCare Miller Children’s and Women’s Hospital, Long Beach, CA90806
| | - Guido Lombardi
- Laboratorio de Paleopatologia, Catedra Pedro Weiss, Universidad Peruana Cayetano Heredia, Lima15102, Peru
| | | | | | | | | | - Bret A. Beheim
- Department of Human Behavior, Ecology and Culture, Max Planck Institute for Evolutionary Anthropology, Leipzig04103, Germany
| | | | - Edmond Seabright
- Department of Anthropology, University of New Mexico, Albuquerque, NM87131
- School of Collective Intelligence, Universite Mohammed 6 Polytechnic, Ben Guerir43150, Morocco
| | - Sarah Alami
- School of Collective Intelligence, Universite Mohammed 6 Polytechnic, Ben Guerir43150, Morocco
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA93106
| | - Angela R. Garcia
- Scientific Research Core, Phoenix Children’s Hospital, Phoenix, AZ85016
- Department of Child Health, University of Arizona, Tucson, AZ85724
| | - Kenneth Buetow
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ85287
| | - Gregory S. Thomas
- MemorialCare Health Systems, Fountain Valley, CA92708
- Division of Cardiology, University of California, Irvine, Orange, CA92868
| | - Caleb E. Finch
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA90089
- Department of Biological Sciences, Anthropology and Psychology, University of Southern California, Los Angeles, CA90089
| | - Jonathan Stieglitz
- Institute for Advanced Study in Toulouse, Toulouse 1 Capitole University, Toulouse31000, France
| | - Benjamin C. Trumble
- Center for Evolution and Medicine, School of Human Evolution and Social Change, Arizona State University, Tempe, AZ85287
| | - Michael D. Gurven
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA93106
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA90089
- Corwin D. Denney Research Center, Department of Biomedical Engineering, University of Southern California, Los Angeles, CA90089
| |
Collapse
|
20
|
Lau CI, Yeh JH, Tsai YF, Hsiao CY, Wu YT, Jao CW. Decreased Brain Structural Network Connectivity in Patients with Mild Cognitive Impairment: A Novel Fractal Dimension Analysis. Brain Sci 2023; 13:brainsci13010093. [PMID: 36672073 PMCID: PMC9856782 DOI: 10.3390/brainsci13010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/18/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Mild cognitive impairment (MCI) is widely regarded to be the intermediate stage to Alzheimer's disease. Cerebral morphological alteration in cortical subregions can provide an accurate predictor for early recognition of MCI. Thirty patients with MCI and thirty healthy control subjects participated in this study. The Desikan-Killiany cortical atlas was applied to segment participants' cerebral cortex into 68 subregions. A complexity measure termed fractal dimension (FD) was applied to assess morphological changes in cortical subregions of participants. The MCI group revealed significantly decreased FD values in the bilateral temporal lobes, right parietal lobe including the medial temporal, fusiform, para hippocampal, and also the orbitofrontal lobes. We further proposed a novel FD-based brain structural network to compare network parameters, including intra- and inter-lobular connectivity between groups. The control group had five modules, and the MCI group had six modules in their brain networks. The MCI group demonstrated shrinkage of modular sizes with fewer components integrated, and significantly decreased global modularity in the brain network. The MCI group had lower intra- and inter-lobular connectivity in all lobes. Between cerebral lobes, the MCI patients may maintain nodal connections between both hemispheres to reduce connectivity loss in the lateral hemispheres. The method and results presented in this study could be a suitable tool for early detection of MCI.
Collapse
Affiliation(s)
- Chi Ieong Lau
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
- Dementia Center, Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
- Applied Cognitive Neuroscience Group, Institute of Cognitive Neuroscience, University College London, London WC1N 3AZ, UK
- Department of Neurology, University Hospital, Taipa 999078, Macau
| | - Jiann-Horng Yeh
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
| | - Yuh-Feng Tsai
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei 242, Taiwan
- Department of Diagnostic Radiology, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan
| | - Chen-Yu Hsiao
- Department of Diagnostic Radiology, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan
| | - Yu-Te Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Correspondence: (Y.-T.W.); (C.-W.J.); Tel.: +886-02-28267169 (Y.-T.W.); +886-02-28267394 (C.-W.J.)
| | - Chi-Wen Jao
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
- Correspondence: (Y.-T.W.); (C.-W.J.); Tel.: +886-02-28267169 (Y.-T.W.); +886-02-28267394 (C.-W.J.)
| |
Collapse
|
21
|
Hopper A, Salans M, Karunamuni R, Hattangadi-Gluth JA. Neurocognitive considerations in the treatment of meningioma with radiation therapy: applications for quantitative neuroimaging and precision radiation medicine. J Neurooncol 2023; 161:277-286. [PMID: 36572802 DOI: 10.1007/s11060-022-04175-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 12/27/2022]
Abstract
This article focuses on the role of radiotherapy in the management of meningioma, in the definitive and adjuvant setting and across the spectrum of meningioma grade. Treatment paradigms, informed by clinical evidence, are discussed. Notably, we focus on the impact of radiotherapy on normal brain tissues and neurocognitive function, particularly the dose-dependent changes in white matter and cerebral cortex thickness. Novel imaging techniques have allowed the identification of microstructural changes to eloquent white matter, cortex, and subcortical regions as biomarkers for understanding RT-induced changes in cognitive functioning. Deficits in multiple domains including attention, memory, language and executive function can become more pronounced following radiation. Longitudinal assessment with imaging and neurocognitive testing pre- and post-radiation have allowed correlation between dose to specific regions of the brain and decline in associated domains of neurocognitive function. These findings suggest incorporation of areas at higher risk for neurocognitive sequelae into precision radiation planning. Volumetric arc therapy, advanced planning with cortical sparing, proton therapy and stereotactic radiosurgery are reviewed as options for delivering therapeutic dose to target volumes while minimizing risk to adjacent sensitive regions. The treatment of meningioma is an evolving area, with improving outcomes for higher grade disease in modern trials, where care must be taken to maximize both disease control as well as quality of life for patients.
Collapse
Affiliation(s)
- Austin Hopper
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 9500 Gilman Dr., La Jolla, Mail Code 0861, San Diego, CA, 92093-0861, USA
| | - Mia Salans
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 9500 Gilman Dr., La Jolla, Mail Code 0861, San Diego, CA, 92093-0861, USA
| | - Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 9500 Gilman Dr., La Jolla, Mail Code 0861, San Diego, CA, 92093-0861, USA
| | - Jona A Hattangadi-Gluth
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 9500 Gilman Dr., La Jolla, Mail Code 0861, San Diego, CA, 92093-0861, USA.
| |
Collapse
|
22
|
Carlos AF, Machulda MM, Rutledge MH, Nguyen AT, Reichard RR, Baker MC, Rademakers R, Dickson DW, Petersen RC, Josephs KA. Comparison of Clinical, Genetic, and Pathologic Features of Limbic and Diffuse Transactive Response DNA-Binding Protein 43 Pathology in Alzheimer's Disease Neuropathologic Spectrum. J Alzheimers Dis 2023; 93:1521-1535. [PMID: 37182869 PMCID: PMC10923399 DOI: 10.3233/jad-221094] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Increasing evidence suggests that TAR DNA-binding protein 43 (TDP-43) pathology in Alzheimer's disease (AD), or AD-TDP, can be diffuse or limbic-predominant. Understanding whether diffuse AD-TDP has genetic, clinical, and pathological features that differ from limbic AD-TDP could have clinical and research implications. OBJECTIVE To better characterize the clinical and pathologic features of diffuse AD-TDP and differentiate it from limbic AD-TDP. METHODS 363 participants from the Mayo Clinic Study of Aging, Alzheimer's Disease Research Center, and Neurodegenerative Research Group with autopsy confirmed AD and TDP-43 pathology were included. All underwent genetic, clinical, neuropsychologic, and neuropathologic evaluations. AD-TDP pathology distribution was assessed using the Josephs 6-stage scale. Stages 1-3 were classified as Limbic, those 4-6 as Diffuse. Multivariable logistic regression was used to identify clinicopathologic features that independently predicted diffuse pathology. RESULTS The cohort was 61% female and old at onset (median: 76 years [IQR:70-82]) and death (median: 88 years [IQR:82-92]). Fifty-four percent were Limbic and 46% Diffuse. Clinically, ∼10-20% increases in odds of being Diffuse associated with 5-year increments in age at onset (p = 0.04), 1-year longer disease duration (p = 0.02), and higher Neuropsychiatric Inventory scores (p = 0.03), while 15-second longer Trailmaking Test-B times (p = 0.02) and higher Block Design Test scores (p = 0.02) independently decreased the odds by ~ 10-15%. There was evidence for association of APOEɛ4 allele with limbic AD-TDP and of TMEM106B rs3173615 C allele with diffuse AD-TDP. Pathologically, widespread amyloid-β plaques (Thal phases: 3-5) decreased the odds of diffuse TDP-43 pathology by 80-90%, while hippocampal sclerosis increased it sixfold (p < 0.001). CONCLUSION Diffuse AD-TDP shows clinicopathologic and genetic features different from limbic AD-TDP.
Collapse
Affiliation(s)
- Arenn F. Carlos
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M. Machulda
- Department of Psychology and Psychiatry, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew C. Baker
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rosa Rademakers
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
- VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Flanders 2000, Belgium
| | - Dennis W. Dickson
- Department of Neuroscience (Neuropathology), Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | |
Collapse
|
23
|
Jiang W, Tian Y, Fan F, Fu F, Wei D, Tang S, Chen J, Li Y, Zhu R, Wang L, Shi Z, Wang D, Zhang XY. Effects of comorbid posttraumatic stress disorder on cognitive dysfunction in Chinese male methamphetamine patients. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110611. [PMID: 35907518 DOI: 10.1016/j.pnpbp.2022.110611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 10/16/2022]
Abstract
OBJECTIVES Cognitive dysfunction and posttraumatic stress disorder (PTSD) are common in methamphetamine patients. However, few studies have investigated the cognitive performance of methamphetamine patients with PTSD. The purpose of this study was to investigate the impact of comorbid PTSD on cognitive function in Chinese male methamphetamine patients. METHODS We analyzed 464 methamphetamine patients and 156 healthy volunteers. The PTSD Screening Scale (PCL-5) was used to assess PTSD and the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) was used to assess cognitive function. RESULTS Compared with healthy controls, methamphetamine patients had more cognitive dysfunction in immediate memory, visuospatial/constructional, language, attention and delayed memory. Moreover, methamphetamine patients with PTSD had less cognitive dysfunction in immediate memory, attention, and delayed memory than methamphetamine patients without PTSD. Further stepwise regression analysis showed that PTSD alterations in arousal and reactivity cluster were risk predictors for language, and PTSD negative alteration in cognition and mood cluster were risk predictors for delayed memory. CONCLUSIONS Our results indicate that methamphetamine patients without PTSD have poorer cognitive dysfunction than those with PTSD. Some demographic and PTSD symptom clusters are protective or risk factors for cognitive dysfunction in methamphetamine patients.
Collapse
Affiliation(s)
- Wei Jiang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Tian
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Fusheng Fan
- Xin Hua Drug Rehabilitation Center, Sichuan, China
| | - Fabing Fu
- Xin Hua Drug Rehabilitation Center, Sichuan, China
| | - Dejun Wei
- Xin Hua Drug Rehabilitation Center, Sichuan, China
| | | | - Jiajing Chen
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yuqing Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Rongrong Zhu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Li Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Zhanbiao Shi
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| | - Dongmei Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| | - Xiang-Yang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
24
|
Svenningsson AL, Stomrud E, Palmqvist S, Hansson O, Ossenkoppele R. Axonal degeneration and amyloid pathology predict cognitive decline beyond cortical atrophy. Alzheimers Res Ther 2022; 14:144. [PMID: 36192766 PMCID: PMC9531524 DOI: 10.1186/s13195-022-01081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 09/11/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Cortical atrophy is associated with cognitive decline, but the association is not perfect. We aimed to identify factors explaining the discrepancy between the degree of cortical atrophy and cognitive decline in cognitively unimpaired elderly. METHODS The discrepancy between atrophy and cognitive decline was measured using the residuals from a linear regression analysis between change in whole brain cortical thickness over time and change in a cognitive composite measure over time in 395 cognitively unimpaired participants from the Swedish BioFINDER study. We tested for bivariate associations of this residual measure with demographic, imaging, and fluid biomarker variables using Pearson correlations and independent-samples t-tests, and for multivariate associations using linear regression models. Mediation analyses were performed to explore possible paths between the included variables. RESULTS In bivariate analyses, older age (r = -0.11, p = 0.029), male sex (t = -3.00, p = 0.003), larger intracranial volume (r = -0.17, p < 0.001), carrying an APOEe4 allele (t = -2.71, p = 0.007), larger white matter lesion volume (r = -0.16, p = 0.002), lower cerebrospinal fluid (CSF) β-amyloid (Aβ) 42/40 ratio (t = -4.05, p < 0.001), and higher CSF levels of phosphorylated tau (p-tau) 181 (r = -0.22, p < 0.001), glial fibrillary acidic protein (GFAP; r = -0.15, p = 0.003), and neurofilament light (NfL; r = -0.34, p < 0.001) were negatively associated with the residual measure, i.e., associated with worse than expected cognitive trajectory given the level of atrophy. In a multivariate analysis, only lower CSF Aβ42/40 ratio and higher CSF NfL levels explained cognition beyond brain atrophy. Mediation analyses showed that associations between the residual measure and APOEe4 allele, CSF Aβ42/40 ratio, and CSF GFAP and p-tau181 levels were mediated by levels of CSF NfL, as were the associations with the residual measure for age, sex, and WML volume. CONCLUSIONS Our results suggest that axonal degeneration and amyloid pathology independently affect the rate of cognitive decline beyond the degree of cortical atrophy. Furthermore, axonal degeneration mediated the negative effects of old age, male sex, and white matter lesions, and in part also amyloid and tau pathology, on cognition over time when accounting for cortical atrophy.
Collapse
Affiliation(s)
- Anna Linnéa Svenningsson
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Erik Stomrud
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Sebastian Palmqvist
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Rik Ossenkoppele
- grid.4514.40000 0001 0930 2361Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE 205 02 Malmö, Sweden ,grid.484519.5Alzheimer Center Amsterdam, Department of Neurology, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
25
|
Fang Y, Doyle MF, Chen J, Alosco ML, Mez J, Satizabal CL, Qiu WQ, Murabito JM, Lunetta KL. Association between inflammatory biomarkers and cognitive aging. PLoS One 2022; 17:e0274350. [PMID: 36083988 PMCID: PMC9462682 DOI: 10.1371/journal.pone.0274350] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Inflammatory cytokines and chemokines related to the innate and adaptive immune system have been linked to neuroinflammation in Alzheimer's Disease, dementia, and cognitive disorders. We examined the association of 11 plasma proteins (CD14, CD163, CD5L, CD56, CD40L, CXCL16, SDF1, DPP4, SGP130, sRAGE, and MPO) related to immune and inflammatory responses with measures of cognitive function, brain MRI and dementia risk. We identified Framingham Heart Study Offspring participants who underwent neuropsychological testing (n = 2358) or brain MRI (n = 2100) within five years of the seventh examination where a blood sample for quantifying the protein biomarkers was obtained; and who were followed for 10 years for incident all-cause dementia (n = 1616). We investigated the association of inflammatory biomarkers with neuropsychological test performance and brain MRI volumes using linear mixed effect models accounting for family relationships. We further used Cox proportional hazards models to examine the association with incident dementia. False discovery rate p-values were used to account for multiple testing. Participants included in the neuropsychological test and MRI samples were on average 61 years old and 54% female. Participants from the incident dementia sample (average 68 years old at baseline) included 124 participants with incident dementia. In addition to CD14, which has an established association, we found significant associations between higher levels of CD40L and myeloperoxidase (MPO) with executive dysfunction. Higher CD5L levels were significantly associated with smaller total brain volumes (TCBV), whereas higher levels of sRAGE were associated with larger TCBV. Associations persisted after adjustment for APOE ε4 carrier status and additional cardiovascular risk factors. None of the studied inflammatory biomarkers were significantly associated with risk of incident all-cause dementia. Higher circulating levels of soluble CD40L and MPO, markers of immune cell activation, were associated with poorer performance on neuropsychological tests, while higher CD5L, a key regulator of inflammation, was associated with smaller total brain volumes. Higher circulating soluble RAGE, a decoy receptor for the proinflammatory RAGE/AGE pathway, was associated with larger total brain volume. If confirmed in other studies, this data indicates the involvement of an activated immune system in abnormal brain aging.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| | - Margaret F. Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Jiachen Chen
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| | - Michael L. Alosco
- Boston University Alzheimer’s Disease Research Center and CTE Center, School of Medicine, Boston University, Boston, Massachusetts, United States of America
- Department of Neurology, School of Medicine, Boston University, Boston, Massachusetts, United States of America
| | - Jesse Mez
- Boston University Alzheimer’s Disease Research Center and CTE Center, School of Medicine, Boston University, Boston, Massachusetts, United States of America
- Department of Neurology, School of Medicine, Boston University, Boston, Massachusetts, United States of America
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, United States of America
| | - Claudia L. Satizabal
- Department of Neurology, School of Medicine, Boston University, Boston, Massachusetts, United States of America
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Wei Qiao Qiu
- Boston University Alzheimer’s Disease Research Center and CTE Center, School of Medicine, Boston University, Boston, Massachusetts, United States of America
- Department of Psychiatry, School of Medicine, Boston University, Boston, Massachusetts, United States of America
- Department of Pharmacology & Experimental Therapeutics, School of Medicine, Boston University, Boston, Massachusetts, United States of America
| | - Joanne M. Murabito
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, United States of America
- Department of Medicine, Section of General Internal Medicine, School of Medicine, Boston University, Boston, Massachusetts, United States of America
- Boston Medical Center, Boston University, Boston, Massachusetts, United States of America
| | - Kathryn L. Lunetta
- Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
26
|
Tosun D, Demir Z, Veitch DP, Weintraub D, Aisen P, Jack CR, Jagust WJ, Petersen RC, Saykin AJ, Shaw LM, Trojanowski JQ, Weiner MW. Contribution of Alzheimer's biomarkers and risk factors to cognitive impairment and decline across the Alzheimer's disease continuum. Alzheimers Dement 2022; 18:1370-1382. [PMID: 34647694 PMCID: PMC9014819 DOI: 10.1002/alz.12480] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/10/2021] [Accepted: 08/15/2021] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Amyloid beta (Aβ), tau, and neurodegeneration jointly with the Alzheimer's disease (AD) risk factors affect the severity of clinical symptoms and disease progression. METHODS Within 248 Aβ-positive elderly with and without cognitive impairment and dementia, partial least squares structural equation pathway modeling was used to assess the direct and indirect effects of imaging biomarkers (global Aβ-positron emission tomography [PET] uptake, regional tau-PET uptake, and regional magnetic resonance imaging-based atrophy) and risk-factors (age, sex, education, apolipoprotein E [APOE], and white-matter lesions) on cross-sectional cognitive impairment and longitudinal cognitive decline. RESULTS Sixteen percent of variance in cross-sectional cognitive impairment was accounted for by Aβ, 46% to 47% by tau, and 25% to 29% by atrophy, although 53% to 58% of total variance in cognitive impairment was explained by incorporating mediated and direct effects of AD risk factors. The Aβ-tau-atrophy pathway accounted for 50% to 56% of variance in longitudinal cognitive decline while Aβ, tau, and atrophy independently explained 16%, 46% to 47%, and 25% to 29% of the variance, respectively. DISCUSSION These findings emphasize that treatments that remove Aβ and completely stop downstream effects on tau and neurodegeneration would only be partially effective in slowing of cognitive decline or reversing cognitive impairment.
Collapse
Affiliation(s)
- Duygu Tosun
- San Francisco Veterans Affairs Medical CenterSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Zeynep Demir
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Dallas P. Veitch
- San Francisco Veterans Affairs Medical CenterSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Daniel Weintraub
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Paul Aisen
- Alzheimer's Therapeutic Research Institute (ATRI)Keck School of MedicineUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | | | - William J. Jagust
- School of Public Health and Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Ronald C. Petersen
- Division of EpidemiologyDepartment of Health Sciences ResearchMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Andrew J. Saykin
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Michael W. Weiner
- San Francisco Veterans Affairs Medical CenterSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | |
Collapse
|
27
|
Egorova-Brumley N, Khlif MS, Werden E, Bird LJ, Brodtmann A. Grey and white matter atrophy one year after stroke aphasia. Brain Commun 2022; 4:fcac061. [PMID: 35368613 PMCID: PMC8971893 DOI: 10.1093/braincomms/fcac061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/23/2021] [Accepted: 03/15/2022] [Indexed: 11/20/2022] Open
Abstract
Dynamic whole-brain changes occur following stroke, and not just in association with recovery. We tested the hypothesis that the presence of a specific behavioural deficit after stroke would be associated with structural decline (atrophy) in the brain regions supporting the affected function, by examining language deficits post-stroke. We quantified whole-brain structural volume changes longitudinally (3–12 months) in stroke participants with (N = 32) and without aphasia (N = 59) as assessed by the Token Test at 3 months post-stroke, compared with a healthy control group (N = 29). While no significant difference in language decline rates (change in Token Test scores from 3 to 12 months) was observed between groups and some participants in the aphasic group improved their scores, stroke participants with aphasia symptoms at 3 months showed significant atrophy (>2%, P = 0.0001) of the left inferior frontal gyrus not observed in either healthy control or non-aphasic groups over the 3–12 months period. We found significant group differences in the inferior frontal gyrus volume, accounting for age, sex, stroke severity at baseline, education and total intracranial volume (Bonferroni-corrected P = 0.0003). In a subset of participants (aphasic N = 14, non-aphasic N = 36, and healthy control N = 25) with available diffusion-weighted imaging data, we found significant atrophy in the corpus callosum and the left superior longitudinal fasciculus in the aphasic compared with the healthy control group. Language deficits at 3 months post-stroke are associated with accelerated structural decline specific to the left inferior frontal gyrus, highlighting that known functional brain reorganization underlying behavioural improvement may occur in parallel with atrophy of brain regions supporting the language function.
Collapse
Affiliation(s)
- Natalia Egorova-Brumley
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| | - Mohamed Salah Khlif
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Emilio Werden
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Laura J. Bird
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Amy Brodtmann
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| |
Collapse
|
28
|
Fang Y, Doyle MF, Alosco ML, Mez J, Satizabal CL, Qiu WQ, Lunetta KL, Murabito JM. Cross-Sectional Association Between Blood Cell Phenotypes, Cognitive Function, and Brain Imaging Measures in the Community-Based Framingham Heart Study. J Alzheimers Dis 2022; 87:1291-1305. [PMID: 35431244 PMCID: PMC9969805 DOI: 10.3233/jad-215533] [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: 11/15/2022]
Abstract
BACKGROUND Peripheral inflammation is associated with increased risk for dementia. Neutrophil to lymphocyte ratio (NLR), red cell distribution width (RDW), and mean platelet volume (MPV), are easily measured circulating blood cell phenotypes reflecting chronic peripheral inflammation, but their association with dementia status is unclear. OBJECTIVE We sought to investigate the cross-sectional association of these inflammatory measures with neuropsychological (NP) test performance, and brain magnetic resonance imaging (MRI) measures in the Framingham Heart Study (FHS) Offspring, Third-generation, and Omni cohorts. METHODS We identified FHS participants who attended an exam that included a complete blood cell count (CBC) and underwent NP testing (n = 3,396) or brain MRI (n = 2,770) within five years of blood draw. We investigated the association between NLR, RDW, and MPV and NP test performance and structural MRI-derived volumetric measurements using linear mixed effect models accounting for family relationships and adjusting for potential confounders. RESULTS Participants were on average 60 years old, 53% female, and about 80% attended some college. Higher NLR was significantly associated with poorer performance on visual memory, and visuospatial abilities, as well as with larger white matter hyperintensity volume. We also observed associations for higher RDW with poorer executive function and smaller total cerebral brain volume. CONCLUSION Chronic peripheral inflammation as measured by NLR and RDW was associated with worse cognitive function, reduced brain volume, and greater microvascular disease in FHS participants. If confirmed in other samples, CBC may provide informative and cost-effective biomarkers of abnormal brain aging in the community.
Collapse
Affiliation(s)
- Yuan Fang
- Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Margaret F. Doyle
- University of Vermont, Larner College of Medicine, Department of Pathology and Laboratory Medicine, Burlington, VT
| | - Michael L. Alosco
- Boston University School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA, USA.,Boston University School of Medicine, Department of Neurology, Boston, MA, USA
| | - Jesse Mez
- Boston University School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA, USA.,Boston University School of Medicine, Department of Neurology, Boston, MA, USA.,Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, MA, USA
| | - Claudia L. Satizabal
- Boston University School of Medicine, Department of Neurology, Boston, MA, USA.,University of Texas Health Science Center at San Antonio, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, USA
| | - Wei Qiao Qiu
- Boston University School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA, USA.,Boston University School of Medicine, Department of Psychiatry, Boston, MA, USA.,Boston University School of Medicine, Department of Pharmacology & Experimental Therapeutics, Boston, MA, USA
| | - Kathryn L. Lunetta
- Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Joanne M. Murabito
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, MA, USA.,Boston University School of Medicine, Department of Medicine, Section of General Internal Medicine, Boston, MA, USA
| |
Collapse
|
29
|
Pettemeridou E, Kallousia E, Constantinidou F. Regional Brain Volume, Brain Reserve and MMSE Performance in Healthy Aging From the NEUROAGE Cohort: Contributions of Sex, Education, and Depression Symptoms. Front Aging Neurosci 2021; 13:711301. [PMID: 34867265 PMCID: PMC8633314 DOI: 10.3389/fnagi.2021.711301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/08/2021] [Indexed: 11/27/2022] Open
Abstract
Objective: The aim of this study was twofold. First, to investigate the relationship between age, gray matter (GM), white matter (WM), and cerebrospinal fluid (CSF) volumes, brain reserve (BR), and specific regions of interest (ROIs) with global cognitive function in healthy older adults participating in a longitudinal study on aging in the island country of Cyprus. Second, to assess the contribution of important demographic and psychosocial factors on brain volume. Specifically, the effects of sex and years of education and the association between depression symptoms on brain volume were also explored in this Mediterranean cohort. Methods: Eighty-seven healthy older adults (males = 37, females = 50) scoring ≥24 on the Mini-Mental State Examination (MMSE) were included, with a mean age of 72.75 years and a mean educational level of 10.48 years. The Geriatric Depression Scale was used to assess depression. T1-weighted magnetic resonance images were used to calculate global and regional volumes. Results: Age was negatively correlated with GM, WM, BR, MMSE scores, and ROIs, including the hippocampus, amygdala, entorhinal cortex, prefrontal cortex, anterior cingulate gyrus, and positively with CSF. Higher MMSE scores positively correlated with GM volume. Women exhibited greater levels of depression than men. Depression was also negatively correlated with GM volume and MMSE scores. Men had greater ventricular size than women and participants with higher education had greater ventricular expansion than those with fewer years in education. Conclusions: The reported structural changes provide evidence on the overlap between age-related brain changes and healthy cognitive aging and suggest that these age changes affect certain regions. Furthermore, sex, depressive symptomatology, and education are significant predictors of the aging brain. Brain reserve and higher education accommodate these changes and works against the development of clinical symptoms.
Collapse
Affiliation(s)
- Eva Pettemeridou
- Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus.,KIOS Innovation and Research Center of Excellence, University of Cyprus, Nicosia, Cyprus.,Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Eleni Kallousia
- Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus
| | - Fofi Constantinidou
- Center for Applied Neuroscience, University of Cyprus, Nicosia, Cyprus.,Department of Psychology, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
30
|
Duarte-Abritta B, Sánchez SM, Abulafia C, Gustafson DR, Vázquez S, Sevlever G, Castro MN, Fiorentini L, Villarreal MF, Guinjoan SM. Amyloid and anatomical correlates of executive functioning in middle-aged offspring of patients with late-onset Alzheimer's disease. Psychiatry Res Neuroimaging 2021; 316:111342. [PMID: 34365076 DOI: 10.1016/j.pscychresns.2021.111342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/02/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
A traditional hallmark of cognitive impairment associated with late-onset Alzheimer´s disease (LOAD) is episodic memory impairment. However, early alterations have been identified in brain regions associated with executive function in asymptomatic, middle-age offspring of patients with LOAD (O-LOAD) compared to those with no family history. We hypothesized that executive function among O-LOAD would correlate with structural and amyloid brain imaging differently from those without a family history of LOAD (control subjects, CS). Executive function, cortical thickness, and in-vivo Aβ deposits were quantified in 30 O-LOAD and 25 CS. Associations were observed among O-LOAD only. Cortical thickness in the left lateral orbitofrontal cortex was positively associated with Design Fluency. The Stroop Color and Word Test, correlated positively with right rostral mid-frontal cortex thickness. Trails Making Test-B was inversely related to left medial orbitofrontal thickness. Tower of London total time was positively associated with β-amyloid deposition in the right precuneus. These results support previous evidence that early executive dysfunction might reflect subtle, early changes in persons at risk of LOAD and suggests that executive function alterations deserve further exploration in the LOAD literature.
Collapse
Affiliation(s)
- Bárbara Duarte-Abritta
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta, Instituto de Neurociencias FLENI-CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Stella-Maris Sánchez
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta, Instituto de Neurociencias FLENI-CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Departamento de Física, Facultad de Cs. Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Carolina Abulafia
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta, Instituto de Neurociencias FLENI-CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Institute for Biomedical Research (BIOMED), Pontifical Catholic University of Argentina, Buenos Aires, Argentina
| | - Deborah R Gustafson
- Department of Neurology, State University of New York University Downstate Health Sciences University, United States
| | - Silvia Vázquez
- Centro de imágenes moleculares (CIM), Fundación FLENI, Argentina
| | - Gustavo Sevlever
- Departamento de Neuropatología y Biología Molecular, Fundación FLENI, Buenos Aires, Argentina
| | - Mariana N Castro
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta, Instituto de Neurociencias FLENI-CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Departamento de Salud Mental, Facultad de Medicina, Universidad de Buenos Aires, Argentina; Servicio de Psiquiatría, Fundación FLENI, Buenos Aires, Argentina
| | - Leticia Fiorentini
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta, Instituto de Neurociencias FLENI-CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Servicio de Psiquiatría, Fundación FLENI, Buenos Aires, Argentina
| | - Mirta F Villarreal
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta, Instituto de Neurociencias FLENI-CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Departamento de Física, Facultad de Cs. Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | - Salvador M Guinjoan
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta, Instituto de Neurociencias FLENI-CONICET, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Argentina; Departamento de Salud Mental, Facultad de Medicina, Universidad de Buenos Aires, Argentina; Neurofisiología I, Facultad de Psicología, Universidad de Buenos Aires, Argentina; Laureate Institute for Brain Research, OK, United States.
| |
Collapse
|
31
|
Connor M, Kim MM, Cao Y, Hattangadi-Gluth J. Precision Radiotherapy for Gliomas: Implementing Novel Imaging Biomarkers to Improve Outcomes With Patient-Specific Therapy. Cancer J 2021; 27:353-363. [PMID: 34570449 PMCID: PMC8480523 DOI: 10.1097/ppo.0000000000000546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT Gliomas are the most common primary brain cancer, yet are extraordinarily challenging to treat because they can be aggressive and infiltrative, locally recurrent, and resistant to standard treatments. Furthermore, the treatments themselves, including radiation therapy, can affect patients' neurocognitive function and quality of life. Noninvasive imaging is the standard of care for primary brain tumors, including diagnosis, treatment planning, and monitoring for treatment response. This article explores the ways in which advanced imaging has and will continue to transform radiation treatment for patients with gliomas, with a focus on cognitive preservation and novel biomarkers, as well as precision radiotherapy and treatment adaptation. Advances in novel imaging techniques continue to push the field forward, to more precisely guided treatment planning, radiation dose escalation, measurement of therapeutic response, and understanding of radiation-associated injury.
Collapse
Affiliation(s)
- Michael Connor
- From the Department of Radiation Medicine and Applied Sciences, UC San Diego, Moores Cancer Center, La Jolla, CA
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Jona Hattangadi-Gluth
- From the Department of Radiation Medicine and Applied Sciences, UC San Diego, Moores Cancer Center, La Jolla, CA
| |
Collapse
|
32
|
Schwarz AJ. The Use, Standardization, and Interpretation of Brain Imaging Data in Clinical Trials of Neurodegenerative Disorders. Neurotherapeutics 2021; 18:686-708. [PMID: 33846962 PMCID: PMC8423963 DOI: 10.1007/s13311-021-01027-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Imaging biomarkers play a wide-ranging role in clinical trials for neurological disorders. This includes selecting the appropriate trial participants, establishing target engagement and mechanism-related pharmacodynamic effect, monitoring safety, and providing evidence of disease modification. In the early stages of clinical drug development, evidence of target engagement and/or downstream pharmacodynamic effect-especially with a clear relationship to dose-can provide confidence that the therapeutic candidate should be advanced to larger and more expensive trials, and can inform the selection of the dose(s) to be further tested, i.e., to "de-risk" the drug development program. In these later-phase trials, evidence that the therapeutic candidate is altering disease-related biomarkers can provide important evidence that the clinical benefit of the compound (if observed) is grounded in meaningful biological changes. The interpretation of disease-related imaging markers, and comparability across different trials and imaging tools, is greatly improved when standardized outcome measures are defined. This standardization should not impinge on scientific advances in the imaging tools per se but provides a common language in which the results generated by these tools are expressed. PET markers of pathological protein aggregates and structural imaging of brain atrophy are common disease-related elements across many neurological disorders. However, PET tracers for pathologies beyond amyloid β and tau are needed, and the interpretability of structural imaging can be enhanced by some simple considerations to guard against the possible confound of pseudo-atrophy. Learnings from much-studied conditions such as Alzheimer's disease and multiple sclerosis will be beneficial as the field embraces rarer diseases.
Collapse
Affiliation(s)
- Adam J Schwarz
- Takeda Pharmaceuticals Ltd., 40 Landsdowne Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
33
|
Xia J, Mazaheri A, Segaert K, Salmon DP, Harvey D, Shapiro K, Kutas M, Olichney JM. Event-related potential and EEG oscillatory predictors of verbal memory in mild cognitive impairment. Brain Commun 2020; 2:fcaa213. [PMID: 33364603 PMCID: PMC7749791 DOI: 10.1093/braincomms/fcaa213] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/20/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022] Open
Abstract
Reliable biomarkers of memory decline are critical for the early detection of Alzheimer's disease. Previous work has found three EEG measures, namely the event-related brain potential P600, suppression of oscillatory activity in the alpha frequency range (∼10 Hz) and cross-frequency coupling between low theta/high delta and alpha/beta activity, each of which correlates strongly with verbal learning and memory abilities in healthy elderly and patients with mild cognitive impairment or prodromal Alzheimer's disease. In the present study, we address the question of whether event-related or oscillatory measures, or a combination thereof, best predict the decline of verbal memory in mild cognitive impairment and Alzheimer's disease. Single-trial correlation analyses show that despite a similarity in their time courses and sensitivities to word repetition, the P600 and the alpha suppression components are minimally correlated with each other on a trial-by-trial basis (generally |r s| < 0.10). This suggests that they are unlikely to stem from the same neural mechanism. Furthermore, event-related brain potentials constructed from bandpass filtered (delta, theta, alpha, beta or gamma bands) single-trial data indicate that only delta band activity (1-4 Hz) is strongly correlated (r = 0.94, P < 0.001) with the canonical P600 repetition effect; event-related potentials in higher frequency bands are not. Importantly, stepwise multiple regression analyses reveal that the three event-related brain potential/oscillatory measures are complementary in predicting California Verbal Learning Test scores (overall R 2 ' s in 0.45-0.63 range). The present study highlights the importance of combining EEG event-related potential and oscillatory measures to better characterize the multiple mechanisms of memory failure in individuals with mild cognitive impairment or prodromal Alzheimer's disease.
Collapse
Affiliation(s)
- Jiangyi Xia
- Center for Mind and Brain and Neurology Department, University of California, Davis, CA, USA
| | - Ali Mazaheri
- School of Psychology, University of Birmingham, Birmingham, UK.,Center for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Katrien Segaert
- School of Psychology, University of Birmingham, Birmingham, UK.,Center for Human Brain Health, University of Birmingham, Birmingham, UK
| | - David P Salmon
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Danielle Harvey
- Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Kim Shapiro
- School of Psychology, University of Birmingham, Birmingham, UK.,Center for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Marta Kutas
- Department of Neurosciences, University of California, San Diego, CA, USA.,Department of Cognitive Sciences, University of California, San Diego, CA, USA
| | - John M Olichney
- Center for Mind and Brain and Neurology Department, University of California, Davis, CA, USA
| |
Collapse
|
34
|
Dadar M, Camicioli R, Duchesne S, Collins DL. The temporal relationships between white matter hyperintensities, neurodegeneration, amyloid beta, and cognition. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2020; 12:e12091. [PMID: 33083512 PMCID: PMC7552231 DOI: 10.1002/dad2.12091] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/15/2020] [Accepted: 07/24/2020] [Indexed: 02/03/2023]
Abstract
Introduction Cognitive decline in Alzheimer's disease is associated with amyloid beta (Aβ) accumulation, neurodegeneration, and cerebral small vessel disease, but the temporal relationships among these factors is not well established. Methods Data included white matter hyperintensity (WMH) load, gray matter (GM) atrophy and Alzheimer's Disease Assessment Scale‐Cognitive‐Plus (ADAS13) scores for 720 participants and cerebrospinal fluid amyloid (Aβ1–42) for 461 participants from the Alzheimer's Disease Neuroimaging Initiative. Linear regressions were used to assess the relationships among baseline WMH, GM, and Aβ1–42 to changes in WMH, GM, Aβ1–42, and cognition at 1‐year follow‐up. Results Baseline WMHs and Aβ1–42 predicted WMH increase and GM atrophy. Baseline WMHs and Aβ1–42 predicted worsening cognition. Only baseline Aβ1–42 predicted change in Aβ1–42. Discussion Baseline WMHs lead to greater future GM atrophy and cognitive decline, suggesting that WM damage precedes neurodegeneration and cognitive decline. Baseline Aβ1–42 predicted WMH increase, suggesting a potential role of amyloid in WM damage.
Collapse
Affiliation(s)
- Mahsa Dadar
- CERVO Brain Research Center Centre intégré universitaire santé et services sociaux de la Capitale Nationale Québec Quebec Canada
| | - Richard Camicioli
- Department of Medicine, Division of Neurology University of Alberta Edmonton Alberta Canada
| | - Simon Duchesne
- CERVO Brain Research Center Centre intégré universitaire santé et services sociaux de la Capitale Nationale Québec Quebec Canada.,Department of Radiology and Nuclear Medicine, Faculty of Medicine Université Laval Québec City Quebec Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute McGill University Montreal Quebec Canada.,Department of Neurology and Neurosurgery, Faculty of Medicine McGill University Montreal Quebec Canada.,Department of Biomedical Engineering, Faculty of Medicine McGill University Montreal Quebec Canada
| | | |
Collapse
|
35
|
Zhang T, Shi M. Multi-modal neuroimaging feature fusion for diagnosis of Alzheimer's disease. J Neurosci Methods 2020; 341:108795. [PMID: 32446943 DOI: 10.1016/j.jneumeth.2020.108795] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Compared with single-modal neuroimages classification of AD, multi-modal classification can achieve better performance by fusing different information. Exploring synergy among various multi-modal neuroimages is contributed to identifying the pathological process of neurological disorders. However, it is still problematic to effectively exploit multi-modal information since the lack of an effective fusion method. NEW METHOD In this paper, we propose a deep multi-modal fusion network based on the attention mechanism, which can selectively extract features from MRI and PET branches and suppress irrelevant information. In the attention model, the fusion ratio of each modality is assigned automatically according to the importance of the data. A hierarchical fusion method is adopted to ensure the effectiveness of Multi-modal Fusion. RESULTS Evaluating the model on the ADNI dataset, the experimental results show that it outperforms the state-of-the-art methods. In particular, the final classification results of the NC/AD, SMCI/PMCI and Four-Class are 95.21 %, 89.79 %, and 86.15 %, respectively. COMPARISON WITH EXISTING METHODS Different from the early fusion and the late fusion, the hierarchical fusion method contributes to learning the synergy between the multi-modal data. Compared with some other prominent algorithms, the attention model enables our network to focus on the regions of interest and effectively fuse the multi-modal data. CONCLUSION Benefit from the hierarchical structure with attention model, the proposed network is capable of exploiting low-level and high-level features extracted from the multi-modal data and improving the accuracy of AD diagnosis. Results show its promising performance.
Collapse
Affiliation(s)
- Tao Zhang
- School of Electronic and Information Engineering, Tianjin University, 300387, Tianjin, China
| | - Mingyang Shi
- School of Electronic and Information Engineering, Tianjin University, 300387, Tianjin, China.
| |
Collapse
|
36
|
Edmonds EC, Weigand AJ, Hatton SN, Marshall AJ, Thomas KR, Ayala DA, Bondi MW, McDonald CR. Patterns of longitudinal cortical atrophy over 3 years in empirically derived MCI subtypes. Neurology 2020; 94:e2532-e2544. [PMID: 32393648 DOI: 10.1212/wnl.0000000000009462] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 12/04/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE We previously identified 4 empirically derived mild cognitive impairment (MCI) subtypes via cluster analysis within the Alzheimer's Disease Neuroimaging Initiative (ADNI) and demonstrated high correspondence between patterns of cortical thinning at baseline and each cognitive subtype. We aimed to determine whether our MCI subtypes demonstrate unique longitudinal atrophy patterns. METHODS ADNI participants (295 with MCI and 134 cognitively normal [CN]) underwent annual structural MRI and neuropsychological assessments. General linear modeling compared vertex-wise differences in cortical atrophy rates between each MCI subtype and the CN group. Linear mixed models examined trajectories of cortical atrophy over 3 years within lobar regions of interest. RESULTS Compared to the CN group, those with amnestic MCI (memory deficit) initially demonstrated greater atrophy rates within medial temporal lobe regions that became more widespread over time. Those with dysnomic/amnestic MCI (naming/memory deficits) showed greater atrophy rates largely localized to temporal lobe regions. The mixed MCI (impairment in all cognitive domains) group showed greater atrophy rates in widespread regions at all time points. The cluster-derived normal group, who had intact neuropsychological performance and normal cortical thickness at baseline despite their MCI diagnosis via conventional diagnostic criteria, continued to show normal cognition and minimal cortical atrophy over 3 years. CONCLUSIONS ADNI's purported amnestic MCI sample produced more refined cognitive subtypes with unique longitudinal cortical atrophy rates. These novel MCI subtypes reliably reflect underlying atrophy, reduce false-positive diagnostic errors, and improve prediction of clinical course. Such improvements have implications for the selection of participants for clinical trials and for providing more precise risk assessment for individuals diagnosed with MCI.
Collapse
Affiliation(s)
- Emily C Edmonds
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA.
| | - Alexandra J Weigand
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA
| | - Sean N Hatton
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA
| | - Anisa J Marshall
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA
| | - Kelsey R Thomas
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA
| | - Daniela A Ayala
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA
| | - Mark W Bondi
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA
| | - Carrie R McDonald
- From the Veterans Affairs San Diego Healthcare System (E.C.E., K.R.T., M.W.B.); Department of Psychiatry (E.C.E., S.N.H., K.R.T., M.W.B., C.R.M.), Center for Multimodal Imaging and Genetics (S.N.H., A.M., C.R.M.), Department of Neurosciences (S.N.H.), and Center for Behavior Genetics of Aging (S.N.H.), University of California San Diego, La Jolla; Joint Doctoral Program in Clinical Psychology (A.J.W., J.E.), San Diego State University/University of California San Diego; Department of Psychology (A.J.M.), University of Southern California, Los Angeles; and Department of Biology (D.A.A.), San Diego State University, CA
| | | |
Collapse
|
37
|
Sakurai R, Bartha R, Montero-Odasso M. Entorhinal Cortex Volume Is Associated With Dual-Task Gait Cost Among Older Adults With MCI: Results From the Gait and Brain Study. J Gerontol A Biol Sci Med Sci 2020; 74:698-704. [PMID: 29767690 PMCID: PMC6477635 DOI: 10.1093/gerona/gly084] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 04/16/2018] [Indexed: 11/12/2022] Open
Abstract
Background Low dual-task gait performance (the slowing of gait speed while performing a demanding cognitive task) is associated with low cognitive performance and an increased risk of progression to dementia in older adults with mild cognitive impairment. However, the reason for this remains unclear. This study aimed to examine the relationship between dual-task cost and regional brain volume, focusing on the hippocampus, parahippocampal gyrus, entorhinal cortex, and motor and lateral frontal cortices in older adults with mild cognitive impairment. Methods Forty older adults with mild cognitive impairment from the “Gait and Brain Study” were included in this study. Gait velocity was measured during single-task (ie, walking alone) and dual-task (ie, counting backwards, subtracting serial sevens, and naming animals, in addition to walking) conditions, using an electronic walkway. Regional brain volumes were derived by automated segmentation, using 3T magnetic resonance imaging. Results Partial rank correlation analyses demonstrated that a smaller volume of the left entorhinal cortex was associated with higher dual-task costs in counting backwards and subtracting serial sevens conditions. Subsequent logistic regression analyses demonstrated that a smaller volume of the left entorhinal cortex was independently associated with higher dual-task cost (slowing down >20% when performing cognitive task) in these two conditions. There were no other significant associations. Conclusions Our results show that lower dual-task gait performance is associated with volume reduction in the entorhinal cortex. Cognitive and motor dysfunction in older adults with mild cognitive impairment may reflect a shared pathogenic mechanism, and dual-task-related gait changes might be a surrogate motor marker for Alzheimer’s disease pathology.
Collapse
Affiliation(s)
- Ryota Sakurai
- Gait and Brain Lab, Parkwood Institute, Lawson Health Research Institute, London, Ontario, Canada.,Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.,Tokyo Metropolitan Institute of Gerontology, Japan
| | - Robert Bartha
- Center for Functional and Metabolic Mapping, Robarts Research Institute, London, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| | - Manuel Montero-Odasso
- Gait and Brain Lab, Parkwood Institute, Lawson Health Research Institute, London, Ontario, Canada.,Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| |
Collapse
|
38
|
Jaarsma-Coes MG, Ghaznawi R, Hendrikse J, Slump C, Witkamp TD, van der Graaf Y, Geerlings MI, de Bresser J. MRI phenotypes of the brain are related to future stroke and mortality in patients with manifest arterial disease: The SMART-MR study. J Cereb Blood Flow Metab 2020; 40:354-364. [PMID: 30547694 PMCID: PMC6985990 DOI: 10.1177/0271678x18818918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neurodegenerative and neurovascular diseases lead to heterogeneous brain abnormalities. A combined analysis of these abnormalities by phenotypes of the brain might give a more accurate representation of the underlying aetiology. We aimed to identify different MRI phenotypes of the brain and assessed the risk of future stroke and mortality within these subgroups. In 1003 patients (59 ± 10 years) from the Second Manifestations of ARTerial disease-Magnetic Resonance (SMART-MR) study, different quantitative 1.5T brain MRI markers were used in a hierarchical clustering analysis to identify 11 distinct subgroups with a different distribution in brain MRI markers and cardiovascular risk factors, and a different risk of stroke (Cox regression: from no increased risk compared to the reference group with relatively few brain abnormalities to HR = 10.34; 95% CI 3.80↔28.12 for the multi-burden subgroup) and mortality (from no increased risk compared to the reference group to HR = 4.00; 95% CI 2.50↔6.40 for the multi-burden subgroup). In conclusion, within a group of patients with manifest arterial disease, we showed that different MRI phenotypes of the brain can be identified and that these were associated with different risks of future stroke and mortality. These MRI phenotypes can possibly classify individual patients and assess their risk of future stroke and mortality.
Collapse
Affiliation(s)
- Myriam G Jaarsma-Coes
- Department of Radiology, University Medical Center Utrecht, and Utrecht University, Utrecht, The Netherlands.,MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rashid Ghaznawi
- Department of Radiology, University Medical Center Utrecht, and Utrecht University, Utrecht, The Netherlands.,Julius Center for Health Sciences and Primary Care, Department of Epidemiology, University Medical Center Utrecht, and Utrecht University, Utrecht, the Netherlands
| | - Jeroen Hendrikse
- Department of Radiology, University Medical Center Utrecht, and Utrecht University, Utrecht, The Netherlands
| | - Cornelis Slump
- MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Theo D Witkamp
- Department of Radiology, University Medical Center Utrecht, and Utrecht University, Utrecht, The Netherlands
| | - Yolanda van der Graaf
- Julius Center for Health Sciences and Primary Care, Department of Epidemiology, University Medical Center Utrecht, and Utrecht University, Utrecht, the Netherlands
| | - Mirjam I Geerlings
- Julius Center for Health Sciences and Primary Care, Department of Epidemiology, University Medical Center Utrecht, and Utrecht University, Utrecht, the Netherlands
| | - Jeroen de Bresser
- Department of Radiology, University Medical Center Utrecht, and Utrecht University, Utrecht, The Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
39
|
Duan Y, Lin Y, Rosen D, Du J, He L, Wang Y. Identifying Morphological Patterns of Hippocampal Atrophy in Patients With Mesial Temporal Lobe Epilepsy and Alzheimer Disease. Front Neurol 2020; 11:21. [PMID: 32038474 PMCID: PMC6989594 DOI: 10.3389/fneur.2020.00021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/08/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose: Mesial temporal lobe epilepsy (MTLE) and Alzheimer's disease (AD) are two distinct neurological disorders associated with hippocampal atrophy. Our goal is to analyze the morphologic patterns of hippocampal atrophy to better understand the underlying pathological and clinical characteristics of the two conditions. Methods: Twenty-five patients with AD and 20 healthy controls with matched age and gender were recruited into the AD group. Twenty-three MTLE patients and 28 healthy controls with matched age and gender were recruited into the MTLE group. All subjects were scanned on 3T-MRI scanner. Automated volumetric analysis was applied to measure and compare the hippocampal volume of the two respective groups. Vertex-based morphologic analysis was applied to characterize the morphologic patterns of hippocampal atrophy within and between groups, and a correlation analysis was performed. Results: Volumetric analysis revealed significantly decreased hippocampal volume in both AD and MTLE patients compared to the controls. In the patients with AD, the mean total hippocampal volume was 32.70% smaller than that of healthy controls, without a significant difference between the left and the right hippocampus (p < 0.05). In patients with MTLE, a significant reduction in unilateral hippocampal volume was observed, with a mean volume reduction of 28.38% as compared with healthy controls (p < 0.05). Vertex-based morphologic analysis revealed a generalized shrinkage of the hippocampi in AD patients, especially in bilateral medial and lateral regions. In MTLE group, atrophy was seen in the ipsilateral head, ipsilateral lateral body and slightly contralateral tail of the hippocampus (FWE-corrected, p < 0.05). Conclusions: MTLE and AD have distinctive morphologic patterns of hippocampal atrophy, which provide new insight into the radiology-pathology correlation in these diseases.
Collapse
Affiliation(s)
- Yiran Duan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yicong Lin
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dennis Rosen
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Jialin Du
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liu He
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuping Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Neuromodulation, Beijing, China.,Center of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Smirnov DS, Stasenko A, Salmon DP, Galasko D, Brewer JB, Gollan TH. Distinct structural correlates of the dominant and nondominant languages in bilinguals with Alzheimer's disease (AD). Neuropsychologia 2019; 132:107131. [PMID: 31271821 PMCID: PMC6702045 DOI: 10.1016/j.neuropsychologia.2019.107131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 05/24/2019] [Accepted: 06/28/2019] [Indexed: 11/26/2022]
Abstract
Structural adaptations in brain regions involved in domain-general cognitive control are associated with life-long bilingualism and may contribute to the executive function advantage of bilinguals over monolinguals. To the degree that these adaptations support bilingualism, their disruption by Alzheimer's disease (AD) may compromise the ability to maintain proficiency in two languages, particularly in the less proficient, or nondominant, language that has greater control demands. The present study assessed this possibility in Spanish-English bilinguals with AD (n = 21) and cognitively normal controls (n = 30) by examining the brain correlates of dominant versus nondominant language performance on the Multilingual Naming Test (MINT), adjusting for age and education. There were no significant structural correlates of naming performance for either language in controls. In patients with AD, dominant language MINT performance was associated with cortical thickness of the entorhinal cortex and middle temporal gyrus, consistent with previous findings of temporal atrophy and related decline of naming abilities in AD. Nondominant language MINT performance, in contrast, was correlated with thickness of the left caudal anterior cingulate cortex (ACC), a central cognitive control region involved in error monitoring and task switching. The relationship between naming in the nondominant language and ACC in patients with AD but not in controls may reflect increased reliance on the ACC for nondominant language use in the face of atrophy of other control network components. The results are consistent with the possibility that the increased burden nondominant language use places on cognitive control systems compromised in AD may account for faster nondominant than dominant language decline in AD.
Collapse
Affiliation(s)
- Denis S Smirnov
- Shiley-Marcos Alzheimer's Disease Research Center, Department of Neurosciences, University of California, San Diego, United States.
| | - Alena Stasenko
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, United States; Department of Psychiatry, University of California, San Diego, United States
| | - David P Salmon
- Shiley-Marcos Alzheimer's Disease Research Center, Department of Neurosciences, University of California, San Diego, United States
| | - Douglas Galasko
- Shiley-Marcos Alzheimer's Disease Research Center, Department of Neurosciences, University of California, San Diego, United States
| | - James B Brewer
- Shiley-Marcos Alzheimer's Disease Research Center, Department of Neurosciences, University of California, San Diego, United States
| | - Tamar H Gollan
- Department of Psychiatry, University of California, San Diego, United States
| |
Collapse
|
41
|
Magnetic resonance imaging measures of brain atrophy from the EXPEDITION3 trial in mild Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:328-337. [PMID: 31388559 PMCID: PMC6675941 DOI: 10.1016/j.trci.2019.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Introduction Solanezumab is a humanized monoclonal antibody that preferentially binds to soluble amyloid β and promotes its clearance from the brain in preclinical studies. The objective of this study was to assess the effect of solanezumab in slowing global and anatomically localized brain atrophy as measured by volumetric magnetic resonance imaging (MRI). Methods In the EXPEDITION3 phase 3 trial, participants with mild Alzheimer's disease were randomized to receive intravenous infusions of either 400 mg of solanezumab or placebo every 4 weeks for 76 weeks. Volumetric MRI scans were acquired at baseline and at 80 weeks from 275 MRI facilities using a standardized imaging protocol. A subset of 1462 patients who completed both MRI and 14-item Alzheimer's Disease Assessment Scale–Cognitive Subscale assessments at both time points were selected for analysis. Longitudinal MRI volume changes were analyzed centrally by tensor-based morphometry with a standard FreeSurfer brain parcellation. Prespecified volumetric measures, including whole brain and ventricles, along with anatomically localized regions in the temporal, parietal, and frontal lobes were evaluated in those participants. Results Group-mean differences in brain atrophy rates were directionally consistent across a number of brain regions but small in magnitude (1.3–6.9% slowing) and not statistically significant when corrected for multiple comparisons. The annualized rates of change of the volumetric measures and the correlation of these changes with cognitive changes in placebo-treated subjects were similar to those reported previously. Discussion In the EXPEDITION3 trial, solanezumab did not significantly slow down rates of global or anatomically localized brain atrophy. Brain volume changes and their relationship to cognition were consistent with previous reports.
Collapse
|
42
|
Li X, Zhang J, Shi C. [Study on the improvement of brain cognitive function status by mind-control game training]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2019; 36:364-370. [PMID: 31232537 PMCID: PMC9929962 DOI: 10.7507/1001-5515.201810030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Indexed: 06/09/2023]
Abstract
This study uses mind-control game training to intervene in patients with mild cognitive impairment to improve their cognitive function. In this study, electroencephalogram (EEG) data of 40 participants were collected before and after two training sessions. The continuous complexity of EEG signals was analyzed to assess the status of cognitive function and explore the effect of mind-control game training on the improvement of cognitive function. The results showed that after two training sessions, the continuous complexity of EEG signal of the subject increased (0.012 44 ± 0.000 29, P < 0.05) and amplitude of curve fluctuation decreased gradually, indicating that with increase of training times, the continuous complexity increased significantly, the cognitive function of brain improved significantly and state was stable. The results of this paper may show that mind-control game training can improve the status of the brain cognitive function, which may provide support and help for the future intervention of cognitive dysfunction.
Collapse
Affiliation(s)
- Xin Li
- Institute of Biomedical Engineering, Yanshan University, Qinhuangdao, Hebei 066004, P.R.China;Measurement Technology and Instrumentation Key Lab of Hebei Province, Qinhuangdao, Hebei 066004,
| | - Jie Zhang
- Institute of Biomedical Engineering, Yanshan University, Qinhuangdao, Hebei 066004, P.R.China;Measurement Technology and Instrumentation Key Lab of Hebei Province, Qinhuangdao, Hebei 066004, P.R.China
| | - Chunyan Shi
- Institute of Biomedical Engineering, Yanshan University, Qinhuangdao, Hebei 066004, P.R.China;Measurement Technology and Instrumentation Key Lab of Hebei Province, Qinhuangdao, Hebei 066004, P.R.China
| |
Collapse
|
43
|
Lundervold AJ, Vik A, Lundervold A. Lateral ventricle volume trajectories predict response inhibition in older age-A longitudinal brain imaging and machine learning approach. PLoS One 2019; 14:e0207967. [PMID: 30939173 PMCID: PMC6445521 DOI: 10.1371/journal.pone.0207967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 03/04/2019] [Indexed: 01/06/2023] Open
Abstract
Objective In a three-wave 6 yrs longitudinal study we investigated if the expansion of lateral ventricle (LV) volumes (regarded as a proxy for brain tissue loss) predicts third wave performance on a test of response inhibition (RI). Participants and methods Trajectories of left and right lateral ventricle volumes across the three waves were quantified using the longitudinal stream in Freesurfer. All participants (N = 74;48 females;mean age 66.0 yrs at the third wave) performed the Color-Word Interference Test (CWIT). Response time on the third condition of CWIT, divided into fast, medium and slow, was used as outcome measure in a machine learning framework. Initially, we performed a linear mixed-effect (LME) analysis to describe subject-specific trajectories of the left and right LV volumes (LVV). These features were input to a multinomial logistic regression classification procedure, predicting individual belongings to one of the three RI classes. To obtain results that might generalize, we evaluated the significance of a k-fold cross-validated f1-score with a permutation test, providing a p-value that approximates the probability that the score would be obtained by chance. We also calculated a corresponding confusion matrix. Results The LME-model showed an annual ∼ 3.0% LVV increase. Evaluation of a cross-validated score using 500 permutations gave an f1-score of 0.462 that was above chance level (p = 0.014). 56% of the fast performers were successfully classified. All these were females, and typically older than 65 yrs at inclusion. For the true slow performers, those being correctly classified had higher LVVs than those being misclassified, and their ages at inclusion were also higher. Conclusion Major contributions were: (i) a longitudinal design, (ii) advanced brain imaging and segmentation procedures with longitudinal data analysis, and (iii) a data driven machine learning approach including cross-validation and permutation testing to predict behaviour, solely from the individual’s brain “signatures” (LVV trajectories).
Collapse
Affiliation(s)
- Astri J. Lundervold
- Department of Biological and Medical Psychology University of Bergen, Norway
| | - Alexandra Vik
- Department of Biological and Medical Psychology University of Bergen, Norway
| | - Arvid Lundervold
- Mohn Medical Imaging and Visualization Centre, Department of Biomedicine, University of Bergen, Norway
- * E-mail:
| |
Collapse
|
44
|
Dicks E, Vermunt L, van der Flier WM, Visser PJ, Barkhof F, Scheltens P, Tijms BM. Modeling grey matter atrophy as a function of time, aging or cognitive decline show different anatomical patterns in Alzheimer's disease. NEUROIMAGE-CLINICAL 2019; 22:101786. [PMID: 30921610 PMCID: PMC6439228 DOI: 10.1016/j.nicl.2019.101786] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/12/2019] [Accepted: 03/16/2019] [Indexed: 01/27/2023]
Abstract
Background Grey matter (GM) atrophy in Alzheimer's disease (AD) is most commonly modeled as a function of time. However, this approach does not take into account inter-individual differences in initial disease severity or changes due to aging. Here, we modeled GM atrophy within individuals across the AD clinical spectrum as a function of time, aging and MMSE, as a proxy for disease severity, and investigated how these models influence estimates of GM atrophy. Methods We selected 523 individuals from ADNI (100 preclinical AD, 288 prodromal AD, 135 AD dementia) with abnormal baseline amyloid PET/CSF and ≥1 year of MRI follow-up. We calculated total and 90 regional GM volumes for 2281 MRI scans (median [IQR]; 4 [3–5] scans per individual over 2 [1.6–4] years) and used linear mixed models to investigate atrophy as a function of time, aging and decline on MMSE. Analyses included clinical stage as interaction with the predictor and were corrected for baseline age, sex, education, field strength and total intracranial volume. We repeated analyses for a sample of participants with normal amyloid (n = 387) to assess whether associations were specific for amyloid pathology. Results Using time or aging as predictors, amyloid abnormal participants annually declined −1.29 ± 0.08 points and − 0.28 ± 0.03 points respectively on the MMSE and −12.23 ± 0.47 cm3 and −8.87 ± 0.34 respectively in total GM volume (p < .001). For the time and age models atrophy was widespread and preclinical and prodromal AD showed similar atrophy patterns. Comparing prodromal AD to AD dementia, AD dementia showed faster atrophy mostly in temporal lobes as modeled with time, while prodromal AD showed faster atrophy in mostly frontoparietal areas as modeled with age (pFDR < 0.05). Modeling change in GM volume as a function of decline on MMSE, slopes were less steep compared to those based on time and aging (−4.1 ± 0.25 cm3 per MMSE point decline; p < .001) and showed steeper atrophy for prodromal AD compared to preclinical AD in the right inferior temporal gyrus (p < .05) and compared to AD dementia mostly in temporal areas (pFDR < 0.05). Associations with time, aging and MMSE remained when accounting for these effects in the other models, suggesting that all measures explain part of the variance in GM atrophy. Repeating analyses in amyloid normal individuals, effects for time and aging showed similar widespread anatomical patterns, while associations with MMSE were largely reduced. Conclusion Effects of time, aging and MMSE all explained variance in GM atrophy slopes within individuals. Associations with MMSE were weaker than those for time or age, but specific for amyloid pathology. This suggests that at least some of the atrophy observed in time or age models may not be specific to AD. Modeling atrophy as a function of time or aging show similar anatomical patterns. GM atrophy as a function of time or aging seems nonspecific for amyloid pathology. GM atrophy as a function of MMSE shows involvement of different anatomical patterns. Atrophy modeled based on time or age was steeper than modeled based on MMSE. Atrophy patterns based on MMSE were specific for amyloid pathology.
Collapse
Affiliation(s)
- Ellen Dicks
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Lisa Vermunt
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands; Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands.
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands; Institutes of Neurology & Healthcare Engineering, UCL London, London, United Kingdom.
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, the Netherlands.
| | | |
Collapse
|
45
|
Alperin N, Wiltshire J, Lee SH, Ramos AR, Hernandez-Cardenache R, Rundek T, Curiel Cid R, Loewenstein D. Effect of sleep quality on amnestic mild cognitive impairment vulnerable brain regions in cognitively normal elderly individuals. Sleep 2019; 42:zsy254. [PMID: 30541112 PMCID: PMC6424074 DOI: 10.1093/sleep/zsy254] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/22/2018] [Accepted: 12/10/2018] [Indexed: 01/01/2023] Open
Abstract
STUDY OBJECTIVES This study aims to evaluate the extent to which sleep quality impacts amnestic mild cognitive impairment (aMCI)-related brain regions in a cognitively normal cohort of individuals. METHODS Seventy-four participants were rigorously evaluated using a battery of cognitive tests and a detailed clinical assessment to verify normal cognitive status. We then screened for sleep quality using the Pittsburgh Sleep Quality Index (PSQI) and depressive symptoms using the Geriatric Depression Scale (GDS). Five subjects were excluded due to mild depression. Overall 38 individuals with mean age 70.7 ± 7 were classified as poor sleepers and 31 with mean age of 69.6 ± 6 years as normal sleepers. Structural MRI and Freesurfer brain parcellation were used to measure aMCI-related brain regions. RESULTS Relative to normal sleepers, poor sleepers exhibited significant reductions in cortical and subcortical volumes bilaterally in the hippocampi, as well as in the superior parietal lobules and left amygdala. The effects were strongest in the left superior parietal lobule (p < .015), followed by the hippocampi. Diffuse patterns of cortical thinning were observed in the frontal lobes, but significant effects were concentrated in the right mesial frontal cortex. Lower sleep duration was most correlated with cortical volume and thickness reductions among all subjects. CONCLUSIONS Atrophy related to poor sleep quality impacted a number of regions implicated in aMCI and Alzheimer's disease (AD). As such, interventions targeted towards improving sleep quality amongst the elderly may prove an effective tool for modulating the course of aMCI and AD.
Collapse
Affiliation(s)
- Noam Alperin
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL
| | - John Wiltshire
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL
| | - Sang H Lee
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL
| | - Alberto R Ramos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL
| | - Rene Hernandez-Cardenache
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL
| | - Tatjana Rundek
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL
| | - Rosie Curiel Cid
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL
| | - David Loewenstein
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
46
|
Examining the relationship between nutrition and cerebral structural integrity in older adults without dementia. Nutr Res Rev 2018; 32:79-98. [PMID: 30378509 DOI: 10.1017/s0954422418000185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The proportion of adults aged 60 years and over is expected to increase over the coming decades. This ageing of the population represents an important health issue, given that marked reductions to cerebral macro- and microstructural integrity are apparent with increasing age. Reduced cerebral structural integrity in older adults appears to predict poorer cognitive performance, even in the absence of clinical disorders such as dementia. As such, it is becoming increasingly important to identify those factors predicting cerebral structural integrity, especially factors that are modifiable. One such factor is nutritional intake. While the literature is limited, data from available cross-sectional studies indicate that increased intake of nutrients such as B vitamins (for example, B6, B12 and folate), choline, n-3 fatty acids and vitamin D, or increased adherence to prudent whole diets (for example, the Mediterranean diet) predicts greater cerebral structural integrity in older adults. There is even greater scarcity of randomised clinical trials investigating the effects of nutritional supplementation on cerebral structure, though it appears that supplementation with B vitamins (B6, B12 and folic acid) or n-3 fatty acids (DHA or EPA) may be beneficial. The current review presents an overview of available research examining the relationship between key nutrients or adherence to select diets and cerebral structural integrity in dementia-free older adults.
Collapse
|
47
|
ten Kate M, Ingala S, Schwarz AJ, Fox NC, Chételat G, van Berckel BNM, Ewers M, Foley C, Gispert JD, Hill D, Irizarry MC, Lammertsma AA, Molinuevo JL, Ritchie C, Scheltens P, Schmidt ME, Visser PJ, Waldman A, Wardlaw J, Haller S, Barkhof F. Secondary prevention of Alzheimer's dementia: neuroimaging contributions. Alzheimers Res Ther 2018; 10:112. [PMID: 30376881 PMCID: PMC6208183 DOI: 10.1186/s13195-018-0438-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/10/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND In Alzheimer's disease (AD), pathological changes may arise up to 20 years before the onset of dementia. This pre-dementia window provides a unique opportunity for secondary prevention. However, exposing non-demented subjects to putative therapies requires reliable biomarkers for subject selection, stratification, and monitoring of treatment. Neuroimaging allows the detection of early pathological changes, and longitudinal imaging can assess the effect of interventions on markers of molecular pathology and rates of neurodegeneration. This is of particular importance in pre-dementia AD trials, where clinical outcomes have a limited ability to detect treatment effects within the typical time frame of a clinical trial. We review available evidence for the use of neuroimaging in clinical trials in pre-dementia AD. We appraise currently available imaging markers for subject selection, stratification, outcome measures, and safety in the context of such populations. MAIN BODY Amyloid positron emission tomography (PET) is a validated in-vivo marker of fibrillar amyloid plaques. It is appropriate for inclusion in trials targeting the amyloid pathway, as well as to monitor treatment target engagement. Amyloid PET, however, has limited ability to stage the disease and does not perform well as a prognostic marker within the time frame of a pre-dementia AD trial. Structural magnetic resonance imaging (MRI), providing markers of neurodegeneration, can improve the identification of subjects at risk of imminent decline and hence play a role in subject inclusion. Atrophy rates (either hippocampal or whole brain), which can be reliably derived from structural MRI, are useful in tracking disease progression and have the potential to serve as outcome measures. MRI can also be used to assess comorbid vascular pathology and define homogeneous groups for inclusion or for subject stratification. Finally, MRI also plays an important role in trial safety monitoring, particularly the identification of amyloid-related imaging abnormalities (ARIA). Tau PET to measure neurofibrillary tangle burden is currently under development. Evidence to support the use of advanced MRI markers such as resting-state functional MRI, arterial spin labelling, and diffusion tensor imaging in pre-dementia AD is preliminary and requires further validation. CONCLUSION We propose a strategy for longitudinal imaging to track early signs of AD including quantitative amyloid PET and yearly multiparametric MRI.
Collapse
Affiliation(s)
- Mara ten Kate
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Alzheimer Center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, PO Box 7056, 1007 MB Amsterdam, the Netherlands
| | - Silvia Ingala
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Adam J. Schwarz
- Takeda Pharmaceuticals Comparny, Cambridge, MA USA
- Eli Lilly and Company, Indianapolis, Indiana USA
| | - Nick C. Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Gaël Chételat
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR-S U1237, Université de Caen-Normandie, GIP Cyceron, Caen, France
| | - Bart N. M. van Berckel
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | | | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | | | | | - Adriaan A. Lammertsma
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Craig Ritchie
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Philip Scheltens
- Alzheimer Center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, PO Box 7056, 1007 MB Amsterdam, the Netherlands
| | | | - Pieter Jelle Visser
- Alzheimer Center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, PO Box 7056, 1007 MB Amsterdam, the Netherlands
| | - Adam Waldman
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joanna Wardlaw
- Centre for Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - Sven Haller
- Affidea Centre de Diagnostic Radiologique de Carouge, Geneva, Switzerland
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
- Insititutes of Neurology and Healthcare Engineering, University College London, London, UK
| |
Collapse
|
48
|
Microstructural brain changes track cognitive decline in mild cognitive impairment. NEUROIMAGE-CLINICAL 2018; 20:883-891. [PMID: 30290303 PMCID: PMC6171091 DOI: 10.1016/j.nicl.2018.09.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/31/2018] [Accepted: 09/25/2018] [Indexed: 11/20/2022]
Abstract
Improved characterization of the microstructural brain changes occurring in the early stages of Alzheimer's disease may permit more timely disease detection. This study examined how longitudinal change in brain microstructure relates to cognitive decline in aging and prodromal Alzheimer's disease. At baseline and two-year follow-up, 29 healthy controls and 21 individuals with mild cognitive impairment or mild Alzheimer's disease underwent neuropsychological evaluation and restriction spectrum imaging (RSI). Microstructural change in the hippocampus, entorhinal cortex, and white matter tracts previously shown to be vulnerable to Alzheimer's disease, was compared between healthy controls and impaired participants. Partial correlations and stepwise linear regressions examined whether baseline RSI metrics predicted subsequent cognitive decline, or change in RSI metrics correlated with cognitive change. In medial temporal gray and white matter, restricted isotropic diffusion and crossing fibers were lower, and free water diffusion was higher, in impaired participants. Restricted isotropic diffusion in the hippocampus declined more rapidly for cognitively impaired participants. Baseline hippocampal restricted isotropic diffusion predicted cognitive decline, and change in hippocampal and entorhinal restricted isotropic diffusion correlated with cognitive decline. Within controls, changes in white matter restricted oriented diffusion and crossing fibers correlated with memory decline. In contrast, there were no correlations between rates of cortical atrophy and cognitive decline in the full sample or within controls. Changes in medial temporal lobe microarchitecture were associated with cognitive decline in prodromal Alzheimer's disease, and these changes were distinct from microstructural changes in normal cognitive aging. RSI metrics of brain microstructure may hold value for predicting cognitive decline in aging and for monitoring the course of Alzheimer's disease. Longitudinal diffusion MRI was conducted in individuals with MCI/AD and controls. Mean restricted diffusion in hippocampus declined more rapidly in MCI/AD. Baseline hippocampal restricted diffusion predicted cognitive decline. Change in medial temporal restricted diffusion correlated with cognitive decline. In healthy controls, white matter microstructure correlated with memory decline.
Collapse
|
49
|
Tuokkola T, Koikkalainen J, Parkkola R, Karrasch M, Lötjönen J, Rinne JO. Longitudinal changes in the brain in mild cognitive impairment: a magnetic resonance imaging study using the visual rating method and tensor-based morphometry. Acta Radiol 2018; 59:973-979. [PMID: 28952780 DOI: 10.1177/0284185117734418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Brain atrophy is associated with mild cognitive impairment (MCI), and by using volumetric and visual analyzing methods, it is possible to differentiate between individuals with progressive MCI (MCIp) and stable MCI (MCIs). Automated analysis methods detect degenerative changes in the brain earlier and more reliably than visual methods. Purpose To detect and evaluate structural brain changes between and within the MCIs, MCIp, and control groups during a two-year follow-up period. Material and Methods Brain magnetic resonance imaging (MRI) scans of 11 participants with MCIs, 18 participants with MCIp, and 84 controls were analyzed by the visual rating method (VRM) and tensor-based morphometry (TBM). Results At baseline, both VRM and TBM differentiated the whole MCI group (combined MCIs and MCIp) and the MCIp group from the control group, but they did not differentiate the MCIs group from the control group. At follow-up, both methods differentiated the MCIp group from the control group, but minor differences between the MCIs and control groups were only seen by TBM. Neuropsychological tests did not find differences between the MCIs and control groups at follow-up. Neither method revealed relevant signs of brain atrophy progression within or between MCI subgroups during the follow-up time. Conclusion Both methods are equally good in the evaluation of structural brain changes in MCI if the groups are sufficiently large and the disease progresses to AD. Only TBM disclosed minor atrophic changes in the MCIs group compared to controls at follow-up. The results need to be confirmed with a large patient group and longer follow-up time.
Collapse
Affiliation(s)
- Terhi Tuokkola
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Juha Koikkalainen
- University of Eastern Finland, Faculty of Health Sciences, Kuopio, Finland
| | - Riitta Parkkola
- Department of Radiology, University Hospital of Turku and Turku University Hospital, Turku, Finland
| | - Mira Karrasch
- Department of Psychology, Abo Akademi University, Turku, Finland
| | - Jyrki Lötjönen
- Aalto University, Department of Neuroscience and Biomedical Engineering, Helsinki, Finland VTT Technical Research Centre of Finland, Tampere, Finland
| | - Juha O Rinne
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Finland Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| |
Collapse
|
50
|
Eliassen CF, Reinvang I, Selnes P, Fladby T, Hessen E. Convergent Results from Neuropsychology and from Neuroimaging in Patients with Mild Cognitive Impairment. Dement Geriatr Cogn Disord 2018; 43:144-154. [PMID: 28152536 DOI: 10.1159/000455832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS To investigate the correspondence between neuropsychological single measures and variation in fludeoxyglucose positron emission tomography (FDG PET) glucose metabolism and magnetic resonance imaging (MRI) cortical thickness in mild cognitive impairment (MCI) patients. METHODS Forty-two elderly controls and 73 MCI subjects underwent FDG PET and MRI scanning. Backward regression analyses with PET and MRI regions were used as dependent variables, while Rey Auditory Verbal Memory Test (RAVLT) recall, Trail Making Test B (TMT B), and a composite test score (RAVLT learning and immediate recall, TMT A, COWAT, and letter-number sequencing) were used as predictor variables. RESULTS The composite score predicted variation in cortical metabolism; supplementary analyses showed that TMT B was significantly correlated with PET metabolism as well. RAVLT and TMT B were significant predictors of variation in MRI cortical thickness. CONCLUSION Our results indicate that RAVLT and TMT B are sensitive to variation in Alzheimer disease neuroimaging markers.
Collapse
|