51
|
Zheng X, Cawood J, Hayre C, Wang S. Computer assisted diagnosis of Alzheimer's disease using statistical likelihood-ratio test. PLoS One 2023; 18:e0279574. [PMID: 36800393 PMCID: PMC9937475 DOI: 10.1371/journal.pone.0279574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 12/11/2022] [Indexed: 02/18/2023] Open
Abstract
The purpose of this work is to present a computer assisted diagnostic tool for radiologists in their diagnosis of Alzheimer's disease. A statistical likelihood-ratio procedure from signal detection theory was implemented in the detection of Alzheimer's disease. The probability density functions of the likelihood ratio were constructed by using medial temporal lobe (MTL) volumes of patients with Alzheimer's disease (AD) and normal controls (NC). The volumes of MTL as well as other anatomical regions of the brains were calculated by the FreeSurfer software using T1 weighted MRI images. The MRI images of AD and NC were downloaded from the database of Alzheimer's disease neuroimaging initiative (ADNI). A separate dataset of minimal interval resonance imaging in Alzheimer's disease (MIRIAD) was used for diagnostic testing. A sensitivity of 89.1% and specificity of 87.0% were achieved for the MIRIAD dataset which are better than the 85% sensitivity and specificity achieved by the best radiologists without input of other patient information.
Collapse
Affiliation(s)
- Xiaoming Zheng
- Medical Radiation Science, School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
- * E-mail:
| | - Justin Cawood
- Medical Radiation Science, School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Chris Hayre
- Medical Radiation Science, School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Shaoyu Wang
- Biomedical Sciences, School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | | |
Collapse
|
52
|
Makdissi S, Parsons BD, Di Cara F. Towards early detection of neurodegenerative diseases: A gut feeling. Front Cell Dev Biol 2023; 11:1087091. [PMID: 36824371 PMCID: PMC9941184 DOI: 10.3389/fcell.2023.1087091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
The gastrointestinal tract communicates with the nervous system through a bidirectional network of signaling pathways called the gut-brain axis, which consists of multiple connections, including the enteric nervous system, the vagus nerve, the immune system, endocrine signals, the microbiota, and its metabolites. Alteration of communications in the gut-brain axis is emerging as an overlooked cause of neuroinflammation. Neuroinflammation is a common feature of the pathogenic mechanisms involved in various neurodegenerative diseases (NDs) that are incurable and debilitating conditions resulting in progressive degeneration and death of neurons, such as in Alzheimer and Parkinson diseases. NDs are a leading cause of global death and disability, and the incidences are expected to increase in the following decades if prevention strategies and successful treatment remain elusive. To date, the etiology of NDs is unclear due to the complexity of the mechanisms of diseases involving genetic and environmental factors, including diet and microbiota. Emerging evidence suggests that changes in diet, alteration of the microbiota, and deregulation of metabolism in the intestinal epithelium influence the inflammatory status of the neurons linked to disease insurgence and progression. This review will describe the leading players of the so-called diet-microbiota-gut-brain (DMGB) axis in the context of NDs. We will report recent findings from studies in model organisms such as rodents and fruit flies that support the role of diets, commensals, and intestinal epithelial functions as an overlooked primary regulator of brain health. We will finish discussing the pivotal role of metabolisms of cellular organelles such as mitochondria and peroxisomes in maintaining the DMGB axis and how alteration of the latter can be used as early disease makers and novel therapeutic targets.
Collapse
Affiliation(s)
- Stephanie Makdissi
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS, Canada
- IWK Health Centre, Department of Pediatrics, Halifax, Canada
| | - Brendon D. Parsons
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS, Canada
| | - Francesca Di Cara
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS, Canada
- IWK Health Centre, Department of Pediatrics, Halifax, Canada
| |
Collapse
|
53
|
Milani A, Morawski M, Bechmann I. Inter-hemispherical comparison of tau-pathology in the human temporal lobe. Ann Anat 2023; 246:152042. [PMID: 36592871 DOI: 10.1016/j.aanat.2022.152042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is characterized histopathologically by hyperphosphorylated and aggregated Tau and amyloid plaques. While the latter appear in a less stringent way throughout the brain in the course of the disease, the former evolve in a highly predictable pattern as described by Braak and Braak (1991). It is, however, not clear if this pattern develops simultaneously in both hemispheres. In this study, we therefore compared Tau-pathology of both hemispheres of the same individual in 36 consecutive brain donations as they arrived in our brain bank. 26 exhibited little differences, in eight cases left hemisphere was clearly more affected and in two cases the right hemisphere. Thus, cases with evident interhemispheric Tau-pathology do exist and interhemispheric comparison in such cases may help to identify driving forces in the progression of AD.
Collapse
Affiliation(s)
| | - Markus Morawski
- Paul-Flechsig-Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Universität Leipzig, Leipzig, Germany.
| |
Collapse
|
54
|
Daniel E, Deng F, Patel SK, Sedrak MS, Kim H, Razavi M, Sun CL, Root JC, Ahles TA, Dale W, Chen BT. Cortical thinning in chemotherapy-treated older long-term breast cancer survivors. Brain Imaging Behav 2023; 17:66-76. [PMID: 36369620 PMCID: PMC10156471 DOI: 10.1007/s11682-022-00743-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2022] [Indexed: 11/13/2022]
Abstract
Cognitive decline is an increasing issue for cancer survivors, especially for older adults, as chemotherapy affects brain structure and function. The purpose of this single center study was to evaluate alterations in cortical thickness and cognition in older long-term survivors of breast cancer who had been treated with chemotherapy years ago. In this prospective cohort study, we enrolled 3 groups of women aged ≥ 65 years with a history of stage I-III breast cancer who had received adjuvant chemotherapy 5 to 15 years ago (chemotherapy group, C +), age-matched women with breast cancer but no chemotherapy (no-chemotherapy group, C-) and healthy controls (HC). All participants underwent brain magnetic resonance imaging and neuropsychological testing with the NIH Toolbox Cognition Battery at time point 1 (TP1) and again at 2 years after enrollment (time point 2 (TP2)). At TP1, there were no significant differences in cortical thickness among the 3 groups. Longitudinally, the C + group showed cortical thinning in the fusiform gyrus (p = 0.006, effect size (d) = -0.60 [ -1.86, -0.66]), pars triangularis (p = 0.026, effect size (d) = -0.43 [-1.68, -0.82]), and inferior temporal lobe (p = 0.026, effect size (d) = -0.38 [-1.62, -0.31]) of the left hemisphere. The C + group also showed decreases in neuropsychological scores such as the total composite score (p = 0.01, effect size (d) = -3.9726 [-0.9656, -6.9796], fluid composite score (p = 0.03, effect size (d) = -4.438 [-0.406, -8.47], and picture vocabulary score (p = 0.04, effect size (d) = -3.7499 [-0.0617, -7.438]. Our results showed that cortical thickness could be a candidate neuroimaging biomarker for cancer-related cognitive impairment and accelerated aging in older long-term cancer survivors.
Collapse
Affiliation(s)
- Ebenezer Daniel
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Frank Deng
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Sunita K Patel
- Department of Population Science, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Mina S Sedrak
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Heeyoung Kim
- Center for Cancer and Aging, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Marianne Razavi
- Department of Supportive Care Medicine, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Can-Lan Sun
- Center for Cancer and Aging, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - James C Root
- Neurocognitive Research Lab, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tim A Ahles
- Neurocognitive Research Lab, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William Dale
- Center for Cancer and Aging, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.,Department of Supportive Care Medicine, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Bihong T Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, 91010, USA. .,Center for Cancer and Aging, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
55
|
Lin CT, Ghosh S, Hinkley LB, Dale CL, Souza ACS, Sabes JH, Hess CP, Adams ME, Cheung SW, Nagarajan SS. Multi-tasking deep network for tinnitus classification and severity prediction from multimodal structural MR images. J Neural Eng 2023; 20. [PMID: 36595270 DOI: 10.1088/1741-2552/acab33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Objective:Subjective tinnitus is an auditory phantom perceptual disorder without an objective biomarker. Fast and efficient diagnostic tools will advance clinical practice by detecting or confirming the condition, tracking change in severity, and monitoring treatment response. Motivated by evidence of subtle anatomical, morphological, or functional information in magnetic resonance images of the brain, we examine data-driven machine learning methods for joint tinnitus classification (tinnitus or no tinnitus) and tinnitus severity prediction.Approach:We propose a deep multi-task multimodal framework for tinnitus classification and severity prediction using structural MRI (sMRI) data. To leverage complementary information multimodal neuroimaging data, we integrate two modalities of three-dimensional sMRI-T1 weighted (T1w) and T2 weighted (T2w) images. To explore the key components in the MR images that drove task performance, we segment both T1w and T2w images into three different components-cerebrospinal fluid, grey matter and white matter, and evaluate performance of each segmented image.Main results:Results demonstrate that our multimodal framework capitalizes on the information across both modalities (T1w and T2w) for the joint task of tinnitus classification and severity prediction.Significance:Our model outperforms existing learning-based and conventional methods in terms of accuracy, sensitivity, specificity, and negative predictive value.
Collapse
Affiliation(s)
- Chieh-Te Lin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, United States of America
| | - Sanjay Ghosh
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, United States of America
| | - Leighton B Hinkley
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, United States of America
| | - Corby L Dale
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, United States of America
| | - Ana C S Souza
- Department of Telecommunication and Mechatronics Engineering, Federal University of Sao Joao del-Rei, Praca Frei Orlando, 170, Sao Joao del Rei 36307, MG, Brazil
| | - Jennifer H Sabes
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, 2380 Sutter St., San Francisco, CA 94115, United States of America
| | - Christopher P Hess
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, United States of America
| | - Meredith E Adams
- Department of Otolaryngology-Head and Neck Surgery, University of Minnesota, Phillips Wangensteen Building, 516 Delaware St., Minneapolis, MN 55455, United States of America
| | - Steven W Cheung
- Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, 2380 Sutter St., San Francisco, CA 94115, United States of America.,Surgical Services, Veterans Affairs, 4150 Clement St., San Francisco, CA 94121, United States of America
| | - Srikantan S Nagarajan
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, United States of America.,Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, 2380 Sutter St., San Francisco, CA 94115, United States of America.,Surgical Services, Veterans Affairs, 4150 Clement St., San Francisco, CA 94121, United States of America
| |
Collapse
|
56
|
Lu H, Li J, Chan SSM, Yue WWY, Lam LCW. Decoding the radiomic features of dorsolateral prefrontal cortex in individuals with accelerated cortical changes: implications for personalized transcranial magnetic stimulation. J Med Imaging (Bellingham) 2023; 10:015001. [PMID: 36619873 PMCID: PMC9811135 DOI: 10.1117/1.jmi.10.1.015001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023] Open
Abstract
Purpose Image-guided transcranial magnetic stimulation (TMS) is an emerging research field in neuroscience and rehabilitation medicine. Cortical morphometry, as a radiomic phenotype of aging, plays a vital role in developing personalized TMS model, yet few studies are afoot to examine the aging effects on region-specific morphometry and use it in the estimation of TMS-induced electric fields. Our study was aimed to investigate the radiomic features of bilateral dorsolateral prefrontal cortex (DLPFC) and quantify the TMS-induced electric fields during aging. Approach Baseline, 1-year and 3-year structural magnetic resonance imaging (MRI) scans from normal aging (NA) adults ( n = 32 ) and mild cognitive impairment (MCI) converters ( n = 22 ) were drawn from the Open Access Series of Imaging Studies. The quantitative measures of radiomics included cortical thickness, folding, and scalp-to-cortex distance. Realistic head models were developed to simulate the impacts of radiomic features on TMS-induced E-fields using the finite-element method. Results A pronounced aging-related decrease was found in the gyrification of left DLPFC in MCI converters ( t = 2.21 , p = 0.035 ), which could predict the decline of global cognition at 3-year follow up. Along with the decreased gyrification in left DLPFC, the magnitude of TMS-induced E-fields was rapidly decreased in MCI converters ( t = 2.56 , p = 0.018 ). Conclusions MRI-informed radiomic features of the treatment targets have significant effects on the intensity and distribution of the stimulation-induced electric fields in prodromal dementia patients. Our findings highlight the importance of region-specific radiomics when conducting the transcranial brain stimulation in individuals with accelerated cortical changes, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Hanna Lu
- The Chinese University of Hong Kong, Department of Psychiatry, Hong Kong, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Centre for Neuromodulation and Rehabilitation, Guangzhou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- The Chinese University of Hong Kong, Department of Psychiatry, Hong Kong, China
| | - Sandra Sau Man Chan
- The Chinese University of Hong Kong, Department of Psychiatry, Hong Kong, China
| | | | - Linda Chiu Wa Lam
- The Chinese University of Hong Kong, Department of Psychiatry, Hong Kong, China
| |
Collapse
|
57
|
Wan K, Yin W, Tang Y, Zhu W, Wang Z, Zhou X, Zhang W, Zhang C, Yu X, Zhao W, Li C, Zhu X, Sun Z. Brain Gray Matter Volume Mediated the Correlation Between Plasma P-Tau and Cognitive Function of Early Alzheimer's Disease in China: A Cross-Sectional Observational Study. J Alzheimers Dis 2023; 92:81-93. [PMID: 36710682 DOI: 10.3233/jad-221100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND The primary manifestations of Alzheimer's disease (AD) include cognitive decline and brain gray matter volume (GMV) atrophy. Recent studies have found that plasma phosphorylated-tau (p-tau) concentrations perform better in diagnosing, differentiating, and monitoring the progression of AD. However, the correlation between plasma p-tau, GMV, and cognition remains unclear. OBJECTIVE To investigate whether GMV plays a mediating role in the association between plasma p-tau concentrations and cognition. METHODS In total, 99 participants (47 patients with AD and 52 cognitively unimpaired [CU] individuals) were included. All participants underwent neuropsychological assessments, laboratory examinations, and magnetic resonance imaging scans. Plasma p-tau217 and p-tau181 concentrations were measured using an enzyme-linked immunosorbent assay kit. Voxel-based morphometry was performed to assess participants' brain GMV. Partial correlation and mediation analyses were conducted in AD group. RESULTS Plasma p-tau concentrations were significantly higher in the AD group than in the CU group. Patients with AD had significant brain GMV atrophy in the right hippocampus, bilateral middle temporal gyrus, and right inferior temporal gyrus. In the AD group, there were significant correlations between plasma p-tau217 concentrations, GMV, and Mini-Mental State Examination (MMSE) scores. Brain GMV of the right hippocampus mediated the association between plasma p-tau217 concentrations and MMSE scores. A significant correlation between plasma p-tau181 and MMSE scores was not identified. CONCLUSION The findings indicate that p-tau217 is a promising biomarker for central processes affecting brain GMV and cognitive function. This may provide potential targets for future intervention and treatment of tau-targeting therapies in the early stages of AD.
Collapse
Affiliation(s)
- Ke Wan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Wenwen Yin
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yating Tang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Wenhao Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhiqiang Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Private Bag 23, Hobart, Tasmania, Australia
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Wei Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Cun Zhang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xianfeng Yu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenming Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenchen Li
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xiaoqun Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
58
|
Bramen JE, Siddarth P, Popa ES, Kress GT, Rapozo MK, Hodes JF, Ganapathi AS, Slyapich CB, Glatt RM, Pierce K, Porter VR, Wong C, Kim M, Dye RV, Panos S, Bookheimer T, Togashi T, Loong S, Raji CA, Bookheimer SY, Roach JC, Merrill DA. Impact of Eating a Carbohydrate-Restricted Diet on Cortical Atrophy in a Cross-Section of Amyloid Positive Patients with Alzheimer's Disease: A Small Sample Study. J Alzheimers Dis 2023; 96:329-342. [PMID: 37742646 PMCID: PMC10657694 DOI: 10.3233/jad-230458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND A carbohydrate-restricted diet aimed at lowering insulin levels has the potential to slow Alzheimer's disease (AD). Restricting carbohydrate consumption reduces insulin resistance, which could improve glucose uptake and neural health. A hallmark feature of AD is widespread cortical thinning; however, no study has demonstrated that lower net carbohydrate (nCHO) intake is linked to attenuated cortical atrophy in patients with AD and confirmed amyloidosis. OBJECTIVE We tested the hypothesis that individuals with AD and confirmed amyloid burden eating a carbohydrate-restricted diet have thicker cortex than those eating a moderate-to-high carbohydrate diet. METHODS A total of 31 patients (mean age 71.4±7.0 years) with AD and confirmed amyloid burden were divided into two groups based on a 130 g/day nCHO cutoff. Cortical thickness was estimated from T1-weighted MRI using FreeSurfer. Cortical surface analyses were corrected for multiple comparisons using cluster-wise probability. We assessed group differences using a two-tailed two-independent sample t-test. Linear regression analyses using nCHO as a continuous variable, accounting for confounders, were also conducted. RESULTS The lower nCHO group had significantly thicker cortex within somatomotor and visual networks. Linear regression analysis revealed that lower nCHO intake levels had a significant association with cortical thickness within the frontoparietal, cingulo-opercular, and visual networks. CONCLUSIONS Restricting carbohydrates may be associated with reduced atrophy in patients with AD. Lowering nCHO to under 130 g/day would allow patients to follow the well-validated MIND diet while benefiting from lower insulin levels.
Collapse
Affiliation(s)
- Jennifer E. Bramen
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Saint John’s Cancer Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Prabha Siddarth
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Emily S. Popa
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - Gavin T. Kress
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Molly K. Rapozo
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - John F. Hodes
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Aarthi S. Ganapathi
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - Colby B. Slyapich
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - Ryan M. Glatt
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - Kyron Pierce
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - Verna R. Porter
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Saint John’s Cancer Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Claudia Wong
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Mihae Kim
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Richelin V. Dye
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Loma Linda University, School of Medicine and School of Behavioral Health, Loma Linda, CA, USA
| | - Stella Panos
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - Tess Bookheimer
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
| | - Tori Togashi
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Loma Linda University, School of Medicine and School of Behavioral Health, Loma Linda, CA, USA
| | - Spencer Loong
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Loma Linda University, School of Medicine and School of Behavioral Health, Loma Linda, CA, USA
| | - Cyrus A. Raji
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Susan Y. Bookheimer
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | | | - David A. Merrill
- Pacific Brain Health Center, Pacific Neuroscience Institute and Foundation, Santa Monica, CA, USA
- Saint John’s Cancer Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Santa Monica, CA, USA
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
59
|
Sur C, Adamczuk K, Scott D, Kost J, Sampat M, Buckley C, Farrar G, Newton B, Suhy J, Bennacef I, Egan MF. Evaluation of 18F-flutemetamol amyloid PET image analysis parameters on the effect of verubecestat on brain amlyoid load in Alzheimer's disease. Mol Imaging Biol 2022; 24:862-873. [PMID: 35794343 DOI: 10.1007/s11307-022-01735-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/17/2022] [Accepted: 04/21/2022] [Indexed: 12/29/2022]
Abstract
PURPOSE The BACE inhibitor verubecestat was previously found to reduce amyloid load as assessed by 18F-flutemetamol positron emission tomography (PET) composite cortical standard uptake value ratio (SUVr) in patients with mild-to-moderate Alzheimer's disease (AD) in a substudy of the EPOCH trial. Here, we report on additional analyses relevant to the EPOCH PET data, to help inform on the use of PET for assessing amlyloid load in AD clinical trials. PROCEDURES The analyses addressed (1) identification of an optimal 18F-flutemetamol reference region, (2) determination of the threshold to characterize the magnitude of the longitudinal change, and (3) the impact of partial volume correction (PVC). Pons and subcortical white matter were evaluated as reference regions. The SUVr cutoffs and final reference region choice were determined using 162 18F-flutemetamol PET scans from the AIBL dataset. 18F-flutemetamol SUVrs were computed at baseline and at Week 78 in EPOCH participants who received verubecestat 12 mg (n = 14), 40 mg (n = 20), or placebo (n = 20). Drug effects on amyloid load were computed using either Meltzer (MZ), or symmetric geometric transfer matrix (SGTM) PVC and compared to uncorrected data. RESULTS The optimal subcortical white matter and pons SUVr cutoffs were determined to be 0.69 and 0.62, respectively. The effect size to detect longitudinal change was higher for subcortical white matter (1.20) than pons (0.45). Hence, subcortical white matter was used as the reference region for the EPOCH PET substudy. In EPOCH, uncorrected baseline SUVr values correlated strongly with MZ PVC (r2 = 0.94) and SGTM PVC (r2 = 0.92) baseline SUVr values, and PVC did not provide improvement for evaluating treatment effects on amyloid load at Week 78. No change from baseline was observed in the placebo group at Week 78, whereas a 0.02 and a 0.04 decrease in SUVr were observed in the 12 mg and 40 mg arms, with the latter representing a 22% reduction in the amyloid load above the detection threshold. CONCLUSIONS Treatment-related 18F-flutemetamol longitudinal changes in AD clinical trials can be quantified using a subcortical white matter reference region without PVC. CLINICAL TRIAL REGISTRATION clinicaltrials.gov NCT01739348.
Collapse
|
60
|
Fani N, Eghbalzad L, Harnett NG, Carter SE, Price M, Stevens JS, Ressler KJ, van Rooij SJH, Bradley B. Racial discrimination associates with lower cingulate cortex thickness in trauma-exposed black women. Neuropsychopharmacology 2022; 47:2230-2237. [PMID: 36100659 PMCID: PMC9630426 DOI: 10.1038/s41386-022-01445-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/14/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022]
Abstract
Racial discrimination (RD) has been consistently linked to adverse brain health outcomes. These may be due in part to RD effects on neural networks involved with threat appraisal and regulation; RD has been linked to altered activity in the rostral anterior cingulate cortex (rACC) and structural decrements in the anterior cingulum bundle and hippocampus. In the present study, we examined associations of RD with cingulate, hippocampus and amygdala gray matter morphology in a sample of trauma-exposed Black women. Eighty-one Black women aged 19-62 years were recruited as part of an ongoing study of trauma. Participants completed assessments of RD, trauma exposure, and posttraumatic stress disorder (PTSD), and underwent T1-weighted anatomical imaging. Cortical thickness, surface area and gray matter volume were extracted from subregions of cingulate cortex, and gray matter volume was extracted from amygdala and hippocampus, and entered into partial correlation analyses that included RD and other socio-environmental variables. After correction for multiple comparisons and accounting for variance associated with other stressors and socio-environmental factors, participants with more RD exposure showed proportionally lower cortical thickness in the left rACC, caudal ACC, and posterior cingulate cortex (ps < = 0.01). These findings suggest that greater experiences of RD are linked to compromised cingulate gray matter thickness. In the context of earlier findings indicating that RD produces increased response in threat neurocircuitry, our data suggest that RD may increase vulnerability for brain health problems via cingulate cortex alterations. Further research is needed to elucidate biological mechanisms for these changes.
Collapse
Affiliation(s)
- Negar Fani
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.
| | - Leyla Eghbalzad
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Nathaniel G Harnett
- Division of Depression and Anxiety, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Sierra E Carter
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | - Matthew Price
- Department of Psychological Science, University of Vermont, Burlington, VT, USA
| | - Jennifer S Stevens
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Kerry J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Division of Depression and Anxiety, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Sanne J H van Rooij
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Bekh Bradley
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
61
|
Miletic-Drakulic S, Miloradovic I, Jankovic V, Azanjac-Arsic A, Lazarevic S. VEP Score of a Left Eye Had Predictive Values for Achieving NEDA-3 over Ten Years in Patients with Multiple Sclerosis. SENSORS (BASEL, SWITZERLAND) 2022; 22:8849. [PMID: 36433445 PMCID: PMC9696926 DOI: 10.3390/s22228849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The aim of this study was to determine the predictive value of visual evoked potentials (VEPs) in patients with relapsing-remitting multiple sclerosis (RRMS) in achieving no evidence of disease activity-3 (NEDA-3) during up to 10 years of first-line immunomodulatory therapy and to determine whether the lateralization of optic nerve damage may have prognostic significance concerning clinical disability and response to therapy. METHODS In a retrospective study, a total of 83 patients (53 female and 30 male) with RRMS participated. The average age of patients was 38.31 ± 9.01. Patients were followed for 2, 5 or 10 years. VEPs were measured at the beginning of the follow-up and after many years of monitoring. Data on optical neuritis (ON) were obtained from medical history. The degree of disability was estimated by the neurologist (independent rater), and magnetic resonance (MR) imaging of the endocranium was performed with gadolinium contrasts. Achieving NEDA-3 is considered a favorable outcome of treatments. RESULTS Among those treated, 19 (22.9%) reached NEDA-3, while 64 (77.1%) did not reach NEDA-3. The values of the evoked potential (EP) score for the left eye (r = 0.008, odds ratio (OR) = 0.344 (0.156-0.757)) and latency for the left eye (r = 0.042, OR = 0.966 (0.934-0.999)) at the onset of disease were predictive factors for achieving NEDA-3. CONCLUSIONS A normal VEP at the beginning of RRMS increases the chance of reaching NEDA-3 by about six times.
Collapse
|
62
|
Bocci T, Baloscio D, Ferrucci R, Briscese L, Priori A, Sartucci F. Interhemispheric Connectivity in Idiopathic Cervical Dystonia and Spinocerebellar Ataxias: A Transcranial Magnetic Stimulation Study. Clin EEG Neurosci 2022; 53:460-466. [PMID: 32938220 DOI: 10.1177/1550059420957487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND RATIONALE Hyperkinetic movement disorders represent a heterogeneous group of diseases, different from a genetic and clinical perspective. In the past, neurophysiological approaches provided different, sometimes contradictory findings, pointing to an impaired cortical inhibition as a common electrophysiological marker. Our aim was to evaluate changes in interhemispheric communication in patients with idiopathic cervical dystonia (ICD) and spinocerebellar ataxias (SCAs). MATERIALS AND METHODS Eleven patients with ICD, 7 with genetically confirmed SCA2 or SCA3, and 10 healthy volunteers were enrolled. The onset latency and duration of the ipsilateral silent period (iSPOL and iSPD, respectively), as well as the so-called transcallosal conduction time (TCT), were then recorded from the abductor pollicis brevis of the right side using an 8-shaped focal coil with wing diameters of 70 mm; all these parameters were evaluated and compared among groups. In SCAs, changes in neurophysiological measures were also correlated to the mutational load. RESULTS iSPD was significantly shorter in patients with SCA2 and SCA3, when compared both to control and ICD (P < .0001); iSPOL and TCT were prolonged in SCAs patients (P < .001). Changes in iSPD, iSPOL, and TCT in SCAs are significantly correlated with the mutational load (P = .01, P = .02, and P = .002, respectively). DISCUSSION This is the first study to assess changes in interhemispheric communication in patients with SCAs and ICD, using a transcranial magnetic stimulation protocol. Together with previous data in Huntington's disease, we suggest that these changes may underlie, at least in part, a common disease mechanism of polyglutamine disorders.
Collapse
Affiliation(s)
- Tommaso Bocci
- "Aldo Ravelli" Center for Neurotechnology and Experiental Brain Therapeutics, Department of Health Sciences, University of Milan & ASST Santi Paolo e Carlo, Milan, Italy
| | - Davide Baloscio
- Section of Neurophysiopathology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberta Ferrucci
- "Aldo Ravelli" Center for Neurotechnology and Experiental Brain Therapeutics, Department of Health Sciences, University of Milan & ASST Santi Paolo e Carlo, Milan, Italy
| | - Lucia Briscese
- Severe Acquired Brain Injuries Unit, Cisanello University Hospital, Pisa, Italy
| | - Alberto Priori
- "Aldo Ravelli" Center for Neurotechnology and Experiental Brain Therapeutics, Department of Health Sciences, University of Milan & ASST Santi Paolo e Carlo, Milan, Italy
| | - Ferdinando Sartucci
- Section of Neurophysiopathology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
63
|
Exploring the brain metabolic correlates of process-specific CSF biomarkers in patients with MCI due to Alzheimer's disease: preliminary data. Neurobiol Aging 2022; 117:212-221. [DOI: 10.1016/j.neurobiolaging.2022.03.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 12/30/2022]
|
64
|
Lin K, Jie B, Dong P, Ding X, Bian W, Liu M. Convolutional Recurrent Neural Network for Dynamic Functional MRI Analysis and Brain Disease Identification. Front Neurosci 2022; 16:933660. [PMID: 35873806 PMCID: PMC9298744 DOI: 10.3389/fnins.2022.933660] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Dynamic functional connectivity (dFC) networks derived from resting-state functional magnetic resonance imaging (rs-fMRI) help us understand fundamental dynamic characteristics of human brains, thereby providing an efficient solution for automated identification of brain diseases, such as Alzheimer's disease (AD) and its prodromal stage. Existing studies have applied deep learning methods to dFC network analysis and achieved good performance compared with traditional machine learning methods. However, they seldom take advantage of sequential information conveyed in dFC networks that could be informative to improve the diagnosis performance. In this paper, we propose a convolutional recurrent neural network (CRNN) for automated brain disease classification with rs-fMRI data. Specifically, we first construct dFC networks from rs-fMRI data using a sliding window strategy. Then, we employ three convolutional layers and long short-term memory (LSTM) layer to extract high-level features of dFC networks and also preserve the sequential information of extracted features, followed by three fully connected layers for brain disease classification. Experimental results on 174 subjects with 563 rs-fMRI scans from the Alzheimer's Disease Neuroimaging Initiative (ADNI) demonstrate the effectiveness of our proposed method in binary and multi-category classification tasks.
Collapse
Affiliation(s)
- Kai Lin
- School of Computer and Information, Anhui Normal University, Wuhu, China
| | - Biao Jie
- School of Computer and Information, Anhui Normal University, Wuhu, China
| | - Peng Dong
- School of Computer and Information, Anhui Normal University, Wuhu, China
| | - Xintao Ding
- School of Computer and Information, Anhui Normal University, Wuhu, China
| | - Weixin Bian
- School of Computer and Information, Anhui Normal University, Wuhu, China
| | - Mingxia Liu
- Department of Radiology and Biomedical Research Imaging Center (BRIC), University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
65
|
Kannappan B, Gunasekaran TI, te Nijenhuis J, Gopal M, Velusami D, Kothandan G, Lee KH. Polygenic score for Alzheimer’s disease identifies differential atrophy in hippocampal subfield volumes. PLoS One 2022; 17:e0270795. [PMID: 35830443 PMCID: PMC9278752 DOI: 10.1371/journal.pone.0270795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/20/2022] [Indexed: 01/18/2023] Open
Abstract
Hippocampal subfield atrophy is a prime structural change in the brain, associated with cognitive aging and neurodegenerative diseases such as Alzheimer’s disease. Recent developments in genome-wide association studies (GWAS) have identified genetic loci that characterize the risk of hippocampal volume loss based on the processes of normal and abnormal aging. Polygenic risk scores are the genetic proxies mimicking the genetic role of the pre-existing vulnerabilities of the underlying mechanisms influencing these changes. Discriminating the genetic predispositions of hippocampal subfield atrophy between cognitive aging and neurodegenerative diseases will be helpful in understanding the disease etiology. In this study, we evaluated the polygenic risk of Alzheimer’s disease (AD PGRS) for hippocampal subfield atrophy in 1,086 individuals (319 cognitively normal (CN), 591 mild cognitively impaired (MCI), and 176 Alzheimer’s disease dementia (ADD)). Our results showed a stronger association of AD PGRS effect on the left hemisphere than on the right hemisphere for all the hippocampal subfield volumes in a mixed clinical population (CN+MCI+ADD). The subfields CA1, CA4, hippocampal tail, subiculum, presubiculum, molecular layer, GC-ML-DG, and HATA showed stronger AD PGRS associations with the MCI+ADD group than with the CN group. The subfields CA3, parasubiculum, and fimbria showed moderately higher AD PGRS associations with the MCI+ADD group than with the CN group. Our findings suggest that the eight subfield regions, which were strongly associated with AD PGRS are likely involved in the early stage ADD and a specific focus on the left hemisphere could enhance the early prediction of ADD.
Collapse
Affiliation(s)
- Balaji Kannappan
- Gwangju Alzheimer’s & Related Dementia Cohort Research Center, Chosun University, Gwangju, Republic of Korea
- Department of Biomedical Science, Chosun University, Gwangju, Republic of Korea
| | - Tamil Iniyan Gunasekaran
- Gwangju Alzheimer’s & Related Dementia Cohort Research Center, Chosun University, Gwangju, Republic of Korea
- Department of Biomedical Science, Chosun University, Gwangju, Republic of Korea
| | - Jan te Nijenhuis
- Gwangju Alzheimer’s & Related Dementia Cohort Research Center, Chosun University, Gwangju, Republic of Korea
- Department of Biomedical Science, Chosun University, Gwangju, Republic of Korea
- * E-mail: (JN); (KHL)
| | - Muthu Gopal
- Health Systems Research & MRHRU, ICMR-National Institute of Epidemiology, Tirunelveli, Tamil Nadu, India
| | - Deepika Velusami
- Department of Physiology, Sri Manakula Vinayagar Medical College and Hospital, Puducherry, Tamil Nadu, India
| | - Gugan Kothandan
- Biopolymer Modeling and Protein Chemistry Laboratory, Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Chennai, Tamil Nadu, India
| | - Kun Ho Lee
- Gwangju Alzheimer’s & Related Dementia Cohort Research Center, Chosun University, Gwangju, Republic of Korea
- Department of Biomedical Science, Chosun University, Gwangju, Republic of Korea
- Korea Brain Research Institute, Daegu, Republic of Korea
- * E-mail: (JN); (KHL)
| | | |
Collapse
|
66
|
Zhu LY, Shi L, Luo Y, Leung J, Kwok T. Brain MRI Biomarkers to Predict Cognitive Decline in Older People with Alzheimer's Disease. J Alzheimers Dis 2022; 88:763-769. [PMID: 35723095 DOI: 10.3233/jad-215189] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Structural magnetic resonance imaging markers predicting symptomatic progression at the individual level can be highly beneficial for early intervention and treatment planning for Alzheimer's disease (AD). However, the correlation between baseline MRI findings and AD progression has not been fully established. OBJECTIVE To explore the correlation between baseline MRI findings and AD progression. METHODS Brain volumetric measures were applied to differentiate the patients at risk of fast deterioration in AD. We included 194 AD patients with a 24-month follow-up: 65 slow decliners, 63 normal decliners, and 66 fast decliners categorized by changes in Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). ANOVA analyses were used to identify baseline brain atrophy between groups. Logistic regressions were further performed to explore the relative merits of AD resemblance structural atrophy index (AD-RAI) and individual regional volumetric measures in prediction of disease progression. RESULTS Atrophy in the temporal and insular lobes was associated with fast cognitive decline over 24 months. Smaller volumes of temporal and insular lobes in the left but not the right brain were associated with fast cognitive decline. Baseline AD-RAI predicted fast versus slow progression of cognitive decline (odds ratio 3.025 (95% CI: 1.064-8.600), high versus low, AUC 0.771). Moreover, AD-RAI was significantly lower among slow decliners when compared with normal decliners (p = 0.039). CONCLUSION AD-RAI on MRI showed potential in identifying clinical AD patients at risk of accelerated cognitive decline.
Collapse
Affiliation(s)
- Liu-Ying Zhu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.,Zhongshan City People's Hospital, Zhongshan, Guangdong Province, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen, Guangdong Province, China.,Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, China
| | - Yishan Luo
- BrainNow Research Institute, Shenzhen, Guangdong Province, China
| | - Jason Leung
- Jockey Club Centre for Osteoporosis Care and Control, The Chinese University of Hong Kong, Hong Kong, Shatin, China
| | - Timothy Kwok
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| |
Collapse
|
67
|
Hazarika RA, Maji AK, Sur SN, Olariu I, Kandar D. A fuzzy membership based comparison of the grey matter (GM) in cognitively normal (CN), mild cognitive impairment (MCI), and Alzheimer’s disease (AD) using brain images. JOURNAL OF INTELLIGENT & FUZZY SYSTEMS 2022. [DOI: 10.3233/jifs-219279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Grey matter (GM) in human brain contains most of the important cells covering the regions involved in neurophysiological operations such as memory, emotions, decision making, etc. Alzheimer’s disease (AD) is a neurological disease that kills the brain cells in regions which are mostly involved in the neurophysiological operations. Mild Cognitive Impairment (MCI) is a stage between Cognitively Normal (CN) and AD, where a significant cognitive declination can be observed. The destruction of brain cells causes a reduction in the size of GM. Evaluation of changes in GM, may help in studying the overall brain transformations and accurate classification of different stages of AD. In this work, firstly skull of brain images is stripped for 5 different slices, then segmentation of GM is performed. Finally, the average number of pixels in grey region and the average atrophy in grey pixels per year is calculated and compared amongst CN, MCI, and AD patients of various ages and genders. It is observed that, for some subjects (in some particular ages) from different dementia stages, pattern of GM changes is almost identical. To solve this issue, we have used the concept of fuzzy membership functions to classify the dementia stages more accurately. It is observed from the comparison that average difference in the number of pixels between CN and MCI= 10.01%, CN and AD= 19.63%, MCI and AD= 10.72%. It can be also observed from the comparison that, the average atrophy in grey matter per year in CN= 1.92%, MCI= 3.13%, and AD= 4.33%.
Collapse
Affiliation(s)
- Ruhul Amin Hazarika
- Department of Information Technology, North Eastern Hill University Shillong, Meghalaya, India
| | - Arnab Kumar Maji
- Department of Information Technology, North Eastern Hill University Shillong, Meghalaya, India
| | - Samarendra Nath Sur
- Department of Electronics and Communication Engineering, Sikkim Manipal Institute of Technology, Sikkim Manipal University, Majitar, Rangpo, East Sikkim, India
| | - Iustin Olariu
- Faculty of Medicine, Vasile Goldis Western University of Arad, Arad, Romania
| | - Debdatta Kandar
- Department of Information Technology, North Eastern Hill University Shillong, Meghalaya, India
| |
Collapse
|
68
|
Li W, Qu G, Huo C, Hu X, Xu G, Li H, Zhang J, Li Z. Identifying Cognitive Impairment in Elderly Using Coupling Functions Between Cerebral Oxyhemoglobin and Arterial Blood Pressure. Front Aging Neurosci 2022; 14:904108. [PMID: 35669465 PMCID: PMC9163710 DOI: 10.3389/fnagi.2022.904108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background This study aimed to assess brain oxygenation status and cerebral autoregulation function in subjects with cognitive dysfunction. Methods The Montreal Cognitive Assessment (MoCA) was applied to divide the subjects into three groups: cognitive impairment (Group CI, 72.50 ± 10.93 y), mild cognitive impairment (Group MCI, 72.02 ± 9.90 y), and normal cognition (Group NC, 70.72 ± 7.66 y). Near-infrared spectroscopy technology and a non-invasive blood pressure device were used to simultaneously measure changes in cerebral tissue oxygenation signals in the bilateral prefrontal lobes (LPFC/RPFC) and arterial blood pressure (ABP) signals from subjects in the resting state (15 min). The coupling between ABP and cerebral oxyhemoglobin concentrations (Δ [O2Hb]) was calculated in very-low-frequency (VLF, 0.02-0.07 Hz) and low-frequency (LF, 0.07-0.2 Hz) bands based on the dynamical Bayesian inference approach. Pearson correlation analyses were used to study the relationships between MoCA scores, tissue oxygenation index, and strength of coupling function. Results In the interval VLF, Group CI (p = 0.001) and Group MCI (p = 0.013) exhibited significantly higher coupling strength from ABP to Δ [O2Hb] in the LPFC than Group NC. In the interval LF, coupling strength from ABP to Δ [O2Hb] in the LPFC was significantly higher in Group CI than in Group NC (p = 0.001). Pearson correlation results showed that MoCA scores had a significant positive correlation with the tissue oxygenation index and a significant negative correlation with the coupling strength from ABP to Δ [O2Hb]. Conclusion The significantly increased coupling strength may be evidence of impaired cerebral autoregulation function in subjects with cognitive dysfunction. The Pearson correlation results suggest that indicators of brain oxygenation status and cerebral autoregulation function can reflect cognitive function. This study provides insights into the mechanisms underlying the pathophysiology of cognitive impairment and provides objective indicators for screening cognitive impairment in the elderly population.
Collapse
Affiliation(s)
- Wenhao Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Guanwen Qu
- Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing, China
- Key Laboratory of Neuro-Functional Information and Rehabilitation Engineering of the Ministry of Civil Affairs, Beijing, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Congcong Huo
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Xiaoling Hu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Gongcheng Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Huiyuan Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Jingsha Zhang
- Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing, China
- Key Laboratory of Neuro-Functional Information and Rehabilitation Engineering of the Ministry of Civil Affairs, Beijing, China
| | - Zengyong Li
- Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing, China
- Key Laboratory of Neuro-Functional Information and Rehabilitation Engineering of the Ministry of Civil Affairs, Beijing, China
| |
Collapse
|
69
|
Zhang X, Guan Q, Li Y, Zhang J, Zhu W, Luo Y, Zhang H. Aberrant Cross-Tissue Functional Connectivity in Alzheimer’s Disease: Static, Dynamic, and Directional Properties. J Alzheimers Dis 2022; 88:273-290. [DOI: 10.3233/jad-215649] [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: BOLD signals in the gray matter (GM) and white matter (WM) are tightly coupled. However, our understanding of the cross-tissue functional network in Alzheimer’s disease (AD) is limited. Objective: We investigated the changes of cross-tissue functional connectivity (FC) metrics for the GM regions susceptible to AD damage. Methods: For each GM region in the default mode (DMN) and limbic networks, we obtained its low-order static FC with any WM region, and the high-order static FC between any two WM regions based on their FC pattern similarity with multiple GM regions. The dynamic and directional properties of cross-tissue FC were then acquired, specifically for the regional pairs whose low- or high-order static FCs showed significant differences between AD and normal control (NC). Moreover, these cross-tissue FC metrics were correlated with voxel-based GM volumes and MMSE in all participants. Results: Compared to NC, AD patients showed decreased low-order static FCs between the intra-hemispheric GM-WM pairs (right ITG-right fornix; left MoFG-left posterior corona radiata), and increased low-order static, dynamic, and directional FCs between the inter-hemispheric GM-WM pairs (right MTG-left superior/posterior corona radiata). The high-order static and directional FCs between the left cingulate bundle-left tapetum were increased in AD, based on their FCs with the GMs of DMN. Those decreased and increased cross-tissue FC metrics in AD had opposite correlations with memory-related GM volumes and MMSE (positive for the decreased and negative for the increased). Conclusion: Cross-tissue FC metrics showed opposite changes in AD, possibly as useful neuroimaging biomarkers to reflect neurodegenerative and compensatory mechanisms.
Collapse
Affiliation(s)
- Xingxing Zhang
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen, China
| | - Qing Guan
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China
- Center for Neuroimaging, Shenzhen Institute of Neuroscience, Shenzhen, China
| | - Yingjia Li
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen, China
| | - Jianfeng Zhang
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen, China
| | - Wanlin Zhu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuejia Luo
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen, China
| | - Haobo Zhang
- Center for Brain Disorders and Cognitive Sciences, School of Psychology, Shenzhen University, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China
- Center for Neuroimaging, Shenzhen Institute of Neuroscience, Shenzhen, China
| | | |
Collapse
|
70
|
Carolus K, Fuchs TA, Bergsland N, Ramasamy D, Tran H, Uher T, Horakova D, Vaneckova M, Havrdova E, Benedict RHB, Zivadinov R, Dwyer MG. Time course of lesion-induced atrophy in multiple sclerosis. J Neurol 2022; 269:4478-4487. [PMID: 35394170 DOI: 10.1007/s00415-022-11094-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND PURPOSE White matter (WM) tract disruption impacts volume loss in connected deep gray matter (DGM) over 5 years in people with multiple sclerosis (PwMS). However, the timeline of this phenomenon remains poorly characterized. MATERIALS AND METHODS Annual serial MRI for 181 PwMS was retrospectively analyzed from a 10-year clinical trial database. Annualized thalamic atrophy, DGM atrophy, and disruption of connected WM tracts were measured. For time series analysis, ~700 epochs were collated using a sliding 5-year window, and regression models predicting 1-year atrophy were applied to characterize the influence of new tract disruption from preceding years, while controlling for whole brain atrophy and other relevant factors. RESULTS Disruptions of WM tracts connected to the thalamus were significantly associated with thalamic atrophy 1 year later (β: 0.048-0.103). This effect was not observed for thalamic tract disruption concurrent with the time of atrophy nor for thalamic tract disruption preceding the atrophy by 2-4 years. Similarly, disruptions of white matter tracts connected to the DGM were significantly associated with DGM atrophy 1 year later (β: 0.078-0.111), but not for tract disruption concurrent with, nor preceding the atrophy by 2-4 years. CONCLUSION Increased rates of thalamic and DGM atrophy were restricted to 1 year following newly developed disruption in connected WM tracts. In research and clinical settings, additional gray matter atrophy may be expected 1 year following new lesion growth in connected white matter.
Collapse
Affiliation(s)
- Keith Carolus
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Tom A Fuchs
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Jacobs Multiple Sclerosis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- IRCCS, Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Deepa Ramasamy
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Hoan Tran
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Tomas Uher
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University, General University Hospital, Prague, Czech Republic
| | - Eva Havrdova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ralph H B Benedict
- Jacobs Multiple Sclerosis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, 100 High Street, Buffalo, NY, 14203, USA
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, 100 High Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
71
|
Molecular Signatures of Mitochondrial Complexes Involved in Alzheimer’s Disease via Oxidative Phosphorylation and Retrograde Endocannabinoid Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9565545. [PMID: 35432724 PMCID: PMC9006080 DOI: 10.1155/2022/9565545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/22/2022] [Indexed: 11/17/2022]
Abstract
Objective The inability to intervene in Alzheimer's disease (AD) forces the search for promising gene-targeted therapies. This study was aimed at exploring molecular signatures and mechanistic pathways to improve the diagnosis and treatment of AD. Methods Microarray datasets were collected to filter differentially expressed genes (DEGs) between AD and nondementia controls. Weight gene correlation network analysis (WGCNA) was employed to analyze the correlation of coexpression modules with AD phenotype. A global regulatory network was established and then visualized using Cytoscape software to determine hub genes and their mechanistic pathways. Receiver operating characteristic (ROC) analysis was conducted to estimate the diagnostic performance of hub genes in AD prediction. Results A total of 2,163 DEGs from 13,049 background genes were screened in AD relative to nondementia controls. Among the six coexpression modules constructed by WGCNA, DEGs of the key modules with the strongest correlation with AD were extracted to build a global regulatory network. According to the Maximal Clique Centrality (MCC) method, five hub genes associated with mitochondrial complexes were chosen. Further pathway enrichment analysis of hub genes, such as oxidative phosphorylation and retrograde endocannabinoid signaling, was identified. According to the area under the curve (AUC) of about 70%, each hub gene exhibited a good diagnostic performance in predicting AD. Conclusions Our findings highlight the perturbation of mitochondrial complexes underlying AD onset, which is mediated by molecular signatures involved in oxidative phosphorylation (COX5A, NDUFAB1, SDHB, UQCRC2, and UQCRFS1) and retrograde endocannabinoid signaling (NDUFAB1) pathways.
Collapse
|
72
|
Balboni E, Nocetti L, Carbone C, Dinsdale N, Genovese M, Guidi G, Malagoli M, Chiari A, Namburete AIL, Jenkinson M, Zamboni G. The impact of transfer learning on 3D deep learning convolutional neural network segmentation of the hippocampus in mild cognitive impairment and Alzheimer disease subjects. Hum Brain Mapp 2022; 43:3427-3438. [PMID: 35373881 PMCID: PMC9248306 DOI: 10.1002/hbm.25858] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/04/2022] [Accepted: 03/21/2022] [Indexed: 11/23/2022] Open
Abstract
Research on segmentation of the hippocampus in magnetic resonance images through deep learning convolutional neural networks (CNNs) shows promising results, suggesting that these methods can identify small structural abnormalities of the hippocampus, which are among the earliest and most frequent brain changes associated with Alzheimer disease (AD). However, CNNs typically achieve the highest accuracy on datasets acquired from the same domain as the training dataset. Transfer learning allows domain adaptation through further training on a limited dataset. In this study, we applied transfer learning on a network called spatial warping network segmentation (SWANS), developed and trained in a previous study. We used MR images of patients with clinical diagnoses of mild cognitive impairment (MCI) and AD, segmented by two different raters. By using transfer learning techniques, we developed four new models, using different training methods. Testing was performed using 26% of the original dataset, which was excluded from training as a hold‐out test set. In addition, 10% of the overall training dataset was used as a hold‐out validation set. Results showed that all the new models achieved better hippocampal segmentation quality than the baseline SWANS model (ps < .001), with high similarity to the manual segmentations (mean dice [best model] = 0.878 ± 0.003). The best model was chosen based on visual assessment and volume percentage error (VPE). The increased precision in estimating hippocampal volumes allows the detection of small hippocampal abnormalities already present in the MCI phase (SD = [3.9 ± 0.6]%), which may be crucial for early diagnosis.
Collapse
Affiliation(s)
- Erica Balboni
- Health Physics Unit, Azienda Ospedaliera di Modena, Modena, Italy.,Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Nocetti
- Health Physics Unit, Azienda Ospedaliera di Modena, Modena, Italy
| | - Chiara Carbone
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Neurosciences and Neurotechnology, Università di Modena e Reggio Emilia, Modena, Italy
| | - Nicola Dinsdale
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Oxford Machine Learning in NeuroImaging Lab, Department of Computer Science, Oxford, UK
| | | | - Gabriele Guidi
- Health Physics Unit, Azienda Ospedaliera di Modena, Modena, Italy
| | | | - Annalisa Chiari
- Neuroradiology Unit, Azienda Ospedaliera di Modena, Modena, Italy
| | - Ana I L Namburete
- Oxford Machine Learning in NeuroImaging Lab, Department of Computer Science, Oxford, UK
| | - Mark Jenkinson
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,Australian Institute for Machine Learning, School of Computer Science, University of Adelaide, Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Giovanna Zamboni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Neurosciences and Neurotechnology, Università di Modena e Reggio Emilia, Modena, Italy.,Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
73
|
Tripathi SM, Murray AD, Wischik CM, Schelter B. Crossed cerebellar diaschisis in Alzheimer's disease. Nucl Med Commun 2022; 43:423-427. [PMID: 35081090 DOI: 10.1097/mnm.0000000000001531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Crossed cerebellar diaschisis (CCD) is characterized by hypometabolism and hypoperfusion on molecular imaging in the cerebellum due to a supratentorial lesion on the contralateral side. CCD is a well-established phenomenon in acute or subacute conditions such as infarction but it has been less well described in chronic conditions such as neurodegenerative dementias. Here, we investigate CCD in a large sample of 830 people meeting research criteria for Alzheimer's disease (AD) using [18F]fluorodeoxyglucose-positron emission tomography (FDG-PET). MATERIALS AND METHODS This study is based on FDG-PET data collected at baseline as part of two large-scale Phase III clinical trials of a novel tau aggregation inhibitor medication, methylthioninium in mild to moderate AD participants. Quantification of FDG-PET hypometabolism was carried out using standardized uptake value ratio (SUVR), with the pons as the comparison region. SUVR was compared in different regions of interest between the right and left hemispheres of the brain and cerebellum in people with mild AD (Mini-Mental State Examination score ≥ 20). RESULTS Comparison of SUVR in different brain regions demonstrated significant differences in the temporal, occipital and cerebellar cortices. Right and left asymmetry was noted with lower SUVR in the left temporal and occipital regions, whereas SUVR was lower in the right side of the cerebellum. CONCLUSION Here, we found robust evidence of CCD in a large sample of people with AD, a chronic neurodegenerative condition. The presence of this phenomenon in AD opens up a new avenue of research in AD pathogenesis and has the potential to change future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Shailendra Mohan Tripathi
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
- King George's Medical University, Lucknow, India
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
- King George's Medical University, Lucknow, India
| | - Claude M Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen
- TauRx Therapeutics Ltd, Aberdeen
| | - Bjoern Schelter
- TauRx Therapeutics Ltd, Aberdeen
- Institute for Complex Systems and Mathematical Biology (ICSMB), Meston Building, Meston Walk, King's College, Old Aberdeen University of Aberdeen, Aberdeen, UK
| |
Collapse
|
74
|
Speranza L, Pulcrano S, Perrone-Capano C, di Porzio U, Volpicelli F. Music affects functional brain connectivity and is effective in the treatment of neurological disorders. Rev Neurosci 2022; 33:789-801. [PMID: 35325516 DOI: 10.1515/revneuro-2021-0135] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 02/25/2022] [Indexed: 11/15/2022]
Abstract
In a million years, under the pressure of natural selection, hominins have acquired the abilities for vocal learning, music, and language. Music is a relevant human activity, highly effective in enhancing sociality, is a universal experience common to all known human cultures, although it varies in rhythmic and melodic complexity. It has been part of human life since the beginning of our history, or almost, and it strengthens the mother-baby relation even within the mother's womb. Music engages multiple cognitive functions, and promotes attention, concentration, imagination, creativity, elicits memories and emotions, and stimulates imagination, and harmony of movement. It changes the chemistry of the brain, by inducing the release of neurotransmitters and hormones (dopamine, serotonin, and oxytocin) and activates the reward and prosocial systems. In addition, music is also used to develop new therapies necessary to alleviate severe illness, especially neurological disorders, and brain injuries.
Collapse
Affiliation(s)
- Luisa Speranza
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Salvatore Pulcrano
- Institute of Genetics and Biophysics, "Adriano Buzzati-Traverso", C.N.R., 80131 Naples, Italy.,Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Carla Perrone-Capano
- Institute of Genetics and Biophysics, "Adriano Buzzati-Traverso", C.N.R., 80131 Naples, Italy.,Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Umberto di Porzio
- Institute of Genetics and Biophysics, "Adriano Buzzati-Traverso", C.N.R., 80131 Naples, Italy
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
75
|
Qiu A, Xu L, Liu C. Predicting diagnosis 4 years prior to Alzheimer's disease incident. Neuroimage Clin 2022; 34:102993. [PMID: 35344803 PMCID: PMC8958535 DOI: 10.1016/j.nicl.2022.102993] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022]
Abstract
This study employed a deep learning longitudinal model, graph convolutional and recurrent neural network (graph-CNN-RNN), on a series of brain structural MRI scans for AD prognosis. It characterized whole-brain morphology via incorporating longitudinal cortical and subcortical morphology and defined a probabilistic risk for the prediction of AD as a function of age prior to clinical diagnosis. The graph-CNN-RNN model was trained on half of the Alzheimer's Disease Neuroimaging Initiative dataset (ADNI, n = 1559) and validated on the other half of the ADNI dataset and the Open Access Series of Imaging Studies-3 (OASIS-3, n = 930). Our findings demonstrated that the graph-CNN-RNN can reliably and robustly diagnose AD at the accuracy rate of 85% and above across all the time points for both datasets. The graph-CNN-RNN predicted the AD conversion from 0 to 4 years before the AD onset at ∼80% of accuracy. The AD probabilistic risk was associated with clinical traits, cognition, and amyloid burden assessed using [18F]-Florbetapir (AV45) positron emission tomography (PET) across all the time points. The graph-CNN-RNN provided the quantitative trajectory of brain morphology from prognosis to overt stages of AD. Such a deep learning tool and the AD probabilistic risk have great potential in clinical applications for AD prognosis.
Collapse
Affiliation(s)
- Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore; Institute of Data Science, National University of Singapore, Singapore; NUS (Suzhou) Research Institute, Suzhou, China; School of Computer Engineering and Science, Shanghai University, China; Department of Biomedical Engineering, the Johns Hopkins University, USA.
| | - Liyuan Xu
- School of Computer Engineering and Science, Shanghai University, China
| | - Chaoqiang Liu
- Department of Biomedical Engineering, National University of Singapore, Singapore
| |
Collapse
|
76
|
Fujita S, Cencini M, Buonincontri G, Takei N, Schulte RF, Fukunaga I, Uchida W, Hagiwara A, Kamagata K, Hagiwara Y, Matsuyama Y, Abe O, Tosetti M, Aoki S. Simultaneous relaxometry and morphometry of human brain structures with 3D magnetic resonance fingerprinting: a multicenter, multiplatform, multifield-strength study. Cereb Cortex 2022; 33:729-739. [PMID: 35271703 PMCID: PMC9890456 DOI: 10.1093/cercor/bhac096] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 02/04/2023] Open
Abstract
Relaxation times and morphological information are fundamental magnetic resonance imaging-derived metrics of the human brain that reflect the status of the underlying tissue. Magnetic resonance fingerprinting (MRF) enables simultaneous acquisition of T1 and T2 maps inherently aligned to the anatomy, allowing whole-brain relaxometry and morphometry in a single scan. In this study, we revealed the feasibility of 3D MRF for simultaneous brain structure-wise morphometry and relaxometry. Comprehensive test-retest scan analyses using five 1.5-T and three 3.0-T systems from a single vendor including different scanner types across 3 institutions demonstrated that 3D MRF-derived morphological information and relaxation times are highly repeatable at both 1.5 T and 3.0 T. Regional cortical thickness and subcortical volume values showed high agreement and low bias across different field strengths. The ability to acquire a set of regional T1, T2, thickness, and volume measurements of neuroanatomical structures with high repeatability and reproducibility facilitates the ability of longitudinal multicenter imaging studies to quantitatively monitor changes associated with underlying pathologies, disease progression, and treatments.
Collapse
Affiliation(s)
- Shohei Fujita
- Corresponding author: Department of Radiology, Juntendo University School of Medicine, 12-1 Hongo, Bunkyo, Tokyo 113-8421, Japan.
| | - Matteo Cencini
- Imago7 Foundation, Pisa, Italy,IRCCS Stella Maris, Pisa, Italy
| | | | | | | | - Issei Fukunaga
- Department of Radiology, Juntendo University, Tokyo, Japan
| | - Wataru Uchida
- Department of Radiology, Juntendo University, Tokyo, Japan
| | | | - Koji Kamagata
- Department of Radiology, Juntendo University, Tokyo, Japan
| | - Yasuhiro Hagiwara
- Department of Biostatistics, School of Public Health, The University of Tokyo, Tokyo, Japan
| | - Yutaka Matsuyama
- Department of Biostatistics, School of Public Health, The University of Tokyo, Tokyo, Japan
| | - Osamu Abe
- Department of Radiology, The University of Tokyo, Tokyo, Japan
| | - Michela Tosetti
- Imago7 Foundation, Pisa, Italy,IRCCS Stella Maris, Pisa, Italy
| | - Shigeki Aoki
- Department of Radiology, Juntendo University, Tokyo, Japan
| |
Collapse
|
77
|
Loessner L, Matthes C, Haussmann R, Brandt MD, Sauer C, Espin M, Noppes F, Werner A, Linn J, Hummel T, Haehner A, Donix M. Predictors of subjective cognitive deficits in patients with mild cognitive impairment. Psychogeriatrics 2022; 22:210-217. [PMID: 34939254 DOI: 10.1111/psyg.12802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/12/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Detailed examination of cognitive deficits in patients with mild cognitive impairment (MCI) yields substantial diagnostic and prognostic value, specifically with respect to memory. Magnitude and characteristics of subjective cognitive deficits, however, often receive less attention in this population at risk for developing dementia. METHODS We investigated predictors of subjective cognitive deficits in patients with MCI, using a detailed assessment for such impairments associated with different cognitive domains, as well as demographic and clinical variables including magnetic resonance imaging data. RESULTS The strongest predictor for subjective memory deficits was depressed mood, whereas subjective performance issues associated with attention or executive functions also corresponded to measurable impairments in the respective cognitive domains. Reduced hippocampal thickness and hemispheric entorhinal cortex thickness asymmetry were associated with objective memory impairment but not with subjective deficits or symptoms of depression. CONCLUSIONS Whereas low objective memory performance and reduced cortical thickness within medial temporal lobe subregions could be associated with neurodegeneration, greater subjective memory deficits in patients with MCI may indicate psychological burden.
Collapse
Affiliation(s)
- Lorenz Loessner
- Department of Psychiatry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Claudia Matthes
- Department of Psychiatry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Robert Haussmann
- Department of Psychiatry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Moritz D Brandt
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Cathrin Sauer
- Department of Psychiatry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Melanie Espin
- Department of Otorhinolaryngology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Felix Noppes
- Department of Psychiatry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Annett Werner
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.,Department of Neuroradiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jennifer Linn
- Department of Neuroradiology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Thomas Hummel
- Department of Otorhinolaryngology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Antje Haehner
- Department of Otorhinolaryngology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Markus Donix
- Department of Psychiatry, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| |
Collapse
|
78
|
Canal-Garcia A, Gómez-Ruiz E, Mijalkov M, Chang YW, Volpe G, Pereira JB. Multiplex Connectome Changes across the Alzheimer’s Disease Spectrum Using Gray Matter and Amyloid Data. Cereb Cortex 2022; 32:3501-3515. [PMID: 35059722 PMCID: PMC9376877 DOI: 10.1093/cercor/bhab429] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022] Open
Abstract
The organization of the Alzheimer’s disease (AD) connectome has been studied using graph theory using single neuroimaging modalities such as positron emission tomography (PET) or structural magnetic resonance imaging (MRI). Although these modalities measure distinct pathological processes that occur in different stages in AD, there is evidence that they are not independent from each other. Therefore, to capture their interaction, in this study we integrated amyloid PET and gray matter MRI data into a multiplex connectome and assessed the changes across different AD stages. We included 135 cognitively normal (CN) individuals without amyloid-β pathology (Aβ−) in addition to 67 CN, 179 patients with mild cognitive impairment (MCI) and 132 patients with AD dementia who all had Aβ pathology (Aβ+) from the Alzheimer’s Disease Neuroimaging Initiative. We found widespread changes in the overlapping connectivity strength and the overlapping connections across Aβ-positive groups. Moreover, there was a reorganization of the multiplex communities in MCI Aβ + patients and changes in multiplex brain hubs in both MCI Aβ + and AD Aβ + groups. These findings offer a new insight into the interplay between amyloid-β pathology and brain atrophy over the course of AD that moves beyond traditional graph theory analyses based on single brain networks.
Collapse
Affiliation(s)
- Anna Canal-Garcia
- Address correspondence to Department of NVS, Division of Clinical Geriatrics, NEO seventh floor, Blickagången 16, 141 52 Huddinge, Sweden. ; Data used in preparation of this article were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database (adni.loni.usc.edu). As such, the investigators within the ADNI contributed to the design and implementation of ADNI and/or provided data but did not participate in analysis or writing of this report. A complete listing of ADNI investigators can be found at: http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf
| | | | - Mite Mijalkov
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Yu-Wei Chang
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Giovanni Volpe
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| | - Joana B Pereira
- Address correspondence to Department of NVS, Division of Clinical Geriatrics, NEO seventh floor, Blickagången 16, 141 52 Huddinge, Sweden. ; Data used in preparation of this article were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database (adni.loni.usc.edu). As such, the investigators within the ADNI contributed to the design and implementation of ADNI and/or provided data but did not participate in analysis or writing of this report. A complete listing of ADNI investigators can be found at: http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf
| | | |
Collapse
|
79
|
Xiao Z, Ren X, Zhao Q, Wu W, Liang X, Tang J, Zhang M, Xue Y, Luo J, Ding D, Fu J. Relation of middle cerebral artery flow velocity and risk of cognitive decline: A prospective community-based study. J Clin Neurosci 2022; 97:56-61. [PMID: 35033782 DOI: 10.1016/j.jocn.2021.12.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/15/2021] [Accepted: 12/23/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Hemodynamic parameters measured by the Transcranial Doppler Ultrasound (TCD) are related to cognitive impairment in many cross-sectional studies, but the longitudinal evidence is scarce. In this study, we aim to verify the association between flow velocity of Middle Cerebral Artery (MCA) and the longitudinal cognitive decline in community dwelling older adults. MATERIALS AND METHODS Participants were administered TCD examination at the baseline. The Peak Systolic Velocity (PSV), Mean Flow Velocity (MFV), and Pulsatility Index (PI) of MCA segments on left middle (LmMCA), left proximal (LpMCA), right middle (RmMCA), and right proximal (RpMCA) were obtained. Mini-mental state examination (MMSE) were conducted at both baseline and follow-up. RESULTS One hundred and thirteen participants without dementia were followed up for 6.3 years in average. The mean annual rate of decline in the MMSE score was 0.15 (min to max: -1.0 to 1.2). LpMCA PSV (β = -0.0034, r = -0.231, P = 0.022) and LpMCA MFV (β = -0.0049, r = -0.217, P = 0.031) were inversely associated with annual rate of decline in the MMSE score after adjusting for age, gender, education year, APOE ε4, obesity, hypertension, diabetes mellitus, stroke, and coronary heart disease. CONCLUSIONS Blood flow velocity of left proximal MCA was inversely related to global cognitive decline. Cerebral blood flow velocity may impact the cognitive function.
Collapse
Affiliation(s)
- Zhenxu Xiao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xue Ren
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qianhua Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanqing Wu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoniu Liang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Tang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Miaoyi Zhang
- Department of Neurology, North Huashan Hospital, Fudan University, No.108 Lu Xiang Road, Shanghai, China
| | - Yang Xue
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianfeng Luo
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China; Key Lab of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Ding Ding
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| | - Jianhui Fu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
80
|
OUP accepted manuscript. Arch Clin Neuropsychol 2022; 37:891-903. [DOI: 10.1093/arclin/acac012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2022] [Indexed: 11/12/2022] Open
|
81
|
Tank R, Ward J, Flegal KE, Smith DJ, Bailey MES, Cavanagh J, Lyall DM. Association between polygenic risk for Alzheimer's disease, brain structure and cognitive abilities in UK Biobank. Neuropsychopharmacology 2022; 47:564-569. [PMID: 34621014 PMCID: PMC8674313 DOI: 10.1038/s41386-021-01190-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/05/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023]
Abstract
Previous studies testing associations between polygenic risk for late-onset Alzheimer's disease (LOAD-PGR) and brain magnetic resonance imaging (MRI) measures have been limited by small samples and inconsistent consideration of potential confounders. This study investigates whether higher LOAD-PGR is associated with differences in structural brain imaging and cognitive values in a relatively large sample of non-demented, generally healthy adults (UK Biobank). Summary statistics were used to create PGR scores for n = 32,790 participants using LDpred. Outcomes included 12 structural MRI volumes and 6 concurrent cognitive measures. Models were adjusted for age, sex, body mass index, genotyping chip, 8 genetic principal components, lifetime smoking, apolipoprotein (APOE) e4 genotype and socioeconomic deprivation. We tested for statistical interactions between APOE e4 allele dose and LOAD-PGR vs. all outcomes. In fully adjusted models, LOAD-PGR was associated with worse fluid intelligence (standardised beta [β] = -0.080 per LOAD-PGR standard deviation, p = 0.002), matrix completion (β = -0.102, p = 0.003), smaller left hippocampal total (β = -0.118, p = 0.002) and body (β = -0.069, p = 0.002) volumes, but not other hippocampal subdivisions. There were no significant APOE x LOAD-PGR score interactions for any outcomes in fully adjusted models. This is the largest study to date investigating LOAD-PGR and non-demented structural brain MRI and cognition phenotypes. LOAD-PGR was associated with smaller hippocampal volumes and aspects of cognitive ability in healthy adults and could supplement APOE status in risk stratification of cognitive impairment/LOAD.
Collapse
Affiliation(s)
- Rachana Tank
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Kristin E Flegal
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - Daniel J Smith
- Centre for Clinical Brain Sciences, Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jonathan Cavanagh
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| |
Collapse
|
82
|
Gutman BA, van Erp TG, Alpert K, Ching CRK, Isaev D, Ragothaman A, Jahanshad N, Saremi A, Zavaliangos‐Petropulu A, Glahn DC, Shen L, Cong S, Alnæs D, Andreassen OA, Doan NT, Westlye LT, Kochunov P, Satterthwaite TD, Wolf DH, Huang AJ, Kessler C, Weideman A, Nguyen D, Mueller BA, Faziola L, Potkin SG, Preda A, Mathalon DH, Bustillo J, Calhoun V, Ford JM, Walton E, Ehrlich S, Ducci G, Banaj N, Piras F, Piras F, Spalletta G, Canales‐Rodríguez EJ, Fuentes‐Claramonte P, Pomarol‐Clotet E, Radua J, Salvador R, Sarró S, Dickie EW, Voineskos A, Tordesillas‐Gutiérrez D, Crespo‐Facorro B, Setién‐Suero E, van Son JM, Borgwardt S, Schönborn‐Harrisberger F, Morris D, Donohoe G, Holleran L, Cannon D, McDonald C, Corvin A, Gill M, Filho GB, Rosa PGP, Serpa MH, Zanetti MV, Lebedeva I, Kaleda V, Tomyshev A, Crow T, James A, Cervenka S, Sellgren CM, Fatouros‐Bergman H, Agartz I, Howells F, Stein DJ, Temmingh H, Uhlmann A, de Zubicaray GI, McMahon KL, Wright M, Cobia D, Csernansky JG, Thompson PM, Turner JA, Wang L. A meta-analysis of deep brain structural shape and asymmetry abnormalities in 2,833 individuals with schizophrenia compared with 3,929 healthy volunteers via the ENIGMA Consortium. Hum Brain Mapp 2022; 43:352-372. [PMID: 34498337 PMCID: PMC8675416 DOI: 10.1002/hbm.25625] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/06/2023] Open
Abstract
Schizophrenia is associated with widespread alterations in subcortical brain structure. While analytic methods have enabled more detailed morphometric characterization, findings are often equivocal. In this meta-analysis, we employed the harmonized ENIGMA shape analysis protocols to collaboratively investigate subcortical brain structure shape differences between individuals with schizophrenia and healthy control participants. The study analyzed data from 2,833 individuals with schizophrenia and 3,929 healthy control participants contributed by 21 worldwide research groups participating in the ENIGMA Schizophrenia Working Group. Harmonized shape analysis protocols were applied to each site's data independently for bilateral hippocampus, amygdala, caudate, accumbens, putamen, pallidum, and thalamus obtained from T1-weighted structural MRI scans. Mass univariate meta-analyses revealed more-concave-than-convex shape differences in the hippocampus, amygdala, accumbens, and thalamus in individuals with schizophrenia compared with control participants, more-convex-than-concave shape differences in the putamen and pallidum, and both concave and convex shape differences in the caudate. Patterns of exaggerated asymmetry were observed across the hippocampus, amygdala, and thalamus in individuals with schizophrenia compared to control participants, while diminished asymmetry encompassed ventral striatum and ventral and dorsal thalamus. Our analyses also revealed that higher chlorpromazine dose equivalents and increased positive symptom levels were associated with patterns of contiguous convex shape differences across multiple subcortical structures. Findings from our shape meta-analysis suggest that common neurobiological mechanisms may contribute to gray matter reduction across multiple subcortical regions, thus enhancing our understanding of the nature of network disorganization in schizophrenia.
Collapse
Affiliation(s)
- Boris A. Gutman
- Department of Biomedical EngineeringIllinois Institute of TechnologyChicagoIllinoisUSA
- Institute for Information Transmission Problems (Kharkevich Institute)MoscowRussia
| | - Theo G.M. van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of California IrvineIrvineCaliforniaUSA
| | - Kathryn Alpert
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Christopher R. K. Ching
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Dmitry Isaev
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Anjani Ragothaman
- Department of biomedical engineeringOregon Health and Science universityPortlandOregonUSA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Arvin Saremi
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Artemis Zavaliangos‐Petropulu
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - David C. Glahn
- Department of PsychiatryBoston Children's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Li Shen
- Department of Biostatistics, Epidemiology and InformaticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Shan Cong
- Department of Biostatistics, Epidemiology and InformaticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Dag Alnæs
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Ole Andreas Andreassen
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Nhat Trung Doan
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
| | - Peter Kochunov
- Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Theodore D. Satterthwaite
- Department of PsychiatryUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Daniel H. Wolf
- Department of PsychiatryUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Alexander J. Huang
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Charles Kessler
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Andrea Weideman
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Dana Nguyen
- Department of PediatricsUniversity of California IrvineIrvineCaliforniaUSA
| | - Bryon A. Mueller
- Department of Psychiatry and Behavioral SciencesUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Lawrence Faziola
- Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Steven G. Potkin
- Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Adrian Preda
- Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Daniel H. Mathalon
- Department of Psychiatry and Weill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Judith Ford Mental HealthVA San Francisco Healthcare SystemSan FranciscoCaliforniaUSA
| | - Juan Bustillo
- Departments of Psychiatry & NeuroscienceUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Vince Calhoun
- Tri‐institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS) [Georgia State University, Georgia Institute of Technology]Emory UniversityAtlantaGeorgiaUSA
- Department of Electrical and Computer EngineeringThe University of New MexicoAlbuquerqueNew MexicoUSA
| | - Judith M. Ford
- Judith Ford Mental HealthVA San Francisco Healthcare SystemSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | | - Stefan Ehrlich
- Division of Psychological & Social Medicine and Developmental NeurosciencesFaculty of Medicine, TU‐DresdenDresdenGermany
| | | | - Nerisa Banaj
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Fabrizio Piras
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Federica Piras
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Gianfranco Spalletta
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of MedicineHoustonTexasUSA
| | | | | | | | - Joaquim Radua
- FIDMAG Germanes Hospitalàries Research FoundationCIBERSAMBarcelonaSpain
- Institut d'Investigacions Biomdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research FoundationCIBERSAMBarcelonaSpain
| | - Salvador Sarró
- FIDMAG Germanes Hospitalàries Research FoundationCIBERSAMBarcelonaSpain
| | - Erin W. Dickie
- Centre for Addiction and Mental Health (CAMH)TorontoCanada
| | | | | | | | | | | | - Stefan Borgwardt
- Department of PsychiatryUniversity of BaselBaselSwitzerland
- Department of Psychiatry and PsychotherapyUniversity of LübeckLübeckGermany
| | | | - Derek Morris
- Centre for Neuroimaging and Cognitive Genomics, Discipline of BiochemistryNational University of Ireland GalwayGalwayIreland
| | - Gary Donohoe
- Centre for Neuroimaging and Cognitive Genomics, School of PsychologyNational University of Ireland GalwayGalwayIreland
| | - Laurena Holleran
- Centre for Neuroimaging and Cognitive Genomics, School of PsychologyNational University of Ireland GalwayGalwayIreland
| | - Dara Cannon
- Clinical Neuroimaging Laboratory, Centre for Neuroimaging and Cognitive GenomicsNational University of Ireland GalwayGalwayIreland
| | - Colm McDonald
- Clinical Neuroimaging Laboratory, Centre for Neuroimaging and Cognitive GenomicsNational University of Ireland GalwayGalwayIreland
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of PsychiatryTrinity College DublinDublinIreland
- Trinity College Institute of NeuroscienceTrinity College DublinDublinIreland
| | - Michael Gill
- Neuropsychiatric Genetics Research Group, Department of PsychiatryTrinity College DublinDublinIreland
- Trinity College Institute of NeuroscienceTrinity College DublinDublinIreland
| | - Geraldo Busatto Filho
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Pedro G. P. Rosa
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Mauricio H. Serpa
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Marcus V. Zanetti
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
- Hospital Sirio‐LibanesSao PauloSPBrazil
| | - Irina Lebedeva
- Laboratory of Neuroimaging and Multimodal AnalysisMental Health Research CenterMoscowRussia
| | - Vasily Kaleda
- Department of Endogenous Mental DisordersMental Health Research CenterMoscowRussia
| | - Alexander Tomyshev
- Laboratory of Neuroimaging and Multimodal AnalysisMental Health Research CenterMoscowRussia
| | - Tim Crow
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Anthony James
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Simon Cervenka
- Centre for Psychiatry Reserach, Department of Clinical NeuroscienceKarolinska Institutet, & Stockholm Health Care Services, Region StockholmStockholmSweden
| | - Carl M Sellgren
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
| | - Helena Fatouros‐Bergman
- Centre for Psychiatry Reserach, Department of Clinical NeuroscienceKarolinska Institutet, & Stockholm Health Care Services, Region StockholmStockholmSweden
| | - Ingrid Agartz
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Fleur Howells
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
- Neuroscience InstituteUniversity of Cape Town, Cape TownWCSouth Africa
| | - Dan J. Stein
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
- Neuroscience InstituteUniversity of Cape Town, Cape TownWCSouth Africa
- SA MRC Unit on Risk & Resilience in Mental DisordersUniversity of Cape TownCape TownWCSouth Africa
| | - Henk Temmingh
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
| | - Anne Uhlmann
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
- Department of Child and Adolescent PsychiatryTU DresdenGermany
| | - Greig I. de Zubicaray
- School of Psychology, Faculty of HealthQueensland University of Technology (QUT)BrisbaneQLDAustralia
| | - Katie L. McMahon
- School of Clinical SciencesQueensland University of Technology (QUT)BrisbaneQLDAustralia
| | - Margie Wright
- Queensland Brain InstituteUniversity of QueenslandBrisbaneQLDAustralia
| | - Derin Cobia
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of Psychology and Neuroscience CenterBrigham Young UniversityProvoUtahUSA
| | - John G. Csernansky
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Lei Wang
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of Psychiatry and Behavioral HealthOhio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
83
|
Quek YE, Fung YL, Cheung MWL, Vogrin SJ, Collins SJ, Bowden SC. Agreement Between Automated and Manual MRI Volumetry in Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Magn Reson Imaging 2021; 56:490-507. [PMID: 34964531 DOI: 10.1002/jmri.28037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Automated magnetic resonance imaging (MRI) volumetry is a promising tool to evaluate regional brain volumes in dementia and especially Alzheimer's disease (AD). PURPOSE To compare automated methods and the gold standard manual segmentation in measuring regional brain volumes on MRI across healthy controls, patients with mild cognitive impairment, and patients with dementia due to AD. STUDY TYPE Systematic review and meta-analysis. DATA SOURCES MEDLINE, Embase, and PsycINFO were searched through October 2021. FIELD STRENGTH 1.0 T, 1.5 T, or 3.0 T. ASSESSMENT Two review authors independently identified studies for inclusion and extracted data. Methodological quality was assessed using the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2). STATISTICAL TESTS Standardized mean differences (SMD; Hedges' g) were pooled using random-effects meta-analysis with robust variance estimation. Subgroup analyses were undertaken to explore potential sources of heterogeneity. Sensitivity analyses were conducted to examine the impact of the within-study correlation between effect estimates on the meta-analysis results. RESULTS Seventeen studies provided sufficient data to evaluate the hippocampus, lateral ventricles, and parahippocampal gyrus. The pooled SMD for the hippocampus, lateral ventricles, and parahippocampal gyrus were 0.22 (95% CI -0.50 to 0.93), 0.12 (95% CI -0.13 to 0.37), and -0.48 (95% CI -1.37 to 0.41), respectively. For the hippocampal data, subgroup analyses suggested that the pooled SMD was invariant across clinical diagnosis and field strength. Subgroup analyses could not be conducted on the lateral ventricles data and the parahippocampal gyrus data due to insufficient data. The results were robust to the selected within-study correlation value. DATA CONCLUSION While automated methods are generally comparable to manual segmentation for measuring hippocampal, lateral ventricle, and parahippocampal gyrus volumes, wide 95% CIs and large heterogeneity suggest that there is substantial uncontrolled variance. Thus, automated methods may be used to measure these regions in patients with AD but should be used with caution. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Yi-En Quek
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Yi Leng Fung
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Mike W-L Cheung
- Department of Psychology, Faculty of Arts and Social Sciences, National University of Singapore, Singapore
| | - Simon J Vogrin
- Department of Clinical Neurosciences, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Steven J Collins
- Department of Clinical Neurosciences, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| | - Stephen C Bowden
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, Victoria, Australia.,Department of Clinical Neurosciences, St Vincent's Hospital Melbourne, Fitzroy, Victoria, Australia
| |
Collapse
|
84
|
Kwak K, Giovanello KS, Bozoki A, Styner M, Dayan E. Subtyping of mild cognitive impairment using a deep learning model based on brain atrophy patterns. Cell Rep Med 2021; 2:100467. [PMID: 35028609 PMCID: PMC8714856 DOI: 10.1016/j.xcrm.2021.100467] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/08/2021] [Accepted: 11/13/2021] [Indexed: 12/28/2022]
Abstract
Trajectories of cognitive decline vary considerably among individuals with mild cognitive impairment (MCI). To address this heterogeneity, subtyping approaches have been developed, with the objective of identifying more homogeneous subgroups. To date, subtyping of MCI has been based primarily on cognitive measures, often resulting in indistinct boundaries between subgroups and limited validity. Here, we introduce a subtyping method for MCI based solely upon brain atrophy. We train a deep learning model to differentiate between Alzheimer's disease (AD) and cognitively normal (CN) subjects based on whole-brain MRI features. We then deploy the trained model to classify MCI subjects based on whole-brain gray matter resemblance to AD-like or CN-like patterns. We subsequently validate the subtyping approach using cognitive, clinical, fluid biomarker, and molecular imaging data. Overall, the results suggest that atrophy patterns in MCI are sufficiently heterogeneous and can thus be used to subtype individuals into biologically and clinically meaningful subgroups.
Collapse
Affiliation(s)
- Kichang Kwak
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kelly S. Giovanello
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Andrea Bozoki
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Martin Styner
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eran Dayan
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
85
|
Yu T, Liu X, Wu J, Wang Q. Electrophysiological Biomarkers of Epileptogenicity in Alzheimer's Disease. Front Hum Neurosci 2021; 15:747077. [PMID: 34916917 PMCID: PMC8669481 DOI: 10.3389/fnhum.2021.747077] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
Cortical network hyperexcitability is an inextricable feature of Alzheimer’s disease (AD) that also might accelerate its progression. Seizures are reported in 10–22% of patients with AD, and subclinical epileptiform abnormalities have been identified in 21–42% of patients with AD without seizures. Accurate identification of hyperexcitability and appropriate intervention to slow the compromise of cognitive functions of AD might open up a new approach to treatment. Based on the results of several studies, epileptiform discharges, especially those with specific features (including high frequency, robust morphology, right temporal location, and occurrence during awake or rapid eye movement states), frequent small sharp spikes (SSSs), temporal intermittent rhythmic delta activities (TIRDAs), and paroxysmal slow wave events (PSWEs) recorded in long-term scalp electroencephalogram (EEG) provide sufficient sensitivity and specificity in detecting cortical network hyperexcitability and epileptogenicity of AD. In addition, magnetoencephalogram (MEG), foramen ovale (FO) electrodes, and computational approaches help to find subclinical seizures that are invisible on scalp EEGs. We performed a comprehensive analysis of the aforementioned electrophysiological biomarkers of AD-related seizures.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiao Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jianping Wu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
86
|
Di Tella S, Cabinio M, Isernia S, Blasi V, Rossetto F, Saibene FL, Alberoni M, Silveri MC, Sorbi S, Clerici M, Baglio F. Neuroimaging Biomarkers Predicting the Efficacy of Multimodal Rehabilitative Intervention in the Alzheimer's Dementia Continuum Pathology. Front Aging Neurosci 2021; 13:735508. [PMID: 34880742 PMCID: PMC8645692 DOI: 10.3389/fnagi.2021.735508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/30/2021] [Indexed: 12/19/2022] Open
Abstract
In this work we aimed to identify neural predictors of the efficacy of multimodal rehabilitative interventions in AD-continuum patients in the attempt to identify ideal candidates to improve the treatment outcome. Subjects in the AD continuum who participated in a multimodal rehabilitative treatment were included in the analysis [n = 82, 38 Males, mean age = 76 ± 5.30, mean education years = 9.09 ± 3.81, Mini Mental State Examination (MMSE) mean score = 23.31 ± 3.81]. All subjects underwent an MRI acquisition (1.5T) at baseline (T0) and a neuropsychological evaluation before (T0) and after intervention (T1). All subjects underwent an intensive multimodal cognitive rehabilitation (8–10 weeks). The MMSE and Neuropsychiatric Inventory (NPI) scores were considered as the main cognitive and behavioral outcome measures, and Delta change scores (T1–T0) were categorized in Improved (ΔMMSE > 0; ΔNPI < 0) and Not Improved (ΔMMSE ≤ 0; ΔNPI ≥ 0). Logistic Regression (LR) and Random Forest classification models were performed including neural markers (Medial Temporal Brain; Posterior Brain (PB); Frontal Brain (FB), Subcortical Brain indexes), neuropsychological (MMSE, NPI, verbal fluencies), and demographical variables (sex, age, education) at baseline. More than 50% of patients showed a positive effect of the treatment (ΔMMSE > 0: 51%, ΔNPI < 0: 52%). LR model on ΔMMSE (Improved vs. Not Improved) indicate a predictive role for MMSE score (p = 0.003) and PB index (p = 0.005), especially the right PB (p = 0.002) at baseline. The Random Forest analysis correctly classified 77% of cognitively improved and not improved AD patients. Concerning the NPI, LR model on ΔNPI (Improved vs. Not Improved) showed a predictive role of sex (p = 0.002), NPI (p = 0.005), PB index (p = 0.006), and FB index (p = 0.039) at baseline. The Random Forest reported a classification accuracy of 86%. Our data indicate that cognitive and behavioral status alone are not sufficient to identify best responders to a multidomain rehabilitation treatment. Increased neural reserve, especially in the parietal areas, is also relevant for the compensatory mechanisms activated by rehabilitative treatment. These data are relevant to support clinical decision by identifying target patients with high probability of success after rehabilitative programs on cognitive and behavioral functioning.
Collapse
Affiliation(s)
- Sonia Di Tella
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy.,Department of Psychology, Università Cattolica del Sacro Cuore, Milan, Italy
| | - Monia Cabinio
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Sara Isernia
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Valeria Blasi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | | | | | | | - Maria Caterina Silveri
- Department of Psychology, Università Cattolica del Sacro Cuore, Milan, Italy.,Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Sandro Sorbi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy.,Università degli Studi di Firenze, NEUROFARBA, Firenze, Italy
| | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy.,Department of Physiopathology and Transplants, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
87
|
Blinkouskaya Y, Caçoilo A, Gollamudi T, Jalalian S, Weickenmeier J. Brain aging mechanisms with mechanical manifestations. Mech Ageing Dev 2021; 200:111575. [PMID: 34600936 PMCID: PMC8627478 DOI: 10.1016/j.mad.2021.111575] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022]
Abstract
Brain aging is a complex process that affects everything from the subcellular to the organ level, begins early in life, and accelerates with age. Morphologically, brain aging is primarily characterized by brain volume loss, cortical thinning, white matter degradation, loss of gyrification, and ventricular enlargement. Pathophysiologically, brain aging is associated with neuron cell shrinking, dendritic degeneration, demyelination, small vessel disease, metabolic slowing, microglial activation, and the formation of white matter lesions. In recent years, the mechanics community has demonstrated increasing interest in modeling the brain's (bio)mechanical behavior and uses constitutive modeling to predict shape changes of anatomically accurate finite element brain models in health and disease. Here, we pursue two objectives. First, we review existing imaging-based data on white and gray matter atrophy rates and organ-level aging patterns. This data is required to calibrate and validate constitutive brain models. Second, we review the most critical cell- and tissue-level aging mechanisms that drive white and gray matter changes. We focuse on aging mechanisms that ultimately manifest as organ-level shape changes based on the idea that the integration of imaging and mechanical modeling may help identify the tipping point when normal aging ends and pathological neurodegeneration begins.
Collapse
Affiliation(s)
- Yana Blinkouskaya
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Andreia Caçoilo
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Trisha Gollamudi
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Shima Jalalian
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States.
| |
Collapse
|
88
|
Anderson JAE, Grundy JG, Grady CL, Craik FIM, Bialystok E. Bilingualism contributes to reserve and working memory efficiency: Evidence from structural and functional neuroimaging. Neuropsychologia 2021; 163:108071. [PMID: 34715120 DOI: 10.1016/j.neuropsychologia.2021.108071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/14/2021] [Accepted: 10/22/2021] [Indexed: 01/26/2023]
Abstract
This study compared brain and behavioral outcomes for monolingual and bilingual older adults who reported no cognitive or memory problems on three types of memory that typically decline in older age, namely, working memory (measured by n-back), item, and associative recognition. The results showed that bilinguals were faster on the two-back working memory task than monolinguals but used a set of frontostriatal regions less than monolinguals. There was no group difference on an item/associative recognition task. In brain structure, gray matter volume and white matter integrity (fractional anisotropy) were generally lower in bilinguals than in monolinguals, but bilinguals had better white matter integrity than monolinguals in the bilateral superior corona radiata and better gray matter density in the left inferior temporal gyrus. These regions may help preserve bilinguals' executive functions despite generally more significant atrophy throughout the brain than monolinguals in that these structures contribute to efficient communication between executive frontal regions and subcortical motor regions, and perceptual pathways. Reliable negative correlations between brain structure and age were only observed in bilinguals, and to the extent that bilinguals (but not monolinguals) had better brain structure, their performance was enhanced. Collectively, the findings provide evidence for reserve in bilingual older adults.
Collapse
Affiliation(s)
- John A E Anderson
- Carleton University, Departments of Cognitive Science and Psychology, Canada.
| | - John G Grundy
- Iowa State University, Department of Psychology, United States
| | - Cheryl L Grady
- Rotman Research Institute at Baycrest Hospital, Canada; University of Toronto, Department of Psychiatry, Canada; University of Toronto, Department of Psychology, Canada
| | - Fergus I M Craik
- Rotman Research Institute at Baycrest Hospital, Canada; University of Toronto, Department of Psychology, Canada
| | - Ellen Bialystok
- Rotman Research Institute at Baycrest Hospital, Canada; York University, Department of Psychology, Canada
| |
Collapse
|
89
|
Panitch R, Hu J, Chung J, Zhu C, Meng G, Xia W, Bennett DA, Lunetta KL, Ikezu T, Au R, Stein TD, Farrer LA, Jun GR. Integrative brain transcriptome analysis links complement component 4 and HSPA2 to the APOE ε2 protective effect in Alzheimer disease. Mol Psychiatry 2021; 26:6054-6064. [PMID: 34480088 PMCID: PMC8758485 DOI: 10.1038/s41380-021-01266-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 07/07/2021] [Accepted: 08/12/2021] [Indexed: 02/08/2023]
Abstract
Mechanisms underlying the protective effect of apolipoprotein E (APOE) ε2 against Alzheimer disease (AD) are not well understood. We analyzed gene expression data derived from autopsied brains donated by 982 individuals including 135 APOE ɛ2/ɛ3 carriers. Complement pathway genes C4A and C4B were among the most significantly differentially expressed genes between ɛ2/ɛ3 AD cases and controls. We also identified an APOE ε2/ε3 AD-specific co-expression network enriched for astrocytes, oligodendrocytes and oligodendrocyte progenitor cells containing the genes C4A, C4B, and HSPA2. These genes were significantly associated with the ratio of phosphorylated tau at position 231 to total Tau but not with amyloid-β 42 level, suggesting this APOE ɛ2 related co-expression network may primarily be involved with tau pathology. HSPA2 expression was oligodendrocyte-specific and significantly associated with C4B protein. Our findings provide the first evidence of a crucial role of the complement pathway in the protective effect of APOE ε2 for AD.
Collapse
Affiliation(s)
- Rebecca Panitch
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Junming Hu
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Jaeyoon Chung
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Congcong Zhu
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA
| | - Gaoyuan Meng
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
| | - Weiming Xia
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Tsuneya Ikezu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Center for Systems Neuroscience, Boston University School of Medicine, Boston, MA, USA
| | - Rhoda Au
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Thor D Stein
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- Department of Pathology & Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA.
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA.
| | - Gyungah R Jun
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA, USA.
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
90
|
Skipper JI, Aliko S, Brown S, Jo YJ, Lo S, Molimpakis E, Lametti DR. Reorganization of the Neurobiology of Language After Sentence Overlearning. Cereb Cortex 2021; 32:2447-2468. [PMID: 34585723 PMCID: PMC9157312 DOI: 10.1093/cercor/bhab354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 11/14/2022] Open
Abstract
It is assumed that there are a static set of "language regions" in the brain. Yet, language comprehension engages regions well beyond these, and patients regularly produce familiar "formulaic" expressions when language regions are severely damaged. These suggest that the neurobiology of language is not fixed but varies with experiences, like the extent of word sequence learning. We hypothesized that perceiving overlearned sentences is supported by speech production and not putative language regions. Participants underwent 2 sessions of behavioral testing and functional magnetic resonance imaging (fMRI). During the intervening 15 days, they repeated 2 sentences 30 times each, twice a day. In both fMRI sessions, they "passively" listened to those sentences, novel sentences, and produced sentences. Behaviorally, evidence for overlearning included a 2.1-s decrease in reaction times to predict the final word in overlearned sentences. This corresponded to the recruitment of sensorimotor regions involved in sentence production, inactivation of temporal and inferior frontal regions involved in novel sentence listening, and a 45% change in global network organization. Thus, there was a profound whole-brain reorganization following sentence overlearning, out of "language" and into sensorimotor regions. The latter are generally preserved in aphasia and Alzheimer's disease, perhaps explaining residual abilities with formulaic expressions in both.
Collapse
Affiliation(s)
| | - Sarah Aliko
- Experimental Psychology, University College London, London, UK.,London Interdisciplinary Biosciences Consortium, University College London, London, UK
| | - Stephen Brown
- Natural Sciences, University College London, London, UK
| | - Yoon Ju Jo
- Experimental Psychology, University College London, London, UK
| | - Serena Lo
- Speech and Language Sciences, University College London, London, UK
| | - Emilia Molimpakis
- Wellcome Centre for Human Neuroimaging, University College London, London, UK
| | - Daniel R Lametti
- Experimental Psychology, University College London, London, UK.,Department of Psychology, Acadia University, Nova Scotia, Canada
| |
Collapse
|
91
|
Jesus B, Cassani R, McGeown WJ, Cecchi M, Fadem KC, Falk TH. Multimodal Prediction of Alzheimer's Disease Severity Level Based on Resting-State EEG and Structural MRI. Front Hum Neurosci 2021; 15:700627. [PMID: 34566600 PMCID: PMC8458963 DOI: 10.3389/fnhum.2021.700627] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
While several biomarkers have been developed for the detection of Alzheimer's disease (AD), not many are available for the prediction of disease severity, particularly for patients in the mild stages of AD. In this paper, we explore the multimodal prediction of Mini-Mental State Examination (MMSE) scores using resting-state electroencephalography (EEG) and structural magnetic resonance imaging (MRI) scans. Analyses were carried out on a dataset comprised of EEG and MRI data collected from 89 patients diagnosed with minimal-mild AD. Three feature selection algorithms were assessed alongside four machine learning algorithms. Results showed that while MRI features alone outperformed EEG features, when both modalities were combined, improved results were achieved. The top-selected EEG features conveyed information about amplitude modulation rate-of-change, whereas top-MRI features comprised information about cortical area and white matter volume. Overall, a root mean square error between predicted MMSE values and true MMSE scores of 1.682 was achieved with a multimodal system and a random forest regression model.
Collapse
Affiliation(s)
- Belmir Jesus
- Institut National de la Recherche Scientifique, University of Quebec, Montreal, QC, Canada
| | - Raymundo Cassani
- Institut National de la Recherche Scientifique, University of Quebec, Montreal, QC, Canada
| | - William J McGeown
- School of Psychological Sciences and Health, University of Strathclyde, Glasgow, United Kingdom
| | | | - K C Fadem
- COGNISION, Louisville, KY, United States
| | - Tiago H Falk
- Institut National de la Recherche Scientifique, University of Quebec, Montreal, QC, Canada
| |
Collapse
|
92
|
Zeng Q, Li K, Luo X, Wang S, Xu X, Li Z, Zhang T, Liu X, Fu Y, Xu X, Wang C, Wang T, Zhou J, Liu Z, Chen Y, Huang P, Zhang M. Distinct Atrophy Pattern of Hippocampal Subfields in Patients with Progressive and Stable Mild Cognitive Impairment: A Longitudinal MRI Study. J Alzheimers Dis 2021; 79:237-247. [PMID: 33252076 DOI: 10.3233/jad-200775] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Predicting the prognosis of mild cognitive impairment (MCI) has outstanding clinical value, and the hippocampal volume is a reliable imaging biomarker of AD diagnosis. OBJECTIVE We aimed to longitudinally assess hippocampal sub-regional difference (volume and asymmetry) among progressive MCI (pMCI), stable MCI (sMCI) patients, and normal elderly. METHODS We identified 29 pMCI, 52 sMCI, and 102 normal controls (NC) from the ADNI database. All participants underwent neuropsychological assessment and 3T MRI scans three times. The time interval between consecutive MRI sessions was about 1 year. Volumes of hippocampal subfield were measured by Freesurfer. Based on the analysis of variance, repeated measures analyses, and receiver operating characteristic curves, we compared cross-sectional and longitudinal alteration sub-regional volume and asymmetry index. RESULTS Compared to NC, both MCI groups showed significant atrophy in all subfields. At baseline, pMCI have a smaller volume than sMCI in the bilateral subiculum, molecular layer (ML), the molecular and granule cell layers of the dentate gyrus, cornu ammonis 4, and right tail. Furthermore, repeated measures analyses revealed that pMCI patients showed a faster volume loss than sMCI in bilateral subiculum and ML. After controlling for age, gender, and education, most results remained unchanged. However, none of the hippocampal sub-regional volumes performed better than the whole hippocampus in ROC analyses, and no asymmetric difference between pMCI and sMCI was found. CONCLUSION The faster volume loss in subiculum and ML suggest a higher risk of disease progression in MCI patients. The hippocampal asymmetry may have smaller value in predicting the MCI prognosis.
Collapse
Affiliation(s)
- Qingze Zeng
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Kaicheng Li
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Xiao Luo
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Shuyue Wang
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Xiaopei Xu
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Zheyu Li
- Department of Neurology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Tianyi Zhang
- Department of Neurology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Xiaocao Liu
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Yanv Fu
- Department of Neurology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Xiaojun Xu
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Chao Wang
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Tao Wang
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Jiong Zhou
- Department of Neurology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Zhirong Liu
- Department of Neurology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Yanxing Chen
- Department of Neurology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Peiyu Huang
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | - Minming Zhang
- Department of Radiology, 2nd Affiliated Hospital of Zhejiang University School of Medicine, China
| | | |
Collapse
|
93
|
Lubben N, Ensink E, Coetzee GA, Labrie V. The enigma and implications of brain hemispheric asymmetry in neurodegenerative diseases. Brain Commun 2021; 3:fcab211. [PMID: 34557668 PMCID: PMC8454206 DOI: 10.1093/braincomms/fcab211] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 01/15/2023] Open
Abstract
The lateralization of the human brain may provide clues into the pathogenesis and progression of neurodegenerative diseases. Though differing in their presentation and underlying pathologies, neurodegenerative diseases are all devastating and share an intriguing theme of asymmetrical pathology and clinical symptoms. Parkinson’s disease, with its distinctive onset of motor symptoms on one side of the body, stands out in this regard, but a review of the literature reveals asymmetries in several other neurodegenerative diseases. Here, we review the lateralization of the structure and function of the healthy human brain and the common genetic and epigenetic patterns contributing to the development of asymmetry in health and disease. We specifically examine the role of asymmetry in Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, and interrogate whether these imbalances may reveal meaningful clues about the origins of these diseases. We also propose several hypotheses for how lateralization may contribute to the distinctive and enigmatic features of asymmetry in neurodegenerative diseases, suggesting a role for asymmetry in the choroid plexus, neurochemistry, protein distribution, brain connectivity and the vagus nerve. Finally, we suggest how future studies may reveal novel insights into these diseases through the lens of asymmetry.
Collapse
Affiliation(s)
- Noah Lubben
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Elizabeth Ensink
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Gerhard A Coetzee
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Viviane Labrie
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
94
|
Karoly HC, Skrzynski CJ, Moe E, Bryan AD, Hutchison KE. Investigating Associations Between Inflammatory Biomarkers, Gray Matter, Neurofilament Light and Cognitive Performance in Healthy Older Adults. Front Aging Neurosci 2021; 13:719553. [PMID: 34539381 PMCID: PMC8446648 DOI: 10.3389/fnagi.2021.719553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/29/2021] [Indexed: 11/18/2022] Open
Abstract
Background: Exploring biological variables that may serve as indicators of the development and progression of cognitive decline is currently a high-priority research area. Recent studies have demonstrated that during normal aging, individuals experience increased inflammation throughout the brain and body, which may be linked to cognitive impairment and reduced gray matter volume in the brain. Neurofilament light polypeptide (NfL), which is released into the circulation following neuronal damage, has been proposed as a biomarker for neurodegenerative diseases, and may also have utility in the context of normal aging. The present study tested associations between age, peripheral levels of the pro-inflammatory cytokine IL-6, peripheral NfL, brain volume, and cognitive performance in a sample of healthy adults over 60 years old. Methods: Of the 273 individuals who participated in this study, 173 had useable neuroimaging data, a subset of whom had useable blood data (used for quantifying IL-6 and NfL) and completed a cognitive task. Gray matter (GM) thickness values were extracted from brain areas of interest using Freesurfer. Regression models were used to test relationships between IL-6, NfL, GM, and cognitive performance. To test putative functional relationships between these variables, exploratory path analytic models were estimated, in which the relationship between age, IL-6, and working memory performance were linked via four different operationalizations of brain health: (1) a latent GM variable composed of several regions linked to cognitive impairment, (2) NfL alone, (3) NfL combined with the GM latent variable, and (4) the hippocampus alone. Results: Regression models showed that IL-6 and NfL were significantly negatively associated with GM volume and that GM was positively associated with cognitive performance. The path analytic models indicated that age and cognitive performance are linked by GM in the hippocampus as well as several other regions previously associated with cognitive impairment, but not by NfL alone. Peripheral IL-6 was not associated with age in any of the path models. Conclusions: Results suggest that among healthy older adults, there are several GM regions that link age and cognitive performance. Notably, NfL alone is not a sufficient marker of brain changes associated with aging, inflammation, and cognitive performance.
Collapse
Affiliation(s)
- Hollis C Karoly
- Institute for Cognitive Science, University of Colorado Boulder, Boulder, CO, United States.,Department of Psychology, Colorado State University, Fort Collins, CO, United States
| | - Carillon J Skrzynski
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, United States
| | - Erin Moe
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angela D Bryan
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, United States
| | - Kent E Hutchison
- Institute for Cognitive Science, University of Colorado Boulder, Boulder, CO, United States.,Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, United States.,Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
95
|
Han F, Brown GL, Zhu Y, Belkin-Rosen AE, Lewis MM, Du G, Gu Y, Eslinger PJ, Mailman RB, Huang X, Liu X. Decoupling of Global Brain Activity and Cerebrospinal Fluid Flow in Parkinson's Disease Cognitive Decline. Mov Disord 2021; 36:2066-2076. [PMID: 33998068 PMCID: PMC8453044 DOI: 10.1002/mds.28643] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Deposition and spreading of misfolded proteins (α-synuclein and tau) have been linked to Parkinson's disease cognitive dysfunction. The glymphatic system may play an important role in the clearance of these toxic proteins via cerebrospinal fluid (CSF) flow through perivascular and interstitial spaces. Recent studies discovered that sleep-dependent global brain activity is coupled to CSF flow, which may reflect glymphatic function. OBJECTIVE The objective of this current study was to determine if the decoupling of brain activity-CSF flow is linked to Parkinson's disease cognitive dysfunction. METHODS Functional and structural MRI data, clinical motor (Unified Parkinson's Disease Rating Scale), and cognitive (Montreal Cognitive Assessment [MoCA]) scores were collected from 60 Parkinson's disease and 58 control subjects. Parkinson's disease patients were subgrouped into those with mild cognitive impairment (MoCA < 26), n = 31, and those without mild cognitive impairment (MoCA ≥ 26), n = 29. The coupling strength between the resting-state global blood-oxygen-level-dependent signal and associated CSF flow was quantified, compared among groups, and associated with clinical and structural measurements. RESULTS Global blood-oxygen-level-dependent signal-CSF coupling decreased significantly (P < 0.006) in Parkinson's disease patients showing mild cognitive impairment, compared with those without mild cognitive impairment and controls. Reduced global blood-oxygen-level-dependent signal-CSF coupling was associated with decreased MoCA scores present in Parkinson's disease patients (P = 0.005) but not in controls (P = 0.65). Weaker global blood-oxygen-level-dependent signal-CSF coupling in Parkinson's disease patients also was associated with a thinner right entorhinal cortex (Spearman's correlation, -0.36; P = 0.012), an early structural change often seen in Alzheimer's disease. CONCLUSIONS The decoupling between global brain activity and associated CSF flow is related to Parkinson's disease cognitive impairment. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Feng Han
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Gregory L. Brown
- Department of Engineering Science and Mechanics, The Pennsylvania State University, PA, USA
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Yalin Zhu
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | | | - Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Yameng Gu
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
| | - Paul J. Eslinger
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Radiology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Richard B. Mailman
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Radiology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neurosurgery, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Kinesiology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Institute for Computational and Data Sciences, The Pennsylvania State University, PA, USA
| | - Xiao Liu
- Department of Biomedical Engineering, The Pennsylvania State University, PA, USA
- Institute for Computational and Data Sciences, The Pennsylvania State University, PA, USA
| |
Collapse
|
96
|
An experimental analysis of different Deep Learning based Models for Alzheimer’s Disease classification using Brain Magnetic Resonance Images. JOURNAL OF KING SAUD UNIVERSITY - COMPUTER AND INFORMATION SCIENCES 2021. [DOI: 10.1016/j.jksuci.2021.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
97
|
Cacciaguerra L, Valsasina P, Meani A, Riccitelli GC, Radaelli M, Rocca MA, Filippi M. Volume of hippocampal subfields and cognitive deficits in neuromyelitis optica spectrum disorders. Eur J Neurol 2021; 28:4167-4177. [PMID: 34415660 DOI: 10.1111/ene.15073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Aquaporin-4 (AQP4) water channel is involved in hippocampal plasticity and is the target of neuromyelitis optica spectrum disorders (NMOSD) autoimmunity. We measured volumes of hippocampal subfields and their association with cognitive performance in AQP4-seropositive NMOSD patients. METHODS Global and regional hippocampal volumes were derived from 28 AQP4-seropositive NMOSD patients and 101 healthy controls (HC) from 3D-T1-weighted images. Normalized brain volumes were also calculated. A neuropsychological evaluation, including the Brief Repeatable Battery of Neuropsychological Tests, was performed in patients. Based on HC data, we estimated mean z-scores of volumes in the whole NMOSD group and compared them according to the status of global and domain-selective cognitive impairment. RESULTS Global cognitive impairment was detected in 46.4% of NMOSD patients, with attentive (60.7%) and executive (21.4%) domains being the most affected. NMOSD patients had left hippocampal atrophy at global (p = 0.012) and regional level (fimbria, Cornu Ammonis [CA] 3, molecular layer, dentate gyrus [DG], and subicular complex, p values ranging between 0.033 and <0.001). On the right side the fimbria and hippocampal tail were atrophic (p = 0.024 for both). Cognitively impaired patients showed atrophy in the left CA3 and CA4 (p = 0.025-0.028), while patients presenting verbal and visual memory impairment had significant CA3 and DG atrophy. Those patients with attentive or executive impairment had preserved brain and hippocampal volumes. CONCLUSIONS NMOSD patients showed hippocampal atrophy associated with verbal and visual memory impairment. Such damage did not explain attention and executive function alterations, which were the most common cognitive deficits in this population.
Collapse
Affiliation(s)
- Laura Cacciaguerra
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Paola Valsasina
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Meani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianna C Riccitelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Radaelli
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
98
|
Scarth M, Rissanen I, Scholten RJPM, Geerlings MI. Biomarkers of Alzheimer's Disease and Cerebrovascular Lesions and Clinical Progression in Patients with Subjective Cognitive Decline: A Systematic Review. J Alzheimers Dis 2021; 83:1089-1111. [PMID: 34397412 DOI: 10.3233/jad-210218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Early identification of Alzheimer's disease (AD) may be extremely beneficial for delaying disease progression. Subjective cognitive decline (SCD) may be an early indicator of AD pathology. Not all individuals with SCD will eventually develop AD, making it critical to identify biomarkers during the SCD stage which indicate likely clinical progression. OBJECTIVE The present review aims to summarize available data on structural MRI and cerebrospinal fluid (CSF) biomarkers and their association with clinical progression to mild cognitive impairment (MCI) or AD in people with SCD. METHODS Database searches were conducted using Embase and PubMed until June 2020. Longitudinal studies assessing biomarkers in individuals with SCD and assessing clinical progression to MCI/AD were included. Two assessors performed data extraction and assessed the risk of bias in the included studies. Data were synthesized narratively. RESULTS An initial search identified 1,065 papers; after screening and review 14 studies were included. Sample size of the included studies ranged from 28-674, mean age was 60.0-68.6 years, and 10.2%-52%of participants converted to MCI/AD. Lower levels of CSF Aβ 42 were consistently associated with clinical progression. Combination measures identifying an AD-like profile of Aβ 42 and tau levels were strongly associated with clinical progression. Biomarkers identified with structural MRI were less conclusive, as some studies found significant associations while others did not. CONCLUSION Biomarkers may be able to predict clinical progression in those with cognitive complaints. Aβ 42, or combinations of Aβ 42 and tau may be useful biomarkers in identifying individuals with SCD who will progress to MCI/AD.
Collapse
Affiliation(s)
- Morgan Scarth
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Ina Rissanen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Rob J P M Scholten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Mirjam I Geerlings
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
99
|
Gajdošík M, Landheer K, Swanberg KM, Adlparvar F, Madelin G, Bogner W, Juchem C, Kirov II. Hippocampal single-voxel MR spectroscopy with a long echo time at 3 T using semi-LASER sequence. NMR IN BIOMEDICINE 2021; 34:e4538. [PMID: 33956374 PMCID: PMC10874619 DOI: 10.1002/nbm.4538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/01/2021] [Accepted: 04/19/2021] [Indexed: 06/12/2023]
Abstract
The hippocampus is one of the most challenging brain regions for proton MR spectroscopy (MRS) applications. Moreover, quantification of J-coupled species such as myo-inositol (m-Ins) and glutamate + glutamine (Glx) is affected by the presence of macromolecular background. While long echo time (TE) MRS eliminates the macromolecules, it also decreases the m-Ins and Glx signal and, as a result, these metabolites are studied mainly with short TE. Here, we investigate the feasibility of reproducibly measuring their concentrations at a long TE of 120 ms, using a semi-adiabatic localization by adiabatic selective refocusing (sLASER) sequence, as this sequence was recently recommended as a standard for clinical MRS. Gradient offset-independent adiabatic refocusing pulses were implemented, and an optimal long TE for the detection of m-Ins and Glx was determined using the T2 relaxation times of macromolecules. Metabolite concentrations and their coefficients of variation (CVs) were obtained for a 3.4-mL voxel centered on the left hippocampus on 3-T MR systems at two different sites with three healthy subjects (aged 32.5 ± 10.2 years [mean ± standard deviation]) per site, with each subject scanned over two sessions, and with each session comprising three scans. Concentrations of m-Ins, choline, creatine, Glx and N-acetyl-aspartate were 5.4 ± 1.5, 1.7 ± 0.2, 5.8 ± 0.3, 11.6 ± 1.2 and 5.9 ± 0.4 mM (mean ± standard deviation), respectively. Their respective mean within-session CVs were 14.5% ± 5.9%, 6.5% ± 5.3%, 6.0% ± 3.4%, 10.6% ± 6.2% and 3.5% ± 1.4%, and their mean within-subject CVs were 17.8% ± 18.2%, 7.5% ± 6.3%, 7.4% ± 6.4%, 12.4% ± 5.3% and 4.8% ± 3.0%. The between-subject CVs were 25.0%, 12.3%, 5.3%, 10.7% and 6.4%, respectively. Hippocampal long-TE sLASER single voxel spectroscopy can provide macromolecule-independent assessment of all major metabolites including Glx and m-Ins.
Collapse
Affiliation(s)
- Martin Gajdošík
- Center for Advanced Imaging Innovation and Research (CAIR), Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science, New York, NY, United States
| | - Karl Landheer
- Department of Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science, New York, NY, United States
| | - Kelley M. Swanberg
- Department of Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science, New York, NY, United States
| | - Fatemeh Adlparvar
- Center for Advanced Imaging Innovation and Research (CAIR), Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Guillaume Madelin
- Center for Advanced Imaging Innovation and Research (CAIR), Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Wolfgang Bogner
- High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christoph Juchem
- Department of Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science, New York, NY, United States
- Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Ivan I. Kirov
- Center for Advanced Imaging Innovation and Research (CAIR), Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
100
|
OmidYeganeh M, Khalili-Mahani N, Bermudez P, Ross A, Lepage C, Vincent RD, Jeon S, Lewis LB, Das S, Zijdenbos AP, Rioux P, Adalat R, Van Eede MC, Evans AC. A Simulation Toolkit for Testing the Sensitivity and Accuracy of Corticometry Pipelines. Front Neuroinform 2021; 15:665560. [PMID: 34381348 PMCID: PMC8350777 DOI: 10.3389/fninf.2021.665560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 11/25/2022] Open
Abstract
In recent years, the replicability of neuroimaging findings has become an important concern to the research community. Neuroimaging pipelines consist of myriad numerical procedures, which can have a cumulative effect on the accuracy of findings. To address this problem, we propose a method for simulating artificial lesions in the brain in order to estimate the sensitivity and specificity of lesion detection, using different automated corticometry pipelines. We have applied this method to different versions of two widely used neuroimaging pipelines (CIVET and FreeSurfer), in terms of coefficients of variation; sensitivity and specificity of detecting lesions in 4 different regions of interest in the cortex, while introducing variations to the lesion size, the blurring kernel used prior to statistical analyses, and different thickness metrics (in CIVET). These variations are tested in a between-subject design (in two random groups, with and without lesions, using T1-weigted MRIs of 152 individuals from the International Consortium of Brain Mapping (ICBM) dataset) and in a within-subject pre-/post-lesion design [using 21 T1-Weighted MRIs of a single adult individual, scanned in the Infant Brain Imaging Study (IBIS)]. The simulation method is sensitive to partial volume effect and lesion size. Comparisons between pipelines illustrate the ability of this method to uncover differences in sensitivity and specificity of lesion detection. We propose that this method be adopted in the workflow of software development and release.
Collapse
Affiliation(s)
- Mona OmidYeganeh
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Najmeh Khalili-Mahani
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada.,PERFORM Centre, Concordia University, Montreal, QC, Canada
| | - Patrick Bermudez
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Alison Ross
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Claude Lepage
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Robert D Vincent
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - S Jeon
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Lindsay B Lewis
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - S Das
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Alex P Zijdenbos
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Pierre Rioux
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | - Reza Adalat
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| | | | - Alan C Evans
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, Montreal, QC, Canada
| |
Collapse
|