1
|
Simfukwe C, Han SH, Jeong HT, Youn YC. qEEG as Biomarker for Alzheimer's Disease: Investigating Relative PSD Difference and Coherence Analysis. Neuropsychiatr Dis Treat 2023; 19:2423-2437. [PMID: 37965528 PMCID: PMC10642578 DOI: 10.2147/ndt.s433207] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/04/2023] [Indexed: 11/16/2023] Open
Abstract
Purpose Electroencephalography (EEG) is a non-intrusive technique that provides comprehensive insights into the electrical activities of the brain's cerebral cortex. The brain signals obtained from EEGs can be used as a neuropsychological biomarker to detect different stages of Alzheimer's disease (AD) through quantitative EEG (qEEG) analysis. This paper investigates the difference in the abnormalities of resting state EEG (rEEG) signals between eyes-open (EOR) and eyes-closed (ECR) in AD by analyzing 19-scalp electrode EEG signals and making a comparison with healthy controls (HC). Participants and Methods The rEEG data from 534 subjects (ages 40-90) consisting of 269 HC and 265 AD subjects in South Korea were used in this study. The qEEG for EOR and ECR states were performed separately for HC and AD subjects to measure the relative power spectrum density (PSD) and coherence with functional connectivity to evaluate abnormalities. The rEEG data were preprocessed and analyzed using EEGlab and Brainstorm toolboxes in MATLAB R2021a software, and statistical analyses were carried out using ANOVA. Results Based on the Welch method, the relative PSD of the EEG EOR and ECR states difference in the AD group showed a significant increase in the delta frequency band of 19 EEG channels, particularly in the frontal, parietal, and temporal, than the HC groups. The delta power band on the source level was increased for the AD group and decreased for the HC group. In contrast, the source activities of alpha, beta, and gamma frequency bands were significantly reduced in the AD group, with a high decrease in the beta frequency band in all brain areas. Furthermore, the coherence of rEEG among different EEG electrodes was analyzed in the beta frequency band. It showed that pair-wise coherence between different brain areas in the AD group is remarkably increased in the ECR state and decreased after subtracting out the EOR state. Conclusion The findings suggest that examining PSD and functional connectivity through coherence analysis could serve as a promising and comprehensive approach to differentiate individuals with AD from normal, which may benefit our understanding of the disease.
Collapse
Affiliation(s)
- Chanda Simfukwe
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Su-Hyun Han
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Ho Tae Jeong
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, South Korea
| |
Collapse
|
2
|
Scheijbeler EP, de Haan W, Stam CJ, Twisk JWR, Gouw AA. Longitudinal resting-state EEG in amyloid-positive patients along the Alzheimer's disease continuum: considerations for clinical trials. Alzheimers Res Ther 2023; 15:182. [PMID: 37858173 PMCID: PMC10585755 DOI: 10.1186/s13195-023-01327-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND To enable successful inclusion of electroencephalography (EEG) outcome measures in Alzheimer's disease (AD) clinical trials, we retrospectively mapped the progression of resting-state EEG measures over time in amyloid-positive patients with mild cognitive impairment (MCI) or dementia due to AD. METHODS Resting-state 21-channel EEG was recorded in 148 amyloid-positive AD patients (MCI, n = 88; dementia due to AD, n = 60). Two or more EEG recordings were available for all subjects. We computed whole-brain and regional relative power (i.e., theta (4-8 Hz), alpha1 (8-10 Hz), alpha2 (10-13 Hz), beta (13-30 Hz)), peak frequency, signal variability (i.e., theta permutation entropy), and functional connectivity values (i.e., alpha and beta corrected amplitude envelope correlation, theta phase lag index, weighted symbolic mutual information, inverted joint permutation entropy). Whole-group linear mixed effects models were used to model the development of EEG measures over time. Group-wise analysis was performed to investigate potential differences in change trajectories between the MCI and dementia subgroups. Finally, we estimated the minimum sample size required to detect different treatment effects (i.e., 50% less deterioration, stabilization, or 50% improvement) on the development of EEG measures over time, in hypothetical clinical trials of 1- or 2-year duration. RESULTS Whole-group analysis revealed significant regional and global oscillatory slowing over time (i.e., increased relative theta power, decreased beta power), with strongest effects for temporal and parieto-occipital regions. Disease severity at baseline influenced the EEG measures' rates of change, with fastest deterioration reported in MCI patients. Only AD dementia patients displayed a significant decrease of the parieto-occipital peak frequency and theta signal variability over time. We estimate that 2-year trials, focusing on amyloid-positive MCI patients, require 36 subjects per arm (2 arms, 1:1 randomization, 80% power) to detect a stabilizing treatment effect on temporal relative theta power. CONCLUSIONS Resting-state EEG measures could facilitate early detection of treatment effects on neuronal function in AD patients. Their sensitivity depends on the region-of-interest and disease severity of the study population. Conventional spectral measures, particularly recorded from temporal regions, present sensitive AD treatment monitoring markers.
Collapse
Affiliation(s)
- Elliz P Scheijbeler
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands.
| | - Willem de Haan
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
| | - Cornelis J Stam
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
| | - Jos W R Twisk
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
| | - Alida A Gouw
- Clinical Neurophysiology and MEG Center, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, Netherlands
| |
Collapse
|
3
|
Chu KT, Lei WC, Wu MH, Fuh JL, Wang SJ, French IT, Chang WS, Chang CF, Huang NE, Liang WK, Juan CH. A holo-spectral EEG analysis provides an early detection of cognitive decline and predicts the progression to Alzheimer's disease. Front Aging Neurosci 2023; 15:1195424. [PMID: 37674782 PMCID: PMC10477374 DOI: 10.3389/fnagi.2023.1195424] [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: 03/28/2023] [Accepted: 07/25/2023] [Indexed: 09/08/2023] Open
Abstract
Aims Our aim was to differentiate patients with mild cognitive impairment (MCI) and Alzheimer's disease (AD) from cognitively normal (CN) individuals and predict the progression from MCI to AD within a 3-year longitudinal follow-up. A newly developed Holo-Hilbert Spectral Analysis (HHSA) was applied to resting state EEG (rsEEG), and features were extracted and subjected to machine learning algorithms. Methods A total of 205 participants were recruited from three hospitals, with CN (n = 51, MMSE > 26), MCI (n = 42, CDR = 0.5, MMSE ≥ 25), AD1 (n = 61, CDR = 1, MMSE < 25), AD2 (n = 35, CDR = 2, MMSE < 16), and AD3 (n = 16, CDR = 3, MMSE < 16). rsEEG was also acquired from all subjects. Seventy-two MCI patients (CDR = 0.5) were longitudinally followed up with two rsEEG recordings within 3 years and further subdivided into an MCI-stable group (MCI-S, n = 36) and an MCI-converted group (MCI-C, n = 36). The HHSA was then applied to the rsEEG data, and features were extracted and subjected to machine-learning algorithms. Results (a) At the group level analysis, the HHSA contrast of MCI and different stages of AD showed augmented amplitude modulation (AM) power of lower-frequency oscillations (LFO; delta and theta bands) with attenuated AM power of higher-frequency oscillations (HFO; beta and gamma bands) compared with cognitively normal elderly controls. The alpha frequency oscillation showed augmented AM power across MCI to AD1 with a reverse trend at AD2. (b) At the individual level of cross-sectional analysis, implementation of machine learning algorithms discriminated between groups with good sensitivity (Sen) and specificity (Spec) as follows: CN elderly vs. MCI: 0.82 (Sen)/0.80 (Spec), CN vs. AD1: 0.94 (Sen)/0.80 (Spec), CN vs. AD2: 0.93 (Sen)/0.90 (Spec), and CN vs. AD3: 0.75 (Sen)/1.00 (Spec). (c) In the longitudinal MCI follow-up, the initial contrasted HHSA between MCI-S and MCI-C groups showed significantly attenuated AM power of alpha and beta band oscillations. (d) At the individual level analysis of longitudinal MCI groups, deploying machine learning algorithms with the best seven features resulted in a sensitivity of 0.9 by the support vector machine (SVM) classifier, with a specificity of 0.8 yielded by the decision tree classifier. Conclusion Integrating HHSA into EEG signals and machine learning algorithms can differentiate between CN and MCI as well as also predict AD progression at the MCI stage.
Collapse
Affiliation(s)
- Kwo-Ta Chu
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Yang-Ming Hospital, Taoyuan, Taiwan
| | - Weng-Chi Lei
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
| | - Ming-Hsiu Wu
- Division of Neurology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Long-Term Care and Health Promotion, Min-Hwei Junior College of Health Care Management, Tainan, Taiwan
| | - Jong-Ling Fuh
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shuu-Jiun Wang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Isobel T. French
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Central University and Academia Sinica, Taipei, Taiwan
| | - Wen-Sheng Chang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Chi-Fu Chang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Norden E. Huang
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
- Key Laboratory of Data Analysis and Applications, First Institute of Oceanography, SOA, Qingdao, China
| | - Wei-Kuang Liang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
| | - Chi-Hung Juan
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
- Department of Psychology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
4
|
Ponomareva NV, Andreeva TV, Protasova MS, Kunizheva SS, Kuznetsova IL, Kolesnikova EP, Malina DD, Mitrofanov AA, Fokin VF, Illarioshkin SN, Rogaev EI. Neuronal Hyperactivation in EEG Data during Cognitive Tasks Is Related to the Apolipoprotein J/Clusterin Genotype in Nondemented Adults. Int J Mol Sci 2023; 24:6790. [PMID: 37047762 PMCID: PMC10095572 DOI: 10.3390/ijms24076790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/24/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
The clusterin (CLU) rs11136000 CC genotype is a probable risk factor for Alzheimer's disease (AD). CLU, also known as the apolipoprotein J gene, shares certain properties with the apolipoprotein E (APOE) gene with a well-established relationship with AD. This study aimed to determine whether the electrophysiological patterns of brain activation during the letter fluency task (LFT) depend on CLU genotypes in adults without dementia. Previous studies have shown that LFT performance involves activation of the frontal cortex. We examined EEG alpha1 and alpha2 band desynchronization in the frontal regions during the LFT in 94 nondemented individuals stratified by CLU (rs11136000) genotype. Starting at 30 years of age, CLU CC carriers exhibited more pronounced task-related alpha2 desynchronization than CLU CT&TT carriers in the absence of any differences in LFT performance. In CLU CC carriers, alpha2 desynchronization was significantly correlated with age. Increased task-related activation in individuals at genetic risk for AD may reflect greater "effort" to perform the task and/or neuronal hyperexcitability. The results show that the CLU genotype is associated with neuronal hyperactivation in the frontal cortex during cognitive tasks performances in nondemented individuals, suggesting systematic vulnerability of LFT related cognitive networks in people carrying unfavorable CLU alleles.
Collapse
Affiliation(s)
- Natalya V. Ponomareva
- Research Center of Neurology, 125367 Moscow, Russia
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354349 Sochi, Russia
| | - Tatiana V. Andreeva
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354349 Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- Centre for Genetics and Genetic Technologies, Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Maria S. Protasova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Svetlana S. Kunizheva
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354349 Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Irina L. Kuznetsova
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354349 Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | | | | | | | | | - Evgeny I. Rogaev
- Center for Genetics and Life Science, Sirius University of Science and Technology, 354349 Sochi, Russia
- Department of Psychiatry, Umass Chan Medical School, Shrewsbury, MA 01545, USA
| |
Collapse
|
5
|
Bourdès V, Dogterom P, Aleman A, Parmantier P, Colas D, Lemarchant S, Marie S, Chou T, Abd-Elaziz K, Godfrin Y. Safety, Tolerability, Pharmacokinetics and Initial Pharmacodynamics of a Subcommissural Organ-Spondin-Derived Peptide: A Randomized, Placebo-Controlled, Double-Blind, Single Ascending Dose First-in-Human Study. Neurol Ther 2022; 11:1353-1374. [PMID: 35779189 PMCID: PMC9338184 DOI: 10.1007/s40120-022-00380-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/08/2022] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION This randomized, double-blind, placebo-controlled study in healthy volunteers assessed the safety, tolerability, and pharmacokinetics of single ascending doses of intravenously administered NX210-a linear peptide derived from subcommissural organ-spondin-and explored the effects on blood/urine biomarkers and cerebral activity. METHODS Participants in five cohorts (n = 8 each) were randomized to receive a single intravenous dose of NX210 (n = 6 each) (0.4, 1.25, 2.5, 5, and 10 mg/kg) or placebo (n = 2 each); in total, 10 and 29 participants received placebo and NX210, respectively. Blood samples were collected for pharmacokinetics within 180 min post dosing. Plasma and urine were collected from participants (cohorts: 2.5, 5, and 10 mg/kg) for biomarker analysis and electroencephalography (EEG) recordings within 48 h post dosing. Safety/tolerability and pharmacokinetic data were assessed before ascending to the next dose. RESULTS The study included 39 participants. All dosages were safe and well tolerated. All treatment-emergent adverse events (n = 17) were of mild severity and resolved spontaneously (except one with unknown outcome). Twelve treatment-emergent adverse events (70.6%) were deemed drug related; seven of those (58.3%) concerned nervous system disorders (dizziness, headache, and somnolence). The pharmacokinetic analysis indicated a short half-life in plasma (6-20 min), high apparent volume of distribution (1870-4120 L), and rapid clearance (7440-16,400 L/h). In plasma, tryptophan and homocysteine showed dose-related increase and decrease, respectively. No drug dose effect was found for the glutamate or glutamine plasma biomarkers. Nevertheless, decreased blood glutamate and increased glutamine were observed in participants treated with NX210 versus placebo. EEG showed a statistically significant decrease in beta and gamma bands and a dose-dependent increasing trend in alpha bands. Pharmacodynamics effects were sustained for several hours (plasma) or 48 h (urine and EEG). CONCLUSION NX210 is safe and well tolerated and may exert beneficial effects on the central nervous system, particularly in terms of cognitive processing.
Collapse
Affiliation(s)
| | | | - André Aleman
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | - Yann Godfrin
- Axoltis Pharma, 60 Avenue Rockefeller, 69008, Lyon, France
- Godfrin Life-Sciences, Caluire-et-Cuire, France
| |
Collapse
|
6
|
Ponomareva NV, Andreeva TV, Protasova M, Konovalov RN, Krotenkova MV, Kolesnikova EP, Malina DD, Kanavets EV, Mitrofanov AA, Fokin VF, Illarioshkin SN, Rogaev EI. Genetic association of apolipoprotein E genotype with EEG alpha rhythm slowing and functional brain network alterations during normal aging. Front Neurosci 2022; 16:931173. [PMID: 35979332 PMCID: PMC9376365 DOI: 10.3389/fnins.2022.931173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022] Open
Abstract
The ε4 allele of the apolipoprotein E (APOE4+) genotype is a major genetic risk factor for Alzheimer’s disease (AD), but the mechanisms underlying its influence remain incompletely understood. The study aimed to investigate the possible effect of the APOE genotype on spontaneous electroencephalogram (EEG) alpha characteristics, resting-state functional MRI (fMRI) connectivity (rsFC) in large brain networks and the interrelation of alpha rhythm and rsFC characteristics in non-demented adults during aging. We examined the EEG alpha subband’s relative power, individual alpha peak frequency (IAPF), and fMRI rsFC in non-demented volunteers (age range 26–79 years) stratified by the APOE genotype. The presence of the APOE4+ genotype was associated with lower IAPF and lower relative power of the 11–13 Hz alpha subbands. The age related decrease in EEG IAPF was more pronounced in the APOE4+ carriers than in the APOE4+ non-carriers (APOE4-). The APOE4+ carriers had a stronger fMRI positive rsFC of the interhemispheric regions of the frontoparietal, lateral visual and salience networks than the APOE4– individuals. In contrast, the negative rsFC in the network between the left hippocampus and the right posterior parietal cortex was reduced in the APOE4+ carriers compared to the non-carriers. Alpha rhythm slowing was associated with the dysfunction of hippocampal networks. Our results show that in adults without dementia APOE4+ genotype is associated with alpha rhythm slowing and that this slowing is age-dependent. Our data suggest predominant alterations of inhibitory processes in large-scale brain network of non-demented APOE4+ carriers. Moreover, dysfunction of large-scale hippocampal network can influence APOE-related alpha rhythm vulnerability.
Collapse
Affiliation(s)
- Natalya V. Ponomareva
- Research Center of Neurology, Moscow, Russia
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
- *Correspondence: Natalya V. Ponomareva,
| | - Tatiana V. Andreeva
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences (RAS), Moscow, Russia
| | - Maria Protasova
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences (RAS), Moscow, Russia
| | | | | | | | | | | | | | | | | | - Evgeny I. Rogaev
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences (RAS), Moscow, Russia
- Brudnick Neuropsychiatric Research Institute (BNRI), University of Massachusetts Medical School, Worcester, MA, United States
- Evgeny I. Rogaev,
| |
Collapse
|
7
|
García Pretelt FJ, Suárez Relevo JX, Aguillón D, Lopera F, Ochoa JF, Tobón Quintero CA. Automatic Classification of Subjects of the PSEN1-E280A Family at Risk of Developing Alzheimer’s Disease Using Machine Learning and Resting State Electroencephalography. J Alzheimers Dis 2022; 87:817-832. [DOI: 10.3233/jad-210148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The study of genetic variant carriers provides an opportunity to identify neurophysiological changes in preclinical stages. Electroencephalography (EEG) is a low-cost and minimally invasive technique which, together with machine learning, provide the possibility to construct systems that classify subjects that might develop Alzheimer’s disease (AD). Objective: The aim of this paper is to evaluate the capacity of the machine learning techniques to classify healthy Non-Carriers (NonCr) from Asymptomatic Carriers (ACr) of PSEN1-E280A variant for autosomal dominant Alzheimer’s disease (ADAD), using spectral features from EEG channels and brain-related independent components (ICs) obtained using independent component analysis (ICA). Methods: EEG was recorded in 27 ACr and 33 NonCr. Statistical significance analysis was applied to spectral information from channels and group ICA (gICA), standardized low-resolution tomography (sLORETA) analysis was applied over the IC as well. Strategies for feature selection and classification like Chi-square, mutual informationm and support vector machines (SVM) were evaluated over the dataset. Results: A test accuracy up to 83% was obtained by implementing a SVM with spectral features derived from gICA. The main findings are related to theta and beta rhythms, generated in the parietal and occipital regions, like the precuneus and superior parietal lobule. Conclusion: Promising models for classification of preclinical AD due to PSEN-1-E280A variant can be trained using spectral features, and the importance of the beta band and precuneus region is highlighted in asymptomatic stages, opening up the possibility of its use as a screening methodology.
Collapse
Affiliation(s)
- Francisco J. García Pretelt
- Bioinstrumentation and Clinical Engineering Research Group (GIBIC), Bioengineering Program, Universidad de Antioquia, Medellín, Colombia
- Neuropsychology and Behavior Group (GRUNECO), Medical School, Universidad de Antioquia, Medellín, Colombia
| | - Jazmín X. Suárez Relevo
- Bioinstrumentation and Clinical Engineering Research Group (GIBIC), Bioengineering Program, Universidad de Antioquia, Medellín, Colombia
- Neuropsychology and Behavior Group (GRUNECO), Medical School, Universidad de Antioquia, Medellín, Colombia
| | - David Aguillón
- Neuroscience Group of Antioquia (GNA), Medical School, Universidad de Antioquia, Medellín, Colombia
- Neuropsychology and Behavior Group (GRUNECO), Medical School, Universidad de Antioquia, Medellín, Colombia
| | - Francisco Lopera
- Neuroscience Group of Antioquia (GNA), Medical School, Universidad de Antioquia, Medellín, Colombia
| | - John Fredy Ochoa
- Bioinstrumentation and Clinical Engineering Research Group (GIBIC), Bioengineering Program, Universidad de Antioquia, Medellín, Colombia
- Neuropsychology and Behavior Group (GRUNECO), Medical School, Universidad de Antioquia, Medellín, Colombia
| | - Carlos A. Tobón Quintero
- Neuroscience Group of Antioquia (GNA), Medical School, Universidad de Antioquia, Medellín, Colombia
- Neuropsychology and Behavior Group (GRUNECO), Medical School, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
8
|
Vijverberg EGB, Axelsen TM, Bihlet AR, Henriksen K, Weber F, Fuchs K, Harrison JE, Kühn-Wache K, Alexandersen P, Prins ND, Scheltens P. Rationale and study design of a randomized, placebo-controlled, double-blind phase 2b trial to evaluate efficacy, safety, and tolerability of an oral glutaminyl cyclase inhibitor varoglutamstat (PQ912) in study participants with MCI and mild AD-VIVIAD. Alzheimers Res Ther 2021; 13:142. [PMID: 34425883 PMCID: PMC8381483 DOI: 10.1186/s13195-021-00882-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Varoglutamstat (formerly PQ912) is a small molecule that inhibits the activity of the glutaminyl cyclase to reduce the level of pyroglutamate-A-beta (pGluAB42). Recent studies confirm that pGluAB42 is a particular amyloid form that is highly synaptotoxic and plays a significant role in the development of AD. METHODS This paper describes the design and methodology behind the phase 2b VIVIAD-trial in AD. The aim of this study is to evaluate varoglutamstat in a state-of-the-art designed, placebo-controlled, double-blind, randomized clinical trial for safety and tolerability, efficacy on cognition, and effects on brain activity and AD biomarkers. In addition to its main purpose, the trial will explore potential associations between novel and established biomarkers and their individual and composite relation to disease characteristics. RESULTS To be expected early 2023 CONCLUSION: This state of the art phase 2b study will yield important results for the field with respect to trial methodology and for the treatment of AD with a small molecule directed against pyroglutamate-A-beta. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04498650.
Collapse
Affiliation(s)
- E. G. B. Vijverberg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Brain Research Center, Amsterdam, The Netherlands
| | - T. M. Axelsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Sanos Clinic A/S, Vejle, Denmark
| | | | | | - F. Weber
- Vivoryon Therapeutics NV, Halle, Germany
| | - K. Fuchs
- Vivoryon Therapeutics NV, Halle, Germany
| | - J. E. Harrison
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Metis Cognition Ltd, Park House, Kilmington Common, Wiltshire, UK
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | | | | | - N. D. Prins
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Brain Research Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Alzheimercentrum Amsterdam, Amsterdam UMC, Locatie VUmc, De Boelelaan 1117/1118, 1091 HZ Amsterdam, The Netherlands
| |
Collapse
|
9
|
Vanneste S, Luckey A, McLeod SL, Robertson IH, To WT. Impaired posterior cingulate cortex-parahippocampus connectivity is associated with episodic memory retrieval problems in amnestic mild cognitive impairment. Eur J Neurosci 2021; 53:3125-3141. [PMID: 33738836 DOI: 10.1111/ejn.15189] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/14/2021] [Accepted: 02/28/2021] [Indexed: 11/27/2022]
Abstract
Episodic memory retention and retrieval decline are the most common impairments observed in amnestic mild cognitive impairment (aMCI) patients who progress to Alzheimer's disease (AD). Clinical electroencephalography research shows that patients with dementia due to AD exhibit a slowing of neural electrical activity in the parietal cortex. Memory research has further suggested that successful memory performance is associated with changes in a posterior cingulate-parahippocampal cortical network together with increased θ-γ oscillatory coupling, where θ oscillations act as carrier waves for γ oscillations, which contain the actual information. However, the neurophysiological link between the memory research and clinical studies investigating aMCI and AD is lacking. In this study, we look at brain activity in aMCI and how it relates to memory performance. We demonstrate decreased γ power in the posterior cingulate cortex and the left and right parahippocampus in aMCI patients in comparison to control participants. This goes together with reduced θ coherence between the posterior cingulate cortex and parahippocampus associated with altered memory performance aMCI patients in comparison to control participants. In addition, comparing patients with aMCI to control participants reveals an effect for θ-γ coupling for the posterior cingulate cortex, and the left and right parahippocampus. Taken together, our results show that parahippocampus and posterior cingulate cortex interact via θ-γ coupling, which is associated with memory recollection and is altered in aMCI patients, offering a potential candidate mechanism for memory decline in aMCI.
Collapse
Affiliation(s)
- Sven Vanneste
- Lab for Clinical & Integrative Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA.,School of Psychology, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute & Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Alison Luckey
- School of Psychology, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute & Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - S Lauren McLeod
- Lab for Clinical & Integrative Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Ian H Robertson
- School of Psychology, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute & Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Wing Ting To
- School of Nursing, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Yu H, Zhu L, Cai L, Wang J, Liu J, Wang R, Zhang Z. Identification of Alzheimer's EEG With a WVG Network-Based Fuzzy Learning Approach. Front Neurosci 2020; 14:641. [PMID: 32848530 PMCID: PMC7396629 DOI: 10.3389/fnins.2020.00641] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022] Open
Abstract
A novel analytical framework combined fuzzy learning and complex network approaches is proposed for the identification of Alzheimer's disease (AD) with multichannel scalp-recorded electroencephalograph (EEG) signals. Weighted visibility graph (WVG) algorithm is first applied to transform each channel EEG into network and its topological parameters were further extracted. Statistical analysis indicates that AD and normal subjects show significant difference in the structure of WVG network and thus can be used to identify Alzheimer's disease. Taking network parameters as input features, a Takagi-Sugeno-Kang (TSK) fuzzy model is established to identify AD's EEG signal. Three feature sets-single parameter from multi-networks, multi-parameters from single network, and multi-parameters from multi-networks-are considered as input vectors. The number and order of input features in each set is optimized with various feature selection methods. Classification results demonstrate the ability of network-based TSK fuzzy classifiers and the feasibility of three input feature sets. The highest accuracy that can be achieved is 95.28% for single parameter from four networks, 93.41% for three parameters from single network. In particular, multi-parameters from the multi-networks set obtained the best result. The highest accuracy, 97.12%, is achieved with five features selected from four networks. The combination of network and fuzzy learning can highly improve the efficiency of AD's EEG identification.
Collapse
Affiliation(s)
- Haitao Yu
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Lin Zhu
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Lihui Cai
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Jiang Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Jing Liu
- Department of Neurology, Tangshan Gongren Hospital, Tangshan, China
| | - Ruofan Wang
- School of Information Technology Engineering, Tianjin University of Technology and Education, Tianjin, China
| | - Zhiyong Zhang
- Department of Pathology, Tangshan Gongren Hospital, Tangshan, China
| |
Collapse
|
11
|
Briels CT, Schoonhoven DN, Stam CJ, de Waal H, Scheltens P, Gouw AA. Reproducibility of EEG functional connectivity in Alzheimer's disease. Alzheimers Res Ther 2020; 12:68. [PMID: 32493476 PMCID: PMC7271479 DOI: 10.1186/s13195-020-00632-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Although numerous electroencephalogram (EEG) studies have described differences in functional connectivity in Alzheimer's disease (AD) compared to healthy subjects, there is no general consensus on the methodology of estimating functional connectivity in AD. Inconsistent results are reported due to multiple methodological factors such as diagnostic criteria, small sample sizes and the use of functional connectivity measures sensitive to volume conduction. We aimed to investigate the reproducibility of the disease-associated effects described by commonly used functional connectivity measures with respect to the amyloid, tau and neurodegeneration (A/T/N) criteria. METHODS Eyes-closed task-free 21-channel EEG was used from patients with probable AD and subjective cognitive decline (SCD), to form two cohorts. Artefact-free epochs were visually selected and several functional connectivity measures (AEC(-c), coherence, imaginary coherence, PLV, PLI, wPLI) were estimated in five frequency bands. Functional connectivity was compared between diagnoses using AN(C)OVA models correcting for sex, age and, additionally, relative power of the frequency band. Another model predicted the Mini-Mental State Exam (MMSE) score of AD patients by functional connectivity estimates. The analysis was repeated in a subpopulation fulfilling the A/T/N criteria, after correction for influencing factors. The analyses were repeated in the second cohort. RESULTS Two large cohorts were formed (SCD/AD; n = 197/214 and n = 202/196). Reproducible effects were found for the AEC-c in the alpha and beta frequency bands (p = 6.20 × 10-7, Cohen's d = - 0.53; p = 5.78 × 10-4, d = - 0.37) and PLI and wPLI in the theta band (p = 3.81 × 10-8, d = 0.59; p = 1.62 × 10-8, d = 0.60, respectively). Only effects of the AEC-c remained significant after statistical correction for the relative power of the selected bandwidth. In addition, alpha band AEC-c correlated with disease severity represented by MMSE score. CONCLUSION The choice of functional connectivity measure and frequency band can have a large impact on the outcome of EEG studies in AD. Our results indicate that in the alpha and beta frequency bands, the effects measured by the AEC-c are reproducible and the most valid in terms of influencing factors, correlation with disease severity and preferable properties such as correction for volume conduction. Phase-based measures with correction for volume conduction, such as the PLI, showed reproducible effects in the theta frequency band.
Collapse
Affiliation(s)
- Casper T Briels
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
- Department of Clinical Neurophysiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Deborah N Schoonhoven
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Neurophysiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Cornelis J Stam
- Department of Clinical Neurophysiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Hanneke de Waal
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Alida A Gouw
- Department of Clinical Neurophysiology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
12
|
Effective differentiation of mild cognitive impairment by functional brain graph analysis and computerized testing. PLoS One 2020; 15:e0230099. [PMID: 32176709 PMCID: PMC7075594 DOI: 10.1371/journal.pone.0230099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 02/21/2020] [Indexed: 11/25/2022] Open
Abstract
Community-dwelling African American elders are twice as likely to develop mild cognitive impairment (MCI) or Alzheimer’s disease and related dementias than older white Americans and therefore represent a significant at-risk group in need of early monitoring. More extensive imaging or cerebrospinal fluid studies represent significant barriers due to cost and burden. We combined functional connectivity and graph theoretical measures, derived from resting-state electroencephalography (EEG) recordings, with computerized cognitive testing to identify differences between persons with MCI and healthy controls based on a sample of community-dwelling African American elders. We found a significant decrease in functional connectivity and a less integrated graph topology in persons with MCI. A combination of functional connectivity, topological and cognition measurements is powerful for prediction of MCI and combined measures are clearly more effective for prediction than using a single approach. Specifically, by combining cognition features with functional connectivity and topological features the prediction improved compared with the classification using features from single cognitive or EEG domains, with an accuracy of 86.5%, compared with the accuracy of 77.5% of the best single approach. Community-dwelling African American elders find EEG and computerized testing acceptable and results are promising in terms of differentiating between healthy controls and persons with MCI living in the community.
Collapse
|
13
|
Colom-Cadena M, Spires-Jones T, Zetterberg H, Blennow K, Caggiano A, DeKosky ST, Fillit H, Harrison JE, Schneider LS, Scheltens P, de Haan W, Grundman M, van Dyck CH, Izzo NJ, Catalano SM. The clinical promise of biomarkers of synapse damage or loss in Alzheimer's disease. Alzheimers Res Ther 2020; 12:21. [PMID: 32122400 PMCID: PMC7053087 DOI: 10.1186/s13195-020-00588-4] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/14/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Synapse damage and loss are fundamental to the pathophysiology of Alzheimer's disease (AD) and lead to reduced cognitive function. The goal of this review is to address the challenges of forging new clinical development approaches for AD therapeutics that can demonstrate reduction of synapse damage or loss. The key points of this review include the following: Synapse loss is a downstream effect of amyloidosis, tauopathy, inflammation, and other mechanisms occurring in AD.Synapse loss correlates most strongly with cognitive decline in AD because synaptic function underlies cognitive performance.Compounds that halt or reduce synapse damage or loss have a strong rationale as treatments of AD.Biomarkers that measure synapse degeneration or loss in patients will facilitate clinical development of such drugs.The ability of methods to sensitively measure synapse density in the brain of a living patient through synaptic vesicle glycoprotein 2A (SV2A) positron emission tomography (PET) imaging, concentrations of synaptic proteins (e.g., neurogranin or synaptotagmin) in the cerebrospinal fluid (CSF), or functional imaging techniques such as quantitative electroencephalography (qEEG) provides a compelling case to use these types of measurements as biomarkers that quantify synapse damage or loss in clinical trials in AD. CONCLUSION A number of emerging biomarkers are able to measure synapse injury and loss in the brain and may correlate with cognitive function in AD. These biomarkers hold promise both for use in diagnostics and in the measurement of therapeutic successes.
Collapse
Affiliation(s)
- Martí Colom-Cadena
- Centre for Discovery Brain Sciences, UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Steven T DeKosky
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Howard Fillit
- Alzheimer's Drug Discovery Foundation, New York, NY, USA
| | - John E Harrison
- Metis Cognition Ltd, Kilmington, UK
- Alzheimer Center, AUmc, Amsterdam, The Netherlands
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | | | - Phillip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Willem de Haan
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Clinical Neurophysiology and MEG, VU University Medical Center, Amsterdam, Netherlands
| | | | - Christopher H van Dyck
- Alzheimer's Disease Research Unit and Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | | | | |
Collapse
|
14
|
Methylphenidate and galantamine in patients with vascular cognitive impairment-the proof-of-principle study STREAM-VCI. ALZHEIMERS RESEARCH & THERAPY 2020; 12:10. [PMID: 31910895 PMCID: PMC6947990 DOI: 10.1186/s13195-019-0567-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/06/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND To date, no symptomatic treatment is available for patients with vascular cognitive impairment (VCI). In the proof-of-principle study Symptomatic Treatment of Vascular Cognitive Impairment (STREAM-VCI), we investigated whether a single dose of a monoaminergic drug (methylphenidate) improves executive functioning and whether a single dose of a cholinergic drug (galantamine) improves memory in VCI patients. METHODS STREAM-VCI is a single-center, double-blind, three-way crossover trial. We included 30 VCI patients (Mini-Mental State Examination (MMSE) ≥ 16 and Clinical Dementia Rating score 0.5-1.0) with cerebrovascular pathology on MRI. All patients received single doses of methylphenidate (10 mg), galantamine (16 mg), and placebo in random order on three separate study visits. We used the NeuroCart®, a computerized test battery, to assess drug-sensitive cognitive effects. Predefined main outcomes, measured directly after a single dose of a study drug, were (i) change in performance on the adaptive tracker for executive functioning and (ii) performance on the Visual Verbal Learning Test-15 (VVLT-15) for memory, compared to placebo. We performed mixed model analysis of variance. RESULTS The study population had a mean age of 67 ± 8 years and MMSE 26 ± 3, and 9 (30%) were female. Methylphenidate improved performance on the adaptive tracker more than placebo (mean difference 1.40%; 95% confidence interval [CI] 0.56-2.25; p = 0.002). In addition, methylphenidate led to better memory performance on the VVLT-15 compared to placebo (mean difference in recalled words 0.59; 95% CI 0.03-1.15; p = 0.04). Galantamine did not improve performance on the adaptive tracker and led to worse performance on delayed recall of the VVLT-15 (mean difference - 0.84; 95% CI - 1.65, - 0.03; p = 0.04). Methylphenidate was well tolerated while galantamine produced gastrointestinal side effects in a considerable number of patients. CONCLUSIONS In this proof-of-principle study, methylphenidate is well tolerated and improves executive functioning and immediate recall in patients with VCI. Galantamine did not improve memory or executive dysfunction. Results might be influenced by the considerable amount of side effects seen. TRIAL REGISTRATION http://www.clinicaltrials.gov. Registration number: NCT02098824. Registration date: March 28, 2014.
Collapse
|
15
|
Briels C, Stam C, Scheltens P, Bruins S, Lues I, Gouw A. In pursuit of a sensitive EEG functional connectivity outcome measure for clinical trials in Alzheimer’s disease. Clin Neurophysiol 2020; 131:88-95. [DOI: 10.1016/j.clinph.2019.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/19/2019] [Accepted: 09/15/2019] [Indexed: 01/01/2023]
|
16
|
Durongbhan P, Zhao Y, Chen L, Zis P, De Marco M, Unwin ZC, Venneri A, He X, Li S, Zhao Y, Blackburn DJ, Sarrigiannis PG. A Dementia Classification Framework Using Frequency and Time-Frequency Features Based on EEG Signals. IEEE Trans Neural Syst Rehabil Eng 2019; 27:826-835. [DOI: 10.1109/tnsre.2019.2909100] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
17
|
Franciotti R, Falasca NW, Arnaldi D, Famà F, Babiloni C, Onofrj M, Nobili FM, Bonanni L. Cortical Network Topology in Prodromal and Mild Dementia Due to Alzheimer's Disease: Graph Theory Applied to Resting State EEG. Brain Topogr 2019; 32:127-141. [PMID: 30145728 PMCID: PMC6326972 DOI: 10.1007/s10548-018-0674-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 08/17/2018] [Indexed: 12/31/2022]
Abstract
Graph theory analysis on resting state electroencephalographic rhythms disclosed topological properties of cerebral network. In Alzheimer's disease (AD) patients, this approach showed mixed results. Granger causality matrices were used as input to the graph theory allowing to estimate the strength and the direction of information transfer between electrode pairs. The number of edges (degree), the number of inward edges (in-degree), of outgoing edges (out-degree) were statistically compared among healthy controls, patients with mild cognitive impairment due to AD (AD-MCI) and AD patients with mild dementia (ADD) to evaluate if degree abnormality could involve low and/or high degree vertices, the so called hubs, in both prodromal and over dementia stage. Clustering coefficient and local efficiency were evaluated as measures of network segregation, path length and global efficiency as measures of integration, the assortativity coefficient as a measure of resilience. Degree, in-degree and out-degree values were lower in AD-MCI and ADD than the control group for non-hubs and hubs vertices. The number of edges was preserved for frontal electrodes, where patients' groups showed an additional hub in F3. Clustering coefficient was lower in ADD compared with AD-MCI in the right occipital electrode, and it was positively correlated with mini mental state examination. Local and global efficiency values were lower in patients' than control groups. Our results show that the topology of the network is altered in AD patients also in its prodromal stage, begins with the reduction of the number of edges and the loss of the local and global efficiency.
Collapse
Affiliation(s)
- Raffaella Franciotti
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Via Luigi Polacchi, 66013, Chieti, Italy
| | - Nicola Walter Falasca
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Via Luigi Polacchi, 66013, Chieti, Italy
- BIND - Behavioral Imaging and Neural Dynamics Center, "G. d'Annunzio" University, Chieti, Italy
| | - Dario Arnaldi
- Dipartimento di Neuroscienze (DINOGMI), Università di Genova, Genoa, Italy
- U.O. Clinica Neurologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Famà
- Dipartimento di Neuroscienze (DINOGMI), Università di Genova, Genoa, Italy
- U.O. Neurofisiopatologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Claudio Babiloni
- Department of Physiology and Pharmacology, University of Rome "La Sapienza", Rome, Italy
- IRCCS S. Raffaele Pisana, Rome, Italy
- IRCCS S. Raffaele Cassino, Cassino, Italy
| | - Marco Onofrj
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Via Luigi Polacchi, 66013, Chieti, Italy
| | - Flavio Mariano Nobili
- Dipartimento di Neuroscienze (DINOGMI), Università di Genova, Genoa, Italy
- U.O. Clinica Neurologica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Bonanni
- Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Via Luigi Polacchi, 66013, Chieti, Italy.
| |
Collapse
|
18
|
Simpraga S, Mansvelder HD, Groeneveld GJ, Prins S, Hart EP, Poil SS, Linkenkaer-Hansen K. An EEG nicotinic acetylcholine index to assess the efficacy of pro-cognitive compounds. Clin Neurophysiol 2018; 129:2325-2332. [DOI: 10.1016/j.clinph.2018.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/27/2018] [Accepted: 08/23/2018] [Indexed: 11/26/2022]
|
19
|
Fraga FJ, Mamani GQ, Johns E, Tavares G, Falk TH, Phillips NA. Early diagnosis of mild cognitive impairment and Alzheimer's with event-related potentials and event-related desynchronization in N-back working memory tasks. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2018; 164:1-13. [PMID: 30195417 DOI: 10.1016/j.cmpb.2018.06.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/24/2018] [Accepted: 06/14/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND AND OBJECTIVE In this study we investigate whether or not event-related potentials (ERP) and/or event-related (de)synchronization (ERD/ERS) can be used to differentiate between 27 healthy elderly (HE), 21 subjects diagnosed with mild cognitive impairment (MCI) and 15 mild Alzheimer's disease (AD) patients. METHODS Using 32-channel EEG recordings, we measured ERP responses to a three-level (N-back, N = 0,1,2) visual working memory task. We also performed ERD analysis over the same EEG data, dividing the full-band signal into the well-known delta, theta, alpha, beta and gamma bands. Both ERP and ERD analyses were followed by cluster analysis with correction for multicomparisons whenever significant differences were found between groups. RESULTS Regarding ERP (full-band analysis), our findings have shown both patient groups (MCI and AD) with reduced P450 amplitude (compared to HE controls) in the execution of the non-match 1-back task at many scalp electrodes, chiefly at parietal and centro-parietal areas. However, no significant differences were found between MCI and AD in ERP analysis whatever was the task. As for sub-band analyses, ERD/ERS measures revealed that HE subjects elicited consistently greater alpha ERD responses than MCI and AD patients during the 1-back task in the match condition, with all differences located at frontal, central and occipital regions. Moreover, in the non-match condition, it was possible to distinguish between MCI and AD patients when they were performing the 0-back task, with MCI presenting more desynchronization than AD on the theta band at temporal and fronto-temporal areas. In summary, ERD analyses have revealed themselves more valuable than ERP, since they showed significant differences in all three group comparisons: HE vs. MCI, HE vs. AD, and MCI vs. AD. CONCLUSIONS Based on these findings, we conclude that ERD responses to working memory (N-back) tasks could be useful not only for early MCI diagnosis or for improved AD diagnosis, but probably also for assessing the likelihood of MCI progression to AD, after further validated by a longitudinal study.
Collapse
Affiliation(s)
- Francisco J Fraga
- Engineering, Modelling and Applied Social Sciences Center, Universidade Federal do ABC, Santo André, São Paulo, Brazil.
| | - Godofredo Quispe Mamani
- Engineering, Modelling and Applied Social Sciences Center, Universidade Federal do ABC, Santo André, São Paulo, Brazil; Departamento de Estadística, Universidad Nacional del Altiplano, Puno, Peru
| | - Erin Johns
- Department of Psychology, Concordia University, Montreal, Quebec, Canada
| | - Guilherme Tavares
- Engineering, Modelling and Applied Social Sciences Center, Universidade Federal do ABC, Santo André, São Paulo, Brazil
| | - Tiago H Falk
- Institut National de la Recherche Scientifique (INRS-EMT), University of Quebec, Montreal, Quebec, Canada
| | - Natalie A Phillips
- Department of Psychology, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
20
|
Alvarez-Jimenez R, Hart EP, Prins S, de Kam M, van Gerven JMA, Cohen AF, Groeneveld GJ. Reversal of mecamylamine-induced effects in healthy subjects by nicotine receptor agonists: Cognitive and (electro) physiological responses. Br J Clin Pharmacol 2018; 84:888-899. [PMID: 29319910 DOI: 10.1111/bcp.13507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 12/24/2017] [Accepted: 12/31/2017] [Indexed: 11/27/2022] Open
Abstract
AIMS Establishing a pharmacological challenge model could yield an important tool to understand the complex role of the nicotinic cholinergic system in cognition and to develop novel compounds acting on the nicotinic acetylcholine receptor. METHODS This randomized, double-blind, double-dummy, placebo-controlled, four-way crossover study examined the effects of the nicotinic antagonist mecamylamine on a battery of cognitive and neurophysiological test with coadministration of a placebo, nicotine or galantamine in order to reverse the cognitive impairment caused by mecamylamine. RESULTS Thirty-three healthy subjects received a single oral dose of 30 mg of mecamylamine (or placebo) in combination with either 16 mg of oral galantamine or 21 mg of transdermal nicotine (or its double-dummy). Mecamylamine 30 mg induced significant disturbances of cognitive functions. Attention and execution of visual (fine) motor tasks was decreased, short- and long-term memory was impaired and the reaction velocity during the test was slower when compared to placebo. Mecamylamine 30 mg produced a decrease in posterior α and β power in the surface electroencephalogram, effects that were reversed by nicotine coadministration. Memory and motor coordination tests could be partially reversed by the coadministration of nicotine. CONCLUSIONS Mecamylamine administration induced slowing of the electroencephalogram and produced decrease in performance of tests evaluating motor coordination, sustained attention and short- and long-term memory. These effects could be partially reversed by the coadministration of nicotine, and to a lesser extent by galantamine.
Collapse
Affiliation(s)
- Ricardo Alvarez-Jimenez
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.,Anesthesiology Department, Vrije Universiteit Medisch Centrum (VU University Medical Center), De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Ellen P Hart
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands
| | - Samantha Prins
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands
| | - Marieke de Kam
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands
| | - Joop M A van Gerven
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.,Neurology Department, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Adam F Cohen
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.,Internal Medicine Department, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Geert Jan Groeneveld
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.,Neurology Department, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Ochoa JF, Alonso JF, Duque JE, Tobón CA, Mañanas MA, Lopera F, Hernández AM. Successful Object Encoding Induces Increased Directed Connectivity in Presymptomatic Early-Onset Alzheimer's Disease. J Alzheimers Dis 2018; 55:1195-1205. [PMID: 27792014 PMCID: PMC5147495 DOI: 10.3233/jad-160803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Recent studies report increases in neural activity in brain regions critical to episodic memory at preclinical stages of Alzheimer's disease (AD). Although electroencephalography (EEG) is widely used in AD studies, given its non-invasiveness and low cost, there is a need to translate the findings in other neuroimaging methods to EEG. OBJECTIVE To examine how the previous findings using functional magnetic resonance imaging (fMRI) at preclinical stage in presenilin-1 E280A mutation carriers could be assessed and extended, using EEG and a connectivity approach. METHODS EEG signals were acquired during resting and encoding in 30 normal cognitive young subjects, from an autosomal dominant early-onset AD kindred from Antioquia, Colombia. Regions of the brain previously reported as hyperactive were used for connectivity analysis. RESULTS Mutation carriers exhibited increasing connectivity at analyzed regions. Among them, the right precuneus exhibited the highest changes in connectivity. CONCLUSION Increased connectivity in hyperactive cerebral regions is seen in individuals, genetically-determined to develop AD, at preclinical stage. The use of a connectivity approach and a widely available neuroimaging technique opens the possibility to increase the use of EEG in early detection of preclinical AD.
Collapse
Affiliation(s)
- John Fredy Ochoa
- Bioinstrumentation and Clinical Engineering Research Group, Bioengineering Program, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Joan Francesc Alonso
- Department of Automatic Control (ESAII), Biomedical Engineering Research Center (CREB), Universitat Politènica de Catalunya (UPC), Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Jon Edinson Duque
- Bioinstrumentation and Clinical Engineering Research Group, Bioengineering Program, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Carlos Andrés Tobón
- Neuroscience Group of Antioquia, Medical School, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.,Neuropsychology and Behavior group, Medical School, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Miguel Angel Mañanas
- Department of Automatic Control (ESAII), Biomedical Engineering Research Center (CREB), Universitat Politènica de Catalunya (UPC), Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Francisco Lopera
- Neuroscience Group of Antioquia, Medical School, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Alher Mauricio Hernández
- Bioinstrumentation and Clinical Engineering Research Group, Bioengineering Program, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| |
Collapse
|
22
|
Simpraga S, Alvarez-Jimenez R, Mansvelder HD, van Gerven JMA, Groeneveld GJ, Poil SS, Linkenkaer-Hansen K. EEG machine learning for accurate detection of cholinergic intervention and Alzheimer's disease. Sci Rep 2017; 7:5775. [PMID: 28720796 PMCID: PMC5515842 DOI: 10.1038/s41598-017-06165-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 06/09/2017] [Indexed: 12/21/2022] Open
Abstract
Monitoring effects of disease or therapeutic intervention on brain function is increasingly important for clinical trials, albeit hampered by inter-individual variability and subtle effects. Here, we apply complementary biomarker algorithms to electroencephalography (EEG) recordings to capture the brain’s multi-faceted signature of disease or pharmacological intervention and use machine learning to improve classification performance. Using data from healthy subjects receiving scopolamine we developed an index of the muscarinic acetylcholine receptor antagonist (mAChR) consisting of 14 EEG biomarkers. This mAChR index yielded higher classification performance than any single EEG biomarker with cross-validated accuracy, sensitivity, specificity and precision ranging from 88–92%. The mAChR index also discriminated healthy elderly from patients with Alzheimer’s disease (AD); however, an index optimized for AD pathophysiology provided a better classification. We conclude that integrating multiple EEG biomarkers can enhance the accuracy of identifying disease or drug interventions, which is essential for clinical trials.
Collapse
Affiliation(s)
- Sonja Simpraga
- Department of Integrative Neurophysiology, CNCR, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, CNCR, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Geert Jan Groeneveld
- Centre for Human Drug Research, Leiden, The Netherlands.,Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Simon-Shlomo Poil
- Department of Integrative Neurophysiology, CNCR, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,NBT Analytics BV, Amsterdam, The Netherlands
| | - Klaus Linkenkaer-Hansen
- Department of Integrative Neurophysiology, CNCR, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Cerebral PET glucose hypometabolism in subjects with mild cognitive impairment and higher EEG high-alpha/low-alpha frequency power ratio. Neurobiol Aging 2017; 58:213-224. [PMID: 28755648 DOI: 10.1016/j.neurobiolaging.2017.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/29/2017] [Accepted: 06/18/2017] [Indexed: 01/18/2023]
Abstract
In Alzheimer's disease (AD) research, both 2-deoxy-2-(18F)fluoro-D-glucose (FDG) positron emission tomography (PET) and electroencephalography (EEG) are reliable investigational modalities. The aim of this study was to investigate the associations between EEG High-alpha/Low-alpha (H-alpha/L-alpha) power ratio and cortical glucose metabolism. A total of 23 subjects with mild cognitive impairment (MCI) underwent FDG-PET and EEG examinations. H-alpha/L-alpha power ratio was computed for each subject and 2 groups were obtained based on the increase of the power ratio. The subjects with higher H-alpha/L-alpha power ratio showed a decrease in glucose metabolism in the hub brain areas previously identified as typically affected by AD pathology. In subjects with higher H-alpha/L-alpha ratio and lower metabolism, a "double alpha peak" was identified in the EEG spectrum and a U-shaped correlation between glucose metabolism and increase of H-alpha/L-alpha power ratio has been found. Moreover, in this group, a conversion rate of 62.5% at 24 months was detected, significantly different from the chance percentage expected. The neurophysiological meaning of the interplay between alpha oscillations and glucose metabolism and the possible interest of the H-alpha/L-alpha power ratio as a clinical biomarker in AD have been discussed.
Collapse
|
24
|
Gouw AA, Alsema AM, Tijms BM, Borta A, Scheltens P, Stam CJ, van der Flier WM. EEG spectral analysis as a putative early prognostic biomarker in nondemented, amyloid positive subjects. Neurobiol Aging 2017. [PMID: 28646686 DOI: 10.1016/j.neurobiolaging.2017.05.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We studied whether electroencephalography (EEG)-derived measures of brain oscillatory activity are related to clinical progression in nondemented, amyloid positive subjects. We included 205 nondemented amyloid positive subjects (63 subjective cognitive decline [SCD]; 142 mild cognitive impairment [MCI]) with a baseline resting-state EEG data and ≥1-year follow-up. Peak frequency and relative power of 4 frequency bands were calculated. Relationships between normalized EEG measures and time to clinical progression (conversion from SCD to MCI/dementia or from MCI to dementia) were analyzed using Cox proportional hazard models. One hundred eight (53%) subjects clinically progressed after 2.1 (IQR 1.3-3.0) years. In the total sample, none of the EEG spectral measures were significant predictors. Stratified for baseline diagnosis, we found that in SCD patients higher delta and theta power (HR [95% CI] = 1.7 [1.0-2.7] resp. 2.3 [1.2-4.4]), and lower alpha power and peak frequency (HR [95% CI] = 0.5 [0.3-1.0] resp. 0.6 [0.4-1.0]) were associated with clinical progression over time. In amyloid positive subjects with normal cognition, slowing of oscillatory brain activity is related to clinical progression.
Collapse
Affiliation(s)
- Alida A Gouw
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands; Department of Clinical Neurophysiology and MEG Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.
| | - Astrid M Alsema
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands; Department of Clinical Neurophysiology and MEG Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Betty M Tijms
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Andreas Borta
- Boehringer Ingelheim Pharma GmbH Co KG, Ingelheim am Rhein, Germany
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Cornelis J Stam
- Department of Clinical Neurophysiology and MEG Center, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands; Department of Epidemiology and Biostatistics, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
25
|
Ochoa JF, Alonso JF, Duque JE, Tobón CA, Baena A, Lopera F, Mañanas MA, Hernández AM. Precuneus Failures in Subjects of the PSEN1 E280A Family at Risk of Developing Alzheimer's Disease Detected Using Quantitative Electroencephalography. J Alzheimers Dis 2017; 58:1229-1244. [PMID: 28550254 DOI: 10.3233/jad-161291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Presenilin-1 (PSEN1) mutations are the most common cause of familial early onset Alzheimer's disease (AD). The PSEN1 E280A (E280A) mutation has an autosomal dominant inheritance and is involved in the production of amyloid-β. The largest family group of carriers with E280A mutation is found in Antioquia, Colombia. The study of mutation carriers provides a unique opportunity to identify brain changes in stages previous to AD. Electroencephalography (EEG) is a low cost and minimally invasiveness technique that enables the following of brain changes in AD. OBJECTIVE To examine how previous reported differences in EEG for Theta and Alpha-2 rhythms in E280A subjects are related to specific regions in cortex and could be tracked across different ages. METHODS EEG signals were acquired during resting state from non-carriers and carriers, asymptomatic and symptomatic subjects from E280A kindred from Antioquia, Colombia. Independent component analysis (ICA) and inverse solution methods were used to locate brain regions related to differences in Theta and Alpha-2 bands. RESULTS ICA identified two components, mainly related to the Precuneus, where the differences in Theta and Alpha-2 exist simultaneously at asymptomatic and symptomatic stages. When the ratio between Theta and Alpha-2 is used, significant correlations exist with age and a composite cognitive scale. CONCLUSION Theta and Alpha-2 rhythms are altered in E280A subjects. The alterations are possible to track at Precuneus regions using EEG, ICA, and inverse solution methods.
Collapse
Affiliation(s)
- John Fredy Ochoa
- Bioinstrumentation and Clinical Engineering Research Group, Bioengineering Program, Universidad de Antioquia, Medellín, Colombia
| | - Joan Francesc Alonso
- Department of Automatic Control (ESAII), Biomedical Engineering Research Center (CREB), Universitat Politènica de Catalunya (UPC), Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Jon Edinson Duque
- Bioinstrumentation and Clinical Engineering Research Group, Bioengineering Program, Universidad de Antioquia, Medellín, Colombia
| | - Carlos Andrés Tobón
- Neuroscience Group of Antioquia, Medical School, Universidad de Antioquia, Medellín, Colombia.,Neuropsychology and Behavior Group, Medical School, Universidad de Antioquia, Medellín, Colombia
| | - Ana Baena
- Neuroscience Group of Antioquia, Medical School, Universidad de Antioquia, Medellín, Colombia
| | - Francisco Lopera
- Neuroscience Group of Antioquia, Medical School, Universidad de Antioquia, Medellín, Colombia
| | - Miguel Angel Mañanas
- Department of Automatic Control (ESAII), Biomedical Engineering Research Center (CREB), Universitat Politènica de Catalunya (UPC), Barcelona, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Alher Mauricio Hernández
- Bioinstrumentation and Clinical Engineering Research Group, Bioengineering Program, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
26
|
Engels MMA, van der Flier WM, Stam CJ, Hillebrand A, Scheltens P, van Straaten ECW. Alzheimer's disease: The state of the art in resting-state magnetoencephalography. Clin Neurophysiol 2017. [PMID: 28622527 DOI: 10.1016/j.clinph.2017.05.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is accompanied by functional brain changes that can be detected in imaging studies, including electromagnetic activity recorded with magnetoencephalography (MEG). Here, we systematically review the studies that have examined resting-state MEG changes in AD and identify areas that lack scientific or clinical progress. Three levels of MEG analysis will be covered: (i) single-channel signal analysis, (ii) pairwise analyses over time series, which includes the study of interdependencies between two time series and (iii) global network analyses. We discuss the findings in the light of other functional modalities, such as electroencephalography (EEG) and functional magnetic resonance imaging (fMRI). Overall, single-channel MEG results show consistent changes in AD that are in line with EEG studies, but the full potential of the high spatial resolution of MEG and advanced functional connectivity and network analysis has yet to be fully exploited. Adding these features to the current knowledge will potentially aid in uncovering organizational patterns of brain function in AD and thereby aid the understanding of neuronal mechanisms leading to cognitive deficits.
Collapse
Affiliation(s)
- M M A Engels
- Alzheimer Centrum and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| | - W M van der Flier
- Alzheimer Centrum and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; Department of Epidemiology and Biostatistics, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - C J Stam
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| | - A Hillebrand
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Ph Scheltens
- Alzheimer Centrum and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - E C W van Straaten
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Ponomareva NV, Andreeva TV, Protasova MS, Shagam LI, Malina DD, Goltsov AY, Fokin VF, Illarioshkin SN, Rogaev EI. Quantitative EEG during normal aging: association with the Alzheimer's disease genetic risk variant in PICALM gene. Neurobiol Aging 2017; 51:177.e1-177.e8. [DOI: 10.1016/j.neurobiolaging.2016.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 11/14/2016] [Accepted: 12/11/2016] [Indexed: 10/20/2022]
|
28
|
Locating Temporal Functional Dynamics of Visual Short-Term Memory Binding using Graph Modular Dirichlet Energy. Sci Rep 2017; 7:42013. [PMID: 28186173 PMCID: PMC5301217 DOI: 10.1038/srep42013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 01/05/2017] [Indexed: 11/09/2022] Open
Abstract
Visual short-term memory binding tasks are a promising early marker for Alzheimer's disease (AD). To uncover functional deficits of AD in these tasks it is meaningful to first study unimpaired brain function. Electroencephalogram recordings were obtained from encoding and maintenance periods of tasks performed by healthy young volunteers. We probe the task's transient physiological underpinnings by contrasting shape only (Shape) and shape-colour binding (Bind) conditions, displayed in the left and right sides of the screen, separately. Particularly, we introduce and implement a novel technique named Modular Dirichlet Energy (MDE) which allows robust and flexible analysis of the functional network with unprecedented temporal precision. We find that connectivity in the Bind condition is less integrated with the global network than in the Shape condition in occipital and frontal modules during the encoding period of the right screen condition. Using MDE we are able to discern driving effects in the occipital module between 100-140 ms, coinciding with the P100 visually evoked potential, followed by a driving effect in the frontal module between 140-180 ms, suggesting that the differences found constitute an information processing difference between these modules. This provides temporally precise information over a heterogeneous population in promising tasks for the detection of AD.
Collapse
|
29
|
Amyloid β Peptide-Induced Changes in Prefrontal Cortex Activity and Its Response to Hippocampal Input. INTERNATIONAL JOURNAL OF PEPTIDES 2017; 2017:7386809. [PMID: 28127312 PMCID: PMC5239987 DOI: 10.1155/2017/7386809] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 11/02/2016] [Indexed: 12/24/2022]
Abstract
Alterations in prefrontal cortex (PFC) function and abnormalities in its interactions with other brain areas (i.e., the hippocampus) have been related to Alzheimer Disease (AD). Considering that these malfunctions correlate with the increase in the brain's amyloid beta (Aβ) peptide production, here we looked for a causal relationship between these pathognomonic signs of AD. Thus, we tested whether or not Aβ affects the activity of the PFC network and the activation of this cortex by hippocampal input stimulation in vitro. We found that Aβ application to brain slices inhibits PFC spontaneous network activity as well as PFC activation, both at the population and at the single-cell level, when the hippocampal input is stimulated. Our data suggest that Aβ can contribute to AD by disrupting PFC activity and its long-range interactions throughout the brain.
Collapse
|
30
|
Kanda PAM, Oliveira EF, Fraga FJ. EEG epochs with less alpha rhythm improve discrimination of mild Alzheimer's. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2017; 138:13-22. [PMID: 27886711 DOI: 10.1016/j.cmpb.2016.09.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/01/2016] [Accepted: 09/23/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Eyes-closed-awake electroencephalogram (EEG) is a useful tool in the diagnosis of Alzheimer's. However, there is eyes-closed-awake EEG with dominant or rare alpha rhythm. In this paper, we show that random selection of EEG epochs disregarding the alpha rhythm will lead to bias concerning EEG-based Alzheimer's Disease diagnosis. METHODS We compared EEG epochs with more than 30% and with less than 30% alpha rhythm of mild Alzheimer's Disease patients and healthy elderly. We classified epochs as dominant alpha scenario and rare alpha scenario according to alpha rhythm (8-13 Hz) percentage in O1, O2 and Oz channels. Accordingly, we divided the probands into four groups: 17 dominant alpha scenario controls, 15 mild Alzheimer's patients with dominant alpha scenario epochs, 12 rare alpha scenario healthy elderly and 15 mild Alzheimer's Disease patients with rare alpha scenario epochs. We looked for group differences using one-way ANOVA tests followed by post-hoc multiple comparisons (p < 0.05) over normalized energy values (%) on the other four well-known frequency bands (delta, theta, beta and gamma) using two different electrode configurations (parieto-occipital and central). RESULTS After carrying out post-hoc multiple comparisons, for both electrode configurations we found significant differences between mild Alzheimer's patients and healthy elderly on beta- and theta-energy (%) only for the rare alpha scenario. No differences were found for the dominant alpha scenario in any of the five frequency bands. CONCLUSIONS This is the first study of Alzheimer's awake-EEG reporting the influence of alpha rhythm on epoch selection, where our results revealed that, contrarily to what was most likely expected, less synchronized EEG epochs (rare alpha scenario) better discriminated mild Alzheimer's than those presenting abundant alpha (dominant alpha scenario). In addition, we find out that epoch selection is a very sensitive issue in qEEG research. Consequently, for Alzheimer's studies dealing with resting state EEG, we propose that epoch selection strategies should always be cautiously designed and thoroughly explained.
Collapse
Affiliation(s)
| | - Eliezyer F Oliveira
- CECS - Engineering, Modelling and Applied Social Sciences Center, UFABC - Universidade Federal do ABC, Santo André, SP, Brazil
| | - Francisco J Fraga
- CECS - Engineering, Modelling and Applied Social Sciences Center, UFABC - Universidade Federal do ABC, Santo André, SP, Brazil.
| |
Collapse
|
31
|
Fraschini M, Demuru M, Hillebrand A, Cuccu L, Porcu S, Di Stefano F, Puligheddu M, Floris G, Borghero G, Marrosu F. EEG functional network topology is associated with disability in patients with amyotrophic lateral sclerosis. Sci Rep 2016; 6:38653. [PMID: 27924954 PMCID: PMC5141491 DOI: 10.1038/srep38653] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/10/2016] [Indexed: 12/27/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is one of the most severe neurodegenerative diseases, which is known to affect upper and lower motor neurons. In contrast to the classical tenet that ALS represents the outcome of extensive and progressive impairment of a fixed set of motor connections, recent neuroimaging findings suggest that the disease spreads along vast non-motor connections. Here, we hypothesised that functional network topology is perturbed in ALS, and that this reorganization is associated with disability. We tested this hypothesis in 21 patients affected by ALS at several stages of impairment using resting-state electroencephalography (EEG) and compared the results to 16 age-matched healthy controls. We estimated functional connectivity using the Phase Lag Index (PLI), and characterized the network topology using the minimum spanning tree (MST). We found a significant difference between groups in terms of MST dissimilarity and MST leaf fraction in the beta band. Moreover, some MST parameters (leaf, hierarchy and kappa) significantly correlated with disability. These findings suggest that the topology of resting-state functional networks in ALS is affected by the disease in relation to disability. EEG network analysis may be of help in monitoring and evaluating the clinical status of ALS patients.
Collapse
Affiliation(s)
- Matteo Fraschini
- Department of Electrical and Electronic Engineering, University of Cagliari, Piazza D’armi, Cagliari, 09123, Italy
| | - Matteo Demuru
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Centre, Amsterdam, The Netherlands
| | - Arjan Hillebrand
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Centre, Amsterdam, The Netherlands
| | - Lorenza Cuccu
- Biomedical Engineering Course, University of Cagliari, Piazza D’armi, Cagliari, 09123, Italy
| | - Silvia Porcu
- Department of Neurology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| | | | - Monica Puligheddu
- Department of Neurology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| | - Gianluca Floris
- Department of Neurology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| | - Giuseppe Borghero
- Department of Neurology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| | - Francesco Marrosu
- Department of Neurology, AOU Cagliari, University of Cagliari, Cagliari, Italy
| |
Collapse
|
32
|
van Straaten ECW, de Waal H, Lansbergen MM, Scheltens P, Maestu F, Nowak R, Hillebrand A, Stam CJ. Magnetoencephalography for the Detection of Intervention Effects of a Specific Nutrient Combination in Patients with Mild Alzheimer's Disease: Results from an Exploratory Double-Blind, Randomized, Controlled Study. Front Neurol 2016; 7:161. [PMID: 27799918 PMCID: PMC5065957 DOI: 10.3389/fneur.2016.00161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/13/2016] [Indexed: 01/01/2023] Open
Abstract
Synaptic loss is an early pathological finding in Alzheimer’s disease (AD) and correlates with memory impairment. Changes in macroscopic brain activity measured with electro- and magnetoencephalography (EEG and MEG) in AD indicate synaptic changes and may therefore serve as markers of intervention effects in clinical trials. EEG peak frequency and functional networks have shown, in addition to improved memory performance, to be sensitive to detect an intervention effect in mild AD patients of the medical food Souvenaid containing the specific nutrient combination Fortasyn® Connect, which is designed to enhance synapse formation and function. Here, we explore the value of MEG, with higher spatial resolution than EEG, in identifying intervention effects of the nutrient combination by comparing MEG spectral measures, functional connectivity, and networks between an intervention and a control group. Quantitative markers describing spectral properties, functional connectivity, and graph theoretical aspects of MEG from the exploratory 24-week, double-blind, randomized, controlled Souvenir II MEG sub-study (NTR1975, http://www.trialregister.nl) in drug naïve patients with mild AD were compared between a test group (n = 27), receiving Souvenaid, and a control group (n = 28), receiving an isocaloric control product. The groups were unbalanced at screening with respect to Mini-Mental State Examination. Peak frequencies of MEG were compared with EEG peak frequencies, recorded in the same patients at similar time points, were compared with respect to sensitivity to intervention effects. No consistent statistically significant intervention effects were detected. In addition, we found no difference in sensitivity between MEG and EEG peak frequency. This exploratory study could not unequivocally establish the value of MEG in detecting interventional effects on brain activity, possibly due to small sample size and unbalanced study groups. We found no indication that the difference could be attributed to a lack of sensitivity of MEG compared with EEG. MEG in randomized controlled trials is feasible but its value to disclose intervention effects of Souvenaid in mild AD patients needs to be studied further.
Collapse
Affiliation(s)
- Elisabeth C W van Straaten
- Department of Clinical Neurophysiology, MEG Center, VU Medical Center, Amsterdam, Netherlands; Nutricia Advanced Medical Nutrition, Nutricia Research, Utrecht, Netherlands
| | - Hanneke de Waal
- Department of Neurology, Alzheimer Center, VU Medical Center , Amsterdam , Netherlands
| | | | - Philip Scheltens
- Department of Neurology, Alzheimer Center, VU Medical Center , Amsterdam , Netherlands
| | - Fernando Maestu
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology , Madrid , Spain
| | - Rafal Nowak
- Magnetoencephalography Unit, Centro Medico Teknon , Barcelona , Spain
| | - Arjan Hillebrand
- Department of Clinical Neurophysiology, MEG Center, VU Medical Center , Amsterdam , Netherlands
| | - Cornelis J Stam
- Department of Clinical Neurophysiology, MEG Center, VU Medical Center , Amsterdam , Netherlands
| |
Collapse
|
33
|
Barzegaran E, van Damme B, Meuli R, Knyazeva MG. Perception-related EEG is more sensitive to Alzheimer's disease effects than resting EEG. Neurobiol Aging 2016; 43:129-39. [DOI: 10.1016/j.neurobiolaging.2016.03.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 01/06/2023]
|
34
|
Engels MMA, Hillebrand A, van der Flier WM, Stam CJ, Scheltens P, van Straaten ECW. Slowing of Hippocampal Activity Correlates with Cognitive Decline in Early Onset Alzheimer's Disease. An MEG Study with Virtual Electrodes. Front Hum Neurosci 2016; 10:238. [PMID: 27242496 PMCID: PMC4873509 DOI: 10.3389/fnhum.2016.00238] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/05/2016] [Indexed: 11/13/2022] Open
Abstract
Pathology in Alzheimer's disease (AD) starts in the entorhinal cortex and hippocampus. Because of their deep location, activity from these areas is difficult to record with conventional electro- or magnetoencephalography (EEG/MEG). The purpose of this study was to explore hippocampal activity in AD patients and healthy controls using "virtual MEG electrodes". We used resting-state MEG recordings from 27 early onset AD patients [age 60.6 ± 5.4, 12 females, mini-mental state examination (MMSE) range: 19-28] and 26 cognitively healthy age- and gender-matched controls (age 61.8 ± 5.5, 14 females). Activity was reconstructed using beamformer-based virtual electrodes for 78 cortical regions and 6 hippocampal regions. Group differences in peak frequency and relative power in six frequency bands were identified using permutation testing. For the patients, spearman correlations between the MMSE scores and peak frequency or relative power were calculated. Moreover, receiver operator characteristic curves were plotted to estimate the diagnostic accuracy. We found a lower hippocampal peak frequency in AD compared to controls, which, in the patients, correlated positively with MMSE [r(25) = 0.61; p < 0.01] whereas hippocampal relative theta power correlated negatively with MMSE [r(25) = -0.54; p < 0.01]. Cortical peak frequency was also lower in AD in association areas. Furthermore, cortical peak frequency correlated positively with MMSE [r(25) = 0.43; p < 0.05]. In line with this finding, relative theta power was higher in AD across the cortex, and relative alpha and beta power was lower in more circumscribed areas. The average cortical relative theta power was the best discriminator between AD and controls (sensitivity 82%; specificity 81%). Using beamformer-based virtual electrodes, we were able to detect hippocampal activity in AD. In AD, this hippocampal activity is slowed, and correlates better with cognition than the (slowed) activity in cortical areas. On the other hand, the average cortical relative power in the theta band was shown to be the best diagnostic discriminator. We postulate that this novel approach using virtual electrodes can be used in future research to quantify functional interactions between the hippocampi and cortical areas.
Collapse
Affiliation(s)
- Marjolein M A Engels
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, Netherlands
| | - Arjan Hillebrand
- Department of Clinical Neurophysiology and Magnetoencephalography Center, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical CenterAmsterdam, Netherlands; Department of Epidemiology and Biostatistics, Neuroscience Campus Amsterdam, VU University Medical CenterAmsterdam, Netherlands
| | - Cornelis J Stam
- Department of Clinical Neurophysiology and Magnetoencephalography Center, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, Netherlands
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, Netherlands
| | - Elisabeth C W van Straaten
- Department of Clinical Neurophysiology and Magnetoencephalography Center, Neuroscience Campus Amsterdam, VU University Medical CenterAmsterdam, Netherlands; Nutricia Advanced Medical Nutrition, Nutricia ResearchUtrecht, Netherlands
| |
Collapse
|
35
|
Yu M, Gouw AA, Hillebrand A, Tijms BM, Stam CJ, van Straaten ECW, Pijnenburg YAL. Different functional connectivity and network topology in behavioral variant of frontotemporal dementia and Alzheimer's disease: an EEG study. Neurobiol Aging 2016; 42:150-62. [PMID: 27143432 DOI: 10.1016/j.neurobiolaging.2016.03.018] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 03/11/2016] [Accepted: 03/15/2016] [Indexed: 10/22/2022]
Abstract
We investigated whether the functional connectivity and network topology in 69 Alzheimer's disease (AD), 48 behavioral variant of frontotemporal dementia (bvFTD) patients, and 64 individuals with subjective cognitive decline are different using resting-state electroencephalography recordings. Functional connectivity between all pairs of electroencephalography channels was assessed using the phase lag index (PLI). We subsequently calculated PLI-weighted networks, from which minimum spanning trees (MSTs) were constructed. Finally, we investigated the hierarchical clustering organization of the MSTs. Functional connectivity analysis showed frequency-dependent results: in the delta band, bvFTD showed highest whole-brain PLI; in the theta band, the whole-brain PLI in AD was higher than that in bvFTD; in the alpha band, AD showed lower whole-brain PLI compared with bvFTD and subjective cognitive decline. The MST results indicate that frontal networks appear to be selectively involved in bvFTD against the background of preserved global efficiency, whereas parietal and occipital loss of network organization in AD is accompanied by global efficiency loss. Our findings suggest different pathophysiological mechanisms in these 2 separate neurodegenerative disorders.
Collapse
Affiliation(s)
- Meichen Yu
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, the Netherlands.
| | - Alida A Gouw
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, the Netherlands; Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| | - Arjan Hillebrand
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Betty M Tijms
- Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| | - Cornelis Jan Stam
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Elisabeth C W van Straaten
- Department of Clinical Neurophysiology and MEG Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
36
|
Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, Bakardjian H, Benali H, Bertram L, Blennow K, Broich K, Cavedo E, Crutch S, Dartigues JF, Duyckaerts C, Epelbaum S, Frisoni GB, Gauthier S, Genthon R, Gouw AA, Habert MO, Holtzman DM, Kivipelto M, Lista S, Molinuevo JL, O'Bryant SE, Rabinovici GD, Rowe C, Salloway S, Schneider LS, Sperling R, Teichmann M, Carrillo MC, Cummings J, Jack CR. Preclinical Alzheimer's disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement 2016; 12:292-323. [PMID: 27012484 PMCID: PMC6417794 DOI: 10.1016/j.jalz.2016.02.002] [Citation(s) in RCA: 1238] [Impact Index Per Article: 137.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During the past decade, a conceptual shift occurred in the field of Alzheimer's disease (AD) considering the disease as a continuum. Thanks to evolving biomarker research and substantial discoveries, it is now possible to identify the disease even at the preclinical stage before the occurrence of the first clinical symptoms. This preclinical stage of AD has become a major research focus as the field postulates that early intervention may offer the best chance of therapeutic success. To date, very little evidence is established on this "silent" stage of the disease. A clarification is needed about the definitions and lexicon, the limits, the natural history, the markers of progression, and the ethical consequence of detecting the disease at this asymptomatic stage. This article is aimed at addressing all the different issues by providing for each of them an updated review of the literature and evidence, with practical recommendations.
Collapse
Affiliation(s)
- Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France.
| | - Harald Hampel
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France; AXA Research Fund & UPMC Chair, Paris, France
| | | | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center and Neuroscience Campus, Amsterdam, The Netherlands
| | - Paul Aisen
- University of Southern California San Diego, CA, USA
| | - Sandrine Andrieu
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France
| | - Hovagim Bakardjian
- IHU-A-ICM-Institut des Neurosciences translationnelles de Paris, Paris, France
| | - Habib Benali
- INSERM U1146-CNRS UMR 7371-UPMC UM CR2, Site Pitié-Salpêtrière, Paris, France
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany; School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Lab, Department of Neuroscience and Physiology, University of Gothenburg, Mölndal Hospital, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Enrica Cavedo
- AXA Research Fund & UPMC Chair, Paris, France; Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Sebastian Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | | | - Charles Duyckaerts
- University Pierre et Marie Curie, Assistance Publique des Hôpitaux de Paris, Alzheimer-Prion Team Institut du Cerveau et de la Moelle (ICM), Paris, France
| | - Stéphane Epelbaum
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Giovanni B Frisoni
- University Hospitals and University of Geneva, Geneva, Switzerland; IRCCS Fatebenefratelli, Brescia, Italy
| | - Serge Gauthier
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Canada
| | - Remy Genthon
- Fondation pour la Recherche sur Alzheimer, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alida A Gouw
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France; Department of Clinical Neurophysiology/MEG Center, VU University Medical Center, Amsterdam
| | - Marie-Odile Habert
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Département de Médecine Nucléaire, Paris, France
| | - David M Holtzman
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, St. Louis, MO, USA; Department of Neurology, Washington University, Knight Alzheimer's Disease Research Center, St. Louis, MO, USA
| | - Miia Kivipelto
- Center for Alzheimer Research, Karolinska Institutet, Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden; Institute of Clinical Medicine/ Neurology, University of Eastern Finland, Kuopio, Finland
| | | | - José-Luis Molinuevo
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Sid E O'Bryant
- Center for Alzheimer's & Neurodegenerative Disease Research, University of North Texas Health Science Center, TX, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Christopher Rowe
- Department of Molecular Imaging, Austin Health, University of Melbourne, Australia
| | - Stephen Salloway
- Memory and Aging Program, Butler Hospital, Alpert Medical School of Brown University, USA; Department of Neurology, Alpert Medical School of Brown University, USA; Department of Psychiatry, Alpert Medical School of Brown University, USA
| | - Lon S Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Reisa Sperling
- Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, USA; Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Massachusetts General Hospital, Boston, USA
| | - Marc Teichmann
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Maria C Carrillo
- The Alzheimer's Association Division of Medical & Scientific Relations, Chicago, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Cliff R Jack
- Department of Radiology, Mayo Clinic, Rochester MN, USA
| |
Collapse
|
37
|
Escudero J. Open your eyes and you will see. Changes in “eyes-open” versus “eyes-closed” small-world properties of EEG functional connectivity in amnesic mild cognitive impairment. Clin Neurophysiol 2016; 127:999-1000. [DOI: 10.1016/j.clinph.2015.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 09/03/2015] [Accepted: 09/05/2015] [Indexed: 01/06/2023]
|
38
|
Cicchese JJ, Berry SD. Hippocampal Non-Theta-Contingent Eyeblink Classical Conditioning: A Model System for Neurobiological Dysfunction. Front Psychiatry 2016; 7:1. [PMID: 26903886 PMCID: PMC4751249 DOI: 10.3389/fpsyt.2016.00001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 01/01/2016] [Indexed: 11/30/2022] Open
Abstract
Typical information processing is thought to depend on the integrity of neurobiological oscillations that may underlie coordination and timing of cells and assemblies within and between structures. The 3-7 Hz bandwidth of hippocampal theta rhythm is associated with cognitive processes essential to learning and depends on the integrity of cholinergic, GABAergic, and glutamatergic forebrain systems. Since several significant psychiatric disorders appear to result from dysfunction of medial temporal lobe (MTL) neurochemical systems, preclinical studies on animal models may be an important step in defining and treating such syndromes. Many studies have shown that the amount of hippocampal theta in the rabbit strongly predicts the acquisition rate of classical eyeblink conditioning and that impairment of this system substantially slows the rate of learning and attainment of asymptotic performance. Our lab has developed a brain-computer interface that makes eyeblink training trials contingent upon the explicit presence or absence of hippocampal theta. The behavioral benefit of theta-contingent training has been demonstrated in both delay and trace forms of the paradigm with a two- to fourfold increase in learning speed over non-theta states. The non-theta behavioral impairment is accompanied by disruption of the amplitude and synchrony of hippocampal local field potentials, multiple-unit excitation, and single-unit response patterns dependent on theta state. Our findings indicate a significant electrophysiological and behavioral impact of the pretrial state of the hippocampus that suggests an important role for this MTL system in associative learning and a significant deleterious impact in the absence of theta. Here, we focus on the impairments in the non-theta state, integrate them into current models of psychiatric disorders, and suggest how improvement in our understanding of neurobiological oscillations is critical for theories and treatment of psychiatric pathology.
Collapse
Affiliation(s)
- Joseph J Cicchese
- Department of Psychology, Center for Neuroscience, Miami University , Oxford, OH , USA
| | - Stephen D Berry
- Department of Psychology, Center for Neuroscience, Miami University , Oxford, OH , USA
| |
Collapse
|
39
|
Cromarty RA, Elder GJ, Graziadio S, Baker M, Bonanni L, Onofrj M, O'Brien JT, Taylor JP. Neurophysiological biomarkers for Lewy body dementias. Clin Neurophysiol 2015; 127:349-359. [PMID: 26183755 PMCID: PMC4727506 DOI: 10.1016/j.clinph.2015.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 06/05/2015] [Accepted: 06/23/2015] [Indexed: 11/07/2022]
Abstract
Biomarkers are needed to improve Lewy body dementia (LBD) diagnosis and measure treatment response. There is substantial heterogeneity in neurophysiology biomarker methodologies limiting comparison. However, there is tentative evidence to suggest neurophysiological approaches may show promise as potential biomarkers of LBD.
Objective Lewy body dementias (LBD) include both dementia with Lewy bodies (DLB) and Parkinson’s disease with dementia (PDD), and the differentiation of LBD from other neurodegenerative dementias can be difficult. Currently, there are few biomarkers which might assist early diagnosis, map onto LBD symptom severity, and provide metrics of treatment response. Traditionally, biomarkers in LBD have focussed on neuroimaging modalities; however, as biomarkers need to be simple, inexpensive and non-invasive, neurophysiological approaches might also be useful as LBD biomarkers. Methods In this review, we searched PubMED and PsycINFO databases in a semi-systematic manner in order to identify potential neurophysiological biomarkers in the LBDs. Results We identified 1491 studies; of these, 37 studies specifically examined neurophysiological biomarkers in LBD patients. We found that there was substantial heterogeneity with respect to methodologies and patient cohorts. Conclusion Generally, many of the findings have yet to be replicated, although preliminary findings reinforce the potential utility of approaches such as quantitative electroencephalography and motor cortical stimulation paradigms. Significance Various neurophysiological techniques have the potential to be useful biomarkers in the LBDs. We recommend that future studies focus on maximising the diagnostic specificity and sensitivity of the most promising neurophysiological biomarkers.
Collapse
Affiliation(s)
- Ruth A Cromarty
- Institute of Neuroscience, Campus for Aging and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| | - Greg J Elder
- Institute of Neuroscience, Campus for Aging and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Sara Graziadio
- Institute of Neuroscience, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Mark Baker
- Institute of Neuroscience, Framlington Place, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Laura Bonanni
- Clinica Neurologica, Dipartimento di Neuroscienze e Imaging, Università "G.D'Annunzio" Chieti-Pescara, Italy
| | - Marco Onofrj
- Clinica Neurologica, Dipartimento di Neuroscienze e Imaging, Università "G.D'Annunzio" Chieti-Pescara, Italy
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0SP, UK
| | - John-Paul Taylor
- Institute of Neuroscience, Campus for Aging and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|