1
|
Ranasinghe T, Cha SH. Open-source software utilization for zebrafish embryos behavior test. Mol Cells 2025; 48:100221. [PMID: 40288494 DOI: 10.1016/j.mocell.2025.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/13/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
This work described simple methods for measuring locomotive activity using open-source software, ImageJ1.54fFiji, and VirtualDub2. The significance of animal behavior is a mirror of brain activity, which can give information implicated with neurological diseases. Commercial behavioral analysis software frequently needs expertise and expenses high costs due to equip a specific instrument to use of software, thereby encouraging a trend toward open-source alternatives that are both accessible and effective. Here, we explained how to convert video format, measure movement, and produce useful locomotive parameters to aid in the assessment of zebrafish embryos. This method could be easily translated for use in other model systems. This methodology seeks to streamline behavioral quantification in research contexts, encouraging broader research aspects.
Collapse
Affiliation(s)
- Thilini Ranasinghe
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan 31962, Republic of Korea.
| | - Seon-Heui Cha
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan 31962, Republic of Korea; Department of AquaLife Medicine, Hanseo University, Seosan 31962, Republic of Korea; Institute for International Fisheries Science, Hanseo University, Seosan 31962, Republic of Korea.
| |
Collapse
|
2
|
Scheffer JA, Levan DT, Wells JL, Gallagher-Thompson D, Grimm KJ, Chen KH, Brown CK, Bullard BM, Yee CI, Newton SL, Chen EY, Merrilees JJ, Moss D, Wang G, Levenson RW. In-Home Assistive Technology May Help Protect Dementia Caregivers from Declining Sleep Efficiency: A Randomized Control Trial. Clin Gerontol 2025:1-14. [PMID: 40400304 DOI: 10.1080/07317115.2025.2499812] [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] [Indexed: 05/23/2025]
Abstract
OBJECTIVES Caregivers for people with dementia (PWDs) often experience sleep problems due to stressors associated with their role (e.g. concern about PWDs' nighttime wandering). We investigated whether a technology system, People Power Caregiver (PPCg), that helps monitor the caregiver's home would benefit caregivers' sleep. METHODS Primary caregivers of PWDs (Study 1: N = 70, Age M = 64.54, SD = 11.82, range = 35-84; Study 2: N = 92, Age M = 62.73, SD = 11.10, range = 32-89) were assigned to a fully activated PPCg condition or control condition (Study 1: partially active PPCg; Study 2: waitlist control). Caregivers completed the Pittsburgh Sleep Quality Index at baseline, three-months, and six-months. RESULTS Caregivers in the control conditions reported significantly worsening sleep efficiency whereas in comparison, those in the active conditions reported improving sleep efficiency. CONCLUSIONS Given how critical sleep is both for caregivers' health and the care they provide, these findings underscore potential benefits of in-home technologies for protecting caregivers' sleep. CLINICAL IMPLICATIONS Technology-based interventions that help monitor the home may support caregivers' sleep. Protecting caregivers' sleep may also preserve their ability to provide high-quality care as their loved one's disease and associated functional decline progresses.
Collapse
Affiliation(s)
- Julian A Scheffer
- Department of Psychology, University of California, Berkeley, California, USA
- Department of Psychology, University of Western Ontario, London, Ontario, Canada
| | - Darius T Levan
- Department of Psychology, University of California, Berkeley, California, USA
| | - Jenna L Wells
- Department of Psychology, Cornell University, Ithaca, New York, USA
| | - Dolores Gallagher-Thompson
- Department of Psychiatry and Behavioral Sciences and School of Medicine, Stanford University, Stanford, California, USA
| | - Kevin J Grimm
- Department of Psychology, Arizona State University, Tempe, Arizona, USA
| | - Kuan-Hua Chen
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Casey K Brown
- Department of Psychology, Georgetown University, Washington, DC, USA
| | - Breanna M Bullard
- Department of Psychology, University of California, Berkeley, California, USA
| | - Claire I Yee
- Department of Quantitative and Health Sciences, Mayo Clinic, Scottsdale, Arizona, USA
| | - Scott L Newton
- Department of Psychology, University of California, Berkeley, California, USA
| | - Enna Y Chen
- Department of Psychology, Stanford University, Stanford, California, USA
| | - Jennifer J Merrilees
- Department of Neurology, Memory and Aging Center, San Francisco, California, USA
| | - David Moss
- People Power Company dba Care Daily Company
| | - Gene Wang
- People Power Company dba Care Daily Company
| | - Robert W Levenson
- Department of Psychology, University of California, Berkeley, California, USA
| |
Collapse
|
3
|
Li Y, Huang J, Liu R, Zhang Y, Wu S, Liu X, Ma W. Behavioural and psychological symptoms among out-patients with different cognitive states: cross-sectional study. BJPsych Open 2025; 11:e73. [PMID: 40164507 PMCID: PMC12052587 DOI: 10.1192/bjo.2025.7] [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: 09/12/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The study examines the behavioural and psychological symptoms (BPSs) associated with dementia and mild cognitive impairment (MCI), highlighting the prevalence and impact of these symptoms on individuals with varying levels of cognitive function, particularly in the context of the increasing incidence of dementia among the ageing population. AIMS To explore the BPSs among out-patients with different cognitive statuses. METHOD This cross-sectional study enrolled out-patients who attended the cognitive assessment out-patient clinic at our hospital between January 2018 and October 2022. The patients' cognitive status was evaluated using the Neuropsychiatric Inventory (NPI), Activities of Daily Living and the Montreal Cognitive Assessment-Basic scales. RESULTS The study enrolled 3273 out-patients, including 688 (21%) with cognitively unimpairment, 1831 (56%) with MCI and 754 (23%) with dementia. The NPI score, the percentage of patients with BPSs and the number of BPSs increased with decreasing cognition level. Unordered logistic regression analysis showed that after adjustment of confounding variables, delusions, depression, euphoria and psychomotor alterations were independently associated with MCI. Delusions, agitation, euphoria, apathy, psychomotor alterations and sleep change were independently associated with dementia. CONCLUSIONS NPI scores, the percentage of patients with BPSs and the numbers of BPSs increased with declining cognitive function.
Collapse
Affiliation(s)
- Yuhang Li
- Department of Geriatric Medicine, Tongji Hospital of Tongji University, Shanghai, China
| | - Junling Huang
- Department of Geriatric Medicine, Tongji Hospital of Tongji University, Shanghai, China
| | - Ruiping Liu
- Department of Geriatric Medicine, Tongji Hospital of Tongji University, Shanghai, China
| | - Youyang Zhang
- Department of Geriatric Medicine, Tongji Hospital of Tongji University, Shanghai, China
| | - Shihao Wu
- International Medical Service, Tongji Hospital of Tongji University, Shanghai, China
| | - Xiaoli Liu
- Department of Geriatric Medicine, Tongji Hospital of Tongji University, Shanghai, China
| | - Wenlin Ma
- Department of Geriatric Medicine, Tongji Hospital of Tongji University, Shanghai, China
- Shanghai Clinical Research Center for Aging and Medicine, Shanghai, China
| |
Collapse
|
4
|
Leffa D, Povala G, Ferreira P, Ferrari-Souza JP, Bauer-Negrini G, Rodrigues M, Amaral L, Lussier F, Medeiros M, Soares C, Aguzzoli CS, Macedo A, Therriault J, Rosa-Neto P, Tudorascu D, Zimmer E, Bellaver B, Pascoal T. In vivo-measured Lewy body pathology is associated with neuropsychiatric symptoms across the Alzheimer's disease continuum. RESEARCH SQUARE 2025:rs.3.rs-6270682. [PMID: 40196010 PMCID: PMC11975041 DOI: 10.21203/rs.3.rs-6270682/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Intracellular alpha-synuclein aggregates, known as Lewy bodies (LB), are commonly observed in Alzheimer's disease (AD) dementia. Post-mortem studies have shown a higher frequency of neuropsychiatric symptoms among individuals with AD and LB co-pathology. However, the effects of in vivo-measured LB pathology on neuropsychiatric symptoms in AD remain underexplored. This study aimed to evaluate cross-sectional and longitudinal effects of in vivo-measured LB pathology on neuropsychiatric symptoms across the AD continuum. We analyzed data from 1,169 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants had in vivo measures of LB pathology (assessed using an alpha-synuclein seed amplification assay), amyloid-beta (Aβ) and phosphorylated tau (p-tau) levels in cerebrospinal fluid (CSF), and neuropsychiatric symptoms evaluated using the Neuropsychiatric Inventory-Questionnaire (NPI-Q). Logistic and Cox proportional hazards regression models were used to assess cross-sectional and longitudinal effects, respectively, adjusting for age, sex, and cognitive status. Participants had a mean baseline age of 73.05 (SD 7.22) years, 47.13% were women, 426 (36.44%) cognitively unimpaired, and 743 (63.56%) cognitively impaired. In cross-sectional analyses, LB pathology was associated with higher rates of anxiety, apathy, motor disturbances, and appetite disturbances. In longitudinal analyses, LB pathology increased the risk of developing psychosis and anxiety. These effects were independent of Aβ and p-tau. Our results suggest that in vivo-measured LB pathology is closely associated with neuropsychiatric symptoms across the AD continuum. These findings underscore the potential of in vivo LB detection as a marker for identifying individuals at increased risk of neuropsychiatric symptoms, both in clinical trials and in clinical practice.
Collapse
|
5
|
Guo R, Li D, Li F, Ji L, Liu H, Qiao H, Lv Z, Tang Y, Wang D. Effects of whole-head 810 nm near-infrared therapy on cognitive and neuropsychiatric symptoms in Alzheimer's disease: A pilot study. J Alzheimers Dis 2025; 104:52-60. [PMID: 39910867 DOI: 10.1177/13872877251313819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
BackgroundAlzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by significant cognitive and behavioral impairments. Near-infrared (NIR) light treatment has shown potential in cognitive improvement in previous studies. However, clinical trials of NIR for AD remain limited.ObjectiveThis study investigated the safety and effects of whole-head 810 nm NIR therapy in AD patients, including long-term efficacy.MethodsAn open-label pilot study on whole-head NIR treatment for AD patients was conducted. Nine AD patients completed 4-month treatment (810 nm, 100 mW/cm², 30 min/session, 6 sessions weekly). Safety and efficacy were evaluated at baseline, months 2 and 4, and 2-month post-treatment.ResultsAfter four months of whole-head NIR treatment, mean changes from baseline on the Mini-Mental State Examination were 3.2 (p = 0.02). Mean changes from baseline on the Alzheimer's Disease Assessment Scale-Cognitive were -5.0 (p = 0.05), mean changes from baseline on the Montreal Cognitive Assessment were 1.9 (p = 0.12). Mean changes from baseline on the Neuropsychiatric Inventory were -4.2 (p = 0.47). These benefits were sustained two months at least. With no device-related adverse effects were reported.ConclusionsWhole-head 810 nm NIR light is safe and offers promising benefits for AD patients. To fully confirm its efficacy, durability, and underlying mechanisms, further large-scale randomized controlled trials are necessary.
Collapse
Affiliation(s)
- Rong Guo
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Deyu Li
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
- State Key Laboratory of Software Development Environment, State Key Laboratory of Virtual Reality Technology and System, Beihang University, Beijing, China
| | - Fang Li
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Linna Ji
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Hongying Liu
- Rehabilitation Hospital Affiliated to National Rehabilitation Assistive Devices Research Center, Beijing, China
| | - Huiting Qiao
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Zeping Lv
- Rehabilitation Hospital Affiliated to National Rehabilitation Assistive Devices Research Center, Beijing, China
- Key Laboratory of Assistive Technology for Rehabilitation of Elderly Dysfunction, Beijing, China
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Daifa Wang
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
- Jiangsu Danyang Huichuang Medical Equipment Co., Ltd, Jiangsu, China
| |
Collapse
|
6
|
Blasutto B, Fattapposta F, Casagrande M. Mild Behavioral Impairment and cognitive functions: A systematic review and meta-analysis. Ageing Res Rev 2025; 105:102668. [PMID: 39875064 DOI: 10.1016/j.arr.2025.102668] [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: 08/21/2024] [Revised: 01/09/2025] [Accepted: 01/19/2025] [Indexed: 01/30/2025]
Abstract
Mild behavioral impairment (MBI) represents a recently introduced diagnostic concept that focuses on behavioral and personality changes occurring in late life and associated with cognitive decline. Nevertheless, the relationship between these dimensions remains unclear. This systematic review and meta-analysis aim to analyze the relationship between MBI and cognitive functioning. The review process was conducted according to the PRISMA-Statement. Restrictions were made, selecting the studies published in peer-review journals, including at least one cognitive measure and presenting the measurement of MBI. Studies that included participants with neurological disorders, dementia, or psychiatric disorders or that only did a neuroimaging or genetic study were excluded. Twenty-two studies were included in the systematic review, while in the meta-analysis seventeen studies featured data to be included in the analyses. The results were classified according to the following cognitive domains: global cognitive functioning, memory, language, attention executive functions, visuospatial skills, and processing speed. In the quantitative analysis, only global cognitive functioning, executive function, attention, and memory were evaluated. The results of both qualitative and quantitative analysis indicate that individuals with MBI exhibited diminished performance on cognitive tasks when compared to those without MBI symptoms. These results are stronger when evaluating the various domains individually (particularly memory and executive functions) than when a global assessment was made. These findings highlight the potential role of MBI symptoms as early indicators of neurodegenerative processes, reinforcing the necessity for comprehensive assessments that encompass both behavioral and cognitive evaluations. The early detection of these symptoms in prodromal phases can be very useful for the development of non-pharmacological interventions and may provide relevant guidelines for clinicians in the management and diagnosis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Barbara Blasutto
- Department of Psychology, University of Rome "Sapienza", Rome 00185, Italy.
| | - Francesco Fattapposta
- Department of Human Neuroscience, "Sapienza" University of Rome, Viale dell'Università 30, Rome 00185, Italy
| | - Maria Casagrande
- Department of Dynamic and Clinical Psychology and Health, University of Rome "Sapienza", Rome 00185, Italy.
| |
Collapse
|
7
|
Morros-Serra M, Melendo-Azuela EM, Garre-Olmo J, Turró-Garriga O, Santaeugènia S. Sex differences in dementia diagnosis: a fourteen-year retrospective analysis of cases using the Registry of Dementia of Girona. J Women Aging 2025; 37:72-85. [PMID: 39225074 DOI: 10.1080/08952841.2024.2397158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/24/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To evaluate the impact of sociodemographic and clinical sex/gender-associated factors on dementia severity at the time of diagnosis. METHODS Retrospective, cross-sectional study using 2007-2020 data from the Registry of Dementia of Girona (ReDeGi), collecting information from new dementia diagnoses in the seven hospitals of the Health Region of Girona (Northeast region of Catalonia, Spain). Sociodemographic and clinical variables were compared by sex and dementia severity at diagnosis. A multivariate analysis stratified by sex evaluated the risk of having a moderate/severe dementia diagnosis. RESULTS Of 9614 new dementia cases, 6040 (62.8%) were women, and 3574 (37.2%) were men. Women and men aged 75-85 years had 27.9% (p = .003) and 43.1% (p < .001) less risk of moderate/severe diagnosis, respectively, than those >85. Being institutionalized increased the risk of moderate/severe diagnosis more in women (159.9%; p < .001) than in men (114.8%; p = .030). Being single and having a higher education increased the risk of moderate/severe diagnosis by 76.2% (p = .039) and 69.8% (p = .021), respectively, only in women. CONCLUSIONS Age, education level, marital status, and place of residence were differentially associated with moderate/severe dementia at the time of diagnosis in women and men, indicating sex/gender differences in dementia severity at diagnosis, with an increased impact on women.
Collapse
Affiliation(s)
- Mònica Morros-Serra
- Equality and Feminisms Department, Generalitat de Catalunya, Barcelona, Spain
| | - Eva M Melendo-Azuela
- Central Catalonia Chronicity Research Group (C3RG), Centre for Health and Social Care Research (CESS), University of Vic/Central University of Catalonia (UVIC-UCC), Vic, Spain
- Intermediate Care Program, Ministry of Health, Generalitat de Catalunya, Barcelona, Spain
| | | | - Oriol Turró-Garriga
- Registry of Dementia of Girona, Health Region of Girona, Girona, Spain
- Research Institute, Fundació Salut Empordà, Figueres, Spain
| | - Sebastià Santaeugènia
- Central Catalonia Chronicity Research Group (C3RG), Centre for Health and Social Care Research (CESS), University of Vic/Central University of Catalonia (UVIC-UCC), Vic, Spain
- Intermediate Care Program, Ministry of Health, Generalitat de Catalunya, Barcelona, Spain
- Parc Sanitari Sant Joan de Deu, Sant Boi de Llobregat, Barcelona, Spain
| |
Collapse
|
8
|
Macedo AC, Therriault J, Tissot C, Aumont É, Servaes S, Rahmouni N, Fernandez-Arias J, Lussier FZ, Wang YT, Ng KP, Vermeiren M, Bezgin G, Socualaya KQ, Stevenson J, Hosseini SA, Chamoun M, Ferrari-Souza JP, Ferreira PCL, Bellaver B, Leffa DT, Vitali P, Zimmer ER, Ismail Z, Pascoal TA, Gauthier S, Rosa-Neto P. Modeling the progression of neuropsychiatric symptoms in Alzheimer's disease with PET-based Braak staging. Neurobiol Aging 2024; 144:127-137. [PMID: 39326302 DOI: 10.1016/j.neurobiolaging.2024.09.009] [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: 04/18/2024] [Revised: 08/15/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024]
Abstract
In Alzheimer's disease (AD), neuropsychiatric symptoms (NPS) correlate with tau deposition in the brain. Here, we investigated the association of PET-based Braak stages with NPS and assessed whether they predict annual changes in NPS. We evaluated 231 individuals in the aging and AD continuum. Participants were assigned a Braak stage at baseline and followed for 1.97 (s.d. 0.62) years. NPS were investigated using the Mild Behavioral Impairment Checklist (MBI-C) and the Neuropsychiatric Inventory Questionnaire severity (NPI-Q-S) and distress (NPI-Q-D) scales. Multiple linear regressions (MLR) assessed the association of Braak stages with baseline NPS and the annual change in NPS scores. At baseline, stages I-II, III-IV, and V-VI were associated with higher MBI-C, NPI-Q-S, and NPI-Q-D scores. Stages V-VI were associated with a significant annual increase in MBI-C scores. These findings suggest that tau accumulation may manifest clinically with an increase in NPS, which seems to be an early event in AD pathophysiology. Moreover, PET-based Braak staging appears to be a good predictor of NPS severity progression.
Collapse
Affiliation(s)
- Arthur C Macedo
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada; Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - Étienne Aumont
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Psychology, University of Québec at Montréal, 100 Rue Sherbrooke O, Montréal, QC H2X 3P2, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Jaime Fernandez-Arias
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, 11 Jln Tan Tock Seng, Singapore 308433, Singapore
| | - Marie Vermeiren
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Kely Quispialaya Socualaya
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Seyyed Ali Hosseini
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Pâmela C L Ferreira
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Douglas Teixeira Leffa
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Paolo Vitali
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | - Eduardo R Zimmer
- Department of Pharmacology, Graduate Program in Biological Sciences: Pharmacology and Therapeutics; and Biochemistry, Universidade Federal do Rio Grande do Sul, 2600 Ramiro Barcelo St, Porto Alegre, RS 90.035-003, Brazil; Brain Institute of Rio Grande do Sul, PUCRS, Av. Ipiranga, 6690, Porto Alegre, RS 90610-000, Brazil
| | - Zahinoor Ismail
- Departments of Psychiatry, Clinical Neurosciences, Community Health Sciences, and Pathology, Hotchkiss Brain Institute and O'Brien Institute of Public Health, University of Calgary, Calgary, AB, Canada; National Institute for Health and Care Research Exeter Biomedical Research Centre, University of Exeter, Exeter, UK
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, 3501 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Mental Health Institute, Montréal, 6875 LaSalle Blvd, Montréal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montréal, QC H3A 1A1, Canada; Montreal Neurological Institute, 3801 University Street, Montréal, QC H3A 2B4, Canada.
| |
Collapse
|
9
|
Huszár Z, Solomon A, Engh MA, Koszovácz V, Terebessy T, Molnár Z, Hegyi P, Horváth A, Mangialasche F, Kivipelto M, Csukly G. Association of modifiable risk factors with progression to dementia in relation to amyloid and tau pathology. Alzheimers Res Ther 2024; 16:238. [PMID: 39462394 PMCID: PMC11515263 DOI: 10.1186/s13195-024-01602-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND Dementia preventive interventions targeting multiple modifiable risk factors are a promising approach. However, the impact of modifiable risk factors in the presence of beta-amyloid or phosphorylated-tau (p-tau) pathology is unclear. METHODS The objective of the study was to examine the role of modifiable risk factors (vascular factors, depression, and smoking) in the progression to mild cognitive impairment (MCI) or dementia among 434 cognitively unimpaired (CU) and 611 individuals with MCI from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Vascular risk factors were summarized with the Cardiovascular Risk Factors, Aging, and Dementia (CAIDE) score, dichotomized into higher versus lower risk. Depression and smoking (yes/no) were categorised according to medical history or current symptoms. Analyses were stratified by beta-amyloid negative (A-) and positive (A +), p-tau negative (T-) and positive (T +), or beta-amyloid and p-tau negative (A-T-) and positive (A + T +) biomarker status. Cox proportional hazard models were adjusted for age, sex, education, baseline MMSE score, baseline hippocampal volume and ApoE4 carrier status. RESULTS Higher CAIDE score was associated with increased risk of progression to all-cause dementia in most MCI subgroups: adjusted hazard ratios (aHR) [95% CI] were 3.1 [1.43; 6.53] in the A- subgroup, 1.7 [1.20-2.27] in T + , 2.6 [1.06-6.59] in A-T-, and 1.6 [1.15-2.22] in the A + T + subgroup. Smoking (yes/no) was associated with increased dementia aHR in the A + MCI subgroup: 1.6 [1.07-2.34]. Depression increased dementia aHR in the T + MCI subgroup: 1.5 [1.06-2.02]. No significant associations were found in the CU biomarker subgroups. CONCLUSION Addressing modifiable risk factors carries an important potential for reducing the risk of dementia even after the onset of Alzheimer's pathology. Knowledge of biomarker status can further optimize prevention strategies.
Collapse
Affiliation(s)
- Zsolt Huszár
- Centre for Translational Medicine, Semmelweis University, Üllői Út 26, Budapest, Hungary
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa U. 6, Budapest, 1083, Hungary
| | - Alina Solomon
- Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Marie Anne Engh
- Centre for Translational Medicine, Semmelweis University, Üllői Út 26, Budapest, Hungary
| | - Vanda Koszovácz
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa U. 6, Budapest, 1083, Hungary
| | - Tamás Terebessy
- Centre for Translational Medicine, Semmelweis University, Üllői Út 26, Budapest, Hungary
| | - Zsolt Molnár
- Centre for Translational Medicine, Semmelweis University, Üllői Út 26, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Üllői 78/A, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 49 Przybyszewskiego St, Poznan, Poland
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Üllői Út 26, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Szigeti U. 12, Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Tömő 25-29, Budapest, Hungary
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation, University of Szeged, 6728, Szeged, Hungary
| | - András Horváth
- Centre for Translational Medicine, Semmelweis University, Üllői Út 26, Budapest, Hungary
- Neurocognitive Research Center, National Institute of Mental Health, Neurology, and Neurosurgery, Budapest, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
- Research Centre for Natural Sciences, Hungarian Research Network, Budapest, Hungary
| | - Francesca Mangialasche
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Gábor Csukly
- Centre for Translational Medicine, Semmelweis University, Üllői Út 26, Budapest, Hungary.
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa U. 6, Budapest, 1083, Hungary.
| |
Collapse
|
10
|
Guha S, Rodriguez-Acosta J, Dinov ID. A Bayesian Multiplex Graph Classifier of Functional Brain Connectivity Across Diverse Tasks of Cognitive Control. Neuroinformatics 2024; 22:457-472. [PMID: 38861097 DOI: 10.1007/s12021-024-09670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2024] [Indexed: 06/12/2024]
Abstract
This article seeks to investigate the impact of aging on functional connectivity across different cognitive control scenarios, particularly emphasizing the identification of brain regions significantly associated with early aging. By conceptualizing functional connectivity within each cognitive control scenario as a graph, with brain regions as nodes, the statistical challenge revolves around devising a regression framework to predict a binary scalar outcome (aging or normal) using multiple graph predictors. Popular regression methods utilizing multiplex graph predictors often face limitations in effectively harnessing information within and across graph layers, leading to potentially less accurate inference and predictive accuracy, especially for smaller sample sizes. To address this challenge, we propose the Bayesian Multiplex Graph Classifier (BMGC). Accounting for multiplex graph topology, our method models edge coefficients at each graph layer using bilinear interactions between the latent effects associated with the two nodes connected by the edge. This approach also employs a variable selection framework on node-specific latent effects from all graph layers to identify influential nodes linked to observed outcomes. Crucially, the proposed framework is computationally efficient and quantifies the uncertainty in node identification, coefficient estimation, and binary outcome prediction. BMGC outperforms alternative methods in terms of the aforementioned metrics in simulation studies. An additional BMGC validation was completed using an fMRI study of brain networks in adults. The proposed BMGC technique identified that sensory motor brain network obeys certain lateral symmetries, whereas the default mode network exhibits significant brain asymmetries associated with early aging.
Collapse
Affiliation(s)
- Sharmistha Guha
- Department of Statistics, Texas A&M University, 3143 TAMU, College Station, 77843, TX, USA.
| | - Jose Rodriguez-Acosta
- Department of Statistics, Texas A&M University, 3143 TAMU, College Station, 77843, TX, USA
| | - Ivo D Dinov
- Statistics Online Computational Resource, University of Michigan, 426 N. Ingalls St., Ann Arbor, 48109, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, 48109, MI, USA
| |
Collapse
|
11
|
Bieger A, Brum WS, Borelli WV, Therriault J, De Bastiani MA, Moreira AG, Benedet AL, Ferrari-Souza JP, Da Costa JC, Souza DO, Castilhos RM, Schumacher Schuh AF, Fagundes Chaves ML, Schöll M, Zetterberg H, Blennow K, Pascoal TA, Gauthier S, Rosa-Neto P, Schilling LP, Zimmer ER. Influence of Different Diagnostic Criteria on Alzheimer Disease Clinical Research. Neurology 2024; 103:e209753. [PMID: 39167736 PMCID: PMC11338500 DOI: 10.1212/wnl.0000000000209753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 06/14/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Updates in Alzheimer disease (AD) diagnostic guidelines by the National Institute on Aging-Alzheimer's Association (NIA-AA) and the International Working Group (IWG) over the past 11 years may affect clinical diagnoses. We assessed how these guidelines affect clinical AD diagnosis in a cohort of cognitively unimpaired (CU) and cognitively impaired (CI) individuals. METHODS We applied clinical and biomarker data in algorithms to classify individuals from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort according to the following diagnostic guidelines for AD: 2011 NIA-AA, 2016 IWG-2, 2018 NIA-AA, and 2021 IWG-3, assigning the following generic diagnostic labels: (1) not AD (nAD), (2) increased risk of developing AD (irAD), and (3) AD. Diagnostic labels were compared according to their frequency, convergence across guidelines, biomarker profiles, and prognostic value. We also evaluated the diagnostic discordance among the criteria. RESULTS A total of 1,195 individuals (mean age 73.2 ± 7.2 years, mean education 16.1 ± 2.7, 44.0% female) presented different repartitions of diagnostic labels according to the 2011 NIA-AA (nAD = 37.8%, irAD = 23.0%, AD = 39.2%), 2016 IWG-2 (nAD = 37.7%, irAD = 28.7%, AD = 33.6%), 2018 NIA-AA (nAD = 40.7%, irAD = 9.3%, AD = 50.0%), and 2021 IWG-3 (nAD = 51.2%, irAD = 8.4%, AD = 48.3%) frameworks. Discordant diagnoses across all guidelines were found in 512 participants (42.8%) (138 [91.4%] occurring in only β-amyloid [CU 65.4%, CI 34.6%] and 191 [78.6%] in only tau-positive [CU 71.7%, CI 28.3%] individuals). Differences in predicting cognitive impairment between nAD and irAD groups were observed with the 2011 NIA-AA (hazard ratio [HR] 2.21, 95% CI 1.34-3.65, p = 0.002), 2016 IWG-2 (HR 2.81, 95% CI 1.59-4.96, p < 0.000), and 2021 IWG-3 (HR 3.61, 95% CI 2.09-6.23, p < 0.000), but not with 2018 NIA-AA (HR 1.69, 95% CI 0.87-3.28, p = 0.115). DISCUSSION Over 42% of the studied population presented discordant diagnoses when using the different examined AD criteria, mostly in individuals with a single positive biomarker. Except for 2018 NIA-AA, all guidelines identified asymptomatic individuals at risk of cognitive impairment. Our findings highlight important differences between the guidelines, emphasizing the necessity for updated criteria with enhanced staging metrics, considering clinical, research, therapeutic, and trial design aspects.
Collapse
Affiliation(s)
- Andrei Bieger
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Wagner S Brum
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Wyllians V Borelli
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Joseph Therriault
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Marco A De Bastiani
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Amanda G Moreira
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Andrea L Benedet
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - João Pedro Ferrari-Souza
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Jaderson C Da Costa
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Diogo O Souza
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Raphael M Castilhos
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Artur Francisco Schumacher Schuh
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Marcia L Fagundes Chaves
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Michael Schöll
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Henrik Zetterberg
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Kaj Blennow
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Tharick A Pascoal
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Serge Gauthier
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Pedro Rosa-Neto
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Lucas P Schilling
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| | - Eduardo R Zimmer
- From the Graduate Program in Biological Sciences: Biochemistry (A.B., W.S.B., M.A.D.B., J.P.F.-S., D.O.S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Wallenberg Centre for Molecular and Translational Medicine (M.S.), University of Gothenburg, Sweden; Pharmacology and Therapeutics Graduate Program (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS); Memory Center (W.V.B.), Moinhos de Vento Hospital; Department of Anatomy (W.V.B.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; The McGill University Research Centre for Studies in Aging (J.T., A.L.B., T.A.P., S.G., P.R.-N., E.R.Z.), McGill University; Douglas Research Institute (J.T., P.R.-N.), Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal, McGill University; Departments of Neurology and Neurosurgery (J.T., S.G., P.R.-N.) and Psychiatry (J.T., S.G., P.R.-N.), McGill University, Montreal, Canada; Graduate Program in Biological Sciences: Pharmacology and Therapeutics (A.G.M., A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Psychiatry and Neurochemistry (W.S.B., A.L.B., M.S., H.Z., K.B.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden; Department of Neurology and Psychiatry (J.P.F.-S., T.A.P.), University of Pittsburgh, PA; Brain Institute of Rio Grande do Sul (J.C.D.C., L.P.S., E.R.Z.), Pontíficia Universidade Católica do Rio Grande do Sul; Department of Biochemistry (D.O.S.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Neurology Service (R.M.C., A.F.S.-S., M.L.F.C.), Hospital de Clínicas de Porto Alegre; Department of Pharmacology (A.F.S.-S., E.R.Z.), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Neurodegenerative Disease (M.S., H.Z.), Queen Square Institute of Neurology, University College London, United Kingdom; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg, Sweden; UK Dementia Research Institute at University College London (H.Z.), United Kingdom; Hong Kong Center for Neurodegenerative Diseases (H.Z.)
| |
Collapse
|
12
|
Wiesman AI, Gallego‐Rudolf J, Villeneuve S, Baillet S, Wilson TW, the PREVENT‐AD Research Group. Neurochemical organization of cortical proteinopathy and neurophysiology along the Alzheimer's disease continuum. Alzheimers Dement 2024; 20:6316-6331. [PMID: 39001629 PMCID: PMC11497661 DOI: 10.1002/alz.14110] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Despite parallel research indicating amyloid-β accumulation, alterations in cortical neurophysiological signaling, and multi-system neurotransmitter disruptions in Alzheimer's disease (AD), the relationships between these phenomena remains unclear. METHODS Using magnetoencephalography, positron emission tomography, and an atlas of 19 neurotransmitters, we studied the alignment between neurophysiological alterations, amyloid-β deposition, and the neurochemical gradients of the cortex. RESULTS In patients with mild cognitive impairment and AD, changes in cortical rhythms were topographically aligned with cholinergic, serotonergic, and dopaminergic systems. These alignments correlated with the severity of clinical impairments. Additionally, cortical amyloid-β plaques were preferentially deposited along neurochemical boundaries, influencing how neurophysiological alterations align with muscarinic acetylcholine receptors. Most of the amyloid-β-neurochemical and alpha-band neuro-physio-chemical alignments replicated in an independent dataset of individuals with asymptomatic amyloid-β accumulation. DISCUSSION Our findings demonstrate that AD pathology aligns topographically with the cortical distribution of chemical neuromodulator systems and scales with clinical severity, with implications for potential pharmacotherapeutic pathways. HIGHLIGHTS Changes in cortical rhythms in Alzheimer's are organized along neurochemical boundaries. The strength of these alignments is related to clinical symptom severity. Deposition of amyloid-β (Aβ) is aligned with similar neurotransmitter systems. Aβ deposition mediates the alignment of beta rhythms with cholinergic systems. Most alignments replicate in participants with pre-clinical Alzheimer's pathology.
Collapse
Affiliation(s)
- Alex I. Wiesman
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Department of Biomedical Physiology & KinesiologySimon Fraser UniversityBurnabyBritish ColumbiaCanada
| | - Jonathan Gallego‐Rudolf
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteMontrealQuebecCanada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteMontrealQuebecCanada
| | - Sylvain Baillet
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Tony W. Wilson
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
- Department of Pharmacology & NeuroscienceCreighton UniversityOmahaNebraskaUSA
| | | |
Collapse
|
13
|
Sun Y, Shi Y, Li C, Shi H. Histidine Protonation Behaviors on Structural Properties and Aggregation Properties of Aβ(1-42) Mature Fibril: Approaching by Edge Effects. J Phys Chem B 2024; 128:7341-7349. [PMID: 39018428 DOI: 10.1021/acs.jpcb.4c02343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
The histidine behavior plays a crucial role in the structural and aggregation properties of protein folding and misfolding. Understanding the histidine behavior at the edge of the protein structure is critical for finding ways to disrupt fibril elongation and growth, but this impact remains poorly understood. In the current study, we used molecular dynamics simulations to investigate the edge substitution effect of histidine protonation on the structural and aggregation properties. Our data showed that ΔG1 contributed the most to binding affinity compared to ΔG2 and ΔG3. The different protonation states at the edge chain significantly impacted the secondary structure properties of the edge chain. Specifically, we found that such protonation behavior significantly affected specific regions, particularly the N-terminus (G9-Q15) and C-terminus (K28-A30). Further analysis confirmed that H6, H13, and H14 were directly involved in H-bonding networks with the C1_H14//C2_H13 interchain interactions critical for maintaining the interchain stability. Furthermore, we confirmed that H6, H13, and H14 were directly involved in the loss of the carbon skeleton contact in the N-terminus. Our findings indicate that the edge condition is more susceptible to changes in structural properties than the middle condition. The current study is helpful for understanding the histidine behavior hypothesis in related misfolding diseases.
Collapse
Affiliation(s)
- Yue Sun
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Yaru Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Changgui Li
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| | - Hu Shi
- School of Chemistry and Chemical Engineering, Institute of Molecular Science, Shanxi University, Taiyuan 030000, China
| |
Collapse
|
14
|
Yasuno F, Kimura Y, Ogata A, Ikenuma H, Abe J, Minami H, Nihashi T, Yokoi K, Hattori S, Shimoda N, Watanabe A, Kasuga K, Ikeuchi T, Takeda A, Sakurai T, Ito K, Kato T. Trait-anxiety and glial-related neuroinflammation of the amygdala and its associated regions in Alzheimer's disease: A significant correlation. Brain Behav Immun Health 2024; 38:100795. [PMID: 38799793 PMCID: PMC11126804 DOI: 10.1016/j.bbih.2024.100795] [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: 02/24/2024] [Revised: 04/28/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024] Open
Abstract
Background Positron emission tomography, which assesses the binding of translocator protein radiotracers, 11C-DPA-713, may be a sensitive method for determining glial-mediated neuroinflammation levels. This study investigated the relationship between regional 11C-DPA713 binding potential (BPND) and anxiety in patients with Alzheimer's disease (AD) continuum. Methods Nineteen patients with AD continuum determined to be amyloid-/p-tau 181-positive via cerebrospinal fluid analysis were included in this cross-sectional study (mild cognitive impairment [MCI, n = 5] and AD [n = 14]). Anxiety was evaluated using the State-Trait Anxiety Inventory (STAI). A whole-brain voxel-based analysis was performed to examine the relationship between 11C-DPA-713-BPND values at each voxel and the STAI score. Stepwise multiple regression analysis was performed to determine the predictors of STAI scores using independent variables, including 11C-DPA-713-BPND values within significant clusters. 11C-DPA-713-BPND values were compared between patients with AD continuum with low-to-moderate and high STAI scores. Results Voxel-based analysis revealed a positive correlation between trait anxiety severity and 11C-DPA713-BPND values in the centromedial amygdala and the left inferior occipital area [P < 0.001 (uncorrected) at the voxel-level]. 11C-DPA713-BPND values in these regions were a strong predictor of the STAI trait anxiety score. Specifically, patients with AD continuum and high trait anxiety had increased 11C-DPA713-BPND values in these regions. Conclusions The amygdala-occipital lobe circuit influences the control of emotional generation, and disruption of this network by AD pathology-induced inflammation may contribute to the expression of anxiety. Our findings suggest that suppression of inflammation can help effectively treat anxiety by attenuating damage to the amygdala and its associated areas.
Collapse
Affiliation(s)
- Fumihiko Yasuno
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yasuyuki Kimura
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Aya Ogata
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Pharmacy, Faculty of Pharmacy, Gifu University of Medical Science, Kani, Japan
| | - Hiroshi Ikenuma
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Junichiro Abe
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hiroyuki Minami
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Nihashi
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kastunori Yokoi
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Saori Hattori
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Nobuyoshi Shimoda
- Functional Genomics Unit, Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Atsushi Watanabe
- Equipment Management Division, Center for Core Facility Administration, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akinori Takeda
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Sakurai
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kengo Ito
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| |
Collapse
|
15
|
Kim D, Yi B, Won Y. Speech Emotion Recognition in People at High Risk of Dementia. Dement Neurocogn Disord 2024; 23:146-160. [PMID: 39113753 PMCID: PMC11300689 DOI: 10.12779/dnd.2024.23.3.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Background and Purpose The emotions of people at various stages of dementia need to be effectively utilized for prevention, early intervention, and care planning. With technology available for understanding and addressing the emotional needs of people, this study aims to develop speech emotion recognition (SER) technology to classify emotions for people at high risk of dementia. Methods Speech samples from people at high risk of dementia were categorized into distinct emotions via human auditory assessment, the outcomes of which were annotated for guided deep-learning method. The architecture incorporated convolutional neural network, long short-term memory, attention layers, and Wav2Vec2, a novel feature extractor to develop automated speech-emotion recognition. Results Twenty-seven kinds of Emotions were found in the speech of the participants. These emotions were grouped into 6 detailed emotions: happiness, interest, sadness, frustration, anger, and neutrality, and further into 3 basic emotions: positive, negative, and neutral. To improve algorithmic performance, multiple learning approaches were applied using different data sources-voice and text-and varying the number of emotions. Ultimately, a 2-stage algorithm-initial text-based classification followed by voice-based analysis-achieved the highest accuracy, reaching 70%. Conclusions The diverse emotions identified in this study were attributed to the characteristics of the participants and the method of data collection. The speech of people at high risk of dementia to companion robots also explains the relatively low performance of the SER algorithm. Accordingly, this study suggests the systematic and comprehensive construction of a dataset from people with dementia.
Collapse
Affiliation(s)
- Dongseon Kim
- Department of Silver Business, Sookmyung Women’s University, Seoul, Korea
| | - Bongwon Yi
- Department of Communication Disorders, Korea Nazarene University, Cheonan, Korea
| | | |
Collapse
|
16
|
Chockanathan U, Padmanabhan K. Differential disruptions in population coding along the dorsal-ventral axis of CA1 in the APP/PS1 mouse model of Aβ pathology. PLoS Comput Biol 2024; 20:e1012085. [PMID: 38709845 PMCID: PMC11098488 DOI: 10.1371/journal.pcbi.1012085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/16/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
Alzheimer's Disease (AD) is characterized by a range of behavioral alterations, including memory loss and psychiatric symptoms. While there is evidence that molecular pathologies, such as amyloid beta (Aβ), contribute to AD, it remains unclear how this histopathology gives rise to such disparate behavioral deficits. One hypothesis is that Aβ exerts differential effects on neuronal circuits across brain regions, depending on the neurophysiology and connectivity of different areas. To test this, we recorded from large neuronal populations in dorsal CA1 (dCA1) and ventral CA1 (vCA1), two hippocampal areas known to be structurally and functionally diverse, in the APP/PS1 mouse model of amyloidosis. Despite similar levels of Aβ pathology, dCA1 and vCA1 showed distinct disruptions in neuronal population activity as animals navigated a virtual reality environment. In dCA1, pairwise correlations and entropy, a measure of the diversity of activity patterns, were decreased in APP/PS1 mice relative to age-matched C57BL/6 controls. However, in vCA1, APP/PS1 mice had increased pair-wise correlations and entropy as compared to age matched controls. Finally, using maximum entropy models, we connected the microscopic features of population activity (correlations) to the macroscopic features of the population code (entropy). We found that the models' performance increased in predicting dCA1 activity, but decreased in predicting vCA1 activity, in APP/PS1 mice relative to the controls. Taken together, we found that Aβ exerts distinct effects across different hippocampal regions, suggesting that the various behavioral deficits of AD may reflect underlying heterogeneities in neuronal circuits and the different disruptions that Aβ pathology causes in those circuits.
Collapse
Affiliation(s)
- Udaysankar Chockanathan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Krishnan Padmanabhan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Center for Visual Sciences, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Intellectual and Developmental Disabilities Research Center, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
17
|
Shaw JS, Richey LN, Gifford MK, Bray MJC, Esagoff AI, Rosenberg PB, Peters ME. Impact of motor dysfunction on neuropsychiatric symptom profile in patients with autopsy-confirmed Alzheimer's disease. Int Rev Psychiatry 2024; 36:208-218. [PMID: 39255020 DOI: 10.1080/09540261.2024.2361764] [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: 04/09/2024] [Accepted: 05/24/2024] [Indexed: 09/11/2024]
Abstract
Motor dysfunction, which includes changes in gait, balance, and/or functional mobility, is a lesser-known feature of Alzheimer's Disease (AD), especially as it relates to the development of neuropsychiatric symptoms (NPS). This study (1) compared rates of NPS between autopsy-confirmed AD patients with and without early-onset motor dysfunction and (2) compared rates of non-AD dementia autopsy pathology (Lewy Body disease, Frontotemporal Lobar degeneration) between these groups. This retrospective longitudinal cohort study utilized National Alzheimer's Coordinating Center (NACC) data. Participants (N = 856) were required to have moderate-to-severe autopsy-confirmed AD, Clinical Dementia Rating-Global scores of ≤1 at their index visit, and NPS and clinician-rated motor data. Early motor dysfunction was associated with significantly higher NPI-Q total scores (T = 4.48, p < .001) and higher odds of delusions (OR [95%CI]: 1.73 [1.02-2.96]), hallucinations (2.45 [1.35-4.56]), depression (1.51 [1.11-2.06]), irritability (1.50 [1.09-2.08]), apathy (1.70 [1.24-2.36]), anxiety (1.38 [1.01-1.90]), nighttime behaviors (1.98 [1.40-2.81]), and appetite/eating problems (1.56 [1.09-2.25]). Early motor dysfunction was also associated with higher Lewy Body disease pathology (1.41 [1.03-1.93]), but not Frontotemporal Lobar degeneration (1.10 [0.71-1.69]), on autopsy. Our results suggest that motor symptoms in early AD are associated with a higher number and severity of NPS, which may be partially explained by comorbid non-AD neuropathology.
Collapse
Affiliation(s)
- Jacob S Shaw
- Department of Psychiatry and Behavioral Sciences, Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa N Richey
- Department of Psychiatry and Behavioral Sciences, Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mia K Gifford
- Department of Psychiatry and Behavioral Sciences, Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael J C Bray
- Department of Psychiatry and Behavioral Sciences, Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aaron I Esagoff
- Department of Psychiatry and Behavioral Sciences, Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences, Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew E Peters
- Department of Psychiatry and Behavioral Sciences, Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Wiesman AI, Gallego-Rudolf J, Villeneuve S, Baillet S, Wilson TW, PREVENT-AD Research Group. Alignments between cortical neurochemical systems, proteinopathy and neurophysiological alterations along the Alzheimer's disease continuum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.13.24305551. [PMID: 38645027 PMCID: PMC11030470 DOI: 10.1101/2024.04.13.24305551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Two neuropathological hallmarks of Alzheimer's disease (AD) are the accumulation of amyloid-β (Aβ) proteins and alterations in cortical neurophysiological signaling. Despite parallel research indicating disruption of multiple neurotransmitter systems in AD, it has been unclear whether these two phenomena are related to the neurochemical organization of the cortex. We leveraged task-free magnetoencephalography and positron emission tomography, with a cortical atlas of 19 neurotransmitters to study the alignment and interactions between alterations of neurophysiological signaling, Aβ deposition, and the neurochemical gradients of the human cortex. In patients with amnestic mild cognitive impairment (N = 18) and probable AD (N = 20), we found that changes in rhythmic, but not arrhythmic, cortical neurophysiological signaling relative to healthy controls (N = 20) are topographically aligned with cholinergic, serotonergic, and dopaminergic neurochemical systems. These neuro-physio-chemical alignments are related to the severity of cognitive and behavioral impairments. We also found that cortical Aβ plaques are preferentially deposited along neurochemical boundaries, and mediate how beta-band rhythmic cortical activity maps align with muscarinic acetylcholine receptors. Finally, we show in an independent dataset that many of these alignments manifest in the asymptomatic stages of cortical Aβ accumulation (N = 33; N = 71 healthy controls), particularly the Aβ-neurochemical alignments (57.1%) and neuro-physio-chemical alignments in the alpha frequency band (62.5%). Overall, the present study demonstrates that the expression of pathology in pre-clinical and clinical AD aligns topographically with the cortical distribution of chemical neuromodulator systems, scaling with clinical severity and with implications for potential pharmacotherapeutic pathways.
Collapse
Affiliation(s)
- Alex I. Wiesman
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Jonathan Gallego-Rudolf
- Montreal Neurological Institute, McGill University, Montreal, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- Montreal Neurological Institute, McGill University, Montreal, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Sylvain Baillet
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Tony W. Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
- Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA
| | | |
Collapse
|
19
|
Zeng Q, Wang Y, Wang S, Luo X, Li K, Xu X, Liu X, Hong L, Li J, Li Z, Zhang X, Zhong S, Liu Z, Huang P, Chen Y, Zhang M. Cerebrospinal fluid amyloid-β and cerebral microbleed are associated with distinct neuropsychiatric sub-syndromes in cognitively impaired patients. Alzheimers Res Ther 2024; 16:69. [PMID: 38570794 PMCID: PMC10988961 DOI: 10.1186/s13195-024-01434-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/23/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Neuropsychiatric symptoms (NPS) are prevalent in cognitively impaired individuals including Alzheimer's disease (AD) dementia and mild cognitive impairment (MCI). Whereas several studies have reported the associations between NPS with AD pathologic biomarkers and cerebral small vessel disease (SVD), but it remains unknown whether AD pathology and SVD contribute to different sub-syndromes independently or aggravate same symptoms synergistically. METHOD We included 445 cognitively impaired individuals (including 316 MCI and 129 AD) with neuropsychiatric, cerebrospinal fluid (CSF) biomarkers (Aβ42, p-tau, and t-tau) and multi-model MRI data. Psychiatric symptoms were accessed by using the Neuropsychiatric Inventory (NPI). Visual assessment of SVD (white matter hyperintensity, microbleed, perivascular space, lacune) on MRI images was performed by experienced radiologist. Linear regression analyses were conducted to test the association between neuropsychiatric symptoms with AD pathology and CSVD burden after adjustment for age, sex, education, apolipoprotein E (APOE) ε4 carrier status, and clinical diagnosis. RESULTS The NPI total scores were related to microbleed (estimate 2.424; 95% CI [0.749, 4.099]; P =0.005). Considering the sub-syndromes, the hyperactivity was associated with microbleed (estimate 0.925; 95% CI [0.115, 1.735]; P =0.025), whereas the affective symptoms were correlated to CSF level of Aβ42 (estimate -0.006; 95% CI [-0.011, -0.002]; P =0.005). Furthermore, we found the apathy sub-syndrome was associated with CSF t-tau/Aβ42 (estimate 0.636; 95% CI [0.078, 1.194]; P =0.041) and microbleed (estimate 0.693; 95% CI [0.046, 1.340]; P =0.036). In addition, we found a significant interactive effect between CSF t-tau/Aβ42 and microbleed (estimate 0.993; 95% CI [0.360, 1.626]; P =0.019) on severity of apathy sub-syndrome. CONCLUSION Our study showed that CSF Aβ42 was associated with affective symptoms, but microbleed was correlated with hyperactivity and apathy, suggesting the effect of AD pathology and SVD on different neuropsychiatric sub-syndromes.
Collapse
Affiliation(s)
- Qingze Zeng
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Yanbo Wang
- Department of Neurology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
- Department of Neurology, Xinhua Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyue Wang
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Xiao Luo
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Kaicheng Li
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Xiaopei Xu
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Xiaocao Liu
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Luwei Hong
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Jixuan Li
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Zheyu Li
- Department of Neurology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Xinyi Zhang
- Department of Neurology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Siyan Zhong
- Department of Neurology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Zhirong Liu
- Department of Neurology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Peiyu Huang
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China
| | - Yanxing Chen
- Department of Neurology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China.
| | - Minming Zhang
- Department of Radiology, Zhejiang University School of Medicine Second Affiliated Hospital, Shangcheng District, No.88 Jiefang Road, Hangzhou, 310009, China.
| |
Collapse
|
20
|
Wu D, Zhang B, Chang Y, Huang S. Apathy Associated with Alzheimer's Disease. Curr Alzheimer Res 2024; 21:527-537. [PMID: 39716787 PMCID: PMC12079319 DOI: 10.2174/0115672050350970241216072400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION/OBJECTIVE Apathy is a multidimensional and complex disease that is the primary neuropsychiatric symptom among those diagnosed with Alzheimer's disease (AD). Yet, apathy in AD is sometimes underestimated. METHODS A systematic literature review was conducted using databases such as PubMed, Scopus, and Web of Science. The search utilized specific keywords related to apathy and Alzheimer's disease (e.g., "apathy," "Alzheimer's disease," "neuropsychiatric symptoms," "front-striatal circuitry"). The studies were selected based on pre-defined criteria, including publication date (within the last 10 years), peer-reviewed status, and relevance to neurobiological, neurochemical, and behavioral aspects of apathy in AD. The articles were screened through title and abstract reviews, followed by full-text evaluations to ensure they met the inclusion criteria, such as relevance to apathy in Alzheimer's patients, study design rigor, and methodological quality. RESULTS Some research on the behavioral and neurobiological characteristics of apathy in AD points to the role of the front-striatal circuitry, particularly the anterior cingulate cortex (ACC). In addition, we reviewed the neurochemical, neuropsychological, and neuropathological characteristics believed to be associated with apathy symptoms. CONCLUSION The findings indicate that understanding the intricate neurobiological underpinnings of apathy in AD is crucial for developing targeted interventions. Our analysis suggests that a multimodal approach, incorporating both pharmacological and personalized non-pharmacological strategies, could enhance therapeutic efficacy and improve patient outcomes. This highlights the need for future research to explore these combined treatment modalities and their potential to alleviate apathy in AD patients.
Collapse
Affiliation(s)
- Dan Wu
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Department of Pathology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Bo Zhang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yajuan Chang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Shuming Huang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
21
|
Li S, Zhang Q, Liu J, Zhang N, Li X, Liu Y, Qiu H, Li J, Cao H. Bibliometric Analysis of Alzheimer's Disease and Depression. Curr Neuropharmacol 2024; 23:98-115. [PMID: 39092642 PMCID: PMC11519817 DOI: 10.2174/1570159x22666240730154834] [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: 10/25/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The link between Alzheimer's disease and depression has been confirmed by clinical and epidemiological research. Therefore, our study examined the literary landscape and prevalent themes in depression-related research works on Alzheimer's disease through bibliometric analysis. METHODS Relevant literature was identified from the Web of Science core collection. Bibliometric parameters were extracted, and the major contributors were defined in terms of countries, institutions, authors, and articles using Microsoft Excel 2019 and VOSviewer. VOSviewer and CiteSpace were employed to visualize the scientific networks and seminal topics. RESULTS The analysis of literature utilised 10,553 articles published from 1991 until 2023. The three countries or regions with the most publications were spread across the United States, China, and England. The University of Toronto and the University of Pittsburgh were the major contributors to the institutions. Lyketsos, Constantine G., Cummings, JL were found to make outstanding contributions. Journal of Alzheimer's Disease was identified as the most productive journal. Furthermore, "Alzheimer's", "depression", "dementia", and "mild cognitive decline" were the main topics of discussion during this period. LIMITATIONS Data were searched from a single database to become compatible with VOSviewer and CiteSpace, leading to a selection bias. Manuscripts in English were considered, leading to a language bias. CONCLUSION Articles on "Alzheimer's" and "depression" displayed an upward trend. The prevalent themes addressed were the mechanisms of depression-associated Alzheimer's disease, the identification of depression and cognitive decline in the early stages of Alzheimer's, alleviating depression and improving life quality in Alzheimer's patients and their caregivers, and diagnosing and treating neuropsychiatric symptoms in Alzheimer. Future research on these hot topics would promote understanding in this field.
Collapse
Affiliation(s)
- Sixin Li
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province), Changsha, Hunan, China
| | - Qian Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Jian Liu
- Center for Medical Research and Innovation, The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, P.R.China
| | - Xinyu Li
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province), Changsha, Hunan, China
| | - Ying Liu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province), Changsha, Hunan, China
| | - Huiwen Qiu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province), Changsha, Hunan, China
| | - Jing Li
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Cao
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Psychiatry, Brain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province), Changsha, Hunan, China
| |
Collapse
|
22
|
Kashibayashi T, Kanemoto H, Takahashi R, Fujito R, Chadani Y, Tagai K, Shinagawa S, Ishii K, Ikeda M, Kazui H. Neural Basis of Agitated Behaviors in Patients with Amnestic Mild Cognitive Impairment and Alzheimer's Disease. J Alzheimers Dis 2024; 100:1399-1406. [PMID: 39093072 DOI: 10.3233/jad-240256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Background Aggression, a common symptom of Alzheimer's disease (AD), can impose a significant burden on caregivers, necessitating early institutionalization. Objective The current study examined the neural basis of aggression and its expression mechanism, to advance the development of effective treatment strategies for aggression in patients with AD. Methods The study sample included 257 patients; 180 were diagnosed with AD and 77 with amnestic mild cognitive impairment (aMCI). Factor analysis of the neuropsychiatric inventory (NPI) aggression scores was performed, and the correlation between each factor and cerebral blood flow (CBF) was examined via diagnosis of AD or aMCI using statistical parametric mapping. Results Refusal of care was correlated with reduced CBF in the right hippocampus of patients with AD while no specific related regions could be identified in patients with aMCI. Violent behavior was associated with decreased CBF in the right temporal pole and medial frontal lobe of patients with AD and aMCI. Conclusions These findings suggest that aggression, measured using NPI includes two distinct symptoms, refusal of care and violent behavior, having different underlying neural bases.
Collapse
Affiliation(s)
- Tetsuo Kashibayashi
- Dementia-related Disease Medical Center, Hyogo Prefectural Rehabilitation Hospital at Nishi-Harima, Tatsuno, Japan
| | - Hideki Kanemoto
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
- Health and Counseling Center, Osaka University, Toyonaka, Japan
| | - Ryuichi Takahashi
- Dementia-related Disease Medical Center, Hyogo Prefectural Rehabilitation Hospital at Nishi-Harima, Tatsuno, Japan
| | - Ryoko Fujito
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Yoshihiro Chadani
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Kenji Tagai
- Department of Psychiatry, School of Medicine, Jikei University, Tokyo, Japan
| | | | - Kazunari Ishii
- Department of Radiology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiroaki Kazui
- Department of Neuropsychiatry, Kochi Medical School, Kochi University, Nankoku, Japan
| |
Collapse
|
23
|
Lanctôt KL, Chen C, Mah E, Kiss A, Li A, Shade D, Scherer RW, Vieira D, Coulibaly H, Rosenberg PB, Lerner AJ, Padala PR, Brawman-Mintzer O, van Dyck CH, Porsteinsson AP, Craft S, Levey A, Burke WJ, Mintzer J, Herrmann N. Cost consequence analysis of Apathy in Dementia Methylphenidate Trial 2 (ADMET 2). Int Psychogeriatr 2023; 35:664-672. [PMID: 37066690 PMCID: PMC10579450 DOI: 10.1017/s1041610223000327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND This paper used data from the Apathy in Dementia Methylphenidate Trial 2 (NCT02346201) to conduct a planned cost consequence analysis to investigate whether treatment of apathy with methylphenidate is economically attractive. METHODS A total of 167 patients with clinically significant apathy randomized to either methylphenidate or placebo were included. The Resource Utilization in Dementia Lite instrument assessed resource utilization for the past 30 days and the EuroQol five dimension five level questionnaire assessed health utility at baseline, 3 months, and 6 months. Resources were converted to costs using standard sources and reported in 2021 USD. A repeated measures analysis of variance compared change in costs and utility over time between the treatment and placebo groups. A binary logistic regression was used to assess cost predictors. RESULTS Costs were not significantly different between groups whether the cost of methylphenidate was excluded (F(2,330) = 0.626, ηp2 = 0.004, p = 0.535) or included (F(2,330) = 0.629, ηp2 = 0.004, p = 0.534). Utility improved with methylphenidate treatment as there was a group by time interaction (F(2,330) = 7.525, ηp2 = 0.044, p < 0.001). DISCUSSION Results from this study indicated that there was no evidence for a difference in resource utilization costs between methylphenidate and placebo treatment. However, utility improved significantly over the 6-month follow-up period. These results can aid in decision-making to improve quality of life in patients with Alzheimer's disease while considering the burden on the healthcare system.
Collapse
Affiliation(s)
| | - Clara Chen
- Faculty of Health Sciences, Western University, London, ON, Canada
| | - Ethan Mah
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Alex Kiss
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Abby Li
- Sunnybrook Research Institute, Toronto, ON, Canada
| | - Dave Shade
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Roberta W. Scherer
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Hamadou Coulibaly
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Alan J. Lerner
- University Hospital – Case Western Reserve University, Cleveland, OH, USA
| | - Prasad R. Padala
- University of Arkansas for Medical Science, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Olga Brawman-Mintzer
- Medical University of South Carolina and Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | | | | | - Suzanne Craft
- Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | | | - Jacobo Mintzer
- Medical University of South Carolina and Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | | |
Collapse
|
24
|
Jazaeri SZ, Taghizadeh G, Babaei JF, Goudarzi S, Saadatmand P, Joghataei MT, Khanahmadi Z. Aquaporin 4 beyond a water channel; participation in motor, sensory, cognitive and psychological performances, a comprehensive review. Physiol Behav 2023; 271:114353. [PMID: 37714320 DOI: 10.1016/j.physbeh.2023.114353] [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: 06/05/2023] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023]
Abstract
Aquaporin 4 (AQP4) is a protein highly expressed in the central nervous system (CNS) and peripheral nervous system (PNS) as well as various other organs, whose different sites of action indicate its importance in various functions. AQP4 has a variety of essential roles beyond water homeostasis. In this article, we have for the first time summarized different roles of AQP4 in motor and sensory functions, besides cognitive and psychological performances, and most importantly, possible physiological mechanisms by which AQP4 can exert its effects. Furthermore, we demonstrated that AQP4 participates in pathology of different neurological disorders, various effects depending on the disease type. Since neurological diseases involve a spectrum of dysfunctions and due to the difficulty of obtaining a treatment that can simultaneously affect these deficits, it is therefore suggested that future studies consider the role of this protein in different functional impairments related to neurological disorders simultaneously or separately by targeting AQP4 expression and/or polarity modulation.
Collapse
Affiliation(s)
- Seyede Zohreh Jazaeri
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ghorban Taghizadeh
- Department of Occupational Therapy, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Javad Fahanik Babaei
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Goudarzi
- Experimental Medicine Research Center, Tehran University of medical Sciences, Tehran, Iran
| | - Pegah Saadatmand
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Innovation in Medical Education, Faculty of Medicine, Ottawa University, Ottawa, Canada.
| | - Zohreh Khanahmadi
- Department of Occupational Therapy, School of Rehabilitation Services, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
25
|
Huang LK, Kuan YC, Lin HW, Hu CJ. Clinical trials of new drugs for Alzheimer disease: a 2020-2023 update. J Biomed Sci 2023; 30:83. [PMID: 37784171 PMCID: PMC10544555 DOI: 10.1186/s12929-023-00976-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/26/2023] [Indexed: 10/04/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, presenting a significant unmet medical need worldwide. The pathogenesis of AD involves various pathophysiological events, including the accumulation of amyloid and tau, neuro-inflammation, and neuronal injury. Clinical trials focusing on new drugs for AD were documented in 2020, but subsequent developments have emerged since then. Notably, the US-FDA has approved Aducanumab and Lecanemab, both antibodies targeting amyloid, marking the end of a nearly two-decade period without new AD drugs. In this comprehensive report, we review all trials listed in clinicaltrials.gov, elucidating their underlying mechanisms and study designs. Ongoing clinical trials are investigating numerous promising new drugs for AD. The main trends in these trials involve pathophysiology-based, disease-modifying therapies and the recruitment of participants in earlier stages of the disease. These trends underscore the significance of conducting fundamental research on pathophysiology, prevention, and intervention prior to the occurrence of brain damage caused by AD.
Collapse
Affiliation(s)
- Li-Kai Huang
- PhD Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, No. 291, Zhong Zheng Road, Zhonghe District, New Taipei City, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan
- Dementia Center and Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Chun Kuan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan
- Dementia Center and Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Ho-Wei Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chaur-Jong Hu
- PhD Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, No. 291, Zhong Zheng Road, Zhonghe District, New Taipei City, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan.
- Dementia Center and Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
26
|
Suliman M, Al-Hawary SIS, Al-Dolaimy F, Hjazi A, Almalki SG, Alkhafaji AT, Alawadi AH, Alsaalamy A, Bijlwan S, Mustafa YF. Inflammatory diseases: Function of LncRNAs in their emergence and the role of mesenchymal stem cell secretome in their treatment. Pathol Res Pract 2023; 249:154758. [PMID: 37660657 DOI: 10.1016/j.prp.2023.154758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023]
Abstract
One of the best treatments for inflammatory diseases such as COVID-19, respiratory diseases and brain diseases is treatment with stem cells. Here we investigate the effect of stem cell therapy in the treatment of brain diseases.Preclinical studies have shown promising results, including improved functional recovery and tissue repair in animal models of neurodegenerative diseases, strokes,and traumatic brain injuries. However,ethical implications, safety concerns, and regulatory frameworks necessitate thorough evaluation before transitioning to clinical applications. Additionally, the complex nature of the brain and its intricate cellular environment present unique obstacles that must be overcome to ensure the successful integration and functionality of genetically engineered MSCs. The careful navigation of this path will determine whether the application of genetically engineered MSCs in brain tissue regeneration ultimately lives up to the hype surrounding it.
Collapse
Affiliation(s)
- Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | | | - Ahmed Hussien Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| | - Sheela Bijlwan
- Uttaranchal School of Computing Sciences, Uttaranchal University, Dehradun, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
27
|
Theleritis C, Siarkos K, Politis A, Smyrnis N, Papageorgiou C, Politis AM. A Systematic Review of Pharmacological Interventions for Apathy in Aging Neurocognitive Disorders. Brain Sci 2023; 13:1061. [PMID: 37508993 PMCID: PMC10377475 DOI: 10.3390/brainsci13071061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVE Apathy, a frequent neuropsychiatric symptom in aging neurocognitive disorders, has been associated with cognitive decline and functional disability. Therefore, timely provision of pharmacological interventions for apathy is greatly needed. DESIGN A systematical literature review of existing studies was conducted up to 30 May 2023 in several databases (PubMed, PsychInfo, Cochrane, Google Scholar, etc.) that included randomized controlled trials (RCTs) and meta-analyses assessing pharmacological treatments for apathy in aging neurocognitive disorders. The quality of the studies was appraised. RESULTS In patients with Alzheimer's Disease (AD), donepezil, galantamine, rivastigmine, methylphenidate, and gingko biloba were proven efficacious for apathy, while rivastigmine, cognitive enhancer IRL752 and piribedil were found to be beneficial in patients with Parkinson's Disease (PD) and agomelatine in patients with Frontotemporal Dementia (FD). The extensive proportion of RCTs in which apathy was used as a secondary outcome measure, along with the considerable methodological heterogeneity, did not allow the evaluation of group effects. CONCLUSIONS Pharmacological interventions for apathy in aging neurocognitive disorders are complex and under-investigated. The continuation of systematic research efforts and the provision of individualized treatment for patients suffering from these disorders is vital.
Collapse
Affiliation(s)
- Christos Theleritis
- First Department of Psychiatry, National and Kapodistrian University of Athens, Eginition Hospital, 74 Vas. Sofias Ave., 11528 Athens, Greece
| | - Kostas Siarkos
- First Department of Psychiatry, National and Kapodistrian University of Athens, Eginition Hospital, 74 Vas. Sofias Ave., 11528 Athens, Greece
| | - Anastasios Politis
- Second Department of Neurosurgery, National and Kapodistrian University of Athens, Attikon Hospital, 1 Rimini Str., 12462 Athens, Greece
| | - Nikolaos Smyrnis
- Second Department of Psychiatry, National and Kapodistrian University of Athens, Attikon Hospital, 1 Rimini Str., 12462 Athens, Greece
| | - Charalabos Papageorgiou
- First Department of Psychiatry, National and Kapodistrian University of Athens, Eginition Hospital, 74 Vas. Sofias Ave., 11528 Athens, Greece
| | - Antonios M Politis
- First Department of Psychiatry, National and Kapodistrian University of Athens, Eginition Hospital, 74 Vas. Sofias Ave., 11528 Athens, Greece
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
28
|
Singh NA, Martin PR, Graff-Radford J, Sintini I, Machulda MM, Duffy JR, Gunter JL, Botha H, Jones DT, Lowe VJ, Jack CR, Josephs KA, Whitwell JL. Altered within- and between-network functional connectivity in atypical Alzheimer's disease. Brain Commun 2023; 5:fcad184. [PMID: 37434879 PMCID: PMC10331277 DOI: 10.1093/braincomms/fcad184] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 05/04/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023] Open
Abstract
Posterior cortical atrophy and logopenic progressive aphasia are atypical clinical presentations of Alzheimer's disease. Resting-state functional connectivity studies have shown functional network disruptions in both phenotypes, particularly involving the language network in logopenic progressive aphasia and the visual network in posterior cortical atrophy. However, little is known about how connectivity differs both within and between brain networks in these atypical Alzheimer's disease phenotypes. A cohort of 144 patients was recruited by the Neurodegenerative Research Group at Mayo Clinic, Rochester, MN, USA, and underwent structural and resting-state functional MRI. Spatially preprocessed data were analysed to explore the default mode network and the salience, sensorimotor, language, visual and memory networks. The data were analysed at the voxel and network levels. Bayesian hierarchical linear models adjusted for age and sex were used to analyse within- and between-network connectivity. Reduced within-network connectivity was observed in the language network in both phenotypes, with stronger evidence of reductions in logopenic progressive aphasia compared to controls. Only posterior cortical atrophy showed reduced within-network connectivity in the visual network compared to controls. Both phenotypes showed reduced within-network connectivity in the default mode and sensorimotor networks. No significant change was noted in the memory network, but a slight increase in the salience within-network connectivity was seen in both phenotypes compared to controls. Between-network analysis in posterior cortical atrophy showed evidence of reduced visual-to-language network connectivity, with reduced visual-to-salience network connectivity, compared to controls. An increase in visual-to-default mode network connectivity was noted in posterior cortical atrophy compared to controls. Between-network analysis in logopenic progressive aphasia showed evidence of reduced language-to-visual network connectivity and an increase in language-to-salience network connectivity compared to controls. Findings from the voxel-level and network-level analysis were in line with the Bayesian hierarchical linear model analysis, showing reduced connectivity in the dominant network based on diagnosis and more crosstalk between networks in general compared to controls. The atypical Alzheimer's disease phenotypes were associated with disruptions in connectivity, both within and between brain networks. Phenotype-specific differences in connectivity patterns were noted in the visual network for posterior cortical atrophy and the language network for logopenic progressive aphasia.
Collapse
Affiliation(s)
| | - Peter R Martin
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Irene Sintini
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Mary M Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Joseph R Duffy
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
29
|
Nissim NR, Pham DVH, Poddar T, Blutt E, Hamilton RH. The impact of gamma transcranial alternating current stimulation (tACS) on cognitive and memory processes in patients with mild cognitive impairment or Alzheimer's disease: A literature review. Brain Stimul 2023; 16:748-755. [PMID: 37028756 PMCID: PMC10862495 DOI: 10.1016/j.brs.2023.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/16/2023] [Accepted: 04/02/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Transcranial alternating current stimulation (tACS)-a noninvasive brain stimulation technique that modulates cortical oscillations through entrainment-has been demonstrated to alter oscillatory activity and enhance cognition in healthy adults. TACS is being explored as a tool to improve cognition and memory in patient populations with mild cognitive impairment (MCI) and Alzheimer's disease (AD). OBJECTIVE To review the growing body of literature and current findings obtained from the application of tACS in patients with MCI or AD, highlighting the effects of gamma tACS on brain function, memory, and cognition. Evidence on the use of brain stimulation in animal models of AD is also discussed. Important parameters of stimulation are underscored for consideration in protocols that aim to apply tACS as a therapeutic tool in patients with MCI/AD. FINDINGS The application of gamma tACS has shown promising results in the improvement of cognitive and memory processes that are impacted in patients with MCI/AD. These data demonstrate the potential for tACS as an interventional stand-alone tool or alongside pharmacological and/or other behavioral interventions in MCI/AD. CONCLUSIONS While the use of tACS in MCI/AD has evidenced encouraging results, the effects of this stimulation technique on brain function and pathophysiology in MCI/AD remains to be fully determined. This review explores the literature and highlights the need for continued research on tACS as a tool to alter the course of the disease by reinstating oscillatory activity, improving cognitive and memory processing, delaying disease progression, and remediating cognitive abilities in patients with MCI/AD.
Collapse
Affiliation(s)
- N R Nissim
- Laboratory for Cognition and Neural Stimulation, Department of Neurology, University of Pennsylvania, Pennsylvania, PA, USA; Moss Rehabilitation Research Institute, Einstein Medical Center, Elkins Park, PA, USA.
| | - D V H Pham
- Laboratory for Cognition and Neural Stimulation, Department of Neurology, University of Pennsylvania, Pennsylvania, PA, USA
| | - T Poddar
- Laboratory for Cognition and Neural Stimulation, Department of Neurology, University of Pennsylvania, Pennsylvania, PA, USA
| | - E Blutt
- Laboratory for Cognition and Neural Stimulation, Department of Neurology, University of Pennsylvania, Pennsylvania, PA, USA
| | - R H Hamilton
- Laboratory for Cognition and Neural Stimulation, Department of Neurology, University of Pennsylvania, Pennsylvania, PA, USA; Moss Rehabilitation Research Institute, Einstein Medical Center, Elkins Park, PA, USA.
| |
Collapse
|
30
|
Pillette L, Moreau G, Normand JM, Perrier M, Lecuyer A, Cogne M. A Systematic Review of Navigation Assistance Systems for People With Dementia. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2023; 29:2146-2165. [PMID: 35007194 DOI: 10.1109/tvcg.2022.3141383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Technological developments provide solutions to alleviate the tremendous impact on the health and autonomy due to the impact of dementia on navigation abilities. We systematically reviewed the literature on devices tested to provide assistance to people with dementia during indoor, outdoor and virtual navigation (PROSPERO ID number: 215585). Medline and Scopus databases were searched from inception. Our aim was to summarize the results from the literature to guide future developments. Twenty-three articles were included in our study. Three types of information were extracted from these studies. First, the types of navigation advice the devices provided were assessed through: (i) the sensorial modality of presentation, e.g., visual and tactile stimuli, (ii) the navigation content, e.g., landmarks, and (iii) the timing of presentation, e.g., systematically at intersections. Second, we analyzed the technology that the devices were based on, e.g., smartphone. Third, the experimental methodology used to assess the devices and the navigation outcome was evaluated. We report and discuss the results from the literature based on these three main characteristics. Finally, based on these considerations, recommendations are drawn, challenges are identified and potential solutions are suggested. Augmented reality-based devices, intelligent tutoring systems and social support should particularly further be explored.
Collapse
|
31
|
Thiel A, Hermanns C, Lauer AA, Reichrath J, Erhardt T, Hartmann T, Grimm MOW, Grimm HS. Vitamin D and Its Analogues: From Differences in Molecular Mechanisms to Potential Benefits of Adapted Use in the Treatment of Alzheimer’s Disease. Nutrients 2023; 15:nu15071684. [PMID: 37049524 PMCID: PMC10096957 DOI: 10.3390/nu15071684] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Lifestyle habits and insufficient sunlight exposure lead to a high prevalence of vitamin D hypovitaminosis, especially in the elderly. Recent studies suggest that in central Europe more than 50% of people over 60 years are not sufficiently supplied with vitamin D. Since vitamin D hypovitaminosis is associated with many diseases, such as Alzheimer’s disease (AD), vitamin D supplementation seems to be particularly useful for this vulnerable age population. Importantly, in addition to vitamin D, several analogues are known and used for different medical purposes. These vitamin D analogues differ not only in their pharmacokinetics and binding affinity to the vitamin D receptor, but also in their potential side effects. Here, we discuss these aspects, especially those of the commonly used vitamin D analogues alfacalcidol, paricalcitol, doxercalciferol, tacalcitol, calcipotriol, and eldecalcitol. In addition to their pleiotropic effects on mechanisms relevant to AD, potential effects of vitamin D analogues on comorbidities common in the context of geriatric diseases are summarized. AD is defined as a complex neurodegenerative disease of the central nervous system and is commonly represented in the elderly population. It is usually caused by extracellular accumulation of amyloidogenic plaques, consisting of amyloid (Aβ) peptides. Furthermore, the formation of intracellular neurofibrillary tangles involving hyperphosphorylated tau proteins contributes to the pathology of AD. In conclusion, this review emphasizes the importance of an adequate vitamin D supply and discusses the specifics of administering various vitamin D analogues compared with vitamin D in geriatric patients, especially those suffering from AD.
Collapse
|
32
|
Mehak SF, Shivakumar AB, Saraf V, Johansson M, Gangadharan G. Apathy in Alzheimer's disease: A neurocircuitry based perspective. Ageing Res Rev 2023; 87:101891. [PMID: 36871779 DOI: 10.1016/j.arr.2023.101891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/25/2023] [Accepted: 02/21/2023] [Indexed: 03/07/2023]
Abstract
In addition to memory deficits and other cognitive disturbances, patients with Alzheimer's disease (AD) experience neuropsychiatric symptoms, notably apathy, which is a state of impaired motivation observed by deficits in goal directed behavior. Apathy is a multifaceted neuropsychiatric condition and appears to be a prognostic indicator, correlating with the progression of AD. Strikingly, recent studies point out that the neurodegenerative pathology of AD may drive apathy independent of cognitive decline. These studies also highlight that neuropsychiatric symptoms, in particular apathy, might manifest early in AD. Here, we review the current understanding of the neurobiological underpinnings of apathy as a neuropsychiatric symptom of AD. Specifically, we highlight the neural circuits and brain regions recognized to be correlated with the apathetic symptomatology. We also discuss the current evidence that supports the notion that apathy and cognitive deficits may develop as independent but concurrent phenomena driven by AD pathology, suggesting its efficacy as an additional outcome measure in Alzheimer's disease clinical trials. The current and prospective therapeutic interventions for apathy in AD from a neurocircuitry based perspective are also reviewed.
Collapse
Affiliation(s)
- Sonam Fathima Mehak
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| | - Apoorva Bettagere Shivakumar
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| | - Vikyath Saraf
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| | - Maurits Johansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SUS, Sweden; Division of Clinical Sciences, Helsingborg, Department of Clinical Sciences Lund, Lund University, Sweden; Department of Psychiatry, Helsingborg Hospital, Sweden.
| | - Gireesh Gangadharan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
33
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Honey and Alzheimer's Disease-Current Understanding and Future Prospects. Antioxidants (Basel) 2023; 12:427. [PMID: 36829985 PMCID: PMC9952506 DOI: 10.3390/antiox12020427] [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: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Alzheimer's disease (AD), a leading cause of dementia, has been a global concern. AD is associated with the involvement of the central nervous system that causes the characteristic impaired memory, cognitive deficits, and behavioral abnormalities. These abnormalities caused by AD is known to be attributed by extracellular aggregates of amyloid beta plaques and intracellular neurofibrillary tangles. Additionally, genetic factors such as abnormality in the expression of APOE, APP, BACE1, PSEN-1, and PSEN-2 play a role in the disease. As the current treatment aims to treat the symptoms and to slow the disease progression, there has been a continuous search for new nutraceutical agent or medicine to help prevent and cure AD pathology. In this quest, honey has emerged as a powerful nootropic agent. Numerous studies have demonstrated that the high flavonoids and phenolic acids content in honey exerts its antioxidant, anti-inflammatory, and neuroprotective properties. This review summarizes the effect of main flavonoid compounds found in honey on the physiological functioning of the central nervous system, and the effect of honey intake on memory and cognition in various animal model. This review provides a new insight on the potential of honey to prevent AD pathology, as well as to ameliorate the damage in the developed AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
34
|
Mielke MM, Aggarwal NT, Vila‐Castelar C, Agarwal P, Arenaza‐Urquijo EM, Brett B, Brugulat‐Serrat A, DuBose LE, Eikelboom WS, Flatt J, Foldi NS, Franzen S, Gilsanz P, Li W, McManus AJ, van Lent DM, Milani SA, Shaaban CE, Stites SD, Sundermann E, Suryadevara V, Trani J, Turner AD, Vonk JMJ, Quiroz YT, Babulal GM, for the Diversity and Disparity Professional Interest Area Sex and Gender Special Interest Group. Consideration of sex and gender in Alzheimer's disease and related disorders from a global perspective. Alzheimers Dement 2022; 18:2707-2724. [PMID: 35394117 PMCID: PMC9547039 DOI: 10.1002/alz.12662] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 01/31/2023]
Abstract
Sex or gender differences in the risk of Alzheimer's disease and related dementias (ADRD) differ by world region, suggesting that there are potentially modifiable risk factors for intervention. However, few epidemiological or clinical ADRD studies examine sex differences; even fewer evaluate gender in the context of ADRD risk. The goals of this perspective are to: (1) provide definitions of gender, biologic sex, and sexual orientation. and the limitations of examining these as binary variables; (2) provide an overview of what is known with regard to sex and gender differences in the risk, prevention, and diagnosis of ADRD; and (3) discuss these sex and gender differences from a global, worldwide perspective. Identifying drivers of sex and gender differences in ADRD throughout the world is a first step in developing interventions unique to each geographical and sociocultural area to reduce these inequities and to ultimately reduce global ADRD risk. HIGHLIGHTS: The burden of dementia is unevenly distributed geographically and by sex and gender. Scientific advances in genetics and biomarkers challenge beliefs that sex is binary. Discrimination against women and sex and gender minority (SGM) populations contributes to cognitive decline. Sociocultural factors lead to gender inequities in Alzheimer's disease and related dementias (ADRD) worldwide.
Collapse
Affiliation(s)
- Michelle M. Mielke
- Division of Epidemiology, Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Neelum T. Aggarwal
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Clara Vila‐Castelar
- Department of Psychiatry, Harvard Medical SchoolMassachusetts General HospitalMassachusettsBostonUSA
| | - Puja Agarwal
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| | - Eider M. Arenaza‐Urquijo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Benjamin Brett
- Department of NeurosurgeryMedical College of WisconsinWisconsinMilwaukeeUSA
| | - Anna Brugulat‐Serrat
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Atlantic Fellow for Equity in Brain HealthThe University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lyndsey E. DuBose
- Department of Medicine, Division of GeriatricsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Willem S. Eikelboom
- Department of NeurologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Jason Flatt
- Social and Behavioral Health Program, School of Public HealthUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Nancy S. Foldi
- Department of Psychology, Queens College and The Graduate CenterCity University of New YorkNew YorkUSA
- Department of PsychiatryNew York University Long Island School of MedicineNew YorkUSA
| | - Sanne Franzen
- Department of NeurologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Paola Gilsanz
- Kaiser Permanente Division of ResearchOaklandCaliforniaUSA
| | - Wei Li
- Department of Clinical and Diagnostic SciencesUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Alison J. McManus
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Debora Melo van Lent
- UT Health San AntonioGlenn Biggs Institute for Alzheimer's and Neurodegenerative diseasesSan AntonioTexasUSA
- Framingham Heart StudyFraminghamMassachusettsUSA
- Department of NeurologyBoston University School of MedicineBostonMassachusettsUSA
| | - Sadaf Arefi Milani
- Division of Geriatrics & Palliative Medicine, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTexasUSA
| | - C. Elizabeth Shaaban
- Department of EpidemiologyGraduate School of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Shana D. Stites
- Department of PsychiatryPerlman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Erin Sundermann
- Department of PsychiatryUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Vidyani Suryadevara
- Department of Orthopedic SurgeryRush University Medical CenterChicagoIllinoisUSA
| | - Jean‐Francoise Trani
- Department of Public HealthWashington University in St. LouisSt. LouisMissouriUSA
| | - Arlener D. Turner
- Department of Psychiatry & Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jet M. J. Vonk
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Julius Center for Health Sciences and Primary CareDepartment of EpidemiologyUniversity Medical Center Utrecht and Utrecht UniversityUtrechtThe Netherlands
| | - Yakeel T. Quiroz
- Department of Psychiatry, Harvard Medical SchoolMassachusetts General HospitalMassachusettsBostonUSA
- Grupo de Neurociencias de Antioquia of Universidad de AntioquiaMedellinColumbiaUSA
| | - Ganesh M. Babulal
- Department of NeurologyWashington University in St. LouisSt. LouisMississippiUSA
- Department of Clinical Research and LeadershipThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
- Department of Psychology, Faculty of HumanitiesUniversity of JohannesburgJohannesburgSouth Africa
| | | |
Collapse
|
35
|
Chockanathan U, Padmanabhan K. From synapses to circuits and back: Bridging levels of understanding in animal models of Alzheimer's disease. Eur J Neurosci 2022; 56:5564-5586. [PMID: 35244297 PMCID: PMC10926359 DOI: 10.1111/ejn.15636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/04/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by behavioural changes that include memory loss and cognitive decline and is associated with the appearance of amyloid-β plaques and neurofibrillary tangles throughout the brain. Although aspects of the disease percolate across multiple levels of neuronal organization, from the cellular to the behavioural, it is increasingly clear that circuits are a critical junction between the cellular pathology and the behavioural phenotypes that bookend these levels of analyses. In this review, we discuss critical aspects of neural circuit research, beginning with synapses and progressing to network activity and how they influence our understanding of disease processed in AD.
Collapse
Affiliation(s)
- Udaysankar Chockanathan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Krishnan Padmanabhan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Center for Visual Science, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Intellectual and Developmental Disabilities Research Center, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
36
|
Baazaoui N, Iqbal K. Alzheimer's Disease: Challenges and a Therapeutic Opportunity to Treat It with a Neurotrophic Compound. Biomolecules 2022; 12:biom12101409. [PMID: 36291618 PMCID: PMC9599095 DOI: 10.3390/biom12101409] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with an insidious onset and multifactorial nature. A deficit in neurogenesis and synaptic plasticity are considered the early pathological features associated with neurofibrillary tau and amyloid β pathologies and neuroinflammation. The imbalance of neurotrophic factors with an increase in FGF-2 level and a decrease in brain derived neurotrophic factor (BDNF) and neurotrophin 4 (NT-4) in the hippocampus, frontal cortex and parietal cortex and disruption of the brain micro-environment are other characteristics of AD. Neurotrophic factors are crucial in neuronal differentiation, maturation, and survival. Several attempts to use neurotrophic factors to treat AD were made, but these trials were halted due to their blood-brain barrier (BBB) impermeability, short-half-life, and severe side effects. In the present review we mainly focus on the major etiopathology features of AD and the use of a small neurotrophic and neurogenic peptide mimetic compound; P021 that was discovered in our laboratory and was found to overcome the difficulties faced in the administration of the whole neurotrophic factor proteins. We describe pre-clinical studies on P021 and its potential as a therapeutic drug for AD and related neurodegenerative disorders. Our study is limited because it focuses only on P021 and the relevant literature; a more thorough investigation is required to review studies on various therapeutic approaches and potential drugs that are emerging in the AD field.
Collapse
Affiliation(s)
- Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
- Correspondence: ; Tel.: +1-718-494-5259; Fax: +1-718-494-1080
| |
Collapse
|
37
|
Maust DT. Ending choose-your-own-adventure care for agitation in dementia. Int Psychogeriatr 2022; 34:867-869. [PMID: 35733284 DOI: 10.1017/s104161022200059x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Donovan T Maust
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
- Center for Clinical Management Research, VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
38
|
Pengo M, Alberici A, Libri I, Benussi A, Gadola Y, Ashton NJ, Zetterberg H, Blennow K, Borroni B. Sex influences clinical phenotype in frontotemporal dementia. Neurol Sci 2022; 43:5281-5287. [PMID: 35672480 PMCID: PMC9385756 DOI: 10.1007/s10072-022-06185-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 05/28/2022] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Frontotemporal dementia (FTD) encompasses a wide spectrum of genetic, clinical, and histological findings. Sex is emerging as a potential biological variable influencing FTD heterogeneity; however, only a few studies explored this issue with nonconclusive results. OBJECTIVE To estimate the role of sex in a single-center large cohort of FTD patients. METHODS Five hundred thirty-one FTD patients were consecutively enrolled. Demographic, clinical, and neuropsychological features, survival rate, and serum neurofilament light (NfL) concentration were determined and compared between sex. RESULTS The behavioral variant of FTD was more common in men, whereas primary progressive aphasia was overrepresented in women (p < 0.001). While global cognitive impairment was comparable, females had a more severe cognitive impairment, namely in Trail Making Test parts A and B (p = 0.003), semantic fluency (p = 0.03), Short Story Recall Test (p = 0.003), and the copy of Rey Complex Figure (p = 0.005). On the other hand, men exhibited more personality/behavioral symptoms (Frontal Behavior Inventory [FBI] AB, p = 0.003), displaying higher scores in positive FBI subscales (FBI B, p < 0.001). In particular, apathy (p = 0.02), irritability (p = 0.006), poor judgment (p = 0.033), aggressivity (p = 0.008), and hypersexuality (p = 0.006) were more common in men, after correction for disease severity. NfL concentration and survival were not statistically different between men and women (p = 0.167 and p = 0.645, respectively). DISCUSSION The present study demonstrated that sex is a potential factor in determining FTD phenotype, while it does not influence survival. Although the pathophysiological contribution of sex in neurodegeneration is not well characterized yet, our findings highlight its role as deserving biological variable in FTD.
Collapse
Affiliation(s)
- Marta Pengo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonella Alberici
- Department of Neurological and Vision Sciences, Neurology Unit, ASST Spedali Civili, Brescia, Italy
| | - Ilenia Libri
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Department of Neurological and Vision Sciences, Neurology Unit, ASST Spedali Civili, Brescia, Italy
| | - Yasmine Gadola
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
| | - Nicholas J Ashton
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mӧlndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Mӧlndal, Sweden
- Institute of Psychiatry, King's College London, Maurice Wohl Clinical Neuroscience Institute, Psychology & Neuroscience, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mӧlndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Diseases, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mӧlndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Barbara Borroni
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy.
- Department of Neurological and Vision Sciences, Neurology Unit, ASST Spedali Civili, Brescia, Italy.
| |
Collapse
|
39
|
Tampi RR, Bhattacharya G, Marpuri P. Managing Behavioral and Psychological Symptoms of Dementia (BPSD) in the Era of Boxed Warnings. Curr Psychiatry Rep 2022; 24:431-440. [PMID: 35781675 DOI: 10.1007/s11920-022-01347-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW To provide a comprehensive overview on the evaluation and management of behavioral and psychological symptoms of dementia (BPSD) using evidence from literature. RECENT FINDINGS Evidence indicates efficacy for some non-pharmacological techniques including education of caregivers and cognitive stimulation therapy and pharmacological agents like antidepressant and antipsychotics for the management of BPSD. The use of antipsychotics has generated controversy due to the recognition of their serious adverse effect profile including the risk of cerebrovascular adverse events and death. BPSD is associated with worsening of cognition and function among individuals with dementia, greater caregiver burden, more frequent institutionalization, overall poorer quality of life, and greater cost of caring for these individuals. Future management strategies for BPSD should include the use of technology for the provision of non-pharmacological interventions and the judicious use of cannabinoids and interventional procedures like ECT for the management of refractory symptoms.
Collapse
Affiliation(s)
- Rajesh R Tampi
- Department of Psychiatry, Creighton University School of Medicine, Omaha, NE, USA. .,Department of Psychiatry &Behavioral Sciences, Cleveland Clinic Akron General, Akron, OH, USA. .,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA. .,Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA. .,Department of Psychiatry, North East Medical University, Rootstown, OH, USA.
| | - Gargi Bhattacharya
- Department of Psychiatry &Behavioral Sciences, Cleveland Clinic Akron General, Akron, OH, USA
| | | |
Collapse
|
40
|
Kshirsagar S, Alvir RV, Pradeepkiran JA, Hindle A, Vijayan M, Ramasubramaniam B, Kumar S, Reddy AP, Reddy PH. A Combination Therapy of Urolithin A+EGCG Has Stronger Protective Effects Than Single Drug Urolithin A in a Humanized Amyloid Beta Knockin Mice for Late-Onset Alzheimer’s Disease. Cells 2022; 11:cells11172660. [PMID: 36078067 PMCID: PMC9454743 DOI: 10.3390/cells11172660] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/03/2022] Open
Abstract
In the current study, for the first time, we study mitophagy enhancer urolithin A and a combination of urolithin A+green tea extract EGCG against human Aβ peptide-induced mitochondrial and synaptic, dendritic, inflammatory toxicities and behavioral changes in humanized homozygous amyloid beta knockin (hAbKI) mice of late-onset Alzheimer’s disease (AD). Our findings reveal significantly increased positive effects of urolithin A and a combination treatment of urolithin A+EGCG in hAbKI mice for phenotypic behavioral changes including motor coordination, locomotion/exploratory activity, spatial learning and working memory. mRNA and protein levels of mitochondrial fusion, synaptic, mitophagy and autophagy genes were upregulated, and mitochondrial fission genes are downregulated in urolithin A and combine treatment in hAbKI mice; however, the effect is stronger in combined treatment. Immunofluorescence analysis of hippocampal brain sections shows similar findings of mRNA and protein levels. Mitochondrial dysfunction is significantly reduced in both treatment groups, but a stronger reduction is observed in combined treatment. Dendritic spines and lengths are significantly increased in both treatment groups, but the effect is stronger in combined treatment. The fragmented number of mitochondria is reduced, and mitochondrial length is increased, and mitophagosomal formations are increased in both the groups, but the effect is stronger in the combined treatment. The levels of amyloid beta (Aβ) 40 and Aβ42 are reduced in both treatments, however, the reduction is higher for combined treatment. These observations suggest that urolithin A is protective against human Aβ peptide-induced toxicities; however, combined treatment of urolithin A+EGCG is effective and stronger, indicating that combined therapy is promising to treat late-onset AD patients.
Collapse
Affiliation(s)
- Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Rainier Vladlen Alvir
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Jangampalli Adi Pradeepkiran
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Ashly Hindle
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Bhagavathi Ramasubramaniam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Subodh Kumar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Arubala P. Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, 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
- 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
- Correspondence: ; Tel.: +1-806-743-3194
| |
Collapse
|
41
|
Pathogenesis and Personalized Interventions for Pharmacological Treatment-Resistant Neuropsychiatric Symptoms in Alzheimer’s Disease. J Pers Med 2022; 12:jpm12091365. [PMID: 36143150 PMCID: PMC9501542 DOI: 10.3390/jpm12091365] [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: 06/27/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, with cognitive impairment as a core symptom. Neuropsychiatric symptoms (NPSs) also occur as non-cognitive symptoms during the disease course, worsening the prognosis. Recent treatment guidelines for NPSs have recommended non-pharmacological treatments as the first line of therapy, followed by pharmacological treatments. However, pharmacological treatment for urgent NPSs can be difficult because of a lack of efficacy or an intolerance, requiring multiple changes in psychotropic prescriptions. One biological factor that might be partly responsible for this difficulty is structural deterioration in elderly people with dementia, which may cause a functional vulnerability affecting the pharmacological response. Other causative factors might include awkward psychosocial interpersonal relations between patients and their caregiver, resulting in distressful vicious circles. Overlapping NPS sub-symptoms can also blur the prioritization of targeted symptoms. Furthermore, consistent neurocognitive reductions cause a primary apathy state and a secondary distorted ideation or perception of present objects, leading to reactions that cannot be treated pharmacologically. The present review defines treatment-resistant NPSs in AD; it may be necessary and helpful for clinicians to discuss the pathogenesis and comprehensive solutions based on three major hypothetical pathophysiological viewpoints: (1) biology, (2) psychosociology, and (3) neurocognition.
Collapse
|
42
|
Toller G, Cobigo Y, Ljubenkov PA, Appleby BS, Dickerson BC, Domoto-Reilly K, Fong JC, Forsberg LK, Gavrilova RH, Ghoshal N, Heuer HW, Knopman DS, Kornak J, Lapid MI, Litvan I, Lucente DE, Mackenzie IR, McGinnis SM, Miller BL, Pedraza O, Rojas JC, Staffaroni AM, Wong B, Wszolek ZK, Boeve BF, Boxer AL, Rosen HJ, Rankin KP. Sensitivity of the Social Behavior Observer Checklist to Early Symptoms of Patients With Frontotemporal Dementia. Neurology 2022; 99:e488-e499. [PMID: 35584922 PMCID: PMC9421596 DOI: 10.1212/wnl.0000000000200582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 03/08/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Changes in social behavior are common symptoms of frontotemporal lobar degeneration (FTLD) and Alzheimer disease syndromes. For early identification of individual patients and differential diagnosis, sensitive clinical measures are required that are able to assess patterns of behaviors and detect syndromic differences in both asymptomatic and symptomatic stages. We investigated whether the examiner-based Social Behavior Observer Checklist (SBOCL) is sensitive to early behavior changes and reflects disease severity within and between neurodegenerative syndromes. METHODS Asymptomatic individuals and patients with neurodegenerative disease were selected from the multisite ALLFTD cohort study. In a sample of participants with at least 1 time point of SBOCL data, we investigated whether the Disorganized, Reactive, and Insensitive subscales of the SBOCL change as a function of disease stage within and between these syndromes. In a longitudinal subsample with both SBOCL and neuroimaging data, we examined whether change over time on each subscale corresponds to progressive gray matter atrophy. RESULTS A total of 1,082 FTLD pathogenic variant carriers and noncarriers were enrolled (282 asymptomatic, 341 behavioral variant frontotemporal dementia, 114 semantic and 95 nonfluent variant primary progressive aphasia, 137 progressive supranuclear palsy, and 113 Alzheimer disease syndrome). The Disorganized score increased between asymptomatic to very mild (p = 0.016, estimate = -1.10, 95% CI = -1.99 to -0.22), very mild to mild (p = 0.013, estimate = -1.17, 95% CI = -2.08 to -0.26), and mild to moderate/severe (p < 0.001, estimate = -2.00, 95% CI = -2.55 to -1.45) disease stages in behavioral variant frontotemporal dementia regardless of pathogenic variant status. Asymptomatic GRN pathogenic gene variant carriers showed more reactive behaviors (preoccupation with time: p = 0.001, estimate = 1.11, 95% CI = 1.06 to 1.16; self-consciousness: p = 0.003, estimate = 1.77, 95% CI = 1.52 to 2.01) than asymptomatic noncarriers (estimate = 1.01, 95% CI = 0.98 to 1.03; estimate = 1.31, 95% CI = 1.20 to 1.41). The Insensitive score increased to a clinically abnormal level in advanced stages of behavioral variant frontotemporal dementia (p = 0.003, estimate = -0.73, 95% CI = -1.18 to -0.29). Higher scores on each subscale corresponded with higher caregiver burden (p < 0.001). Greater change over time corresponded to greater fronto-subcortical atrophy in the semantic-appraisal and fronto-parietal intrinsically connected networks. DISCUSSION The SBOCL is sensitive to early symptoms and reflects disease severity, with some evidence for progression across asymptomatic and symptomatic stages of FTLD syndromes; thus, it may hold promise for early measurement and monitoring of behavioral symptoms in clinical practice and treatment trials. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that the SBOCL is sensitive to early behavioral changes in FTLD pathogenic variants and early symptomatic individuals in a highly educated patient cohort.
Collapse
Affiliation(s)
- Gianina Toller
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL.
| | - Yann Cobigo
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Peter A Ljubenkov
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Brian S Appleby
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Bradford C Dickerson
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Kimiko Domoto-Reilly
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Jamie C Fong
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Leah K Forsberg
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Ralitza H Gavrilova
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Nupur Ghoshal
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Hilary W Heuer
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - David S Knopman
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - John Kornak
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Maria I Lapid
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Irene Litvan
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Diane E Lucente
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Ian R Mackenzie
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Scott M McGinnis
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Bruce L Miller
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Otto Pedraza
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Julio C Rojas
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Adam M Staffaroni
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Bonnie Wong
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Zbigniew K Wszolek
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Brad F Boeve
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Adam L Boxer
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Howard J Rosen
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| | - Katherine P Rankin
- From the Department of Neurology (G.T., Y.C., P.A.L., J.C.F., H.W.H., B.L.M., J.C.R., A.M.S., A.L.B., H.J.R., K.P.R.), Memory and Aging Center, University of California, San Francisco; Department of Neurology (B.S.A.), Case Western Reserve University, Cleveland, OH; Frontotemporal Disorders Unit (B.C.D., D.E.L., S.M.M., B.W.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; Department of Neurology (K.D.-R.), University of Washington, Seattle; Department of Neurology (L.K.F., R.H.G., D.S.K., B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (N.G.), Washington University, St. Louis, MO; Department of Epidemiology and Biostatistics (J.K.), University of California, San Francisco; Department of Psychiatry and Psychology (M.I.L.), Mayo Clinic, Rochester, MN; Department of Neurology (I.L.), Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Pathology and Laboratory Medicine (I.R.M.), University of British Columbia, Vancouver, Canada; Departments of Psychiatry and Psychology (O.P.), and Neurology (Z.K.W.), Mayo Clinic, Jacksonville, FL
| |
Collapse
|
43
|
Poon E. A Systematic Review and Meta-Analysis of Dyadic Psychological Interventions for BPSD, Quality of Life and/or Caregiver Burden in Dementia or MCI. Clin Gerontol 2022; 45:777-797. [PMID: 31752633 DOI: 10.1080/07317115.2019.1694117] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objectives: This systematic review and meta-analysis assesses the effectiveness of psychological interventions that involve people with dementia or mild cognitive impairment (MCI) and their informal caregivers, and target improvements in the management of the behavioral and psychological symptoms of dementia (BPSD); quality of life; and/or burden reduction for people with either dementia or MCI and their informal caregivers.Methods: Studies were identified through database searches (Cochrane Library, CENTRAL, CINAHL, EMBASE, MEDLINE and PsychINFO) and clinical trials registers (ClinicalTrials.gov and http://apps.who.int/trialsearch/). Data were pooled for meta-analysis.Results: Database and reference list searches identified 1,878 references, of which fourteen studies were included. Positive effects were found on the anxiety symptoms of people with dementia on the RAID scale; on the quality of life of people with dementia on the self-rated QoL-AD scale; and on informal caregiver burden on the Zarit Burden Interview.Conclusions: Psychological interventions involving whole dyads have some promise for both people with dementia and informal caregivers, but are still far from uniformly effective across BPSD, quality of life, and caregiver burden. Further research directions are discussed.Clinical Implications: The results suggest that clinicians should routinely involve both halves of the dyad when delivering psychological interventions targeting anxiety or quality of life for people with dementia, or burden for informal caregivers.
Collapse
Affiliation(s)
- Emma Poon
- Clinical Education Development and Research (CEDAR) Group, Psychology: College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
44
|
Shenoy A, Banerjee M, Upadhya A, Bagwe-Parab S, Kaur G. The Brilliance of the Zebrafish Model: Perception on Behavior and Alzheimer's Disease. Front Behav Neurosci 2022; 16:861155. [PMID: 35769627 PMCID: PMC9234549 DOI: 10.3389/fnbeh.2022.861155] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) has become increasingly prevalent in the elderly population across the world. It's pathophysiological markers such as overproduction along with the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFT) are posing a serious challenge to novel drug development processes. A model which simulates the human neurodegenerative mechanism will be beneficial for rapid screening of potential drug candidates. Due to the comparable neurological network with humans, zebrafish has emerged as a promising AD model. This model has been thoroughly validated through research in aspects of neuronal pathways analogous to the human brain. The cholinergic, glutamatergic, and GABAergic pathways, which play a role in the manifested behavior of the zebrafish, are well defined. There are several behavioral models in both adult zebrafish and larvae to establish various aspects of cognitive impairment including spatial memory, associative memory, anxiety, and other such features that are manifested in AD. The zebrafish model eliminates the shortcomings of previously recognized mammalian models, in terms of expense, extensive assessment durations, and the complexity of imaging the brain to test the efficacy of therapeutic interventions. This review highlights the various models that analyze the changes in the normal behavioral patterns of the zebrafish when exposed to AD inducing agents. The mechanistic pathway adopted by drugs and novel therapeutic strategies can be explored via these behavioral models and their efficacy to slow the progression of AD can be evaluated.
Collapse
Affiliation(s)
| | | | | | | | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s Narsee Monjee Institute of Management Studies, Mumbai, India
| |
Collapse
|
45
|
Rahmani B, Ghashghayi E, Zendehdel M, Baghbanzadeh A, Khodadadi M. Molecular mechanisms highlighting the potential role of COVID-19 in the development of neurodegenerative diseases. Physiol Int 2022; 109:135-162. [DOI: 10.1556/2060.2022.00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/17/2022] [Accepted: 04/11/2022] [Indexed: 01/08/2023]
Abstract
Abstract
Coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to the pulmonary manifestations, COVID-19 patients may present a wide range of neurological disorders as extrapulmonary presentations. In this view, several studies have recently documented the worsening of neurological symptoms within COVID-19 morbidity in patients previously diagnosed with neurodegenerative diseases (NDs). Moreover, several cases have also been reported in which the patients presented parkinsonian features after initial COVID-19 symptoms. These data raise a major concern about the possibility of communication between SARS-CoV-2 infection and the initiation and/or worsening of NDs. In this review, we have collected compelling evidence suggesting SARS-CoV-2, as an environmental factor, may be capable of developing NDs. In this respect, the possible links between SARS-CoV-2 infection and molecular pathways related to most NDs and the pathophysiological mechanisms of the NDs such as Alzheimer's disease, vascular dementia, frontotemporal dementia, Parkinson's disease, and amyotrophic lateral sclerosis will be explained.
Collapse
Affiliation(s)
- Behrouz Rahmani
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Elham Ghashghayi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Morteza Zendehdel
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Ali Baghbanzadeh
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| | - Mina Khodadadi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tehran, 14155-6453 Tehran, Iran
| |
Collapse
|
46
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
47
|
Benmelouka AY, Ouerdane Y, Outani O, Alnasser YT, Alghamdi BS, Perveen A, Ashraf GM, Ebada MA. Alzheimer's Disease-Related Psychosis: An Overview of Clinical Manifestations, Pathogenesis, and Current Treatment. Curr Alzheimer Res 2022; 19:285-301. [PMID: 35440308 DOI: 10.2174/1567205019666220418151914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/25/2022] [Accepted: 03/06/2022] [Indexed: 11/22/2022]
Abstract
Behavioral and psychotic manifestations, including aggression, delusions, and hallucinations, are frequent comorbidities in patients with debilitating nervous illnesses such as Alzheimer's disease (AD), Amyotrophic Lateral Sclerosis, Multiple Sclerosis, and Parkinson's disease. AD-related psychosis may be linked to a poor disease prognosis, highlighting that early detection and management are mandatory. The manifestations are variable and may be very heterogeneous, imposing a real diagnostic issue. Some assessment tools such as BEHAVE-AD, CERAD-BRSD, and the Psycho-Sensory Hallucinations Scale have been designed to facilitate the diagnosis. The mechanisms behind neurodegeneration-related psychosis are complex and are not fully understood, imposing a burden on researchers to find appropriate management modalities. Familial history and some genetic disturbances may have a determinant role in these delusions and hallucinations in cases with AD. The loss of neuronal cells, atrophy in some regions of the central nervous, and synaptic dysfunction may also contribute to these comorbidities. Furthermore, inflammatory disturbances triggered by pro-inflammatory agents such as interleukins and tumor necrosis factors are stratified among the potential risk factors of the onset of numerous psychotic symptoms in Alzheimer's patients. Little is known about the possible management tools; therefore, it is urgent to conduct well-designed trials to investigate pharmacological and non-pharmacological interventions that can improve the care process of these patients. This review summarizes the current findings regarding the AD-related psychosis symptoms, pathological features, assessment, and management.
Collapse
Affiliation(s)
| | | | - Oumaima Outani
- Faculty of Medicine and Pharmacy of Rabat, Mohammed 5 University
| | | | - Badrah S Alghamdi
- Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah.,Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah
| | - Asma Perveen
- Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah.,Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah
| | - Mahmoud Ahmed Ebada
- Faculty of Medicine, Zagazig University, Zagazig, Al-Sharkia.,Internal Medicine Resident, Ministry of Health and Population of Egypt, Cairo
| |
Collapse
|
48
|
Jenkins LM, Wang L, Rosen H, Weintraub S. A transdiagnostic review of neuroimaging studies of apathy and disinhibition in dementia. Brain 2022; 145:1886-1905. [PMID: 35388419 PMCID: PMC9630876 DOI: 10.1093/brain/awac133] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/18/2022] [Accepted: 03/13/2022] [Indexed: 11/12/2022] Open
Abstract
Apathy and disinhibition are common and highly distressing neuropsychiatric symptoms associated with negative outcomes in persons with dementia. This paper is a critical review of functional and structural neuroimaging studies of these symptoms transdiagnostically in dementia of the Alzheimer type, which is characterized by prominent amnesia early in the disease course, and behavioural variant frontotemporal dementia, characterized by early social-comportmental deficits. We describe the prevalence and clinical correlates of these symptoms and describe methodological issues, including difficulties with symptom definition and different measurement instruments. We highlight the heterogeneity of findings, noting however, a striking similarity of the set of brain regions implicated across clinical diagnoses and symptoms. These regions involve several key nodes of the salience network, and we describe the functions and anatomical connectivity of these brain areas, as well as present a new theoretical account of disinhibition in dementia. Future avenues for research are discussed, including the importance of transdiagnostic studies, measuring subdomains of apathy and disinhibition, and examining different units of analysis for deepening our understanding of the networks and mechanisms underlying these extremely distressing symptoms.
Collapse
Affiliation(s)
- Lisanne M Jenkins
- Correspondence to: Lisanne Jenkins 710 N Lakeshore Drive, Suite 1315 Chicago, IL 60611, USA E-mail:
| | - Lei Wang
- Department of Psychiatry and Behavioral Health, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Howie Rosen
- Weill Institute for Neurosciences, School of Medicine, University of California, San Francisco, CA, USA 94158
| | - Sandra Weintraub
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA,Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA 60611
| |
Collapse
|
49
|
Alves SS, da Silva Junior RMP, Delfino-Pereira P, Pereira MGAG, Vasconcelos I, Schwaemmle H, Mazzei RF, Carlos ML, Espreafico EM, Tedesco AC, Sebollela A, Almeida SS, de Oliveira JAC, Garcia-Cairasco N. A Genetic Model of Epilepsy with a Partial Alzheimer's Disease-Like Phenotype and Central Insulin Resistance. Mol Neurobiol 2022; 59:3721-3737. [PMID: 35378696 DOI: 10.1007/s12035-022-02810-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/22/2022] [Indexed: 12/20/2022]
Abstract
Studies have suggested an important connection between epilepsy and Alzheimer's disease (AD), mostly due to the high number of patients diagnosed with AD who develop epileptic seizures later on. However, this link is not well understood. Previous studies from our group have identified memory impairment and metabolic abnormalities in the Wistar audiogenic rat (WAR) strain, a genetic model of epilepsy. Our goal was to investigate AD behavioral and molecular alterations, including brain insulin resistance, in naïve (seizure-free) animals of the WAR strain. We used the Morris water maze (MWM) test to evaluate spatial learning and memory performance and hippocampal tissue to verify possible molecular and immunohistochemical alterations. WARs presented worse performance in the MWM test (p < 0.0001), higher levels of hyperphosphorylated tau (S396) (p < 0.0001) and phosphorylated glycogen synthase kinase 3 (S21/9) (p < 0.05), and lower insulin receptor levels (p < 0.05). Conversely, WARs and Wistar controls present progressive increase in amyloid fibrils (p < 0.0001) and low levels of soluble amyloid-β. Interestingly, the detected alterations were age-dependent, reaching larger differences in aged than in young adult animals. In summary, the present study provides evidence of a partial AD-like phenotype, including altered regulation of insulin signaling, in a genetic model of epilepsy. Together, these data contribute to the understanding of the connection between epilepsy and AD as comorbidities. Moreover, since both tau hyperphosphorylation and altered insulin signaling have already been reported in epilepsy and AD, these two events should be considered as important components in the interconnection between epilepsy and AD pathogenesis and, therefore, potential therapeutic targets in this field.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | | | - Polianna Delfino-Pereira
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | | | - Israel Vasconcelos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Hanna Schwaemmle
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Rodrigo Focosi Mazzei
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - Maiko Luiz Carlos
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - Enilza Maria Espreafico
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Antônio Claudio Tedesco
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Sebastião Sousa Almeida
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - José Antônio Cortes de Oliveira
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Av. Dos Bandeirantes 3900, Ribeirao Preto, SP, 14049-900, Brazil
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil.
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Av. Dos Bandeirantes 3900, Ribeirao Preto, SP, 14049-900, Brazil.
| |
Collapse
|
50
|
Sex differences in neuropsychiatric symptoms in Alzheimer's disease dementia: a meta-analysis. Alzheimers Res Ther 2022; 14:48. [PMID: 35379344 PMCID: PMC8978393 DOI: 10.1186/s13195-022-00991-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/17/2022] [Indexed: 12/25/2022]
Abstract
Background Neuropsychiatric symptoms (NPS) are common in individuals with Alzheimer’s disease (AD) dementia, but substantial heterogeneity exists in the manifestation of NPS. Sex differences may explain this clinical variability. We aimed to investigate the sex differences in the prevalence and severity of NPS in AD dementia. Methods Literature searches were conducted in Embase, MEDLINE/PubMed, Web of Science Core Collection, Cochrane Central Register of Controlled Trials, PsycINFO, and Google Scholar from inception to February 2021. Study selection, data extraction, and quality assessment were conducted in duplicate. Effect sizes were calculated as odds ratios (OR) for NPS prevalence and Hedges’ g for NPS severity. Data were pooled using random-effects models. Sources of heterogeneity were examined using meta-regression analyses. Results Sixty-two studies were eligible representing 21,554 patients (61.2% females). The majority of the included studies had an overall rating of fair quality (71.0%), with ten studies of good quality (16.1%) and eight studies of poor quality (12.9%). There was no sex difference in the presence of any NPS (k = 4, OR = 1.35 [95% confidence interval 0.78, 2.35]) and overall NPS severity (k = 13, g = 0.04 [− 0.04, 0.12]). Regarding specific symptoms, female sex was associated with more prevalent depressive symptoms (k = 20, OR = 1.60 [1.28, 1.98]), psychotic symptoms (general psychosis k = 4, OR = 1.62 [1.12, 2.33]; delusions k = 12, OR = 1.56 [1.28, 1.89]), and aberrant motor behavior (k = 6, OR = 1.47 [1.09, 1.98]). In addition, female sex was related to more severe depressive symptoms (k = 16, g = 0.24 [0.14, 0.34]), delusions (k = 10, g = 0.19 [0.04, 0.34]), and aberrant motor behavior (k = 9, g = 0.17 [0.08, 0.26]), while apathy was more severe among males compared to females (k = 11, g = − 0.10 [− 0.18, − 0.01]). There was no association between sex and the prevalence and severity of agitation, anxiety, disinhibition, eating behavior, euphoria, hallucinations, irritability, and sleep disturbances. Meta-regression analyses revealed no consistent association between the effect sizes across studies and method of NPS assessment and demographic and clinical characteristics. Discussion Female sex was associated with a higher prevalence and greater severity of several specific NPS, while male sex was associated with more severe apathy. While more research is needed into factors underlying these sex differences, our findings may guide tailored treatment approaches of NPS in AD dementia. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00991-z.
Collapse
|