551
|
Bhattacharyya S, Egerton A, Kim E, Rosso L, Riano Barros D, Hammers A, Brammer M, Turkheimer FE, Howes OD, McGuire P. Acute induction of anxiety in humans by delta-9-tetrahydrocannabinol related to amygdalar cannabinoid-1 (CB1) receptors. Sci Rep 2017; 7:15025. [PMID: 29101333 PMCID: PMC5670208 DOI: 10.1038/s41598-017-14203-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022] Open
Abstract
Use of Cannabis, the most widely used illicit drug worldwide, is associated with acute anxiety, and anxiety disorders following regular use. The precise neural and receptor basis of these effects have not been tested in man. Employing a combination of functional MRI (fMRI) and positron emission tomography (PET), we investigated whether the effects of delta-9-tetrahydrocannabinol (delta-9-THC), the main psychoactive ingredient of cannabis, on anxiety and on amygdala response while processing fearful stimuli were related to local availability of its main central molecular target, cannabinoid-1 (CB1) receptors in man. Fourteen healthy males were studied with fMRI twice, one month apart, following an oral dose of either delta-9-THC (10 mg) or placebo, while they performed a fear-processing task. Baseline availability of the CB1 receptor was studied using PET with [11C]MePPEP, a CB1 inverse agonist radioligand. Relative to the placebo condition, delta-9-THC induced anxiety and modulated right amygdala activation while processing fear. Both these effects were positively correlated with CB1 receptor availability in the right amygdala. These results suggest that the acute effects of cannabis on anxiety in males are mediated by the modulation of amygdalar function by delta-9-THC and the extent of these effects are related to local availability of CB1 receptors.
Collapse
Affiliation(s)
- Sagnik Bhattacharyya
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK.
| | - Alice Egerton
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Euitae Kim
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Lula Rosso
- Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | | | - Alexander Hammers
- King's College London & Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, 4th floor Lambeth Wing, St Thomas' Hospital, Westminster Bridge Road, London, SE1 7EH, UK
| | - Michael Brammer
- Department of Neuroimaging, Centre for Neuroimaging Sciences, PO Box 089, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Federico E Turkheimer
- Department of Neuroimaging, Centre for Neuroimaging Sciences, PO Box 089, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Oliver D Howes
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
- Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| |
Collapse
|
552
|
Vomacka L, Albert NL, Lindner S, Unterrainer M, Mahler C, Brendel M, Ermoschkin L, Gosewisch A, Brunegraf A, Buckley C, Kümpfel T, Rupprecht R, Ziegler S, Kerschensteiner M, Bartenstein P, Böning G. TSPO imaging using the novel PET ligand [ 18F]GE-180: quantification approaches in patients with multiple sclerosis. EJNMMI Res 2017; 7:89. [PMID: 29150726 PMCID: PMC5693838 DOI: 10.1186/s13550-017-0340-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/20/2017] [Indexed: 01/12/2023] Open
Abstract
Background PET ligands targeting the translocator protein (TSPO) represent promising tools to visualise neuroinflammation. Here, we analysed parameters obtained in dynamic and static PET images using the novel TSPO ligand [18F]GE-180 in patients with relapsing remitting multiple sclerosis (RRMS) and an approach for semi-quantitative assessment of this disease in clinical routine. Seventeen dynamic [18F]GE-180 PET scans of RRMS patients were evaluated (90 min). A pseudo-reference region (PRR) was defined after identification of the least disease-affected brain area by voxel-based comparison with six healthy controls (HC) and upon exclusion of voxels suspected of being affected in static 60–90 min p.i. images. Standardised uptake value ratios (SUVR) obtained from static images normalised to PRR were correlated to the distribution volume ratios (DVR) derived from dynamic data with Logan reference tissue model. Results Group comparison with HC revealed white matter and thalamus as most affected regions. Fewest differences were found in grey matter, and normalisation to frontal cortex (FC) yielded the greatest reduction in variability of healthy grey and white matter. Hence, FC corrected for affected voxels was chosen as PRR, leading to time-activity curves of FC which were congruent to HC data (SUV60–90 0.37, U test P = 0.42). SUVR showed a very strong correlation with DVR (Pearson ρ > 0.9). Focal MS lesions exhibited a high SUVR (range, 1.3–3.2). Conclusions This comparison with parameters from dynamic data suggests that SUVR normalised to corrected frontal cortex as PRR is suitable for the quantification of [18F]GE-180 uptake in lesions and different brain regions of RRMS patients. This efficient diagnostic protocol based on static [18F]GE-180 PET scans acquired 60–90 min p.i. allows the semi-quantitative assessment of neuroinflammation in RRMS patients in clinical routine.
Collapse
Affiliation(s)
- Lena Vomacka
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Nathalie Lisa Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Christoph Mahler
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Larissa Ermoschkin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Astrid Gosewisch
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Anika Brunegraf
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | | | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Guido Böning
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
553
|
A hitchhiker's guide to lesion-behaviour mapping. Neuropsychologia 2017; 115:5-16. [PMID: 29066325 DOI: 10.1016/j.neuropsychologia.2017.10.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 01/09/2023]
Abstract
Lesion-behaviour mapping is an influential and popular approach to anatomically localise cognitive brain functions in the human brain. Multiple considerations, ranging from patient selection, assessment of lesion location and patient behaviour, spatial normalisation, statistical testing, to the anatomical interpretation of obtained results, are necessary to optimize a lesion-behaviour mapping study and arrive at meaningful conclusions. Here, we provide a hitchhiker's guide, giving practical guidelines and references for each step of the typical lesion-behaviour mapping study pipeline.
Collapse
|
554
|
Multimodal correlation of dynamic [ 18F]-AV-1451 perfusion PET and neuronal hypometabolism in [ 18F]-FDG PET. Eur J Nucl Med Mol Imaging 2017; 44:2249-2256. [PMID: 29026951 DOI: 10.1007/s00259-017-3840-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/22/2017] [Indexed: 01/30/2023]
Abstract
PURPOSE Cerebral glucose metabolism measured with [18F]-FDG PET is a well established marker of neuronal dysfunction in neurodegeneration. The tau-protein tracer [18F]-AV-1451 PET is currently under evaluation and shows promising results. Here, we assess the feasibility of early perfusion imaging with AV-1451 as a substite for FDG PET in assessing neuronal injury. METHODS Twenty patients with suspected neurodegeneration underwent FDG and early phase AV-1451 PET imaging. Ten one-minute timeframes were acquired after application of 200 MBq AV-1451. FDG images were acquired on a different date according to clinical protocol. Early AV-1451 timeframes were coregistered to individual FDG-scans and spatially normalized. Voxel-wise intermodal correlations were calculated on within-subject level for every possible time window. The window with highest pooled correlation was considered optimal. Z-transformed deviation maps (ZMs) were created from both FDG and early AV-1451 images, comparing against FDG images of healthy controls. RESULTS Regional patterns and extent of perfusion deficits were highly comparable to metabolic deficits. Best results were observed in a time window from 60 to 360 s (r = 0.86). Correlation strength ranged from r = 0.96 (subcortical gray matter) to 0.83 (frontal lobe) in regional analysis. ZMs of early AV-1451 and FDG images were highly similar. CONCLUSION Perfusion imaging with AV-1451 is a valid biomarker for assessment of neuronal dysfunction in neurodegenerative diseases. Radiation exposure and complexity of the diagnostic workup could be reduced significantly by routine acquisition of early AV-1451 images, sparing additional FDG PET.
Collapse
|
555
|
Wegrzyk J, Kebets V, Richiardi J, Galli S, de Ville DV, Aybek S. Identifying motor functional neurological disorder using resting-state functional connectivity. NEUROIMAGE-CLINICAL 2017; 17:163-168. [PMID: 29071210 PMCID: PMC5651543 DOI: 10.1016/j.nicl.2017.10.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/07/2017] [Accepted: 10/10/2017] [Indexed: 12/02/2022]
Abstract
Background Motor functional neurological disorder (mFND) is a clinical diagnosis with reliable features; however, patients are reluctant to accept the diagnosis and physicians themselves bear doubts on potential misdiagnoses. The identification of a positive biomarker could help limiting unnecessary costs of multiple referrals and investigations, thus promoting early diagnosis and allowing early engagement in appropriate therapy. Objectives To test whether resting-state (RS) functional magnetic resonance imaging could discriminate patients suffering from mFND from healthy controls. Methods We classified 23 mFND patients and 25 age- and gender-matched healthy controls based on whole-brain RS functional connectivity (FC) data, using a support vector machine classifier and the standard Automated Anatomic Labeling (AAL) atlas, as well as two additional atlases for validation. Results Accuracy, specificity and sensitivity were over 68% (p = 0.004) to discriminate between mFND patients and controls, with consistent findings between the three tested atlases. The most discriminative connections comprised the right caudate, amygdala, prefrontal and sensorimotor regions. Post-hoc seed connectivity analyses showed that these regions were hyperconnected in patients compared to controls. Conclusions The good accuracy to discriminate patients from controls suggests that RS FC could be used as a biomarker with high diagnostic value in future clinical practice to identify mFND patients at the individual level. We classified FND patients from controls with 68% accuracy using resting-state fMRI. Right caudate-amygdala connectivity best discriminated patients with FND from controls. Future additional steps are needed before translation to clinical use. This could be used in complement of clinical signs for the diagnosis of FND.
Collapse
Affiliation(s)
- Jennifer Wegrzyk
- Department of Clinical Neuroscience, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva, Switzerland
| | - Valeria Kebets
- Department of Neuroscience, University of Geneva, Campus Biotech, Chemin des Mines 9, 1202 Geneva, Switzerland
| | - Jonas Richiardi
- Department of Neuroscience, University of Geneva, Campus Biotech, Chemin des Mines 9, 1202 Geneva, Switzerland
| | - Silvio Galli
- Department of Clinical Neuroscience, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva, Switzerland
| | - Dimitri Van de Ville
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Campus Biotech, Chemin des Mines 9, 1202 Geneva, Switzerland; Department of Radiology and Medical Informatics, University of Geneva, Campus Biotech, Chemin des Mines 9, 1202 Geneva, Switzerland
| | - Selma Aybek
- Department of Clinical Neuroscience, Geneva University Hospital, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva, Switzerland; Department of Neuroscience, University of Geneva, Campus Biotech, Chemin des Mines 9, 1202 Geneva, Switzerland; Neurology University Clinic, InselSpital, Department of Clinical Neuroscience, 3010 Bern, Switzerland.
| |
Collapse
|
556
|
Lidzba K, de Haan B, Wilke M, Krägeloh-Mann I, Staudt M. Lesion characteristics driving right-hemispheric language reorganization in congenital left-hemispheric brain damage. BRAIN AND LANGUAGE 2017; 173:1-9. [PMID: 28549234 DOI: 10.1016/j.bandl.2017.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 04/19/2017] [Accepted: 04/28/2017] [Indexed: 06/07/2023]
Abstract
Pre- or perinatally acquired ("congenital") left-hemispheric brain lesions can be compensated for by reorganizing language into homotopic brain regions in the right hemisphere. Language comprehension may be hemispherically dissociated from language production. We investigated the lesion characteristics driving inter-hemispheric reorganization of language comprehension and language production in 19 patients (7-32years; eight females) with congenital left-hemispheric brain lesions (periventricular lesions [n=11] and middle cerebral artery infarctions [n=8]) by fMRI. 16/17 patients demonstrated reorganized language production, while 7/19 patients had reorganized language comprehension. Lesions to the insular cortex and the temporo-parietal junction (predominantly supramarginal gyrus) were significantly more common in patients in whom both, language production and comprehension were reorganized. These areas belong to the dorsal stream of the language network, participating in the auditory-motor integration of language. Our data suggest that the integrity of this stream might be crucial for a normal left-lateralized language development.
Collapse
Affiliation(s)
- Karen Lidzba
- University Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, University of Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany; Experimental Pediatric Neuroimaging Group, Department of Pediatric Neurology and Developmental Medicine & Department of Neuroradiology, University Hospital Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany.
| | - Bianca de Haan
- Center of Neurology, Division of Neuropsychology, Hertie-Institute of Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Marko Wilke
- University Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, University of Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany; Experimental Pediatric Neuroimaging Group, Department of Pediatric Neurology and Developmental Medicine & Department of Neuroradiology, University Hospital Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Ingeborg Krägeloh-Mann
- University Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, University of Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Martin Staudt
- University Children's Hospital, Department of Pediatric Neurology and Developmental Medicine, University of Tübingen, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany; Clinic for Neuropediatrics and Neurorehabilitation, Epilepsy Centre for Children and Adolescents, Schön Klinik Vogtareuth, Krankenhausstr. 20, 83569 Vogtareuth, Germany
| |
Collapse
|
557
|
Michels L, Muthuraman M, Anwar AR, Kollias S, Leh SE, Riese F, Unschuld PG, Siniatchkin M, Gietl AF, Hock C. Changes of Functional and Directed Resting-State Connectivity Are Associated with Neuronal Oscillations, ApoE Genotype and Amyloid Deposition in Mild Cognitive Impairment. Front Aging Neurosci 2017; 9:304. [PMID: 29081745 PMCID: PMC5646353 DOI: 10.3389/fnagi.2017.00304] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 09/04/2017] [Indexed: 01/03/2023] Open
Abstract
The assessment of effects associated with cognitive impairment using electroencephalography (EEG) power mapping allows the visualization of frequency-band specific local changes in oscillatory activity. In contrast, measures of coherence and dynamic source synchronization allow for the study of functional and effective connectivity, respectively. Yet, these measures have rarely been assessed in parallel in the context of mild cognitive impairment (MCI) and furthermore it has not been examined if they are related to risk factors of Alzheimer’s disease (AD) such as amyloid deposition and apolipoprotein ε4 (ApoE) allele occurrence. Here, we investigated functional and directed connectivities with Renormalized Partial Directed Coherence (RPDC) in 17 healthy controls (HC) and 17 participants with MCI. Participants underwent ApoE-genotyping and Pittsburgh compound B positron emission tomography (PiB-PET) to assess amyloid deposition. We observed lower spectral source power in MCI in the alpha and beta bands. Coherence was stronger in HC than MCI across different neuronal sources in the delta, theta, alpha, beta and gamma bands. The directed coherence analysis indicated lower information flow between fronto-temporal (including the hippocampus) sources and unidirectional connectivity in MCI. In MCI, alpha and beta RPDC showed an inverse correlation to age and gender; global amyloid deposition was inversely correlated to alpha coherence, RPDC and beta and gamma coherence. Furthermore, the ApoE status was negatively correlated to alpha coherence and RPDC, beta RPDC and gamma coherence. A classification analysis of cognitive state revealed the highest accuracy using EEG power, coherence and RPDC as input. For this small but statistically robust (Bayesian power analyses) sample, our results suggest that resting EEG related functional and directed connectivities are sensitive to the cognitive state and are linked to ApoE and amyloid burden.
Collapse
Affiliation(s)
- Lars Michels
- Clinic of Neuroradiology, University Hospital of ZurichZurich, Switzerland.,MR-Center, University Children's Hospital ZurichZurich, Switzerland
| | - Muthuraman Muthuraman
- Clinic for Neurology, University of KielKiel, Germany.,Clinic for Neurology, University of MainzMainz, Germany
| | - Abdul R Anwar
- Clinic for Neurology, University of KielKiel, Germany
| | - Spyros Kollias
- Clinic of Neuroradiology, University Hospital of ZurichZurich, Switzerland
| | - Sandra E Leh
- Division of Psychiatry Research and Psychogeriatric Medicine, University of ZurichZurich, Switzerland
| | - Florian Riese
- Division of Psychiatry Research and Psychogeriatric Medicine, University of ZurichZurich, Switzerland
| | - Paul G Unschuld
- Division of Psychiatry Research and Psychogeriatric Medicine, University of ZurichZurich, Switzerland
| | - Michael Siniatchkin
- Institute of Medical Psychology and Medical Sociology, Christian-Albrechts-University of KielKiel, Germany
| | - Anton F Gietl
- Division of Psychiatry Research and Psychogeriatric Medicine, University of ZurichZurich, Switzerland
| | - Christoph Hock
- Division of Psychiatry Research and Psychogeriatric Medicine, University of ZurichZurich, Switzerland
| |
Collapse
|
558
|
Ding SL, Royall JJ, Sunkin SM, Ng L, Facer BAC, Lesnar P, Guillozet-Bongaarts A, McMurray B, Szafer A, Dolbeare TA, Stevens A, Tirrell L, Benner T, Caldejon S, Dalley RA, Dee N, Lau C, Nyhus J, Reding M, Riley ZL, Sandman D, Shen E, van der Kouwe A, Varjabedian A, Wright M, Zöllei L, Dang C, Knowles JA, Koch C, Phillips JW, Sestan N, Wohnoutka P, Zielke HR, Hohmann JG, Jones AR, Bernard A, Hawrylycz MJ, Hof PR, Fischl B, Lein ES. Comprehensive cellular-resolution atlas of the adult human brain. J Comp Neurol 2017; 524:3127-481. [PMID: 27418273 PMCID: PMC5054943 DOI: 10.1002/cne.24080] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022]
Abstract
Detailed anatomical understanding of the human brain is essential for unraveling its functional architecture, yet current reference atlases have major limitations such as lack of whole‐brain coverage, relatively low image resolution, and sparse structural annotation. We present the first digital human brain atlas to incorporate neuroimaging, high‐resolution histology, and chemoarchitecture across a complete adult female brain, consisting of magnetic resonance imaging (MRI), diffusion‐weighted imaging (DWI), and 1,356 large‐format cellular resolution (1 µm/pixel) Nissl and immunohistochemistry anatomical plates. The atlas is comprehensively annotated for 862 structures, including 117 white matter tracts and several novel cyto‐ and chemoarchitecturally defined structures, and these annotations were transferred onto the matching MRI dataset. Neocortical delineations were done for sulci, gyri, and modified Brodmann areas to link macroscopic anatomical and microscopic cytoarchitectural parcellations. Correlated neuroimaging and histological structural delineation allowed fine feature identification in MRI data and subsequent structural identification in MRI data from other brains. This interactive online digital atlas is integrated with existing Allen Institute for Brain Science gene expression atlases and is publicly accessible as a resource for the neuroscience community. J. Comp. Neurol. 524:3127–3481, 2016. © 2016 The Authors The Journal of Comparative Neurology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Song-Lin Ding
- Allen Institute for Brain Science, Seattle, Washington, 98109.
| | - Joshua J Royall
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Susan M Sunkin
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | | | - Phil Lesnar
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | | | - Bergen McMurray
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Aaron Szafer
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Tim A Dolbeare
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Allison Stevens
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | - Lee Tirrell
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | - Thomas Benner
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | | | - Rachel A Dalley
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Christopher Lau
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Melissa Reding
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Zackery L Riley
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - David Sandman
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Elaine Shen
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Andre van der Kouwe
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | - Ani Varjabedian
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | - Michelle Wright
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | - Lilla Zöllei
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | - Chinh Dang
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - James A Knowles
- Zilkha Neurogenetic Institute, and Department of Psychiatry, University of Southern California, Los Angeles, California, 90033
| | - Christof Koch
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - John W Phillips
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Nenad Sestan
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, Connecticut, 06510
| | - Paul Wohnoutka
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - H Ronald Zielke
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - John G Hohmann
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Allan R Jones
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | - Amy Bernard
- Allen Institute for Brain Science, Seattle, Washington, 98109
| | | | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 11029
| | - Bruce Fischl
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, 02129
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, Washington, 98109.
| |
Collapse
|
559
|
Lidzba K, Küpper H, Kluger G, Staudt M. The time window for successful right-hemispheric language reorganization in children. Eur J Paediatr Neurol 2017. [PMID: 28648758 DOI: 10.1016/j.ejpn.2017.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
AIM To identify, in a retrospective, observational study, the time window during which successful right-hemispheric language reorganization is possible after left-hemispheric brain damage. METHOD 25 patients (10 females; age 6-41 years; ≥12 months after insult; age at insult 0;3-15;11 years) with acute, language-relevant left-hemispheric insults acquired during childhood and adolescence completed questionnaires for self-assessment of language problems. 12 patients of those reporting no (n = 8) or only moderate (n = 4) language problems participated in language fMRI. RESULTS Language outcome of lesions occurring before 5 years of age (n = 7) was always favorable, and language was right-lateralized (2 patients: age at lesion < 2 years) or bilateral (3 patients: age at lesion 2-5 years). Following lesions occurring after 5 years of age, language outcome was often unfavorable (11/18 patients: moderate or severe problems), and of the 7 patients without problems, none showed right-hemispheric reorganization (fMRI available in 4). INTERPRETATION The combination of normal language outcome and right-hemispheric language reorganization after a left-hemispheric lesion sustained after the neonatal period is extremely rare. Functionally sufficient right-hemispheric language was documented in only two patients with lesions acquired before two years of age.
Collapse
Affiliation(s)
- Karen Lidzba
- Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany.
| | - Hanna Küpper
- Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany
| | - Gerhard Kluger
- Clinic for Neuropediatrics and Neurorehabilitation, Epilepsy Center for Children and Adolescents, Schön Klinik, Krankenhausstraße 20, 83569 Vogtareuth, Germany; Institute of Rehabilitation, Transition and Palliation of Children with Neurological Illnesses, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria
| | - Martin Staudt
- Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital, Hoppe-Seyler-Str. 1, 72076 Tübingen, Germany; Clinic for Neuropediatrics and Neurorehabilitation, Epilepsy Center for Children and Adolescents, Schön Klinik, Krankenhausstraße 20, 83569 Vogtareuth, Germany
| |
Collapse
|
560
|
Alves IL, Meles SK, Willemsen ATM, Dierckx RA, Marques da Silva AM, Leenders KL, Koole M. Dual time point method for the quantification of irreversible tracer kinetics: A reference tissue approach applied to [ 18F]-FDOPA brain PET. J Cereb Blood Flow Metab 2017; 37:3124-3134. [PMID: 28156211 PMCID: PMC5584692 DOI: 10.1177/0271678x16684137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The Patlak graphical analysis (PGAREF) for quantification of irreversible tracer binding with a reference tissue model was approximated by a dual time point imaging approach (DTPREF). The DTPREF was applied to 18 [18F]-FDOPA brain scans using the occipital cortex as reference region (DTPOCC) and compared to both PGAOCC and striatal-to-occipital ratios (SOR). Pearson correlation analysis and Bland-Altman plots showed an excellent correlation and good agreement between DTPOCC and PGAOCC, while correlations between SOR and PGAOCC were consistently lower. Linear discriminant analysis (LDA) demonstrated a similar performance for all methods in differentiating patients with Parkinson's disease (PD) from healthy controls (HC). Specifically for [18F]-FDOPA brain imaging, these findings validate DTPOCC as an approximation for PGAOCC, providing the same quantitative information while reducing the acquisition time to two short static scans. For PD patients, this approach can greatly improve patient comfort while reducing motion artifacts and increasing image quality. In general, DTPREF can improve the clinical applicability of tracers with irreversible binding characteristics when a reference tissue is available.
Collapse
Affiliation(s)
- Isadora L Alves
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Sanne K Meles
- Department of Neurology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Antoon TM Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Rudi A Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Ana M Marques da Silva
- Laboratory of Medical Imaging, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Klaus L Leenders
- Department of Neurology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Michel Koole
- Department of Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
- Michel Koole, KU Leuven, Herestraat 49 - Bus 7003, B-3000 Leuven, Belgium.
| |
Collapse
|
561
|
Parbo P, Ismail R, Hansen KV, Amidi A, Mårup FH, Gottrup H, Brændgaard H, Eriksson BO, Eskildsen SF, Lund TE, Tietze A, Edison P, Pavese N, Stokholm MG, Borghammer P, Hinz R, Aanerud J, Brooks DJ. Brain inflammation accompanies amyloid in the majority of mild cognitive impairment cases due to Alzheimer's disease. Brain 2017; 140:2002-2011. [PMID: 28575151 DOI: 10.1093/brain/awx120] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/12/2017] [Indexed: 01/19/2023] Open
Abstract
See Kreisl (doi:10.1093/awx151) for a scientific commentary on this article.Subjects with mild cognitive impairment associated with cortical amyloid-β have a greatly increased risk of progressing to Alzheimer's disease. We hypothesized that neuroinflammation occurs early in Alzheimer's disease and would be present in most amyloid-positive mild cognitive impairment cases. 11C-Pittsburgh compound B and 11C-(R)-PK11195 positron emission tomography was used to determine the amyloid load and detect the extent of neuroinflammation (microglial activation) in 42 mild cognitive impairment cases. Twelve age-matched healthy control subjects had 11C-Pittsburgh compound B and 10 healthy control subjects had 11C-(R)-PK11195 positron emission tomography for comparison. Amyloid-positivity was defined as 11C-Pittsburgh compound B target-to-cerebellar ratio above 1.5 within a composite cortical volume of interest. Supervised cluster analysis was used to generate parametric maps of 11C-(R)-PK11195 binding potential. Levels of 11C-(R)-PK11195 binding potential were measured in a selection of cortical volumes of interest and at a voxel level. Twenty-six (62%) of 42 mild cognitive impairment cases showed a raised cortical amyloid load compared to healthy controls. Twenty-two (85%) of the 26 amyloid-positive mild cognitive impairment cases showed clusters of increased cortical microglial activation accompanying the amyloid. There was a positive correlation between levels of amyloid load and 11C-(R)-PK11195 binding potentials at a voxel level within subregions of frontal, parietal and temporal cortices. 11C-(R)-PK11195 positron emission tomography reveals increased inflammation in a majority of amyloid positive mild cognitive impairment cases, its cortical distribution overlapping that of amyloid deposition.
Collapse
Affiliation(s)
- Peter Parbo
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Kim V Hansen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Ali Amidi
- Department of Psychology and Behavioural Sciences and Department of Oncology Aarhus University and Aarhus University Hospital, Denmark
| | - Frederik H Mårup
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Hanne Gottrup
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Hans Brændgaard
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, Aarhus, Denmark
| | - Torben E Lund
- Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, Aarhus, Denmark
| | - Anna Tietze
- Institute of Neuroradiology, Charité, Universitätsmedizin, Berlin, Germany
| | - Paul Edison
- Division of Neuroscience, Department of Medicine, Imperial College London, London, UK
| | - Nicola Pavese
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Division of Neuroscience, Newcastle University, Newcastle, UK
| | - Morten G Stokholm
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Per Borghammer
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Joel Aanerud
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus, Denmark.,Division of Neuroscience, Department of Medicine, Imperial College London, London, UK.,Division of Neuroscience, Newcastle University, Newcastle, UK
| |
Collapse
|
562
|
Wild HM, Heckemann RA, Studholme C, Hammers A. Gyri of the human parietal lobe: Volumes, spatial extents, automatic labelling, and probabilistic atlases. PLoS One 2017; 12:e0180866. [PMID: 28846692 PMCID: PMC5573296 DOI: 10.1371/journal.pone.0180866] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 06/22/2017] [Indexed: 01/16/2023] Open
Abstract
Accurately describing the anatomy of individual brains enables interlaboratory communication of functional and developmental studies and is crucial for possible surgical interventions. The human parietal lobe participates in multimodal sensory integration including language processing and also contains the primary somatosensory area. We describe detailed protocols to subdivide the parietal lobe, analyze morphological and volumetric characteristics, and create probabilistic atlases in MNI152 stereotaxic space. The parietal lobe was manually delineated on 3D T1 MR images of 30 healthy subjects and divided into four regions: supramarginal gyrus (SMG), angular gyrus (AG), superior parietal lobe (supPL) and postcentral gyrus (postCG). There was the expected correlation of male gender with larger brain and intracranial volume. We examined a wide range of anatomical features of the gyri and the sulci separating them. At least a rudimentary primary intermediate sulcus of Jensen (PISJ) separating SMG and AG was identified in nearly all (59/60) hemispheres. Presence of additional gyri in SMG and AG was related to sulcal features and volumetric characteristics. The parietal lobe was slightly (2%) larger on the left, driven by leftward asymmetries of the postCG and SMG. Intersubject variability was highest for SMG and AG, and lowest for postCG. Overall the morphological characteristics tended to be symmetrical, and volumes also tended to covary between hemispheres. This may reflect developmental as well as maturation factors. To assess the accuracy with which the labels can be used to segment newly acquired (unlabelled) T1-weighted brain images, we applied multi-atlas label propagation software (MAPER) in a leave-one-out experiment and compared the resulting automatic labels with the manually prepared ones. The results showed strong agreement (mean Jaccard index 0.69, corresponding to a mean Dice index of 0.82, average mean volume error of 0.6%). Stereotaxic probabilistic atlases of each subregion were obtained. They illustrate the physiological brain torque, with structures in the right hemisphere positioned more anteriorly than in the left, and right/left positional differences of up to 10 mm. They also allow an assessment of sulcal variability, e.g. low variability for parietooccipital fissure and cingulate sulcus. Illustrated protocols, individual label sets, probabilistic atlases, and a maximum-probability atlas which takes into account surrounding structures are available for free download under academic licences.
Collapse
Affiliation(s)
- Heather M. Wild
- Neurodis Foundation, Lyon, France
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Rolf A. Heckemann
- Neurodis Foundation, Lyon, France
- MedTech West at Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Colin Studholme
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
| | - Alexander Hammers
- Neurodis Foundation, Lyon, France
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
563
|
Theruvath AJ, Ilivitzki A, Muehe A, Theruvath J, Gulaka P, Kim C, Luna-Fineman S, Sakamoto KM, Yeom KW, Yang P, Moseley M, Chan F, Daldrup-Link HE. A PET/MR Imaging Approach for the Integrated Assessment of Chemotherapy-induced Brain, Heart, and Bone Injuries in Pediatric Cancer Survivors: A Pilot Study. Radiology 2017; 285:971-979. [PMID: 28777701 DOI: 10.1148/radiol.2017170073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose To develop a positron emission tomography (PET)/magnetic resonance (MR) imaging protocol for evaluation of the brain, heart, and joints of pediatric cancer survivors for chemotherapy-induced injuries in one session. Materials and Methods Three teams of experts in neuroimaging, cardiac imaging, and bone imaging were tasked to develop a 20-30-minute PET/MR imaging protocol for detection of chemotherapy-induced tissue injuries of the brain, heart, and bone. In an institutional review board-approved, HIPAA-compliant, prospective study from April to July 2016, 10 pediatric cancer survivors who completed chemotherapy underwent imaging of the brain, heart, and bone with a 3-T PET/MR imager. Cumulative chemotherapy doses and clinical symptoms were correlated with the severity of MR imaging abnormalities by using linear regression analyses. MR imaging measures of brain perfusion and metabolism were compared among eight patients who were treated with methotrexate and eight untreated age-matched control subjects by using Wilcoxon rank-sum tests. Results Combined brain, heart, and bone examinations were completed within 90 minutes. Eight of 10 cancer survivors had abnormal findings on brain, heart, and bone images, including six patients with and two patients without clinical symptoms. Cumulative chemotherapy doses correlated significantly with MR imaging measures of left ventricular ejection fraction and end-systolic volume, but not with the severity of brain or bone abnormalities. Methotrexate-treated cancer survivors had significantly lower cerebral blood flow and metabolic activity in key brain areas compared with control subjects. Conclusion The feasibility of a single examination for assessment of chemotherapy-induced injuries of the brain, heart, and joints was shown. Earlier detection of tissue injuries may enable initiation of timely interventions and help to preserve long-term health of pediatric cancer survivors. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ashok J Theruvath
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Anat Ilivitzki
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Anne Muehe
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Johanna Theruvath
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Praveen Gulaka
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Christine Kim
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Sandra Luna-Fineman
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Kathleen M Sakamoto
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Kristen W Yeom
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Phillip Yang
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Michael Moseley
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Frandics Chan
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| | - Heike E Daldrup-Link
- From the Department of Radiology and the Molecular Imaging Program (A.J.T., A.I., A.M., J.T., P.G., C.K., K.W.Y., M.M., F.C., H.E.D.L.), Department of Pediatrics, Division of Pediatric Hematology/Oncology (S.L.F., K.M.S., H.E.D.L.), and Department of Medicine, Division of Cardiology (P.Y.), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Rd, Stanford, CA 94305-5654
| |
Collapse
|
564
|
Lambert B, Declerck CH, Boone C, Parizel PM. A functional MRI study on how oxytocin affects decision making in social dilemmas: Cooperate as long as it pays off, aggress only when you think you can win. Horm Behav 2017; 94:145-152. [PMID: 28676252 DOI: 10.1016/j.yhbeh.2017.06.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 06/15/2017] [Accepted: 06/29/2017] [Indexed: 02/02/2023]
Abstract
We investigate if the neuropeptide oxytocin (OT), known to moderate social behaviour, influences strategic decision making in social dilemmas by facilitating the integration of incentives and social cues. Participants (N=29) played two economic games with different incentive structures in the fMRI scanner after receiving OT or placebo (following a double blind, within-subject design). Pictures of angry or neutral faces (the social cues) were displayed alongside the game matrices. Consistent with a priori hypotheses based on the modulatory role of OT in mesolimbic dopaminergic brain regions, the results indicate that, compared to placebo, OT significantly increases the activation of the nucleus accumbens during an assurance (coordination) game that rewards mutual cooperation. This increases appetitive motivation so that cooperative behaviour is facilitated for risk averse individuals. OT also significantly attenuates the amygdala, thereby reducing the orienting response to social cues. The corresponding change in behaviour is only apparent in the chicken (or anti-coordination) game, where aggression is incentivized but fatal if the partner also aggresses. Because of this ambiguity, decision making can be improved by additional information, and OT steers decisions in the chicken game in accordance with the valence of the facial cue: aggress when face is neutral; retreat when it is angry. Through its combined influence on amygdala and nucleus accumbens, OT improves the selection of a cooperative or aggressive strategy in function of the best match between the incentives of the game and the social cues present in the decision environment.
Collapse
Affiliation(s)
- Bruno Lambert
- Faculty of economic sciences, University of Antwerp, Prinsstraat 13, 2000 Antwerp, Belgium.
| | - Carolyn H Declerck
- Faculty of economic sciences, University of Antwerp, Prinsstraat 13, 2000 Antwerp, Belgium
| | - Christophe Boone
- Faculty of economic sciences, University of Antwerp, Prinsstraat 13, 2000 Antwerp, Belgium
| | - Paul M Parizel
- Department of radiology, Antwerp University Hospital and University of Antwerp, Wilrijkstraat 10, 2650 Antwerp (Edegem), Belgium
| |
Collapse
|
565
|
Bernard-Gauthier V, Bailey JJ, Mossine AV, Lindner S, Vomacka L, Aliaga A, Shao X, Quesada CA, Sherman P, Mahringer A, Kostikov A, Grand’Maison M, Rosa-Neto P, Soucy JP, Thiel A, Kaplan DR, Fricker G, Wängler B, Bartenstein P, Schirrmacher R, Scott PJH. A Kinome-Wide Selective Radiolabeled TrkB/C Inhibitor for in Vitro and in Vivo Neuroimaging: Synthesis, Preclinical Evaluation, and First-in-Human. J Med Chem 2017; 60:6897-6910. [DOI: 10.1021/acs.jmedchem.7b00396] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Vadim Bernard-Gauthier
- Department of Oncology,
Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Justin J. Bailey
- Department of Oncology,
Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Andrew V. Mossine
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Simon Lindner
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, Munich 81377, Germany
| | - Lena Vomacka
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, Munich 81377, Germany
| | - Arturo Aliaga
- Translational Neuroimaging Laboratory, McGill Centre
for Studies in Aging, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, Quebec H4H 1R3, Canada
| | - Xia Shao
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Carole A. Quesada
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Phillip Sherman
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Anne Mahringer
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg 69120, Germany
| | - Alexey Kostikov
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | | | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre
for Studies in Aging, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, Quebec H4H 1R3, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Alexander Thiel
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
- Jewish General Hospital, Lady Davis Institute, Montreal, Quebec HT3 1E2, Canada
| | - David R. Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular
Genetics, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg 69120, Germany
| | - Björn Wängler
- Molecular
Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear
Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer
1-3, Mannheim 68167, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, Munich 81377, Germany
| | - Ralf Schirrmacher
- Department of Oncology,
Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Peter J. H. Scott
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- The Interdepartmental Program in Medicinal
Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
566
|
Lassen ML, Muzik O, Beyer T, Hacker M, Ladefoged CN, Cal-González J, Wadsak W, Rausch I, Langer O, Bauer M. Reproducibility of Quantitative Brain Imaging Using a PET-Only and a Combined PET/MR System. Front Neurosci 2017; 11:396. [PMID: 28769742 PMCID: PMC5511842 DOI: 10.3389/fnins.2017.00396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/23/2017] [Indexed: 01/02/2023] Open
Abstract
The purpose of this study was to test the feasibility of migrating a quantitative brain imaging protocol from a positron emission tomography (PET)-only system to an integrated PET/MR system. Potential differences in both absolute radiotracer concentration as well as in the derived kinetic parameters as a function of PET system choice have been investigated. Five healthy volunteers underwent dynamic (R)-[11C]verapamil imaging on the same day using a GE-Advance (PET-only) and a Siemens Biograph mMR system (PET/MR). PET-emission data were reconstructed using a transmission-based attenuation correction (AC) map (PET-only), whereas a standard MR-DIXON as well as a low-dose CT AC map was applied to PET/MR emission data. Kinetic modeling based on arterial blood sampling was performed using a 1-tissue-2-rate constant compartment model, yielding kinetic parameters (K1 and k2) and distribution volume (V T ). Differences for parametric values obtained in the PET-only and the PET/MR systems were analyzed using a 2-way Analysis of Variance (ANOVA). Comparison of DIXON-based AC (PET/MR) with emission data derived from the PET-only system revealed average inter-system differences of -33 ± 14% (p < 0.05) for the K1 parameter and -19 ± 9% (p < 0.05) for k2. Using a CT-based AC for PET/MR resulted in slightly lower systematic differences of -16 ± 18% for K1 and -9 ± 10% for k2. The average differences in V T were -18 ± 10% (p < 0.05) for DIXON- and -8 ± 13% for CT-based AC. Significant systematic differences were observed for kinetic parameters derived from emission data obtained from PET/MR and PET-only imaging due to different standard AC methods employed. Therefore, a transfer of imaging protocols from PET-only to PET/MR systems is not straightforward without application of proper correction methods. Clinical Trial Registration: www.clinicaltrialsregister.eu, identifier 2013-001724-19.
Collapse
Affiliation(s)
- Martin L Lassen
- Center for Medical Physics and Biomedical Engineering, Medical University of ViennaVienna, Austria
| | - Otto Muzik
- Department of Radiology, Detroit Medical Center, Children's Hospital of Michigan, Wayne State University School of MedicineDetroit, MI, United States
| | - Thomas Beyer
- Center for Medical Physics and Biomedical Engineering, Medical University of ViennaVienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of ViennaVienna, Austria
| | - Claes Nøhr Ladefoged
- Department of Clinical Physiology, Nuclear Medicine and PETRigshospitalet, Copenhagen, Denmark
| | - Jacobo Cal-González
- Center for Medical Physics and Biomedical Engineering, Medical University of ViennaVienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of ViennaVienna, Austria.,CBmed GmbH, Center for Biomarker Research in MedicineGraz, Austria
| | - Ivo Rausch
- Center for Medical Physics and Biomedical Engineering, Medical University of ViennaVienna, Austria
| | - Oliver Langer
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of ViennaVienna, Austria.,Department for Clinical Pharmacology, Medical University of ViennaVienna, Austria.,Health and Environment Department, AIT Austrian Institute of Technology GmbHSeibersdorf, Austria
| | - Martin Bauer
- Department for Clinical Pharmacology, Medical University of ViennaVienna, Austria
| |
Collapse
|
567
|
Buzzell GA, Richards JE, White LK, Barker TV, Pine DS, Fox NA. Development of the error-monitoring system from ages 9-35: Unique insight provided by MRI-constrained source localization of EEG. Neuroimage 2017; 157:13-26. [PMID: 28549796 DOI: 10.1016/j.neuroimage.2017.05.045] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 04/24/2017] [Accepted: 05/19/2017] [Indexed: 11/18/2022] Open
Abstract
The ability to self-detect errors and dynamically adapt behavior is a cornerstone of higher-level cognition, requiring coordinated activity from a network of neural regions. However, disagreement exists over how the error-monitoring system develops throughout adolescence and early adulthood. The present report leveraged MRI-constrained EEG source localization to detail typical development of the error-monitoring system in a sample of 9-35 year-olds (n = 43). Participants performed a flanker task while high-density EEG was recorded; structural MRIs were also acquired for all participants. Analysis of the scalp-recorded EEG data revealed a frontocentral negativity (error-related negativity; ERN) immediately following errors for all participants, although the topography of the ERN varied with age. Source localization of the ERN time range revealed maximal activity within the posterior cingulate cortex (PCC) for all ages, consistent with recent evidence that the PCC provides a substantial contribution to the scalp-recorded ERN. Activity within a network of brain regions, including dorsal anterior cingulate, PCC, and parietal cortex, was predictive of improved performance following errors, regardless of age. However, additional activity within insula, orbitofrontal cortex and inferior frontal gyrus linearly increased with age. Together, these data suggest that the core error-monitoring system is online by early adolescence and remains relatively stable into adulthood. However, additional brain regions become embedded within this core network with age. These results serve as a model of typical development of the error-monitoring system from early adolescence into adulthood.
Collapse
Affiliation(s)
- George A Buzzell
- Department of Human Development and Quantitative Methodology, University of Maryland, College Park, MD 20742, United States.
| | - John E Richards
- Department of Psychology, University of South Carolina, Columbia, SC 29208, United States
| | - Lauren K White
- Department of Child and Adolescent Psychiatry, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Tyson V Barker
- Department of Human Development and Quantitative Methodology, University of Maryland, College Park, MD 20742, United States
| | - Daniel S Pine
- Emotion and Development Branch, Intramural Research Program, National Institute of Mental Health, Bethesda, MD 20814, United States
| | - Nathan A Fox
- Department of Human Development and Quantitative Methodology, University of Maryland, College Park, MD 20742, United States
| |
Collapse
|
568
|
McGinnity CJ, Riaño Barros DA, Rosso L, Veronese M, Rizzo G, Bertoldo A, Hinz R, Turkheimer FE, Koepp MJ, Hammers A. Test-retest reproducibility of quantitative binding measures of [ 11C]Ro15-4513, a PET ligand for GABA A receptors containing alpha5 subunits. Neuroimage 2017; 152:270-282. [PMID: 28292717 PMCID: PMC5440177 DOI: 10.1016/j.neuroimage.2016.12.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/20/2016] [Accepted: 12/14/2016] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Alteration of γ-aminobutyric acid "A" (GABAA) receptor-mediated neurotransmission has been associated with various neurological and psychiatric disorders. [11C]Ro15-4513 is a PET ligand with high affinity for α5-subunit-containing GABAA receptors, which are highly expressed in limbic regions of the human brain (Sur et al., 1998). We quantified the test-retest reproducibility of measures of [11C]Ro15-4513 binding derived from six different quantification methods (12 variants). METHODS Five healthy males (median age 40 years, range 38-49 years) had a 90-min PET scan on two occasions (median interval 12 days, range 11-30 days), after injection of a median dose of 441 MegaBequerels of [11C]Ro15-4513. Metabolite-corrected arterial plasma input functions (parent plasma input functions, ppIFs) were generated for all scans. We quantified regional binding using six methods (12 variants), some of which were region-based (applied to the average time-activity curve within a region) and others were voxel-based: 1) Models requiring arterial ppIFs - regional reversible compartmental models with one and two tissue compartments (2kbv and 4kbv); 2) Regional and voxelwise Logan's graphical analyses (Logan et al., 1990), which required arterial ppIFs; 3) Model-free regional and voxelwise (exponential) spectral analyses (SA; (Cunningham and Jones, 1993)), which also required arterial ppIFs; 4) methods not requiring arterial ppIFs - voxelwise standardised uptake values (Kenney et al., 1941), and regional and voxelwise simplified reference tissue models (SRTM/SRTM2) using brainstem or alternatively cerebellum as pseudo-reference regions (Lammertsma and Hume, 1996; Gunn et al., 1997). To compare the variants, we sampled the mean values of the outcome parameters within six bilateral, non-reference grey matter regions-of-interest. Reliability was quantified in terms of median absolute percentage test-retest differences (MA-TDs; preferentially low) and between-subject coefficient of variation (BS-CV, preferentially high), both compounded by the intraclass correlation coefficient (ICC). These measures were compared between variants, with particular interest in the hippocampus. RESULTS Two of the six methods (5/12 variants) yielded reproducible data (i.e. MA-TD <10%): regional SRTMs and voxelwise SRTM2s, both using either the brainstem or the cerebellum; and voxelwise SA. However, the SRTMs using the brainstem yielded a lower median BS-CV (7% for regional, 7% voxelwise) than the other variants (8-11%), resulting in lower ICCs. The median ICCs across six regions were 0.89 (interquartile range 0.75-0.90) for voxelwise SA, 0.71 (0.64-0.84) for regional SRTM-cerebellum and 0.83 (0.70-0.86) for voxelwise SRTM-cerebellum. The ICCs for the hippocampus were 0.89 for voxelwise SA, 0.95 for regional SRTM-cerebellum and 0.93 for voxelwise SRTM-cerebellum. CONCLUSION Quantification of [11C]Ro15-4513 binding shows very good to excellent reproducibility with SRTM and with voxelwise SA which, however, requires an arterial ppIF. Quantification in the α5 subunit-rich hippocampus is particularly reliable. The very low expression of the α5 in the cerebellum (Fritschy and Mohler, 1995; Veronese et al., 2016) and the substantial α1 subunit density in this region may hamper the application of reference tissue methods.
Collapse
Affiliation(s)
- Colm J McGinnity
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK; Division of Imaging Sciences & Biomedical Engineering, King's College London, London, UK.
| | - Daniela A Riaño Barros
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | - Lula Rosso
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Gaia Rizzo
- Department of Information Engineering, University of Padova, Padova, Italy
| | | | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Federico E Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Matthias J Koepp
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, UK; Epilepsy Society, Chalfont St Peter, UK
| | - Alexander Hammers
- Centre for Neuroscience, Department of Medicine, Imperial College London, London, UK; Medical Research Council Clinical Sciences Centre, Hammersmith Hospital, London, UK; Division of Imaging Sciences & Biomedical Engineering, King's College London, London, UK; The Neurodis Foundation, CERMEP - Imagerie du Vivant, Lyon, France
| |
Collapse
|
569
|
Höglinger GU, Schöpe J, Stamelou M, Kassubek J, Del Ser T, Boxer AL, Wagenpfeil S, Huppertz HJ. Longitudinal magnetic resonance imaging in progressive supranuclear palsy: A new combined score for clinical trials. Mov Disord 2017; 32:842-852. [PMID: 28436538 DOI: 10.1002/mds.26973] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/16/2017] [Accepted: 02/19/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Two recent, randomized, placebo-controlled phase II/III trials (clinicaltrials.gov: NCT01110720, NCT01049399) of davunetide and tideglusib in progressive supranuclear palsy (PSP) generated prospective, 1-year longitudinal datasets of high-resolution T1-weighted three-dimensional MRI. OBJECTIVE The objective of this study was to develop a quantitative MRI disease progression measurement for clinical trials. METHODS The authors performed a fully automated quantitative MRI analysis employing atlas-based volumetry and provide sample size calculations based on data collected in 99 PSP patients assigned to placebo in these trials. Based on individual volumes of 44 brain compartments and structures at baseline and 52 weeks of follow-up, means and standard deviations of annualized percentage volume changes were used to estimate standardized effect sizes and the required sample sizes per group for future 2-armed, placebo-controlled therapeutic trials. RESULTS The highest standardized effect sizes were found for midbrain, frontal lobes, and the third ventricle. Using the annualized percentage volume change of these structures to detect a 50% change in the 1-year progression (80% power, significance level 5%) required lower numbers of patients per group (third ventricle, n = 32; midbrain, n = 37; frontal lobe, n = 43) than the best clinical scale (PSP rating scale total score, n = 58). A combination of volume changes in these 3 structures reduced the number of required patients to only 20 and correlated best with the progression in the clinical scales. CONCLUSIONS We propose the 1-year change in the volumes of third ventricle, midbrain, and frontal lobe as combined imaging read-out for clinical trials in PSP that require the least number of patients for detecting efficacy to reduce brain atrophy. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Günter U Höglinger
- Department of Neurology, Technische Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany.,Department of Neurology, University Hospital Gießen and Marburg, Marburg, Germany
| | - Jakob Schöpe
- Institute for Medical Biometry, Epidemiology and Medical Informatics, Saarland University, Campus Homburg, Germany
| | - Maria Stamelou
- Department of Neurology, University Hospital Gießen and Marburg, Marburg, Germany.,Second Department of Neurology, Attikon University Hospital, University of Athens, Athens, Greece
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, USA
| | - Stefan Wagenpfeil
- Institute for Medical Biometry, Epidemiology and Medical Informatics, Saarland University, Campus Homburg, Germany
| | | | | | | | | |
Collapse
|
570
|
Golla SSV, Lubberink M, van Berckel BNM, Lammertsma AA, Boellaard R. Partial volume correction of brain PET studies using iterative deconvolution in combination with HYPR denoising. EJNMMI Res 2017; 7:36. [PMID: 28432674 PMCID: PMC5400775 DOI: 10.1186/s13550-017-0284-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 04/07/2017] [Indexed: 11/10/2022] Open
Abstract
Background Accurate quantification of PET studies depends on the spatial resolution of the PET data. The commonly limited PET resolution results in partial volume effects (PVE). Iterative deconvolution methods (IDM) have been proposed as a means to correct for PVE. IDM improves spatial resolution of PET studies without the need for structural information (e.g. MR scans). On the other hand, deconvolution also increases noise, which results in lower signal-to-noise ratios (SNR). The aim of this study was to implement IDM in combination with HighlY constrained back-PRojection (HYPR) denoising to mitigate poor SNR properties of conventional IDM. Methods An anthropomorphic Hoffman brain phantom was filled with an [18F]FDG solution of ~25 kBq mL−1 and scanned for 30 min on a Philips Ingenuity TF PET/CT scanner (Philips, Cleveland, USA) using a dynamic brain protocol with various frame durations ranging from 10 to 300 s. Van Cittert IDM was used for PVC of the scans. In addition, HYPR was used to improve SNR of the dynamic PET images, applying it both before and/or after IDM. The Hoffman phantom dataset was used to optimise IDM parameters (number of iterations, type of algorithm, with/without HYPR) and the order of HYPR implementation based on the best average agreement of measured and actual activity concentrations in the regions. Next, dynamic [11C]flumazenil (five healthy subjects) and [11C]PIB (four healthy subjects and four patients with Alzheimer’s disease) scans were used to assess the impact of IDM with and without HYPR on plasma input-derived distribution volumes (VT) across various regions of the brain. Results In the case of [11C]flumazenil scans, Hypr-IDM-Hypr showed an increase of 5 to 20% in the regional VT whereas a 0 to 10% increase or decrease was seen in the case of [11C]PIB depending on the volume of interest or type of subject (healthy or patient). References for these comparisons were the VTs from the PVE-uncorrected scans. Conclusions IDM improved quantitative accuracy of measured activity concentrations. Moreover, the use of IDM in combination with HYPR (Hypr-IDM-Hypr) was able to correct for PVE without increasing noise. Electronic supplementary material The online version of this article (doi:10.1186/s13550-017-0284-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sandeep S V Golla
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007MB, Amsterdam, The Netherlands.
| | - Mark Lubberink
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007MB, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007MB, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007MB, Amsterdam, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
571
|
Castellaro M, Rizzo G, Tonietto M, Veronese M, Turkheimer FE, Chappell MA, Bertoldo A. A Variational Bayesian inference method for parametric imaging of PET data. Neuroimage 2017; 150:136-149. [PMID: 28213113 DOI: 10.1016/j.neuroimage.2017.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/22/2017] [Accepted: 02/04/2017] [Indexed: 12/15/2022] Open
Abstract
In dynamic Positron Emission Tomography (PET) studies, compartmental models provide the richest information on the tracer kinetics of the tissue. Inverting such models at the voxel level is however quite challenging due to the low signal-to-noise ratio of the time activity curves. In this study, we propose the use of a Variational Bayesian (VB) approach to efficiently solve this issue and thus obtain robust quantitative parametric maps. VB was adapted to the non-uniform noise distribution of PET data. Moreover, we propose a novel hierarchical scheme to define the model parameter priors directly from the images in case such information are not available from the literature, as often happens with new PET tracers. VB was initially tested on synthetic data generated using compartmental models of increasing complexity, providing accurate (%bias<2%±2%, root mean square error<15%±5%) parameter estimates. When applied to real data on a paradigmatic set of PET tracers (L-[1-11C]leucine, [11C]WAY100635 and [18F]FDG), VB was able to generate reliable parametric maps even in presence of high noise in the data (unreliable estimates<11%±5%).
Collapse
Affiliation(s)
- M Castellaro
- Department of Information Engineering, University of Padova, Italy
| | - G Rizzo
- Department of Information Engineering, University of Padova, Italy
| | - M Tonietto
- Department of Information Engineering, University of Padova, Italy
| | - M Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - F E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - M A Chappell
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Old Road Campus, Headington, Oxford, United Kingdom
| | - A Bertoldo
- Department of Information Engineering, University of Padova, Italy.
| |
Collapse
|
572
|
Faillenot I, Heckemann RA, Frot M, Hammers A. Macroanatomy and 3D probabilistic atlas of the human insula. Neuroimage 2017; 150:88-98. [DOI: 10.1016/j.neuroimage.2017.01.073] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 12/16/2016] [Accepted: 01/30/2017] [Indexed: 11/28/2022] Open
|
573
|
Lambert B, Declerck CH, Emonds G, Boone C. Trust as commodity: social value orientation affects the neural substrates of learning to cooperate. Soc Cogn Affect Neurosci 2017; 12:609-617. [PMID: 28119509 PMCID: PMC5390759 DOI: 10.1093/scan/nsw170] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 09/16/2016] [Accepted: 11/24/2016] [Indexed: 12/02/2022] Open
Abstract
Individuals differ in their motives and strategies to cooperate in social dilemmas. These differences are reflected by an individual's social value orientation: proselfs are strategic and motivated to maximize self-interest, while prosocials are more trusting and value fairness. We hypothesize that when deciding whether or not to cooperate with a random member of a defined group, proselfs, more than prosocials, adapt their decisions based on past experiences: they 'learn' instrumentally to form a base-line expectation of reciprocity. We conducted an fMRI experiment where participants (19 proselfs and 19 prosocials) played 120 sequential prisoner's dilemmas against randomly selected, anonymous and returning partners who cooperated 60% of the time. Results indicate that cooperation levels increased over time, but that the rate of learning was steeper for proselfs than for prosocials. At the neural level, caudate and precuneus activation were more pronounced for proselfs relative to prosocials, indicating a stronger reliance on instrumental learning and self-referencing to update their trust in the cooperative strategy.
Collapse
|
574
|
Blautzik J, Brendel M, Sauerbeck J, Kotz S, Scheiwein F, Bartenstein P, Seibyl J, Rominger A. Reference region selection and the association between the rate of amyloid accumulation over time and the baseline amyloid burden. Eur J Nucl Med Mol Imaging 2017; 44:1364-1374. [PMID: 28326436 DOI: 10.1007/s00259-017-3666-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/24/2017] [Indexed: 10/19/2022]
Abstract
Relative quantitative analysis of amyloid plaque burden in Alzheimer's disease (AD) patients can be reported as standardized uptake value ratio (SUVR) from positron emission tomography (PET). Here, the SUVR is the ratio of the mean amyloid radioligand retention in a composite (COMP) neocortical volume of interest (VOI) to that in a reference VOI, such as the cerebellum, brainstem (BST)/pons, or white matter (WM). Some longitudinal PET investigations show that the rate of amyloid accumulation to follow-up has an inverted U relationship with baseline amyloid SUVR relative to cerebellar or brainstem/pons reference VOIs. The corresponding association with SUVR relative to WM is unknown. To test the possible benefits of WM normalization, we analyzed [18F]-AV45 PET data from 404 subjects in the AD Neuroimaging Initiative (ADNI) database at baseline and 2-year follow-up (144 cognitively normal controls, 225 patients with mild cognitive impairment, and 35 AD patients). Reference regions included subcortical WM as well as conventional cerebellar gray matter (CBL), and BST. We tested associations between each subject's inter-session change (∆) of SUVR and their baseline SUVR by applying linear, logarithmic, and quadratic regression analyses. Unscaled standardized uptake values (SUVs) were correlated between VOIs at baseline and follow-up, and within VOIs in the longitudinal run. The association between ∆SUVR and baseline SUVR relative to WM reference was best described by an inverted U-shaped function. Correlation analyses demonstrated a high regional and temporal correlation between COMP and WM VOI SUVs. For WM normalization, we confirm that the rate of amyloid accumulation over time follows an inverted U-shaped function of baseline amyloid burden. Reference region selection, however, has substantial effects on SUVR results. This reflects the extent of covariance between SUVs in the COMP VOI and those in the various reference VOIs. We speculate that PET labeling of amyloid deposition within target regions is partially confounded by effects of longitudinal changes of cerebral blood flow (CBF) on tracer delivery. Indeed, CBF may be the leading factor influencing longitudinal SUV changes. We suggest that SUVR relative to WM may be more robust to changes in CBF, and thus fitter for sensitive detection of amyloid accumulation in intervention studies.
Collapse
Affiliation(s)
- Janusch Blautzik
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | - Julia Sauerbeck
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | - Sebastian Kotz
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | | | - Axel Rominger
- Department of Nuclear Medicine, University of Munich, Munich, Germany.
| | | |
Collapse
|
575
|
Parkinson's disease: diagnostic utility of volumetric imaging. Neuroradiology 2017; 59:367-377. [PMID: 28303376 DOI: 10.1007/s00234-017-1808-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/08/2017] [Indexed: 10/20/2022]
Abstract
PURPOSE This paper aims to examine the effectiveness of structural imaging as an aid in the diagnosis of Parkinson's disease (PD). METHODS High-resolution T 1-weighted magnetic resonance imaging was performed in 72 patients with idiopathic PD (mean age, 61.08 years) and 73 healthy subjects (mean age, 58.96 years). The whole brain was parcellated into 95 regions of interest using composite anatomical atlases, and region volumes were calculated. Three diagnostic classifiers were constructed using binary multiple logistic regression modeling: the (i) basal ganglion prior classifier, (ii) data-driven classifier, and (iii) basal ganglion prior/data-driven hybrid classifier. Leave-one-out cross validation was used to unbiasedly evaluate the predictive accuracy of imaging features. Pearson's correlation analysis was further performed to correlate outcome measurement using the best PD classifier with disease severity. RESULTS Smaller volume in susceptible regions is diagnostic for Parkinson's disease. Compared with the other two classifiers, the basal ganglion prior/data-driven hybrid classifier had the highest diagnostic reliability with a sensitivity of 74%, specificity of 75%, and accuracy of 74%. Furthermore, outcome measurement using this classifier was associated with disease severity. CONCLUSIONS Brain structural volumetric analysis with multiple logistic regression modeling can be a complementary tool for diagnosing PD.
Collapse
|
576
|
Mérida I, Reilhac A, Redouté J, Heckemann RA, Costes N, Hammers A. Multi-atlas attenuation correction supports full quantification of static and dynamic brain PET data in PET-MR. Phys Med Biol 2017; 62:2834-2858. [PMID: 28181479 DOI: 10.1088/1361-6560/aa5f6c] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
577
|
Hansen AK, Damholdt MF, Fedorova TD, Knudsen K, Parbo P, Ismail R, Østergaard K, Brooks DJ, Borghammer P. In Vivo cortical tau in Parkinson's disease using 18F-AV-1451 positron emission tomography. Mov Disord 2017; 32:922-927. [PMID: 28256006 DOI: 10.1002/mds.26961] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/22/2017] [Accepted: 01/27/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Alzheimer's disease copathology is common in PD at autopsy. In non-PD subjects with mild cognitive impairment, tau depositions can be detected using 18F-AV-1451 PET. We hypothesized that 18F-AV-1451 PET would show tau aggregation in PD with mild cognitive impairment and correlate with cognitive dysfunction. OBJECTIVES To describe tau aggregation in PD patients. METHODS Twenty-six PD patients and 23 controls had 18F-AV-1451 PET and neuropsychological assessment to detect mild cognitive impairment. RESULTS Nine PD patients (35%) were identified with mild cognitive impairment. Regional analyses showed no significant differences between groups. Voxel-wise analyses showed no correlation with cognitive domain z-scores within patients. One patient with mild cognitive impairment was estimated Braak tau stage 5; all other patients were stage 0. CONCLUSION Our results indicate that tau pathology, as detected by 18F-AV-1451, is uncommon in PD with mild cognitive impairment and shows no significant correlation with cognitive dysfunction at this stage. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Allan K Hansen
- Dept of Nuclear Medicine & PET Centre, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Malene Flensborg Damholdt
- Unit of Psychooncology and Health psychology, Dept of Psychology and Behavioral Sciences, Aarhus University, Aarhus, Denmark
| | - Tatyana D Fedorova
- Dept of Nuclear Medicine & PET Centre, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karoline Knudsen
- Dept of Nuclear Medicine & PET Centre, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Peter Parbo
- Dept of Nuclear Medicine & PET Centre, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rola Ismail
- Dept of Nuclear Medicine & PET Centre, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karen Østergaard
- Dept of Neurology, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Dept of Nuclear Medicine & PET Centre, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Division of Neuroscience, Dept of Medicine, Imperial College London, United Kingdom.,Division of Neuroscience, Newcastle University, United Kingdom
| | - Per Borghammer
- Dept of Nuclear Medicine & PET Centre, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
578
|
Egerton A, Howes OD, Houle S, McKenzie K, Valmaggia LR, Bagby MR, Tseng HH, Bloomfield MAP, Kenk M, Bhattacharyya S, Suridjan I, Chaddock CA, Winton-Brown TT, Allen P, Rusjan P, Remington G, Meyer-Lindenberg A, McGuire PK, Mizrahi R. Elevated Striatal Dopamine Function in Immigrants and Their Children: A Risk Mechanism for Psychosis. Schizophr Bull 2017; 43:293-301. [PMID: 28057720 PMCID: PMC5605255 DOI: 10.1093/schbul/sbw181] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Migration is a major risk factor for schizophrenia but the neurochemical processes involved are unknown. One candidate mechanism is through elevations in striatal dopamine synthesis and release. The objective of this research was to determine whether striatal dopamine function is elevated in immigrants compared to nonimmigrants and the relationship with psychosis. Two complementary case-control studies of in vivo dopamine function (stress-induced dopamine release and dopamine synthesis capacity) in immigrants compared to nonimmigrants were performed in Canada and the United Kingdom. The Canadian dopamine release study included 25 immigrant and 31 nonmigrant Canadians. These groups included 23 clinical high risk (CHR) subjects, 9 antipsychotic naïve patients with schizophrenia, and 24 healthy volunteers. The UK dopamine synthesis study included 32 immigrants and 44 nonimmigrant British. These groups included 50 CHR subjects and 26 healthy volunteers. Both striatal stress-induced dopamine release and dopamine synthesis capacity were significantly elevated in immigrants compared to nonimmigrants, independent of clinical status. These data provide the first evidence that the effect of migration on the risk of developing psychosis may be mediated by an elevation in brain dopamine function.
Collapse
Affiliation(s)
- Alice Egerton
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK;,These authors are joint first authors
| | - Oliver D. Howes
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK;,These authors are joint first authors
| | - Sylvain Houle
- Research Imaging Center, CAMH, PET Centre, Toronto, ON, Canada;,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada;,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada;,These authors are joint first authors
| | - Kwame McKenzie
- Research Imaging Center, CAMH, PET Centre, Toronto, ON, Canada;,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Lucia R. Valmaggia
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Michael R. Bagby
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada;,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada;,Department of Psychology, University of Toronto
| | - Huai-Hsuan Tseng
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK;,Research Imaging Center, CAMH, PET Centre, Toronto, ON, Canada
| | - Michael A. P. Bloomfield
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK;,Division of Psychiatry, University College London, London, UK;,Psychiatric Imaging Group, MRC Clinical Sciences Centre, Hammersmith Hospital, London, UK
| | - Miran Kenk
- Research Imaging Center, CAMH, PET Centre, Toronto, ON, Canada
| | - Sagnik Bhattacharyya
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Ivonne Suridjan
- Research Imaging Center, CAMH, PET Centre, Toronto, ON, Canada
| | | | - Toby T. Winton-Brown
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Paul Allen
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK;,Department of Psychology, Whitelands College, University of Roehampton, London, UK
| | - Pablo Rusjan
- Research Imaging Center, CAMH, PET Centre, Toronto, ON, Canada;,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Gary Remington
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada;,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Andreas Meyer-Lindenberg
- Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
| | - Philip K. McGuire
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK;,These authors are joint last authors
| | - Romina Mizrahi
- Research Imaging Center, CAMH, PET Centre, Toronto, ON, Canada;,Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada;,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada;,These authors are joint last authors
| |
Collapse
|
579
|
Passamonti L, Vázquez Rodríguez P, Hong YT, Allinson KSJ, Williamson D, Borchert RJ, Sami S, Cope TE, Bevan-Jones WR, Jones PS, Arnold R, Surendranathan A, Mak E, Su L, Fryer TD, Aigbirhio FI, O’Brien JT, Rowe JB. 18F-AV-1451 positron emission tomography in Alzheimer's disease and progressive supranuclear palsy. Brain 2017; 140:781-791. [PMID: 28122879 PMCID: PMC5382948 DOI: 10.1093/brain/aww340] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/02/2016] [Accepted: 11/24/2016] [Indexed: 12/30/2022] Open
Abstract
The ability to assess the distribution and extent of tau pathology in Alzheimer's disease and progressive supranuclear palsy in vivo would help to develop biomarkers for these tauopathies and clinical trials of disease-modifying therapies. New radioligands for positron emission tomography have generated considerable interest, and controversy, in their potential as tau biomarkers. We assessed the radiotracer 18F-AV-1451 with positron emission tomography imaging to compare the distribution and intensity of tau pathology in 15 patients with Alzheimer's pathology (including amyloid-positive mild cognitive impairment), 19 patients with progressive supranuclear palsy, and 13 age- and sex-matched controls. Regional analysis of variance and a support vector machine were used to compare and discriminate the clinical groups, respectively. We also examined the 18F-AV-1451 autoradiographic binding in post-mortem tissue from patients with Alzheimer's disease, progressive supranuclear palsy, and a control case to assess the 18F-AV-1451 binding specificity to Alzheimer's and non-Alzheimer's tau pathology. There was increased 18F-AV-1451 binding in multiple regions in living patients with Alzheimer's disease and progressive supranuclear palsy relative to controls [main effect of group, F(2,41) = 17.5, P < 0.0001; region of interest × group interaction, F(2,68) = 7.5, P < 0.00001]. More specifically, 18F-AV-1451 binding was significantly increased in patients with Alzheimer's disease, relative to patients with progressive supranuclear palsy and with control subjects, in the hippocampus and in occipital, parietal, temporal, and frontal cortices (t's > 2.2, P's < 0.04). Conversely, in patients with progressive supranuclear palsy, relative to patients with Alzheimer's disease, 18F-AV-1451 binding was elevated in the midbrain (t = 2.1, P < 0.04); while patients with progressive supranuclear palsy showed, relative to controls, increased 18F-AV-1451 uptake in the putamen, pallidum, thalamus, midbrain, and in the dentate nucleus of the cerebellum (t's > 2.7, P's < 0.02). The support vector machine assigned patients' diagnoses with 94% accuracy. The post-mortem autoradiographic data showed that 18F-AV-1451 strongly bound to Alzheimer-related tau pathology, but less specifically in progressive supranuclear palsy. 18F-AV-1451 binding to the basal ganglia was strong in all groups in vivo. Postmortem histochemical staining showed absence of neuromelanin-containing cells in the basal ganglia, indicating that off-target binding to neuromelanin is an insufficient explanation of 18F-AV-1451 positron emission tomography data in vivo, at least in the basal ganglia. Overall, we confirm the potential of 18F-AV-1451 as a heuristic biomarker, but caution is indicated in the neuropathological interpretation of its binding. Off-target binding may contribute to disease profiles of 18F-AV-1451 positron emission tomography, especially in primary tauopathies such as progressive supranuclear palsy. We suggest that 18F-AV-1451 positron emission tomography is a useful biomarker to assess tau pathology in Alzheimer's disease and to distinguish it from other tauopathies with distinct clinical and pathological characteristics such as progressive supranuclear palsy.
Collapse
Affiliation(s)
- Luca Passamonti
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- 2 Istituto di Bioimmagini e Fisiologia Molecolare, Consiglio Nazionale delle Ricerche, Milano, Italy
| | | | - Young T. Hong
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- 3 Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | | | - David Williamson
- 3 Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Robin J. Borchert
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Saber Sami
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Thomas E. Cope
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - P. Simon Jones
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Robert Arnold
- 5 Department of Psychiatry, University of Cambridge, Cambridge, UK
| | | | - Elijah Mak
- 5 Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Li Su
- 5 Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Tim D. Fryer
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- 3 Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Franklin I. Aigbirhio
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- 3 Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - John T. O’Brien
- 5 Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B. Rowe
- 1 Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
580
|
Lopes Alves I, Willemsen AT, Dierckx RA, da Silva AMM, Koole M. Dual time-point imaging for post-dose binding potential estimation applied to a [ 11C]raclopride PET dose occupancy study. J Cereb Blood Flow Metab 2017; 37:866-876. [PMID: 27073203 PMCID: PMC5363466 DOI: 10.1177/0271678x16644463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Receptor occupancy studies performed with PET often require time-consuming dynamic imaging for baseline and post-dose scans. Shorter protocol approximations based on standard uptake value ratios have been proposed. However, such methods depend on the time-point chosen for the quantification and often lead to overestimation and bias. The aim of this study was to develop a shorter protocol for the quantification of post-dose scans using a dual time-point approximation, which employs kinetic parameters from the baseline scan. Dual time-point was evaluated for a [11C]raclopride PET dose occupancy study with the D2 antagonist JNJ-37822681, obtaining estimates for binding potential and receptor occupancy. Results were compared to standard simplified reference tissue model and standard uptake value ratios-based estimates. Linear regression and Bland-Altman analysis demonstrated excellent correlation and agreement between dual time-point and the standard simplified reference tissue model approach. Moreover, the stability of dual time-point-based estimates is shown to be independent of the time-point chosen for quantification. Therefore, a dual time-point imaging protocol can be applied to post-dose [11C]raclopride PET scans, resulting in a significant reduction in total acquisition time while maintaining accuracy in the quantification of both the binding potential and the receptor occupancy.
Collapse
Affiliation(s)
- Isadora Lopes Alves
- 1 Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Antoon Tm Willemsen
- 1 Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudi A Dierckx
- 1 Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ana Maria M da Silva
- 2 Laboratory of Medical Imaging, School of Physics, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Michel Koole
- 1 Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,3 Department of Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium
| |
Collapse
|
581
|
Kim E, Howes OD, Veronese M, Beck K, Seo S, Park JW, Lee JS, Lee YS, Kwon JS. Presynaptic Dopamine Capacity in Patients with Treatment-Resistant Schizophrenia Taking Clozapine: An [ 18F]DOPA PET Study. Neuropsychopharmacology 2017; 42:941-950. [PMID: 27857125 PMCID: PMC5312074 DOI: 10.1038/npp.2016.258] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 11/09/2016] [Accepted: 11/11/2016] [Indexed: 01/20/2023]
Abstract
Some patients with schizophrenia show poor response to first-line antipsychotic treatments and this is termed treatment-resistant schizophrenia. The differential response to first-line antipsychotic drugs may reflect a different underlying neurobiology. Indeed, a previous study found dopamine synthesis capacity was significantly lower in patients with treatment-resistant schizophrenia. However, in this study, the treatment-resistant patients were highly symptomatic, whereas the responsive patients showed no or minimal symptoms. The study could not distinguish whether this was a trait effect or reflected the difference in symptom levels. Thus, we aimed to test whether dopaminergic function is altered in patients with a history of treatment resistance to first-line drugs relative to treatment responders when both groups are matched for symptom severity levels by recruiting treatment-resistant patients currently showed low symptom severity with the clozapine treatment. Healthy controls (n=12), patients treated with clozapine (n=12) who had not responded to first-line antipsychotics, and patients who had responded to first-line antipsychotics (n=12) were recruited. Participants were matched for age and sex and symptomatic severity level in patient groups. Participants' dopamine synthesis capacity was measured by using [18F]DOPA PET. We found that patients treated with clozapine show lower dopamine synthesis capacity than patients who have responded to first-line treatment (Cohen's d=0.9191 (whole striatum), 0.7781 (associative striatum), 1.0344 (limbic striatum), and 1.0189 (sensorimotor striatum) in line with the hypothesis that the dopaminergic function is linked to treatment response. This suggests that a different neurobiology may underlie treatment-resistant schizophrenia and that dopamine synthesis capacity may be a useful biomarker to predict treatment responsiveness.
Collapse
Affiliation(s)
- Euitae Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Gyeonggi-do, Korea
| | - Oliver D Howes
- Psychiatric Imaging, Medical Research Council Clinical Sciences Centre, Imperial College London, Hammersmith Hospital Campus, London, UK,Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Mattia Veronese
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Katherine Beck
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Seongho Seo
- Department of Brain & Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Korea,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Woo Park
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Sung Lee
- Department of Brain & Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Korea,Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jun Soo Kwon
- Department of Brain & Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Korea,Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea,Department of Psychiatry, Seoul National University College of Medicine and Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, 28 Yeongon-dong, Chongno-gu, Seoul 110-744, Korea, Tel: +82 2 2072 2972, Fax: +82 2 747 9063, E-mail:
| |
Collapse
|
582
|
Ziegler G, Ridgway GR, Blakemore SJ, Ashburner J, Penny W. Multivariate dynamical modelling of structural change during development. Neuroimage 2017; 147:746-762. [PMID: 27979788 PMCID: PMC5315058 DOI: 10.1016/j.neuroimage.2016.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/28/2016] [Accepted: 12/08/2016] [Indexed: 01/07/2023] Open
Abstract
Here we introduce a multivariate framework for characterising longitudinal changes in structural MRI using dynamical systems. The general approach enables modelling changes of states in multiple imaging biomarkers typically observed during brain development, plasticity, ageing and degeneration, e.g. regional gray matter volume of multiple regions of interest (ROIs). Structural brain states follow intrinsic dynamics according to a linear system with additional inputs accounting for potential driving forces of brain development. In particular, the inputs to the system are specified to account for known or latent developmental growth/decline factors, e.g. due to effects of growth hormones, puberty, or sudden behavioural changes etc. Because effects of developmental factors might be region-specific, the sensitivity of each ROI to contributions of each factor is explicitly modelled. In addition to the external effects of developmental factors on regional change, the framework enables modelling and inference about directed (potentially reciprocal) interactions between brain regions, due to competition for space, or structural connectivity, and suchlike. This approach accounts for repeated measures in typical MRI studies of development and aging. Model inversion and posterior distributions are obtained using earlier established variational methods enabling Bayesian evidence-based comparisons between various models of structural change. Using this approach we demonstrate dynamic cortical changes during brain maturation between 6 and 22 years of age using a large openly available longitudinal paediatric dataset with 637 scans from 289 individuals. In particular, we model volumetric changes in 26 bilateral ROIs, which cover large portions of cortical and subcortical gray matter. We account for (1) puberty-related effects on gray matter regions; (2) effects of an early transient growth process with additional time-lag parameter; (3) sexual dimorphism by modelling parameter differences between boys and girls. There is evidence that the regional pattern of sensitivity to dynamic hidden growth factors in late childhood is similar across genders and shows a consistent anterior-posterior gradient with strongest impact to prefrontal cortex (PFC) brain changes. Finally, we demonstrate the potential of the framework to explore the coupling of structural changes across a priori defined subnetworks using an example of previously established resting state functional connectivity.
Collapse
Affiliation(s)
- Gabriel Ziegler
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany.
| | - Gerard R Ridgway
- FMRIB Centre, University of Oxford, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK; Wellcome Trust Centre for Neuroimaging, University College, London WC1N 3BG, UK
| | | | - John Ashburner
- Wellcome Trust Centre for Neuroimaging, University College, London WC1N 3BG, UK
| | - Will Penny
- Wellcome Trust Centre for Neuroimaging, University College, London WC1N 3BG, UK
| |
Collapse
|
583
|
Emberson LL, Cannon G, Palmeri H, Richards JE, Aslin RN. Using fNIRS to examine occipital and temporal responses to stimulus repetition in young infants: Evidence of selective frontal cortex involvement. Dev Cogn Neurosci 2017; 23:26-38. [PMID: 28012401 PMCID: PMC5253300 DOI: 10.1016/j.dcn.2016.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/06/2016] [Accepted: 11/11/2016] [Indexed: 12/24/2022] Open
Abstract
How does the developing brain respond to recent experience? Repetition suppression (RS) is a robust and well-characterized response of to recent experience found, predominantly, in the perceptual cortices of the adult brain. We use functional near-infrared spectroscopy (fNIRS) to investigate how perceptual (temporal and occipital) and frontal cortices in the infant brain respond to auditory and visual stimulus repetitions (spoken words and faces). In Experiment 1, we find strong evidence of repetition suppression in the frontal cortex but only for auditory stimuli. In perceptual cortices, we find only suggestive evidence of auditory RS in the temporal cortex and no evidence of visual RS in any ROI. In Experiments 2 and 3, we replicate and extend these findings. Overall, we provide the first evidence that infant and adult brains respond differently to stimulus repetition. We suggest that the frontal lobe may support the development of RS in perceptual cortices.
Collapse
|
584
|
Rodriguez-Vieitez E, Leuzy A, Chiotis K, Saint-Aubert L, Wall A, Nordberg A. Comparability of [ 18F]THK5317 and [ 11C]PIB blood flow proxy images with [ 18F]FDG positron emission tomography in Alzheimer's disease. J Cereb Blood Flow Metab 2017; 37:740-749. [PMID: 27107028 PMCID: PMC5381463 DOI: 10.1177/0271678x16645593] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
For amyloid positron emission tomography tracers, the simplified reference tissue model derived ratio of influx rate in target relative to reference region (R1) has been shown to serve as a marker of brain perfusion, and, due to the strong coupling between perfusion and metabolism, as a proxy for glucose metabolism. In the present study, 11 prodromal Alzheimer's disease and nine Alzheimer's disease dementia patients underwent [18F]THK5317, carbon-11 Pittsburgh Compound-B ([11C]PIB), and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography to assess the possible use of early-phase [18F]THK5317 and R1 as proxies for brain perfusion, and thus, for glucose metabolism. Discriminative performance (prodromal vs Alzheimer's disease dementia) of [18F]THK5317 (early-phase SUVr and R1) was compared with that of [11C]PIB (early-phase SUVr and R1) and [18F]FDG. Strong positive correlations were found between [18F]THK5317 (early-phase, R1) and [18F]FDG, particularly in frontal and temporoparietal regions. Differences in correlations between early-phase and R1 ([18F]THK5317 and [11C]PIB) and [18F]FDG, were not statistically significant, nor were differences in area under the curve values in the discriminative analysis. Our findings suggest that early-phase [18F]THK5317 and R1 provide information on brain perfusion, closely related to glucose metabolism. As such, a single positron emission tomography study with [18F]THK5317 may provide information about both tau pathology and brain perfusion in Alzheimer's disease, with potential clinical applications.
Collapse
Affiliation(s)
| | - Antoine Leuzy
- 1 Department NVS, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Anders Wall
- 2 Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Agneta Nordberg
- 1 Department NVS, Karolinska Institutet, Stockholm, Sweden.,3 Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
585
|
Interference from related actions in spoken word production: Behavioural and fMRI evidence. Neuropsychologia 2017; 96:78-88. [DOI: 10.1016/j.neuropsychologia.2017.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 11/18/2022]
|
586
|
Kumar VJ, van Oort E, Scheffler K, Beckmann CF, Grodd W. Functional anatomy of the human thalamus at rest. Neuroimage 2017; 147:678-691. [DOI: 10.1016/j.neuroimage.2016.12.071] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/25/2022] Open
|
587
|
van der Lee SJ, Roshchupkin GV, Adams HHH, Schmidt H, Hofer E, Saba Y, Schmidt R, Hofman A, Amin N, van Duijn CM, Vernooij MW, Ikram MA, Niessen WJ. Gray matter heritability in family-based and population-based studies using voxel-based morphometry. Hum Brain Mapp 2017; 38:2408-2423. [PMID: 28145022 DOI: 10.1002/hbm.23528] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 10/27/2016] [Accepted: 01/12/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The combination of genetics and imaging has improved their understanding of the brain through studies of aggregate measures obtained from high-resolution structural imaging. Voxel-wise analyses have the potential to provide more detailed information of genetic influences on the brain. Here they report a large-scale study of the heritability of gray matter at voxel resolution (1 × 1 × 1 mm). METHODS Validated voxel-based morphometry (VBM) protocols were applied to process magnetic resonance imaging data of 3,239 unrelated subjects from a population-based study and 491 subjects from two family-based studies. Genome-wide genetic data was used to estimate voxel-wise gray matter heritability of the unrelated subjects and pedigree-structure was used to estimate heritability in families. They subsequently associated two genetic variants, known to be linked with subcortical brain volume, with most heritable voxels to determine if this would enhance their association signals. RESULTS Voxels significantly heritable in both estimates mapped to subcortical structures, but also voxels in the language areas of the left hemisphere were found significantly heritable. When comparing regional patterns of heritability, family-based estimates were higher than population-based estimates. However, regional consistency of the heritability measures across study designs was high (Pearson's correlation coefficient = 0.73, P = 2.6 × 10-13 ). They further show enhancement of the association signal of two previously discovered genetic loci with subcortical volume by using only the most heritable voxels. CONCLUSION Gray matter voxel-wise heritability can be reliably estimated with different methods. Combining heritability estimates from multiple studies is feasible to construct reliable heritability maps of gray matter voxels. Hum Brain Mapp 38:2408-2423, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sven J van der Lee
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Gennady V Roshchupkin
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, Austria.,Department of Neurology, Medical University Graz, Graz, Austria
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University Graz, Graz, Austria.,Institute of Medical Informatics, Statistics and Documentation, Medical University Graz, Graz, Austria
| | - Yasaman Saba
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University Graz, Graz, Austria
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Meike W Vernooij
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wiro J Niessen
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Medical Informatics, Erasmus Medical Center, Rotterdam, The Netherlands.,Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
588
|
Dickie DA, Shenkin SD, Anblagan D, Lee J, Blesa Cabez M, Rodriguez D, Boardman JP, Waldman A, Job DE, Wardlaw JM. Whole Brain Magnetic Resonance Image Atlases: A Systematic Review of Existing Atlases and Caveats for Use in Population Imaging. Front Neuroinform 2017; 11:1. [PMID: 28154532 PMCID: PMC5244468 DOI: 10.3389/fninf.2017.00001] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/04/2017] [Indexed: 11/17/2022] Open
Abstract
Brain MRI atlases may be used to characterize brain structural changes across the life course. Atlases have important applications in research, e.g., as registration and segmentation targets to underpin image analysis in population imaging studies, and potentially in future in clinical practice, e.g., as templates for identifying brain structural changes out with normal limits, and increasingly for use in surgical planning. However, there are several caveats and limitations which must be considered before successfully applying brain MRI atlases to research and clinical problems. For example, the influential Talairach and Tournoux atlas was derived from a single fixed cadaveric brain from an elderly female with limited clinical information, yet is the basis of many modern atlases and is often used to report locations of functional activation. We systematically review currently available whole brain structural MRI atlases with particular reference to the implications for population imaging through to emerging clinical practice. We found 66 whole brain structural MRI atlases world-wide. The vast majority were based on T1, T2, and/or proton density (PD) structural sequences, had been derived using parametric statistics (inappropriate for brain volume distributions), had limited supporting clinical or cognitive data, and included few younger (>5 and <18 years) or older (>60 years) subjects. To successfully characterize brain structural features and their changes across different stages of life, we conclude that whole brain structural MRI atlases should include: more subjects at the upper and lower extremes of age; additional structural sequences, including fluid attenuation inversion recovery (FLAIR) and T2* sequences; a range of appropriate statistics, e.g., rank-based or non-parametric; and detailed cognitive and clinical profiles of the included subjects in order to increase the relevance and utility of these atlases.
Collapse
Affiliation(s)
- David Alexander Dickie
- Brain Research Imaging Centre, Neuroimaging Sciences, Centre for Clinical Brain Sciences, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationGlasgow, UK
| | - Susan D. Shenkin
- Brain Research Imaging Centre, Neuroimaging Sciences, Centre for Clinical Brain Sciences, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
- Geriatric Medicine Unit, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of EdinburghEdinburgh, UK
| | - Devasuda Anblagan
- Brain Research Imaging Centre, Neuroimaging Sciences, Centre for Clinical Brain Sciences, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationGlasgow, UK
- MRC Centre for Reproductive Health, Queen's Medical Research InstituteEdinburgh, UK
| | - Juyoung Lee
- Graduate Training Centre of Neuroscience, International Max Planck Research School, University of TübingenTübingen, Germany
| | - Manuel Blesa Cabez
- MRC Centre for Reproductive Health, Queen's Medical Research InstituteEdinburgh, UK
| | - David Rodriguez
- Brain Research Imaging Centre, Neuroimaging Sciences, Centre for Clinical Brain Sciences, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationGlasgow, UK
| | - James P. Boardman
- MRC Centre for Reproductive Health, Queen's Medical Research InstituteEdinburgh, UK
| | - Adam Waldman
- Brain Research Imaging Centre, Neuroimaging Sciences, Centre for Clinical Brain Sciences, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
| | - Dominic E. Job
- Brain Research Imaging Centre, Neuroimaging Sciences, Centre for Clinical Brain Sciences, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationGlasgow, UK
| | - Joanna M. Wardlaw
- Brain Research Imaging Centre, Neuroimaging Sciences, Centre for Clinical Brain Sciences, Royal Infirmary of Edinburgh, The University of EdinburghEdinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) CollaborationGlasgow, UK
- Department of Psychology, Centre for Cognitive Ageing and Cognitive Epidemiology, The University of EdinburghEdinburgh, UK
| |
Collapse
|
589
|
A Discriminative Event Based Model for Alzheimer’s Disease Progression Modeling. LECTURE NOTES IN COMPUTER SCIENCE 2017. [DOI: 10.1007/978-3-319-59050-9_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
590
|
Laviña B. Brain Vascular Imaging Techniques. Int J Mol Sci 2016; 18:ijms18010070. [PMID: 28042833 PMCID: PMC5297705 DOI: 10.3390/ijms18010070] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/13/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022] Open
Abstract
Recent major improvements in a number of imaging techniques now allow for the study of the brain in ways that could not be considered previously. Researchers today have well-developed tools to specifically examine the dynamic nature of the blood vessels in the brain during development and adulthood; as well as to observe the vascular responses in disease situations in vivo. This review offers a concise summary and brief historical reference of different imaging techniques and how these tools can be applied to study the brain vasculature and the blood-brain barrier integrity in both healthy and disease states. Moreover, it offers an overview on available transgenic animal models to study vascular biology and a description of useful online brain atlases.
Collapse
Affiliation(s)
- Bàrbara Laviña
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden.
| |
Collapse
|
591
|
Gonzalez-Escamilla G, Lange C, Teipel S, Buchert R, Grothe MJ. PETPVE12: an SPM toolbox for Partial Volume Effects correction in brain PET - Application to amyloid imaging with AV45-PET. Neuroimage 2016; 147:669-677. [PMID: 28039094 DOI: 10.1016/j.neuroimage.2016.12.077] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 12/15/2022] Open
Abstract
Positron emission tomography (PET) allows detecting molecular brain changes in vivo. However, the accuracy of PET is limited by partial volume effects (PVE) that affects quantitative analysis and visual interpretation of the images. Although PVE-correction methods have been shown to effectively increase the correspondence of the measured signal with the true regional tracer uptake, these procedures are still not commonly applied, neither in clinical nor in research settings. Here, we present an implementation of well validated PVE-correction procedures as a SPM toolbox, PETPVE12, for automated processing. We demonstrate its utility by a comprehensive analysis of the effects of PVE-correction on amyloid-sensitive AV45-PET data from 85 patients with Alzheimer's disease (AD) and 179 cognitively normal (CN) elderly. Effects of PVE-correction on global cortical standard uptake value ratios (SUVR) and the power of diagnostic group separation were assessed for the region-wise geometric transfer matrix method (PVEc-GTM), as well as for the 3-compartmental voxel-wise "Müller-Gärtner" method (PVEc-MG). Both PVE-correction methods resulted in decreased global cortical SUVRs in the low to middle range of SUVR values, and in increased global cortical SUVRs at the high values. As a consequence, average SUVR of the CN group was reduced, whereas average SUVR of the AD group was increased by PVE-correction. These effects were also reflected in increased accuracies of group discrimination after PVEc-GTM (AUC=0.86) and PVEc-MG (AUC=0.89) compared to standard non-corrected SUVR (AUC=0.84). Voxel-wise analyses of PVEc-MG corrected data also demonstrated improved detection of regionally increased AV45 SUVR values in AD patients. These findings complement the growing evidence for a beneficial effect of PVE-correction in quantitative analysis of amyloid-sensitive PET data. The novel PETPVE12 toolbox significantly facilitates the application of PVE-correction, particularly within SPM-based processing pipelines. This is expected to foster the use of PVE-correction in brain PET for more widespread use. The toolbox is freely available at http://www.fil.ion.ucl.ac.uk/spm/ext/#PETPVE12.
Collapse
Affiliation(s)
| | - Catharina Lange
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Ralph Buchert
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany.
| |
Collapse
|
592
|
Mendelson AF, Zuluaga MA, Lorenzi M, Hutton BF, Ourselin S. Selection bias in the reported performances of AD classification pipelines. NEUROIMAGE-CLINICAL 2016; 14:400-416. [PMID: 28271040 PMCID: PMC5322215 DOI: 10.1016/j.nicl.2016.12.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/09/2016] [Accepted: 12/16/2016] [Indexed: 12/26/2022]
Abstract
The last decade has seen a great proliferation of supervised learning pipelines for individual diagnosis and prognosis in Alzheimer's disease. As more pipelines are developed and evaluated in the search for greater performance, only those results that are relatively impressive will be selected for publication. We present an empirical study to evaluate the potential for optimistic bias in classification performance results as a result of this selection. This is achieved using a novel, resampling-based experiment design that effectively simulates the optimisation of pipeline specifications by individuals or collectives of researchers using cross validation with limited data. Our findings indicate that bias can plausibly account for an appreciable fraction (often greater than half) of the apparent performance improvement associated with the pipeline optimisation, particularly in small samples. We discuss the consistency of our findings with patterns observed in the literature and consider strategies for bias reduction and mitigation. Demonstration and measurement of selection bias in AD classification experiments Bias accounts for much of the performance improvement seen with pipeline optimisation. Assessment of key risk factors and guidance on best research practices Evidence of selection bias in results collected by a recent literature review
Collapse
Affiliation(s)
- Alex F Mendelson
- Translational Imaging Group, Centre for Medical Image Computing University College London, London, UK
| | - Maria A Zuluaga
- Translational Imaging Group, Centre for Medical Image Computing University College London, London, UK
| | - Marco Lorenzi
- Translational Imaging Group, Centre for Medical Image Computing University College London, London, UK
| | - Brian F Hutton
- Institute of Nuclear Medicine, University College London, London, UK; Centre for Medical Radiation Physics, University of Wollongong, NSW, Australia
| | - Sébastien Ourselin
- Translational Imaging Group, Centre for Medical Image Computing University College London, London, UK; Dementia Research Centre, University College London, UK
| | | |
Collapse
|
593
|
Bang L, Rø Ø, Endestad T. Threat-Detection and Attentional Bias to Threat in Women Recovered from Anorexia Nervosa: Neural Alterations in Extrastriate and Medial Prefrontal Cortices. EUROPEAN EATING DISORDERS REVIEW 2016; 25:80-88. [DOI: 10.1002/erv.2494] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Lasse Bang
- Regional Department for Eating Disorders, Division of Mental Health and Addiction; Oslo University Hospital; Oslo Norway
| | - Øyvind Rø
- Regional Department for Eating Disorders, Division of Mental Health and Addiction; Oslo University Hospital; Oslo Norway
- Division of Mental Health and Addiction, Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - Tor Endestad
- Department of Psychology; University of Oslo; Oslo Norway
| |
Collapse
|
594
|
Schreiner SJ, Kirchner T, Wyss M, Van Bergen JM, Quevenco FC, Steininger SC, Griffith EY, Meier I, Michels L, Gietl AF, Leh SE, Brickman AM, Hock C, Nitsch RM, Pruessmann KP, Henning A, Unschuld PG. Low episodic memory performance in cognitively normal elderly subjects is associated with increased posterior cingulate gray matter N-acetylaspartate: a 1H MRSI study at 7 Tesla. Neurobiol Aging 2016; 48:195-203. [DOI: 10.1016/j.neurobiolaging.2016.08.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/03/2016] [Accepted: 08/24/2016] [Indexed: 11/30/2022]
|
595
|
Barret O, Alagille D, Sanabria S, Comley RA, Weimer RM, Borroni E, Mintun M, Seneca N, Papin C, Morley T, Marek K, Seibyl JP, Tamagnan GD, Jennings D. Kinetic Modeling of the Tau PET Tracer 18F-AV-1451 in Human Healthy Volunteers and Alzheimer Disease Subjects. J Nucl Med 2016; 58:1124-1131. [PMID: 27908967 DOI: 10.2967/jnumed.116.182881] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/22/2016] [Indexed: 01/01/2023] Open
Abstract
18F-AV-1451 is currently the most widely used of several experimental tau PET tracers. The objective of this study was to evaluate 18F-AV-1451 binding with full kinetic analysis using a metabolite-corrected arterial input function and to compare parameters derived from kinetic analysis with SUV ratio (SUVR) calculated over different imaging time intervals. Methods:18F-AV-1451 PET brain imaging was completed in 16 subjects: 4 young healthy volunteers (YHV), 4 aged healthy volunteers (AHV), and 8 Alzheimer disease (AD) subjects. Subjects were imaged for 3.5 h, with arterial blood samples obtained throughout. PET data were analyzed using plasma and reference tissue-based methods to estimate the distribution volume, binding potential (BPND), and SUVR. BPND and SUVR were calculated using the cerebellar cortex as a reference region and were compared across the different methods and across the 3 groups (YHV, AHV, and AD). Results: AD demonstrated increased 18F-AV-1451 retention compared with YHV and AHV based on both invasive and noninvasive analyses in cortical regions in which paired helical filament tau accumulation is expected in AD. A correlation of R2 > 0.93 was found between BPND (130 min) and SUVR-1 at all time intervals. Cortical SUVR curves reached a relative plateau around 1.0-1.2 for YHV and AHV by approximately 50 min, but increased in AD by up to approximately 20% at 110-130 min and approximately 30% at 160-180 min relative to 80-100 min. Distribution volume (130 min) was lower by 30%-35% in the YHV than AHV. Conclusion: Our data suggest that although 18F-AV-1451 SUVR curves do not reach a plateau and are still increasing in AD, an SUVR calculated over an imaging window of 80-100 min (as currently used in clinical studies) provides estimates of paired helical filament tau burden in good correlation with BPND, whereas SUVR sensitivity to regional cerebral blood changes needs further investigation.
Collapse
Affiliation(s)
| | | | - Sandra Sanabria
- Genentech Research and Early Development, Genentech, South San Francisco, California
| | - Robert A Comley
- Pharma Research and Early Development, F. Hoffmann-La Roche, Konzern-Hauptsitz, Basel, Switzerland
| | - Robby M Weimer
- Genentech Research and Early Development, Genentech, South San Francisco, California
| | - Edilio Borroni
- Pharma Research and Early Development, F. Hoffmann-La Roche, Konzern-Hauptsitz, Basel, Switzerland
| | - Mark Mintun
- Avid Radiopharmaceuticals, Philadelphia, Pennsylvania; and
| | - Nicholas Seneca
- Product Development, F. Hoffmann-La Roche, Konzern-Hauptsitz, Basel, Switzerland
| | | | | | - Ken Marek
- Molecular NeuroImaging LLC, New Haven, Connecticut
| | | | | | | |
Collapse
|
596
|
Holmes SE, Hinz R, Drake RJ, Gregory CJ, Conen S, Matthews JC, Anton-Rodriguez JM, Gerhard A, Talbot PS. In vivo imaging of brain microglial activity in antipsychotic-free and medicated schizophrenia: a [ 11C](R)-PK11195 positron emission tomography study. Mol Psychiatry 2016; 21:1672-1679. [PMID: 27698434 DOI: 10.1038/mp.2016.180] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) imaging of the 18 kDa translocator protein (TSPO) has been used to investigate whether microglial activation, an indication of neuroinflammation, is evident in the brain of adults with schizophrenia. Interpretation of these studies is confounded by potential modulatory effects of antipsychotic medication on microglial activity. In the first such study in antipsychotic-free schizophrenia, we have used [11C](R)-PK11195 PET to compare TSPO availability in a predominantly antipsychotic-naive group of moderate-to-severely symptomatic unmedicated patients (n=8), similarly symptomatic medicated patients with schizophrenia taking risperidone or paliperidone by regular intramuscular injection (n=8), and healthy comparison subjects (n=16). We found no evidence for increased TSPO availability in antipsychotic-free patients compared with healthy controls (mean difference 4%, P=0.981). However, TSPO availability was significantly elevated in medicated patients (mean increase 88%, P=0.032) across prefrontal (dorsolateral, ventrolateral, orbital), anterior cingulate and parietal cortical regions. In the patients, TSPO availability was also strongly correlated with negative symptoms measured using the Positive and Negative Syndrome Scale across all the brain regions investigated (r=0.651-0.741). We conclude that the pathophysiology of schizophrenia is not associated with microglial activation in the 2-6 year period following diagnosis. The elevation in the medicated patients may be a direct effect of the antipsychotic, although this study cannot exclude treatment resistance and/or longer illness duration as potential explanations. It also remains to be determined whether it is present only in a subset of patients, represents a pro- or anti-inflammatory state, its association with primary negative symptoms, and whether there are significant differences between antipsychotics.
Collapse
Affiliation(s)
- S E Holmes
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - R Hinz
- Wolfson Molecular Imaging Centre, Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - R J Drake
- Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - C J Gregory
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - S Conen
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - J C Matthews
- Wolfson Molecular Imaging Centre, Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - J M Anton-Rodriguez
- Wolfson Molecular Imaging Centre, Division of Informatics, Imaging & Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Gerhard
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - P S Talbot
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
597
|
Healthy brain ageing assessed with 18F-FDG PET and age-dependent recovery factors after partial volume effect correction. Eur J Nucl Med Mol Imaging 2016; 44:838-849. [PMID: 27878594 DOI: 10.1007/s00259-016-3569-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/03/2016] [Indexed: 12/26/2022]
|
598
|
Politis M, Sauerbier A, Loane C, Pavese N, Martin A, Corcoran B, Brooks DJ, Ray-Chaudhuri K, Piccini P. Sustained striatal dopamine levels following intestinal levodopa infusions in Parkinson's disease patients. Mov Disord 2016; 32:235-240. [PMID: 27859651 DOI: 10.1002/mds.26848] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The objective of this study was to investigate in vivo the ability of levodopa/carbidopa intestinal gel infusions to produce sustained striatal dopamine levels and to improve clinical outcomes in Parkinson's disease patients. METHODS Six advanced Parkinson's disease patients had serial [11 C]raclopride PET to assess levodopa/carbidopa intestinal gel infusion-induced rises in striatal dopamine as reflected by a fall in dopamine-D2/3 receptor availability. Parkinson's disease patients had baseline scan OFF-dopaminergic stimulation and 2 scans following initiation of levodopa/carbidopa intestinal gel infusions. Striatal D2/3 binding was measured in striatal subregions corresponding to sensorimotor, limbic, and cognitive/associative function. RESULTS Mean striatal [11 C]raclopride nondisplaceable binding potential decreased by 14.0% to 16.7% in sensorimotor, 12.0%-14.4% in limbic, and 8.7%-11.6% in cognitive/associative function subregions at 1- to 10-hour points (P < 0.01). Sensorimotor subregion [11 C]raclopride nondisplaceable binding potential reductions correlated with reductions in Unified Parkinson's Disease Rating Scale Part III scores over the course of the infusion (r = 0.81; P < 0.05). CONCLUSIONS Levodopa/carbidopa intestinal gel infusions generate a stable rise in striatal dopamine levels and are associated with improvements in motor manifestations. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marios Politis
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Parkinson's Centre of Excellence, Department of Neurology, King's College Hospital Foundation Trust, London, UK.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Anna Sauerbier
- Parkinson's Centre of Excellence, Department of Neurology, King's College Hospital Foundation Trust, London, UK.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Clare Loane
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK.,Memory Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU
| | - Nicola Pavese
- Centre for Neurodegeneration and Neuroinflammation, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK.,Department of Nuclear Medicine, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne Martin
- Parkinson's Centre of Excellence, Department of Neurology, King's College Hospital Foundation Trust, London, UK
| | - Benjamin Corcoran
- Department of Nuclear Medicine, King's College Hospital Foundation Trust, London, UK
| | - David J Brooks
- Centre for Neurodegeneration and Neuroinflammation, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK.,Department of Nuclear Medicine, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Division of Neurology, Newcastle University, Newcastle upon Tyne, UK
| | - K Ray-Chaudhuri
- Parkinson's Centre of Excellence, Department of Neurology, King's College Hospital Foundation Trust, London, UK.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Paola Piccini
- Centre for Neurodegeneration and Neuroinflammation, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
599
|
Ikawa M, Lohith TG, Shrestha S, Telu S, Zoghbi SS, Castellano S, Taliani S, Da Settimo F, Fujita M, Pike VW, Innis RB. 11C-ER176, a Radioligand for 18-kDa Translocator Protein, Has Adequate Sensitivity to Robustly Image All Three Affinity Genotypes in Human Brain. J Nucl Med 2016; 58:320-325. [PMID: 27856631 DOI: 10.2967/jnumed.116.178996] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/29/2016] [Indexed: 11/16/2022] Open
Abstract
For PET imaging of 18-kDa translocator protein (TSPO), a biomarker of neuroinflammation, most second-generation radioligands are sensitive to the single nucleotide polymorphism rs6971; however, this is probably not the case for the prototypical agent 11C-PK11195 (11C-labeled N-butan-2-yl-1-(2-chlorophenyl)-N-methylisoquinoline-3-carboxamide), which has a relatively lower signal-to-noise ratio. We recently found that 11C-ER176 (11C-(R)-N-sec-butyl-4-(2-chlorophenyl)-N-methylquinazoline-2-carboxamide), a new analog of 11C-(R)-PK11195, showed little sensitivity to rs6971 when tested in vitro and had high specific binding in monkey brain. This study sought, first, to determine whether the sensitivity of 11C-ER176 in humans is similar to the low sensitivity measured in vitro and, second, to measure the nondisplaceable binding potential (BPND, or the ratio of specific-to-nondisplaceable uptake) of 11C-ER176 in human brain. METHODS Nine healthy volunteers-3 high-affinity binders (HABs), 3 mixed-affinity binders (MABs), and 3 low-affinity binders (LABs)-were studied with whole-body 11C-ER176 PET imaging. SUVs from 60 to 120 min after injection derived from each organ were compared between genotypes. Eight separate healthy volunteers-3 HABs, 3 MABs, and 2 LABs-underwent brain PET imaging. The 3 HABs underwent a repeated brain scan after TSPO blockade with XBD173 (N-benzyl-N-ethyl-2-(7-methyl-8-oxo-2-phenylpurin-9-yl)acetamide) to determine nondisplaceable distribution volume (VND) via Lassen occupancy plotting and thereby estimate BPND in brain. RESULTS Regional SUV averaged from 60 to 120 min after injection in brain and peripheral organs with high TSPO densities such as lung and spleen were greater in HABs than in LABs. On the basis of VND determined via the occupancy plot, the whole-brain BPND for LABs was estimated to be 1.4 ± 0.8, which was much lower than that for HABs (4.2 ± 1.3) but about the same as that for HABs with 11C-PBR28 ([methyl-11C]N-acetyl-N-(2-methoxybenzyl)-2-phenoxy-5-pyridinamine)) (∼1.2). CONCLUSION Obvious in vivo sensitivity to rs6971 was observed in 11C-ER176 that had not been expected from in vitro studies, suggesting that the future development of any improved radioligand for TSPO should consider the possibility that in vitro properties will not be reflected in vivo. We also found that 11C-ER176 has adequately high BPND for all rs6971 genotypes. Thus, the new radioligand would likely have greater sensitivity in detecting abnormalities in patients.
Collapse
Affiliation(s)
- Masamichi Ikawa
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Talakad G Lohith
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Stal Shrestha
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Sabrina Castellano
- Department of Medicine and Surgery, University of Salerno, Fisciano, Italy; and
| | | | | | - Masahiro Fujita
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
600
|
Abstract
Although music is ubiquitous in human societies, there are some people for whom music holds no reward value despite normal perceptual ability and preserved reward-related responses in other domains. The study of these individuals with specific musical anhedonia may be crucial to understand better the neural correlates underlying musical reward. Previous neuroimaging studies have shown that musically induced pleasure may arise from the interaction between auditory cortical networks and mesolimbic reward networks. If such interaction is critical for music-induced pleasure to emerge, then those individuals who do not experience it should show alterations in the cortical-mesolimbic response. In the current study, we addressed this question using fMRI in three groups of 15 participants, each with different sensitivity to music reward. We demonstrate that the music anhedonic participants showed selective reduction of activity for music in the nucleus accumbens (NAcc), but normal activation levels for a monetary gambling task. Furthermore, this group also exhibited decreased functional connectivity between the right auditory cortex and ventral striatum (including the NAcc). In contrast, individuals with greater than average response to music showed enhanced connectivity between these structures. Thus, our results suggest that specific musical anhedonia may be associated with a reduction in the interplay between the auditory cortex and the subcortical reward network, indicating a pivotal role of this interaction for the enjoyment of music.
Collapse
|