1
|
Mori AD, Tauber C, Alzheimer’s Disease Neuroimaging Initiative. Longitudinal dementia trajectories for Alzheimer's Disease characterization and prediction. Comput Biol Med 2025; 192:110241. [PMID: 40345134 DOI: 10.1016/j.compbiomed.2025.110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/19/2025] [Accepted: 04/19/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Alzheimer's Disease (AD) remains one of the most significant neurodegenerative diseases globally, affecting approximately 38.5 million people in 2023. Early identification of individuals at risk of developing AD is essential to managing disease progression and implementing timely interventions. Despite extensive research on predicting individual AD progression and diagnosis conversion, the dynamic and multimodal characterization of AD evolution remains an open challenge. APPROACH This study introduces a model leveraging multimodal clinical data to capture and predict both population-level and individual trajectories of dementia over extended timescales. We define a Static Dementia Score (SDS) as a metric representing the current state of dementia, with its temporal evolution modeled using a Bayesian nonlinear mixed-effects approach. This method generates a single continuous curve that delineates the average dementia progression pattern across the population. Additionally, our approach allows for patient-specific SDS progression modeling, enabling the prediction of individual dementia trajectories over a specified time horizon. RESULTS The model was trained on a dataset comprising 5,033 observations, including MP-RAGE MRI scans, cognitive assessments, and demographic data from 883 individuals with at least four observations and stable diagnostic trajectories. Population-level curves produced by our model align with established AD progression trends, capturing key stages of disease evolution. Individual-specific deformation parameters were effective in characterizing personalized disease progression, achieving a theoretical diagnosis prediction accuracy of 0.952±0.013 three years in advance and an AD conversion prediction accuracy of 0.916±0.022 within the same period. CONCLUSION Our findings underscore the potential of this approach for AD classification and forecasting disease progression. These results emphasize its utility for clinicians and researchers in detecting atypical disease trajectories early in patients with longitudinal follow-up, enhancing decision-making and therapeutic planning.
Collapse
Affiliation(s)
- Antoine de Mori
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France
| | - Clovis Tauber
- Université de Tours, INSERM, Imaging Brain & Neuropsychiatry iBraiN U1253, 37032, Tours, France.
| | | |
Collapse
|
2
|
Wang R, Li J, Li X, Guo Y, Chen P, Peng T. Exercise-induced modulation of miRNAs and gut microbiome: a holistic approach to neuroprotection in Alzheimer's disease. Rev Neurosci 2025:revneuro-2025-0013. [PMID: 40366727 DOI: 10.1515/revneuro-2025-0013] [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: 01/24/2025] [Accepted: 03/28/2025] [Indexed: 05/15/2025]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, is marked by cognitive decline, neuroinflammation, and neuronal loss. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression, influencing key pathways involved in neuroinflammation and neurodegeneration in AD. This review delves into the multifaceted role of exercise in modulating miRNA expression and its interplay with the gut microbiome, proposing a comprehensive framework for neuroprotection in AD. By synthesizing current research, we elucidate how exercise-induced changes in miRNA profiles can mitigate inflammatory responses, promote neurogenesis, and reduce amyloid-beta and tau pathologies. Additionally, we explore the gut-brain axis, highlighting how exercise-driven alterations in gut microbiota composition can further influence miRNA expression, thereby enhancing cognitive function and reducing neuroinflammatory markers. This holistic approach underscores the potential of targeting exercise-regulated miRNAs and gut microbiome interactions as a novel, noninvasive therapeutic strategy to decelerate AD progression and improve quality of life for patients. This approach aims to decelerate disease progression and improve patient outcomes, offering a promising avenue for enhancing the effectiveness of AD management.
Collapse
Affiliation(s)
- Rui Wang
- College of Physical Education, Guizhou Normal University, GuiYang 550025, China
| | - Juan Li
- Hanyang University Erica, AnSan 15588, Korea
| | - Xiaochen Li
- School of Physical Education, Huaibei Normal University, HuaiBei 235000, China
| | - Yan Guo
- Sichuan University Jinjiang College, ChengDu 610000, China
| | - Pei Chen
- School of Physical Education, Huaibei Normal University, HuaiBei 235000, China
| | - Tian Peng
- Department of Physical Education, 12377 Zhejiang University of Science and Technology , HangZhou 310023, China
| |
Collapse
|
3
|
Goel SA, Singh R, John DR, Suthar P, Singh JS. Advances in Molecular Imaging for the Early Detection and Management of Alzheimer's Disease. Cureus 2025; 17:e83127. [PMID: 40438850 PMCID: PMC12118448 DOI: 10.7759/cureus.83127] [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] [Accepted: 04/27/2025] [Indexed: 06/01/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by impairments in memory and cognitive abilities. The development of new immunotherapies targeting beta-amyloid (Aβ) and tau protein deposition in the brain is ushering in great advances in clinical management. Advances in molecular imaging techniques, particularly positron emission tomography (PET) with amyloid and tau tracers, have facilitated the early detection of Alzheimer's pathology. We report a case of a 67-year-old patient presenting with mild cognitive impairment (MCI) who was confirmed to have AD on newer PET imaging. This report highlights the important role of molecular imaging in the early diagnosis of AD before significant clinical and functional decline sets in, thereby emphasizing its role in clinical practice.
Collapse
Affiliation(s)
- Shiv A Goel
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, USA
| | - Rishab Singh
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, USA
| | | | - Pokhraj Suthar
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, USA
| | - Jagadeesh S Singh
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, USA
| |
Collapse
|
4
|
Bagnati PM, Londoño Castaño M, Fernández ML, Henao BM, Chrem P, Aguillón D, Varela LE, Barbaran JD, Leon Y, Surace E, Madrigal CC, Picasso JP, Ramos CP, Fernández CMR, Vigo G, Aguilar LR, VargasCuadros GA, Arcos‐Burgos M, Longoria EM, Ziegemeier E, McDade E, Bateman RJ, Allegri RF, Lopera F, Llibre‐Guerra JJ. Impact of genetic counseling and testing in individuals at high risk of familial Alzheimer's disease from Latin America: a non-randomized controlled trial. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70102. [PMID: 40201594 PMCID: PMC11973255 DOI: 10.1002/dad2.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION This study involved evaluating a tailored genetic counseling and testing (GCT) protocol for families at risk of autosomal dominant Alzheimer's disease (ADAD) in Latin America (LatAm), focusing on essential cultural and regional adaptations. METHODS We conducted a non-randomized controlled trial among ADAD families in Colombia and Argentina. Participants were categorized based on their decision to learn their genetic status (GS), with further comparisons between mutation-positive versus mutation-negative participants who learned their status. Psychological impacts were measured using validated scales for anxiety and depression. RESULTS Of the 122 eligible participants, 97 completed the GCT protocol, and 87 opted to learn their GS. There were no clinically significant differences in psychological distress between those who learned their status and those who did not, nor between mutation-positive and mutation-negative individuals. DISCUSSION The GCT protocol effectively managed psychological impacts in ADAD families and was positively received, demonstrating the importance of culturally adapted GCT protocols. Highlights We examined the adaptation and efficacy of a GCT protocol in LatAm for families at risk of ADAD.The GCT protocol mitigated psychological distress among at-risk ADAD families.The study confirms the protocol's cultural appropriateness and psychological safety.Future studies should explore the long-term psychological and public health impacts of GCT and use of GCT for treatment options.
Collapse
Affiliation(s)
- Pablo M. Bagnati
- Department of Cognitive NeurologyNeuropsychiatry and NeuropsychologyFleni Neurological Research InstituteBuenos AiresArgentina
| | - Marisol Londoño Castaño
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
- Facultad de Teología, Filosofía y HumanidadesUniversidad Pontifícia BolivarianaMedellinColombia
| | - María Laura Fernández
- Department of Cognitive NeurologyNeuropsychiatry and NeuropsychologyFleni Neurological Research InstituteBuenos AiresArgentina
| | | | - Patricio Chrem
- Department of Cognitive NeurologyNeuropsychiatry and NeuropsychologyFleni Neurological Research InstituteBuenos AiresArgentina
| | - David Aguillón
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
| | - Luz Estela Varela
- Fundación Médica de Enfermedades RarasMedellínColombia
- Facultad de EnfermeríaUniversidad de AntioquiaMedellinColombia
| | | | - Yudy Leon
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
| | - Ezequiel Surace
- Department of Cognitive NeurologyNeuropsychiatry and NeuropsychologyFleni Neurological Research InstituteBuenos AiresArgentina
| | - Claudia C. Madrigal
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
| | - Juan Pablo Picasso
- Department of Cognitive NeurologyNeuropsychiatry and NeuropsychologyFleni Neurological Research InstituteBuenos AiresArgentina
| | - Claudia P. Ramos
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
| | | | - Gabriela Vigo
- Servicio de NeurologíaHospital San BernardoSaltaArgentina
| | - Laura Ramirez Aguilar
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
| | | | - Mauricio Arcos‐Burgos
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
| | - Erika Mariana Longoria
- Cognitive Neurosciences LabNational Institute of Neurology and NeurosurgeryMexico CityMexico
| | - Ellen Ziegemeier
- Department of NeurologyWashington University School of MedicineSt LouisMissouriUSA
| | - Eric McDade
- Department of NeurologyWashington University School of MedicineSt LouisMissouriUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt LouisMissouriUSA
| | - Ricardo F. Allegri
- Department of Cognitive NeurologyNeuropsychiatry and NeuropsychologyFleni Neurological Research InstituteBuenos AiresArgentina
| | - Francisco Lopera
- Grupo de Neurociencias de AntioquiaFacultad de MedicinaUniversidad de AntioquiaMedellínColombia
| | | |
Collapse
|
5
|
Onifade IA, Umar HI, Aborode AT, Awaji AA, Jegede ID, Adeleye BH, Fatoba DO, Bello RO, Fakorede S, Idowu N. In silico study of selected alkaloids as dual inhibitors of β- and γ-secretases for Alzheimer's disease. J Alzheimers Dis 2025; 103:1191-1215. [PMID: 39956948 DOI: 10.1177/13872877241313049] [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: 02/18/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) has become common as the number of aged people increases making it as a socioeconomic problem lately. To date, no success is recorded for disease-modifying therapies for AD but only drugs for symptomatic relief exist. Research has been centered on the role of amyloid-β on the pathogenesis of AD, which has led to the development of drugs that target Aβ (β- and γ-secretase inhibitors) to reduce the amount of Aβ formed. However, the existing β and γ-secretase inhibitors were associated with harmful side effects, low efficacy, and inability to cross the blood-brain barrier. OBJECTIVE This study therefore used in silico approach to predict the inhibitory properties of alkaloids as potential drug targets against AD. METHODS Thus, in this current study, 54 alkaloids from the PhytoHub server (phytohub.eu), and two approved drugs were docked against β-secretases. Additionally, galantamine and 5 alkaloids with the utmost binding potential with β-secretase were subjected to pharmacokinetics evaluation and docked against γ-secretase. RESULTS From the result, 5 compounds displayed for both docking periods, with demissidine, solasodine, tomatidine, and solanidine having better BE than the control drugs. Based on the pharmacokinetics evaluation, 4 compounds possessed good pharmacokinetic evaluation and biological activities than galantamine. CONCLUSIONS This study suggests that demissidine, solasodine, tomatidine, and solanidine are promising dual inhibitors against β- and γ-secretase proteins in silico. However, there is an urgent need to carry out in vitro and in vivo experiments on these new leads to validate the findings of this study.
Collapse
Affiliation(s)
- Isreal Ayobami Onifade
- Department of Biological Sciences, University at Albany - State University of New York, Albany, NY, USA
| | - Haruna Isiyaku Umar
- Department of Biochemistry, Federal University of Technology, Akure, Nigeria
- Computer-Aided Therapeutic Discovery and Design Platform, Federal University of Technology, Akure, Nigeria
| | | | - Aeshah A Awaji
- Department of Biology, Faculty of Science, University College of Taymaa, University of Tabuk, Tabuk, Saudi Arabia
| | | | | | | | | | - Sodiq Fakorede
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nike Idowu
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
6
|
Sohn E, Lim HS, Jin Kim Y, Kim BY, Yoon J, Kim JH, Jeong SJ. Exploring the therapeutic potential of Potentilla fragarioides var. major (Rosaceae) extract in Alzheimer's disease using in vitro and in vivo models: A multi-faceted approach. Neuroscience 2024; 559:77-90. [PMID: 39179018 DOI: 10.1016/j.neuroscience.2024.08.029] [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: 04/23/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and is caused by various factors including amyloid-beta (Aβ) aggregation. We investigated the pharmacological effects of the ethanol extract of Potentilla fragarioides var. major (Rosaceae) (EEPF) on AD-related pathogenesis, which remain elusive. We observed the effects of EEPF on Aβ disaggregation and free-radical scavenging activities for 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) using in vitro assays, evaluated the effects of EEPF on memory loss in two animal models, and examined the molecular regulatory mechanisms of EEPF using an antibody-protein microarray in EEPF-treated neuronal cell lines. EEPF inhibited Aβ aggregation in a concentration-dependent manner and enhanced free-radical scavenging activities for ABTS and DPPH. EEPF significantly inhibited memory impairment in the passive avoidance task, Y-maze test, and Morris water maze test in scopolamine-induced short-term memory loss mice and Aβ-injected AD-like mice. Nissl staining and immunohistochemistry for NeuN and Iba-1 confirmed the neuroprotective and anti-inflammatory effects of EEPF in both animal models. In H2O2-treated HT22 hippocampal cells, EEPF significantly prevented cell damage, enhanced CaMK2, and reduced ferric reductase. In lipopolysaccharide (LPS)-stimulated BV-2 microglia, EEPF significantly inhibited LPS-induced production of inflammatory factors, such as nitric oxide, prostaglandin E2, tumor necrosis factor-α, and interleukin-6, and decreased the phosphorylation of Smad3 and cyclin D3. High-performance liquid chromatography confirmed that EEPF has five major components: neochlorogenic acid, chlorogenic acid, polydatin, isochlorogenic acid A, and buddleoside, with amounts ranging across 1.91-9.41 mg/g. EEPF may be a promising drug for treatment of AD and AD-related brain disorders.
Collapse
Affiliation(s)
- Eunjin Sohn
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Hye-Sun Lim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 111 Geonjae-ro, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Yu Jin Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Bu-Yeo Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Jiyeon Yoon
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Joo-Hwan Kim
- Department of Life Science, Gachon University, 1342, Seongnam-daero, Sujeong-gu, Seongnam, Gyeonggi-do 13120, Republic of Korea.
| | - Soo-Jin Jeong
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| |
Collapse
|
7
|
Li M, Mo Y, Yu Q, Anayyat U, Yang H, Zhang F, Wei Y, Wang X. Rotating magnetic field improves cognitive and memory impairments in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway. Exp Neurol 2024; 383:115029. [PMID: 39461710 DOI: 10.1016/j.expneurol.2024.115029] [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: 06/08/2024] [Revised: 08/16/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Alzheimer's disease (AD) is a geriatric disorder that can be roughly classified into sporadic AD and hereditary AD. The latter is strongly associated with genetic factors, and its treatment poses greater challenges compared to sporadic AD. Rotating magnetic fields (RMF) is a non-invasive treatment known to have diverse biological effects, including the modulation of the central nervous system and aging. However, the impact of RMF on hereditary AD and its underlying mechanism remain unexplored. In this study, we exposed APP/PS1 mice to RMF (2 h/day, 0.2 T, 4 Hz) for a duration of 6 months. The results demonstrated that RMF treatment significantly ameliorated their cognitive and memory impairments, attenuated neuronal damage, and reduced amyloid deposition. Furthermore, RNA-sequencing analysis revealed a significant enrichment of autophagy-related genes and the PI3K/AKT-mTOR signaling pathway. Western blotting further confirmed that RMF activated autophagy and suppressed the phosphorylation of proteins associated with the PI3K/AKT/mTOR signaling pathway in APP/PS1 mice. These protective effects and the underlying mechanism were also observed in Aβ25-35-exposed HT22 cells. Collectively, our findings indicate that RMF improves cognitive and memory dysfunction in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway, thus highlighting the potential of RMF as a clinical treatment for hereditary AD.
Collapse
Affiliation(s)
- Mengqing Li
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Yaxian Mo
- Songgang People's Hospital, Shenzhen, Guangdong 518105, China
| | - Qinyao Yu
- School of Pharmacy, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Umer Anayyat
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Hua Yang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Fen Zhang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China
| | - Yunpeng Wei
- Songgang People's Hospital, Shenzhen, Guangdong 518105, China.
| | - Xiaomei Wang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518061, China; International Cancer Center, Shenzhen University Health Sciences Center, Shenzhen, Guangdong 518061, China.
| |
Collapse
|
8
|
Dang J, Yong ACW, Fong ZH, Ang K, Ng ASL. A Systematic Review of Palliative Care Needs in Young-Onset Dementia. J Am Med Dir Assoc 2024; 25:105219. [PMID: 39155044 DOI: 10.1016/j.jamda.2024.105219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND The distinctive differences in clinical needs and disease trajectory between people with young-onset (YOD) and late-onset dementia (LOD) make dementia palliative care unique. Limited studies have reported on the differences in palliative care needs between YOD and LOD, and the optimal time point to introduce palliative care in YOD remains controversial. We performed a systematic review to summarize key issues surrounding palliative care in YOD and highlight unmet needs in this pertinent area. METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we searched the PubMed database for all studies published between January 2000 and July 2022 that reported on palliative care in YOD. RESULTS Of 32 records identified, 8 articles were eligible for inclusion. The top 3 themes extracted centered around (1) clinical differences between YOD and LOD, (2) symptoms and causes of death in end-stage YOD, and (3) the importance of early advanced care planning (ACP). YOD diagnosis is often delayed and people with YOD have fewer somatic comorbidities but more neuropsychiatric symptoms, longer survival times, and a more malignant disease course. People with YOD and their families face unique psychosocial challenges when symptoms start at a younger age. End-stage YOD is not dissimilar to LOD where patients suffer from a broad spectrum of physical and psychological symptoms requiring palliation. Early initiation of ACP discussion is crucial in YOD given the more rapid progression of disease affecting cognition and decision-making capacity; however, rates of ACP completion in YOD remain low. CONCLUSIONS Given the complex care needs and more rapid disease trajectory in YOD, palliative care in YOD should be considered from the time of diagnosis, and to be incorporated into routine dementia care.
Collapse
Affiliation(s)
- Jiaojiao Dang
- Department of Neurology, National Neuroscience Institute, Singapore.
| | - Alisa Cui Wen Yong
- Department of Psychology, Faculty of Arts and Social Sciences, National University of Singapore, Singapore
| | - Zhi Hui Fong
- Department of Neurology, National Neuroscience Institute, Singapore
| | - Kexin Ang
- Department of Neurology, National Neuroscience Institute, Singapore
| | - Adeline Su Lyn Ng
- Department of Neurology, National Neuroscience Institute, Singapore; Neuroscience and Behavioural Disorders Unit, Duke-NUS Medical School, Singapore
| |
Collapse
|
9
|
Li YB, Fu Q, Guo M, Du Y, Chen Y, Cheng Y. MicroRNAs: pioneering regulators in Alzheimer's disease pathogenesis, diagnosis, and therapy. Transl Psychiatry 2024; 14:367. [PMID: 39256358 PMCID: PMC11387755 DOI: 10.1038/s41398-024-03075-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
This article delves into Alzheimer's disease (AD), a prevalent neurodegenerative condition primarily affecting the elderly. It is characterized by progressive memory and cognitive impairments, severely disrupting daily life. Recent research highlights the potential involvement of microRNAs in the pathogenesis of AD. MicroRNAs (MiRNAs), short non-coding RNAs comprising 20-24 nucleotides, significantly influence gene regulation by hindering translation or promoting degradation of target genes. This review explores the role of specific miRNAs in AD progression, focusing on their impact on β-amyloid (Aβ) peptide accumulation, intracellular aggregation of hyperphosphorylated tau proteins, mitochondrial dysfunction, neuroinflammation, oxidative stress, and the expression of the APOE4 gene. Our insights contribute to understanding AD's pathology, offering new avenues for identifying diagnostic markers and developing novel therapeutic targets.
Collapse
Affiliation(s)
- Yao-Bo Li
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Qiang Fu
- Institute of National Security, Minzu University of China, Beijing, China
| | - Mei Guo
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yang Du
- Institute of National Security, Minzu University of China, Beijing, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China.
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
- Institute of National Security, Minzu University of China, Beijing, China.
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China.
| |
Collapse
|
10
|
Marmor A, Vakil E, Kahana Merhavi S, Meiner Z. The complex interplay between cognitive reserve, age of diagnosis and cognitive decline in Alzheimer's disease: a retrospective study. J Clin Exp Neuropsychol 2024; 46:683-692. [PMID: 39235435 DOI: 10.1080/13803395.2024.2400109] [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] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE The present study examined the cognitive reserve (CR) theory at late stages of Alzheimer's disease (AD). The objective is to replicate previous studies and examine the complex role of education and family size as indicators of CR. PARTICIPANTS AND METHODS This is a retrospective study included 642 patients diagnosed with AD after age 65, categorized into low education (LE, ≤ 8 years, n = 141) and medium-high education (MHE, ≥ 9 years, n = 442) groups. Participants were followed up longitudinally using the Mini Mental State Examination. RESULTS Higher education in the MHE group, but not in the LE group, correlated with delayed diagnosis. In both groups, higher education correlated with accelerated cognitive decline. In the MHE group, country of origin was associated with cognitive decline, while in the LE group, it was linked to family size. CONCLUSIONS This study shows that in patients with MHE but not in LE, higher education resulted in delayed diagnosis. Conversely, in cases of LE, this measure may not fully reflect CR and abilities. Additionally, higher education was associated with faster deterioration, a finding that has not been replicated often in the literature. The study illustrates the complex impact of CR proxies on age of diagnosis and cognitive decline.
Collapse
Affiliation(s)
- Anat Marmor
- Department of Physical & Medical Rehabilitation, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Psychology, and Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Eli Vakil
- Department of Psychology, and Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Shlomzion Kahana Merhavi
- Department of Neurology, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Zeev Meiner
- Department of Physical & Medical Rehabilitation, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
11
|
McInvale JJ, Canoll P, Hargus G. Induced pluripotent stem cell models as a tool to investigate and test fluid biomarkers in Alzheimer's disease and frontotemporal dementia. Brain Pathol 2024; 34:e13231. [PMID: 38246596 PMCID: PMC11189780 DOI: 10.1111/bpa.13231] [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: 10/03/2023] [Accepted: 11/29/2023] [Indexed: 01/23/2024] Open
Abstract
Neurodegenerative diseases are increasing in prevalence and comprise a large socioeconomic burden on patients and their caretakers. The need for effective therapies and avenues for disease prevention and monitoring is of paramount importance. Fluid biomarkers for neurodegenerative diseases have gained a variety of uses, including informing participant selection for clinical trials, lending confidence to clinical diagnosis and disease staging, determining prognosis, and monitoring therapeutic response. Their role is expected to grow as disease-modifying therapies start to be available to a broader range of patients and as prevention strategies become established. Many of the underlying molecular mechanisms of currently used biomarkers are incompletely understood. Animal models and in vitro systems using cell lines have been extensively employed but face important translatability limitations. Induced pluripotent stem cell (iPSC) technology, where a theoretically unlimited range of cell types can be reprogrammed from peripheral cells sampled from patients or healthy individuals, has gained prominence over the last decade. It is a promising avenue to study physiological and pathological biomarker function and response to experimental therapeutics. Such systems are amenable to high-throughput drug screening or multiomics readouts such as transcriptomics, lipidomics, and proteomics for biomarker discovery, investigation, and validation. The present review describes the current state of biomarkers in the clinical context of neurodegenerative diseases, with a focus on Alzheimer's disease and frontotemporal dementia. We include a discussion of how iPSC models have been used to investigate and test biomarkers such as amyloid-β, phosphorylated tau, neurofilament light chain or complement proteins, and even nominate novel biomarkers. We discuss the limitations of current iPSC methods, mentioning alternatives such as coculture systems and three-dimensional organoids which address some of these concerns. Finally, we propose exciting prospects for stem cell transplantation paradigms using animal models as a preclinical tool to study biomarkers in the in vivo context.
Collapse
Affiliation(s)
- Julie J. McInvale
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
- Medical Scientist Training Program, Columbia UniversityNew YorkNew YorkUSA
| | - Peter Canoll
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | - Gunnar Hargus
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
12
|
Valdez-Gaxiola CA, Rosales-Leycegui F, Gaxiola-Rubio A, Moreno-Ortiz JM, Figuera LE. Early- and Late-Onset Alzheimer's Disease: Two Sides of the Same Coin? Diseases 2024; 12:110. [PMID: 38920542 PMCID: PMC11202866 DOI: 10.3390/diseases12060110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/04/2024] [Accepted: 05/18/2024] [Indexed: 06/27/2024] Open
Abstract
Early-onset Alzheimer's disease (EOAD), defined as Alzheimer's disease onset before 65 years of age, has been significantly less studied than the "classic" late-onset form (LOAD), although EOAD often presents with a more aggressive disease course, caused by variants in the APP, PSEN1, and PSEN2 genes. EOAD has significant differences from LOAD, including encompassing diverse phenotypic manifestations, increased genetic predisposition, and variations in neuropathological burden and distribution. Phenotypically, EOAD can be manifested with non-amnestic variants, sparing the hippocampi with increased tau burden. The aim of this article is to review the different genetic bases, risk factors, pathological mechanisms, and diagnostic approaches between EOAD and LOAD and to suggest steps to further our understanding. The comprehension of the monogenic form of the disease can provide valuable insights that may serve as a roadmap for understanding the common form of the disease.
Collapse
Affiliation(s)
- César A. Valdez-Gaxiola
- División de Genética, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara 44340, Jalisco, Mexico; (C.A.V.-G.); (F.R.-L.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Frida Rosales-Leycegui
- División de Genética, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara 44340, Jalisco, Mexico; (C.A.V.-G.); (F.R.-L.)
- Maestría en Ciencias del Comportamiento, Instituto de Neurociencias, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Abigail Gaxiola-Rubio
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
- Facultad de Medicina, Universidad Autónoma de Guadalajara, Zapopan 45129, Jalisco, Mexico
| | - José Miguel Moreno-Ortiz
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Instituto de Genética Humana “Dr. Enrique Corona Rivera”, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Luis E. Figuera
- División de Genética, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara 44340, Jalisco, Mexico; (C.A.V.-G.); (F.R.-L.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
13
|
Pérez-Millan A, Borrego-Écija S, Falgàs N, Juncà-Parella J, Bosch B, Tort-Merino A, Antonell A, Bargalló N, Rami L, Balasa M, Lladó A, Sala-Llonch R, Sánchez-Valle R. Cortical thickness modeling and variability in Alzheimer's disease and frontotemporal dementia. J Neurol 2024; 271:1428-1438. [PMID: 38012398 PMCID: PMC10896866 DOI: 10.1007/s00415-023-12087-1] [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: 06/09/2023] [Revised: 09/29/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND AND OBJECTIVE Alzheimer's disease (AD) and frontotemporal dementia (FTD) show different patterns of cortical thickness (CTh) loss compared with healthy controls (HC), even though there is relevant heterogeneity between individuals suffering from each of these diseases. Thus, we developed CTh models to study individual variability in AD, FTD, and HC. METHODS We used the baseline CTh measures of 379 participants obtained from the structural MRI processed with FreeSurfer. A total of 169 AD patients (63 ± 9 years, 65 men), 88 FTD patients (64 ± 9 years, 43 men), and 122 HC (62 ± 10 years, 47 men) were studied. We fitted region-wise temporal models of CTh using Support Vector Regression. Then, we studied associations of individual deviations from the model with cerebrospinal fluid levels of neurofilament light chain (NfL) and 14-3-3 protein and Mini-Mental State Examination (MMSE). Furthermore, we used real longitudinal data from 144 participants to test model predictivity. RESULTS We defined CTh spatiotemporal models for each group with a reliable fit. Individual deviation correlated with MMSE for AD and with NfL for FTD. AD patients with higher deviations from the trend presented higher MMSE values. In FTD, lower NfL levels were associated with higher deviations from the CTh prediction. For AD and HC, we could predict longitudinal visits with the presented model trained with baseline data. For FTD, the longitudinal visits had more variability. CONCLUSION We highlight the value of CTh models for studying AD and FTD longitudinal changes and variability and their relationships with cognitive features and biomarkers.
Collapse
Affiliation(s)
- Agnès Pérez-Millan
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
- Institut de Neurociències, University of Barcelona, 08036, Barcelona, Spain
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, 08036, Barcelona, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
- Atlantic Fellow for Equity in Brain Health, Global Brain Health Institute, University of California San Francisco, San Francisco, 94143, USA
| | - Jordi Juncà-Parella
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
| | - Adrià Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
| | - Nuria Bargalló
- Image Diagnostic Centre, CIBER de Salud Mental, Instituto de Salud Carlos III, Magnetic Resonance Image Core Facility, IDIBAPS, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lorena Rami
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
- Atlantic Fellow for Equity in Brain Health, Global Brain Health Institute, University of California San Francisco, San Francisco, 94143, USA
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain
- Institut de Neurociències, University of Barcelona, 08036, Barcelona, Spain
| | - Roser Sala-Llonch
- Institut de Neurociències, University of Barcelona, 08036, Barcelona, Spain
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 08036, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit. Service of Neurology, Hospital Clínic de Barcelona. Fundació Recerca Clínic Barcelona-IDIBAPS, Villarroel, 170, 08036, Barcelona, Spain.
- Institut de Neurociències, University of Barcelona, 08036, Barcelona, Spain.
- Departament de Medicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036, Barcelona, Spain.
| |
Collapse
|
14
|
Harun-Or-Roshid M, Maeda K, Phan LT, Manavalan B, Kurata H. Stack-DHUpred: Advancing the accuracy of dihydrouridine modification sites detection via stacking approach. Comput Biol Med 2024; 169:107848. [PMID: 38145601 DOI: 10.1016/j.compbiomed.2023.107848] [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: 08/26/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 12/27/2023]
Abstract
Dihydrouridine (DHU, D) is one of the most abundant post-transcriptional uridine modifications found in tRNA, mRNA, and snoRNA, closely associated with disease pathogenesis and various biological processes in eukaryotes. Identifying D sites is important for understanding the modification mechanisms and/or epigenetic regulation. However, biological experiments for detecting D sites are time-consuming and expensive. Given these challenges, computational methods have been developed for accurately identifying the D sites in genome-wide datasets. However, existing methods have some limitations, and their prediction performance needs to be improved. In this work, we have developed a new computational predictor for accurately identifying D sites called Stack-DHUpred. Briefly, we trained 66 baseline models or single-feature models by connecting six machine learning classifiers with eleven different feature encoding methods and stacked different baseline models to build stacked ensemble learning models. Subsequently, the optimal combination of the baseline models was identified for the construction of the final stacked model. Remarkably, the Stack-DHUpred outperformed the existing predictors on our new independent dataset, indicating that the stacking approach significantly improved the prediction performance. We have made Stack-DHUpred available to the public through a web server (http://kurata35.bio.kyutech.ac.jp/Stack-DHUpred) and a standalone program (https://github.com/kuratahiroyuki/Stack-DHUpred). We believe that Stack-DHUpred will be a valuable tool for accelerating the discovery of D modifications and understanding their role in post-transcriptional regulation.
Collapse
Affiliation(s)
- Md Harun-Or-Roshid
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Kazuhiro Maeda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan
| | - Le Thi Phan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Balachandran Manavalan
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Hiroyuki Kurata
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka, Fukuoka 820-8502, Japan.
| |
Collapse
|
15
|
Hu YH, Su T, Wu L, Wu JF, Liu D, Zhu LQ, Yuan M. Deregulation of the Glymphatic System in Alzheimer's Disease: Genetic and Non-Genetic Factors. Aging Dis 2024; 16:AD.2023.1229. [PMID: 38270115 PMCID: PMC11745449 DOI: 10.14336/ad.2023.1229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/29/2023] [Indexed: 01/26/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia and is characterized by progressive degeneration of brain function. AD gradually affects the parts of the brain that control thoughts, language, behavior and mental function, severely impacting a person's ability to carry out daily activities and ultimately leading to death. The accumulation of extracellular amyloid-β peptide (Aβ) and the aggregation of intracellular hyperphosphorylated tau are the two key pathological hallmarks of AD. AD is a complex condition that involves both non-genetic risk factors (35%) and genetic risk factors (58-79%). The glymphatic system plays an essential role in clearing metabolic waste, transporting tissue fluid, and participating in the immune response. Both non-genetic and genetic risk factors affect the glymphatic system to varying degrees. The main purpose of this review is to summarize the underlying mechanisms involved in the deregulation of the glymphatic system during the progression of AD, especially concerning the diverse contributions of non-genetic and genetic risk factors. In the future, new targets and interventions that modulate these interrelated mechanisms will be beneficial for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yan-Hong Hu
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Ting Su
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Lin Wu
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Jun-Fang Wu
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Dan Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Mei Yuan
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
16
|
Llibre-Guerra JJ, Iaccarino L, Coble D, Edwards L, Li Y, McDade E, Strom A, Gordon B, Mundada N, Schindler SE, Tsoy E, Ma Y, Lu R, Fagan AM, Benzinger TLS, Soleimani-Meigooni D, Aschenbrenner AJ, Miller Z, Wang G, Kramer JH, Hassenstab J, Rosen HJ, Morris JC, Miller BL, Xiong C, Perrin RJ, Allegri R, Chrem P, Surace E, Berman SB, Chhatwal J, Masters CL, Farlow MR, Jucker M, Levin J, Fox NC, Day G, Gorno-Tempini ML, Boxer AL, La Joie R, Rabinovici GD, Bateman R. Longitudinal clinical, cognitive and biomarker profiles in dominantly inherited versus sporadic early-onset Alzheimer's disease. Brain Commun 2023; 5:fcad280. [PMID: 37942088 PMCID: PMC10629466 DOI: 10.1093/braincomms/fcad280] [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: 06/01/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 11/10/2023] Open
Abstract
Approximately 5% of Alzheimer's disease cases have an early age at onset (<65 years), with 5-10% of these cases attributed to dominantly inherited mutations and the remainder considered as sporadic. The extent to which dominantly inherited and sporadic early-onset Alzheimer's disease overlap is unknown. In this study, we explored the clinical, cognitive and biomarker profiles of early-onset Alzheimer's disease, focusing on commonalities and distinctions between dominantly inherited and sporadic cases. Our analysis included 117 participants with dominantly inherited Alzheimer's disease enrolled in the Dominantly Inherited Alzheimer Network and 118 individuals with sporadic early-onset Alzheimer's disease enrolled at the University of California San Francisco Alzheimer's Disease Research Center. Baseline differences in clinical and biomarker profiles between both groups were compared using t-tests. Differences in the rates of decline were compared using linear mixed-effects models. Individuals with dominantly inherited Alzheimer's disease exhibited an earlier age-at-symptom onset compared with the sporadic group [43.4 (SD ± 8.5) years versus 54.8 (SD ± 5.0) years, respectively, P < 0.001]. Sporadic cases showed a higher frequency of atypical clinical presentations relative to dominantly inherited (56.8% versus 8.5%, respectively) and a higher frequency of APOE-ε4 (50.0% versus 28.2%, P = 0.001). Compared with sporadic early onset, motor manifestations were higher in the dominantly inherited cohort [32.5% versus 16.9% at baseline (P = 0.006) and 46.1% versus 25.4% at last visit (P = 0.001)]. At baseline, the sporadic early-onset group performed worse on category fluency (P < 0.001), Trail Making Test Part B (P < 0.001) and digit span (P < 0.001). Longitudinally, both groups demonstrated similar rates of cognitive and functional decline in the early stages. After 10 years from symptom onset, dominantly inherited participants experienced a greater decline as measured by Clinical Dementia Rating Sum of Boxes [3.63 versus 1.82 points (P = 0.035)]. CSF amyloid beta-42 levels were comparable [244 (SD ± 39.3) pg/ml dominantly inherited versus 296 (SD ± 24.8) pg/ml sporadic early onset, P = 0.06]. CSF phosphorylated tau at threonine 181 levels were higher in the dominantly inherited Alzheimer's disease cohort (87.3 versus 59.7 pg/ml, P = 0.005), but no significant differences were found for t-tau levels (P = 0.35). In summary, sporadic and inherited Alzheimer's disease differed in baseline profiles; sporadic early onset is best distinguished from dominantly inherited by later age at onset, high frequency of atypical clinical presentations and worse executive performance at baseline. Despite these differences, shared pathways in longitudinal clinical decline and CSF biomarkers suggest potential common therapeutic targets for both populations, offering valuable insights for future research and clinical trial design.
Collapse
Affiliation(s)
| | - Leonardo Iaccarino
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dean Coble
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63108, USA
| | - Lauren Edwards
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yan Li
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63108, USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Amelia Strom
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Brian Gordon
- Malinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Nidhi Mundada
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Elena Tsoy
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yinjiao Ma
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63108, USA
| | - Ruijin Lu
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63108, USA
| | - Anne M Fagan
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Tammie L S Benzinger
- Malinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO 63108, USA
| | - David Soleimani-Meigooni
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Zachary Miller
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Guoqiao Wang
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63108, USA
| | - Joel H Kramer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Howard J Rosen
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - John C Morris
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| | - Bruce L Miller
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chengjie Xiong
- Division of Biostatistics, Washington University in St Louis, St Louis, MO 63108, USA
| | - Richard J Perrin
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
- Department of Pathology and Immunology, Washington University in St Louis, St. Louis, MO 63108, USA
| | - Ricardo Allegri
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Patricio Chrem
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Ezequiel Surace
- Department of Cognitive Neurology, Institute for Neurological Research Fleni, Buenos Aires, Argentina
| | - Sarah B Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jasmeer Chhatwal
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Colin L Masters
- Florey Institute, The University of Melbourne, Melbourne 3052, Australia
| | - Martin R Farlow
- Neuroscience Center, Indiana University School of Medicine at Indianapolis, IN 46202, USA
| | - Mathias Jucker
- DZNE-German Center for Neurodegenerative Diseases, Tübingen 72076, Germany
- Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich 80539, Germany
- German Center for Neurodegenerative Diseases, Munich 81377, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich 81377, Germany
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, UK
| | - Gregory Day
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL 33224, USA
| | - Maria Luisa Gorno-Tempini
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Adam L Boxer
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Renaud La Joie
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Gil D Rabinovici
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Randall Bateman
- Department of Neurology, Washington University in St Louis, St Louis, MO 63108, USA
| |
Collapse
|
17
|
Bhatti JS, Kaur S, Mishra J, Dibbanti H, Singh A, Reddy AP, Bhatti GK, Reddy PH. Targeting dynamin-related protein-1 as a potential therapeutic approach for mitochondrial dysfunction in Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166798. [PMID: 37392948 DOI: 10.1016/j.bbadis.2023.166798] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that manifests its pathology through synaptic damage, mitochondrial abnormalities, microRNA deregulation, hormonal imbalance, increased astrocytes & microglia, accumulation of amyloid β (Aβ) and phosphorylated Tau in the brains of AD patients. Despite extensive research, the effective treatment of AD is still unknown. Tau hyperphosphorylation and mitochondrial abnormalities are involved in the loss of synapses, defective axonal transport and cognitive decline in patients with AD. Mitochondrial dysfunction is evidenced by enhanced mitochondrial fragmentation, impaired mitochondrial dynamics, mitochondrial biogenesis and defective mitophagy in AD. Hence, targeting mitochondrial proteins might be a promising therapeutic strategy in treating AD. Recently, dynamin-related protein 1 (Drp1), a mitochondrial fission protein, has gained attention due to its interactions with Aβ and hyperphosphorylated Tau, altering mitochondrial morphology, dynamics, and bioenergetics. These interactions affect ATP production in mitochondria. A reduction in Drp1 GTPase activity protects against neurodegeneration in AD models. This article provides a comprehensive overview of Drp1's involvement in oxidative damage, apoptosis, mitophagy, and axonal transport of mitochondria. We also highlighted the interaction of Drp1 with Aβ and Tau, which may contribute to AD progression. In conclusion, targeting Drp1 could be a potential therapeutic approach for preventing AD pathology.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India.
| | - Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Jayapriya Mishra
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | | | - Arti Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India.
| | - P Hemachandra Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA; Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, 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 Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
18
|
Tureson KN, Beam CR, Medina LD, Segal-Gidan F, D'Orazio LM, Chui H, Torres M, Varma R, Ringman JM. Use of the Spanish English Neuropsychological Assessment Scale in older adult Latines and those at risk for autosomal dominant Alzheimer's disease. J Clin Exp Neuropsychol 2023; 45:553-569. [PMID: 37990912 PMCID: PMC10926998 DOI: 10.1080/13803395.2023.2284971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 11/12/2023] [Indexed: 11/23/2023]
Abstract
OBJECTIVE The Spanish English Neuropsychological Assessment Scale (SENAS) is a cognitive battery with English and Spanish versions for use with persons for whom either language is predominant. Few studies have examined its utility outside the normative sample. The current study examined SENAS performance in samples of older adult Latines and Latines with or at risk for autosomal dominant Alzheimer's disease (ADAD) mutations. METHOD The SENAS was administered to 202 older adults from the Los Angeles Latino Eye Study (LALES) and 29 adults with (carriers) or without (non-carriers) mutations causing ADAD. We examined associations between SENAS, age, education, and language (LALES) and between SENAS, estimated years from familial age of dementia diagnosis, education, language, and acculturation (ADAD). Partial correlations were used to examine differences in correlational strength between estimated years from familial age of dementia diagnosis and SENAS scores among ADAD carriers compared to chronological age and SENAS in the LALES sample. Exploratory t-tests were performed to examine SENAS performance differences between ADAD carriers and non-carriers. RESULTS In an older adult sample (LALES), increased age correlated with worse verbal delayed recall; English fluency and higher education correlated with better naming and visuospatial subtest performance. Among ADAD carriers, verbal and nonverbal delayed recall and object naming subtest performance worsened as they approached their familial age of dementia diagnosis. English fluency and higher U.S.-acculturation were related to better SENAS performance among carriers and non-carriers. Tests of verbal delayed recall and object naming best distinguished ADAD carriers from their familial non-carrier counterparts. CONCLUSIONS Verbal delayed recall and object naming measures appear to be most sensitive to age-related changes in older adult samples and mutation-related changes in distinguishing ADAD carriers from non-carriers. Future research should examine the sensitivity of SENAS in other samples, such as larger samples of symptomatic ADAD carriers and other AD subtypes.
Collapse
Affiliation(s)
- Kayla N Tureson
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Christopher R Beam
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
- USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Luis D Medina
- Department of Psychology, University of Houston, Houston, TX, USA
| | - Freddi Segal-Gidan
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Lina M D'Orazio
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Helena Chui
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Mina Torres
- CHA Hollywood Presbyterian Medical Center, Southern California Eye Institute, Los Angeles, CA, USA
| | - Rohit Varma
- CHA Hollywood Presbyterian Medical Center, Southern California Eye Institute, Los Angeles, CA, USA
| | - John M Ringman
- Alzheimer's Disease Research Center, Department of Neurology, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
19
|
Noel RL, Gorman SL, Batts AJ, Konofagou EE. Getting ahead of Alzheimer's disease: early intervention with focused ultrasound. Front Neurosci 2023; 17:1229683. [PMID: 37575309 PMCID: PMC10412991 DOI: 10.3389/fnins.2023.1229683] [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: 05/26/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
The amyloid-β (Aβ) hypothesis implicates Aβ protein accumulation in Alzheimer's disease (AD) onset and progression. However, therapies targeting Aβ have proven insufficient in achieving disease reversal, prompting a shift to focus on early intervention and alternative therapeutic targets. Focused ultrasound (FUS) paired with systemically-introduced microbubbles (μB) is a non-invasive technique for targeted and transient blood-brain barrier opening (BBBO), which has demonstrated Aβ and tau reduction, as well as memory improvement in models of late-stage AD. However, similar to drug treatments for AD, this approach is not sufficient for complete reversal of advanced, symptomatic AD. Here we aim to determine whether early intervention with FUS-BBBO in asymptomatic AD could delay disease onset. Thus, the objective of this study is to measure the protective effects of FUS-BBBO on anxiety, memory and AD-associated protein levels in female and male triple transgenic (3xTg) AD mice treated at an early age and disease state. Here we show that early, repeated intervention with FUS-BBBO decreased anxiety-associated behaviors in the open field test by 463.02 and 37.42% in male and female cohorts, respectively. FUS-BBBO preserved female aptitude for learning in the active place avoidance paradigm, reducing the shock quadrant time by 30.03 and 31.01% in the final long-term and reversal learning trials, respectively. Finally, FUS-BBBO reduced hippocampal accumulation of Aβ40, Aβ42, and total tau in females by 12.54, 13.05, and 3.57%, respectively, and reduced total tau in males by 18.98%. This demonstration of both cognitive and pathological protection could offer a solution for carriers of AD-associated mutations as a safe, non-invasive technique to delay the onset of the cognitive and pathological effects of AD.
Collapse
Affiliation(s)
- Rebecca L. Noel
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Samantha L. Gorman
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Alec J. Batts
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Elisa E. Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
- Department of Radiology, Columbia University, New York, NY, United States
| |
Collapse
|
20
|
Taléns-Visconti R, de Julián-Ortiz JV, Vila-Busó O, Diez-Sales O, Nácher A. Intranasal Drug Administration in Alzheimer-Type Dementia: Towards Clinical Applications. Pharmaceutics 2023; 15:pharmaceutics15051399. [PMID: 37242641 DOI: 10.3390/pharmaceutics15051399] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer-type dementia (ATD) treatments face limitations in crossing the blood-brain barrier and systemic adverse effects. Intranasal administration offers a direct route to the brain via the nasal cavity's olfactory and trigeminal pathways. However, nasal physiology can hinder drug absorption and limit bioavailability. Therefore, the physicochemical characteristics of formulations must be optimized by means of technological strategies. Among the strategies that have been explored, lipid-based nanosystems, particularly nanostructured lipid carriers, are promising in preclinical investigations with minimal toxicity and therapeutic efficacy due to their ability to overcome challenges associated with other nanocarriers. We review the studies of nanostructured lipid carriers for intranasal administration in the treatment of ATD. Currently, no drugs for intranasal administration in ATD have marketing approval, with only three candidates, insulin, rivastigmine and APH-1105, being clinically investigated. Further studies with different candidates will eventually confirm the potential of the intranasal route of administration in the treatment of ATD.
Collapse
Affiliation(s)
- Raquel Taléns-Visconti
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain
| | - Jesus Vicente de Julián-Ortiz
- Molecular Topology and Drug Design Research Unit, Department of Physical Chemistry, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain
| | - Ofelia Vila-Busó
- Colloids Research Unit, Department of Physical Chemistry, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain
| | - Octavio Diez-Sales
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Av. Vicent Andrés Estellés s/n, Burjassot, 46100 Valencia, Spain
| | - Amparo Nácher
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Av. Vicent Andrés Estellés s/n, Burjassot, 46100 Valencia, Spain
| |
Collapse
|
21
|
Edahiro A, Okamura T, Arai T, Ikeuchi T, Ikeda M, Utsumi K, Ota H, Kakuma T, Kawakatsu S, Konagaya Y, Suzuki K, Tanimukai S, Miyanaga K, Awata S. Initial symptoms of early-onset dementia in Japan: nationwide survey. Psychogeriatrics 2023; 23:422-433. [PMID: 36814116 DOI: 10.1111/psyg.12949] [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: 11/11/2022] [Revised: 01/24/2023] [Accepted: 02/06/2023] [Indexed: 02/24/2023]
Abstract
AIM The aim of this study was to investigate initial symptoms of early-onset dementia (EOD) for each dementia subtype. METHOD We conducted a nationwide, population-based EOD prevalence study in Japan. Data were collected through service providers for people with EOD. Initial symptoms were assessed in six domains: loss of memory, difficulty in word generation, irritability, loss of motivation, increased mistakes in the workplace or domestically, and unusual behaviours or attitudes other than those listed. RESULTS Participants were 770 people with EOD. Characteristic initial symptoms were observed for each EOD subtype. Loss of memory was more common in early-onset Alzheimer's disease (75.7%, P < 0.001), difficulty in word generation was more common in early-onset vascular dementia (41.3%, P < 0.001), and loss of motivation, increased mistakes in the workplace or domestically, and unusual behaviours or attitudes other than those listed were more common in early-onset frontotemporal dementia (34.9%, P < 0.001; 49.4%, P < 0.001; 34.9%, P < 0.001, respectively). In addition, we observed gender differences whereby loss of memory was more common among women and irritability was more common among men. More than half of the participants were employed at symptom onset, and 57.2% of those who were employed at the onset had initial symptoms of increased mistakes in the workplace or domestically. CONCLUSION This report reveals differences in the frequency of initial symptoms by EOD subtype. The results contribute to increasing public awareness of the initial symptoms of EOD, which will facilitate early diagnosis and social support.
Collapse
Affiliation(s)
- Ayako Edahiro
- Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | | | - Tetsuaki Arai
- Department of Psychiatry, Division of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kumiko Utsumi
- Department of Psychiatry, Sunagawa City Medical Centre, Sunagawa, Japan
| | - Hidetaka Ota
- Advanced Research Centre for Geriatric and Gerontology, Akita University, Akita, Japan
| | - Tatsuyuki Kakuma
- Biostatistics Centre, Kurume University School of Medicine, Kurume, Japan
| | - Shinobu Kawakatsu
- Department of Neuropsychiatry, Aizu Medical Centre, Fukushima Medical University, Aizu, Japan
| | | | - Kyoko Suzuki
- Department of Behavioural Neurology and Cognitive Neuroscience, Tohoku University School of Medicine, Sendai, Japan
| | - Satoshi Tanimukai
- Department of Comprehensive Community Care for Elderly, Nursing and Health Science, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Kazuo Miyanaga
- Yukiguni-Yamato Dementia Care Centre, Yukiguni-Yamato Hospital, Niigata, Japan
| | - Shuichi Awata
- Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
22
|
Heikkinen AL, Paajanen TI, Hublin C, Valtonen T, Krüger J, Tikkanen V, Saari T, Koivisto AM, Hänninen T, Remes AM. The Cognitive Function at Work Questionnaire in memory clinic setting: a validation study. J Clin Exp Neuropsychol 2023; 45:365-376. [PMID: 37561064 DOI: 10.1080/13803395.2023.2239508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/14/2023] [Indexed: 08/11/2023]
Abstract
INTRODUCTION As there is a trend toward more people seeking medical help due to cognitive symptoms, validated and targeted questionnaires are increasingly important in the clinical evaluation process. The Cognitive Function at Work Questionnaire (CFWQ) was developed to identify and rate subjective cognitive symptoms of individuals active in working life. However, its psychometric characteristics have not been previously studied in a memory clinic setting. METHOD The factorial structure, internal consistency, test-retest reliability, and convergent validity of the CFWQ were studied in a memory clinic setting (N = 113). We also investigated the instrument's ability to identify cognitive symptoms in a cohort of early-onset dementia (EOD, N = 22), mild cognitive impairment-neurological (MCI-n, N = 18), MCI due to mood, sleep, or other physical health problems (MCI-o, N = 59), and subjective cognitive decline (SCD, N = 14) patients. RESULTS Based on factor analysis, eight cognitive subscales were identified covering main cognitive domains: Memory, Language, Executive Function, Speed of Processing, Cognitive Control, Name Memory, Visuospatial/Praxis and Attention. The internal consistency (α = .93) and the test-retest reliability (ICC = .91) were high. Several correlations (r = .19 - .33, p < .05) were documented between neuropsychological impairment level and CFWQ scores. EOD, MCI-n, MCI-o, and SCD groups did not differ statistically significantly in the levels of cognitive symptoms as measured by the CFWQ Total score. EOD group scored higher (p = .009) than other patient groups on the Visuospatial/Praxis subscale, but the difference between EOD and MCI-o groups turned insignificant after correcting for multiple testing. CONCLUSIONS The results of the study support the validity and reliability characteristics of the CFWQ in a memory clinic setting. The instrument is easy-to-use and has clinical utility in capturing the subjective cognitive symptoms of patients active in working life and who need a referral to a more detailed evaluation.
Collapse
Affiliation(s)
- Anna-Leena Heikkinen
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
- Work Ability and Working Careers, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Teemu I Paajanen
- Work Ability and Working Careers, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Christer Hublin
- Work Ability and Working Careers, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Teppo Valtonen
- Work Ability and Working Careers, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Johanna Krüger
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
| | - Veera Tikkanen
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
| | - Toni Saari
- Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anne M Koivisto
- Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Geriatrics, Helsinki University Hospital, Helsinki, Finland
- Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Tuomo Hänninen
- Neurocenter, Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Anne M Remes
- Research Unit of Clinical Medicine, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
- Neurocenter, Neurology, Oulu University Hospital, Oulu, Finland
- Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Lei D, Mao C, Li J, Huang X, Sha L, Liu C, Dong L, Xu Q, Gao J. CSF biomarkers for early-onset Alzheimer's disease in Chinese population from PUMCH dementia cohort. Front Neurol 2023; 13:1030019. [PMID: 36698871 PMCID: PMC9868908 DOI: 10.3389/fneur.2022.1030019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Alzheimer's disease (AD) is one of the highly concerned degenerative disorders in recent decades. Though vast amount of researches has been done in various aspects, early-onset subtype, however, needs more investigation in diagnosis for its atypical manifestations and progression process. Fundamental CSF biomarkers of early-onset AD are explored in PUMCH dementia cohort to depict its laboratory characteristics. Materials and methods A total of 125 individuals (age of onset <65 years old) from PUMCH dementia cohort were recruited consecutively and classified into AD, non-AD dementia, and control groups. Levels of amyloid-β 42 (Aβ42), total tau (t-tau) and phosphorylated tau (p-tau) were measured using ELISA INNOTEST (Fujirebio, Ghent, Belgium). Students' t-test or non-parametric test are used to evaluate the differences between groups. Area under curve (AUC) of receiver operating characteristic (ROC) curve was introduced to prove the diagnostic powers of corresponding markers. Logistic regression is used to establish diagnostic model to combine several markers together to promote the diagnostic power. Results The average of all three biomarkers and two calculated ratios (t-tau/Aβ42, p-tau/Aβ42) were statistically different in the AD group compared with the other two groups (Ps < 0.01). From our data, we were able to provide cutoff values (Aβ42 < 570.9 pg/mL; p-tau > 56.49 pg/mL; t-tau > 241.6 pg/mL; t-tau/Aβ42 > 0.529; p-tau/Aβ42 > 0.0846) with acceptable diagnostic accuracy compared to other studies. Using a combination of biomarkers and logistic regression (area under curve 0.951), we were able to further improve diagnostic efficacy. Discussion Our study supports the diagnostic usefulness of biomarkers and defined cutoff values to diagnose early-onset AD. We showed that the ratios of t-tau/Aβ42 and p-tau/Aβ42 are more sensitive than relying on Aβ42 levels alone, and that we can further improve diagnostic accuracy by combining biomarkers.
Collapse
Affiliation(s)
- Dan Lei
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Chenhui Mao
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jie Li
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xinying Huang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Longze Sha
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Caiyan Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Liling Dong
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Gao
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China,*Correspondence: Jing Gao ✉
| |
Collapse
|
24
|
Maurya R, Bhattacharjee G, Khambhati K, Gohil N, Singh P, Mani I, Chu DT, Ramakrishna S, Show PL, Singh V. Amyloid precursor protein in Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:261-270. [PMID: 36813361 DOI: 10.1016/bs.pmbts.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Amyloid precursor protein (APP) is a membrane protein expressed in several tissues. The occurrence of APP is predominant in synapses of nerve cells. It acts as a cell surface receptor and plays a vital role as a regulator of synapse formation, iron export and neural plasticity. It is encoded by the APP gene that is regulated by substrate presentation. APP is a precursor protein activated by proteolytic cleavage and thereby generating amyloid beta (Aβ) peptides which eventually form amyloid plaques that accumulate in Alzheimer's disease patients' brains. In this chapter, we highlight basic mechanism, structure, expression patterns and cleavage of amyloid plaques, and its diagnosis and potential treatment for Alzheimer's disease.
Collapse
Affiliation(s)
- Rupesh Maurya
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Gargi Bhattacharjee
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Khushal Khambhati
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Nisarg Gohil
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Priyanka Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India
| | - Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Suresh Ramakrishna
- College of Medicine, Hanyang University, Seoul, South Korea; Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Pau-Loke Show
- Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India.
| |
Collapse
|
25
|
Suleman MT, Alturise F, Alkhalifah T, Khan YD. iDHU-Ensem: Identification of dihydrouridine sites through ensemble learning models. Digit Health 2023; 9:20552076231165963. [PMID: 37009307 PMCID: PMC10064468 DOI: 10.1177/20552076231165963] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/09/2023] [Indexed: 04/04/2023] Open
Abstract
Background Dihydrouridine (D) is one of the most significant uridine modifications that have a prominent occurrence in eukaryotes. The folding and conformational flexibility of transfer RNA (tRNA) can be attained through this modification. Objective The modification also triggers lung cancer in humans. The identification of D sites was carried out through conventional laboratory methods; however, those were costly and time-consuming. The readiness of RNA sequences helps in the identification of D sites through computationally intelligent models. However, the most challenging part is turning these biological sequences into distinct vectors. Methods The current research proposed novel feature extraction mechanisms and the identification of D sites in tRNA sequences using ensemble models. The ensemble models were then subjected to evaluation using k-fold cross-validation and independent testing. Results The results revealed that the stacking ensemble model outperformed all the ensemble models by revealing 0.98 accuracy, 0.98 specificity, 0.97 sensitivity, and 0.92 Matthews Correlation Coefficient. The proposed model, iDHU-Ensem, was also compared with pre-existing predictors using an independent test. The accuracy scores have shown that the proposed model in this research study performed better than the available predictors. Conclusion The current research contributed towards the enhancement of D site identification capabilities through computationally intelligent methods. A web-based server, iDHU-Ensem, was also made available for the researchers at https://taseersuleman-idhu-ensem-idhu-ensem.streamlit.app/.
Collapse
Affiliation(s)
- Muhammad Taseer Suleman
- Department of Computer Science, School of systems and technology, University of Management and Technology, Lahore, Pakistan
| | - Fahad Alturise
- Department of Computer, College of Science and Arts in Ar Rass, Qassim University, Ar Rass, Qassim, Saudi Arabia
- Fahad Alturise, Department of Computer, College of Science and Arts in Ar Rass, Qassim University, Ar Rass, Qassim, Saudi Arabia.
| | - Tamim Alkhalifah
- Department of Computer, College of Science and Arts in Ar Rass, Qassim University, Ar Rass, Qassim, Saudi Arabia
| | - Yaser Daanial Khan
- Department of Computer Science, School of systems and technology, University of Management and Technology, Lahore, Pakistan
| |
Collapse
|
26
|
Forno G, Saranathan M, Contador J, Guillen N, Falgàs N, Tort-Merino A, Balasa M, Sanchez-Valle R, Hornberger M, Lladó A. Thalamic nuclei changes in early and late onset Alzheimer's disease. CURRENT RESEARCH IN NEUROBIOLOGY 2023. [DOI: 10.1016/j.crneur.2023.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
|
27
|
Rayff da Silva P, de Andrade JC, de Sousa NF, Portela ACR, Oliveira Pires HF, Remígio MCRB, da Nóbrega Alves D, de Andrade HHN, Dias AL, Salvadori MGDSS, de Oliveira Golzio AMF, de Castro RD, Scotti MT, Felipe CFB, de Almeida RN, Scotti L. Computational Studies Applied to Linalool and Citronellal Derivatives Against Alzheimer's and Parkinson's Disorders: A Review with Experimental Approach. Curr Neuropharmacol 2023; 21:842-866. [PMID: 36809939 PMCID: PMC10227923 DOI: 10.2174/1570159x21666230221123059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 02/24/2023] Open
Abstract
Alzheimer's and Parkinson's are neurodegenerative disorders that affect a great number of people around the world, seriously compromising the quality of life of individuals, due to motor and cognitive damage. In these diseases, pharmacological treatment is used only to alleviate symptoms. This emphasizes the need to discover alternative molecules for use in prevention. Using Molecular Docking, this review aimed to evaluate the anti-Alzheimer's and anti-Parkinson's activity of linalool and citronellal, as well as their derivatives. Before performing Molecular Docking simulations, the compounds' pharmacokinetic characteristics were evaluated. For Molecular Docking, 7 chemical compounds derived from citronellal, and 10 compounds derived from linalool, and molecular targets involved in Alzheimer's and Parkinson's pathophysiology were selected. According to the Lipinski rules, the compounds under study presented good oral absorption and bioavailability. For toxicity, some tissue irritability was observed. For Parkinson-related targets, the citronellal and linalool derived compounds revealed excellent energetic affinity for α-Synuclein, Adenosine Receptors, Monoamine Oxidase (MAO), and Dopamine D1 receptor proteins. For Alzheimer disease targets, only linalool and its derivatives presented promise against BACE enzyme activity. The compounds studied presented high probability of modulatory activity against the disease targets under study, and are potential candidates for future drugs.
Collapse
Affiliation(s)
- Pablo Rayff da Silva
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Jéssica Cabral de Andrade
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Natália Ferreira de Sousa
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Anne Caroline Ribeiro Portela
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Hugo Fernandes Oliveira Pires
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Maria Caroline Rodrigues Bezerra Remígio
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Danielle da Nóbrega Alves
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Humberto Hugo Nunes de Andrade
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Arthur Lins Dias
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | | | | | - Ricardo Dias de Castro
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Marcus T. Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Cícero Francisco Bezerra Felipe
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Reinaldo Nóbrega de Almeida
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-085, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| | - Luciana Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, 58051-900, Via Ipê Amarelo, S/N, João Pessoa, Paraíba, Brazil
| |
Collapse
|
28
|
Isella V, Licciardo D, Nastasi G, Impagnatiello V, Ferri F, Mapelli C, Crivellaro C, Musarra M, Morzenti S, Appollonio I, Ferrarese C. Clinical and metabolic imaging features of late-onset and early-onset posterior cortical atrophy. Eur J Neurol 2022; 29:3147-3157. [PMID: 35950612 PMCID: PMC9804481 DOI: 10.1111/ene.15520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/12/2022] [Accepted: 08/04/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE Late-onset (LO) and early-onset (EO) dementia show neurobiological and clinical differences. Clinical and 18 fluoro-deoxy-glucose positron emission tomography (FDG-PET) features of LO and EO posterior cortical atrophy (LO_PCA, EO_PCA), the visual variant of Alzheimer's disease (AD), were compared. LO_PCA patients were also compared with a group of patients with LO typical AD (tAD). METHODS Thirty-seven LO_PCA patients (onset age ≥ 65 years), 29 EO_PCA patients and 40 tAD patients who all underwent a standard neuropsychological battery were recruited; PCA patients were also assessed for the presence of posterior signs and symptoms. Brain FDG-PET was available in 32 LO_PCA cases, 23 EO_PCA cases and all tAD cases, and their scans were compared with scans from 30 healthy elderly controls. Within the entire PCA sample FDG uptake was also correlated with age at onset as a continuous variable. RESULTS The main difference between the two PCA groups was a higher prevalence of Bálint-Holmes symptoms in EO cases, which was associated with the presence of severe bilateral occipito-temporo-parietal hypometabolism, whilst LO_PCA patients showed reduction of FDG uptake mainly in the right posterior regions. The latter group also showed mesial temporal hypometabolism, similarly to the tAD group, although with a right rather than left lateralization. Correlation analysis confirmed the association between older age and decreased limbic metabolism and between younger age and decreased left parietal metabolism. CONCLUSIONS The major involvement of the temporal cortex in LO cases and of the parietal cortex in EO cases reported previously within the AD spectrum holds true also for the visual variant of AD.
Collapse
Affiliation(s)
- Valeria Isella
- Neurology, School of MedicineUniversity of Milano—BicoccaMonzaItaly
- Milan Center for NeurosciencesNeuroMIMilanItaly
| | - Daniele Licciardo
- Milan Center for NeurosciencesNeuroMIMilanItaly
- Neurology DepartmentSan Gerardo HospitalMonzaLombardiaItaly
| | - Giulia Nastasi
- Neurology DepartmentASST of VimercateVimercateLombardiaItaly
| | - Valentina Impagnatiello
- Milan Center for NeurosciencesNeuroMIMilanItaly
- Neurology DepartmentSan Gerardo HospitalMonzaLombardiaItaly
| | - Francesca Ferri
- Milan Center for NeurosciencesNeuroMIMilanItaly
- Neurology DepartmentSan Gerardo HospitalMonzaLombardiaItaly
| | | | | | - Monica Musarra
- Nuclear Medicine UnitSan Gerardo HospitalMonzaLombardiaItaly
| | | | - Ildebrando Appollonio
- Neurology, School of MedicineUniversity of Milano—BicoccaMonzaItaly
- Milan Center for NeurosciencesNeuroMIMilanItaly
- Neurology DepartmentSan Gerardo HospitalMonzaLombardiaItaly
| | - Carlo Ferrarese
- Neurology, School of MedicineUniversity of Milano—BicoccaMonzaItaly
- Milan Center for NeurosciencesNeuroMIMilanItaly
- Neurology DepartmentSan Gerardo HospitalMonzaLombardiaItaly
| |
Collapse
|
29
|
Shade RD, Ross JA, Van Bockstaele EJ. Targeting the cannabinoid system to counteract the deleterious effects of stress in Alzheimer’s disease. Front Aging Neurosci 2022; 14:949361. [PMID: 36268196 PMCID: PMC9577232 DOI: 10.3389/fnagi.2022.949361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder characterized histologically in postmortem human brains by the presence of dense protein accumulations known as amyloid plaques and tau tangles. Plaques and tangles develop over decades of aberrant protein processing, post-translational modification, and misfolding throughout an individual’s lifetime. We present a foundation of evidence from the literature that suggests chronic stress is associated with increased disease severity in Alzheimer’s patient populations. Taken together with preclinical evidence that chronic stress signaling can precipitate cellular distress, we argue that chronic psychological stress renders select circuits more vulnerable to amyloid- and tau- related abnormalities. We discuss the ongoing investigation of systemic and cellular processes that maintain the integrity of protein homeostasis in health and in degenerative conditions such as Alzheimer’s disease that have revealed multiple potential therapeutic avenues. For example, the endogenous cannabinoid system traverses the central and peripheral neural systems while simultaneously exerting anti-inflammatory influence over the immune response in the brain and throughout the body. Moreover, the cannabinoid system converges on several stress-integrative neuronal circuits and critical regions of the hypothalamic-pituitary-adrenal axis, with the capacity to dampen responses to psychological and cellular stress. Targeting the cannabinoid system by influencing endogenous processes or exogenously stimulating cannabinoid receptors with natural or synthetic cannabis compounds has been identified as a promising route for Alzheimer’s Disease intervention. We build on our foundational framework focusing on the significance of chronic psychological and cellular stress on the development of Alzheimer’s neuropathology by integrating literature on cannabinoid function and dysfunction within Alzheimer’s Disease and conclude with remarks on optimal strategies for treatment potential.
Collapse
Affiliation(s)
- Ronnie D. Shade
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Jennifer A. Ross
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA, United States
- *Correspondence: Jennifer A. Ross,
| | - Elisabeth J. Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
30
|
Contador J, Pérez-Millan A, Guillen N, Sarto J, Tort-Merino A, Balasa M, Falgàs N, Castellví M, Borrego-Écija S, Juncà-Parella J, Bosch B, Fernández-Villullas G, Ramos-Campoy O, Antonell A, Bargalló N, Sanchez-Valle R, Sala Llonch R, Lladó A. Sex differences in early-onset Alzheimer's disease. Eur J Neurol 2022; 29:3623-3632. [PMID: 36005384 DOI: 10.1111/ene.15531] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Sex is believed to drive heterogeneity in Alzheimer's disease (AD), although evidence in early-onset AD (<65 years, EOAD) is scarce. METHODS We included 62 EOAD patients and 44 healthy controls (HC) with cerebrospinal fluid (CSF) AD's core biomarkers and neurofilament light chain levels, neuropsychological assessment, and 3T-MRI. We measured cortical thickness (CTh) and hippocampal subfield volumes (HpS) using Freesurfer. Adjusted linear models were used to analyze sex-differences and the relationship between atrophy and cognition. RESULTS Compared to same-sex HC, female-EOAD showed greater cognitive impairment and broader atrophy burden than male-EOAD. In a direct female-EOAD and male-EOAD comparison, there were slight differences in temporal CTh, with no differences in cognition or HpS. CSF tau levels were higher in female-EOAD than in male-EOAD. Greater atrophy was associated with worse cognition in female-EOAD. CONCLUSIONS At diagnosis, there are sex-differences in the pattern of cognitive impairment, atrophy burden and CSF tau in EOAD, suggesting there is an influence of sex on pathology spreading and susceptibility to the disease in EOAD.
Collapse
Affiliation(s)
- José Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Agnès Pérez-Millan
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Nuria Guillen
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Adrià Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute
| | - Magdalena Castellví
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Juncà-Parella
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Guadalupe Fernández-Villullas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Oscar Ramos-Campoy
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Nuria Bargalló
- Image Diagnostic Centre Radiology Department, Hospital Clínic de Barcelona, Magnetic Resonance Image Core facility Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM., Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Roser Sala Llonch
- Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Biomedical Imaging Group, Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Chanda K, Jana NR, Mukhopadhyay D. Long non-coding RNA MALAT1 protects against Aβ 1-42 induced toxicity by regulating the expression of receptor tyrosine kinase EPHA2 via quenching miR-200a/26a/26b in Alzheimer's disease. Life Sci 2022; 302:120652. [PMID: 35598655 DOI: 10.1016/j.lfs.2022.120652] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/04/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
Altered expressions of Receptor Tyrosine Kinases (RTK) and non-coding (nc) RNAs are known to regulate the pathophysiology of Alzheimer's disease (AD). However, specific understanding of the roles played, especially the mechanistic and functional roles, by long ncRNAs in AD is still elusive. Using mouse tissue qPCR assays we observe changes in the expression levels of 41 lncRNAs in AD mice of which only 7 genes happen to have both human orthologs and AD associations. Post validation of these 7 human lncRNA genes, MEG3 and MALAT1 shows consistent and significant decrease in AD cell, animal models and human AD brain tissues, but MALAT1 showed a more pronounced decrease. Using bioinformatics, qRT-PCR, RNA FISH and RIP techniques, we could establish MALAT1 as an interactor and regulator of miRs-200a, -26a and -26b, all of which are naturally elevated in AD. We could further show that these miRNAs target the RTK EPHA2 and several of its downstream effectors. Expectedly EPHA2 over expression protects against Aβ1-42 induced cytotoxicity. Transiently knocking down MALAT1 validates these unique regulatory facets of AD at the miRNA and protein levels. Although the idea of sponging of miRNAs by lncRNAs in other pathologies is gradually gaining credibility, this novel MALAT1- miR-200a/26a/26b - EPHA2 regulation mechanism in the context of AD pathophysiology promises to become a significant strategy in controlling the disease.
Collapse
Affiliation(s)
- Kaushik Chanda
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, HBNI, Kolkata 700 064, India; Department of Neuroscience, UF Scripps Biomedical Research, 120 Scripps Way, Jupiter, FL 33458, United States of America
| | - Nihar Ranjan Jana
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon 122 050, India
| | - Debashis Mukhopadhyay
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, HBNI, Kolkata 700 064, India.
| |
Collapse
|
32
|
Kim KW, Choi J, Chin J, Lee BH, Na DL. Eye-Tracking Metrics for Figure-Copying Processes in Early- vs. Late-Onset Alzheimer's Disease. Front Neurol 2022; 13:844341. [PMID: 35651346 PMCID: PMC9149280 DOI: 10.3389/fneur.2022.844341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/12/2022] [Indexed: 12/04/2022] Open
Abstract
Visuospatial dysfunction is a common symptom in patients with Alzheimer's disease (AD). To more focus on copying processes rather than on finally completed figures, we conceptually split the copying processes into three stages: visuoperceptual function, visuoconstructional function, and working memory function. We constructed perceptual and working spaces to investigate the different stages of copying, and then, we compared the number and duration of fixations and saccades and the number of switches across the two spaces. We used eye-tracking glasses to assess eye-tracking metrics in patients with early-onset AD (EOAD), patients with late-onset AD (LOAD), and normal control (NC) participants while they copied the simplified Rey–Osterrieth complex figure test (RCFT). Regarding eye metrics on the perceptual space, the number and duration of fixations were greater in both groups of patients with AD than in the NC participants group (number: EOAD vs. NC: p < 0.001, LOAD vs. NC: p = 0. 003/ duration: EOAD vs. NC: p < 0.001, LOAD vs. NC: p < 0.001). On the working space, the number and duration of fixations were greater in the patients with EOAD than in the patients with LOAD and NC participants (number: EOAD vs. LOAD: p = 0. 007, EOAD vs. NC: p = 0. 001/duration: EOAD vs. LOAD: p = 0. 008, EOAD vs. NC: p = 0. 002). The number of saccades and switching was higher in patients with EOAD than in NC participants (p < 0.001). The eye-tracking metrics from the simplified RCFT correlated with the neuropsychological test scores. Patients with EOAD and LOAD achieved the same level of performance at the simplified and original RCFT scores. However, patients with EOAD than LOAD showed a greater number and duration of fixations on the working space and more frequent switching between the perceptual and working spaces, which may reflect more cognitive efforts to achieve the same level of performance.
Collapse
Affiliation(s)
- Ko Woon Kim
- Department of Neurology, Jeonbuk National University Medical School and Hospital, Jeonju, South Korea.,Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, South Korea.,Biomedical Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Jongdoo Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Juhee Chin
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Byung Hwa Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Neuroscience Center, Samsung Medical Center, Seoul, South Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea.,Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
33
|
Ning P, Luo A, Mu X, Xu Y, Li T. Exploring the dual character of metformin in Alzheimer's disease. Neuropharmacology 2022; 207:108966. [PMID: 35077762 DOI: 10.1016/j.neuropharm.2022.108966] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/17/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which results in dementia typically in the elderly. The disease is mainly characterized by the deposition of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. However, only few drugs are available for AD because of its unknown pathological mechanism which limits the development of new drugs. Therefore, it is urgent to identify potential therapeutic strategies for AD. Moreover, research have showed that there is a significant association between Type 2 diabetes mellites (T2DM) and AD, suggesting that the two diseases may share common pathophysiological mechanisms. Such mechanisms include impaired insulin signaling, altered glucose metabolism, inflammation, oxidative stress, and premature aging, which strongly affect cognitive function and increased risk of dementia. Consequently, as a widely used drug for T2DM, metformin also has therapeutic potential for AD in vivo. It has been confirmed that metformin is beneficial on the brain of AD animal models. The mechanisms underlying the effects of metformin in Alzheimer's disease are complex and multifaceted. Metformin may work through mechanisms involving homeostasis of glucose metabolism, decrease of amyloid plaque deposition, normalization of tau protein phosphorylation and enhancement of autophagy. However, in clinical trials, metformin had little effects on patients with mild cognitive impairment or mild AD. Pathological effects and negative clinical results of metformin on AD make the current topic quite controversial. By reviewing the latest progress of related research, this paper summarizes the possible role of metformin in AD. The purpose of this study is not only to determine the potential treatment of AD, but also other related neurodegenerative diseases.
Collapse
Affiliation(s)
- Pingping Ning
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China.
| | - Anling Luo
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China.
| | - Xin Mu
- Department of Neurology, Chengdu First People's Hospital, 18 Wanxiang North Road, Chengdu, Sichuan Province, 610041, PR China.
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University/Air Force Medical University, No. 169 Changle West Rd, Xi'an, 710032, PR China.
| |
Collapse
|
34
|
Alegret M, Sotolongo-Grau O, de Antonio EE, Pérez-Cordón A, Orellana A, Espinosa A, Gil S, Jiménez D, Ortega G, Sanabria A, Roberto N, Hernández I, Rosende-Roca M, Tartari JP, Alarcon-Martin E, de Rojas I, Montrreal L, Morató X, Cano A, Rentz DM, Tárraga L, Ruiz A, Valero S, Marquié M, Boada M. Automatized FACEmemory® scoring is related to Alzheimer's disease phenotype and biomarkers in early-onset mild cognitive impairment: the BIOFACE cohort. Alzheimers Res Ther 2022; 14:43. [PMID: 35303916 PMCID: PMC8933921 DOI: 10.1186/s13195-022-00988-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/10/2022] [Indexed: 11/13/2022]
Abstract
Background FACEmemory® is the first computerized, self-administered verbal episodic memory test with voice recognition. It can be conducted under minimal supervision and contains an automatic scoring system to avoid administrator errors. Moreover, it is suitable for discriminating between cognitively healthy and amnestic mild cognitive impairment (MCI) individuals, and it is associated with Alzheimer’s disease (AD) cerebrospinal fluid (CSF) biomarkers. This study aimed to determine whether FACEmemory scoring is related to performance on classical memory tests and to AD biomarkers of brain magnetic resonance imaging (MRI) and CSF in patients with early-onset MCI (EOMCI). Methods Ninety-four patients with EOMCI from the BIOFACE study completed FACEmemory, classical memory tests (the Spanish version of the Word Free and Cued Selective Reminding Test -FCSRT-, the Word List from the Wechsler Memory Scale, third edition, and the Spanish version of the Rey–Osterrieth Complex Figure Test), and a brain MRI. Eighty-two individuals also underwent a lumbar puncture. Results FACEmemory scoring was moderately correlated with FCSRT scoring. With regard to neuroimaging MRI results, worse execution on FACEmemory was associated with lower cortical volume in the right prefrontal and inferior parietal areas, along with the left temporal and associative occipital areas. Moreover, the total FACEmemory score correlated with CSF AD biomarkers (Aβ1-42/Aβ1-40 ratio, p181-tau, and Aβ1-42/p181-tau ratio). When performance on FACEmemory was compared among the ATN classification groups, significant differences between the AD group and normal and SNAP groups were found. Conclusions FACEmemory is a promising tool for detecting memory deficits sensitive to early-onset AD, but it also allows the detection of memory-impaired cases due to other etiologies. Our findings suggest that FACEmemory scoring can detect the AD endophenotype and that it is also associated with AD-related changes in MRI and CSF in patients with EOMCI. The computerized FACEmemory tool might be an opportunity to facilitate early detection of MCI in younger people than 65, who have a growing interest in new technologies.
Collapse
Affiliation(s)
- Montserrat Alegret
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain. .,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Oscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ester Esteban de Antonio
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Alba Pérez-Cordón
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Espinosa
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Gil
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Jiménez
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Gemma Ortega
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Angela Sanabria
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Natalia Roberto
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Hernández
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Maitee Rosende-Roca
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Emilio Alarcon-Martin
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Xavier Morató
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain
| | - Dorene M Rentz
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Gran Via de Carles III, 85 bis, 08028, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
35
|
Gautherot M, Kuchcinski G, Bordier C, Sillaire AR, Delbeuck X, Leroy M, Leclerc X, Pruvo JP, Pasquier F, Lopes R. Longitudinal Analysis of Brain-Predicted Age in Amnestic and Non-amnestic Sporadic Early-Onset Alzheimer's Disease. Front Aging Neurosci 2021; 13:729635. [PMID: 34803654 PMCID: PMC8596466 DOI: 10.3389/fnagi.2021.729635] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/27/2021] [Indexed: 01/28/2023] Open
Abstract
Objective: Predicted age difference (PAD) is a score computed by subtracting chronological age from "brain" age, which is estimated using neuroimaging data. The goal of this study was to evaluate the PAD as a marker of phenotypic heterogeneity and severity among early-onset Alzheimer's disease (EOAD) patients. Methods: We first used 3D T1-weighted (3D-T1) magnetic resonance images (MRI) of 3,227 healthy subjects aged between 18 and 85 years to train, optimize, and evaluate the brain age model. A total of 123 participants who met the criteria for early-onset (<65 years) sporadic form of probable Alzheimer's disease (AD) and presented with two distinctive clinical presentations [an amnestic form (n = 74) and a non-amnestic form (n = 49)] were included at baseline and followed-up for a maximum period of 4 years. All the participants underwent a work-up at baseline and every year during the follow-up period, which included clinical examination, neuropsychological testing and genotyping, and structural MRI. In addition, cerebrospinal fluid biomarker assay was recorded at baseline. PAD score was calculated by applying brain age model to 3D-T1 images of the EOAD patients and healthy controls, who were matched based on age and sex. At baseline, between-group differences for neuropsychological and PAD scores were assessed using linear models. Regarding longitudinal analysis of neuropsychological and PAD scores, differences between amnestic and non-amnestic participants were analyzed using linear mixed-effects modeling. Results: PAD score was significantly higher for non-amnestic patients (2.35 ± 0.91) when compared to amnestic patients (2.09 ± 0.74) and controls (0.00 ± 1). Moreover, PAD score was linearly correlated with the Mini-Mental State Examination (MMSE) and the Clinical Dementia Rating Sum of Boxes (CDR-SB), for both amnestic and non-amnestic sporadic forms. Longitudinal analyses showed that the gradual development of the disease in patients was accompanied by a significant increase in PAD score over time, for both amnestic and non-amnestic patients. Conclusion: PAD score was able to separate amnestic and non-amnestic sporadic forms. Regardless of the clinical presentation, as PAD score was a way of quantifying an early brain age acceleration, it was an appropriate method to detect the development of AD and follow the evolution of the disease as a marker of severity as MMSE and CDR-SB.
Collapse
Affiliation(s)
- Morgan Gautherot
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
| | - Grégory Kuchcinski
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Neuroradiology Department, Lille University Medical Centre, Lille, France
| | - Cécile Bordier
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
| | - Adeline Rollin Sillaire
- Memory Center, DISTALZ, Lille, France
- Neurology Department, Lille University Medical Centre, Lille, France
| | | | - Mélanie Leroy
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Memory Center, DISTALZ, Lille, France
| | - Xavier Leclerc
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Neuroradiology Department, Lille University Medical Centre, Lille, France
| | - Jean-Pierre Pruvo
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Neuroradiology Department, Lille University Medical Centre, Lille, France
| | - Florence Pasquier
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
- Memory Center, DISTALZ, Lille, France
- Neurology Department, Lille University Medical Centre, Lille, France
| | - Renaud Lopes
- UMS 2014–US 41–PLBS–Plateformes Lilloises en Biologie & Santé, University of Lille, Lille, France
- Inserm, U1172–LilNCog–Lille Neuroscience & Cognition, University of Lille, Lille, France
| |
Collapse
|
36
|
Baseline MRI atrophy predicts 2-year cognitive outcomes in early-onset Alzheimer's disease. J Neurol 2021; 269:2573-2583. [PMID: 34665329 DOI: 10.1007/s00415-021-10851-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND MRI atrophy predicts cognitive status in AD. However, this relationship has not been investigated in early-onset AD (EOAD, < 65 years) patients with a biomarker-based diagnosis. METHODS Forty eight EOAD (MMSE ≥ 15; A + T + N +) and forty two age-matched healthy controls (HC; A - T - N -) from a prospective cohort underwent full neuropsychological assessment, 3T-MRI scan and lumbar puncture at baseline. Participants repeated the cognitive assessment annually. We used linear mixed models to investigate whether baseline cortical thickness (CTh) or subcortical volume predicts two-year cognitive outcomes in the EOAD group. RESULTS In EOAD, hemispheric CTh and ventricular volume at baseline were associated with global cognition, language and attentional/executive functioning 2 years later (p < 0.0028). Regional CTh was related to most cognitive outcomes (p < 0.0028), except verbal/visual memory subtests. Amygdalar volume was associated with letter fluency test (p < 0.0028). Hippocampal volume did not show significant associations. CONCLUSION Baseline hemispheric/regional CTh, ventricular and amygdalar volume, but not the hippocampus, predict two-year cognitive outcomes in EOAD.
Collapse
|
37
|
Monterey MD, Wei H, Wu X, Wu JQ. The Many Faces of Astrocytes in Alzheimer's Disease. Front Neurol 2021; 12:619626. [PMID: 34531807 PMCID: PMC8438135 DOI: 10.3389/fneur.2021.619626] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 07/20/2021] [Indexed: 01/11/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and is the most common cause of dementia in an aging population. The majority of research effort has focused on the role of neurons in neurodegeneration and current therapies have limited ability to slow disease progression. Recently more attention has been given to the role of astrocytes in the process of neurodegeneration. Specifically, reactive astrocytes have both advantageous and adverse effects during neurodegeneration. The ability to isolate and depict astrocyte phenotype has been challenging. However, with the recent development of single-cell sequencing technologies researchers are provided with the resource to delineate specific biomarkers associated with reactive astrocytes in AD. In this review, we will focus on the role of astrocytes in normal conditions and the pathological development of AD. We will further review recent developments in the understanding of astrocyte heterogeneity and associated biomarkers. A better understanding of astrocyte contributions and phenotypic changes in AD can ultimately lead to more effective therapeutic targets.
Collapse
Affiliation(s)
- Michael D Monterey
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Haichao Wei
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, United States
| | - Xizi Wu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, United States
| | - Jia Qian Wu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
38
|
Pollet M, Skrobala E, Lopes R, Kuchcinski G, Bordier C, Rollin-Sillaire A, Bombois S, Pasquier F, Delbeuck X. A multimodal, longitudinal study of cognitive heterogeneity in early-onset Alzheimer's disease. Eur J Neurol 2021; 28:3990-3998. [PMID: 34490682 DOI: 10.1111/ene.15097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is a heterogeneous pathology. Young patients with AD are particularly likely to have an atypical presentation. The objectives of the present cluster analysis were to determine whether patients with early-onset AD (EOAD) had several distinct cognitive profiles and to compare the resulting clusters with regard to clinical, neuroimaging, and laboratory characteristics. METHODS We collected cognitive, behavioural, functional, neuroimaging, and laboratory data on 72 patients meeting the criteria for probable mild EOAD. The patients were first classified into clinical phenotype groups by a multidisciplinary board of clinicians. The patients' cognitive and functional decline was monitored for 24 months. A k-means clustering analysis was then used to determine clusters on the basis of the patients' neuropsychological test results. RESULTS Two distinct clusters were identified: the patients in the first cluster (C1, n = 38) had a predominant memory impairment, whereas patients in the second (C2, n = 34) did not. Dyslipidaemia and the presence of ɛ4 apolipoprotein E allele were more frequent in C1, whereas the cognitive and functional decline was faster in the patients in C2. Moreover, posterior brain abnormalities were more severe in patients in C2 than in patients in C1. CONCLUSIONS By applying a k-means clustering analysis, we identified two clusters of patients in an EOAD cohort. The clusters differed with regard to certain clinical, imaging, and laboratory characteristics. This clustering procedure might be of value for managing patients with EOAD in general and for identifying those at risk of more rapid decline in particular.
Collapse
Affiliation(s)
- Marianne Pollet
- Department of Neurology, Lille University Hospital Centre, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France
| | - Emilie Skrobala
- Lille University Hospital Centre, DISTALZ, Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease, Lille, France
| | - Renaud Lopes
- University of Lille, French National Institute of Health and Medical Research U1172, Lille Neuroscience & Cognition, Degenerative and Vascular Cognitive Disorders, Lille, France.,Department of Neuroradiology, Lille University Hospital Centre, Lille, France.,University of Lille, French National Centre for Scientific Research, French National Institute of Health and Medical Research, Pasteur Institute of Lille, US41-UMS 2014 - PLBS, Lille, France
| | - Grégory Kuchcinski
- Lille University Hospital Centre, DISTALZ, Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease, Lille, France.,University of Lille, French National Institute of Health and Medical Research U1172, Lille Neuroscience & Cognition, Degenerative and Vascular Cognitive Disorders, Lille, France.,Department of Neuroradiology, Lille University Hospital Centre, Lille, France
| | - Cécile Bordier
- University of Lille, French National Institute of Health and Medical Research U1172, Lille Neuroscience & Cognition, Degenerative and Vascular Cognitive Disorders, Lille, France.,Department of Neuroradiology, Lille University Hospital Centre, Lille, France
| | - Adeline Rollin-Sillaire
- Department of Neurology, Lille University Hospital Centre, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France.,Lille University Hospital Centre, DISTALZ, Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease, Lille, France.,University of Lille, French National Institute of Health and Medical Research U1172, Lille Neuroscience & Cognition, Degenerative and Vascular Cognitive Disorders, Lille, France
| | - Stéphanie Bombois
- Department of Neurology, Lille University Hospital Centre, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France.,University of Lille, French National Institute of Health and Medical Research U1172, Lille Neuroscience & Cognition, Degenerative and Vascular Cognitive Disorders, Lille, France
| | - Florence Pasquier
- Department of Neurology, Lille University Hospital Centre, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France.,Lille University Hospital Centre, DISTALZ, Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease, Lille, France.,University of Lille, French National Institute of Health and Medical Research U1172, Lille Neuroscience & Cognition, Degenerative and Vascular Cognitive Disorders, Lille, France
| | - Xavier Delbeuck
- Department of Neurology, Lille University Hospital Centre, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France.,University of Lille, French National Institute of Health and Medical Research U1172, Lille Neuroscience & Cognition, Degenerative and Vascular Cognitive Disorders, Lille, France
| |
Collapse
|
39
|
Zhan L, Li J, Jew B, Sul JH. Rare variants in the endocytic pathway are associated with Alzheimer's disease, its related phenotypes, and functional consequences. PLoS Genet 2021; 17:e1009772. [PMID: 34516545 PMCID: PMC8460036 DOI: 10.1371/journal.pgen.1009772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/23/2021] [Accepted: 08/10/2021] [Indexed: 11/19/2022] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common type of dementia causing irreversible brain damage to the elderly and presents a major public health challenge. Clinical research and genome-wide association studies have suggested a potential contribution of the endocytic pathway to AD, with an emphasis on common loci. However, the contribution of rare variants in this pathway to AD has not been thoroughly investigated. In this study, we focused on the effect of rare variants on AD by first applying a rare-variant gene-set burden analysis using genes in the endocytic pathway on over 3,000 individuals with European ancestry from three large whole-genome sequencing (WGS) studies. We identified significant associations of rare-variant burden within the endocytic pathway with AD, which were successfully replicated in independent datasets. We further demonstrated that this endocytic rare-variant enrichment is associated with neurofibrillary tangles (NFTs) and age-related phenotypes, increasing the risk of obtaining severer brain damage, earlier age-at-onset, and earlier age-of-death. Next, by aggregating rare variants within each gene, we sought to identify single endocytic genes associated with AD and NFTs. Careful examination using NFTs revealed one significantly associated gene, ANKRD13D. To identify functional associations, we integrated bulk RNA-Seq data from over 600 brain tissues and found two endocytic expression genes (eGenes), HLA-A and SLC26A7, that displayed significant influences on their gene expressions. Differential expressions between AD patients and controls of these three identified genes were further examined by incorporating scRNA-Seq data from 48 post-mortem brain samples and demonstrated distinct expression patterns across cell types. Taken together, our results demonstrated strong rare-variant effect in the endocytic pathway on AD risk and progression and functional effect of gene expression alteration in both bulk and single-cell resolution, which may bring more insight and serve as valuable resources for future AD genetic studies, clinical research, and therapeutic targeting.
Collapse
Affiliation(s)
- Lingyu Zhan
- Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jiajin Li
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Brandon Jew
- Interdepartmental Program in Bioinformatics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Jae Hoon Sul
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
40
|
Contador J, Pérez-Millán A, Tort-Merino A, Balasa M, Falgàs N, Olives J, Castellví M, Borrego-Écija S, Bosch B, Fernández-Villullas G, Ramos-Campoy O, Antonell A, Bargalló N, Sanchez-Valle R, Sala-Llonch R, Lladó A. Longitudinal brain atrophy and CSF biomarkers in early-onset Alzheimer's disease. NEUROIMAGE-CLINICAL 2021; 32:102804. [PMID: 34474317 PMCID: PMC8405839 DOI: 10.1016/j.nicl.2021.102804] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 01/09/2023]
Abstract
There is evidence of longitudinal atrophy in posterior brain areas in early-onset Alzheimer's disease (EOAD; aged < 65 years), but no studies have been conducted in an EOAD cohort with fluid biomarkers characterization. We used 3T-MRI and Freesurfer 6.0 to investigate cortical and subcortical gray matter loss at two years in 12 EOAD patients (A + T + N + ) compared to 19 controls (A-T-N-) from the Hospital Clínic Barcelona cohort. We explored group differences in atrophy patterns and we correlated atrophy and baseline CSF-biomarkers levels in EOAD. We replicated the correlation analyses in 14 EOAD (A + T + N + ) and 55 late-onset AD (LOAD; aged ≥ 75 years; A + T + N + ) participants from the Alzheimer's disease Neuroimaging Initiative. We found that EOAD longitudinal atrophy spread with a posterior-to-anterior gradient and beyond hippocampus/amygdala. In EOAD, higher initial CSF NfL levels correlated with higher ventricular volumes at baseline. On the other hand, higher initial CSF Aβ42 levels (within pathological range) predicted higher rates of cortical loss in EOAD. In EOAD and LOAD subjects, higher CSF t-tau values at baseline predicted higher rates of subcortical atrophy. CSF p-tau did not show any significant correlation. In conclusion, posterior cortices, hippocampus and amygdala capture EOAD atrophy from early stages. CSF Aβ42 might predict cortical thinning and t-tau/NfL subcortical atrophy.
Collapse
Affiliation(s)
- José Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Agnès Pérez-Millán
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Adrià Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain; Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain; Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute; Department of Neurology, Memory & Aging Center, Weill Institute for Neurosciences, University of California, 675 Nelson Rising Lane, Suite 190, San Francisco, CA 94158, USA
| | - Jaume Olives
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Magdalena Castellví
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Guadalupe Fernández-Villullas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Oscar Ramos-Campoy
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain
| | - Nuria Bargalló
- Image Diagnostic Centre, IDIBAPS, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Spain
| | - Roser Sala-Llonch
- Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, 08036, Spain; Biomedical Imaging Group, Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona 08036, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas. CIBERNED, Spain.
| |
Collapse
|
41
|
Janoutová J, Kovalová M, Machaczka O, Ambroz P, Zatloukalová A, Němček K, Janout V. Risk Factors for Alzheimer's Disease: An Epidemiological Study. Curr Alzheimer Res 2021; 18:372-379. [PMID: 34420505 DOI: 10.2174/1567205018666210820124135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/04/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Dementia becomes a major public health challenge in both the Czech Republic and worldwide. The most common form of dementia is Alzheimer's disease (AD). OBJECTIVE We conducted two successive epidemiological projects in 2012-2015 and 2016-2019. Their aim was to study the effect of selected potential genetic, vascular and psychosocial risk factors on the development of AD by comparing their frequencies in AD patients and controls. METHODS Epidemiological case-control studies were conducted. In total, data from 2106 participants (1096 cases, 1010 controls) were analyzed. RESULTS Three times more females than males suffered from AD. The highest proportion of cases were those with primary education, unlike controls. There were statistically significantly more manual workers among cases than among controls. Of selected vascular risk factors, coronary heart disease was found to be statistically significantly more frequent in cases than in controls. The onset of hypertension and diabetes mellitus was earlier in controls than in cases. As for hobbies and interests, there were statistically significant differences in physical activity, reading and solving crosswords between the groups, with these activities being more common in controls. CONCLUSION The prevalence of chronic neurodegenerative diseases, in particular AD, is currently increasing. Given the aging of the population, these conditions may be expected to rise in prevalence. Potential risk of AD needs to be studied, analyzed and confirmed; a detailed knowledge of the risks of AD and early detection of the pathology may therefore be very beneficial for prevention and early treatment of this condition.
Collapse
Affiliation(s)
- Jana Janoutová
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Martina Kovalová
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Ondřej Machaczka
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Petr Ambroz
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Anna Zatloukalová
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Kateřina Němček
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Vladimír Janout
- Department Of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| |
Collapse
|
42
|
Esteban de Antonio E, Pérez-Cordón A, Gil S, Orellana A, Cano A, Alegret M, Espinosa A, Alarcón-Martín E, Valero S, Martínez J, de Rojas I, Sotolongo-Grau Ó, Martín E, Vivas A, Gomez-Chiari M, Tejero MÁ, Bernuz M, Tárraga L, Ruiz A, Marquié M, Boada M. BIOFACE: A Prospective Study of Risk Factors, Cognition, and Biomarkers in a Cohort of Individuals with Early-Onset Mild Cognitive Impairment. Study Rationale and Research Protocols. J Alzheimers Dis 2021; 83:1233-1249. [PMID: 34420953 PMCID: PMC8543256 DOI: 10.3233/jad-210254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background: Mild cognitive impairment (MCI) due to Alzheimer’s disease (AD) diagnosis is based on cerebrospinal fluid (CSF) or neuroimaging biomarkers. Currently, non-invasive and inexpensive blood-based biomarkers are being investigated, such as neuronal-derived plasma exosomes (NPEs). Neuroinflammation and early vascular changes have been described in AD pathogenesis and can be traced in plasma and NPEs. However, they have not been studied in early onset MCI (EOMCI). Objective: To describe the rationale, design, and baseline characteristics of the participants from the BIOFACE cohort, a two-year observational study on EOMCI conducted at Fundació ACE. The study goal is to characterize the different phenotypes from a clinical, neuropsychological, and biomarker point of view and to investigate the CSF and plasma proteomics as well as the role of NPEs as early biomarkers of AD. Methods: Participants underwent extended neurological and neuropsychological batteries, multimodal biomarkers including brain MRI, blood, saliva, CSF, anthropometric, and neuro-ophthalmological examinations. Results: Ninety-seven patients with EOMCI were recruited. 59.8%were women. Mean age at symptom onset was 57 years; mean MMSE was 28. First degree and presenile family history of dementia was present in 60.8%and 15.5%, respectively. Depressive and anxiety disorders along with vascular risk factors were the most frequent comorbidities. 29%of participants were APOE ɛ4 carriers, and 67%showed a CSF normal ATN profile. Conclusion: BIOFACE is a two-year study of clinical, cognition, and biomarkers that will shed light on the physiopathology and the potential utility of plasma and NPEs as non-invasive early diagnostic and prognostic biomarkers in people younger than 65 years.
Collapse
Affiliation(s)
- Ester Esteban de Antonio
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Alba Pérez-Cordón
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Silvia Gil
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Amanda Cano
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Montserrat Alegret
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Espinosa
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Emilio Alarcón-Martín
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Sergi Valero
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Martínez
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Óscar Sotolongo-Grau
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Elvira Martín
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Assumpta Vivas
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | - Marta Gomez-Chiari
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | | | - Mireia Bernuz
- Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lluis Tárraga
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
43
|
Movement Analysis for Neurological and Musculoskeletal Disorders Using Graph Convolutional Neural Network. FUTURE INTERNET 2021. [DOI: 10.3390/fi13080194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Using optical motion capture and wearable sensors is a common way to analyze impaired movement in individuals with neurological and musculoskeletal disorders. However, using optical motion sensors and wearable sensors is expensive and often requires highly trained professionals to identify specific impairments. In this work, we proposed a graph convolutional neural network that mimics the intuition of physical therapists to identify patient-specific impairments based on video of a patient. In addition, two modeling approaches are compared: a graph convolutional network applied solely on skeleton input data and a graph convolutional network accompanied with a 1-dimensional convolutional neural network (1D-CNN). Experiments on the dataset showed that the proposed method not only improves the correlation of the predicted gait measure with the ground truth value (speed = 0.791, gait deviation index (GDI) = 0.792) but also enables faster training with fewer parameters. In conclusion, the proposed method shows that the possibility of using video-based data to treat neurological and musculoskeletal disorders with acceptable accuracy instead of depending on the expensive and labor-intensive optical motion capture systems.
Collapse
|
44
|
Li H, Zou L, Shi J, Han X. Bioinformatics analysis of differentially expressed genes and identification of an miRNA-mRNA network associated with entorhinal cortex and hippocampus in Alzheimer's disease. Hereditas 2021; 158:25. [PMID: 34243818 PMCID: PMC8272337 DOI: 10.1186/s41065-021-00190-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/28/2021] [Indexed: 01/09/2023] Open
Abstract
Background Alzheimer’s disease (AD) is a fatal neurodegenerative disorder, and the lesions originate in the entorhinal cortex (EC) and hippocampus (HIP) at the early stage of AD progression. Gaining insight into the molecular mechanisms underlying AD is critical for the diagnosis and treatment of this disorder. Recent discoveries have uncovered the essential roles of microRNAs (miRNAs) in aging and have identified the potential of miRNAs serving as biomarkers in AD diagnosis. Methods We sought to apply bioinformatics tools to investigate microarray profiles and characterize differentially expressed genes (DEGs) in both EC and HIP and identify specific candidate genes and pathways that might be implicated in AD for further analysis. Furthermore, we considered that DEGs might be dysregulated by miRNAs. Therefore, we investigated patients with AD and healthy controls by studying the gene profiling of their brain and blood samples to identify AD-related DEGs, differentially expressed miRNAs (DEmiRNAs), along with gene ontology (GO) analysis, KEGG pathway analysis, and construction of an AD-specific miRNA–mRNA interaction network. Results Our analysis identified 10 key hub genes in the EC and HIP of patients with AD, and these hub genes were focused on energy metabolism, suggesting that metabolic dyshomeostasis contributed to the progression of the early AD pathology. Moreover, after the construction of an miRNA–mRNA network, we identified 9 blood-related DEmiRNAs, which regulated 10 target genes in the KEGG pathway. Conclusions Our findings indicated these DEmiRNAs having the potential to act as diagnostic biomarkers at an early stage of AD. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-021-00190-0.
Collapse
Affiliation(s)
- Haoming Li
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center, Neuroregeneration of Nantong University, Nantong, 226001, Jiangsu, China
| | - Linqing Zou
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Jinhong Shi
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Xiao Han
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China. .,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center, Neuroregeneration of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
45
|
Ling TS, Chandrasegaran S, Xuan LZ, Suan TL, Elaine E, Nathan DV, Chai YH, Gunasekaran B, Salvamani S. The Potential Benefits of Nanotechnology in Treating Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5550938. [PMID: 34285915 PMCID: PMC8275379 DOI: 10.1155/2021/5550938] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease is a neurodegenerative disorder that is caused by the accumulation of beta-amyloid plaques in the brain. Currently, there is no definitive cure available to treat Alzheimer's disease. The available medication in the market has the ability to only slow down its progression. However, nanotechnology has shown its superiority that can be applied for medical usage and it has a great potential in the therapy of Alzheimer's disease, specifically in the disease diagnosis and providing an alternative approach to treat Alzheimer's disease. This is done by increasing the efficiency of drug delivery by penetrating and overcoming the blood-brain barrier. Having said that, there are limitations that need to be further investigated and researched in order to minimize the adverse effects and potential toxicity and to improve drug bioavailability. The recent advances in the treatment of Alzheimer's disease using nanotechnology include the regeneration of stem cells, nanomedicine, and neuroprotection. In this review, we will discuss the advancement of nanotechnology which helps in the diagnosis and treatment of neurodegenerative disorders such as Alzheimer's disease as well as its challenges.
Collapse
Affiliation(s)
- Tan Sook Ling
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Shanthini Chandrasegaran
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Low Zhi Xuan
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Tong Li Suan
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Elaine Elaine
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Durrgashini Visva Nathan
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Yam Hok Chai
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Baskaran Gunasekaran
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Shamala Salvamani
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
46
|
Robbins CB, Grewal DS, Stinnett SS, Soundararajan S, Yoon SP, Polascik BW, Liu AJ, Burke JR, Fekrat S. Assessing the Retinal Microvasculature in Individuals With Early and Late-Onset Alzheimer's Disease. Ophthalmic Surg Lasers Imaging Retina 2021; 52:336-344. [PMID: 34185588 DOI: 10.3928/23258160-20210528-06] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVE To evaluate retinal microvascular changes in early and late-onset Alzheimer's disease (AD). PATIENTS AND METHODS Eighty-six eyes of 50 late-onset AD participants, 27 eyes of 15 early onset AD participants, and 111 eyes of 57 cognitively normal controls were included. Optical coherence tomography angiography (OCTA) vessel density (VD) and perfusion density (PD) in Early Treatment Diabetic Retinopathy Study 3-mm and 6-mm circles and rings were assessed. RESULTS There was decreased PD in early onset AD 3-mm circle (P = .026) and ring (P = .026) versus controls as well as in late-onset AD 3-mm circle (P = .023) and ring (P = .023) versus controls. There was decreased VD in late-onset AD 3-mm circle (P = .012) and ring (P = .006). No parameters differed between early and late-onset AD (P > .05). CONCLUSIONS AD eyes exhibited decreased retinal microvascular density compared to controls. Retinal parameters may not differ between early onset AD and late-onset AD after adjusting for age. [Ophthalmic Surg Lasers Imaging Retina. 2021;52:336-344.].
Collapse
|
47
|
Ullah MA, Johora FT, Sarkar B, Araf Y, Ahmed N, Nahar AN, Akter T. Computer-assisted evaluation of plant-derived β-secretase inhibitors in Alzheimer’s disease. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00150-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Abstract
Background
Alzheimer’s disease (AD) is a progressive neurodegenerative age-related dementia that results in memory loss of elderly people. Many hypotheses have been formally articulated till now to decipher the pathogenesis of this disease. According to the compelling amyloidogenic hypothesis, β-secretase is a key regulatory enzyme in AD development and is therefore considered as one of the major targets for the development of drugs to treat AD. In this study, 40 plant-derived phytocompounds, proven to have β-secretase inhibitory activity in different laboratory experiments, were evaluated using computational approaches in order to identify the best possible β-secretase inhibitor(s).
Results
Amentoflavone (IFD score: − 7.842 Kcal/mol), Bilobetin (IFD score: − 7.417 Kcal/mol), and Ellagic acid (IFD score: − 6.923 Kcal/mol) showed highest β-secretase inhibitory activities with high binding affinity among all the selected phytocompounds and interacted with key amino acids, i.e., Asp32, Tyr71, and Asp228 in the catalytic site of β-secretase. Moreover, these three molecules exhibited promising results in different drug potential assessment experiments and displayed signs of correlation with significant pharmacological and biological activities.
Conclusion
Amentoflavone, Biolbetin, and Ellagic acid could be investigated further in developing β-secretase-dependent drug for the effective treatment of AD. However, additional in vivo and in vitro experiments might be required to strengthen the findings of this experiment.
Collapse
|
48
|
Ali M, Saleem U, Anwar F, Imran M, Nadeem H, Ahmad B, Ali T, Ismail T. Screening of Synthetic Isoxazolone Derivative Role in Alzheimer's Disease: Computational and Pharmacological Approach. Neurochem Res 2021; 46:905-920. [PMID: 33486698 DOI: 10.1007/s11064-021-03229-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/16/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is age-dependent neurological disorder with progressive loss of cognition and memory. This multifactorial disease is characterized by intracellular neurofibrillary tangles, beta amyloid plaques, neuroinflammation, and increased oxidative stress. The increased cellular manifestations of these markers play a critical role in neurodegeneration and pathogenesis of AD. Therefore, reducing neurodegeneration by decreasing one or more of these markers may provide a potential therapeutic roadmap for the treatment of AD. AD causes a devastating loss of cognition with no conclusive and effective treatment. Many synthetic compound containing isoxazolone nucleus have been reported as neuroprotective agents. The aim of this study was to explore the anti-Alzheimer's potential of a newly synthesized 3,4,5-trimethoxy isoxazolone derivative (TMI) that attenuated the beta amyloid (Aβ1-42) and tau protein levels in streptozotocin (STZ) induced Alzheimer's disease mouse model. Molecular analysis revealed increased beta amyloid (Aβ1-42) protein levels, increased tau protein levels, increased cellular oxidative stress and reduced antioxidant enzymes in STZ exposed mice brains. Furthermore, ELISA and PCR were used to validate the expression of Aβ1-42. Pre-treatment with TMI significantly improved the memory and cognitive behavior along with ameliorated levels of Aβ1-42 proteins. TMI treated mice further showed marked increase in GSH, CAT, SOD levels while decreased levels of acetylcholinesterase inhibitors (AChEI's) and MDA intermediate. The multidimensional nature of isoxazolone derivatives and its versatile affinity towards various targets highpoint its multistep targeting nature. These results indicated the neuroprotective potential of TMI which may be considered for the treatment of neurodegenerative disease specifically in AD.
Collapse
Affiliation(s)
- Meissam Ali
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, 54000, Pakistan
| | - Uzma Saleem
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, 38000, Pakistan
| | - Fareeha Anwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, 54000, Pakistan.
| | - Muhammad Imran
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Bashir Ahmad
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, 54000, Pakistan
| | - Tahir Ali
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, 54000, Pakistan
| | - Tariq Ismail
- Department of Pharmacy, COMSATS University, Abbottabad, 22060, Pakistan
| |
Collapse
|
49
|
Gumus M, Multani N, Mack ML, Tartaglia MC. Progression of neuropsychiatric symptoms in young-onset versus late-onset Alzheimer's disease. GeroScience 2021; 43:213-223. [PMID: 33420706 DOI: 10.1007/s11357-020-00304-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022] Open
Abstract
Young-onset and late-onset Alzheimer's disease has different clinical presentations with late-onset presenting most often with memory deficits while young-onset often presents with a non-amnestic syndrome. However, it is unknown whether there are differences in presentation and progression of neuropsychiatric symptoms in young- versus late-onset Alzheimer's disease. We aimed to investigate differences in the prevalence and severity of neuropsychiatric symptoms in patients with young- and late-onset Alzheimer's disease longitudinally with and without accounting for the effect of medication usage. Sex differences were also considered in these patient groups. We included 126 young-onset and 505 late-onset Alzheimer's disease patients from National Alzheimer's Coordinating Center-Uniform Data Set (NACC-UDS) and Alzheimer's Disease Neuroimaging Initiative (ADNI). We investigated the prevalence and severity of neuropsychiatric symptoms using the Neuropsychiatric Inventory-Questionnaire over 4 visits with 1-year intervals, using a linear mixed-effects model. The prevalence of depression was significantly higher in young-onset than late-onset Alzheimer's disease over a 4-year interval when antidepressant usage was included in our analyses. Our findings suggest that neuropsychiatric symptom profiles of young- and late-onset Alzheimer's disease differ cross-sectionally but also display significant differences in progression.
Collapse
Affiliation(s)
- Melisa Gumus
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Ave., Toronto, ON, M5T 2S8, Canada
| | - Namita Multani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Ave., Toronto, ON, M5T 2S8, Canada
| | - Michael L Mack
- Department of Psychology, Faculty of Arts and Science, University of Toronto, Sidney Smith Hall, 100 St. George Street, Toronto, ON, M5S 3G3, Canada
| | - Maria Carmela Tartaglia
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Ave., Toronto, ON, M5T 2S8, Canada. .,Division of Neurology, Toronto Western Hospital, University Health Network, 399 Bathurst St., Toronto, ON, M5T 2S8, Canada.
| | | |
Collapse
|
50
|
Liu M, Beckett TL, Thomason LAM, Dorr A, Stefanovic B, McLaurin J. Covert strokes prior to Alzheimer's disease onset accelerate peri-lesional pathology but not cognitive deficits in an inducible APP mouse model. Brain Res 2021; 1754:147233. [PMID: 33412147 DOI: 10.1016/j.brainres.2020.147233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022]
Abstract
It is estimated that up to 1 in 3 healthy middle-aged adults will have had a covert stroke during their lifetime. Furthermore, post-stroke, survivors are more than twice as likely to develop dementia. In the present study, we aimed to model the impact of focal subclinical ischemia prior to the onset of AD pathogenesis in a preclinical model. We utilized endothelin-1 to induce ischemia in an iducible transgenic mouse model of Alzheimer's disease, APPsi:tTA, allowing for temporal control of APP gene expression. We induced the focal subclinical ischemic events in the absence of APP expression, thus prior to AD onset. T2 structural magnetic resonance imaging confirmed the volume and location of focal subclinical ischemic lesions to the medial prefrontal cortex. Following recovery from surgery and 7 weeks of APP expression, we found that two subclinical ischemic lesions resulted in a significant localized increase in amyloid load and in microglial activation proximal to the lesion. However, no differences were found in astrogliosis. A battery of behaviour tests was conducted, in which no significant differences were detected in activities of daily living and cognitive function between stroked and sham cohorts. Overall, our results demonstrated that APP expression was the sole driving force behind behavioural deficits. In conclusion, our results suggest that a history of two subclinical strokes prior to AD onset does not worsen early disease trajectory in a mouse model.
Collapse
Affiliation(s)
- Mingzhe Liu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.
| | - Tina L Beckett
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Adrienne Dorr
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Bojana Stefanovic
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - JoAnne McLaurin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|