201
|
Marastoni D, Crescenzo F, Pisani AI, Zuco C, Schiavi G, Benedetti G, Ricciardi GK, Montemezzi S, Pizzini FB, Tamanti A, Calabrese M. Two years' effect of dimethyl fumarate on focal and diffuse gray matter pathology in multiple sclerosis. Mult Scler 2022; 28:2090-2098. [PMID: 35765211 DOI: 10.1177/13524585221104014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Data on the effect of dimethyl fumarate (DMF) on focal and diffuse gray matter (GM) damage, a relevant pathological substrate of multiple sclerosis (MS)-related disability are lacking. OBJECTIVE To evaluate the DMF effect on cortical lesions (CLs) accumulation and global and regional GM atrophy in subjects with relapsing-remitting MS. METHODS A total of 148 patients (mean age 38.1 ± 9.7 years) treated with DMF ended a 2-year longitudinal study. All underwent regular Expanded Disability Status Scale (EDSS assessment), and at least two 3T-magnetic resonance imaging (MRI) at 3 and 24 months after DMF initiation. CLs and changes in global and regional atrophy of several brain regions were compared with 47 untreated age and sex-matched patients. RESULTS DMF-treated patients showed lower CLs accumulation (median 0[0-3] vs 2[0-7], p < 0.001) with respect to controls. Global cortical thickness (p < 0.001) and regional thickness and volume were lower in treated group (cerebellum, hippocampus, caudate, and putamen: p < 0.001; thalamus p = 0.03). Lower relapse rate (14% vs 40%, p < 0.001), EDSS change (0.2 ± 0.4 vs 0.4 ± 0.9, p < 0.001), and new WM lesions (median 0[0-5] vs 2[0-6], p < 0.001) were reported. No severe adverse drug reactions occurred. CONCLUSIONS Beyond the well-known effect on disease activity, these results provide evidence of the effect of DMF through reduced progression of focal and diffuse GM damage.
Collapse
Affiliation(s)
- Damiano Marastoni
- Regional Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | | | - Anna I Pisani
- Regional Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Carmela Zuco
- Neurology Unit, "Carlo Poma" Hospital, ASST Mantua, Mantua, Italy
| | - Gianmarco Schiavi
- Regional Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Giulia Benedetti
- Regional Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Giuseppe K Ricciardi
- Neuroradiology & Radiology Units, Department of Diagnostic and Pathology, Integrated University Hospital of Verona, Verona, Italy
| | - Stefania Montemezzi
- Neuroradiology & Radiology Units, Department of Diagnostic and Pathology, Integrated University Hospital of Verona, Verona, Italy
| | - Francesca B Pizzini
- Radiology Unit, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Agnese Tamanti
- Regional Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimiliano Calabrese
- Regional Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
202
|
Katsumi Y, Wong B, Cavallari M, Fong TG, Alsop DC, Andreano JM, Carvalho N, Brickhouse M, Jones R, Libermann TA, Marcantonio ER, Schmitt E, Shafi MM, Pascual-Leone A, Travison T, Barrett LF, Inouye SK, Dickerson BC, Touroutoglou A. Structural integrity of the anterior mid-cingulate cortex contributes to resilience to delirium in SuperAging. Brain Commun 2022; 4:fcac163. [PMID: 35822100 PMCID: PMC9272062 DOI: 10.1093/braincomms/fcac163] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/24/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Despite its devastating clinical and societal impact, approaches to treat delirium in older adults remain elusive, making it important to identify factors that may confer resilience to this syndrome. Here, we investigated a cohort of 93 cognitively normal older patients undergoing elective surgery recruited as part of the Successful Aging after Elective Surgery study. Each participant was classified either as a SuperAger (n = 19) or typically aging older adult (n = 74) based on neuropsychological criteria, where the former was defined as those older adults whose memory function rivals that of young adults. We compared these subgroups to examine the role of preoperative memory function in the incidence and severity of postoperative delirium. We additionally investigated the association between indices of postoperative delirium symptoms and cortical thickness in functional networks implicated in SuperAging based on structural magnetic resonance imaging data that were collected preoperatively. We found that SuperAging confers the real-world benefit of resilience to delirium, as shown by lower (i.e. zero) incidence of postoperative delirium and decreased severity scores compared with typical older adults. Furthermore, greater baseline cortical thickness of the anterior mid-cingulate cortex-a key node of the brain's salience network that is also consistently implicated in SuperAging-predicted lower postoperative delirium severity scores in all patients. Taken together, these findings suggest that baseline memory function in older adults may be a useful predictor of postoperative delirium risk and severity and that superior memory function may contribute to resilience to delirium. In particular, the integrity of the anterior mid-cingulate cortex may be a potential biomarker of resilience to delirium, pointing to this region as a potential target for preventive or therapeutic interventions designed to mitigate the risk or consequences of developing this prevalent clinical syndrome.
Collapse
Affiliation(s)
- Yuta Katsumi
- Harvard Medical School, Boston MA, USA
- Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston MA, USA
| | - Bonnie Wong
- Harvard Medical School, Boston MA, USA
- Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston MA, USA
| | - Michele Cavallari
- Harvard Medical School, Boston MA, USA
- Center for Neurologlical Imaging, Department of Radiology, Brigham and Women’s Hospital, Boston MA, USA
| | - Tamara G Fong
- Harvard Medical School, Boston MA, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston MA, USA
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston MA, USA
| | - David C Alsop
- Harvard Medical School, Boston MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston MA, USA
| | - Joseph M Andreano
- Harvard Medical School, Boston MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston MA, USA
| | - Nicole Carvalho
- Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston MA, USA
| | - Michael Brickhouse
- Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston MA, USA
| | - Richard Jones
- Department of Psychiatry and Human Behavior and Neurology, Brown University Warren Alpert Medical School, Providence RI, USA
| | - Towia A Libermann
- Harvard Medical School, Boston MA, USA
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston MA, USA
| | - Edward R Marcantonio
- Harvard Medical School, Boston MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston MA, USA
| | - Eva Schmitt
- Harvard Medical School, Boston MA, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston MA, USA
| | - Mouhsin M Shafi
- Harvard Medical School, Boston MA, USA
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston MA, USA
- Berenson-Allen Center for Non-Invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston MA, USA
| | - Alvaro Pascual-Leone
- Harvard Medical School, Boston MA, USA
- Berenson-Allen Center for Non-Invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Boston MA, USA
| | - Thomas Travison
- Harvard Medical School, Boston MA, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston MA, USA
| | - Lisa Feldman Barrett
- Harvard Medical School, Boston MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston MA, USA
- Department of Psychology, Northeastern University, Boston MA, USA
| | - Sharon K Inouye
- Harvard Medical School, Boston MA, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston MA, USA
| | - Bradford C Dickerson
- Harvard Medical School, Boston MA, USA
- Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston MA, USA
- Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Boston MA, USA
| | - Alexandra Touroutoglou
- Harvard Medical School, Boston MA, USA
- Frontotemporal Disorders Unit, Massachusetts General Hospital, Boston MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston MA, USA
| |
Collapse
|
203
|
Moran C, Xu ZY, Mehta H, Gillies M, Karayiannis C, Beare R, Chen C, Srikanth V. Neuroimaging and cognitive correlates of retinal Optical Coherence Tomography (OCT) measures at late middle age in a twin sample. Sci Rep 2022; 12:9562. [PMID: 35688899 PMCID: PMC9187769 DOI: 10.1038/s41598-022-13662-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/26/2022] [Indexed: 11/09/2022] Open
Abstract
Sharing in embryology and function between the eye and brain has led to interest in whether assessments of the eye reflect brain changes seen in neurodegeneration. We aimed to examine the associations between measures of retinal layer thickness using optical coherence tomography (OCT) and multimodal measures of brain structure and function. Using a convenient sample of twins discordant for type 2 diabetes, we performed cognitive testing, structural brain MRI (tissue volumetry), diffusion tensor imaging (white matter microstructure), and arterial spin labelling (cerebral blood flow). OCT images were recorded and retinal thickness maps generated. We used mixed level modelling to examine the relationship between retinal layer thicknesses and brain measures. We enrolled 35 people (18 pairs, mean age 63.8 years, 63% female). Ganglion cell layer thickness was positively associated with memory, speed, gray matter volume, and altered mean diffusivity. Ganglion cell layer thickness was strongly positively associated with regional cerebral blood flow. We found only a limited number of associations between other retinal layer thickness and measures of brain structure or function. Ganglion cell layer thickness showed consistent associations with a range of brain measures suggesting it may have utility as a marker for future dementia risk.
Collapse
Affiliation(s)
- Chris Moran
- National Centre for Healthy Ageing, Melbourne, Australia.,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia.,Department of Aged Care, Alfred Health, Melbourne, Australia
| | - Zheng Yang Xu
- Royal Free London NHS Foundation Trust, London, UK.,UCL Medical School, London, UK
| | - Hemal Mehta
- Royal Free London NHS Foundation Trust, London, UK.,Macular Research Group, University of Sydney, Sydney, Australia
| | - Mark Gillies
- Macular Research Group, University of Sydney, Sydney, Australia
| | - Chris Karayiannis
- National Centre for Healthy Ageing, Melbourne, Australia.,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia
| | - Richard Beare
- National Centre for Healthy Ageing, Melbourne, Australia.,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia
| | - Christine Chen
- Department of Ophthalmology, Monash Health, Melbourne, Australia
| | - Velandai Srikanth
- National Centre for Healthy Ageing, Melbourne, Australia. .,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia.
| |
Collapse
|
204
|
Backhausen LL, Herting MM, Tamnes CK, Vetter NC. Best Practices in Structural Neuroimaging of Neurodevelopmental Disorders. Neuropsychol Rev 2022; 32:400-418. [PMID: 33893904 PMCID: PMC9090677 DOI: 10.1007/s11065-021-09496-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 03/02/2021] [Indexed: 11/25/2022]
Abstract
Structural magnetic resonance imaging (sMRI) offers immense potential for increasing our understanding of how anatomical brain development relates to clinical symptoms and functioning in neurodevelopmental disorders. Clinical developmental sMRI may help identify neurobiological risk factors or markers that may ultimately assist in diagnosis and treatment. However, researchers and clinicians aiming to conduct sMRI studies of neurodevelopmental disorders face several methodological challenges. This review offers hands-on guidelines for clinical developmental sMRI. First, we present brain morphometry metrics and review evidence on typical developmental trajectories throughout adolescence, together with atypical trajectories in selected neurodevelopmental disorders. Next, we discuss challenges and good scientific practices in study design, image acquisition and analysis, and recent options to implement quality control. Finally, we discuss choices related to statistical analysis and interpretation of results. We call for greater completeness and transparency in the reporting of methods to advance understanding of structural brain alterations in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lea L. Backhausen
- Department of Child and Adolescent Psychiatry, Faculty of Medicine of the Technische Universitaet Dresden, Dresden, Germany
| | - Megan M. Herting
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Christian K. Tamnes
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Nora C. Vetter
- Department of Child and Adolescent Psychiatry, Faculty of Medicine of the Technische Universitaet Dresden, Dresden, Germany
| |
Collapse
|
205
|
Kang Y, Kang W, Han KM, Tae WS, Ham BJ. Associations between cognitive impairment and cortical thickness alterations in patients with euthymic and depressive bipolar disorder. Psychiatry Res Neuroimaging 2022; 322:111462. [PMID: 35231679 DOI: 10.1016/j.pscychresns.2022.111462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 10/19/2022]
Affiliation(s)
- Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Wooyoung Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo-Suk Tae
- Korea University, Brain Convergence Research Center
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
206
|
Schiffer F, Khan A, Ohashi K, Hernandez Garcia LC, Anderson CM, Nickerson LD, Teicher MH. Individual Differences in Hemispheric Emotional Valence by Computerized Test Correlate with Lateralized Differences in Nucleus Accumbens, Hippocampal and Amygdala Volumes. Psychol Res Behav Manag 2022; 15:1371-1384. [PMID: 35673325 PMCID: PMC9167593 DOI: 10.2147/prbm.s357138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/17/2022] [Indexed: 12/31/2022] Open
Abstract
Purpose Conventional theories of hemispheric emotional valence (HEV) postulate fixed hemispheric differences in emotional processing. Schiffer’s dual brain psychology proposes that there are prominent individual differences with a substantial subset showing a reversed laterality pattern. He further proposed that hemispheric differences were more akin to differences in personality than in emotional processing. This theory is supported by findings that unilateral treatments, such as transcranial magnetic stimulation, are effective if they accurately target individual differences in laterality. The aim of this paper was to assess if a computer test of hemispheric emotional valence (CTHEV) could effectively identify individual differences in HEV and to ascertain if these individual differences were associated with underlying differences in brain structure and connectivity. Patients and Methods The CTHEV was administered to 50 (18 male/32 female) right-handed participants, aged 18–19 years, enrolled in a study assessing the neurobiological effects of childhood maltreatment. Based on a literature review, we determined whether CTHEV correlated with lateralized volumes of the nucleus accumbens, amygdala, hippocampus, and subgenual anterior cingulate as well as volume of the corpus callosum. Results CTHEV scores correlated with laterality indices of the nucleus accumbens (p = 0.00016), amygdala (p = 0.0138) and hippocampus (p = 0.031). A positive left hemispheric valence was associated with a larger left-sided nucleus accumbens and hippocampus and a smaller left amygdala. We identified four eigenvector network centrality DTI measures that predict CTHEV, most notably the left amygdala, and found that CTHEV results correlated with total and segment-specific corpus callosal volumes. Conclusion Individual differences in HEV can be readily assessed by computer test and correlate with differences in brain structure and connectivity that could provide a mechanistic understanding. These findings provide further support for a revised understanding of HEV and provide a tool that could be used to guide lateralized brain treatments.
Collapse
Affiliation(s)
- Fredric Schiffer
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
- Correspondence: Fredric Schiffer, Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA, Tel +1 617 855 2970, Fax +1 617 855 3712, Email
| | - Alaptagin Khan
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Kyoko Ohashi
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Laura C Hernandez Garcia
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Carl M Anderson
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Lisa D Nickerson
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Imaging Center, McLean Hospital, Belmont, MA, USA
| | - Martin H Teicher
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
207
|
Ly MT, Scarneo-Miller SE, Lepley AS, Coleman K, Hirschhorn R, Yeargin S, Casa DJ, Chen CM. Combining MRI and cognitive evaluation to classify concussion in university athletes. Brain Imaging Behav 2022; 16:2175-2187. [PMID: 35639240 DOI: 10.1007/s11682-022-00687-w] [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] [Accepted: 05/09/2022] [Indexed: 11/26/2022]
Abstract
Current methods of concussion assessment lack the objectivity and reliability to detect neurological injury. This multi-site study uses combinations of neuroimaging (diffusion tensor imaging and resting state functional MRI) and cognitive measures to train algorithms to detect the presence of concussion in university athletes. Athletes (29 concussed, 48 controls) completed symptom reports, brief cognitive evaluation, and MRI within 72 h of injury. Hierarchical linear regression compared groups on cognitive and neuroimaging measures while controlling for sex and data collection site. Logistic regression and support vector machine models were trained using cognitive and neuroimaging measures and evaluated for overall accuracy, sensitivity, and specificity. Concussed athletes reported greater symptoms than controls (∆R2 = 0.32, p < .001), and performed worse on tests of concentration (∆R2 = 0.07, p < .05) and delayed memory (∆R2 = 0.17, p < .001). Concussed athletes showed lower functional connectivity within the frontoparietal and primary visual networks (p < .05), but did not differ on mean diffusivity and fractional anisotropy. Of the cognitive measures, classifiers trained using delayed memory yielded the best performance with overall accuracy of 71%, though sensitivity was poor at 46%. Of the neuroimaging measures, classifiers trained using mean diffusivity yielded similar accuracy. Combining cognitive measures with mean diffusivity increased overall accuracy to 74% and sensitivity to 64%, comparable to the sensitivity of symptom report. Trained algorithms incorporating both MRI and cognitive performance variables can reliably detect common neurobiological sequelae of acute concussion. The integration of multi-modal data can serve as an objective, reliable tool in the assessment and diagnosis of concussion.
Collapse
Affiliation(s)
- Monica T Ly
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA.
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.
- Department of Psychiatry, University of California San Diego, School of Medicine, San Diego, CA, USA.
| | - Samantha E Scarneo-Miller
- Department of Kinesiology, Korey Stringer Institute, University of Connecticut, Storrs, CT, USA
- Division of Athletic Training, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Adam S Lepley
- Department of Kinesiology, Korey Stringer Institute, University of Connecticut, Storrs, CT, USA
- School of Kinesiology, Exercise and Sport Science Initiative, University of Michigan, Ann Arbor, MI, USA
| | - Kelly Coleman
- Department of Kinesiology, Korey Stringer Institute, University of Connecticut, Storrs, CT, USA
- Department of Health & Movement Sciences, Southern Connecticut State University, New Haven, CT, USA
| | - Rebecca Hirschhorn
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
- School of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| | - Susan Yeargin
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Douglas J Casa
- Department of Kinesiology, Korey Stringer Institute, University of Connecticut, Storrs, CT, USA
| | - Chi-Ming Chen
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
208
|
Loehrer PA, Weber I, Oehrn CR, Nettersheim FS, Dafsari HS, Knake S, Tittgemeyer M, Timmermann L, Belke M. Microstructural alterations predict impaired bimanual control in Parkinson’s disease. Brain Commun 2022; 4:fcac137. [PMID: 35702729 PMCID: PMC9185383 DOI: 10.1093/braincomms/fcac137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/25/2022] [Accepted: 05/20/2022] [Indexed: 11/25/2022] Open
Abstract
Bimanual coordination is impaired in Parkinson’s disease affecting patients’ ability to perform activities of daily living and to maintain independence. Conveyance of information between cortical and subcortical areas is essential for bimanual coordination and relies on the integrity of cerebral microstructure. As pathological deposition of alpha-synuclein compromises microstructure in Parkinson’s disease, we investigated the relationship between microstructural integrity and bimanual coordination using diffusion-weighted MRI in 23 patients with Parkinson’s disease (mean age ± standard deviation: 56.0 ± 6.45 years; 8 female) and 26 older adults (mean age ± standard deviation: 58.5 ± 5.52 years). Whole-brain analysis revealed specific microstructural alterations between patients and healthy controls matched for age, sex, handedness, and cognitive status congruent with the literature and known Parkinson’s disease pathology. A general linear model revealed distinct microstructural alterations associated with poor bimanual coordination in Parkinson’s disease, corrected for multiple comparisons using a permutation-based approach. Integrating known functional topography, we conclude that distinct changes in microstructure cause an impediment of structures involved in attention, working memory, executive function, motor planning, motor control, and visual processing contributing to impaired bimanual coordination in Parkinson’s disease.
Collapse
Affiliation(s)
- Philipp A. Loehrer
- Correspondence to: Philipp A. Loehrer Department of Neurology Philipps-University Marburg, Baldinger Str 35043 Marburg, Germany E-mail:
| | - Immo Weber
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps-University Marburg, Marburg, Germany
| | - Carina R. Oehrn
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps-University Marburg, Marburg, Germany
- Department of Cardiology, University Hospital Cologne, Cologne, Germany
| | | | - Haidar S. Dafsari
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Susanne Knake
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps-University Marburg, Marburg, Germany
- Center for Personalized Translational Epilepsy Research (CePTER) Consortium, Frankfurt am Main, Germany
| | - Marc Tittgemeyer
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Lars Timmermann
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps-University Marburg, Marburg, Germany
| | - Marcus Belke
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
- Center for Personalized Translational Epilepsy Research (CePTER) Consortium, Frankfurt am Main, Germany
| |
Collapse
|
209
|
Han KM, Choi KW, Kim A, Kang W, Kang Y, Tae WS, Han MR, Ham BJ. Association of DNA Methylation of the NLRP3 Gene with Changes in Cortical Thickness in Major Depressive Disorder. Int J Mol Sci 2022; 23:ijms23105768. [PMID: 35628578 PMCID: PMC9143533 DOI: 10.3390/ijms23105768] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
The Nod-like receptor pyrin containing 3 (NLRP3) inflammasome has been reported to be a convergent point linking the peripheral immune response induced by psychological stress and neuroinflammatory processes in the brain. We aimed to identify differences in the methylation profiles of the NLRP3 gene between major depressive disorder (MDD) patients and healthy controls (HCs). We also investigated the correlation of the methylation score of loci in NLRP3 with cortical thickness in the MDD group using magnetic resonance imaging (MRI) data. A total of 220 patients with MDD and 82 HCs were included in the study, and genome-wide DNA methylation profiling of the NLRP3 gene was performed. Among the total sample, 88 patients with MDD and 74 HCs underwent T1-weighted structural MRI and were included in the neuroimaging–methylation analysis. We identified five significant differentially methylated positions (DMPs) in NLRP3. In the MDD group, the methylation scores of cg18793688 and cg09418290 showed significant positive or negative correlations with cortical thickness in the occipital, parietal, temporal, and frontal regions, which showed significant differences in cortical thickness between the MDD and HC groups. Our findings suggest that NLRP3 DNA methylation may predispose to depression-related brain structural changes by increasing NLRP3 inflammasome-related neuroinflammatory processes in MDD.
Collapse
Affiliation(s)
- Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (K.-M.H.); (K.W.C.)
| | - Kwan Woo Choi
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (K.-M.H.); (K.W.C.)
| | - Aram Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea; (A.K.); (W.K.); (Y.K.)
| | - Wooyoung Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea; (A.K.); (W.K.); (Y.K.)
| | - Youbin Kang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea; (A.K.); (W.K.); (Y.K.)
| | - Woo-Suk Tae
- Brain Convergence Research Center, Korea University College of Medicine, Seoul 02841, Korea;
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
- Correspondence: (M.-R.H.); (B.-J.H.)
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (K.-M.H.); (K.W.C.)
- Correspondence: (M.-R.H.); (B.-J.H.)
| |
Collapse
|
210
|
SCN1A overexpression, associated with a genomic region marked by a risk variant for a common epilepsy, raises seizure susceptibility. Acta Neuropathol 2022; 144:107-127. [PMID: 35551471 PMCID: PMC9217876 DOI: 10.1007/s00401-022-02429-0] [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: 03/16/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/01/2022]
Abstract
Mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures is associated with common variation at rs7587026, located in the promoter region of SCN1A. We sought to explore possible underlying mechanisms. SCN1A expression was analysed in hippocampal biopsy specimens of individuals with mesial temporal lobe epilepsy with hippocampal sclerosis who underwent surgical treatment, and hippocampal neuronal cell loss was quantitatively assessed using immunohistochemistry. In healthy individuals, hippocampal volume was measured using MRI. Analyses were performed stratified by rs7587026 type. To study the functional consequences of increased SCN1A expression, we generated, using transposon-mediated bacterial artificial chromosome transgenesis, a zebrafish line expressing exogenous scn1a, and performed EEG analysis on larval optic tecta at 4 day post-fertilization. Finally, we used an in vitro promoter analysis to study whether the genetic motif containing rs7587026 influences promoter activity. Hippocampal SCN1A expression differed by rs7587026 genotype (Kruskal-Wallis test P = 0.004). Individuals homozygous for the minor allele showed significantly increased expression compared to those homozygous for the major allele (Dunn's test P = 0.003), and to heterozygotes (Dunn's test P = 0.035). No statistically significant differences in hippocampal neuronal cell loss were observed between the three genotypes. Among 597 healthy participants, individuals homozygous for the minor allele at rs7587026 displayed significantly reduced mean hippocampal volume compared to major allele homozygotes (Cohen's D = - 0.28, P = 0.02), and to heterozygotes (Cohen's D = - 0.36, P = 0.009). Compared to wild type, scn1lab-overexpressing zebrafish larvae exhibited more frequent spontaneous seizures [one-way ANOVA F(4,54) = 6.95 (P < 0.001)]. The number of EEG discharges correlated with the level of scn1lab overexpression [one-way ANOVA F(4,15) = 10.75 (P < 0.001]. Finally, we showed that a 50 bp promoter motif containing rs7587026 exerts a strong regulatory role on SCN1A expression, though we could not directly link this to rs7587026 itself. Our results develop the mechanistic link between rs7587026 and mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures. Furthermore, we propose that quantitative precision may be important when increasing SCN1A expression in current strategies aiming to treat seizures in conditions involving SCN1A haploinsufficiency, such as Dravet syndrome.
Collapse
|
211
|
Dimick MK, Kennedy KG, Mitchell RHB, Sinyor M, MacIntosh BJ, Goldstein BI. Neurostructural differences associated with self-harm in youth bipolar disorder. Bipolar Disord 2022; 24:275-285. [PMID: 34596314 DOI: 10.1111/bdi.13137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/19/2021] [Accepted: 09/25/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Youth with bipolar disorder (BD) are at greatly elevated risk for suicide. Self-harm, encompassing all self-injurious behaviors regardless of suicidal intent, is among one of the greatest risk factors for death by suicide. This study aims to extend the sparse literature regarding the neurostructural correlates of self-harm in youth with BD. METHODS Participants included 156 youth (17.14 ± 1.61 years): 38 BD with lifetime history of self-harm (BDSH+ ), 43 BD without history of self-harm (BDSH- ), and 75 healthy controls (HC). Measures of cortical thickness, surface area (SA), and volume were obtained using 3 T magnetic resonance imaging. Orbitofrontal and ventrolateral prefrontal cortices were examined in region-of-interest (ROI) analyses, complemented by exploratory vertex-wise analyses using a general linear model controlling for age, sex, and intracranial volume. RESULTS In ROI analyses, there were no between-group differences after correction for multiple comparisons. Vertex-wise analysis revealed three significant clusters in precentral gyrus SA, inferior temporal gyrus SA, and caudal middle frontal gyrus volume. Post-hoc vertex-wise analyses showed BDSH+ had lower cortical SA and volume compared with both BDSH- and HC for all clusters. CONCLUSIONS Significant vertex-wise findings were observed in frontotemporal regions relevant to BD and self-harm, with smaller neurostructural measures among BDSH+ compared with both BDSH- and HC. Future studies are needed to evaluate the temporal nature of the relationship of these neurostructural differences (i.e., potential risk indicators) to self-harm and to identify mechanisms underlying these findings.
Collapse
Affiliation(s)
- Mikaela K Dimick
- Centre for Youth Bipolar Disorder, Child and Youth Psychiatry Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada
| | - Kody G Kennedy
- Centre for Youth Bipolar Disorder, Child and Youth Psychiatry Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada
| | - Rachel H B Mitchell
- Department of Psychiatry, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada.,Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Mark Sinyor
- Department of Psychiatry, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada.,Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Bradley J MacIntosh
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Hurvitz Brain Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Child and Youth Psychiatry Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Pharmacology and Toxicology, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada.,Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
212
|
Zheng G, Zhang Y, Zhao Z, Wang Y, Liu X, Shang Y, Cong Z, Dimitriadis S, Yao Z, Hu B. A Transformer-based Multi-features Fusion Model for Prediction of Conversion in Mild Cognitive Impairment. Methods 2022; 204:241-248. [PMID: 35487442 DOI: 10.1016/j.ymeth.2022.04.015] [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] [Received: 01/31/2022] [Revised: 03/26/2022] [Accepted: 04/24/2022] [Indexed: 12/21/2022] Open
Abstract
Mild cognitive impairment (MCI) is usually considered the early stage of Alzheimer's disease (AD). Therefore, the accurate identification of MCI individuals with high risk in converting to AD is essential for the potential prevention and treatment of AD. Recently, the great success of deep learning has sparked interest in applying deep learning to neuroimaging field. However, deep learning techniques are prone to overfitting since available neuroimaging datasets are not sufficiently large. Therefore, we proposed a deep learning model fusing cortical features to address the issue of fusion and classification blocks. To validate the effectiveness of the proposed model, we compared seven different models on the same dataset in the literature. The results show that our proposed model outperformed the competing models in the prediction of MCI conversion with an accuracy of 83.3% in the testing dataset. Subsequently, we used deep learning to characterize the contribution of brain regions and different cortical features to MCI progression. The results revealed that the caudal anterior cingulate and pars orbitalis contributed most to the classification task, and our model pays more attention to volume features and cortical thickness features.
Collapse
Affiliation(s)
- Guowei Zheng
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China
| | - Yu Zhang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China
| | - Ziyang Zhao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China
| | - Yin Wang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China
| | - Xia Liu
- School of Computer Science, Qinghai Normal University, Xining, China
| | - Yingying Shang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China
| | - Zhaoyang Cong
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China
| | - Stavros Dimitriadis
- Integrative Neuroimaging Lab, 55133, Thessaloniki (Makedonia), Greece; Neuroinformatics Group, Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, -College of Biomedical and Life Sciences, Cardiff, Wales, United Kingdom; 1st Department of Neurology, G.H. "AHEPA " School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (AUTH), Thessaloniki, Greece; Greek Association of Alzheimer's Disease and Related Disorders, Thessaloniki, Makedonia, Greece; Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, United Kingdom; Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, United Kingdom; School of Psychology, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, United Kingdom; Neuroscience and Mental Health Research Institute, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Zhijun Yao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China.
| | - Bin Hu
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Joint Research Center for Cognitive Neurosensor Technology of Lanzhou University & Institute of Semiconductors, Chinese Academy of Sciences, Lanzhou, China; Engineering Research Center of Open Source Software and Real-Time System (Lanzhou University), Ministry of Education, Lanzhou, China
| |
Collapse
|
213
|
Kim K, Joo YY, Ahn G, Wang HH, Moon SY, Kim H, Ahn WY, Cha J. The sexual brain, genes, and cognition: A machine-predicted brain sex score explains individual differences in cognitive intelligence and genetic influence in young children. Hum Brain Mapp 2022; 43:3857-3872. [PMID: 35471639 PMCID: PMC9294341 DOI: 10.1002/hbm.25888] [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: 10/04/2021] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/06/2022] Open
Abstract
Sex impacts the development of the brain and cognition differently across individuals. However, the literature on brain sex dimorphism in humans is mixed. We aim to investigate the biological underpinnings of the individual variability of sexual dimorphism in the brain and its impact on cognitive performance. To this end, we tested whether the individual difference in brain sex would be linked to that in cognitive performance that is influenced by genetic factors in prepubertal children (N = 9,658, ages 9-10 years old; the Adolescent Brain Cognitive Development study). To capture the interindividual variability of the brain, we estimated the probability of being male or female based on the brain morphometry and connectivity features using machine learning (herein called a brain sex score). The models accurately classified the biological sex with a test ROC-AUC of 93.32%. As a result, a greater brain sex score correlated significantly with greater intelligence (pfdr < .001, η p 2 $$ {\eta}_p^2 $$ = .011-.034; adjusted for covariates) and higher cognitive genome-wide polygenic scores (GPSs) (pfdr < .001, η p 2 $$ {\eta}_p^2 $$ < .005). Structural equation models revealed that the GPS-intelligence association was significantly modulated by the brain sex score, such that a brain with a higher maleness score (or a lower femaleness score) mediated a positive GPS effect on intelligence (indirect effects = .006-.009; p = .002-.022; sex-stratified analysis). The finding of the sex modulatory effect on the gene-brain-cognition relationship presents a likely biological pathway to the individual and sex differences in the brain and cognitive performance in preadolescence.
Collapse
Affiliation(s)
- Kakyeong Kim
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | | | - Gun Ahn
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul, South Korea
| | - Hee-Hwan Wang
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Seo-Yoon Moon
- College of Liberal Studies, Seoul National University, Seoul, South Korea
| | - Hyeonjin Kim
- Department of Psychology, College of Social Sciences, Seoul National University, Seoul, South Korea
| | - Woo-Young Ahn
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Psychology, College of Social Sciences, Seoul National University, Seoul, South Korea.,AI Institute, Seoul National University, Seoul, South Korea
| | - Jiook Cha
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea.,Department of Psychology, College of Social Sciences, Seoul National University, Seoul, South Korea.,AI Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
214
|
Chen J, Tam A, Kebets V, Orban C, Ooi LQR, Asplund CL, Marek S, Dosenbach NUF, Eickhoff SB, Bzdok D, Holmes AJ, Yeo BTT. Shared and unique brain network features predict cognitive, personality, and mental health scores in the ABCD study. Nat Commun 2022; 13:2217. [PMID: 35468875 PMCID: PMC9038754 DOI: 10.1038/s41467-022-29766-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 03/18/2022] [Indexed: 12/30/2022] Open
Abstract
How individual differences in brain network organization track behavioral variability is a fundamental question in systems neuroscience. Recent work suggests that resting-state and task-state functional connectivity can predict specific traits at the individual level. However, most studies focus on single behavioral traits, thus not capturing broader relationships across behaviors. In a large sample of 1858 typically developing children from the Adolescent Brain Cognitive Development (ABCD) study, we show that predictive network features are distinct across the domains of cognitive performance, personality scores and mental health assessments. On the other hand, traits within each behavioral domain are predicted by similar network features. Predictive network features and models generalize to other behavioral measures within the same behavioral domain. Although tasks are known to modulate the functional connectome, predictive network features are similar between resting and task states. Overall, our findings reveal shared brain network features that account for individual variation within broad domains of behavior in childhood.
Collapse
Affiliation(s)
- Jianzhong Chen
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore.,Centre for Sleep and Cognition, National University of Singapore, Singapore, Singapore.,Centre for Translational MR Research, National University of Singapore, Singapore, Singapore.,N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Angela Tam
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore.,Centre for Sleep and Cognition, National University of Singapore, Singapore, Singapore.,Centre for Translational MR Research, National University of Singapore, Singapore, Singapore.,N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Valeria Kebets
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore.,Centre for Sleep and Cognition, National University of Singapore, Singapore, Singapore.,Centre for Translational MR Research, National University of Singapore, Singapore, Singapore.,N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Csaba Orban
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore.,Centre for Sleep and Cognition, National University of Singapore, Singapore, Singapore.,Centre for Translational MR Research, National University of Singapore, Singapore, Singapore.,N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore
| | - Leon Qi Rong Ooi
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore.,Centre for Sleep and Cognition, National University of Singapore, Singapore, Singapore.,Centre for Translational MR Research, National University of Singapore, Singapore, Singapore.,N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore.,Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore
| | - Christopher L Asplund
- Centre for Sleep and Cognition, National University of Singapore, Singapore, Singapore.,Centre for Translational MR Research, National University of Singapore, Singapore, Singapore.,N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore.,Division of Social Sciences, Yale-NUS College, Singapore, Singapore.,Department of Psychology, National University of Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Scott Marek
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Nico U F Dosenbach
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Simon B Eickhoff
- Institute for Systems Neuroscience, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.,Institute of Neuroscience and Medicine, Brain & Behaviours (INM-7), Research Center Jülich, Jülich, Germany
| | - Danilo Bzdok
- Department of Biomedical Engineering, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Mila - Quebec AI Institute, Montreal, QC, Canada
| | - Avram J Holmes
- Yale University, Departments of Psychology and Psychiatry, New Haven, CT, USA
| | - B T Thomas Yeo
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore, Singapore. .,Centre for Sleep and Cognition, National University of Singapore, Singapore, Singapore. .,Centre for Translational MR Research, National University of Singapore, Singapore, Singapore. .,N.1 Institute for Health & Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore. .,Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore. .,Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
215
|
Demro C, Shen C, Hendrickson TJ, Arend JL, Disner SG, Sponheim SR. Advanced Brain-Age in Psychotic Psychopathology: Evidence for Transdiagnostic Neurodevelopmental Origins. Front Aging Neurosci 2022; 14:872867. [PMID: 35527740 PMCID: PMC9074783 DOI: 10.3389/fnagi.2022.872867] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is characterized by abnormal brain structure such as global reductions in gray matter volume. Machine learning models trained to estimate the age of brains from structural neuroimaging data consistently show advanced brain-age to be associated with schizophrenia. Yet, it is unclear whether advanced brain-age is specific to schizophrenia compared to other psychotic disorders, and whether evidence that brain structure is "older" than chronological age actually reflects neurodevelopmental rather than atrophic processes. It is also unknown whether advanced brain-age is associated with genetic liability for psychosis carried by biological relatives of people with schizophrenia. We used the Brain-Age Regression Analysis and Computation Utility Software (BARACUS) prediction model and calculated the residualized brain-age gap of 332 adults (163 individuals with psychotic disorders: 105 schizophrenia, 17 schizoaffective disorder, 41 bipolar I disorder with psychotic features; 103 first-degree biological relatives; 66 controls). The model estimated advanced brain-ages for people with psychosis in comparison to controls and relatives, with no differences among psychotic disorders or between relatives and controls. Specifically, the model revealed an enlarged brain-age gap for schizophrenia and bipolar disorder with psychotic features. Advanced brain-age was associated with lower cognitive and general functioning in the full sample. Among relatives, cognitive performance and schizotypal symptoms were related to brain-age gap, suggesting that advanced brain-age is associated with the subtle expressions associated with psychosis. Exploratory longitudinal analyses suggested that brain aging was not accelerated in individuals with a psychotic disorder. In sum, we found that people with psychotic disorders, irrespective of specific diagnosis or illness severity, show indications of non-progressive, advanced brain-age. These findings support a transdiagnostic, neurodevelopmental formulation of structural brain abnormalities in psychotic psychopathology.
Collapse
Affiliation(s)
- Caroline Demro
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
| | - Chen Shen
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
| | | | - Jessica L. Arend
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
| | - Seth G. Disner
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| | - Scott R. Sponheim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
- Department of Psychology, University of Minnesota, Minneapolis, MN, United States
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, United States
| |
Collapse
|
216
|
Brown EM, Salat DH, Milberg WP, Fortier CB, McGlinchey RE. Accelerated longitudinal cortical atrophy in
OEF
/
OIF
/
OND
veterans with severe
PTSD
and the impact of comorbid
TBI. Hum Brain Mapp 2022; 43:3694-3705. [PMID: 35426972 PMCID: PMC9294300 DOI: 10.1002/hbm.25877] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/04/2022] [Accepted: 04/05/2022] [Indexed: 12/02/2022] Open
Abstract
Veterans who deployed in support of Operation Enduring Freedom (OEF), Iraqi Freedom (OIF), and New Dawn (OND) commonly experience severe psychological trauma, often accompanied by physical brain trauma resulting in mild traumatic brain injury (mTBI). Prior studies of individuals with posttraumatic stress disorder (PTSD) have revealed alterations in brain structure, accelerated cellular aging, and impacts on cognition following exposure to severe psychological trauma and potential interactive effects of military‐related mTBI. To date, however, little is known how such deployment‐related trauma changes with time and age of injury of the affected veteran. In this study, we explored changes in cortical thickness, volume, and surface area after an average interval of approximately 2 years in a cohort of 254 OEF/OIF/OND Veterans ranging in age from 19 to 67 years. Whole‐brain vertex‐wise analyses revealed that veterans who met criteria for severe PTSD (Clinician‐Administered PTSD Scale ≥60) at baseline showed greater negative longitudinal changes in cortical thickness, volume, and area over time. Analyses also revealed a significant severe‐PTSD by age interaction on cortical measures with severe‐PTSD individuals exhibiting accelerated cortical degeneration with increasing age. Interaction effects of comorbid military‐related mTBI within the severe‐PTSD group were also observed in several cortical regions. These results suggest that those exhibiting severe PTSD symptomatology have accelerated atrophy that is exacerbated with increasing age and history of mTBI.
Collapse
Affiliation(s)
- Emma M. Brown
- Neuroimaging Research for Veterans (NeRVe) Center VA Boston Healthcare System Boston Massachusetts USA
- Translational Research Center for TBI and Stress Disorders (TRACTS) VA Boston Healthcare System Boston Massachusetts USA
| | - David H. Salat
- Neuroimaging Research for Veterans (NeRVe) Center VA Boston Healthcare System Boston Massachusetts USA
- Translational Research Center for TBI and Stress Disorders (TRACTS) VA Boston Healthcare System Boston Massachusetts USA
- Brain Aging and Dementia (BAnD) Laboratory, A. A. Martinos Center for Biomedical Imaging, Department of Radiology Massachusetts General Hospital Charlestown Massachusetts USA
| | - William P. Milberg
- Neuroimaging Research for Veterans (NeRVe) Center VA Boston Healthcare System Boston Massachusetts USA
- Translational Research Center for TBI and Stress Disorders (TRACTS) VA Boston Healthcare System Boston Massachusetts USA
- Department of Psychiatry Harvard Medical School Boston Massachusetts USA
- Geriatric Research, Education, and Clinical Center (GRECC) VA Boston Healthcare System Boston Massachusetts USA
| | - Catherine B. Fortier
- Translational Research Center for TBI and Stress Disorders (TRACTS) VA Boston Healthcare System Boston Massachusetts USA
- Department of Psychiatry Harvard Medical School Boston Massachusetts USA
- Geriatric Research, Education, and Clinical Center (GRECC) VA Boston Healthcare System Boston Massachusetts USA
| | - Regina E. McGlinchey
- Neuroimaging Research for Veterans (NeRVe) Center VA Boston Healthcare System Boston Massachusetts USA
- Translational Research Center for TBI and Stress Disorders (TRACTS) VA Boston Healthcare System Boston Massachusetts USA
- Department of Psychiatry Harvard Medical School Boston Massachusetts USA
- Geriatric Research, Education, and Clinical Center (GRECC) VA Boston Healthcare System Boston Massachusetts USA
| |
Collapse
|
217
|
Petrican R, Fornito A, Jones N. Psychological Resilience and Neurodegenerative Risk: A Connectomics-Transcriptomics Investigation in Healthy Adolescent and Middle-Aged Females. Neuroimage 2022; 255:119209. [PMID: 35429627 DOI: 10.1016/j.neuroimage.2022.119209] [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: 09/14/2021] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
Adverse life events can inflict substantial long-term damage, which, paradoxically, has been posited to stem from initially adaptative responses to the challenges encountered in one's environment. Thus, identification of the mechanisms linking resilience against recent stressors to longer-term psychological vulnerability is key to understanding optimal functioning across multiple timescales. To address this issue, our study tested the relevance of neuro-reproductive maturation and senescence, respectively, to both resilience and longer-term risk for pathologies characterised by accelerated brain aging, specifically, Alzheimer's Disease (AD). Graph theoretical and partial least squares analyses were conducted on multimodal imaging, reported biological aging and recent adverse experience data from the Lifespan Human Connectome Project (HCP). Availability of reproductive maturation/senescence measures restricted our investigation to adolescent (N =178) and middle-aged (N=146) females. Psychological resilience was linked to age-specific brain senescence patterns suggestive of precocious functional development of somatomotor and control-relevant networks (adolescence) and earlier aging of default mode and salience/ventral attention systems (middle adulthood). Biological aging showed complementary associations with the neural patterns relevant to resilience in adolescence (positive relationship) versus middle-age (negative relationship). Transcriptomic and expression quantitative trait locus data analyses linked the neural aging patterns correlated with psychological resilience in middle adulthood to gene expression patterns suggestive of increased AD risk. Our results imply a partially antagonistic relationship between resilience against proximal stressors and longer-term psychological adjustment in later life. They thus underscore the importance of fine-tuning extant views on successful coping by considering the multiple timescales across which age-specific processes may unfold.
Collapse
Affiliation(s)
- Raluca Petrican
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, United Kingdom.
| | - Alex Fornito
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| | - Natalie Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, United Kingdom
| |
Collapse
|
218
|
Ramirez-Garcia G, Galvez V, Diaz R, Campos-Romo A, Fernandez-Ruiz J. Montreal Cognitive Assessment (MoCA) performance in Huntington's disease patients correlates with cortical and caudate atrophy. PeerJ 2022; 10:e12917. [PMID: 35402100 PMCID: PMC8988933 DOI: 10.7717/peerj.12917] [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: 09/03/2021] [Accepted: 01/20/2022] [Indexed: 01/11/2023] Open
Abstract
Huntington's Disease (HD) is an autosomal neurodegenerative disease characterized by motor, cognitive, and psychiatric symptoms. Cognitive impairment develops gradually in HD patients, progressing later into a severe cognitive dysfunction. The Montreal Cognitive Assessment (MoCA) is a brief screening test commonly employed to detect mild cognitive impairment, which has also been useful to assess cognitive decline in HD patients. However, the relationship between MoCA performance and brain structural integrity in HD patients remains unclear. Therefore, to explore this relationship we analyzed if cortical thinning and subcortical nuclei volume differences correlated with HD patients' MoCA performance. Twenty-two HD patients and twenty-two healthy subjects participated in this study. T1-weighted images were acquired to analyze cortical thickness and subcortical nuclei volumes. Group comparison analysis showed a significantly lower score in the MoCA global performance of HD patients. Also, the MoCA total score correlated with cortical thinning of fronto-parietal and temporo-occipital cortices, as well as with bilateral caudate volume differences in HD patients. These results provide new insights into the effectiveness of using the MoCA test to detect cognitive impairment and the brain atrophy pattern associated with the cognitive status of prodromal/early HD patients.
Collapse
Affiliation(s)
- Gabriel Ramirez-Garcia
- Departamento de Fisiología, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | - Victor Galvez
- Escuela de Psicología, Universidad Panamericana, Ciudad de Mexico, Mexico
| | - Rosalinda Diaz
- Departamento de Fisiología, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | - Aurelio Campos-Romo
- Facultad de Medicina, Unidad Periférica de Neurociencias, Universidad Nacional Autónoma de México/Instituto Nacional de Neurologia y Neurocirugia, Ciudad de Mexico, Mexico
| | - Juan Fernandez-Ruiz
- Departamento de Fisiología, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| |
Collapse
|
219
|
Langan MT, Smith DA, Verma G, Khegai O, Saju S, Rashid S, Ranti D, Markowitz M, Belani P, Jette N, Mathew B, Goldstein J, Kirsch CFE, Morris LS, Becker JH, Delman BN, Balchandani P. Semi-automated Segmentation and Quantification of Perivascular Spaces at 7 Tesla in COVID-19. Front Neurol 2022; 13:846957. [PMID: 35432151 PMCID: PMC9010775 DOI: 10.3389/fneur.2022.846957] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/10/2022] [Indexed: 01/12/2023] Open
Abstract
While COVID-19 is primarily considered a respiratory disease, it has been shown to affect the central nervous system. Mounting evidence shows that COVID-19 is associated with neurological complications as well as effects thought to be related to neuroinflammatory processes. Due to the novelty of COVID-19, there is a need to better understand the possible long-term effects it may have on patients, particularly linkage to neuroinflammatory processes. Perivascular spaces (PVS) are small fluid-filled spaces in the brain that appear on MRI scans near blood vessels and are believed to play a role in modulation of the immune response, leukocyte trafficking, and glymphatic drainage. Some studies have suggested that increased number or presence of PVS could be considered a marker of increased blood-brain barrier permeability or dysfunction and may be involved in or precede cascades leading to neuroinflammatory processes. Due to their size, PVS are better detected on MRI at ultrahigh magnetic field strengths such as 7 Tesla, with improved sensitivity and resolution to quantify both concentration and size. As such, the objective of this prospective study was to leverage a semi-automated detection tool to identify and quantify differences in perivascular spaces between a group of 10 COVID-19 patients and a similar subset of controls to determine whether PVS might be biomarkers of COVID-19-mediated neuroinflammation. Results demonstrate a detectable difference in neuroinflammatory measures in the patient group compared to controls. PVS count and white matter volume were significantly different in the patient group compared to controls, yet there was no significant association between PVS count and symptom measures. Our findings suggest that the PVS count may be a viable marker for neuroinflammation in COVID-19, and other diseases which may be linked to neuroinflammatory processes.
Collapse
Affiliation(s)
- Mackenzie T. Langan
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
- *Correspondence: Mackenzie T. Langan
| | - Derek A. Smith
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
| | - Gaurav Verma
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
| | - Oleksandr Khegai
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
| | - Sera Saju
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
| | - Shams Rashid
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
| | - Daniel Ranti
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
| | - Matthew Markowitz
- The Graduate Center, City University of New York, New York, NY, United States
| | - Puneet Belani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nathalie Jette
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Brian Mathew
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jonathan Goldstein
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Claudia F. E. Kirsch
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
- Department of Radiology, Zucker Hofstra School of Medicine at Northwell Health, Uniondale, NY, United States
| | - Laurel S. Morris
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
- Department of Psychiatry at the Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jacqueline H. Becker
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bradley N. Delman
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Priti Balchandani
- Biomedical Engineering and Imaging Institute at Mount Sinai School of Medicine, New York, NY, United States
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
220
|
McKay NS, Dincer A, Mehrotra V, Aschenbrenner AJ, Balota D, Hornbeck RC, Hassenstab J, Morris JC, Benzinger TLS, Gordon BA. Beta-amyloid moderates the relationship between cortical thickness and attentional control in middle- and older-aged adults. Neurobiol Aging 2022; 112:181-190. [PMID: 35227946 PMCID: PMC9208719 DOI: 10.1016/j.neurobiolaging.2021.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/13/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022]
Abstract
Although often unmeasured in studies of cognition, many older adults possess Alzheimer disease (AD) pathologies such as beta-amyloid (Aβ) deposition, despite being asymptomatic. We were interested in examining whether the behavior-structure relationship observed in later life was altered by the presence of preclinical AD pathology. A total of 511 cognitively unimpaired adults completed magnetic resonance imaging and three attentional control tasks; a subset (n = 396) also underwent Aβ-positron emissions tomography. A vertex-wise model was conducted to spatially represent the relationship between cortical thickness and average attentional control accuracy, while moderation analysis examined whether Aβ deposition impacted this relationship. First, we found that reduced cortical thickness in temporal, medial- and lateral-parietal, and dorsolateral prefrontal cortex, predicted worse performance on the attention task composite. Subsequent moderation analyses observed that levels of Aβ significantly influence the relationship between cortical thickness and attentional control. Our results support the hypothesis that preclinical AD, as measured by Aβ deposition, is partially driving what would otherwise be considered general aging in a cognitively normal adult population.
Collapse
Affiliation(s)
- Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO.
| | - Aylin Dincer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO
| | | | - Andrew J Aschenbrenner
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Neurology, Washington School of Medicine, St. Louis, MO
| | - David Balota
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Psychological and Brain Sciences, Washington University in St. Louis, MO
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO
| | - Jason Hassenstab
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Neurology, Washington School of Medicine, St. Louis, MO; Department of Psychological and Brain Sciences, Washington University in St. Louis, MO
| | - John C Morris
- Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Neurology, Washington School of Medicine, St. Louis, MO
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, MO; Department of Psychological and Brain Sciences, Washington University in St. Louis, MO
| |
Collapse
|
221
|
Radiomics-Based Artificial Intelligence Differentiation of Neurodegenerative Diseases with Reference to the Volumetry. Life (Basel) 2022; 12:life12040514. [PMID: 35455005 PMCID: PMC9024778 DOI: 10.3390/life12040514] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 01/06/2023] Open
Abstract
This study aimed to build automated detection models—one by brain regional volume (V-model), and the other by radiomics features of the whole brain (R-model)—to differentiate mild cognitive impairment (MCI) from cognitive normal (CN), and Alzheimer’s Disease (AD) from mild cognitive impairment (MCI). The objectives are to compare the models and identify whether radiomics or volumetry can provide a better prediction for differentiating different types of dementia. Method: 582 MRI T1-weighted images were retrieved from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database, which is a multicenter operating open source database for AD. In total, 97 images of AD, 293 images of MCI patient and 192 images of cognitive normal were divided into a training, a validation and a test group at a ratio of 70:15:15. For each T1-weighted image, volumetric segmentation was performed with the image analysis software FreeSurfer, and radiomics features were retrieved by imaging research software 3D slicers. Brain regional volume and radiomics features were used to build the V-model and R-model, respectively, using the random forest algorithm by R. The receiver operating characteristics (ROC) curve of both models were used to evaluate their diagnostic accuracy and reliability to differentiate AD, MCI and CN. Results: To differentiate MCI and CN, both V-model and R-model achieved excellent performance, with an AUC of 0.9992 ± 0.0022 and 0.9850 ± 0.0032, respectively. No significant difference was found between the two AUCs, indicating both models attained similar good performance. In MCI and AD differentiation, the V-model and R-model yielded AUC of 0.9986 ± 0.0013 and 0.9714 ± 0.0175, respectively. The best performance was to differentiate AD from CN, where the V-model and R-model yielded AUC of 0.9994 ± 0.0019 and 0.9830 ± 0.009, respectively. The results suggested that both volumetry and radiomics approaches could be used in differentiating AD, MCI and CN, based on T1 weighted MR images using random forest algorithm successfully. Conclusion: This study showed that the radiomics features from T1-weighted MR images achieved excellence performance in differentiating AD, MCI and CN. Compared to the volumetry method, the accuracy, sensitivity and specificity are slightly lower in using radiomics, but still attained very good and reliable classification of the three stages of neurodegenerations. In view of the convenience and operator independence in feature extraction, radiomics can be a quantitative biomarker to differentiate the disease groups.
Collapse
|
222
|
Rus-Oswald OG, Benner J, Reinhardt J, Bürki C, Christiner M, Hofmann E, Schneider P, Stippich C, Kressig RW, Blatow M. Musicianship-Related Structural and Functional Cortical Features Are Preserved in Elderly Musicians. Front Aging Neurosci 2022; 14:807971. [PMID: 35401149 PMCID: PMC8990841 DOI: 10.3389/fnagi.2022.807971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Professional musicians are a model population for exploring basic auditory function, sensorimotor and multisensory integration, and training-induced neuroplasticity. The brain of musicians exhibits distinct structural and functional cortical features; however, little is known about how these features evolve during aging. This multiparametric study aimed to examine the functional and structural neural correlates of lifelong musical practice in elderly professional musicians. Methods Sixteen young musicians, 16 elderly musicians (age >70), and 15 elderly non-musicians participated in the study. We assessed gray matter metrics at the whole-brain and region of interest (ROI) levels using high-resolution magnetic resonance imaging (MRI) with the Freesurfer automatic segmentation and reconstruction pipeline. We used BrainVoyager semiautomated segmentation to explore individual auditory cortex morphotypes. Furthermore, we evaluated functional blood oxygenation level-dependent (BOLD) activations in auditory and non-auditory regions by functional MRI (fMRI) with an attentive tone-listening task. Finally, we performed discriminant function analyses based on structural and functional ROIs. Results A general reduction of gray matter metrics distinguished the elderly from the young subjects at the whole-brain level, corresponding to widespread natural brain atrophy. Age- and musicianship-dependent structural correlations revealed group-specific differences in several clusters including superior, middle, and inferior frontal as well as perirolandic areas. In addition, the elderly musicians exhibited increased gyrification of auditory cortex like the young musicians. During fMRI, the elderly non-musicians activated predominantly auditory regions, whereas the elderly musicians co-activated a much broader network of auditory association areas, primary and secondary motor areas, and prefrontal and parietal regions like, albeit weaker, the young musicians. Also, group-specific age- and musicianship-dependent functional correlations were observed in the frontal and parietal regions. Moreover, discriminant function analysis could separate groups with high accuracy based on a set of specific structural and functional, mainly temporal and occipital, ROIs. Conclusion In conclusion, despite naturally occurring senescence, the elderly musicians maintained musicianship-specific structural and functional cortical features. The identified structural and functional brain regions, discriminating elderly musicians from non-musicians, might be of relevance for the aging musicians’ brain. To what extent lifelong musical activity may have a neuroprotective impact needs to be addressed further in larger longitudinal studies.
Collapse
Affiliation(s)
- Oana G. Rus-Oswald
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland
- *Correspondence: Oana G. Rus-Oswald,
| | - Jan Benner
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
- Jan Benner,
| | - Julia Reinhardt
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zürich, Switzerland
- Division of Diagnostic and Interventional Neuroradiology, Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Orthopedic Surgery and Traumatology, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Céline Bürki
- University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland
| | - Markus Christiner
- Centre for Systematic Musicology, University of Graz, Graz, Austria
- Vitols Jazeps Latvian Academy of Music, Riga, Latvia
| | - Elke Hofmann
- Academy of Music, University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Basel, Switzerland
| | - Peter Schneider
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
- Centre for Systematic Musicology, University of Graz, Graz, Austria
- Vitols Jazeps Latvian Academy of Music, Riga, Latvia
| | - Christoph Stippich
- Department of Neuroradiology and Radiology, Kliniken Schmieder, Allensbach, Germany
| | - Reto W. Kressig
- University Department of Geriatric Medicine FELIX PLATTER, Basel, Switzerland
| | - Maria Blatow
- Section of Neuroradiology, Department of Radiology and Nuclear Medicine, Neurocenter, Cantonal Hospital Lucerne, University of Lucerne, Lucerne, Switzerland
| |
Collapse
|
223
|
Ozzoude M, Varriano B, Beaton D, Ramirez J, Holmes MF, Scott CJM, Gao F, Sunderland KM, McLaughlin P, Rabin J, Goubran M, Kwan D, Roberts A, Bartha R, Symons S, Tan B, Swartz RH, Abrahao A, Saposnik G, Masellis M, Lang AE, Marras C, Zinman L, Shoesmith C, Borrie M, Fischer CE, Frank A, Freedman M, Montero-Odasso M, Kumar S, Pasternak S, Strother SC, Pollock BG, Rajji TK, Seitz D, Tang-Wai DF, Turnbull J, Dowlatshahi D, Hassan A, Casaubon L, Mandzia J, Sahlas D, Breen DP, Grimes D, Jog M, Steeves TDL, Arnott SR, Black SE, Finger E, Tartaglia MC. Investigating the contribution of white matter hyperintensities and cortical thickness to empathy in neurodegenerative and cerebrovascular diseases. GeroScience 2022; 44:1575-1598. [PMID: 35294697 DOI: 10.1007/s11357-022-00539-x] [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] [Received: 08/17/2021] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Change in empathy is an increasingly recognised symptom of neurodegenerative diseases and contributes to caregiver burden and patient distress. Empathy impairment has been associated with brain atrophy but its relationship to white matter hyperintensities (WMH) is unknown. We aimed to investigate the relationships amongst WMH, brain atrophy, and empathy deficits in neurodegenerative and cerebrovascular diseases. Five hundred thirteen participants with Alzheimer's disease/mild cognitive impairment, amyotrophic lateral sclerosis, frontotemporal dementia (FTD), Parkinson's disease, or cerebrovascular disease (CVD) were included. Empathy was assessed using the Interpersonal Reactivity Index. WMH were measured using a semi-automatic segmentation and FreeSurfer was used to measure cortical thickness. A heterogeneous pattern of cortical thinning was found between groups, with FTD showing thinning in frontotemporal regions and CVD in left superior parietal, left insula, and left postcentral. Results from both univariate and multivariate analyses revealed that several variables were associated with empathy, particularly cortical thickness in the fronto-insulo-temporal and cingulate regions, sex (female), global cognition, and right parietal and occipital WMH. Our results suggest that cortical atrophy and WMH may be associated with empathy deficits in neurodegenerative and cerebrovascular diseases. Future work should consider investigating the longitudinal effects of WMH and atrophy on empathy deficits in neurodegenerative and cerebrovascular diseases.
Collapse
Affiliation(s)
- Miracle Ozzoude
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th floor 6KD-407, Toronto, ON, M5T 0S8, Canada.,L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Brenda Varriano
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th floor 6KD-407, Toronto, ON, M5T 0S8, Canada
| | - Derek Beaton
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada
| | - Joel Ramirez
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Melissa F Holmes
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Christopher J M Scott
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Fuqiang Gao
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | - Paula McLaughlin
- Nova Scotia Health and Dalhousie University, Halifax, NS, Canada
| | - Jennifer Rabin
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Maged Goubran
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Donna Kwan
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.,Queen's University, Kingston, ON, Canada
| | - Angela Roberts
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA.,School of Communication Sciences and Disorders, Faculty of Health Sciences, Western University, London, ON, Canada
| | - Robert Bartha
- Robarts Research Institute, Western University, London, ON, Canada
| | - Sean Symons
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Brian Tan
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada
| | - Richard H Swartz
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Agessandro Abrahao
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Mario Masellis
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Anthony E Lang
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Edmond J Safra Program for Parkinson Disease, Movement Disorder Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Connie Marras
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Edmond J Safra Program for Parkinson Disease, Movement Disorder Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Lorne Zinman
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Christen Shoesmith
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada
| | - Michael Borrie
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,St. Joseph's Healthcare Centre, London, ON, Canada
| | - Corinne E Fischer
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Andrew Frank
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Bruyère Research Institute, Ottawa, ON, Canada
| | - Morris Freedman
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Baycrest Health Sciences, Toronto, ON, Canada
| | - Manuel Montero-Odasso
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Lawson Health Research Institute, London, ON, Canada.,Gait and Brain Lab, Parkwood Institute, London, ON, Canada
| | - Sanjeev Kumar
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Stephen Pasternak
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada
| | - Stephen C Strother
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Bruce G Pollock
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tarek K Rajji
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada
| | - Dallas Seitz
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - David F Tang-Wai
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Memory Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - John Turnbull
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Dar Dowlatshahi
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Ayman Hassan
- Thunder Bay Regional Health Research Institute, Thunder Bay, ON, Canada
| | - Leanne Casaubon
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jennifer Mandzia
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada.,Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Demetrios Sahlas
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.,Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - David P Breen
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK.,Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - David Grimes
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mandar Jog
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada.,Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,London Health Sciences Centre, London, ON, Canada
| | - Thomas D L Steeves
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Stephen R Arnott
- Rotman Research Institute of Baycrest Centre, Toronto, ON, Canada
| | - Sandra E Black
- L.C. Campbell Cognitive Neurology Unit, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Toronto Dementia Research Alliance, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, Western University, London, ON, Canada.,Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | | | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, 6th floor 6KD-407, Toronto, ON, M5T 0S8, Canada. .,Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada. .,Memory Clinic, Toronto Western Hospital, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
224
|
Muret D, Root V, Kieliba P, Clode D, Makin TR. Beyond body maps: Information content of specific body parts is distributed across the somatosensory homunculus. Cell Rep 2022; 38:110523. [PMID: 35294887 PMCID: PMC8938902 DOI: 10.1016/j.celrep.2022.110523] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/10/2021] [Accepted: 02/21/2022] [Indexed: 11/23/2022] Open
Abstract
The homunculus in primary somatosensory cortex (S1) is famous for its body part selectivity, but this dominant feature may eclipse other representational features, e.g., information content, also relevant for S1 organization. Using multivariate fMRI analysis, we ask whether body part information content can be identified in S1 beyond its primary region. Throughout S1, we identify significant representational dissimilarities between body parts but also subparts in distant non-primary regions (e.g., between the hand and the lips in the foot region and between different face parts in the foot region). Two movements performed by one body part (e.g., the hand) could also be dissociated well beyond its primary region (e.g., in the foot and face regions), even within Brodmann area 3b. Our results demonstrate that information content is more distributed across S1 than selectivity maps suggest. This finding reveals underlying information contents in S1 that could be harnessed for rehabilitation and brain-machine interfaces.
Collapse
Affiliation(s)
- Dollyane Muret
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London WC1N 3AZ, UK.
| | - Victoria Root
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London WC1N 3AZ, UK; Wellcome Centre of Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| | - Paulina Kieliba
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London WC1N 3AZ, UK
| | - Danielle Clode
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London WC1N 3AZ, UK; Dani Clode Design, 40 Hillside Road, London SW2 3HW, UK
| | - Tamar R Makin
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London WC1N 3AZ, UK; Wellcome Trust Centre for Neuroimaging, University College London, London WC1N 3AR, UK
| |
Collapse
|
225
|
Owens-Walton C, Adamson C, Walterfang M, Hall S, van Westen D, Hansson O, Shaw M, Looi JCL. Midsagittal corpus callosal thickness and cognitive impairment in Parkinson's disease. Eur J Neurosci 2022; 55:1859-1872. [PMID: 35274408 PMCID: PMC9314988 DOI: 10.1111/ejn.15640] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 11/27/2022]
Abstract
People diagnosed with Parkinson's disease (PD) can experience significant neuropsychiatric symptoms, including cognitive impairment and dementia, the neuroanatomical substrates of which are not fully characterised. Symptoms associated with cognitive impairment and dementia in PD may relate to direct structural changes to the corpus callosum via primary white matter pathology, or as a secondary outcome due to the degeneration of cortical regions. Using magnetic resonance imaging, the corpus callosum can be investigated at the midsagittal plane, where it converges to a contiguous mass and is not intertwined with other tracts. The objective of this project was thus twofold; first, we investigated possible changes in the thickness of the midsagittal callosum and cortex in patients with PD with varying levels of cognitive impairment; and secondly, we investigated the relationship between the thickness of the midsagittal corpus callosum and the thickness of the cortex. Study participants included cognitively unimpaired PD participants (n = 35), PD participants with mild cognitive impairment (n = 22), PD participants with dementia (n = 17) and healthy controls (n = 27). We found thinning of the callosum in PD-related dementia compared to PD-related mild cognitive impairment and cognitively unimpaired PD participants. Regression analyses found thickness of the left medial orbitofrontal cortex to be positively correlated with thickness of the anterior callosum in PD-related mild cognitive impairment. This study suggests that a midsagittal thickness model can uncover changes to the corpus callosum in PD-related dementia, which occur in line with changes to the cortex in this advanced disease stage.
Collapse
Affiliation(s)
- Conor Owens-Walton
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, School of Clinical Medicine, Medical School, Australian National University, Canberra, Australia.,Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Neuroinformatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, United States of America
| | - Chris Adamson
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Mark Walterfang
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne Neuropsychiatry Centre, University of Melbourne and Melbourne Health, Melbourne, Australia.,Florey Institute of Neurosciences and Mental Health, University of Melbourne, Melbourne, Australia
| | - Sara Hall
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.,Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Danielle van Westen
- Centre for Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden.,Diagnostic Radiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Oskar Hansson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.,Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Marnie Shaw
- College of Engineering and Computer Science, The Australian National University, Canberra, Australia
| | - Jeffrey C L Looi
- Research Centre for the Neurosciences of Ageing, Academic Unit of Psychiatry and Addiction Medicine, School of Clinical Medicine, Medical School, Australian National University, Canberra, Australia
| |
Collapse
|
226
|
Nivins S, Kennedy E, Thompson B, Gamble GD, Alsweiler JM, Metcalfe R, McKinlay CJD, Harding JE. Associations between neonatal hypoglycaemia and brain volumes, cortical thickness and white matter microstructure in mid-childhood: An MRI study. Neuroimage Clin 2022; 33:102943. [PMID: 35063925 PMCID: PMC8856905 DOI: 10.1016/j.nicl.2022.102943] [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: 10/04/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/11/2022]
Abstract
Neonatal hypoglycaemia is associated with damage to the brain in the acute phase. In mid-childhood, neonatal hypoglycaemia is associated with smaller brain regions. Deep grey matter regions such as the caudate and thalamus are implicated. Children with neonatal hypoglycemia had smaller occipital lobe cortical thickness. Grey matter may be especially vulnerable to long-term effects of neonatal hypoglycemia.
Neonatal hypoglycaemia is a common metabolic disorder that may cause brain damage, most visible in parieto-occipital regions on MRI in the acute phase. However, the long term effects of neonatal hypoglycaemia on the brain are not well understood. We investigated the association between neonatal hypoglycaemia and brain volumes, cortical thickness and white matter microstructure at 9–10 years. Children born at risk of neonatal hypoglycaemia at ≥ 36 weeks’ gestation who took part in a prospective cohort study underwent brain MRI at 9–10 years. Neonatal hypoglycaemia was defined as at least one hypoglycaemic episode (at least one consecutive blood glucose concentration < 2.6 mmol/L) or interstitial episode (at least 10 min of interstitial glucose concentrations < 2.6 mmol/L). Brain volumes and cortical thickness were computed using Freesurfer. White matter microstructure was assessed using tract-based spatial statistics. Children who had (n = 75) and had not (n = 26) experienced neonatal hypoglycaemia had similar combined parietal and occipital lobe volumes and no differences in white matter microstructure at nine years of age. However, those who had experienced neonatal hypoglycaemia had smaller caudate volumes (mean difference: −557 mm3, 95% confidence interval (CI), −933 to −182, p = 0.004) and smaller thalamus (−0.03%, 95%CI, −0.06 to 0.00; p = 0.05) and subcortical grey matter (−0.10%, 95%CI −0.20 to 0.00, p = 0.05) volumes as percentage of total brain volume, and thinner occipital lobe cortex (−0.05 mm, 95%CI −0.10 to 0.00, p = 0.05) than those who had not. The finding of smaller caudate volumes after neonatal hypoglycaemia was consistent across analyses of pre-specified severity groups, clinically detected hypoglycaemic episodes, and severity and frequency of hypoglycaemic events. Neonatal hypoglycaemia is associated with smaller deep grey matter brain regions and thinner occipital lobe cortex but not altered white matter microstructure in mid-childhood.
Collapse
Affiliation(s)
- Samson Nivins
- Liggins Institute, University of Auckland, New Zealand
| | | | - Benjamin Thompson
- Liggins Institute, University of Auckland, New Zealand; School of Optometry and Vision Science, University of Waterloo, Waterloo, Ontario, Canada; Centre for Eye and Vision Research, 17W Science Park, Hong Kong
| | | | - Jane M Alsweiler
- Auckland District Health Board, Auckland, New Zealand; Department of Paediatrics: Child and Youth Health, University of Auckland, New Zealand
| | | | - Christopher J D McKinlay
- Liggins Institute, University of Auckland, New Zealand; Kidz First Neonatal Care, Counties Manukau Health, New Zealand
| | | | | |
Collapse
|
227
|
Chilla GS, Yeow LY, Chew QH, Sim K, Prakash KNB. Machine learning classification of schizophrenia patients and healthy controls using diverse neuroanatomical markers and Ensemble methods. Sci Rep 2022; 12:2755. [PMID: 35177708 PMCID: PMC8854385 DOI: 10.1038/s41598-022-06651-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is a major psychiatric disorder that imposes enormous clinical burden on patients and their caregivers. Determining classification biomarkers can complement clinical measures and improve understanding of the neural basis underlying schizophrenia. Using neuroanatomical features, several machine learning based investigations have attempted to classify schizophrenia from healthy controls but the range of neuroanatomical measures employed have been limited in range to date. In this study, we sought to classify schizophrenia and healthy control cohorts using a diverse set of neuroanatomical measures (cortical and subcortical volumes, cortical areas and thickness, cortical mean curvature) and adopted Ensemble methods for better performance. Additionally, we correlated such neuroanatomical features with Quality of Life (QoL) assessment scores within the schizophrenia cohort. With Ensemble methods and diverse neuroanatomical measures, we achieved classification accuracies ranging from 83 to 87%, sensitivities and specificities varying between 90-98% and 65-70% respectively. In addition to lower QoL scores within schizophrenia cohort, significant correlations were found between specific neuroanatomical measures and psychological health, social relationship subscale domains of QoL. Our results suggest the utility of inclusion of subcortical and cortical measures and Ensemble methods to achieve better classification performance and their potential impact of parsing out neurobiological correlates of quality of life in schizophrenia.
Collapse
Affiliation(s)
- Geetha Soujanya Chilla
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore, 138667.
| | - Ling Yun Yeow
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore, 138667
| | - Qian Hui Chew
- Institute of Mental Health, Singapore, Singapore, 539747
| | - Kang Sim
- Institute of Mental Health, Singapore, Singapore, 539747
| | - K N Bhanu Prakash
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore, 138667.
| |
Collapse
|
228
|
Kennedy KG, Grigorian A, Mitchell RHB, McCrindle BW, MacIntosh BJ, Goldstein BI. Association of blood pressure with brain structure in youth with and without bipolar disorder. J Affect Disord 2022; 299:666-674. [PMID: 34920038 DOI: 10.1016/j.jad.2021.12.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/25/2021] [Accepted: 12/12/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND We previously found that blood pressure (BP) is elevated, and associated with poorer neurocognition, in youth with bipolar disorder (BD). While higher BP is associated with smaller brain structure in adults, studies have not examined this topic in BD or youth. METHODS Participants were 154 youth, ages 13-20 (n = 81 BD, n = 73 HC). Structural magnetic resonance imaging and diastolic (DBP), and systolic (SBP) pressure were obtained. Region of interest (ROI; anterior cingulate cortex [ACC], insular cortex, hippocampus) and vertex-wise analyses controlling for age, sex, body-mass-index, and intracranial volume investigated BP-neurostructural associations; a group-by-BP interaction was also assessed. RESULTS In ROI analyses, higher DBP in the overall sample was associated with smaller insular cortex area (β=-0.18 p = 0.007) and was associated with smaller ACC area to a significantly greater extent in HC vs. BD (β=-0.14 p = 0.015). In vertex-wise analyses, higher DBP and SBP were associated with smaller area and volume in the insular cortex, frontal, parietal, and temporal regions in the overall sample. Additionally, higher SBP was associated with greater thickness in temporal and parietal regions. Finally, higher SBP was associated with smaller area and volume in frontal, parietal, and temporal regions to a significantly greater extent in BD vs. HC. LIMITATIONS Cross-sectional design, single assessment of BP. CONCLUSION BP is associated with brain structure in youth, with variability related to structural phenotype (volume vs. thickness) and psychiatric diagnosis (BD vs. HC). Future studies evaluating temporality of these findings, and the association of BP changes on brain structure in youth, are warranted.
Collapse
Affiliation(s)
- Kody G Kennedy
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Rm 4326, 100 stokes street Way, Toronto, ON M6J 1H4, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Anahit Grigorian
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Rm 4326, 100 stokes street Way, Toronto, ON M6J 1H4, Canada
| | - Rachel H B Mitchell
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Brian W McCrindle
- Division of Pediatric Cardiology, Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Bradley J MacIntosh
- Brain Sciences, Sunnybrook Health Sciences Centre, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Centre for Addiction and Mental Health, Rm 4326, 100 stokes street Way, Toronto, ON M6J 1H4, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada.
| |
Collapse
|
229
|
Grasing M, Sharma P, Lepping RJ, Honea R, Burns JM, Brooks WM, Gupta A. Association between the Estimated Glomerular Filtration Rate and Brain Atrophy in Older Adults. Am J Nephrol 2022; 53:176-181. [PMID: 35130538 PMCID: PMC9060294 DOI: 10.1159/000521892] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/04/2022] [Indexed: 11/19/2022]
Abstract
End-stage kidney disease has been associated with cognitive impairment and brain atrophy. It remains unclear if mild to moderate kidney dysfunction is associated with brain atrophy, especially in older adults. We used cross-sectional data from the Alzheimer's Disease Neuroimaging Initiative (ADNI), an NIH-funded multicenter longitudinal cohort study, to better understand the association between estimated glomerular filtration rate (eGFR) and brain volumes. We included all ADNI participants with both baseline serum creatinine values and MRI brain volume assessments. We used multiple linear regression modeling to assess cross-sectional associations between eGFR and whole-brain gray matter, hippocampus, entorhinal, fusiform, and middle temporal brain volumes. Participants (n = 1,596) were 74 ± 7 years old with a mean eGFR of 69.4 ± 14.8 mL/min/1.73 m2; 53% had mild cognitive impairment, and 19% had dementia. Unadjusted analysis showed an association between lower eGFR and smaller brain volumes. After adjusting for age, sex, and education, there was no association between eGFR brain volumes (p > 0.05 for all). These results remained consistent after subgroup analysis by age stratification and baseline cognitive status. Age was a confounding variable in the unadjusted association between the eGFR and brain volumes. Thus, a mild to moderately reduced eGFR was not associated with brain atrophy in ADNI participants.
Collapse
Affiliation(s)
- Michael Grasing
- The School of medicine, University of Kansas Medical Center, Kansas City, KS
| | - Palash Sharma
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS
| | - Rebecca J Lepping
- Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS
- Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS
| | - Robyn Honea
- Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
| | - Jeffrey M Burns
- Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
- Frontiers Clinical & Translational Science Institute at the University of Kansas, University of Kansas Medical Center, Kansas City, KS
| | - William M Brooks
- Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS
- Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
- Frontiers Clinical & Translational Science Institute at the University of Kansas, University of Kansas Medical Center, Kansas City, KS
| | - Aditi Gupta
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
- Alzheimer’s Disease Center, University of Kansas Medical Center, Kansas City, KS
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
- Division of Nephrology and Hypertension, University of Kansas Medical Center, Kansas City, KS
| | | |
Collapse
|
230
|
Lucibello S, Bertè G, Verdolotti T, Lucignani M, Napolitano A, D’Abronzo R, Cicala MG, Pede E, Chieffo D, Mariotti P, Colosimo C, Mercuri E, Battini R. Cortical Thickness and Clinical Findings in Prescholar Children With Autism Spectrum Disorder. Front Neurosci 2022; 15:776860. [PMID: 35197818 PMCID: PMC8858962 DOI: 10.3389/fnins.2021.776860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
The term autism spectrum disorder (ASD) includes a wide variability of clinical presentation, and this clinical heterogeneity seems to reflect a still unclear multifactorial etiopathogenesis, encompassing different genetic risk factors and susceptibility to environmental factors. Several studies and many theories recognize as mechanisms of autism a disruption of brain development and maturation time course, suggesting the existence of common neurobiological substrates, such as defective synaptic structure and aberrant brain connectivity. Magnetic resonance imaging (MRI) plays an important role in both assessment of region-specific structural changes and quantification of specific alterations in gray or white matter, which could lead to the identification of an MRI biomarker. In this study, we performed measurement of cortical thickness in a selected well-known group of preschool ASD subjects with the aim of finding correlation between cortical metrics and clinical scores to understand the underlying mechanism of symptoms and to support early clinical diagnosis. Our results confirm that recent brain MRI techniques combined with clinical data can provide some useful information in defining the cerebral regions involved in ASD although large sample studies with homogeneous analytical and multisite approaches are needed.
Collapse
Affiliation(s)
- Simona Lucibello
- Pediatric Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanna Bertè
- Dipartimento di Diagnostica per Immagini, Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tommaso Verdolotti
- UOC Radiologia e Neuroradiologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Martina Lucignani
- Medical Physics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Antonio Napolitano
- Medical Physics Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Rosa D’Abronzo
- Dipartimento di Diagnostica per Immagini, Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria G. Cicala
- Pediatric Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elisa Pede
- Pediatric Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daniela Chieffo
- Pediatric Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paolo Mariotti
- Pediatric Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cesare Colosimo
- Dipartimento di Diagnostica per Immagini, Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
- UOC Radiologia e Neuroradiologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberta Battini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa, Italy
- *Correspondence: Roberta Battini,
| |
Collapse
|
231
|
O'Neill A, Dooley N, Healy C, Carey E, Roddy D, Frodl T, O’Hanlon E, Cannon M. Longitudinal grey matter development associated with psychotic experiences in young people. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 3:264-273. [PMID: 37124352 PMCID: PMC10140460 DOI: 10.1016/j.bpsgos.2022.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/21/2022] [Accepted: 02/03/2022] [Indexed: 10/19/2022] Open
Abstract
Background Gray matter abnormalities are observed across the psychosis spectrum. The trajectory of these abnormalities in healthy adolescents reporting subthreshold psychotic experiences (PEs) may provide insight into the neural mechanisms underlying psychotic symptoms. The risk of psychosis and additional psychopathology is even higher among these individuals who also report childhood adversity/DSM-5 diagnoses. Thus, the aims of this longitudinal study were to investigate PE-related volumetric changes in young people, noting any effects of childhood adversity/DSM-5 diagnosis. Methods A total of 211 young people 11 to 13 years of age participated in the initial Adolescent Brain Development study. PE classification was determined by expert consensus at each time point. Participants underwent neuroimaging at 3 time points over 6 years. A total of 76 participants with at least one scan were included in the final sample; 34 who met criteria for PEs at least once across all the time points (PE group) and 42 control subjects. Data from 20 bilateral regions of interest were extracted for linear mixed-effects analyses. Results Right hippocampal volume increased over time in the control group, with no increase in the PE group (p = .00352). DSM-5 diagnosis and childhood adversity were not significantly associated with right hippocampal volume. There was no significant effect of group or interaction in any other region. Conclusions These findings further implicate right hippocampal volumetric abnormalities in the pathophysiology underlying PEs. Furthermore, as suggested by previous studies in those at clinical high risk for psychosis and those with first-episode psychosis, it is possible that these deficits may be a marker for later clinical outcomes.
Collapse
|
232
|
Brady B, Bardouille T. Periodic/Aperiodic Parameterization of Transient Oscillations (PAPTO): Implications for Healthy Ageing. Neuroimage 2022; 251:118974. [DOI: 10.1016/j.neuroimage.2022.118974] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 12/11/2022] Open
|
233
|
Diversity of daily activities is associated with greater hippocampal volume. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2022; 22:75-87. [PMID: 34599488 PMCID: PMC8792192 DOI: 10.3758/s13415-021-00942-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 02/03/2023]
Abstract
Greater engagement in a range of daily activities is associated with better cognitive functioning (Lee et al., Lee et al., 2020). The hippocampus, a subcortical brain structure implicated in learning, memory, spatial navigation and other aspects of cognitive functioning, may be structurally sensitive to exposure to and engagement with novel experiences and environments. The present study tested whether greater activity diversity, defined as the range of common daily activities engaged in and the proportion of time spent in each, is associated with larger hippocampal volume. Greater diversity of activities, as measured using daily diaries across an 8-day period, was related to greater hippocampal volume averaged across the left and right hemispheres, even when adjusting for estimated intracranial volume, total activity time, sociodemographic factors, and self-reported physical health. These findings are broadly consistent with nonhuman animal studies, demonstrating a link between enriched environments and structural changes to the hippocampus. Future longitudinal and experimental work can elucidate causal and directional relationships between diversity of daily activities and hippocampal volume.
Collapse
|
234
|
Zhang W, Yang C, Li Z, Ren J. A Comparison of Three Brain Atlases for Temporal Lobe Epilepsy Prediction. J Med Biol Eng 2022. [DOI: 10.1007/s40846-021-00676-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
235
|
Danilin LK, Spindler M, Sörös P, Bantel C. Heart rate and heart rate variability in patients with chronic inflammatory joint disease: the role of pain duration and the insular cortex. BMC Musculoskelet Disord 2022; 23:75. [PMID: 35062938 PMCID: PMC8783425 DOI: 10.1186/s12891-022-05009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Chronic inflammatory joint diseases (CIJD) have been linked to increased cardiovascular morbidity and mortality. A decisive reason could be a dysregulation of the autonomic nervous system, which is responsible for the control of cardiovascular function. So far, the cause of changes in autonomic nervous system functions remains elusive. In this study, we investigate the role of chronic pain and the insular cortex in autonomic control of cardiac functioning in patients with CIJD.
Methods
We studied the autonomic nervous system through the assessment of heart rate and heart rate variability (HRV) at rest and under cognitive stimulation. Furthermore, we investigated insular cortex volume by performing surface-based brain morphometry with FreeSurfer. For this study, 47 participants were recruited, 22 individual age- and sex-matched pairs for the magnetic resonance imaging analyses and 14 for the HRV analyses. All available patients’ data were used for analysis.
Results
Pain duration was negatively correlated with the resting heart rate in patients with chronic inflammatory joint diseases (n = 20). In a multiple linear regression model including only CIJD patients with heart rate at rest as a dependent variable, we found a significant positive relationship between heart rate at rest and the volume of the left insular cortex and a significant negative relationship between heart rate at rest and the volume of the right insular cortex. However, we found no significant differences in HRV parameters or insular cortex volumes between both groups.
Conclusions
In this study we provide evidence to suggest insular cortex involvement in the process of ANS changes due to chronic pain in CIJD patients.
The study was preregistered with the German Clinical Trials Register (https://www.drks.de; DRKS00012791; date of registration: 28 July 2017).
Collapse
|
236
|
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: 11] [Impact Index Per Article: 3.7] [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
|
237
|
Fitzroy AB, Jones BJ, Kainec KA, Seo J, Spencer RMC. Aging-Related Changes in Cortical Sources of Sleep Oscillatory Neural Activity Following Motor Learning Reflect Contributions of Cortical Thickness and Pre-sleep Functional Activity. Front Aging Neurosci 2022; 13:787654. [PMID: 35087393 PMCID: PMC8786737 DOI: 10.3389/fnagi.2021.787654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/25/2021] [Indexed: 01/01/2023] Open
Abstract
Oscillatory neural activity during sleep, such as that in the delta and sigma bands, is important for motor learning consolidation. This activity is reduced with typical aging, and this reduction may contribute to aging-related declines in motor learning consolidation. Evidence suggests that brain regions involved in motor learning contribute to oscillatory neural activity during subsequent sleep. However, aging-related differences in regional contributions to sleep oscillatory activity following motor learning are unclear. To characterize these differences, we estimated the cortical sources of consolidation-related oscillatory activity using individual anatomical information in young and older adults during non-rapid eye movement sleep after motor learning and analyzed them in light of cortical thickness and pre-sleep functional brain activation. High-density electroencephalogram was recorded from young and older adults during a midday nap, following completion of a functional magnetic resonance imaged serial reaction time task as part of a larger experimental protocol. Sleep delta activity was reduced with age in a left-weighted motor cortical network, including premotor cortex, primary motor cortex, supplementary motor area, and pre-supplementary motor area, as well as non-motor regions in parietal, temporal, occipital, and cingulate cortices. Sleep theta activity was reduced with age in a similar left-weighted motor network, and in non-motor prefrontal and middle cingulate regions. Sleep sigma activity was reduced with age in left primary motor cortex, in a non-motor right-weighted prefrontal-temporal network, and in cingulate regions. Cortical thinning mediated aging-related sigma reductions in lateral orbitofrontal cortex and frontal pole, and partially mediated delta reductions in parahippocampal, fusiform, and lingual gyri. Putamen, caudate, and inferior parietal cortex activation prior to sleep predicted frontal and motor cortical contributions to sleep delta and theta activity in an age-moderated fashion, reflecting negative relationships in young adults and positive or absent relationships in older adults. Overall, these results support the local sleep hypothesis that brain regions active during learning contribute to consolidation-related neural activity during subsequent sleep and demonstrate that sleep oscillatory activity in these regions is reduced with aging.
Collapse
Affiliation(s)
- Ahren B. Fitzroy
- Neuroscience & Behavior Program, University of Massachusetts Amherst, Amherst, MA, United States
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Bethany J. Jones
- Neuroscience & Behavior Program, University of Massachusetts Amherst, Amherst, MA, United States
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Kyle A. Kainec
- Neuroscience & Behavior Program, University of Massachusetts Amherst, Amherst, MA, United States
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Jeehye Seo
- Neuroscience & Behavior Program, University of Massachusetts Amherst, Amherst, MA, United States
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Rebecca M. C. Spencer
- Neuroscience & Behavior Program, University of Massachusetts Amherst, Amherst, MA, United States
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
238
|
Ali MT, ElNakieb Y, Elnakib A, Shalaby A, Mahmoud A, Ghazal M, Yousaf J, Abu Khalifeh H, Casanova M, Barnes G, El-Baz A. The Role of Structure MRI in Diagnosing Autism. Diagnostics (Basel) 2022; 12:165. [PMID: 35054330 PMCID: PMC8774643 DOI: 10.3390/diagnostics12010165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/30/2021] [Accepted: 01/05/2022] [Indexed: 12/30/2022] Open
Abstract
This study proposes a Computer-Aided Diagnostic (CAD) system to diagnose subjects with autism spectrum disorder (ASD). The CAD system identifies morphological anomalies within the brain regions of ASD subjects. Cortical features are scored according to their contribution in diagnosing a subject to be ASD or typically developed (TD) based on a trained machine-learning (ML) model. This approach opens the hope for developing a new CAD system for early personalized diagnosis of ASD. We propose a framework to extract the cerebral cortex from structural MRI as well as identifying the altered areas in the cerebral cortex. This framework consists of the following five main steps: (i) extraction of cerebral cortex from structural MRI; (ii) cortical parcellation to a standard atlas; (iii) identifying ASD associated cortical markers; (iv) adjusting feature values according to sex and age; (v) building tailored neuro-atlases to identify ASD; and (vi) artificial neural networks (NN) are trained to classify ASD. The system is tested on the Autism Brain Imaging Data Exchange (ABIDE I) sites achieving an average balanced accuracy score of 97±2%. This paper demonstrates the ability to develop an objective CAD system using structure MRI and tailored neuro-atlases describing specific developmental patterns of the brain in autism.
Collapse
Affiliation(s)
- Mohamed T. Ali
- Bioengineering Department, University of Louisville, Louisville, KY 40208, USA; (M.T.A.); (Y.E.); (A.E.); (A.S.); (A.M.)
| | - Yaser ElNakieb
- Bioengineering Department, University of Louisville, Louisville, KY 40208, USA; (M.T.A.); (Y.E.); (A.E.); (A.S.); (A.M.)
| | - Ahmed Elnakib
- Bioengineering Department, University of Louisville, Louisville, KY 40208, USA; (M.T.A.); (Y.E.); (A.E.); (A.S.); (A.M.)
| | - Ahmed Shalaby
- Bioengineering Department, University of Louisville, Louisville, KY 40208, USA; (M.T.A.); (Y.E.); (A.E.); (A.S.); (A.M.)
| | - Ali Mahmoud
- Bioengineering Department, University of Louisville, Louisville, KY 40208, USA; (M.T.A.); (Y.E.); (A.E.); (A.S.); (A.M.)
| | - Mohammed Ghazal
- Department of Electrical and Computer Engineering, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates; (M.G.); (J.Y.); (H.A.K.)
| | - Jawad Yousaf
- Department of Electrical and Computer Engineering, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates; (M.G.); (J.Y.); (H.A.K.)
| | - Hadil Abu Khalifeh
- Department of Electrical and Computer Engineering, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates; (M.G.); (J.Y.); (H.A.K.)
| | - Manuel Casanova
- Department of Biomedical Sciences, School of Medicine Greenville, University of South Carolina, Greenville, SC 29425, USA;
| | - Gregory Barnes
- Department of Neurology, Norton Children’s Autism Center, University of Louisville, Louisville, KY 40208, USA;
| | - Ayman El-Baz
- Bioengineering Department, University of Louisville, Louisville, KY 40208, USA; (M.T.A.); (Y.E.); (A.E.); (A.S.); (A.M.)
| |
Collapse
|
239
|
Oltra J, Uribe C, Campabadal A, Inguanzo A, Monté-Rubio GC, Martí MJ, Compta Y, Valldeoriola F, Junque C, Segura B. Sex Differences in Brain and Cognition in de novo Parkinson's Disease. Front Aging Neurosci 2022; 13:791532. [PMID: 35069180 PMCID: PMC8770804 DOI: 10.3389/fnagi.2021.791532] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/13/2021] [Indexed: 12/31/2022] Open
Abstract
Background and Objective: Brain atrophy and cognitive impairment in neurodegenerative diseases are influenced by sex. We aimed to investigate sex differences in brain atrophy and cognition in de novo Parkinson's disease (PD) patients. Methods: Clinical, neuropsychological and T1-weighted MRI data from 205 PD patients (127 males: 78 females) and 69 healthy controls (40 males: 29 females) were obtained from the PPMI dataset. Results: PD males had a greater motor and rapid eye movement sleep behavior disorder symptomatology than PD females. They also showed cortical thinning in postcentral and precentral regions, greater global cortical and subcortical atrophy and smaller volumes in thalamus, caudate, putamen, pallidum, hippocampus, and brainstem, compared with PD females. Healthy controls only showed reduced hippocampal volume in males compared to females. PD males performed worse than PD females in global cognition, immediate verbal recall, and mental processing speed. In both groups males performed worse than females in semantic verbal fluency and delayed verbal recall; as well as females performed worse than males in visuospatial function. Conclusions: Sex effect in brain and cognition is already evident in de novo PD not explained by age per se, being a relevant factor to consider in clinical and translational research in PD.
Collapse
Affiliation(s)
- Javier Oltra
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carme Uribe
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Research Imaging Centre, Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, University of Toronto, Toronto, ON, Canada
| | - Anna Campabadal
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Anna Inguanzo
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gemma C. Monté-Rubio
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Maria J. Martí
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Hospital Clínic de Barcelona, Barcelona, Spain
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Yaroslau Compta
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Hospital Clínic de Barcelona, Barcelona, Spain
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Francesc Valldeoriola
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Hospital Clínic de Barcelona, Barcelona, Spain
- Parkinson's Disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Carme Junque
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Barbara Segura
- Medical Psychology Unit, Department of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
240
|
Lucas JT, Faught AM, Hsu CY, Wilson LJ, Guo Y, Li Y, Khan R, Becksfort JB, LeVine DA, Ismael Y, Darrow K, Moskvin VP, Pirlepesov F, Klimo P, Elijovich L, Indelicato DJ, Boop FA, Merchant TE. Pre- and Post-therapy Risk Factors for Vasculopathy in Pediatric Craniopharyngioma Patients Treated with Surgery and Proton Radiotherapy. Int J Radiat Oncol Biol Phys 2022; 113:152-160. [PMID: 34990778 DOI: 10.1016/j.ijrobp.2021.12.172] [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: 08/12/2021] [Revised: 10/26/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Vasculopathy (VAS) is a significant complication associated with radiotherapy in patients treated for brain tumors. We studied the type, location, severity, timing, and resolution of VAS in children with craniopharyngioma treated with proton radiotherapy (PRT) and evaluated predictors of stenosis (STN) using a novel patient and imaging-based modelling approach. MATERIALS/METHODS Children with craniopharyngioma (N=94) were treated with 54 GyRBE PRT on a clinical trial NCTXXXXXXXX.1 We evaluated VAS type, location, severity, and resolution. VAS events were segmented and related to their location, operative corridor, PRT dose, and vascular territory to facilitate Mixed Effect Logistic Regression Modelling of spatial predictors of STN events. RESULTS Forty-five (47.9%) patients had 111 instances of confirmed VAS (pre-PRT N = 37, 33.3%). The median time to post-PRT VAS was 3.41 years (95% CI 1.86-6.11). Stenosis events were observed post-PRT in 23.4% (N=22) patients. Post-PRT VAS was detected by cerebral angiogram in 9.6% (N=9), severe in 4.3% (N=4), and compensated on perfusion in 2.1% (N=2). Revascularization was required for 5 (5.3%) patients. Post-surgical, pre-PRT VAS, and PRT dose to unperturbed vessels were predictive of STN. The impact of PRT on STN was negligible within the surgical corridor. CONCLUSIONS VAS often precedes PRT and was the strongest predictor of post-PRT STN. The adverse impact of PRT on STN was only apparent in unperturbed vasculature beyond the operative corridor.
Collapse
Affiliation(s)
- John T Lucas
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105.
| | - Austin M Faught
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| | - Chih Yang Hsu
- GlaxoSmithKline, 812 Springdale Drive, Exton, PA 19341
| | - Lydia J Wilson
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| | - Yian Guo
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| | - Yimei Li
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105.
| | - Raja Khan
- Department of Neurology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Jared B Becksfort
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| | - David A LeVine
- University of TN Health Sciences Center, 881 Madison Ave Ste 1020, Memphis, TN 38163
| | - Yousef Ismael
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| | - Kaleb Darrow
- University of TN Health Sciences Center, 881 Madison Ave Ste 1020, Memphis, TN 38163
| | - Vadim P Moskvin
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| | - Fakhriddin Pirlepesov
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| | - Paul Klimo
- Department of Surgery, Semmes Murphy, 6325 Humphreys Blvd, Memphis, TN 38120; Division of Pediatric Neurosurgery and Associate Professor, The University of Tennessee Health Science Center
| | - Lucas Elijovich
- Department of Neurology, University of TN Health Sciences Center, 847 Monroe Avenue, Suite 226, Memphis, TN 38163
| | - Daniel J Indelicato
- Department of Radiation Oncology, University of Florida, Jacksonville, FL 32206
| | - Fredrick A Boop
- Department of Surgery, Semmes Murphy, 6325 Humphreys Blvd, Memphis, TN 38120; Division of Pediatric Neurosurgery and Associate Professor, The University of Tennessee Health Science Center
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, Memphis, TN 38105
| |
Collapse
|
241
|
Interaction of childhood abuse and depressive symptoms on cortical thickness: a general population study. Eur Arch Psychiatry Clin Neurosci 2022; 272:1523-1534. [PMID: 35217912 PMCID: PMC9653317 DOI: 10.1007/s00406-022-01387-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 02/01/2022] [Indexed: 11/29/2022]
Abstract
Childhood abuse was inconsistently related to whole-brain cortical thickness in former studies. However, both childhood abuse and cortical thickness have been associated with depressive symptoms. We hypothesised that childhood abuse moderates the association between depressive symptoms and cortical thickness. In 1551 individuals of the general population, associations between whole-brain cortical thickness and the interaction of childhood abuse (emotional, physical, and sexual) and depressive symptoms were analysed using an ANCOVA. Linear regression analyses were used to estimate the same effect on the cortical thickness of 34 separate regions (Desikan-Killiany-atlas). A significant interaction effect of childhood abuse and depressive symptoms was observed for whole-brain cortical thickness (F(2, 1534) = 5.28, p = 0.007). A thinner cortex was associated with depressive symptoms in abused (t value = 2.78, p = 0.025) but not in non-abused participants (t value = - 1.50, p = 0.224). Focussing on non-depressed participants, a thicker whole-brain cortex was found in abused compared to non-abused participants (t value = - 2.79, p = 0.025). Similar interaction effects were observed in 12 out of 34 cortical regions. Our results suggest that childhood abuse is associated with reduced cortical thickness in subjects with depressive symptoms. In abused subjects without depressive symptoms, larger cortical thickness might act compensatory and thus reflect resilience against depressive symptoms.
Collapse
|
242
|
Tang S, Cao P, Huang M, Liu X, Zaiane O. Dual feature correlation guided multi-task learning for Alzheimer's disease prediction. Comput Biol Med 2022; 140:105090. [PMID: 34875406 DOI: 10.1016/j.compbiomed.2021.105090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a gradually progressive neurodegenerative disease affecting cognition functions. Predicting the cognitive scores from neuroimage measures and identifying relevant imaging biomarkers are important research topics in the study of AD. Despite machine learning algorithms having many successful applications, the prediction model suffers from the so-called curse of dimensionality. Multi-task feature learning (MTFL) has helped tackle this problem incorporating the correlations among multiple clinical cognitive scores. However, MTFL neglects the inherent correlation among brain imaging measures. In order to better predict the cognitive scores and identify stable biomarkers, we first propose a generalized multi-task formulation framework that incorporates the task and feature correlation structures simultaneously. Second, we present a novel feature-aware sparsity-inducing norm (FAS-norm) penalty to incorporate a useful correlation between brain regions by exploiting correlations among features. Three multi-task learning models that incorporate the FAS-norm penalty are proposed following our framework. Finally, the algorithm based on the alternating direction method of multipliers (ADMM) is developed to optimize the non-smooth problems. We comprehensively evaluate the proposed models on the cross-sectional and longitudinal Alzheimer's disease neuroimaging initiative datasets. The inputs are the thickness measures and the volume measures of the cortical regions of interest. Compared with MTFL, our methods achieve an average decrease of 4.28% in overall error in the cross-sectional analysis and an average decrease of 7.97% in the Alzheimer's Disease Assessment Scale cognitive total score longitudinal analysis. Moreover, our methods identify sensitive and stable biomarkers to physicians, such as the hippocampus, lateral ventricle, and corpus callosum.
Collapse
Affiliation(s)
- Shanshan Tang
- College of Information Science and Engineering, State Key Laboratory of Synthetical Automation for Process Industries, Northeastern University, Shenyang, Liaoning, 110819, China
| | - Peng Cao
- College of Computer Science and Engineering, Northeastern University, Shenyang, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, China.
| | - Min Huang
- College of Information Science and Engineering, State Key Laboratory of Synthetical Automation for Process Industries, Northeastern University, Shenyang, Liaoning, 110819, China.
| | - Xiaoli Liu
- Department of Chemical and Biomolecular Engineering, Faculty of Engineering, National University of Singapore, Singapore
| | - Osmar Zaiane
- Department of Computing Science, University of Alberta, Edmonton, Canada
| |
Collapse
|
243
|
Drouin SM, McFall GP, Potvin O, Bellec P, Masellis M, Duchesne S, Dixon RA. Data-Driven Analyses of Longitudinal Hippocampal Imaging Trajectories: Discrimination and Biomarker Prediction of Change Classes. J Alzheimers Dis 2022; 88:97-115. [PMID: 35570482 PMCID: PMC9277685 DOI: 10.3233/jad-215289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Hippocampal atrophy is a well-known biomarker of neurodegeneration, such as that observed in Alzheimer's disease (AD). Although distributions of hippocampal volume trajectories for asymptomatic individuals often reveal substantial heterogeneity, it is unclear whether interpretable trajectory classes can be objectively detected and used for prediction analyses. OBJECTIVE To detect and predict hippocampal trajectory classes in a computationally competitive context using established AD-related risk factors/biomarkers. METHODS We used biomarker/risk factor and longitudinal MRI data in asymptomatic adults from the AD Neuroimaging Initiative (n = 351; Mean = 75 years; 48.7% female). First, we applied latent class growth analyses to left (LHC) and right (RHC) hippocampal trajectory distributions to identify distinct classes. Second, using random forest analyses, we tested 38 multi-modal biomarkers/risk factors for their relative importance in discriminating the lower (potentially elevated atrophy risk) from the higher (potentially reduced risk) class. RESULTS For both LHC and RHC trajectory distribution analyses, we observed three distinct trajectory classes. Three biomarkers/risk factors predicted membership in LHC and RHC lower classes: male sex, higher education, and lower plasma Aβ1-42. Four additional factors selectively predicted membership in the lower LHC class: lower plasma tau and Aβ1-40, higher depressive symptomology, and lower body mass index. CONCLUSION Data-driven analyses of LHC and RHC trajectories detected three classes underlying the heterogeneous distributions. Machine learning analyses determined three common and four unique biomarkers/risk factors discriminating the higher and lower LHC/RHC classes. Our sequential analytic approach produced evidence that the dynamics of preclinical hippocampal trajectories can be predicted by AD-related biomarkers/risk factors from multiple modalities.
Collapse
Affiliation(s)
- Shannon M. Drouin
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
| | - G. Peggy McFall
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Pierre Bellec
- Département de Psychologie, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montreal, QC, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Research Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Medicine (Neurology), University of Toronto, Toronto, ON, Canada
| | - Simon Duchesne
- CERVO Brain Research Centre, Quebec, QC, Canada
- Radiology and Nuclear Medicine Department, Université Laval, Quebec, QC, Canada
| | - Roger A. Dixon
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
244
|
Likitlersuang J, Brown EM, Salat DH, Iverson KM, Werner K, McGlinchey RE, Galovski TE, Fortier CB. Neural Correlates of Traumatic Brain Injury in Women Survivors of Intimate Partner Violence: A Structural and Functional Connectivity Neuroimaging Study. J Head Trauma Rehabil 2022; 37:E30-E38. [PMID: 34985038 DOI: 10.1097/htr.0000000000000758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE More than one-third of women in the United States experience intimate partner violence (IPV) in their lifetime, increasing their risk for traumatic brain injury (TBI). Despite the prevalence of TBI among IPV survivors, research is sparse in comparison with parallel populations (eg, military, accidents, sports). This pilot study aimed to provide a preliminary investigation of the effect of TBI on brain morphometry and resting-state functional connectivity in women who experience IPV. PARTICIPANTS A total of 45 community-dwelling women survivors of IPV who screened positive for posttraumatic stress disorder (PTSD). DESIGN Participants completed comprehensive assessments of trauma exposure, PTSD, TBI history, and brain neurological health. Twenty-three participants (51.1%) met diagnostic criteria for lifetime TBI. Of these, 15 participants experienced 1 or more TBIs resulting from IPV. The remaining participants experienced TBI from non-IPV exposures (eg, sports/motor vehicle accident). Surface-based neuroimaging analyses were performed to examine group differences in cortical thickness and in functional connectivity of amygdala and isthmus cingulate seeds to examine emotion regulation and the default mode network, respectively. MAIN MEASURES Boston Assessment of Traumatic Brain Injury-Lifetime for Intimate Partner Violence (BAT-L/IPV); Clinician Administered PTSD Scale (CAPS); structural and functional neuroimaging. RESULTS History of lifetime TBI in women IPV survivors was associated with differences in cortical thickness as well as functional connectivity between the isthmus cingulate seed and a variety of regions, including superior parietal and frontal cortices. Individuals with IPV-related TBI showed lower cortical thickness in the right paracentral gyrus than individuals with TBI from other non-IPV etiologies. CONCLUSION Significant differences in brain structure and connectivity were observed in individuals with IPV and TBI. A lower mean cortical thickness of the paracentral gyrus was associated with TBI due to IPV than TBI from other etiologies. Although preliminary, findings from this pilot study present a step toward identifying potential mechanisms by which IPV and TBI secondary to IPV impact brain health in women.
Collapse
Affiliation(s)
- Jirapat Likitlersuang
- Neuroimaging Research for Veterans (NeRVe) Center (Drs Likitlersuang and Salat and Ms Brown), Translational Research Center for TBI and Stress Disorders (TRACTS) (Drs Likitlersuang, McGlinchey, Fortier, and Salat and Ms Brown), Women's Health Sciences Division of the National Center for PTSD (Drs Iverson and Galovski), and Geriatric Research, Educational and Clinical Center (GRECC) (Drs McGlinchey and Fortier), VA Boston Healthcare System, Boston, Massachusetts; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital Department of Radiology, Charlestown, Massachusetts (Dr Salat); Department of Psychiatry (Drs Iverson, McGlinchey, and Galovski), Boston University School of Medicine, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (Dr Likitlersuang, McGlinchey, and Fortier); and College of Nursing, University of Missouri-St Louis (Dr Werner)
| | | | | | | | | | | | | | | |
Collapse
|
245
|
Zugman A, Harrewijn A, Cardinale EM, Zwiebel H, Freitag GF, Werwath KE, Bas‐Hoogendam JM, Groenewold NA, Aghajani M, Hilbert K, Cardoner N, Porta‐Casteràs D, Gosnell S, Salas R, Blair KS, Blair JR, Hammoud MZ, Milad M, Burkhouse K, Phan KL, Schroeder HK, Strawn JR, Beesdo‐Baum K, Thomopoulos SI, Grabe HJ, Van der Auwera S, Wittfeld K, Nielsen JA, Buckner R, Smoller JW, Mwangi B, Soares JC, Wu M, Zunta‐Soares GB, Jackowski AP, Pan PM, Salum GA, Assaf M, Diefenbach GJ, Brambilla P, Maggioni E, Hofmann D, Straube T, Andreescu C, Berta R, Tamburo E, Price R, Manfro GG, Critchley HD, Makovac E, Mancini M, Meeten F, Ottaviani C, Agosta F, Canu E, Cividini C, Filippi M, Kostić M, Munjiza A, Filippi CA, Leibenluft E, Alberton BAV, Balderston NL, Ernst M, Grillon C, Mujica‐Parodi LR, van Nieuwenhuizen H, Fonzo GA, Paulus MP, Stein MB, Gur RE, Gur RC, Kaczkurkin AN, Larsen B, Satterthwaite TD, Harper J, Myers M, Perino MT, Yu Q, Sylvester CM, Veltman DJ, Lueken U, Van der Wee NJA, Stein DJ, Jahanshad N, Thompson PM, Pine DS, Winkler AM. Mega-analysis methods in ENIGMA: The experience of the generalized anxiety disorder working group. Hum Brain Mapp 2022; 43:255-277. [PMID: 32596977 PMCID: PMC8675407 DOI: 10.1002/hbm.25096] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/26/2020] [Accepted: 05/31/2020] [Indexed: 12/15/2022] Open
Abstract
The ENIGMA group on Generalized Anxiety Disorder (ENIGMA-Anxiety/GAD) is part of a broader effort to investigate anxiety disorders using imaging and genetic data across multiple sites worldwide. The group is actively conducting a mega-analysis of a large number of brain structural scans. In this process, the group was confronted with many methodological challenges related to study planning and implementation, between-country transfer of subject-level data, quality control of a considerable amount of imaging data, and choices related to statistical methods and efficient use of resources. This report summarizes the background information and rationale for the various methodological decisions, as well as the approach taken to implement them. The goal is to document the approach and help guide other research groups working with large brain imaging data sets as they develop their own analytic pipelines for mega-analyses.
Collapse
Affiliation(s)
- André Zugman
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Anita Harrewijn
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Elise M. Cardinale
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Hannah Zwiebel
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Gabrielle F. Freitag
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Katy E. Werwath
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Janna M. Bas‐Hoogendam
- Leiden University Medical Center, Department of PsychiatryLeidenThe Netherlands
- Leiden Institute for Brain and Cognition (LIBC)LeidenThe Netherlands
- Leiden University, Institute of Psychology, Developmental and Educational PsychologyLeidenThe Netherlands
| | - Nynke A. Groenewold
- Department of Psychiatry & Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Moji Aghajani
- Department. of PsychiatryAmsterdam UMC/VUMCAmsterdamThe Netherlands
- GGZ InGeestDepartment of Research & InnovationAmsterdamThe Netherlands
| | - Kevin Hilbert
- Department of PsychologyHumboldt‐Universität zu BerlinBerlinGermany
| | - Narcis Cardoner
- Department of Mental HealthUniversity Hospital Parc Taulí‐I3PTBarcelonaSpain
- Department of Psychiatry and Forensic MedicineUniversitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red de Salud MentalCarlos III Health InstituteMadridSpain
| | - Daniel Porta‐Casteràs
- Department of Mental HealthUniversity Hospital Parc Taulí‐I3PTBarcelonaSpain
- Department of Psychiatry and Forensic MedicineUniversitat Autònoma de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red de Salud MentalCarlos III Health InstituteMadridSpain
| | - Savannah Gosnell
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of MedicineHoustonTexasUSA
| | - Ramiro Salas
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of MedicineHoustonTexasUSA
| | - Karina S. Blair
- Center for Neurobehavioral ResearchBoys Town National Research HospitalBoys TownNebraskaUSA
| | - James R. Blair
- Center for Neurobehavioral ResearchBoys Town National Research HospitalBoys TownNebraskaUSA
| | - Mira Z. Hammoud
- Department of PsychiatryNew York UniversityNew YorkNew YorkUSA
| | - Mohammed Milad
- Department of PsychiatryNew York UniversityNew YorkNew YorkUSA
| | - Katie Burkhouse
- Department of PsychiatryUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - K. Luan Phan
- Department of Psychiatry and Behavioral HealthThe Ohio State UniversityColumbusOhioUSA
| | - Heidi K. Schroeder
- Department of Psychiatry & Behavioral NeuroscienceUniversity of CincinnatiCincinnatiOhioUSA
| | - Jeffrey R. Strawn
- Department of Psychiatry & Behavioral NeuroscienceUniversity of CincinnatiCincinnatiOhioUSA
| | - Katja Beesdo‐Baum
- Behavioral EpidemiologyInstitute of Clinical Psychology and Psychotherapy, Technische Universität DresdenDresdenGermany
| | - Sophia I. Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Hans J. Grabe
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
- German Center for Neurodegenerative Diseases (DZNE)Site Rostock/GreifswaldGreifswaldGermany
| | - Sandra Van der Auwera
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
- German Center for Neurodegenerative Diseases (DZNE)Site Rostock/GreifswaldGreifswaldGermany
| | - Katharina Wittfeld
- Department of Psychiatry and PsychotherapyUniversity Medicine GreifswaldGreifswaldGermany
- German Center for Neurodegenerative Diseases (DZNE)Site Rostock/GreifswaldGreifswaldGermany
| | - Jared A. Nielsen
- Department of PsychologyHarvard UniversityCambridgeMassachusettsUSA
- Center for Brain ScienceHarvard UniversityCambridgeMassachusettsUSA
| | - Randy Buckner
- Department of PsychologyHarvard UniversityCambridgeMassachusettsUSA
- Center for Brain ScienceHarvard UniversityCambridgeMassachusettsUSA
- Department of PsychiatryMassachusetts General HospitalBostonMassachusettsUSA
| | - Jordan W. Smoller
- Department of PsychiatryMassachusetts General HospitalBostonMassachusettsUSA
| | - Benson Mwangi
- Center Of Excellence On Mood Disorders, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Jair C. Soares
- Center Of Excellence On Mood Disorders, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Mon‐Ju Wu
- Center Of Excellence On Mood Disorders, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Giovana B. Zunta‐Soares
- Center Of Excellence On Mood Disorders, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Andrea P. Jackowski
- LiNC, Department of PsychiatryFederal University of São PauloSão PauloSão PauloBrazil
| | - Pedro M. Pan
- LiNC, Department of PsychiatryFederal University of São PauloSão PauloSão PauloBrazil
| | - Giovanni A. Salum
- Section on Negative Affect and Social Processes, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreRio Grande do SulBrazil
| | - Michal Assaf
- Olin Neuropsychiatry Research CenterInstitute of Living, Hartford HospitalHartfordConnecticutUSA
- Department of PsychiatryYale School of MedicineNew HavenConnecticutUSA
| | - Gretchen J. Diefenbach
- Anxiety Disorders CenterInstitute of Living, Hartford HospitalHartfordConnecticutUSA
- Yale School of MedicineNew HavenConnecticutUSA
| | - Paolo Brambilla
- Department of Neurosciences and Mental HealthFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Eleonora Maggioni
- Department of Neurosciences and Mental HealthFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - David Hofmann
- Institute of Medical Psychology and Systems Neuroscience, University of MuensterMuensterGermany
| | - Thomas Straube
- Institute of Medical Psychology and Systems Neuroscience, University of MuensterMuensterGermany
| | - Carmen Andreescu
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rachel Berta
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Erica Tamburo
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rebecca Price
- Department of Psychiatry & PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Gisele G. Manfro
- Anxiety Disorder ProgramHospital de Clínicas de Porto AlegrePorto AlegreRio Grande do SulBrazil
- Department of PsychiatryFederal University of Rio Grande do SulPorto AlegreRio Grande do SulBrazil
| | - Hugo D. Critchley
- Department of NeuroscienceBrighton and Sussex Medical School, University of SussexBrightonUK
| | - Elena Makovac
- Centre for Neuroimaging ScienceKings College LondonLondonUK
| | - Matteo Mancini
- Department of NeuroscienceBrighton and Sussex Medical School, University of SussexBrightonUK
| | | | | | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
| | - Elisa Canu
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Camilla Cividini
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Vita‐Salute San Raffaele UniversityMilanItaly
- Neurology and Neurophysiology UnitIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Milutin Kostić
- Institute of Mental Health, University of BelgradeBelgradeSerbia
- Department of Psychiatry, School of MedicineUniversity of BelgradeBelgradeSerbia
| | - Ana Munjiza
- Institute of Mental Health, University of BelgradeBelgradeSerbia
| | - Courtney A. Filippi
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Ellen Leibenluft
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Bianca A. V. Alberton
- Graduate Program in Electrical and Computer Engineering, Universidade Tecnológica Federal do ParanáCuritibaPuerto RicoBrazil
| | - Nicholas L. Balderston
- Center for Neuromodulation in Depression and StressUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Monique Ernst
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Christian Grillon
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | | | | | - Gregory A. Fonzo
- Department of PsychiatryThe University of Texas at Austin Dell Medical SchoolAustinTexasUSA
| | | | - Murray B. Stein
- Department of Psychiatry & Family Medicine and Public HealthUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Raquel E. Gur
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ruben C. Gur
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Bart Larsen
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Jennifer Harper
- Department of PsychiatryWashington UniversitySt. LouisMissouriUSA
| | - Michael Myers
- Department of PsychiatryWashington UniversitySt. LouisMissouriUSA
| | | | - Qiongru Yu
- Department of PsychiatryWashington UniversitySt. LouisMissouriUSA
| | | | - Dick J. Veltman
- Department. of PsychiatryAmsterdam UMC/VUMCAmsterdamThe Netherlands
| | - Ulrike Lueken
- Department of PsychologyHumboldt‐Universität zu BerlinBerlinGermany
| | - Nic J. A. Van der Wee
- Leiden University Medical Center, Department of PsychiatryLeidenThe Netherlands
- Leiden Institute for Brain and Cognition (LIBC)LeidenThe Netherlands
| | - Dan J. Stein
- Department of Psychiatry & Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
- SAMRC Unite on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Daniel S. Pine
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Anderson M. Winkler
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH)BethesdaMarylandUSA
| |
Collapse
|
246
|
Tüngler A, Van der Auwera S, Wittfeld K, Frenzel S, Terock J, Röder N, Homuth G, Völzke H, Bülow R, Grabe HJ, Janowitz D. Body mass index but not genetic risk is longitudinally associated with altered structural brain parameters. Sci Rep 2021; 11:24246. [PMID: 34930940 PMCID: PMC8688483 DOI: 10.1038/s41598-021-03343-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022] Open
Abstract
Evidence from previous studies suggests that elevated body mass index (BMI) and genetic risk for obesity is associated with reduced brain volume, particularly in areas of reward-related cognition, e.g. the medial prefrontal cortex (AC-MPFC), the orbitofrontal cortex (OFC), the striatum and the thalamus. However, only few studies examined the interplay between these factors in a joint approach. Moreover, previous findings are based on cross-sectional data. We investigated the longitudinal relationship between increased BMI, brain structural magnetic resonance imaging (MRI) parameters and genetic risk scores in a cohort of n = 502 community-dwelling participants from the Study of Health in Pomerania (SHIP) with a mean follow-up-time of 4.9 years. We found that (1) increased BMI values at baseline were associated with decreased brain parameters at follow-up. These effects were particularly pronounced for the OFC and AC-MPFC. (2) The genetic predisposition for BMI had no effect on brain parameters at baseline or follow-up. (3) The interaction between the genetic score for BMI and brain parameters had no effect on BMI at baseline. Finding a significant impact of overweight, but not genetic predisposition for obesity on altered brain structure suggests that metabolic mechanisms may underlie the relationship between obesity and altered brain structure.
Collapse
Affiliation(s)
- Anne Tüngler
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany.,German Center for Neurodegenerative Diseases DZNE, Site Rostock/Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany.,German Center for Neurodegenerative Diseases DZNE, Site Rostock/Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Jan Terock
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany.,Department of Psychiatry and Psychotherapy, HELIOS Hanseklinikum Stralsund, Rostocker Chaussee 70, 18437, Stralsund, Germany
| | - Nele Röder
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Walther-Rathenau-Str. 48, 17487, Greifswald, Germany
| | - Robin Bülow
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Str., 17475, Greifswald, Germany
| | - Hans Jörgen Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany.,German Center for Neurodegenerative Diseases DZNE, Site Rostock/Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany
| | - Deborah Janowitz
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1-2, 17475, Greifswald, Germany. .,Department of Psychiatry and Psychotherapy, HELIOS Hanseklinikum Stralsund, Rostocker Chaussee 70, 18437, Stralsund, Germany.
| |
Collapse
|
247
|
Bieneck V, Bletsch A, Mann C, Schäfer T, Seelemeyer H, Herøy N, Zimmermann J, Pretzsch CM, Hattingen E, Ecker C. Longitudinal Changes in Cortical Thickness in Adolescents with Autism Spectrum Disorder and Their Association with Restricted and Repetitive Behaviors. Genes (Basel) 2021; 12:2024. [PMID: 34946972 PMCID: PMC8701312 DOI: 10.3390/genes12122024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/01/2023] Open
Abstract
The neuroanatomy of autism spectrum disorder (ASD) shows highly heterogeneous developmental trajectories across individuals. Mapping atypical brain development onto clinical phenotypes, and establishing their molecular underpinnings, is therefore crucial for patient stratification and subtyping. In this longitudinal study we examined intra- and inter-individual differences in the developmental trajectory of cortical thickness (CT) in childhood and adolescence, and their genomic underpinnings, in 33 individuals with ASD and 37 typically developing controls (aged 11-18 years). Moreover, we aimed to link regional atypical CT development to intra-individual variations in restricted and repetitive behavior (RRB) over a two-year time period. Individuals with ASD showed significantly reduced cortical thinning in several of the brain regions functionally related to wider autism symptoms and traits (e.g., fronto-temporal and cingulate cortices). The spatial patterns of the neuroanatomical differences in CT were enriched for genes known to be associated with ASD at a genetic and transcriptomic level. Further, intra-individual differences in CT correlated with within-subject variability in the severity of RRBs. Our findings represent an important step towards characterizing the neuroanatomical underpinnings of ASD across development based upon measures of CT. Moreover, our findings provide important novel insights into the link between microscopic and macroscopic pathology in ASD, as well as their relationship with different clinical ASD phenotypes.
Collapse
Affiliation(s)
- Valentina Bieneck
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
| | - Anke Bletsch
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
| | - Caroline Mann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
| | - Tim Schäfer
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
| | - Hanna Seelemeyer
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
| | - Njål Herøy
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
| | - Jennifer Zimmermann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
| | - Charlotte Marie Pretzsch
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London SE5 8AF, UK;
| | - Elke Hattingen
- Institute for Neuroradiology, University Hospital, Goethe University, 60528 Frankfurt, Germany;
| | - Christine Ecker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Deutschordenstrasse 50, 60528 Frankfurt, Germany; (A.B.); (C.M.); (T.S.); (H.S.); (N.H.); (J.Z.); (C.E.)
- Brain Imaging Center, Schleusenweg 2-16, Haus 95H, Goethe University, 60528 Frankfurt, Germany
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College, London SE5 8AF, UK;
| |
Collapse
|
248
|
Dwivedi M, Dubey N, Pansari AJ, Bapi RS, Das M, Guha M, Banerjee R, Pramanick G, Basu J, Ghosh A. Effects of Meditation on Structural Changes of the Brain in Patients With Mild Cognitive Impairment or Alzheimer's Disease Dementia. Front Hum Neurosci 2021; 15:728993. [PMID: 34867239 PMCID: PMC8633496 DOI: 10.3389/fnhum.2021.728993] [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: 06/22/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Previous cross-sectional studies reported positive effects of meditation on the brain areas related to attention and executive function in the healthy elderly population. Effects of long-term regular meditation in persons with mild cognitive impairment (MCI) and Alzheimer's disease dementia (AD) have rarely been studied. In this study, we explored changes in cortical thickness and gray matter volume in meditation-naïve persons with MCI or mild AD after long-term meditation intervention. MCI or mild AD patients underwent detailed clinical and neuropsychological assessment and were assigned into meditation or non-meditation groups. High resolution T1-weighted magnetic resonance images (MRI) were acquired at baseline and after 6 months. Longitudinal symmetrized percentage changes (SPC) in cortical thickness and gray matter volume were estimated. Left caudal middle frontal, left rostral middle frontal, left superior parietal, right lateral orbitofrontal, and right superior frontal cortices showed changes in both cortical thickness and gray matter volume; the left paracentral cortex showed changes in cortical thickness; the left lateral occipital, left superior frontal, left banks of the superior temporal sulcus (bankssts), and left medial orbitofrontal cortices showed changes in gray matter volume. All these areas exhibited significantly higher SPC values in meditators as compared to non-meditators. Conversely, the left lateral occipital, and right posterior cingulate cortices showed significantly lower SPC values for cortical thickness in the meditators. In hippocampal subfields analysis, we observed significantly higher SPC in gray matter volume of the left CA1, molecular layer HP, and CA3 with a trend for increased gray matter volume in most other areas. No significant changes were found for the hippocampal subfields in the right hemisphere. Analysis of the subcortical structures revealed significantly increased volume in the right thalamus in the meditation group. The results of the study point out that long-term meditation practice in persons with MCI or mild AD leads to salutary changes in cortical thickness and gray matter volumes. Most of these changes were observed in the brain areas related to executive control and memory that are prominently at risk in neurodegenerative diseases.
Collapse
Affiliation(s)
- Madhukar Dwivedi
- Cognitive Science Lab, International Institute of Information Technology, Hyderabad, India
| | - Neha Dubey
- Department of Neurology, Apollo Gleneagles Hospital, Kolkata, India.,Department of Applied Psychology, University of Calcutta, Kolkata, India
| | - Aditya Jain Pansari
- Cognitive Science Lab, International Institute of Information Technology, Hyderabad, India
| | - Raju Surampudi Bapi
- Cognitive Science Lab, International Institute of Information Technology, Hyderabad, India
| | - Meghoranjani Das
- Department of Neurology, Apollo Gleneagles Hospital, Kolkata, India
| | - Maushumi Guha
- Department of Philosophy, Jadavpur University, Kolkata, India
| | - Rahul Banerjee
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India
| | | | - Jayanti Basu
- Department of Applied Psychology, University of Calcutta, Kolkata, India
| | - Amitabha Ghosh
- Department of Neurology, Apollo Gleneagles Hospital, Kolkata, India
| |
Collapse
|
249
|
Faw TD, Lakhani B, Schmalbrock P, Knopp MV, Lohse KR, Kramer JLK, Liu H, Nguyen HT, Phillips EG, Bratasz A, Fisher LC, Deibert RJ, Boyd LA, McTigue DM, Basso DM. Eccentric rehabilitation induces white matter plasticity and sensorimotor recovery in chronic spinal cord injury. Exp Neurol 2021; 346:113853. [PMID: 34464653 PMCID: PMC10084731 DOI: 10.1016/j.expneurol.2021.113853] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/04/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Experience-dependent white matter plasticity offers new potential for rehabilitation-induced recovery after neurotrauma. This first-in-human translational experiment combined myelin water imaging in humans and genetic fate-mapping of oligodendrocyte lineage cells in mice to investigate whether downhill locomotor rehabilitation that emphasizes eccentric muscle actions promotes white matter plasticity and recovery in chronic, incomplete spinal cord injury (SCI). In humans, of 20 individuals with SCI that enrolled, four passed the imaging screen and had myelin water imaging before and after a 12-week (3 times/week) downhill locomotor treadmill training program (SCI + DH). One individual was excluded for imaging artifacts. Uninjured control participants (n = 7) had two myelin water imaging sessions within the same day. Changes in myelin water fraction (MWF), a histopathologically-validated myelin biomarker, were analyzed in a priori motor learning and non-motor learning brain regions and the cervical spinal cord using statistical approaches appropriate for small sample sizes. PDGFRα-CreERT2:mT/mG mice, that express green fluorescent protein on oligodendrocyte precursor cells and subsequent newly-differentiated oligodendrocytes upon tamoxifen-induced recombination, were either naive (n = 6) or received a moderate (75 kilodyne), contusive SCI at T9 and were randomized to downhill training (n = 6) or unexercised groups (n = 6). We initiated recombination 29 days post-injury, seven days prior to downhill training. Mice underwent two weeks of daily downhill training on the same 10% decline grade used in humans. Between-group comparison of functional (motor and sensory) and histological (oligodendrogenesis, oligodendroglial/axon interaction, paranodal structure) outcomes occurred post-training. In humans with SCI, downhill training increased MWF in brain motor learning regions (postcentral, precuneus) and mixed motor and sensory tracts of the ventral cervical spinal cord compared to control participants (P < 0.05). In mice with thoracic SCI, downhill training induced oligodendrogenesis in cervical dorsal and lateral white matter, increased axon-oligodendroglial interactions, and normalized paranodal structure in dorsal column sensory tracts (P < 0.05). Downhill training improved sensorimotor recovery in mice by normalizing hip and knee motor control and reducing hyperalgesia, both of which were associated with new oligodendrocytes in the cervical dorsal columns (P < 0.05). Our findings indicate that eccentric-focused, downhill rehabilitation promotes white matter plasticity and improved function in chronic SCI, likely via oligodendrogenesis in nervous system regions activated by the training paradigm. Together, these data reveal an exciting role for eccentric training in white matter plasticity and sensorimotor recovery after SCI.
Collapse
Affiliation(s)
- Timothy D Faw
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA
| | - Bimal Lakhani
- Department of Physical Therapy, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Petra Schmalbrock
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Michael V Knopp
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Keith R Lohse
- Department of Health, Kinesiology, and Recreation, University of Utah, Salt Lake City, UT 84112, USA; Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT 84108, USA
| | - John L K Kramer
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Hanwen Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Huyen T Nguyen
- Department of Radiology, The Ohio State University, Columbus, OH 43210, USA
| | - Eileen G Phillips
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Anna Bratasz
- Small Animal Imaging Shared Resources, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Lesley C Fisher
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Rochelle J Deibert
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Lara A Boyd
- Department of Physical Therapy, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Dana M McTigue
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - D Michele Basso
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH 43210, USA; School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
250
|
King DJ, Seri S, Catroppa C, Anderson VA, Wood AG. Structural-covariance networks identify topology-based cortical-thickness changes in children with persistent executive function impairments after traumatic brain injury. Neuroimage 2021; 244:118612. [PMID: 34563681 PMCID: PMC8591373 DOI: 10.1016/j.neuroimage.2021.118612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/14/2021] [Accepted: 09/20/2021] [Indexed: 11/05/2022] Open
Abstract
Paediatric traumatic brain injury (pTBI) results in inconsistent changes to regional morphometry of the brain across studies. Structural-covariance networks represent the degree to which the morphology (typically cortical-thickness) of cortical-regions co-varies with other regions, driven by both biological and developmental factors. Understanding how heterogeneous regional changes may influence wider cortical network organization may more appropriately capture prognostic information in terms of long term outcome following a pTBI. The current study aimed to investigate the relationships between cortical organisation as measured by structural-covariance, and long-term cognitive impairment following pTBI. T1-weighted magnetic resonance imaging (MRI) from n = 83 pTBI patients and 33 typically developing controls underwent 3D-tissue segmentation using Freesurfer to estimate cortical-thickness across 68 cortical ROIs. Structural-covariance between regions was estimated using Pearson's correlations between cortical-thickness measures across 68 regions-of-interest (ROIs), generating a group-level 68 × 68 adjacency matrix for patients and controls. We grouped a subset of patients who underwent executive function testing at 2-years post-injury using a neuropsychological impairment (NPI) rule, defining impaired- and non-impaired subgroups. Despite finding no significant reductions in regional cortical-thickness between the control and pTBI groups, we found specific reductions in graph-level strength of the structural covariance graph only between controls and the pTBI group with executive function (EF) impairment. Node-level differences in strength for this group were primarily found in frontal regions. We also investigated whether the top n nodes in terms of effect-size of cortical-thickness reductions were nodes that had significantly greater strength in the typically developing brain than n randomly selected regions. We found that acute cortical-thickness reductions post-pTBI are loaded onto regions typically high in structural covariance. This association was found in those patients with persistent EF impairment at 2-years post-injury, but not in those for whom these abilities were spared. This study posits that the topography of post-injury cortical-thickness reductions in regions that are central to the typical structural-covariance topology of the brain, can explain which patients have poor EF at follow-up.
Collapse
Affiliation(s)
- Daniel J King
- College of Health and Life Sciences and Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK.
| | - Stefano Seri
- College of Health and Life Sciences and Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK; Department of Clinical Neurophysiology, Birmingham Women's and Children's Hospital NHS Foundation Trust, UK
| | - Cathy Catroppa
- Brain and Mind Research, Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Australia; Department of Psychology, Royal Children's Hospital, Melbourne, Australia
| | - Vicki A Anderson
- Brain and Mind Research, Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Australia; Department of Psychology, Royal Children's Hospital, Melbourne, Australia
| | - Amanda G Wood
- College of Health and Life Sciences and Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK; Brain and Mind Research, Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Australia; School of Psychology, Faculty of Health, Melbourne Burwood Campus, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|