1
|
Safiri S, Ghaffari Jolfayi A, Fazlollahi A, Morsali S, Sarkesh A, Daei Sorkhabi A, Golabi B, Aletaha R, Motlagh Asghari K, Hamidi S, Mousavi SE, Jamalkhani S, Karamzad N, Shamekh A, Mohammadinasab R, Sullman MJM, Şahin F, Kolahi AA. Alzheimer's disease: a comprehensive review of epidemiology, risk factors, symptoms diagnosis, management, caregiving, advanced treatments and associated challenges. Front Med (Lausanne) 2024; 11:1474043. [PMID: 39736972 PMCID: PMC11682909 DOI: 10.3389/fmed.2024.1474043] [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: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Background Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired reasoning. It is the leading cause of dementia in older adults, marked by the pathological accumulation of amyloid-beta plaques and neurofibrillary tangles. These pathological changes lead to widespread neuronal damage, significantly impacting daily functioning and quality of life. Objective This comprehensive review aims to explore various aspects of Alzheimer's disease, including its epidemiology, risk factors, clinical presentation, diagnostic advancements, management strategies, caregiving challenges, and emerging therapeutic interventions. Methods A systematic literature review was conducted across multiple electronic databases, including PubMed, MEDLINE, Cochrane Library, and Scopus, from their inception to May 2024. The search strategy incorporated a combination of keywords and Medical Subject Headings (MeSH) terms such as "Alzheimer's disease," "epidemiology," "risk factors," "symptoms," "diagnosis," "management," "caregiving," "treatment," and "novel therapies." Boolean operators (AND, OR) were used to refine the search, ensuring a comprehensive analysis of the existing literature on Alzheimer's disease. Results AD is significantly influenced by genetic predispositions, such as the apolipoprotein E (APOE) ε4 allele, along with modifiable environmental factors like diet, physical activity, and cognitive engagement. Diagnostic approaches have evolved with advances in neuroimaging techniques (MRI, PET), and biomarker analysis, allowing for earlier detection and intervention. The National Institute on Aging and the Alzheimer's Association have updated diagnostic criteria to include biomarker data, enhancing early diagnosis. Conclusion The management of AD includes pharmacological treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, which provide symptomatic relief but do not slow disease progression. Emerging therapies, including amyloid-beta and tau-targeting treatments, gene therapy, and immunotherapy, offer potential for disease modification. The critical role of caregivers is underscored, as they face considerable emotional, physical, and financial burdens. Support programs, communication strategies, and educational interventions are essential for improving caregiving outcomes. While significant advancements have been made in understanding and managing AD, ongoing research is necessary to identify new therapeutic targets and enhance diagnostic and treatment strategies. A holistic approach, integrating clinical, genetic, and environmental factors, is essential for addressing the multifaceted challenges of Alzheimer's disease and improving outcomes for both patients and caregivers.
Collapse
Affiliation(s)
- Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaffari Jolfayi
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asra Fazlollahi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Morsali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnam Golabi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Aletaha
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kimia Motlagh Asghari
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sana Hamidi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Jamalkhani
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Karamzad
- Department of Persian Medicine, School of Traditional, Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark J. M. Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Zhang X, Huang Y, Liu S, Ma S, Li M, Zhu Z, Wang W, Zhang X, Liu J, Tang S, Hu Y, Ge Z, Yu H, He M, Shang X. Machine learning based metabolomic and genetic profiles for predicting multiple brain phenotypes. J Transl Med 2024; 22:1098. [PMID: 39627804 PMCID: PMC11613467 DOI: 10.1186/s12967-024-05868-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/09/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND It is unclear regarding the association between metabolomic state/genetic risk score(GRS) and brain volumes and how much of variance of brain volumes is attributable to metabolomic state or GRS. METHODS Our analysis included 8635 participants (52.5% females) aged 40-70 years at baseline from the UK Biobank. Metabolomic profiles were assessed using nuclear magnetic resonance at baseline (between 2006 and 2010). Brain volumes were measured using magnetic resonance imaging between 2014 and 2019. Machine learning was used to generate metabolomic state and GRS for each of 21 brain phenotypes. RESULTS Individuals in the top 20% of metabolomic state had 2.4-35.7% larger volumes of 21 individual brain phenotypes compared to those in the bottom 20% while the corresponding number for GRS ranged from 1.5 to 32.8%. The proportion of variance of brain volumes (R [2]) explained by the corresponding metabolomic state ranged from 2.2 to 19.4%, and the corresponding number for GRS ranged from 0.8 to 8.7%. Metabolomic state provided no or minimal additional prediction values of brain volumes to age and sex while GRS provided moderate additional prediction values (ranging from 0.8 to 8.8%). No significant interplay between metabolomic state and GRS was observed, but the association between metabolomic state and some regional brain volumes was stronger in men or younger individuals. Individual metabolomic profiles including lipids and fatty acids were strong predictors of brain volumes. CONCLUSIONS In conclusion, metabolomic state is strongly associated with multiple brain volumes but provides minimal additional prediction value of brain volumes to age + sex. Although GRS is a weaker contributor to brain volumes than metabolomic state, it provides moderate additional prediction value of brain volumes to age + sex. Our findings suggest metabolomic state and GRS are important predictors for multiple brain phenotypes.
Collapse
Affiliation(s)
- Xueli Zhang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Yu Huang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Shunming Liu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shuo Ma
- Medical Big Data Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences, Southern Medical University), Guangzhou, China
| | - Min Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhuoting Zhu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xiayin Zhang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jiahao Liu
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia
| | - Shulin Tang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yijun Hu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zongyuan Ge
- Monash e-Research Center, Faculty of Engineering, Airdoc Research, Nvidia AI Technology Research Center, Monash University, Melbourne, VIC, 3800, Australia
| | - Honghua Yu
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Mingguang He
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China.
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong, China.
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong, Science Park, Hong Kong, China.
| | - Xianwen Shang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Centre for Eye Research Australia, Melbourne, VIC, 3002, Australia.
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Melbourne, VIC, 3050, Australia.
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China.
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong, China.
| |
Collapse
|
3
|
Tonietto M, Sotolongo-Grau O, Roé-Vellvé N, Bullich S, Tartari JP, Sanabria Á, García-Sánchez A, Borroni E, Galli C, Pérez-Martínez E, Castell-Conesa J, Roca I, Tárraga L, Ruiz A, Stephens AW, Boada M, Klein G, Marquié M. Head-to-head comparison of tau PET tracers [ 18F]PI-2620 and [ 18F]RO948 in non-demented individuals with brain amyloid deposition: the TAU-PET FACEHBI cohort. Alzheimers Res Ther 2024; 16:257. [PMID: 39605030 PMCID: PMC11603960 DOI: 10.1186/s13195-024-01622-5] [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: 07/15/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Second-generation tau tracers for positron emission tomography (PET) show high affinity for paired helical filaments tau deposits characteristic of Alzheimer´s disease and low off-target binding. Differences in their chemical structure though may lead to variations in their regional tau uptake and off-target signal. In this work, we aimed to compare the in-vivo uptake of tau tracers [18F]PI-2620 and [18F]RO948 in the early stages of the AD continuum. METHODS Data from the TAU-PET FACEHBI clinical trial (EUDRA-CT 2021-000473-83) were analyzed. All participants were non-demented and underwent tau imaging with [18F]PI-2620 and [18F]RO948 PET within 3 months, amyloid imaging with [18F]Florbetaben and brain magnetic resonance imaging. Tau PET standardized uptake values ratios (SUVR) were calculated in Braak and typical off-target regions using the inferior cerebellar cortex as a reference region. RESULTS The cohort consisted of 18 individuals with subjective cognitive decline (n = 13) and mild cognitive impairment (n = 5), with centiloid values ranging from 17 to 159. Both tau tracers showed similar tau pathology distribution but presented a distinct off-target signal pattern on visual read. SUVR measurements for [18F]PI-2620 and [18F]RO948 were highly correlated in all Braak regions (R2 range [0.65-0.80]). Regarding off-target signal, [18F]PI-2620 had higher SUVRs in vascular structures, and [18F]RO948 had higher SUVRs in the skull/meninges. CONCLUSIONS In a cohort of individuals at early stages of the AD continuum, tau PET tracers [18F]PI-2620 and [18F]RO948 showed similar in-vivo uptake in all Braak regions and distinct off-target signal. These preliminary results support the development of standardized quantification scales for tau deposition that are tracer-independent. TRIAL REGISTRATION AEMPS EudraCT 2021-000473-83. Registered 30 December 2021.
Collapse
Affiliation(s)
- Matteo Tonietto
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F.Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Oscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
| | | | | | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ainhoa García-Sánchez
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
| | - Edilio Borroni
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F.Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Christopher Galli
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F.Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | | | | - Lluís Tárraga
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Mercè Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Gregory Klein
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F.Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marta Marquié
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain.
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Liu Z, Xu Z, Yan A, Zhang P, Wei W. The association between precuneus cortex thickness and mild behavioral impairment in patients with mild stroke. Brain Imaging Behav 2024:10.1007/s11682-024-00955-x. [PMID: 39531165 DOI: 10.1007/s11682-024-00955-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The objective of this research was to examine the association between precuneus cortex thickness and mild behavioral impairment (MBI) in patients with mild stroke. Seventy-two patients were evaluated by high-resolution 3 T magnetic resonance and the mild behavioral impairment checklist (MBI-C). To determine the association between precuneus cortex thickness and MBI, we adjusted for demographics, vascular risk factors, and laboratory examination indicators in logistic regression analysis. In addition, we used mendelian randomization to further study the association through genetic databases. Of the 72 mild stroke patients in this study, 26 had MBI. We found a strong negative connection between precuneus cortex thickness and MBI after adjusting for any confounding variables. In patients with an initial mild stroke, the thinner the precuneus cortex, the higher the risk of MBI (OR: 0.02; 95% CI: 0.00-0.39; P < 0.05). Our study has uncovered a significant negative association between the thickness of the precuneus cortex and MBI. This finding provides a novel viewpoint for the radiological diagnosis of MBI, thereby augmenting the contribution of imaging to the diagnostic process of MBI and advancing the prediction of dementia. Specifically, in patients who have suffered mild stroke, a reduction in the cortical thickness of the precuneus has been pinpointed as crucial radiographic evidence of preclinical cognitive impairment. This insight could potentially facilitate earlier detection and intervention strategies for cognitive decline.
Collapse
Affiliation(s)
- Zhengxin Liu
- Department of Neurology, Stroke Center / Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, China
| | - Ziwei Xu
- Department of Neurology, Stroke Center / Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, China
| | - Aijuan Yan
- Department of Neurology, Stroke Center / Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, China
| | - Panpan Zhang
- Department of Neurology, Stroke Center / Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, China
| | - Wenshi Wei
- Department of Neurology, Stroke Center / Cognitive Disorders Center, Huadong Hospital Affiliated to Fudan University, No. 221, West Yan An Road, Shanghai, China.
| |
Collapse
|
5
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [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: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
6
|
Smolek T, Vince-Kazmerova Z, Hanes J, Stevens E, Palus V, Hajek I, Katina S, Novak P, Zilka N. On the utility of cerebrospinal fluid biomarkers in canine neurological disorders. Sci Rep 2024; 14:24129. [PMID: 39406773 PMCID: PMC11480401 DOI: 10.1038/s41598-024-73812-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
The cerebral biomarkers, neurofilament light chain (NfL), amyloid-β, tau, and neuron specific enolase (NSE) reflect a wide spectrum of neurological damage in the brain and spinal cord. With this study, we aimed to assess whether these biomarkers hold any potential diagnostic value for the three most common canine neurological diseases. Canines suffering from meningoencephalitis of unknown origin (MUO), brain tumors, and selected non-infectious myelopathies were included. For each diagnosis, we analyzed these biomarkers in the cerebrospinal fluid collected via cranial puncture from the cisterna magna. Elevated levels of CSF tau, NfL, and NSE were observed in MUO, with all three biomarkers being intercorrelated. Tau and NSE were increased while amyloid-β was decreased in dogs suffering from tumors. In contrast, no biomarker changes were observed in dogs with myelopathies. Covariates such as age, sex, or castration had minimal impact. CSF biomarkers may reflect molecular changes related to MUO and tumors, but not to non-infectious myelopathies. The combination of NfL, tau, and NSE may represent useful biomarkers for MUO as they reflect the same pathology and are not influenced by age.
Collapse
Affiliation(s)
- Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská Cesta 9, Bratislava, Slovak Republic
- Neuroimmunology Institute, n.p.o., Dvořákovo nábrežie 7527/10, 811 02, Bratislava, Slovak Republic
| | - Zuzana Vince-Kazmerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská Cesta 9, Bratislava, Slovak Republic
| | - Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská Cesta 9, Bratislava, Slovak Republic
| | - Eva Stevens
- Axon Neuroscience R&D Services SE, Dvořakovo Nabrezie 10, Bratislava, Slovak Republic
| | - Viktor Palus
- Neurovet -Referral Center for Veterinary Neurology, Bratislavska 2196/32, Trencin, Slovak Republic
| | - Ivo Hajek
- Small Animal Referral Centre Sibra, Na Vrátkach 13, Bratislava, Slovak Republic
| | - Stanislav Katina
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská Cesta 9, Bratislava, Slovak Republic
- Institute of Mathematics and Statistics, Masaryk University, Kotlářská 267/2, 611 37, Brno, Czech Republic
| | - Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská Cesta 9, Bratislava, Slovak Republic.
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská Cesta 9, Bratislava, Slovak Republic
- Neuroimmunology Institute, n.p.o., Dvořákovo nábrežie 7527/10, 811 02, Bratislava, Slovak Republic
| |
Collapse
|
7
|
Huang B, Zheng W, Mu R, Yang P, Li X, Liu F, Qin X, Zhu X. Disparities in the diagnostic efficacy of radiomics models in predicting various degrees of cognitive impairment in patients with cerebral small vessel disease. BMC Med Imaging 2024; 24:257. [PMID: 39333913 PMCID: PMC11428886 DOI: 10.1186/s12880-024-01431-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Aim to validate the diagnostic efficacy of radiomics models for predicting various degrees of cognitive impairment in patients with cerebral small vessel disease (CSVD). METHODS Participants were divided into mild cognitive impairment group (mild-CSVD group) and sever cognitive impairment group (sever-CSVD group) according to Montreal Cognitive Assessment (MoCA) performance, 98 gender-age-education matched subjects served as normal controls. Radiomic features were extracted from the segmented hippocampus using PyRadiomics. The feature preprocessing involved replacing missing values with the mean, applying stratified random sampling to allocate subjects into training (80%) and testing (20%) sets, ensuring balance among the three classes (normal controls, mild-CSVD group, and sever-CSVD group). A feature selection method was applied to identify discriminative radiomic features, with the optimal texture feature chosen for developing diagnostic models. Performance was evaluated in both the training and testing sets using receiver operating characteristic (ROC) curve analysis. RESULTS The radiomics model achieved an accuracy of 0.625, an AUC of 0.593, a sensitivity of 0.828, and a specificity of 0.316 in distinguishing mild-CSVD group from normal controls. When distinguishing mild-CSVD group from sever-CSVD group, the radiomics model reached an accuracy of 0.683, an AUC of 0.660, a sensitivity of 0.167, and a specificity of 0.897. Similarly, in distinguishing sever-CSVD group from normal controls, the radiomics model exhibited an accuracy of 0.781, an AUC of 0.818, a sensitivity of 0.538, and a specificity of 0.947. CONCLUSION Radiomics model based on hippocampal texture had disparities in the diagnostic efficacy of radiomics models in predicting various degrees of cognitive impairment in patients with CSVD.
Collapse
Affiliation(s)
- Bingqin Huang
- Graduate School, Guilin Medical University, Guilin, 541002, China
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Wei Zheng
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Ronghua Mu
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Peng Yang
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Xin Li
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Fuzhen Liu
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China
| | - Xiaoyan Qin
- Department of Radiology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541004, China.
| | - Xiqi Zhu
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China.
- Life Science and Clinical Medicine Research Center, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China.
| |
Collapse
|
8
|
Saville L, Wu L, Habtewold J, Cheng Y, Gollen B, Mitchell L, Stuart-Edwards M, Haight T, Mohajerani M, Zovoilis A. NERD-seq: a novel approach of Nanopore direct RNA sequencing that expands representation of non-coding RNAs. Genome Biol 2024; 25:233. [PMID: 39198865 PMCID: PMC11351768 DOI: 10.1186/s13059-024-03375-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Non-coding RNAs (ncRNAs) are frequently documented RNA modification substrates. Nanopore Technologies enables the direct sequencing of RNAs and the detection of modified nucleobases. Ordinarily, direct RNA sequencing uses polyadenylation selection, studying primarily mRNA gene expression. Here, we present NERD-seq, which enables detection of multiple non-coding RNAs, excluded by the standard approach, alongside natively polyadenylated transcripts. Using neural tissues as a proof of principle, we show that NERD-seq expands representation of frequently modified non-coding RNAs, such as snoRNAs, snRNAs, scRNAs, srpRNAs, tRNAs, and rRFs. NERD-seq represents an RNA-seq approach to simultaneously study mRNA and ncRNA epitranscriptomes in brain tissues and beyond.
Collapse
Affiliation(s)
- Luke Saville
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E3N4, Canada
- Paul Albrechtsen Research Institute, CCMB, Winnipeg, MB, R3E3N4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Li Wu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E3N4, Canada
- Paul Albrechtsen Research Institute, CCMB, Winnipeg, MB, R3E3N4, Canada
| | - Jemaneh Habtewold
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E3N4, Canada
- Paul Albrechtsen Research Institute, CCMB, Winnipeg, MB, R3E3N4, Canada
| | - Yubo Cheng
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Babita Gollen
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Liam Mitchell
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E3N4, Canada
- Paul Albrechtsen Research Institute, CCMB, Winnipeg, MB, R3E3N4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Matthew Stuart-Edwards
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E3N4, Canada
- Paul Albrechtsen Research Institute, CCMB, Winnipeg, MB, R3E3N4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Travis Haight
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E3N4, Canada
- Paul Albrechtsen Research Institute, CCMB, Winnipeg, MB, R3E3N4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Majid Mohajerani
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada
| | - Athanasios Zovoilis
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E3N4, Canada.
- Paul Albrechtsen Research Institute, CCMB, Winnipeg, MB, R3E3N4, Canada.
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada.
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, AB, T1K3M4, Canada.
| |
Collapse
|
9
|
Torso M, Fumagalli G, Ridgway GR, Contarino VE, Hardingham I, Scarpini E, Galimberti D, Chance SA, Arighi A. Clinical utility of diffusion MRI-derived measures of cortical microstructure in a real-world memory clinic setting. Ann Clin Transl Neurol 2024; 11:1964-1976. [PMID: 39049198 PMCID: PMC11330221 DOI: 10.1002/acn3.52097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/09/2024] [Accepted: 05/12/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE To investigate cortical microstructural measures from diffusion MRI as "neurodegeneration" markers that could improve prognostic accuracy in mild cognitive impairment (MCI). METHODS The prognostic power of Amyloid/Tau/Neurodegeneration (ATN) biomarkers to predict progression from MCI to AD or non-AD dementia was investigated. Ninety patients underwent clinical evaluation (follow-up interval 32 ± 18 months), lumbar puncture, and MRI. Participants were grouped by clinical stage and cerebrospinal fluid Amyloid and Tau status. T1-structural and diffusion MRI scans were analyzed to calculate diffusion metrics related to cortical columnar structure (AngleR, ParlPD, PerpPD+), cortical mean diffusivity, and fractional anisotropy. Statistical tests were corrected for multiple comparisons. Prognostic power was assessed using receiver operating characteristic (ROC) analysis and related indices. RESULTS A progressive increase of whole-brain cortical diffusion values was observed along the AD continuum, with all A+ groups showing significantly higher AngleR than A-T-. Investigating clinical progression to dementia, the AT biomarkers together showed good positive predictive value (with 90.91% of MCI A+T+ converting to dementia) but poor negative predictive value (with 40% of MCI A-T- progressing to a mix of AD and non-AD dementias). Adding whole-brain AngleR as an N marker, produced good differentiation between stable and converting MCI A-T- patients (0.8 area under ROC curve) and substantially improved negative predictive value (+21.25%). INTERPRETATION Results support the clinical utility of cortical microstructure to aid prognosis, especially in A-T- patients. Further work will investigate other complexities of the real-world clinical setting, including A-T+ groups. Diffusion MRI measures of neurodegeneration may complement fluid AT markers to support clinical decision-making.
Collapse
Affiliation(s)
| | - Giorgio Fumagalli
- Center For Mind/Brain Sciences‐CIMeCUniversity of TrentoRoveretoItaly
| | | | | | | | - Elio Scarpini
- Neurodegenerative Disease UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Daniela Galimberti
- Neurodegenerative Disease UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Dept. of Biomedical, Surgical and Dental SciencesUniversity of MilanMilanItaly
| | | | - Andrea Arighi
- Neurodegenerative Disease UnitFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
10
|
Xu Q, Kim Y, Chung K, Schulz P, Gottlieb A. Prediction of Mild Cognitive Impairment Status: Pilot Study of Machine Learning Models Based on Longitudinal Data From Fitness Trackers. JMIR Form Res 2024; 8:e55575. [PMID: 39024003 PMCID: PMC11294783 DOI: 10.2196/55575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/15/2024] [Accepted: 06/08/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Early signs of Alzheimer disease (AD) are difficult to detect, causing diagnoses to be significantly delayed to time points when brain damage has already occurred and current experimental treatments have little effect on slowing disease progression. Tracking cognitive decline at early stages is critical for patients to make lifestyle changes and consider new and experimental therapies. Frequently studied biomarkers are invasive and costly and are limited for predicting conversion from normal to mild cognitive impairment (MCI). OBJECTIVE This study aimed to use data collected from fitness trackers to predict MCI status. METHODS In this pilot study, fitness trackers were worn by 20 participants: 12 patients with MCI and 8 age-matched controls. We collected physical activity, heart rate, and sleep data from each participant for up to 1 month and further developed a machine learning model to predict MCI status. RESULTS Our machine learning model was able to perfectly separate between MCI and controls (area under the curve=1.0). The top predictive features from the model included peak, cardio, and fat burn heart rate zones; resting heart rate; average deep sleep time; and total light activity time. CONCLUSIONS Our results suggest that a longitudinal digital biomarker differentiates between controls and patients with MCI in a very cost-effective and noninvasive way and hence may be very useful for identifying patients with very early AD who can benefit from clinical trials and new, disease-modifying therapies.
Collapse
Affiliation(s)
- Qidi Xu
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yejin Kim
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karen Chung
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Paul Schulz
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Assaf Gottlieb
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
11
|
Jiang J, Zhuo Z, Wang A, Li W, Jiang S, Duan Y, Ren Q, Zhao M, Wang L, Yang S, Awan MUN, Liu Y, Xu J. Choroid plexus volume as a novel candidate neuroimaging marker of the Alzheimer's continuum. Alzheimers Res Ther 2024; 16:149. [PMID: 38961406 PMCID: PMC11221040 DOI: 10.1186/s13195-024-01520-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Enlarged choroid plexus (ChP) volume has been reported in patients with Alzheimer's disease (AD) and inversely correlated with cognitive performance. However, its clinical diagnostic and predictive value, and mechanisms by which ChP impacts the AD continuum remain unclear. METHODS This prospective cohort study enrolled 607 participants [healthy control (HC): 110, mild cognitive impairment (MCI): 269, AD dementia: 228] from the Chinese Imaging, Biomarkers, and Lifestyle study between January 1, 2021, and December 31, 2022. Of the 497 patients on the AD continuum, 138 underwent lumbar puncture for cerebrospinal fluid (CSF) hallmark testing. The relationships between ChP volume and CSF pathological hallmarks (Aβ42, Aβ40, Aβ42/40, tTau, and pTau181), neuropsychological tests [Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), Neuropsychiatric Inventory (NPI), and Activities of Daily Living (ADL) scores], and multimodal neuroimaging measures [gray matter volume, cortical thickness, and corrected cerebral blood flow (cCBF)] were analyzed using partial Spearman's correlation. The mediating effects of four neuroimaging measures [ChP volume, hippocampal volume, lateral ventricular volume (LVV), and entorhinal cortical thickness (ECT)] on the relationship between CSF hallmarks and neuropsychological tests were examined. The ability of the four neuroimaging measures to identify cerebral Aβ42 changes or differentiate among patients with AD dementia, MCI and HCs was determined using receiver operating characteristic analysis, and their associations with neuropsychological test scores at baseline were evaluated by linear regression. Longitudinal associations between the rate of change in the four neuroimaging measures and neuropsychological tests scores were evaluated on the AD continuum using generalized linear mixed-effects models. RESULTS The participants' mean age was 65.99 ± 8.79 years. Patients with AD dementia exhibited the largest baseline ChP volume than the other groups (P < 0.05). ChP volume enlargement correlated with decreased Aβ42 and Aβ40 levels; lower MMSE and MoCA and higher NPI and ADL scores; and lower volume, cortical thickness, and cCBF in other cognition-related regions (all P < 0.05). ChP volume mediated the association of Aβ42 and Aβ40 levels with MMSE scores (19.08% and 36.57%), and Aβ42 levels mediated the association of ChP volume and MMSE or MoCA scores (39.49% and 34.36%). ChP volume alone better identified cerebral Aβ42 changes than LVV alone (AUC = 0.81 vs. 0.67, P = 0.04) and EC thickness alone (AUC = 0.81 vs.0.63, P = 0.01) and better differentiated patients with MCI from HCs than hippocampal volume alone (AUC = 0.85 vs. 0.81, P = 0.01), and LVV alone (AUC = 0.85 vs.0.82, P = 0.03). Combined ChP and hippocampal volumes significantly increased the ability to differentiate cerebral Aβ42 changes and patients among AD dementia, MCI, and HCs groups compared with hippocampal volume alone (all P < 0.05). After correcting for age, sex, years of education, APOE ε4 status, eTIV, and hippocampal volume, ChP volume was associated with MMSE, MoCA, NPI, and ADL score at baseline, and rapid ChP volume enlargement was associated with faster deterioration in NPI scores with an average follow-up of 10.03 ± 4.45 months (all P < 0.05). CONCLUSIONS ChP volume may be a novel neuroimaging marker associated with neurodegenerative changes and clinical AD manifestations. It could better detect the early stages of the AD and predict prognosis, and significantly enhance the differential diagnostic ability of hippocampus on the AD continuum.
Collapse
Affiliation(s)
- Jiwei Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhizheng Zhuo
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Anxin Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wenyi Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shirui Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yunyun Duan
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiwei Ren
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Min Zhao
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Linlin Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shiyi Yang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Neurological Diseases, Beijing, China
| | | | - Yaou Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Jun Xu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
12
|
Bi XA, Wang Y, Luo S, Chen K, Xing Z, Xu L. Hypergraph Structural Information Aggregation Generative Adversarial Networks for Diagnosis and Pathogenetic Factors Identification of Alzheimer's Disease With Imaging Genetic Data. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2024; 35:7420-7434. [PMID: 36264725 DOI: 10.1109/tnnls.2022.3212700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with profound pathogenetic causes. Imaging genetic data analysis can provide comprehensive insights into its causes. To fully utilize the multi-level information in the data, this article proposes a hypergraph structural information aggregation model, and constructs a novel deep learning method named hypergraph structural information aggregation generative adversarial networks (HSIA-GANs) for the automatic sample classification and accurate feature extraction. Specifically, HSIA-GAN is composed of generator and discriminator. The generator has three main functions. First, vertex graph and edge graph are constructed based on the input hypergraph to present the low-order relations. Second, the low-order structural information of hypergraph is extracted by the designed vertex convolution layers and edge convolution layers. Finally, the synthetic hypergraph is generated as the input of the discriminator. The discriminator can extract the high-order structural information directly from hypergraph through vertex-edge convolution, fuse the high and low-order structural information, and finalize the results through the full connection (FC) layers. Based on the data acquired from AD neuroimaging initiative, HSIA-GAN shows significant advantages in three classification tasks, and extracts discriminant features conducive to better disease classification.
Collapse
|
13
|
Holleman J, Daniilidou M, Kåreholt I, Aspö M, Hagman G, Udeh-Momoh CT, Spulber G, Kivipelto M, Solomon A, Matton A, Sindi S. Diurnal cortisol, neuroinflammation, and neuroimaging visual rating scales in memory clinic patients. Brain Behav Immun 2024; 118:499-509. [PMID: 38503394 DOI: 10.1016/j.bbi.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/18/2024] [Accepted: 03/16/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Neuroinflammation is a hallmark of the Alzheimer's disease (AD) pathogenic process. Cortisol dysregulation may increase AD risk and is related to brain atrophy. This cross-sectional study aims to examine interactions of cortisol patterns and neuroinflammation markers in their association with neuroimaging correlates. METHOD 134 participants were recruited from the Karolinska University Hospital memory clinic (Stockholm, Sweden). Four visual rating scales were applied to magnetic resonance imaging or computed tomography scans: medial temporal lobe atrophy (MTA), global cortical atrophy (GCA), white matter lesions (WML), and posterior atrophy. Participants provided saliva samples for assessment of diurnal cortisol patterns, and underwent lumbar punctures for cerebrospinal fluid (CSF) sampling. Three cortisol measures were used: the cortisol awakening response, total daily output, and the ratio of awakening to bedtime levels. Nineteen CSF neuroinflammation markers were categorized into five composite scores: proinflammatory cytokines, other cytokines, angiogenesis markers, vascular injury markers, and glial activation markers. Ordinal logistic regressions were conducted to assess associations between cortisol patterns, neuroinflammation scores, and visual rating scales, and interactions between cortisol patterns and neuroinflammation scores in relation to visual rating scales. RESULT Higher levels of angiogenesis markers were associated with more severe WML. Some evidence was found for interactions between dysregulated diurnal cortisol patterns and greater neuroinflammation-related biomarkers in relation to more severe GCA and WML. No associations were found between cortisol patterns and visual rating scales. CONCLUSION This study suggests an interplay between diurnal cortisol patterns and neuroinflammation in relation to brain structure. While this cross-sectional study does not provide information on causality or temporality, these findings suggest that neuroinflammation may be involved in the relationship between HPA-axis functioning and AD.
Collapse
Affiliation(s)
- Jasper Holleman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden.
| | - Makrina Daniilidou
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ingemar Kåreholt
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden; Institute of Gerontology, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Malin Aspö
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden
| | - Göran Hagman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden
| | - Chinedu T Udeh-Momoh
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK; Division of Public Health Sciences, Wake Forest University School of Medicine, North Carolina, USA; Brain and Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Gabriela Spulber
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Alina Solomon
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK; Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anna Matton
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK
| | - Shireen Sindi
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK
| |
Collapse
|
14
|
Park B, Kim Y, Park J, Choi H, Kim SE, Ryu H, Seo K. Integrating Biomarkers From Virtual Reality and Magnetic Resonance Imaging for the Early Detection of Mild Cognitive Impairment Using a Multimodal Learning Approach: Validation Study. J Med Internet Res 2024; 26:e54538. [PMID: 38631021 PMCID: PMC11063880 DOI: 10.2196/54538] [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: 11/15/2023] [Revised: 12/29/2023] [Accepted: 03/09/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Early detection of mild cognitive impairment (MCI), a transitional stage between normal aging and Alzheimer disease, is crucial for preventing the progression of dementia. Virtual reality (VR) biomarkers have proven to be effective in capturing behaviors associated with subtle deficits in instrumental activities of daily living, such as challenges in using a food-ordering kiosk, for early detection of MCI. On the other hand, magnetic resonance imaging (MRI) biomarkers have demonstrated their efficacy in quantifying observable structural brain changes that can aid in early MCI detection. Nevertheless, the relationship between VR-derived and MRI biomarkers remains an open question. In this context, we explored the integration of VR-derived and MRI biomarkers to enhance early MCI detection through a multimodal learning approach. OBJECTIVE We aimed to evaluate and compare the efficacy of VR-derived and MRI biomarkers in the classification of MCI while also examining the strengths and weaknesses of each approach. Furthermore, we focused on improving early MCI detection by leveraging multimodal learning to integrate VR-derived and MRI biomarkers. METHODS The study encompassed a total of 54 participants, comprising 22 (41%) healthy controls and 32 (59%) patients with MCI. Participants completed a virtual kiosk test to collect 4 VR-derived biomarkers (hand movement speed, scanpath length, time to completion, and the number of errors), and T1-weighted MRI scans were performed to collect 22 MRI biomarkers from both hemispheres. Analyses of covariance were used to compare these biomarkers between healthy controls and patients with MCI, with age considered as a covariate. Subsequently, the biomarkers that exhibited significant differences between the 2 groups were used to train and validate a multimodal learning model aimed at early screening for patients with MCI among healthy controls. RESULTS The support vector machine (SVM) using only VR-derived biomarkers achieved a sensitivity of 87.5% and specificity of 90%, whereas the MRI biomarkers showed a sensitivity of 90.9% and specificity of 71.4%. Moreover, a correlation analysis revealed a significant association between MRI-observed brain atrophy and impaired performance in instrumental activities of daily living in the VR environment. Notably, the integration of both VR-derived and MRI biomarkers into a multimodal SVM model yielded superior results compared to unimodal SVM models, achieving higher accuracy (94.4%), sensitivity (100%), specificity (90.9%), precision (87.5%), and F1-score (93.3%). CONCLUSIONS The results indicate that VR-derived biomarkers, characterized by their high specificity, can be valuable as a robust, early screening tool for MCI in a broader older adult population. On the other hand, MRI biomarkers, known for their high sensitivity, excel at confirming the presence of MCI. Moreover, the multimodal learning approach introduced in our study provides valuable insights into the improvement of early MCI detection by integrating a diverse set of biomarkers.
Collapse
Affiliation(s)
- Bogyeom Park
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Yuwon Kim
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hojin Choi
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Seong-Eun Kim
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Hokyoung Ryu
- Graduate School of Technology and Innovation Management, Hanyang University, Seoul, Republic of Korea
| | - Kyoungwon Seo
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
15
|
Said A, Göker H. Spectral analysis and Bi-LSTM deep network-based approach in detection of mild cognitive impairment from electroencephalography signals. Cogn Neurodyn 2024; 18:597-614. [PMID: 38699612 PMCID: PMC11061085 DOI: 10.1007/s11571-023-10010-y] [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: 05/17/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 05/05/2024] Open
Abstract
Mild cognitive impairment (MCI) is a neuropsychological syndrome that is characterized by cognitive impairments. It typically affects adults 60 years of age and older. It is a noticeable decline in the cognitive function of the patient, and if left untreated it gets converted to Alzheimer's disease (AD). For that reason, early diagnosis of MCI is important as it slows down the conversion of the disease to AD. Early and accurate diagnosis of MCI requires recognition of the clinical characteristics of the disease, extensive testing, and long-term observations. These observations and tests can be subjective, expensive, incomplete, or inaccurate. Electroencephalography (EEG) is a powerful choice for the diagnosis of diseases with its advantages such as being non-invasive, based on findings, less costly, and getting results in a short time. In this study, a new EEG-based model is developed which can effectively detect MCI patients with higher accuracy. For this purpose, a dataset consisting of EEG signals recorded from a total of 34 subjects, 18 of whom were MCI and 16 control groups was used, and their ages ranged from 40 to 77. To conduct the experiment, the EEG signals were denoised using Multiscale Principal Component Analysis (MSPCA), and to increase the size of the dataset Data Augmentation (DA) method was performed. The tenfold cross-validation method was used to validate the model, moreover, the power spectral density (PSD) of the EEG signals was extracted from the EEG signals using three spectral analysis methods, the periodogram, welch, and multitaper. The PSD graphs of the EEG signals showed signal differences between the subjects of control and the MCI group, indicating that the signal power of MCI patients is lower compared to control groups. To classify the subjects, one of the best classifiers of deep learning algorithms called the Bi-directional long-short-term-memory (Bi-LSTM) was used, and several machine learning algorithms, such as decision tree (DT), support vector machine (SVM), and k-nearest neighbor (KNN). These algorithms were trained and tested using the extracted feature vectors from the control and the MCI groups. Additionally, the values of the coefficient matrix of those algorithms were compared and evaluated with the performance evaluation matrix to determine which one performed the best overall. According to the experimental results, the proposed deep learning model of multitaper spectral analysis approach with Bi-LSTM deep learning algorithm attained the highest number of correctly classified samples for diagnosing MCI patients and achieved a remarkable accuracy compared to the other proposed models. The achieved classification results of the deep learning model are reported to be 98.97% accuracy, 98.34% sensitivity, 99.67% specificity, 99.70% precision, 99.02% f1 score, and 97.94% Matthews correlation coefficient (MCC).
Collapse
Affiliation(s)
- Afrah Said
- Department of Electrical Electronics Engineering, Faculty of Simav Technology, Dumlupınar University, 43500 Kütahya, Turkey
| | - Hanife Göker
- Health Services Vocational College, Gazi University, 06830 Ankara, Turkey
| |
Collapse
|
16
|
Yeshaw Y, Madakkatel I, Mulugeta A, Lumsden A, Hyppönen E. Uncovering Predictors of Low Hippocampal Volume: Evidence from a Large-Scale Machine-Learning-Based Study in the UK Biobank. Neuroepidemiology 2024; 58:369-382. [PMID: 38560977 PMCID: PMC11449190 DOI: 10.1159/000538565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Hippocampal atrophy is an established biomarker for conversion from the normal ageing process to developing cognitive impairment and dementia. This study used a novel hypothesis-free machine-learning approach, to uncover potential risk factors of lower hippocampal volume using information from the world's largest brain imaging study. METHODS A combination of machine learning and conventional statistical methods were used to identify predictors of low hippocampal volume. We run gradient boosting decision tree modelling including 2,891 input features measured before magnetic resonance imaging assessments (median 9.2 years, range 4.2-13.8 years) using data from 42,152 dementia-free UK Biobank participants. Logistic regression analyses were run on 87 factors identified as important for prediction based on Shapley values. False discovery rate-adjusted p value <0.05 was used to declare statistical significance. RESULTS Older age, male sex, greater height, and whole-body fat-free mass were the main predictors of low hippocampal volume with the model also identifying associations with lung function and lifestyle factors including smoking, physical activity, and coffee intake (corrected p < 0.05 for all). Red blood cell count and several red blood cell indices such as haemoglobin concentration, mean corpuscular haemoglobin, mean corpuscular volume, mean reticulocyte volume, mean sphered cell volume, and red blood cell distribution width were among many biomarkers associated with low hippocampal volume. CONCLUSION Lifestyles, physical measures, and biomarkers may affect hippocampal volume, with many of the characteristics potentially reflecting oxygen supply to the brain. Further studies are required to establish causality and clinical relevance of these findings.
Collapse
Affiliation(s)
- Yigizie Yeshaw
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia,
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia,
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia,
- Department of Epidemiology and Biostatistics, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia,
| | - Iqbal Madakkatel
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Anwar Mulugeta
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa, Ethiopia
| | - Amanda Lumsden
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
17
|
Zhao J, Jiao Y, Wang H, Song P, Gao Z, Bing X, Zhang C, Ouyang A, Yao J, Wang S, Jiang H. Radiomic features of the hippocampal based on magnetic resonance imaging in the menopausal mouse model linked to neuronal damage and cognitive deficits. Brain Imaging Behav 2024; 18:368-377. [PMID: 38102441 PMCID: PMC11156756 DOI: 10.1007/s11682-023-00808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2023] [Indexed: 12/17/2023]
Abstract
Estrogen deficiency in the early postmenopausal phase is associated with an increased long-term risk of cognitive decline or dementia. Non-invasive characterization of the pathological features of the pathological hallmarks in the brain associated with postmenopausal women (PMW) could enhance patient management and the development of therapeutic strategies. Radiomics is a means to quantify the radiographic phenotype of a diseased tissue via the high-throughput extraction and mining of quantitative features from images acquired from modalities such as CT and magnetic resonance imaging (MRI). This study set out to explore the correlation between radiomics features based on MRI and pathological features of the hippocampus and cognitive function in the PMW mouse model. Ovariectomized (OVX) mice were used as PWM models. MRI scans were performed two months after surgery. The brain's hippocampal region was manually annotated, and the radiomic features were extracted with PyRadiomics. Chemiluminescence was used to evaluate the peripheral blood estrogen level of mice, and the Morris water maze test was used to evaluate the cognitive ability of mice. Nissl staining and immunofluorescence were used to quantify neuronal damage and COX1 expression in brain sections of mice. The OVX mice exhibited marked cognitive decline, brain neuronal damage, and increased expression of mitochondrial complex IV subunit COX1, which are pathological phenomena commonly observed in the brains of AD patients, and these phenotypes were significantly correlated with radiomics features (p < 0.05, |r|>0.5), including Original_firstorder_Interquartile Range, Original_glcm_Difference Average, Original_glcm_Difference Average and Wavelet-LHH_glszm_Small Area Emphasis. Meanwhile, the above radiomics features were significantly different between the sham-operated and OVX groups (p < 0.01) and were associated with decreased serum estrogen levels (p < 0.05, |r|>0.5). This initial study indicates that the above radiomics features may have a role in the assessment of the pathology of brain damage caused by estrogen deficiency using routinely acquired structural MR images.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Jiao
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hui Wang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Peiji Song
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhen Gao
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xue Bing
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chunling Zhang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Aimei Ouyang
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jian Yao
- Department of Radiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Song Wang
- Department of Radiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725, South Wanping Road, Shanghai, 200032, China.
| | - Huijie Jiang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
18
|
Wang X, Peng L, Zhan S, Yin X, Huang L, Huang J, Yang J, Zhang Y, Zeng Y, Liang S. Alterations in hippocampus-centered morphological features and function of the progression from normal cognition to mild cognitive impairment. Asian J Psychiatr 2024; 93:103921. [PMID: 38237533 DOI: 10.1016/j.ajp.2024.103921] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 03/08/2024]
Abstract
Mild cognitive impairment (MCI) is a significant precursor to dementia, highlighting the critical need for early identification of individuals at high risk of MCI to prevent cognitive decline. The study aimed to investigate the changes in brain structure and function before the onset of MCI. This study enrolled 19 older adults with progressive normal cognition (pNC) to MCI and 19 older adults with stable normal cognition (sNC). The gray matter (GM) volume and functional connectivity (FC) were estimated via magnetic resonance imaging during their normal cognition state 3 years prior. Additionally, spatial associations between FC maps and neurochemical profiles were examined using JuSpace. Compared to the sNC group, the pNC group showed decreased volume in the left hippocampus and left amygdala. The significantly positive correlation was observed between the GM volume of the left hippocampus and the MMSE scores after 3 years in pNC group. Besides, it showed that the pNC group had increased FC between the left hippocampus and the anterior-posterior cingulate gyrus, which was significantly correlated with the spatial distribution of dopamine D2 and noradrenaline transporter. Taken together, the study identified the abnormal brain characteristics before the onset of MCI, which might provide insight into clinical research.
Collapse
Affiliation(s)
- Xiuxiu Wang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Lixin Peng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Shiqi Zhan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Xiaolong Yin
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Li Huang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jiayang Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Junchao Yang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Yusi Zhang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Yi Zeng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Shengxiang Liang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; Fujian Key Laboratory of Cognitive Rehabilitation, Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fuzhou 350001, China.
| |
Collapse
|
19
|
Kluckert C, Hüll M. [Dementia Prevention]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2024; 92:90-106. [PMID: 38490216 DOI: 10.1055/a-2230-1845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Dementia is common and will continue to grow in importance and numbers in the future. However, as causal treatment is not possible in most cases, prevention is particularly important. This is not only aimed at cognitively healthy people, but is also a central element in all phases of the disease.
Collapse
|
20
|
Huang L, Li Q, Lu Y, Pan F, Cui L, Wang Y, Miao Y, Chen T, Li Y, Wu J, Chen X, Jia J, Guo Q. Consensus on rapid screening for prodromal Alzheimer's disease in China. Gen Psychiatr 2024; 37:e101310. [PMID: 38313393 PMCID: PMC10836380 DOI: 10.1136/gpsych-2023-101310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024] Open
Abstract
Alzheimer's disease (AD) is a common cause of dementia, characterised by cerebral amyloid-β deposition, pathological tau and neurodegeneration. The prodromal stage of AD (pAD) refers to patients with mild cognitive impairment (MCI) and evidence of AD's pathology. At this stage, disease-modifying interventions should be used to prevent the progression to dementia. Given the inherent heterogeneity of MCI, more specific biomarkers are needed to elucidate the underlying AD's pathology. Although the uses of cerebrospinal fluid and positron emission tomography are widely accepted methods for detecting AD's pathology, their clinical applications are limited by their high costs and invasiveness, particularly in low-income areas in China. Therefore, to improve the early detection of Alzheimer's disease (AD) pathology through cost-effective screening methods, a panel of 45 neurologists, psychiatrists and gerontologists was invited to establish a formal consensus on the screening of pAD in China. The supportive evidence and grades of recommendations are based on a systematic literature review and focus group discussion. National meetings were held to allow participants to review, vote and provide their expert opinions to reach a consensus. A majority (two-thirds) decision was used for questions for which consensus could not be reached. Recommended screening methods are presented in this publication, including neuropsychological assessment, peripheral biomarkers and brain imaging. In addition, a general workflow for screening pAD in China is established, which will help clinicians identify individuals at high risk and determine therapeutic targets.
Collapse
Affiliation(s)
- Lin Huang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qinjie Li
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Lu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengfeng Pan
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Cui
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Miao
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianlu Chen
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yatian Li
- Shanghai BestCovered, Shanghai, China
| | | | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianping Jia
- Department of Neurology, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Yin Y, Tam HL, Quint J, Chen M, Ding R, Zhang X. Epidemiology of Dementia in China in 2010-2020: A Systematic Review and Meta-Analysis. Healthcare (Basel) 2024; 12:334. [PMID: 38338219 PMCID: PMC10855047 DOI: 10.3390/healthcare12030334] [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: 12/12/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Dementia has become one of the leading causes of death across the world. AIMS The aim of this study was to investigate the incidence, prevalence, and mortality of dementia in China between 2010 and 2020, and to investigate any geographical, age, and sex differences in the prevalence and incidence of dementia. METHODS Five databases were searched. The Joanna Briggs Institute (JBI) critical appraisal tool was used to assess the quality of the included studies. A random-effects meta-analysis was performed to estimate the pooled prevalence of dementia. Subgroup analysis was based on the type of dementia. The incidence and mortality of dementia were synthesized qualitatively. RESULTS A total of 19 studies were included. The meta-analysis showed that the prevalence of dementia was 6% (95%CI 5%, 8%), the prevalence of Alzheimer's disease (AD) was 5% (95%CI 4%, 6%), and the prevalence of vascular dementia (VaD) was 1% (95%CI 0%, 2%). The subgroup analysis showed that the prevalence rates of dementia in rural (6%, 95%CI 4%, 8%) and urban areas were similar (6%, 95%CI 4%, 8%). Deaths due to dementia increased over time. CONCLUSION The prevalence, incidence, and mortality of dementia increased with age and over time. Applying consistent criteria to the diagnosis of cognitive impairment and dementia is necessary to help with disease monitoring. Promoting dementia knowledge and awareness at the community level is necessary.
Collapse
Affiliation(s)
- Yueheng Yin
- School of Nursing, Nanjing Medical University, Nanjing 210029, China;
| | - Hon Lon Tam
- The Nethersole School of Nursing, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China;
| | - Jennifer Quint
- School of Public Health, National Heart and Lung Institute, Imperial College London, London W12 7RQ, UK; (J.Q.); (R.D.)
| | - Mengyun Chen
- School of Nursing, Lanzhou University, Lanzhou 730000, China;
| | - Rong Ding
- School of Public Health, National Heart and Lung Institute, Imperial College London, London W12 7RQ, UK; (J.Q.); (R.D.)
| | - Xiubin Zhang
- School of Public Health, National Heart and Lung Institute, Imperial College London, London W12 7RQ, UK; (J.Q.); (R.D.)
| |
Collapse
|
22
|
Mohammadi-Pilehdarboni H, Shenagari M, Joukar F, Naziri H, Mansour-Ghanaei F. Alzheimer's disease and microorganisms: the non-coding RNAs crosstalk. Front Cell Neurosci 2024; 17:1256100. [PMID: 38249527 PMCID: PMC10796784 DOI: 10.3389/fncel.2023.1256100] [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: 07/10/2023] [Accepted: 10/25/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's disease (AD) is a complex, multifactorial disorder, influenced by a multitude of variables ranging from genetic factors, age, and head injuries to vascular diseases, infections, and various other environmental and demographic determinants. Among the environmental factors, the role of the microbiome in the genesis of neurodegenerative disorders (NDs) is gaining increased recognition. This paradigm shift is substantiated by an extensive body of scientific literature, which underscores the significant contributions of microorganisms, encompassing viruses and gut-derived bacteria, to the pathogenesis of AD. The mechanism by which microbial infection exerts its influence on AD hinges primarily on inflammation. Neuroinflammation, activated in response to microbial infections, acts as a defense mechanism for the brain but can inadvertently lead to unexpected neuropathological perturbations, ultimately contributing to NDs. Given the ongoing uncertainty surrounding the genetic factors underpinning ND, comprehensive investigations into environmental factors, particularly the microbiome and viral agents, are imperative. Recent advances in neuroscientific research have unveiled the pivotal role of non-coding RNAs (ncRNAs) in orchestrating various pathways integral to neurodegenerative pathologies. While the upstream regulators governing the pathological manifestations of microorganisms remain elusive, an in-depth exploration of the nuanced role of ncRNAs holds promise for the development of prospective therapeutic interventions. This review aims to elucidate the pivotal role of ncRNAs as master modulators in the realm of neurodegenerative conditions, with a specific focus on Alzheimer's disease.
Collapse
Affiliation(s)
- Hanieh Mohammadi-Pilehdarboni
- Faculty of Medicine and Dentistry and the School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shenagari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamed Naziri
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
23
|
Cui Y, Liu C, Wang Y, Xie H. Multimodal magnetic resonance scans of patients with mild cognitive impairment. Dement Neuropsychol 2023; 17:e20230017. [PMID: 38111592 PMCID: PMC10727029 DOI: 10.1590/1980-5764-dn-2023-0017] [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: 03/14/2023] [Revised: 09/04/2023] [Accepted: 10/20/2023] [Indexed: 12/20/2023] Open
Abstract
The advancement of neuroimaging technology offers a pivotal reference for the early detection of mild cognitive impairment (MCI), a significant area of focus in contemporary cognitive function research. Structural MRI scans present visual and quantitative manifestations of alterations in brain tissue, whereas functional MRI scans depict the metabolic and functional state of brain tissues from diverse perspectives. As various magnetic resonance techniques possess both strengths and constraints, this review examines the methodologies and outcomes of multimodal magnetic resonance technology in MCI diagnosis, laying the groundwork for subsequent diagnostic and therapeutic interventions for MCI.
Collapse
Affiliation(s)
- Yu Cui
- Shandong First Medical University, The Second Affiliated Hospital, Department of Neurosurgery, Tai’an, Shandong, China
| | - Chenglong Liu
- Shandong First Medical University, The Second Affiliated Hospital, Department of Radiology, Tai’an, Shandong, China
| | - Ying Wang
- Shandong First Medical University, Department of Scientific Research, Ji’nan, Shandong, China
| | - Hongyan Xie
- Shandong First Medical University, The Second Affiliated Hospital, Department of Neurology, Tai’an, Shandong, China
| |
Collapse
|
24
|
Guo Y, Zhao T, Chu X, Cheng Z. Development of a diagnostic and risk prediction model for Alzheimer's disease through integration of single-cell and bulk transcriptomic analysis of glutamine metabolism. Front Aging Neurosci 2023; 15:1275793. [PMID: 38020758 PMCID: PMC10667556 DOI: 10.3389/fnagi.2023.1275793] [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: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Background In this study, we present a novel system for quantifying glutamine metabolism (GM) to enhance the effectiveness of Alzheimer's disease (AD) diagnosis and risk prediction. Methods Single-cell RNA sequencing (scRNA-seq) analysis was utilized to comprehensively assess the expression patterns of GM. The WGCNA algorithm was applied to investigate the most significant genes related to GM. Subsequently, three machine learning algorithms (Boruta, LASSO, and SVM-RFE) were employed to identify GM-associated characteristic genes and develop a risk model. Patients were divided into high- and low-risk groups based on this model. Moreover, we explored biological properties, distinct signaling pathways, and immunological characteristics of AD patients at different risk levels. Finally, in vitro and in vivo models of AD were constructed to validate the characteristics of the feature genes. Results Both scRNA-seq and bulk transcriptomic analyses revealed increased GM activity in AD patients, specifically in certain cell subsets (pDC, Tem/Effector helper T cells (LTB), and plasma cells). Cells with higher GM scores demonstrated more significant numbers and strengths of interactions with other cell types. The WGCNA algorithm identified 360 genes related to GM, and a risk score was constructed based on nine characteristic genes (ATP13A4, PIK3C2A, CD164, PHF1, CES2, PDGFB, LCOR, TMEM30A, and PLXNA1) identified through multiple machine learning algorithms displayed reliable diagnostic efficacy for AD onset. Nomograms, calibration curves, and decision curve analysis (DCA) based on these characteristic genes provided significant clinical benefits for AD patients. High-risk AD patients exhibited higher levels of immune-related functions and pathways, increased immune cell infiltration, and elevated expressions of immune modulators. RT-qPCR analysis revealed that the majority of the nine characteristic genes were differentially expressed in AD-induced rat neurons. Knocking down PHF1 could protect against neurite loss and alleviate cell injury in AD neurons. In vivo, down-regulation of PHF1 in AD models decreases GM metabolism levels and modulates the immunoinflammatory response in the brain. Conclusion This comprehensive identification of gene expression patterns contributes to a deeper understanding of the underlying pathological mechanisms driving AD pathogenesis. Furthermore, the risk model based on the nine-gene signature offers a promising theoretical foundation for developing individualized treatments for AD patients.
Collapse
Affiliation(s)
- Yan Guo
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingru Zhao
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xi Chu
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenyun Cheng
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
25
|
Ren X, Dong B, Luan Y, Wu Y, Huang Y. Alterations via inter-regional connective relationships in Alzheimer's disease. Front Hum Neurosci 2023; 17:1276994. [PMID: 38021241 PMCID: PMC10672243 DOI: 10.3389/fnhum.2023.1276994] [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: 08/13/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Disruptions in the inter-regional connective correlation within the brain are believed to contribute to memory impairment. To detect these corresponding correlation networks in Alzheimer's disease (AD), we conducted three types of inter-regional correlation analysis, including structural covariance, functional connectivity and group-level independent component analysis (group-ICA). The analyzed data were obtained from the Alzheimer's Disease Neuroimaging Initiative, comprising 52 cognitively normal (CN) participants without subjective memory concerns, 52 individuals with late mild cognitive impairment (LMCI) and 52 patients with AD. We firstly performed vertex-wise cortical thickness analysis to identify brain regions with cortical thinning in AD and LMCI patients using structural MRI data. These regions served as seeds to construct both structural covariance networks and functional connectivity networks for each subject. Additionally, group-ICA was performed on the functional data to identify intrinsic brain networks at the cohort level. Through a comparison of the structural covariance and functional connectivity networks with ICA networks, we identified several inter-regional correlation networks that consistently exhibited abnormal connectivity patterns among AD and LMCI patients. Our findings suggest that reduced inter-regional connectivity is predominantly observed within a subnetwork of the default mode network, which includes the posterior cingulate and precuneus regions, in both AD and LMCI patients. This disruption of connectivity between key nodes within the default mode network provides evidence supporting the hypothesis that impairments in brain networks may contribute to memory deficits in AD and LMCI.
Collapse
Affiliation(s)
- Xiaomei Ren
- College of Electrical Engineering, Sichuan University, Chengdu, China
| | - Bowen Dong
- College of Electrical Engineering, Sichuan University, Chengdu, China
| | - Ying Luan
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ye Wu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Yunzhi Huang
- Institute for AI in Medicine, School of Artificial Intelligence (School of Future Technology), Nanjing University of Information Science and Technology, Nanjing, China
| | | |
Collapse
|
26
|
Xiong S, Hou N, Tang F, Li J, Deng H. Association of cardiometabolic multimorbidity and adherence to a healthy lifestyle with incident dementia: a large prospective cohort study. Diabetol Metab Syndr 2023; 15:208. [PMID: 37876001 PMCID: PMC10594816 DOI: 10.1186/s13098-023-01186-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND The co-occurrence of cardiometabolic diseases (CMDs) is increasingly prevalent and has been associated with an additive risk of dementia in older adults, but the extent to which this risk can be offset by a healthy lifestyle is unknown. We aimed to examine the associations of cardiometabolic multimorbidity and lifestyle with incident dementia and related brain structural changes. METHODS This prospective study extracted health and lifestyle data from 171 538 UK Biobank participants aged 60 years or older without dementia at baseline between 2006 and 2010 and followed up until July 2021, as well as brain structural data in a nested imaging subsample of 11 972 participants. Cardiometabolic multimorbidity was defined as the presence of two or more CMDs among type 2 diabetes, coronary heart disease, stroke, and hypertension. Lifestyle patterns were determined based on 7 modifiable lifestyle factors including smoking, alcohol consumption, physical activity, diet, sleep duration, sedentary behavior, and social contact. RESULTS Over a median follow-up of 12.3 years, 4479 (2.6%) participants developed dementia. The presence of CMDs was dose-dependently associated with an increased risk of dementia. Compared with participants with no CMDs and a favourable lifestyle, those with ≥ 3 CMDs and an unfavourable lifestyle had a five times greater risk of developing dementia (HR 5.33, 95% CI 4.26-6.66). A significant interaction was found between CMD status and lifestyle (Pinteraction=0.001). The absolute difference in incidence rates of dementia per 1000 person years comparing favourable versus unfavourable lifestyle was - 0.65 (95% CI - 1.02 to - 0.27) among participants with no CMDs and - 5.64 (- 8.11 to - 3.17) among participants with ≥ 3 CMDs, corresponding to a HR of 0.71 (0.58-0.88) and 0.42 (0.28-0.63), respectively. In the imaging subsample, a favourable lifestyle was associated with larger total brain, grey matter, and hippocampus volumes across CMD status. CONCLUSION Our findings suggest that adherence to a healthy lifestyle might substantially attenuate dementia risk and adverse brain structural changes associated with cardiometabolic multimorbidity.
Collapse
Affiliation(s)
- Sizheng Xiong
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Ningxin Hou
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feifei Tang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongping Deng
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
27
|
Kim SY, Park J, Choi H, Loeser M, Ryu H, Seo K. Digital Marker for Early Screening of Mild Cognitive Impairment Through Hand and Eye Movement Analysis in Virtual Reality Using Machine Learning: First Validation Study. J Med Internet Res 2023; 25:e48093. [PMID: 37862101 PMCID: PMC10625097 DOI: 10.2196/48093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/07/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND With the global rise in Alzheimer disease (AD), early screening for mild cognitive impairment (MCI), which is a preclinical stage of AD, is of paramount importance. Although biomarkers such as cerebrospinal fluid amyloid level and magnetic resonance imaging have been studied, they have limitations, such as high cost and invasiveness. Digital markers to assess cognitive impairment by analyzing behavioral data collected from digital devices in daily life can be a new alternative. In this context, we developed a "virtual kiosk test" for early screening of MCI by analyzing behavioral data collected when using a kiosk in a virtual environment. OBJECTIVE We aimed to investigate key behavioral features collected from a virtual kiosk test that could distinguish patients with MCI from healthy controls with high statistical significance. Also, we focused on developing a machine learning model capable of early screening of MCI based on these behavioral features. METHODS A total of 51 participants comprising 20 healthy controls and 31 patients with MCI were recruited by 2 neurologists from a university hospital. The participants performed a virtual kiosk test-developed by our group-where we recorded various behavioral data such as hand and eye movements. Based on these time series data, we computed the following 4 behavioral features: hand movement speed, proportion of fixation duration, time to completion, and the number of errors. To compare these behavioral features between healthy controls and patients with MCI, independent-samples 2-tailed t tests were used. Additionally, we used these behavioral features to train and validate a machine learning model for early screening of patients with MCI from healthy controls. RESULTS In the virtual kiosk test, all 4 behavioral features showed statistically significant differences between patients with MCI and healthy controls. Compared with healthy controls, patients with MCI had slower hand movement speed (t49=3.45; P=.004), lower proportion of fixation duration (t49=2.69; P=.04), longer time to completion (t49=-3.44; P=.004), and a greater number of errors (t49=-3.77; P=.001). All 4 features were then used to train a support vector machine to distinguish between healthy controls and patients with MCI. Our machine learning model achieved 93.3% accuracy, 100% sensitivity, 83.3% specificity, 90% precision, and 94.7% F1-score. CONCLUSIONS Our research preliminarily suggests that analyzing hand and eye movements in the virtual kiosk test holds potential as a digital marker for early screening of MCI. In contrast to conventional biomarkers, this digital marker in virtual reality is advantageous as it can collect ecologically valid data at an affordable cost and in a short period (5-15 minutes), making it a suitable means for early screening of MCI. We call for further studies to confirm the reliability and validity of this approach.
Collapse
Affiliation(s)
- Se Young Kim
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hojin Choi
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Martin Loeser
- Department of Computer Science, Electrical Engineering and Mechatronics, ZHAW Zurich University of Applied Sciences, Winterthur, Switzerland
| | - Hokyoung Ryu
- Graduate School of Technology and Innovation Management, Hanyang University, Seoul, Republic of Korea
| | - Kyoungwon Seo
- Department of Applied Artificial Intelligence, Seoul National University of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
28
|
Chouliaras L, O'Brien JT. The use of neuroimaging techniques in the early and differential diagnosis of dementia. Mol Psychiatry 2023; 28:4084-4097. [PMID: 37608222 PMCID: PMC10827668 DOI: 10.1038/s41380-023-02215-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
Dementia is a leading cause of disability and death worldwide. At present there is no disease modifying treatment for any of the most common types of dementia such as Alzheimer's disease (AD), Vascular dementia, Lewy Body Dementia (LBD) and Frontotemporal dementia (FTD). Early and accurate diagnosis of dementia subtype is critical to improving clinical care and developing better treatments. Structural and molecular imaging has contributed to a better understanding of the pathophysiology of neurodegenerative dementias and is increasingly being adopted into clinical practice for early and accurate diagnosis. In this review we summarise the contribution imaging has made with particular focus on multimodal magnetic resonance imaging (MRI) and positron emission tomography imaging (PET). Structural MRI is widely used in clinical practice and can help exclude reversible causes of memory problems but has relatively low sensitivity for the early and differential diagnosis of dementia subtypes. 18F-fluorodeoxyglucose PET has high sensitivity and specificity for AD and FTD, while PET with ligands for amyloid and tau can improve the differential diagnosis of AD and non-AD dementias, including recognition at prodromal stages. Dopaminergic imaging can assist with the diagnosis of LBD. The lack of a validated tracer for α-synuclein or TAR DNA-binding protein 43 (TDP-43) imaging remain notable gaps, though work is ongoing. Emerging PET tracers such as 11C-UCB-J for synaptic imaging may be sensitive early markers but overall larger longitudinal multi-centre cross diagnostic imaging studies are needed.
Collapse
Affiliation(s)
- Leonidas Chouliaras
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Specialist Dementia and Frailty Service, Essex Partnership University NHS Foundation Trust, St Margaret's Hospital, Epping, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
29
|
Zhang L, Wu J, Wang L, Wang L, Steffens DC, Qiu S, Potter GG, Liu M. Brain Anatomy-Guided MRI Analysis for Assessing Clinical Progression of Cognitive Impairment with Structural MRI. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION : MICCAI ... INTERNATIONAL CONFERENCE ON MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION 2023; 14227:109-119. [PMID: 38390033 PMCID: PMC10883230 DOI: 10.1007/978-3-031-43993-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Brain structural MRI has been widely used for assessing future progression of cognitive impairment (CI) based on learning-based methods. Previous studies generally suffer from the limited number of labeled training data, while there exists a huge amount of MRIs in large-scale public databases. Even without task-specific label information, brain anatomical structures provided by these MRIs can be used to boost learning performance intuitively. Unfortunately, existing research seldom takes advantage of such brain anatomy prior. To this end, this paper proposes a brain anatomy-guided representation (BAR) learning framework for assessing the clinical progression of cognitive impairment with T1-weighted MRIs. The BAR consists of a pretext model and a downstream model, with a shared brain anatomy-guided encoder for MRI feature extraction. The pretext model also contains a decoder for brain tissue segmentation, while the downstream model relies on a predictor for classification. We first train the pretext model through a brain tissue segmentation task on 9,544 auxiliary T1-weighted MRIs, yielding a generalizable encoder. The downstream model with the learned encoder is further fine-tuned on target MRIs for prediction tasks. We validate the proposed BAR on two CI-related studies with a total of 391 subjects with T1-weighted MRIs. Experimental results suggest that the BAR outperforms several state-of-the-art (SOTA) methods. The source code and pre-trained models are available at https://github.com/goodaycoder/BAR.
Collapse
Affiliation(s)
- Lintao Zhang
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinjian Wu
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lihong Wang
- Department of Psychiatry, University of Connecticut School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Li Wang
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David C Steffens
- Department of Psychiatry, University of Connecticut School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Shijun Qiu
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guy G Potter
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Mingxia Liu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
30
|
Richter N, Brand S, Nellessen N, Dronse J, Gramespacher H, Schmieschek MHT, Fink GR, Kukolja J, Onur OA. Fine-grained age-matching improves atrophy-based detection of mild cognitive impairment more than amyloid-negative reference subjects. Neuroimage Clin 2023; 40:103508. [PMID: 37717383 PMCID: PMC10514218 DOI: 10.1016/j.nicl.2023.103508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
INTRODUCTION In clinical practice, differentiating between age-related gray matter (GM) atrophy and neurodegeneration-related atrophy at early disease stages, such as mild cognitive impairment (MCI), remains challenging. We hypothesized that fined-grained adjustment for age effects and using amyloid-negative reference subjects could increase classification accuracy. METHODS T1-weighted magnetic resonance imaging (MRI) data of 131 cognitively normal (CN) individuals and 91 patients with MCI from the Alzheimer's disease neuroimaging initiative (ADNI) characterized concerning amyloid status, as well as 19 CN individuals and 19 MCI patients from an independent validation sample were segmented, spatially normalized and analyzed in the framework of voxel-based morphometry (VBM). For each participant, statistical maps of GM atrophy were computed as the deviation from the GM of CN reference groups at the voxel level. CN reference groups composed with different degrees of age-matching, and mixed and strictly amyloid-negative CN reference groups were examined regarding their effect on the accuracy in distinguishing between CN and MCI. Furthermore, the effects of spatial smoothing and atrophy threshold were assessed. RESULTS Approaches with a specific reference group for each age significantly outperformed all other age-adjustment strategies with a maximum area under the curve of 1.0 in the ADNI sample and 0.985 in the validation sample. Accounting for age in a regression-based approach improved classification accuracy over that of a single CN reference group in the age range of the patient sample. Using strictly amyloid-negative reference groups improved classification accuracy only when age was not considered. CONCLUSION Our results demonstrate that VBM can differentiate between age-related and MCI-associated atrophy with high accuracy. Crucially, age-specific reference groups significantly increased accuracy, more so than regression-based approaches and using amyloid-negative reference groups.
Collapse
Affiliation(s)
- Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany.
| | - Stefanie Brand
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Nils Nellessen
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany; Department of Neurology and Clinical Neurophysiology, Helios University Hospital Wuppertal, 42283 Wuppertal, Germany; Faculty of Health, Witten/Herdecke University, 58448 Witten, Germany
| | - Julian Dronse
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Hannes Gramespacher
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Maximilian H T Schmieschek
- Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Gereon R Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Juraj Kukolja
- Department of Neurology and Clinical Neurophysiology, Helios University Hospital Wuppertal, 42283 Wuppertal, Germany; Faculty of Health, Witten/Herdecke University, 58448 Witten, Germany
| | - Oezguer A Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Department of Neurology, University Hospital Cologne and Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
31
|
Simic T, Desjardins MÈ, Courson M, Bedetti C, Houzé B, Brambati SM. Treatment-induced neuroplasticity after anomia therapy in post-stroke aphasia: A systematic review of neuroimaging studies. BRAIN AND LANGUAGE 2023; 244:105300. [PMID: 37633250 DOI: 10.1016/j.bandl.2023.105300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 05/02/2023] [Accepted: 06/21/2023] [Indexed: 08/28/2023]
Abstract
We systematically reviewed the literature on neural changes following anomia treatment post-stroke. We conducted electronic searches of CINAHL, Cochrane Trials, Embase, Ovid MEDLINE, MEDLINE-in-Process and PsycINFO databases; two independent raters assessed all abstracts and full texts. Accepted studies reported original data on adults with post-stroke aphasia, who received behavioural treatment for anomia, and magnetic resonance brain imaging (MRI) pre- and post-treatment. Search results yielded 2481 citations; 33 studies were accepted. Most studies employed functional MRI and the quality of reporting neuroimaging methodology was variable, particularly for pre-processing steps and statistical analyses. The most methodologically robust data were synthesized, focusing on pre- versus post-treatment contrasts. Studies more commonly reported increases (versus decreases) in activation following naming therapy, primarily in the left supramarginal gyrus, and left/bilateral precunei. Our findings highlight the methodological heterogeneity across MRI studies, and the paucity of robust evidence demonstrating direct links between brain and behaviour in anomia rehabilitation.
Collapse
Affiliation(s)
- Tijana Simic
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), 4545 Queen Mary R.d., Montréal, QC H3W 1W4, Canada; Département de Psychologie, Université de Montréal, 90 Vincent-d'Indy Avenue, Montréal, QC H2V 2S9, Canada; Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 Boul Gouin O, Montréal, QC H4J 1C5, Canada.
| | - Marie-Ève Desjardins
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), 4545 Queen Mary R.d., Montréal, QC H3W 1W4, Canada; Département de Psychologie, Université de Montréal, 90 Vincent-d'Indy Avenue, Montréal, QC H2V 2S9, Canada; Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 Boul Gouin O, Montréal, QC H4J 1C5, Canada
| | - Melody Courson
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), 4545 Queen Mary R.d., Montréal, QC H3W 1W4, Canada; Département de Psychologie, Université de Montréal, 90 Vincent-d'Indy Avenue, Montréal, QC H2V 2S9, Canada
| | - Christophe Bedetti
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), 4545 Queen Mary R.d., Montréal, QC H3W 1W4, Canada
| | - Bérengère Houzé
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), 4545 Queen Mary R.d., Montréal, QC H3W 1W4, Canada; Département de Psychologie, Université de Montréal, 90 Vincent-d'Indy Avenue, Montréal, QC H2V 2S9, Canada
| | - Simona Maria Brambati
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal (CRIUGM), 4545 Queen Mary R.d., Montréal, QC H3W 1W4, Canada; Département de Psychologie, Université de Montréal, 90 Vincent-d'Indy Avenue, Montréal, QC H2V 2S9, Canada; Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 Boul Gouin O, Montréal, QC H4J 1C5, Canada
| |
Collapse
|
32
|
Yosypyshyn D, Kučikienė D, Ramakers I, Schulz JB, Reetz K, Costa AS. Clinical characteristics of patients with suspected Alzheimer's disease within a CSF Aß-ratio grey zone. Neurol Res Pract 2023; 5:40. [PMID: 37533121 PMCID: PMC10398972 DOI: 10.1186/s42466-023-00262-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND The AT(N) research framework for Alzheimer's disease (AD) remains unclear on how to best deal with borderline cases. Our aim was to characterise patients with suspected AD with a borderline Aß1-42/Aß1-40 ratio in cerebrospinal fluid. METHODS We analysed retrospective data from two cohorts (memory clinic cohort and ADNI) of patients (n = 63) with an Aß1-42/Aß1-40 ratio within a predefined borderline area-Q1 above the validated cut-off value(grey zone). We compared demographic, clinical, neuropsychological and neuroimaging features between grey zone patients and patients with low Aß1-42 (normal Aß ratio but pathological Aß1-42, n = 42) and patients with AD (pathological Aß, P-Tau, und T-Tau, n = 80). RESULTS Patients had mild cognitive impairment or mild dementia and a median age of 72 years. Demographic and general clinical characteristics did not differ between the groups. Patients in the grey zone group were the least impaired in cognition. However, they overlapped with the low Aß1-42 group in verbal episodic memory performance, especially in delayed recall and recognition. The grey zone group had less severe medial temporal atrophy, but mild posterior atrophy and mild white matter hyperintensities, similar to the low Aß1-42 group. CONCLUSIONS Patients in the Aß ratio grey zone were less impaired, but showed clinical overlap with patients on the AD continuum. These borderline patients may be at an earlier disease stage. Assuming an increased risk of AD and progressive cognitive decline, careful consideration of clinical follow-up is recommended when using dichotomous approaches to classify Aß status.
Collapse
Affiliation(s)
- Dariia Yosypyshyn
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Domantė Kučikienė
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Inez Ramakers
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Jörg B Schulz
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany
| | - Kathrin Reetz
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany.
| | - Ana Sofia Costa
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
- JARA Institute Molecular Neuroscience and Neuroimaging, RWTH Aachen & Forschungszentrum Jülich, Aachen, Germany
| |
Collapse
|
33
|
Ismail Z, Leon R, Creese B, Ballard C, Robert P, Smith EE. Optimizing detection of Alzheimer's disease in mild cognitive impairment: a 4-year biomarker study of mild behavioral impairment in ADNI and MEMENTO. Mol Neurodegener 2023; 18:50. [PMID: 37516848 PMCID: PMC10386685 DOI: 10.1186/s13024-023-00631-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/05/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Disease-modifying drug use necessitates better Alzheimer disease (AD) detection. Mild cognitive impairment (MCI) leverages cognitive decline to identify the risk group; similarly, mild behavioral impairment (MBI) leverages behavioral change. Adding MBI to MCI improves dementia prognostication over conventional approaches of incorporating neuropsychiatric symptoms (NPS). Here, to determine if adding MBI would better identify AD, we interrogated associations between MBI in MCI, and cerebrospinal fluid biomarkers [β-amyloid (Aβ), phosphorylated-tau (p-tau), and total-tau (tau)-ATN], cross-sectionally and longitudinally. METHODS Data were from two independent referral-based cohorts, ADNI (mean[SD] follow-up 3.14[1.07] years) and MEMENTO (4.25[1.40] years), collected 2003-2021. Exposure was based on three-group stratification: 1) NPS meeting MBI criteria; 2) conventionally measured NPS (NPSnotMBI); and 3) noNPS. Cohorts were analyzed separately for: 1) cross-sectional associations between NPS status and ATN biomarkers (linear regressions); 2) 4-year longitudinal repeated-measures associations of MBI and NPSnotMBI with ATN biomarkers (hierarchical linear mixed-effects models-LMEs); and 3) rates of incident dementia (Cox proportional hazards regressions). RESULTS Of 510 MCI participants, 352 were from ADNI (43.5% females; mean [SD] age, 71.68 [7.40] years), and 158 from MEMENTO (46.2% females; 68.98 [8.18] years). In ADNI, MBI was associated with lower Aβ42 (standardized β [95%CI], -5.52% [-10.48-(-0.29)%]; p = 0.039), and Aβ42/40 (p = 0.01); higher p-tau (9.67% [3.96-15.70%]; p = 0.001), t-tau (7.71% [2.70-12.97%]; p = 0.002), p-tau/Aβ42 (p < 0.001), and t-tau/Aβ42 (p = 0.001). NPSnotMBI was associated only with lower Aβ42/40 (p = 0.045). LMEs revealed a similar 4-year AD-specific biomarker profile for MBI, with NPSnotMBI associated only with higher t-tau. MBI had a greater rate of incident dementia (HR [95%CI], 3.50 [1.99-6.17; p < 0.001). NPSnotMBI did not differ from noNPS (HR 0.96 [0.49-1.89]; p = 0.916). In MEMENTO, MBI demonstrated a similar magnitude and direction of effect for all biomarkers, but with a greater reduction in Aβ40. HR for incident dementia was 3.93 (p = 0.004) in MBI, and 1.83 (p = 0.266) in NPSnotMBI. Of MBI progressors to dementia, 81% developed AD dementia. CONCLUSIONS These findings support a biological basis for NPS that meet MBI criteria, the continued inclusion of MBI in NIA-AA ATN clinical staging, and the utility of MBI criteria to improve identification of patients for enrollment in disease-modifying drug trials or for clinical care.
Collapse
Affiliation(s)
- Zahinoor Ismail
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada.
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada.
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada.
- O'Brien Institute for Public Health, Cumming School of Medicine, University of Calgary, 3280 Hospital Dr NW, Calgary, AB, T2N 4Z6, Canada.
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, B3183, Exeter, EX1 2HZ, UK.
| | - Rebeca Leon
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - Byron Creese
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, B3183, Exeter, EX1 2HZ, UK
| | - Clive Ballard
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, B3183, Exeter, EX1 2HZ, UK
| | | | - Eric E Smith
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
34
|
Yi F, Yang H, Chen D, Qin Y, Han H, Cui J, Bai W, Ma Y, Zhang R, Yu H. XGBoost-SHAP-based interpretable diagnostic framework for alzheimer's disease. BMC Med Inform Decis Mak 2023; 23:137. [PMID: 37491248 PMCID: PMC10369804 DOI: 10.1186/s12911-023-02238-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 07/13/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Due to the class imbalance issue faced when Alzheimer's disease (AD) develops from normal cognition (NC) to mild cognitive impairment (MCI), present clinical practice is met with challenges regarding the auxiliary diagnosis of AD using machine learning (ML). This leads to low diagnosis performance. We aimed to construct an interpretable framework, extreme gradient boosting-Shapley additive explanations (XGBoost-SHAP), to handle the imbalance among different AD progression statuses at the algorithmic level. We also sought to achieve multiclassification of NC, MCI, and AD. METHODS We obtained patient data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, including clinical information, neuropsychological test results, neuroimaging-derived biomarkers, and APOE-ε4 gene statuses. First, three feature selection algorithms were applied, and they were then included in the XGBoost algorithm. Due to the imbalance among the three classes, we changed the sample weight distribution to achieve multiclassification of NC, MCI, and AD. Then, the SHAP method was linked to XGBoost to form an interpretable framework. This framework utilized attribution ideas that quantified the impacts of model predictions into numerical values and analysed them based on their directions and sizes. Subsequently, the top 10 features (optimal subset) were used to simplify the clinical decision-making process, and their performance was compared with that of a random forest (RF), Bagging, AdaBoost, and a naive Bayes (NB) classifier. Finally, the National Alzheimer's Coordinating Center (NACC) dataset was employed to assess the impact path consistency of the features within the optimal subset. RESULTS Compared to the RF, Bagging, AdaBoost, NB and XGBoost (unweighted), the interpretable framework had higher classification performance with accuracy improvements of 0.74%, 0.74%, 1.46%, 13.18%, and 0.83%, respectively. The framework achieved high sensitivity (81.21%/74.85%), specificity (92.18%/89.86%), accuracy (87.57%/80.52%), area under the receiver operating characteristic curve (AUC) (0.91/0.88), positive clinical utility index (0.71/0.56), and negative clinical utility index (0.75/0.68) on the ADNI and NACC datasets, respectively. In the ADNI dataset, the top 10 features were found to have varying associations with the risk of AD onset based on their SHAP values. Specifically, the higher SHAP values of CDRSB, ADAS13, ADAS11, ventricle volume, ADASQ4, and FAQ were associated with higher risks of AD onset. Conversely, the higher SHAP values of LDELTOTAL, mPACCdigit, RAVLT_immediate, and MMSE were associated with lower risks of AD onset. Similar results were found for the NACC dataset. CONCLUSIONS The proposed interpretable framework contributes to achieving excellent performance in imbalanced AD multiclassification tasks and provides scientific guidance (optimal subset) for clinical decision-making, thereby facilitating disease management and offering new research ideas for optimizing AD prevention and treatment programs.
Collapse
Affiliation(s)
- Fuliang Yi
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Hui Yang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Durong Chen
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Yao Qin
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Hongjuan Han
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Jing Cui
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Wenlin Bai
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Yifei Ma
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Rong Zhang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
| | - Hongmei Yu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, 56 South XinJian Road, Taiyuan, 030001 P.R. China
- Shanxi Provincial Key Laboratory of Major Diseases Risk Assessment, Taiyuan, China
| |
Collapse
|
35
|
Dang C, Wang Y, Li Q, Lu Y. Neuroimaging modalities in the detection of Alzheimer's disease-associated biomarkers. PSYCHORADIOLOGY 2023; 3:kkad009. [PMID: 38666112 PMCID: PMC11003434 DOI: 10.1093/psyrad/kkad009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/04/2023] [Accepted: 06/20/2023] [Indexed: 04/28/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Neuropathological changes in AD patients occur up to 10-20 years before the emergence of clinical symptoms. Specific diagnosis and appropriate intervention strategies are crucial during the phase of mild cognitive impairment (MCI) and AD. The detection of biomarkers has emerged as a promising tool for tracking the efficacy of potential therapies, making an early disease diagnosis, and prejudging treatment prognosis. Specifically, multiple neuroimaging modalities, including magnetic resonance imaging (MRI), positron emission tomography, optical imaging, and single photon emission-computed tomography, have provided a few potential biomarkers for clinical application. The MRI modalities described in this review include structural MRI, functional MRI, diffusion tensor imaging, magnetic resonance spectroscopy, and arterial spin labelling. These techniques allow the detection of presymptomatic diagnostic biomarkers in the brains of cognitively normal elderly people and might also be used to monitor AD disease progression after the onset of clinical symptoms. This review highlights potential biomarkers, merits, and demerits of different neuroimaging modalities and their clinical value in MCI and AD patients. Further studies are necessary to explore more biomarkers and overcome the limitations of multiple neuroimaging modalities for inclusion in diagnostic criteria for AD.
Collapse
Affiliation(s)
- Chun Dang
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanchao Wang
- Department of Neurology, Chifeng University of Affiliated Hospital, Chifeng 024000, China
| | - Qian Li
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Yaoheng Lu
- Department of General Surgery, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Chengdu 610000, China
| |
Collapse
|
36
|
Aramadaka S, Mannam R, Sankara Narayanan R, Bansal A, Yanamaladoddi VR, Sarvepalli SS, Vemula SL. Neuroimaging in Alzheimer's Disease for Early Diagnosis: A Comprehensive Review. Cureus 2023; 15:e38544. [PMID: 37273363 PMCID: PMC10239271 DOI: 10.7759/cureus.38544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, affecting roughly half of those over the age of 85. We briefly discussed the risk factors, epidemiology, and treatment options for AD. The development of therapeutic therapies operating very early in the disease cascade has been spurred by the realization that the disease process begins at least a decade or more before the manifestation of symptoms. Thus, the clinical significance of early diagnosis was emphasized. Using various keywords, a literature search was carried out using PubMed and other databases. For inclusion, pertinent articles were chosen and reviewed. This article has reviewed different neuroimaging techniques that are considered advanced tools to aid in establishing a diagnosis and highlighted the advantages as well as disadvantages of those techniques. Besides, the prevalence of several in vivo biomarkers aided in discriminating affected individuals from healthy controls in the early stages of the disease. Each imaging method has its advantages and disadvantages, hence no single imaging approach can be the optimum modality for diagnosis. This article also commented on a better approach to using these techniques to increase the likelihood of an early diagnosis.
Collapse
Affiliation(s)
| | - Raam Mannam
- Research, Narayana Medical College, Nellore, IND
| | | | - Arpit Bansal
- Research, Narayana Medical College, Nellore, IND
| | | | | | | |
Collapse
|
37
|
Optimizing Primary Healthcare in Hong Kong: Strategies for the Successful Integration of Radiology Services. Cureus 2023; 15:e37022. [PMID: 37016673 PMCID: PMC10066850 DOI: 10.7759/cureus.37022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2023] [Indexed: 04/04/2023] Open
Abstract
The primary healthcare system in Hong Kong plays a crucial role in addressing the healthcare needs of its population. However, the integration of radiology services into primary care settings has not been fully realized, and there is significant potential for improvement. Incorporating radiology services into primary healthcare can enhance patient care, promote cost-effectiveness, and increase the overall efficiency of the healthcare system by enabling earlier diagnosis and intervention for various health conditions. To successfully integrate radiology services, key strategies include the establishment of public-private partnerships, the adoption of teleradiology and telemedicine services, the development of comprehensive regulatory and policy frameworks, and the exploration of innovative financial models and incentives. By embracing these strategies, Hong Kong can optimize its primary healthcare system and ensure more equitable, effective care for its population.
Collapse
|
38
|
Vik A, Kociński M, Rye I, Lundervold AJ, Lundervold AS. Functional activity level reported by an informant is an early predictor of Alzheimer's disease. BMC Geriatr 2023; 23:205. [PMID: 37003981 PMCID: PMC10067216 DOI: 10.1186/s12877-023-03849-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 02/24/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Loss of autonomy in day-to-day functioning is one of the feared outcomes of Alzheimer's disease (AD), and relatives may have been worried by subtle behavioral changes in ordinary life situations long before these changes are given medical attention. In the present study, we ask if such subtle changes should be given weight as an early predictor of a future AD diagnosis. METHODS Longitudinal data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were used to define a group of adults with a mild cognitive impairment (MCI) diagnosis remaining stable across several visits (sMCI, n=360; 55-91 years at baseline), and a group of adults who over time converted from having an MCI diagnosis to an AD diagnosis (cAD, n=320; 55-88 years at baseline). Eleven features were used as input in a Random Forest (RF) binary classifier (sMCI vs. cAD) model. This model was tested on an unseen holdout part of the dataset, and further explored by three different permutation-driven importance estimates and a comprehensive post hoc machine learning exploration. RESULTS The results consistently showed that measures of daily life functioning, verbal memory function, and a volume measure of hippocampus were the most important predictors of conversion from an MCI to an AD diagnosis. Results from the RF classification model showed a prediction accuracy of around 70% in the test set. Importantly, the post hoc analyses showed that even subtle changes in everyday functioning noticed by a close informant put MCI patients at increased risk for being on a path toward the major cognitive impairment of an AD diagnosis. CONCLUSION The results showed that even subtle changes in everyday functioning should be noticed when reported by relatives in a clinical evaluation of patients with MCI. Information of these changes should also be included in future longitudinal studies to investigate different pathways from normal cognitive aging to the cognitive decline characterizing different stages of AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Alexandra Vik
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Marek Kociński
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ingrid Rye
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Astri J Lundervold
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| | - Alexander S Lundervold
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway.
- Department of Computer Science, Electrical Engineering and Mathematical Sciences, Western Norway University of Applied Sciences, Bergen, Norway.
| |
Collapse
|
39
|
Shi R, Sheng C, Jin S, Zhang Q, Zhang S, Zhang L, Ding C, Wang L, Wang L, Han Y, Jiang J. Generative adversarial network constrained multiple loss autoencoder: A deep learning-based individual atrophy detection for Alzheimer's disease and mild cognitive impairment. Hum Brain Mapp 2023; 44:1129-1146. [PMID: 36394351 PMCID: PMC9875916 DOI: 10.1002/hbm.26146] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/02/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Exploring individual brain atrophy patterns is of great value in precision medicine for Alzheimer's disease (AD) and mild cognitive impairment (MCI). However, the current individual brain atrophy detection models are deficient. Here, we proposed a framework called generative adversarial network constrained multiple loss autoencoder (GANCMLAE) for precisely depicting individual atrophy patterns. The GANCMLAE model was trained using normal controls (NCs) from the Alzheimer's Disease Neuroimaging Initiative cohort, and the Xuanwu cohort was employed to validate the robustness of the model. The potential of the model for identifying different atrophy patterns of MCI subtypes was also assessed. Furthermore, the clinical application potential of the GANCMLAE model was investigated. The results showed that the model can achieve good image reconstruction performance on the structural similarity index measure (0.929 ± 0.003), peak signal-to-noise ratio (31.04 ± 0.09), and mean squared error (0.0014 ± 0.0001) with less latent loss in the Xuanwu cohort. The individual atrophy patterns extracted from this model are more precise in reflecting the clinical symptoms of MCI subtypes. The individual atrophy patterns exhibit a better discriminative power in identifying patients with AD and MCI from NCs than those of the t-test model, with areas under the receiver operating characteristic curve of 0.867 (95%: 0.837-0.897) and 0.752 (95%: 0.71-0.790), respectively. Similar findings are also reported in the AD and MCI subgroups. In conclusion, the GANCMLAE model can serve as an effective tool for individualised atrophy detection.
Collapse
Affiliation(s)
- Rong Shi
- School of Information and Communication EngineeringShanghai UniversityShanghaiChina
| | - Can Sheng
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Shichen Jin
- School of Information and Communication EngineeringShanghai UniversityShanghaiChina
| | - Qi Zhang
- School of Information and Communication EngineeringShanghai UniversityShanghaiChina
| | - Shuoyan Zhang
- School of Information and Communication EngineeringShanghai UniversityShanghaiChina
| | - Liang Zhang
- Key Laboratory of Biomedical Engineering of Hainan ProvinceSchool of Biomedical Engineering, Hainan UniversityHaikouChina
| | - Changchang Ding
- School of Information and Communication EngineeringShanghai UniversityShanghaiChina
| | - Luyao Wang
- School of Information and Communication EngineeringShanghai UniversityShanghaiChina
| | - Lei Wang
- College of Computing and InformaticsDrexel UniversityPhiladelphiaPennsylvaniaUSA
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- Key Laboratory of Biomedical Engineering of Hainan ProvinceSchool of Biomedical Engineering, Hainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
| | - Jiehui Jiang
- Institute of Biomedical EngineeringSchool of Life Science, Shanghai UniversityShanghaiChina
| |
Collapse
|
40
|
Zhou B, Zhao Q, Kojima S, Ding D, Higashide S, Fukushima M, Hong Z. Early Detection of Dementia using Risk Classification in MCI: Outcomes of Shanghai Mild Cognitive Impairment Cohort Study. Curr Alzheimer Res 2023; 20:431-439. [PMID: 37711110 DOI: 10.2174/1567205020666230914161034] [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: 02/27/2023] [Revised: 07/27/2023] [Accepted: 08/07/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION The purpose of this study is to identify the risk factors and risk classification associated with the conversion from mild cognitive impairment (MCI) to Alzheimer's disease (AD) dementia to facilitate early intervention and the design of clinical trials for AD. METHODS The study comprised a prospective cohort study of 400 subjects with MCI who had annual follow-ups for 3 years. RESULTS During an average follow-up period of 3.5 years, 109 subjects were diagnosed with all cause of dementia, of whom 104 subjects converted to Alzheimer's dementia and 5 subjects converted to other types of dementia. The cumulative conversion rate was 5.5% (95% CI: 3.4, 8.6), 16.3% (95% CI: 12.9, 21.1), and 31.0% (95% CI: 25.4, 36.5) in each of the first 3 follow-up years, respectively. The factors associated with a greater risk of conversion from MCI to AD included smoking status, ApoE4 carrier status, right hippocampal volume (rt. HV), left temporal lobe volume, and scores on the Revised Chinese version of the Alzheimer's Disease Assessment Scale-Cognitive Subscale 13 (ADAS-Cog-C). The risk classification of the ADAS-Cog-C or Preclinical Alzheimer Cognitive Composite (PACC) score combined with the rt. HV showed a conversion difference among the groups at every annual follow-up. CONCLUSION A simple risk classification using the rt. HV and neuropsychological test scores, including those from the ADAS-Cog-C and PACC, could be a practicable and efficient approach to indentify individuals at risk of all-cause dementia.
Collapse
Affiliation(s)
- Bin Zhou
- Foundation of Learning Health Society Institute, Nagoya, Japan
| | - Qianhua Zhao
- Institute of Neurology, Huashan Hospital Fudan University, Shanghai, China
| | - Shinsuke Kojima
- Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Ding Ding
- Institute of Neurology, Huashan Hospital Fudan University, Shanghai, China
| | - Satoshi Higashide
- Translational Research Informatics Center, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | | | - Zhen Hong
- Institute of Neurology, Huashan Hospital Fudan University, Shanghai, China
| |
Collapse
|
41
|
Li KR, Wu AG, Tang Y, He XP, Yu CL, Wu JM, Hu GQ, Yu L. The Key Role of Magnetic Resonance Imaging in the Detection of Neurodegenerative Diseases-Associated Biomarkers: A Review. Mol Neurobiol 2022; 59:5935-5954. [PMID: 35829831 DOI: 10.1007/s12035-022-02944-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 06/28/2022] [Indexed: 11/30/2022]
Abstract
Neurodegenerative diseases (NDs), including chronic disease such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis, and acute diseases like traumatic brain injury and ischemic stroke are characterized by progressive degeneration, brain tissue damage and loss of neurons, accompanied by behavioral and cognitive dysfunctions. So far, there are no complete cures for NDs; thus, early and timely diagnoses are essential and beneficial to patients' treatment. Magnetic resonance imaging (MRI) has become one of the advanced medical imaging techniques widely used in the clinical examination of NDs due to its non-invasive diagnostic value. In this review, research published in English in current decade from PubMed electronic database on the use of MRI to detect specific biomarkers of NDs was collected, summarized, and discussed, which provides valuable suggestions for the early diagnosis, prevention, and treatment of NDs in the clinic.
Collapse
Affiliation(s)
- Ke-Ru Li
- Department of Human Anatomy, School of Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
- Department of Radiology, Chongqing University Fuling Hospital, Chongqing, 408000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
| | - Xiao-Peng He
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chong-Lin Yu
- Department of Human Anatomy, School of Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Guang-Qiang Hu
- Department of Human Anatomy, School of Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, 646000, China.
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Department of Chemistry, School of Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
42
|
Custodio N, Malaga M, Chambergo-Michilot D, Montesinos R, Moron E, Vences MA, Huilca JC, Lira D, Failoc-Rojas VE, Diaz MM. Combining visual rating scales to identify prodromal Alzheimer's disease and Alzheimer's disease dementia in a population from a low and middle-income country. Front Neurol 2022; 13:962192. [PMID: 36119675 PMCID: PMC9477244 DOI: 10.3389/fneur.2022.962192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Background Many low- and middle-income countries, including Latin America, lack access to biomarkers for the diagnosis of prodromal Alzheimer's Disease (AD; mild cognitive impairment due to AD) and AD dementia. MRI visual rating scales may serve as an ancillary diagnostic tool for identifying prodromal AD or AD in Latin America. We investigated the ability of brain MRI visual rating scales to distinguish between cognitively healthy controls, prodromal AD and AD. Methods A cross-sectional study was conducted from a multidisciplinary neurology clinic in Lima, Peru using neuropsychological assessments, brain MRI and cerebrospinal fluid amyloid and tau levels. Medial temporal lobe atrophy (MTA), posterior atrophy (PA), white matter hyperintensity (WMH), and MTA+PA composite MRI scores were compared. Sensitivity, specificity, and area under the curve (AUC) were determined. Results Fifty-three patients with prodromal AD, 69 with AD, and 63 cognitively healthy elderly individuals were enrolled. The median age was 75 (8) and 42.7% were men. Neither sex, mean age, nor years of education were significantly different between groups. The MTA was higher in patients with AD (p < 0.0001) compared with prodromal AD and controls, and MTA scores adjusted by age range (p < 0.0001) and PA scores (p < 0.0001) were each significantly associated with AD diagnosis (p < 0.0001) but not the WMH score (p=0.426). The MTA had better performance among ages <75 years (AUC 0.90 [0.85-0.95]), while adjusted MTA+PA scores performed better among ages>75 years (AUC 0.85 [0.79-0.92]). For AD diagnosis, MTA+PA had the best performance (AUC 1.00) for all age groups. Conclusions Combining MTA and PA scores demonstrates greater discriminative ability to differentiate controls from prodromal AD and AD, highlighting the diagnostic value of visual rating scales in daily clinical practice, particularly in Latin America where access to advanced neuroimaging and CSF biomarkers is limited in the clinical setting.
Collapse
Affiliation(s)
- Nilton Custodio
- Servicio de Neurología, Instituto Peruano de Neurociencias, Lima, Peru
- Unidad de diagnóstico de deterioro cognitivo y prevención de demencia, Instituto Peruano de Neurociencias, Lima, Peru
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Lima, Peru
| | - Marco Malaga
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
- San Martin de Porres University, Lima, Peru
| | - Diego Chambergo-Michilot
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
- Universidad Científica del Sur, Lima, Peru
| | - Rosa Montesinos
- Unidad de diagnóstico de deterioro cognitivo y prevención de demencia, Instituto Peruano de Neurociencias, Lima, Peru
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
| | - Elizabeth Moron
- Departamento de Radiología, Hospital Nacional Edgardo Rebagliati Martins, EsSalud, Lima, Peru
- Servicio de Radiología, Centro de Diagnóstico por Imagen-DPI, Lima, Peru
| | - Miguel A. Vences
- Unidad de diagnóstico de deterioro cognitivo y prevención de demencia, Instituto Peruano de Neurociencias, Lima, Peru
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
- Departamento de Neurología, Hospital Nacional Edgardo Rebagliati Martins, EsSalud, Lima, Peru
| | - José Carlos Huilca
- Unidad de diagnóstico de deterioro cognitivo y prevención de demencia, Instituto Peruano de Neurociencias, Lima, Peru
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
- Servicio de Neurología, Hospital Guillermo Kaelin de La Fuente, Lima, Peru
| | - David Lira
- Servicio de Neurología, Instituto Peruano de Neurociencias, Lima, Peru
- Unidad de diagnóstico de deterioro cognitivo y prevención de demencia, Instituto Peruano de Neurociencias, Lima, Peru
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
| | - Virgilio E. Failoc-Rojas
- Unidad de Investigación, Instituto Peruano de Neurociencias, Lima, Peru
- Centro de Investigación en Medicina Traslacional, Universidad Privada Norbert Wiener, Lima, Peru
| | - Monica M. Diaz
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
43
|
Schilling LP, Balthazar MLF, Radanovic M, Forlenza OV, Silagi ML, Smid J, Barbosa BJAP, Frota NAF, de Souza LC, Vale FAC, Caramelli P, Bertolucci PHF, Chaves MLF, Brucki SMD, Damasceno BP, Nitrini R. Diagnosis of Alzheimer's disease: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022; 16:25-39. [PMID: 36533157 PMCID: PMC9745995 DOI: 10.1590/1980-5764-dn-2022-s102pt] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/22/2021] [Accepted: 04/27/2022] [Indexed: 01/25/2023] Open
Abstract
This paper presents the consensus of the Scientific Department of Cognitive Neurology and Aging from the Brazilian Academy of Neurology on the diagnostic criteria for Alzheimer's disease (AD) in Brazil. The authors conducted a literature review regarding clinical and research criteria for AD diagnosis and proposed protocols for use at primary, secondary, and tertiary care levels. Within this clinical scenario, the diagnostic criteria for typical and atypical AD are presented as well as clinical, cognitive, and functional assessment tools and complementary propaedeutics with laboratory and neuroimaging tests. The use of biomarkers is also discussed for both clinical diagnosis (in specific conditions) and research.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Pontifícia Universidade do Rio Grande do Sul, Escola de Medicina, Serviço de Neurologia, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Instituto do Cérebro do Rio Grande do Sul, Porto Alegre RS, Brasil
- Pontifícia Universidade do Rio Grande do Sul, Programa de Pós-Graduação em Gerontologia Biomédica, Porto Alegre RS, Brasil
| | | | - Márcia Radanovic
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
| | - Orestes Vicente Forlenza
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Instituto de Psiquiatria, Laboratório de Neurociências, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Psiquiatria, São Paulo SP, Brasil
| | - Marcela Lima Silagi
- Universidade Federal de São Paulo, Departamento de Fonoaudiologia, São Paulo SP, Brasil
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Jerusa Smid
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Breno José Alencar Pires Barbosa
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
- Universidade Federal de Pernambuco, Centro de Ciências Médicas, Área Acadêmica de Neuropsiquiatria, Recife PE, Brasil
- Instituto de Medicina Integral Prof. Fernando Figueira, Recife PE, Brasil
| | | | - Leonardo Cruz de Souza
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | - Francisco Assis Carvalho Vale
- Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Medicina, São Carlos SP, Brasil
| | - Paulo Caramelli
- Universidade Federal de Minas Gerais, Departamento de Clínica Médica, Belo Horizonte MG, Brasil
| | | | - Márcia Lorena Fagundes Chaves
- Hospital de Clínicas de Porto Alegre, Serviço de Neurologia, Porto Alegre RS, Brasil
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre RS, Brasil
| | - Sonia Maria Dozzi Brucki
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| | - Benito Pereira Damasceno
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Neurologia, Campinas SP, Brasil
| | - Ricardo Nitrini
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brasil
| |
Collapse
|
44
|
He Q, Shi L, Luo Y, Wan C, Malone IB, Mok VCT, Cole JH, Anatürk M. Validation of the Alzheimer's disease-resemblance atrophy index in classifying and predicting progression in Alzheimer's disease. Front Aging Neurosci 2022; 14:932125. [PMID: 36062150 PMCID: PMC9435378 DOI: 10.3389/fnagi.2022.932125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/14/2022] [Indexed: 12/01/2022] Open
Abstract
Background Automated tools for characterising dementia risk have the potential to aid in the diagnosis, prognosis, and treatment of Alzheimer's disease (AD). Here, we examined a novel machine learning-based brain atrophy marker, the AD-resemblance atrophy index (AD-RAI), to assess its test-retest reliability and further validate its use in disease classification and prediction. Methods Age- and sex-matched 44 probable AD (Age: 69.13 ± 7.13; MMSE: 27-30) and 22 non-demented control (Age: 69.38 ± 7.21; MMSE: 27-30) participants were obtained from the Minimal Interval Resonance Imaging in Alzheimer's Disease (MIRIAD) dataset. Serial T1-weighted images (n = 678) from up to nine time points over a 2-year period, including 179 pairs of back-to-back scans acquired on same participants on the same day and 40 pairs of scans acquired at 2-week intervals were included. All images were automatically processed with AccuBrain® to calculate the AD-RAI. Its same-day repeatability and 2-week reproducibility were first assessed. The discriminative performance of AD-RAI was evaluated using the receiver operating characteristic curve, where DeLong's test was used to evaluate its performance against quantitative medial temporal lobe atrophy (QMTA) and hippocampal volume adjusted by intracranial volume (ICV)-proportions and ICV-residuals methods, respectively (HVR and HRV). Linear mixed-effects modelling was used to investigate longitudinal trajectories of AD-RAI and baseline AD-RAI prediction of cognitive decline. Finally, the longitudinal associations between AD-RAI and MMSE scores were assessed. Results AD-RAI had excellent same-day repeatability and excellent 2-week reproducibility. AD-RAI's AUC (99.8%; 95%CI = [99.3%, 100%]) was equivalent to that of QMTA (96.8%; 95%CI = [92.9%, 100%]), and better than that of HVR (86.8%; 95%CI = [78.2%, 95.4%]) or HRV (90.3%; 95%CI = [83.0%, 97.6%]). While baseline AD-RAI was significantly higher in the AD group, it did not show detectable changes over 2 years. Baseline AD-RAI was negatively associated with MMSE scores and the rate of the change in MMSE scores over time. A negative longitudinal association was also found between AD-RAI values and the MMSE scores among AD patients. Conclusions The AD-RAI represents a potential biomarker that may support AD diagnosis and be used to predict the rate of future cognitive decline in AD patients.
Collapse
Affiliation(s)
- Qiling He
- UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Lin Shi
- Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yishan Luo
- BrainNow Research Institute, Shenzhen, China
| | - Chao Wan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ian B. Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Vincent C. T. Mok
- Gerald Choa Neuroscience Centre, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - James H. Cole
- Department of Computer Science, Faculty of Engineering Science, University College London, London, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Melis Anatürk
- Department of Computer Science, Faculty of Engineering Science, University College London, London, United Kingdom
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Jiang Y, Wang P, Wen J, Wang J, Li H, Biswal BB. Hippocampus-based static functional connectivity mapping within white matter in mild cognitive impairment. Brain Struct Funct 2022; 227:2285-2297. [PMID: 35864361 DOI: 10.1007/s00429-022-02521-x] [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: 11/23/2021] [Accepted: 06/04/2022] [Indexed: 11/28/2022]
Abstract
Mild cognitive impairment (MCI) is clinically characterized by memory loss and cognitive impairment closely associated with the hippocampal atrophy. Accumulating studies have confirmed the presence of neural signal changes within white matter (WM) in resting-state functional magnetic resonance imaging (fMRI). However, it remains unclear how abnormal hippocampus activity affects the WM regions in MCI. The current study employs 43 MCI, 71 very MCI (VMCI) and 87 age-, gender-, and education-matched healthy controls (HCs) from the public OASIS-3 dataset. Using the left and right hippocampus as seed points, we obtained the whole-brain functional connectivity (FC) maps for each subject. We then perform one-way ANOVA analysis to investigate the abnormal FC regions among HCs, VMCI, and MCI. We further performed probabilistic tracking to estimate whether the abnormal FC correspond to structural connectivity disruptions. Compared to HCs, MCI and VMCI groups exhibited reduced FC in the right middle temporal gyrus within gray matter, and right temporal pole, right inferior frontal gyrus within white matter. Specific dysconnectivity is shown in the cerebellum Crus II, left inferior temporal gyrus within gray matter, and right frontal gyrus within white matter. In addition, the fiber bundles connecting the left hippocampus and right temporal pole within white matter show abnormally increased mean diffusivity in MCI. The current study proposes a new functional imaging direction for exploring the mechanism of memory decline and pathophysiological mechanisms in different stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Yuan Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Pan Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.
| | - Jiaping Wen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianlin Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Hongyi Li
- The Fourth People's Hospital of Chengdu, Chengdu, China
| | - Bharat B Biswal
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China. .,Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
| |
Collapse
|
46
|
Ryu DW, Hong YJ, Cho JH, Kwak K, Lee JM, Shim YS, Youn YC, Yang DW. Automated brain volumetric program measuring regional brain atrophy in diagnosis of mild cognitive impairment and Alzheimer's disease dementia. Brain Imaging Behav 2022; 16:2086-2096. [PMID: 35697957 DOI: 10.1007/s11682-022-00678-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 11/27/2022]
Abstract
A quantitative analysis of brain volume can assist in the diagnosis of Alzheimer's disease (AD) which is ususally accompanied by brain atrophy. With an automated analysis program Quick Brain Volumetry (QBraVo) developed for volumetric measurements, we measured regional volumes and ratios to evaluate their performance in discriminating AD dementia (ADD) and mild cognitive impairment (MCI) patients from normal controls (NC). Validation of QBraVo was based on intra-rater and inter-rater reliability with a manual measurement. The regional volumes and ratios to total intracranial volume (TIV) and to total brain volume (TBV) or total cerebrospinal fluid volume (TCV) were compared among subjects. The regional volume to total cerebellar volume ratio named Standardized Atrophy Volume Ratio (SAVR) was calculated to compare brain atrophy. Diagnostic performances to distinguish among NC, MCI, and ADD were compared between MMSE, SAVR, and the predictive model. In total, 56 NCs, 44 MCI, and 45 ADD patients were enrolled. The average run time of QBraVo was 5 min 36 seconds. Intra-rater reliability was 0.999. Inter-rater reliability was high for TBV, TCV, and TIV (R = 0.97, 0.89 and 0.93, respectively). The medial temporal SAVR showed the highest performance for discriminating ADD from NC (AUC = 0.808, diagnostic accuracy = 80.2%). The predictive model using both MMSE and medial temporal SAVR improved the diagnostic performance for MCI in NC (AUC = 0.844, diagnostic accuracy = 79%). Our results demonstrated QBraVo is a fast and accurate method to measure brain volume. The regional volume calculated as SAVR could help to diagnose ADD and MCI and increase diagnostic accuracy for MCI.
Collapse
Affiliation(s)
- Dong-Woo Ryu
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yun Jeong Hong
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Jung Hee Cho
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Kichang Kwak
- Department of Biomedical Engineering, Hanyang University, Seoul, Republic of Korea
| | - Jong-Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul, Republic of Korea
| | - Yong S Shim
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Dong Won Yang
- Department of Neurology, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
47
|
Effects of Palmitoylethanolamide on Neurodegenerative Diseases: A Review from Rodents to Humans. Biomolecules 2022; 12:biom12050667. [PMID: 35625595 PMCID: PMC9138306 DOI: 10.3390/biom12050667] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 02/06/2023] Open
Abstract
Palmitoylethanolamide (PEA) stands out among endogenous lipid mediators for its neuroprotective, anti-inflammatory, and analgesic functions. PEA belonging to the N-acetylanolamine class of phospholipids was first isolated from soy lecithin, egg yolk, and peanut flour. It is currently used for the treatment of different types of neuropathic pain, such as fibromyalgia, osteoarthritis, carpal tunnel syndrome, and many other conditions. The properties of PEA, especially of its micronized or ultra-micronized forms maximizing bioavailability and efficacy, have sparked a series of innovative research to evaluate its possible application as therapeutic agent for neurodegenerative diseases. Neurodegenerative diseases are widespread throughout the world, and although they are numerous and different, they share common patterns of conditions that result from progressive damage to the brain areas involved in mobility, muscle coordination and strength, mood, and cognition. The present review is aimed at illustrating in vitro and in vivo research, as well as human studies, using PEA treatment, alone or in combination with other compounds, in the presence of neurodegeneration. Namely, attention has been paid to the effects of PEA in counteracting neuroinflammatory conditions and in slowing down the progression of diseases, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, Frontotemporal dementia, Amyotrophic Lateral Sclerosis, and Multiple Sclerosis. Literature research demonstrated the efficacy of PEA in addressing the damage typical of major neurodegenerative diseases.
Collapse
|
48
|
Zhu Y, Pan D, He L, Rong X, Li H, Li Y, Pi Y, Xu Y, Tang Y. China Registry Study on Cognitive Impairment in the Elderly: Protocol of a Prospective Cohort Study. Front Aging Neurosci 2022; 13:797704. [PMID: 35002683 PMCID: PMC8733254 DOI: 10.3389/fnagi.2021.797704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/25/2021] [Indexed: 11/21/2022] Open
Abstract
Introduction: To develop appropriate strategies for early diagnosis and intervention of cognitive impairment, the identification of minimally invasive and cost-effective biomarkers for the early diagnosis of cognitive impairment is crucial and desirable. Therefore, the CHina registry study on cOgnitive imPairment in the Elderly (HOPE) study is designed to investigate the natural course of cognitive decline and explore the clinical, imaging, and biochemical markers for the detection and diagnosis of cognitive impairment on its earliest stage. Methods: Approximately 5,000 Chinese elderly aged more than 50 years were recruited from Sun Yat-sen Memorial Hospital, Sun Yat-sen University in Guangzhou, China by the year 2024. All subjects were invited to complete the clinical assessment, neuropsychological assessment, the biological samples collection (blood and cerebrospinal fluid (CSF)], magnetic resonance imaging (MRI) examination, and optional amyloid and tau PET. The follow-up survey was conducted every 1 year to repeat these assessments for 20 years. To better clarify the relationship between potential risk factors and endpoint events [changes in cognitive score or incidence of mild cognitive impairment (MCI) and/or dementia], appropriate statistical methods were used to analyze the data, including but not limited to, such as linear mixed-effect model, competing risk model, or the least absolute shrinkage and selection operator model. Significance: The CHina registry study on cOgnitive imPairment in the Elderly study is designed to explore the longitudinal changes in characteristics of participants with cognitive decline and to identify potential plasma and imaging biomarkers with cost-benefit and scalability advantages. The results will enable broader clinical access and efficient population screening and then improve the development of treatment and the quality of life for cognitive impairment at the early stage. Trial registration number: NCT04360200.
Collapse
Affiliation(s)
- Yingying Zhu
- Clinical Research Design Division, Clinical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Pan
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei He
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoming Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Honghong Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yaxuan Pi
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
49
|
Hemrungrojn S, Tangwongchai S, Charoenboon T, Panasawat M, Supasitthumrong T, Chaipresertsud P, Maleevach P, Likitjaroen Y, Phanthumchinda K, Maes M. Use of the Montreal Cognitive Assessment Thai Version to Discriminate Amnestic Mild Cognitive Impairment from Alzheimer's Disease and Healthy Controls: Machine Learning Results. Dement Geriatr Cogn Disord 2021; 50:183-194. [PMID: 34325427 DOI: 10.1159/000517822] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/11/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The Montreal Cognitive Assessment (MoCA) is an effective and applicable screening instrument to confirm the diagnosis of amnestic mild cognitive impairment (aMCI) from patients with Alzheimer's disease (AD) and healthy controls (HCs). OBJECTIVES This study aimed to determine the reliability and validity of the following: (a) Thai translation of the MoCA (MoCA-Thai) and (b) delineate the key features of aMCI based on the MoCA subdomains. METHODS This study included 60 HCs, 61 aMCI patients, and 60 AD patients. The MoCA-Thai shows adequate psychometric properties including internal consistency, concurrent validity, test-retest validity, and inter-rater reliability. RESULTS The MoCA-Thai may be employed as a diagnostic criterion to make the diagnosis of aMCI, whereby aMCI patients are discriminated from HC with an area under the receiver-operating characteristic (AUC-ROC) curve of 0.813 and from AD patients with an AUC-ROC curve of 0.938. The best cutoff scores of the MoCA-Thai to discriminate aMCI from HC is ≤24 and from AD > 16. Neural network analysis showed that (a) aberrations in recall was the most important feature of aMCI versus HC with impairments in language and orientation being the second and third most important features and (b) aberrations in visuospatial skills and executive functions were the most important features of AD versus aMCI and that impairments in recall, language, and orientation but not attention, concentration, and working memory, further discriminated AD from aMCI. CONCLUSIONS The MoCA-Thai is an appropriate cognitive assessment tool to be used in the Thai population for the diagnosis of aMCI and AD.
Collapse
Affiliation(s)
- Solaphat Hemrungrojn
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand, .,Cognitive Fitness Research Group, Chulalongkorn University, Bangkok, Thailand,
| | | | - Thammanard Charoenboon
- Department of Clinical Epidemiology, Faculty of Medicine, Thammasat University, Prathumthani, Thailand
| | - Muthita Panasawat
- Department of Psychiatry, Faculty of Medicine, Thammasat University, Prathumthani, Thailand
| | | | | | | | - Yuttachai Likitjaroen
- Division of Neurology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kammant Phanthumchinda
- Division of Neurology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Department of Psychiatry, Medical University of Plovdiv and Technological Center for Emergency Medicine, Plovdiv, Bulgaria.,IMPACT Strategic Research Centre, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
50
|
Ranson JM, Rittman T, Hayat S, Brayne C, Jessen F, Blennow K, van Duijn C, Barkhof F, Tang E, Mummery CJ, Stephan BCM, Altomare D, Frisoni GB, Ribaldi F, Molinuevo JL, Scheltens P, Llewellyn DJ. Modifiable risk factors for dementia and dementia risk profiling. A user manual for Brain Health Services-part 2 of 6. Alzheimers Res Ther 2021; 13:169. [PMID: 34635138 PMCID: PMC8507172 DOI: 10.1186/s13195-021-00895-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022]
Abstract
We envisage the development of new Brain Health Services to achieve primary and secondary dementia prevention. These services will complement existing memory clinics by targeting cognitively unimpaired individuals, where the focus is on risk profiling and personalized risk reduction interventions rather than diagnosing and treating late-stage disease. In this article, we review key potentially modifiable risk factors and genetic risk factors and discuss assessment of risk factors as well as additional fluid and imaging biomarkers that may enhance risk profiling. We then outline multidomain measures and risk profiling and provide practical guidelines for Brain Health Services, with consideration of outstanding uncertainties and challenges. Users of Brain Health Services should undergo risk profiling tailored to their age, level of risk, and availability of local resources. Initial risk assessment should incorporate a multidomain risk profiling measure. For users aged 39-64, we recommend the Cardiovascular Risk Factors, Aging, and Incidence of Dementia (CAIDE) Dementia Risk Score, whereas for users aged 65 and older, we recommend the Brief Dementia Screening Indicator (BDSI) and the Australian National University Alzheimer's Disease Risk Index (ANU-ADRI). The initial assessment should also include potentially modifiable risk factors including sociodemographic, lifestyle, and health factors. If resources allow, apolipoprotein E ɛ4 status testing and structural magnetic resonance imaging should be conducted. If this initial assessment indicates a low dementia risk, then low intensity interventions can be implemented. If the user has a high dementia risk, additional investigations should be considered if local resources allow. Common variant polygenic risk of late-onset AD can be tested in middle-aged or older adults. Rare variants should only be investigated in users with a family history of early-onset dementia in a first degree relative. Advanced imaging with 18-fluorodeoxyglucose positron emission tomography (FDG-PET) or amyloid PET may be informative in high risk users to clarify the nature and burden of their underlying pathologies. Cerebrospinal fluid biomarkers are not recommended for this setting, and blood-based biomarkers need further validation before clinical use. As new technologies become available, advances in artificial intelligence are likely to improve our ability to combine diverse data to further enhance risk profiling. Ultimately, Brain Health Services have the potential to reduce the future burden of dementia through risk profiling, risk communication, personalized risk reduction, and cognitive enhancement interventions.
Collapse
Affiliation(s)
- Janice M Ranson
- College of Medicine and Health, University of Exeter, Exeter, UK
- Deep Dementia Phenotyping (DEMON) Network, Exeter, UK
| | - Timothy Rittman
- Deep Dementia Phenotyping (DEMON) Network, Exeter, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Shabina Hayat
- Department of Public Health and Primary Care, Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Department of Public Health and Primary Care, Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Frederik Barkhof
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Eugene Tang
- Deep Dementia Phenotyping (DEMON) Network, Exeter, UK
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Catherine J Mummery
- Deep Dementia Phenotyping (DEMON) Network, Exeter, UK
- Dementia Research Centre, Institute of Neurology, University College London, and National Hospital for Neurology and Neurosurgery, University College London Hospital, London, UK
| | - Blossom C M Stephan
- Institute of Mental Health, Division of Psychiatry and Applied Psychology, School of Medicine, Nottingham University, Nottingham, UK
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), Saint John of God Clinical Research Centre, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Life Science Partners, Amsterdam, The Netherlands
| | - David J Llewellyn
- College of Medicine and Health, University of Exeter, Exeter, UK.
- Deep Dementia Phenotyping (DEMON) Network, Exeter, UK.
- Alan Turing Institute, London, UK.
- 2.04 College House, St Luke's Campus, University of Exeter Medical School, Exeter, EX1 2 LU, UK.
| |
Collapse
|