501
|
van der Aart J, Golla SSV, van der Pluijm M, Schwarte LA, Schuit RC, Klein PJ, Metaxas A, Windhorst AD, Boellaard R, Lammertsma AA, van Berckel BNM. First in human evaluation of [ 18F]PK-209, a PET ligand for the ion channel binding site of NMDA receptors. EJNMMI Res 2018; 8:69. [PMID: 30054846 PMCID: PMC6063804 DOI: 10.1186/s13550-018-0424-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/06/2018] [Indexed: 12/05/2022] Open
Abstract
Background Efforts to develop suitable positron emission tomography (PET) tracers for the ion channel site of human N-methyl-d-aspartate (NMDA) receptors have had limited success. [18F]PK-209 is a GMOM derivative that binds to the intrachannel phencyclidine site with high affinity and selectivity. Primate PET studies have shown that the volume of distribution in the brain was reduced by administration of the NMDA receptor antagonist MK-801, consistent with substantial specific binding. The purpose of the present study was to evaluate [18F]PK-209 in 10 healthy humans by assessing test–retest reproducibility and binding specificity following intravenous S-ketamine administration (0.5 mg ∙ kg−1). Five healthy subjects underwent a test–retest protocol, and five others a baseline-ketamine protocol. In all cases dynamic, 120-min PET scans were acquired together with metabolite-corrected arterial plasma input functions. Additional input functions were tested based on within-subject and population-average parent fractions. Results Best fits of the brain time-activity curves were obtained using an irreversible two-tissue compartment model with additional blood volume parameter. Mean test–retest variability of the net rate of influx Ki varied between 7 and 24% depending on the input function. There were no consistent changes in [18F]PK-209 PET parameters following ketamine administration, which may be a consequence of the complex endogenous ligand processes that affect channel gating. Conclusions The molecular interaction between [18F]PK-209 and the binding site within the NMDA receptor ion channel is insufficiently reproducible and specific to be a reliable imaging agent for its quantification. Trial registration EudraCT 2014-001735-36. Registered 28 April 2014
Collapse
Affiliation(s)
- Jasper van der Aart
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands. .,Centre for Human Drug Research, Leiden, The Netherlands.
| | - Sandeep S V Golla
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Marieke van der Pluijm
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Lothar A Schwarte
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Pieter J Klein
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Athanasios Metaxas
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
502
|
Ledig C, Schuh A, Guerrero R, Heckemann RA, Rueckert D. Structural brain imaging in Alzheimer's disease and mild cognitive impairment: biomarker analysis and shared morphometry database. Sci Rep 2018; 8:11258. [PMID: 30050078 PMCID: PMC6062561 DOI: 10.1038/s41598-018-29295-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/06/2018] [Indexed: 12/22/2022] Open
Abstract
Magnetic resonance (MR) imaging is a powerful technique for non-invasive in-vivo imaging of the human brain. We employed a recently validated method for robust cross-sectional and longitudinal segmentation of MR brain images from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort. Specifically, we segmented 5074 MR brain images into 138 anatomical regions and extracted time-point specific structural volumes and volume change during follow-up intervals of 12 or 24 months. We assessed the extracted biomarkers by determining their power to predict diagnostic classification and by comparing atrophy rates to published meta-studies. The approach enables comprehensive analysis of structural changes within the whole brain. The discriminative power of individual biomarkers (volumes/atrophy rates) is on par with results published by other groups. We publish all quality-checked brain masks, structural segmentations, and extracted biomarkers along with this article. We further share the methodology for brain extraction (pincram) and segmentation (MALPEM, MALPEM4D) as open source projects with the community. The identified biomarkers hold great potential for deeper analysis, and the validated methodology can readily be applied to other imaging cohorts.
Collapse
Affiliation(s)
- Christian Ledig
- Imperial College London, Department of Computing, London, SW7 2AZ, UK.
| | - Andreas Schuh
- Imperial College London, Department of Computing, London, SW7 2AZ, UK
| | - Ricardo Guerrero
- Imperial College London, Department of Computing, London, SW7 2AZ, UK
| | - Rolf A Heckemann
- MedTech West, Sahlgrenska University Hospital, 413 45, Gothenburg, Sweden.,Department of Radiation Therapy, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Division of Brain Sciences, Imperial College London, London, UK
| | - Daniel Rueckert
- Imperial College London, Department of Computing, London, SW7 2AZ, UK
| |
Collapse
|
503
|
Groot C, Tolboom N, Ikonomovic MD, Lammertsma AA, Boon BDC, Barkhof F, Scheltens P, Klunk WE, Rozemuller AJM, Ossenkoppele R, van Berckel BNM. Quantitative PET and Histology of Brain Biopsy Reveal Lack of Selective Pittsburgh Compound-B Binding to Intracerebral Amyloidoma. J Alzheimers Dis 2018; 65:71-77. [PMID: 30040724 DOI: 10.3233/jad-180316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This single case study examines selective Pittsburgh compound-B (PiB) binding to an intracerebral light-chain amyloidoma using a 90-minute dynamic [11C]PiB-PET scan and brain biopsy tissue. Parametric non-displaceable binding potential (BPND) images showed low specific binding in the amyloidoma (BPND = 0.23), while relative tracer delivery was adequate (R1 = 0.44). Histology of the tissue revealed strong coloring with Congo-red, thioflavin-S, and X-34, indicating presence of amyloid. However, immunological staining with 6F/3D revealed absence of amyloid-β and histofluorescence of 6-CN-PiB, a highly fluorescent derivative of PiB, was at background levels. Our results suggest that PiB does not detect the atypical amyloid pathology associated with an intracerebral light-chain amyloidoma. These findings are of interest to clinicians and researchers applying [11C]PiB-PET to detect atypical forms of amyloid pathology.
Collapse
Affiliation(s)
- Colin Groot
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Geriatric Research Education and Clinical Center, Veterans Administration Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Baayla D C Boon
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands.,Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.,Institutes of Neurology and Healthcare Engineering, UCL, London, UK
| | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - William E Klunk
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Rik Ossenkoppele
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
504
|
Delso G, Kemp B, Kaushik S, Wiesinger F, Sekine T. Improving PET/MR brain quantitation with template-enhanced ZTE. Neuroimage 2018; 181:403-413. [PMID: 30010010 DOI: 10.1016/j.neuroimage.2018.07.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 06/20/2018] [Accepted: 07/12/2018] [Indexed: 10/28/2022] Open
Abstract
PURPOSE The impact of MR-based attenuation correction on PET quantitation accuracy is an ongoing cause of concern for advanced brain research with PET/MR. The purpose of this study was to evaluate a new, template-enhanced zero-echo-time attenuation correction method for PET/MR scanners. METHODS 30 subjects underwent a clinically-indicated 18F-FDG-PET/CT, followed by PET/MR on a GE SIGNA PET/MR. For each patient, a 42-s zero echo time (ZTE) sequence was used to generate two attenuation maps: one with the standard ZTE segmentation-based method; and another with a modification of the method, wherein pre-registered anatomical templates and CT data were used to enhance the segmentation. CT data, was used as gold standard. Reconstructed PET images were qualified visually and quantified in 68 volumes-of-interest using a standardized brain atlas. RESULTS Attenuation maps were successfully generated in all cases, without manual intervention or parameter tuning. One patient was excluded from the quantitative analysis due to the presence of multiple brain metastases. The PET bias with template-enhanced ZTE attenuation correction was measured to be -0.9% ± 0.9%, compared with -1.4% ± 1.1% with regular ZTE attenuation correction. In terms of absolute bias, the new method yielded 1.1% ± 0.7%, compared with 1.6% ± 0.9% with regular ZTE. Statistically significant bias reduction was obtained in the frontal region (from -2.0% to -1.0%), temporal (from -1.2% to -0.2%), parietal (from -1.9% to -1.1%), occipital (from -2.0% to -1.1%) and insula (from -1.4% to -1.1%). CONCLUSION These results indicate that the co-registration of pre-recorded anatomical templates to ZTE data is feasible in clinical practice and can be effectively used to improve the performance of segmentation-based attenuation correction.
Collapse
Affiliation(s)
| | - Bradley Kemp
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Tetsuro Sekine
- Department of Radiology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
505
|
Golla SSV, Timmers T, Ossenkoppele R, Groot C, Verfaillie S, Scheltens P, van der Flier WM, Schwarte L, Mintun MA, Devous M, Schuit RC, Windhorst AD, Lammertsma AA, Boellaard R, van Berckel BNM, Yaqub M. Quantification of Tau Load Using [ 18F]AV1451 PET. Mol Imaging Biol 2018; 19:963-971. [PMID: 28374171 PMCID: PMC5662681 DOI: 10.1007/s11307-017-1080-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Purpose The tau tracer [18F]AV1451, also known as flortaucipir, is a promising ligand for imaging tau accumulation in Alzheimer’s disease (AD). Most of the previous studies have quantified tau load using standardized uptake value ratios (SUVr) derived from a static [18F]AV1451 scan. SUVr may, however, be flow dependent and, especially for longitudinal studies, should be validated against a fully quantitative approach. The objective of this study was to identify the optimal tracer kinetic model for measuring tau load using [18F]AV1451. Procedures Following intravenous injection of 225 ± 16 MBq [18F]AV1451, 130 min dynamic PET scans were performed in five biomarker confirmed AD patients and five controls. Arterial blood sampling was performed to obtain a metabolite-corrected plasma input function. Next, regional time–activity curves were generated using PVElab software. These curves were analysed using several pharmacokinetic models. Results The reversible single tissue compartment model (1T2k_VB) was the preferred model for all but one control. For AD patients, however, model preference shifted towards a reversible two tissue compartmental model (2T4k_VB). The simplified reference tissue model (SRTM) derived binding potential (BPND) showed good correlation (AD: r2 = 0.87, slope = 1.06; controls: r2 = 0.87, slope = 0.86) with indirect plasma input binding (distribution volume ratio-1). Standardized uptake value ratios (80–100 min) correlated well with DVR (r2 = 0.93, slope = 1.07) and SRTM-derived BPND (r2 = 0.84, slope = 0.95). In addition, regional differences in tracer binding between subject groups in different tau-specific regions were observed. Conclusions Model preference of [18F]AV1451 appears to depend on subject status and, in particular, VT. The relationship between model preference and VT suggests that (higher) tau load may be reflected by a second tissue compartment. Nevertheless, consistent results can be obtained using a 2T4k_VB model. In addition, SRTM can be used to derive BPND. Electronic supplementary material The online version of this article (doi:10.1007/s11307-017-1080-z) 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, Amsterdam, Netherlands.
| | - Tessa Timmers
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands.,Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands.,Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, Netherlands
| | - Colin Groot
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Sander Verfaillie
- Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, Netherlands
| | - Philip Scheltens
- Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, Netherlands.,Department of Epidemiology & Biostatistics, VU University Medical Center, Amsterdam, Netherlands
| | - Lothar Schwarte
- Department of Anaesthesiology, VU University Medical center, Amsterdam, Netherlands
| | | | | | - Robert C Schuit
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands.,Department of Nuclear Medicine & Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands.,Alzheimer Center & Department of Neurology, VU University Medical Center, Amsterdam, Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
506
|
Trebaul L, Deman P, Tuyisenge V, Jedynak M, Hugues E, Rudrauf D, Bhattacharjee M, Tadel F, Chanteloup-Foret B, Saubat C, Reyes Mejia GC, Adam C, Nica A, Pail M, Dubeau F, Rheims S, Trébuchon A, Wang H, Liu S, Blauwblomme T, Garcés M, De Palma L, Valentin A, Metsähonkala EL, Petrescu AM, Landré E, Szurhaj W, Hirsch E, Valton L, Rocamora R, Schulze-Bonhage A, Mindruta I, Francione S, Maillard L, Taussig D, Kahane P, David O. Probabilistic functional tractography of the human cortex revisited. Neuroimage 2018; 181:414-429. [PMID: 30025851 PMCID: PMC6150949 DOI: 10.1016/j.neuroimage.2018.07.039] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/21/2018] [Accepted: 07/15/2018] [Indexed: 12/13/2022] Open
Abstract
In patients with pharmaco-resistant focal epilepsies investigated with intracranial electroencephalography (iEEG), direct electrical stimulations of a cortical region induce cortico-cortical evoked potentials (CCEP) in distant cerebral cortex, which properties can be used to infer large scale brain connectivity. In 2013, we proposed a new probabilistic functional tractography methodology to study human brain connectivity. We have now been revisiting this method in the F-TRACT project (f-tract.eu) by developing a large multicenter CCEP database of several thousand stimulation runs performed in several hundred patients, and associated processing tools to create a probabilistic atlas of human cortico-cortical connections. Here, we wish to present a snapshot of the methods and data of F-TRACT using a pool of 213 epilepsy patients, all studied by stereo-encephalography with intracerebral depth electrodes. The CCEPs were processed using an automated pipeline with the following consecutive steps: detection of each stimulation run from stimulation artifacts in raw intracranial EEG (iEEG) files, bad channels detection with a machine learning approach, model-based stimulation artifact correction, robust averaging over stimulation pulses. Effective connectivity between the stimulated and recording areas is then inferred from the properties of the first CCEP component, i.e. onset and peak latency, amplitude, duration and integral of the significant part. Finally, group statistics of CCEP features are implemented for each brain parcel explored by iEEG electrodes. The localization (coordinates, white/gray matter relative positioning) of electrode contacts were obtained from imaging data (anatomical MRI or CT scans before and after electrodes implantation). The iEEG contacts were repositioned in different brain parcellations from the segmentation of patients' anatomical MRI or from templates in the MNI coordinate system. The F-TRACT database using the first pool of 213 patients provided connectivity probability values for 95% of possible intrahemispheric and 56% of interhemispheric connections and CCEP features for 78% of intrahemisheric and 14% of interhemispheric connections. In this report, we show some examples of anatomo-functional connectivity matrices, and associated directional maps. We also indicate how CCEP features, especially latencies, are related to spatial distances, and allow estimating the velocity distribution of neuronal signals at a large scale. Finally, we describe the impact on the estimated connectivity of the stimulation charge and of the contact localization according to the white or gray matter. The most relevant maps for the scientific community are available for download on f-tract. eu (David et al., 2017) and will be regularly updated during the following months with the addition of more data in the F-TRACT database. This will provide an unprecedented knowledge on the dynamical properties of large fiber tracts in human.
Collapse
Affiliation(s)
- Lena Trebaul
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Pierre Deman
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Viateur Tuyisenge
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Maciej Jedynak
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Etienne Hugues
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - David Rudrauf
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Manik Bhattacharjee
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - François Tadel
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Blandine Chanteloup-Foret
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Carole Saubat
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Gina Catalina Reyes Mejia
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France
| | - Claude Adam
- Epilepsy Unit, Dept of Neurology, Pitié-Salpêtrière Hospital, APHP, Paris, France
| | - Anca Nica
- Neurology Department, CHU, Rennes, France
| | - Martin Pail
- Brno Epilepsy Center, Department of Neurology, St. Anne's University Hospital and Medical Faculty of Masaryk University, Brno, Czech Republic
| | - François Dubeau
- Montreal Neurological Institute and Hospital, Montreal, Canada
| | - Sylvain Rheims
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and University of Lyon, Lyon, France
| | - Agnès Trébuchon
- Service de Neurophysiologie Clinique, APHM, Hôpitaux de la Timone, Marseille, France
| | - Haixiang Wang
- Yuquan Hospital Epilepsy Center, Tsinghua University, Beijing, China
| | - Sinclair Liu
- Canton Sanjiu Brain Hospital Epilepsy Center, Jinan University, Guangzhou, China
| | - Thomas Blauwblomme
- Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, Université Paris V Descartes, Sorbonne Paris Cité, Paris, France
| | - Mercedes Garcés
- Multidisciplinary Epilepsy Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Luca De Palma
- Department of Neuroscience, Bambino Gesù Children's Hospital, IRRCS, Rome, Italy
| | - Antonio Valentin
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), London, UK
| | | | | | | | - William Szurhaj
- Epilepsy Unit, Department of Clinical Neurophysiology, Lille University Medical Center, Lille, France
| | - Edouard Hirsch
- University Hospital, Department of Neurology, Strasbourg, France
| | - Luc Valton
- University Hospital, Department of Neurology, Toulouse, France
| | - Rodrigo Rocamora
- Epilepsy Monitoring Unit, Department of Neurology, Hospital del Mar-IMIM, Barcelona, Spain
| | - Andreas Schulze-Bonhage
- Epilepsy Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Ioana Mindruta
- Neurology Department, University Emergency Hospital, Bucharest, Romania
| | | | - Louis Maillard
- Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - Delphine Taussig
- Service de neurochirurgie pédiatrique, Fondation Rothschild, Paris, France
| | - Philippe Kahane
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France; CHU Grenoble Alpes, Neurology Department, Grenoble, France
| | - Olivier David
- Inserm, U1216, Grenoble, F-38000, France; Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, F-38000, France.
| |
Collapse
|
507
|
Deman P, Bhattacharjee M, Tadel F, Job AS, Rivière D, Cointepas Y, Kahane P, David O. IntrAnat Electrodes: A Free Database and Visualization Software for Intracranial Electroencephalographic Data Processed for Case and Group Studies. Front Neuroinform 2018; 12:40. [PMID: 30034332 PMCID: PMC6043781 DOI: 10.3389/fninf.2018.00040] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 06/05/2018] [Indexed: 11/13/2022] Open
Abstract
In some cases of pharmaco-resistant and focal epilepsies, intracranial recordings performed epidurally (electrocorticography, ECoG) and/or in depth (stereoelectroencephalography, SEEG) can be required to locate the seizure onset zone and the eloquent cortex before surgical resection. In SEEG, each electrode contact records brain’s electrical activity in a spherical volume of 3 mm diameter approximately. The spatial coverage is around 1% of the brain and differs between patients because the implantation of electrodes is tailored for each case. Group studies thus need a large number of patients to reach a large spatial sampling, which can be achieved more easily using a multicentric approach such as implemented in our F-TRACT project (f-tract.eu). To facilitate group studies, we developed a software—IntrAnat Electrodes—that allows to perform virtual electrode implantation in patients’ neuroanatomy and to overlay results of epileptic and functional mapping, as well as resection masks from the surgery. IntrAnat Electrodes is based on a patient database providing multiple search criteria to highlight various group features. For each patient, the anatomical processing is based on a series of software publicly available. Imaging modalities (Positron Emission Tomography (PET), anatomical MRI pre-implantation, post-implantation and post-resection, functional MRI, diffusion MRI, Computed Tomography (CT) with electrodes) are coregistered. The 3D T1 pre-implantation MRI gray/white matter is segmented and spatially normalized to obtain a series of cortical parcels using different neuroanatomical atlases. On post-implantation images, the user can position 3D models of electrodes defined by their geometry. Each electrode contact is then labeled according to its position in the anatomical atlases, to the class of tissue (gray or white matter, cerebro-spinal fluid) and to its presence inside or outside the resection mask. Users can add more functionally informed labels on contact, such as clinical responses after electrical stimulation, cortico-cortical evoked potentials, gamma band activity during cognitive tasks or epileptogenicity. IntrAnat Electrodes software thus provides a means to visualize multimodal data. The contact labels allow to search for patients in the database according to multiple criteria representing almost all available data, which is to our knowledge unique in current SEEG software. IntrAnat Electrodes will be available in the forthcoming release of BrainVisa software and tutorials can be found on the F-TRACT webpage.
Collapse
Affiliation(s)
- Pierre Deman
- Inserm, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, Grenoble, France
| | - Manik Bhattacharjee
- Inserm, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, Grenoble, France
| | - François Tadel
- Inserm, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, Grenoble, France
| | - Anne-Sophie Job
- Inserm, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, Grenoble, France.,Neurology Department, Grenoble-Alpes University Hospital, Grenoble, France
| | - Denis Rivière
- Neurospin-CEA Saclay, UNATI Lab, Gif sur Yvette, France.,Multicenter Neuroimaging Platform, CATI, Paris, France
| | - Yann Cointepas
- Neurospin-CEA Saclay, UNATI Lab, Gif sur Yvette, France.,Multicenter Neuroimaging Platform, CATI, Paris, France
| | - Philippe Kahane
- Inserm, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, Grenoble, France.,Neurology Department, Grenoble-Alpes University Hospital, Grenoble, France
| | - Olivier David
- Inserm, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,Grenoble Institut des Neurosciences (GIN), University of Grenoble Alpes, Grenoble, France
| |
Collapse
|
508
|
Rate of β-amyloid accumulation varies with baseline amyloid burden: Implications for anti-amyloid drug trials. Alzheimers Dement 2018; 14:1387-1396. [PMID: 30420035 DOI: 10.1016/j.jalz.2018.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/06/2018] [Accepted: 05/28/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION This study examined a longitudinal trajectory of β-amyloid (Aβ) accumulation at the predementia stage of Alzheimer's disease in the context of clinical trials. METHODS Analyzed were baseline (BL) and 2 years' follow-up 18F-florbetapir positron emission tomography data of 246 Aβ-positive subjects with normal cognition and mild cognitive impairment. We studied the relationship between annual accumulation rates of 18F-florbetapir and BL standard uptake value ratios in whole gray matter (SUVRGM). RESULTS Subjects with BL SUVRGM of 0.56 to 0.92 (n = 134) appeared to accumulate Aβ approximately 1.5 times faster than remaining subjects. In subjects with SUVRGM above 0.95, most regions with the highest annual accumulation rate were outside the established set of Alzheimer's disease typical regions. CONCLUSION There are global and regional variations in annual accumulation rate at the predementia stage of Alzheimer's disease. When taken into account, the sample size in anti-amyloid trials can be substantially reduced. Critically, treated and placebo groups should be matched for BL SUVRGM.
Collapse
|
509
|
Richards EM, Zanotti-Fregonara P, Fujita M, Newman L, Farmer C, Ballard ED, Machado-Vieira R, Yuan P, Niciu MJ, Lyoo CH, Henter ID, Salvadore G, Drevets WC, Kolb H, Innis RB, Zarate Jr CA. PET radioligand binding to translocator protein (TSPO) is increased in unmedicated depressed subjects. EJNMMI Res 2018; 8:57. [PMID: 29971587 PMCID: PMC6029989 DOI: 10.1186/s13550-018-0401-9] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/30/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Inflammation is associated with major depressive disorder (MDD). Translocator protein 18 kDa (TSPO), a putative biomarker of neuroinflammation, is quantified using positron emission tomography (PET) and 11C-PBR28, a TSPO tracer. We sought to (1) investigate TSPO binding in MDD subjects currently experiencing a major depressive episode, (2) investigate the effects of antidepressants on TSPO binding, and (3) determine the relationship of peripheral and central inflammatory markers to cerebral TSPO binding. Twenty-eight depressed MDD subjects (unmedicated (n = 12) or medicated (n = 16)) and 20 healthy controls (HC) underwent PET imaging using 11C-PBR28. Total distribution volume (VT, proportional to Bmax/Kd) was measured and corrected with the free fraction in plasma (fp). The subgenual prefrontal cortex (sgPFC) and anterior cingulate cortex (ACC) were the primary regions of interest. Peripheral blood samples and cerebrospinal fluid were analyzed to investigate the relationship between TSPO binding and peripheral and central inflammatory markers, including interleukins and neurotrophic factors previously linked to depression. RESULTS TSPO binding was higher in MDD versus HC in the sgPFC (Cohen's d = 0.64, p = .038, 95% CI 0.04-1.24) and ACC (d = 0.60, p = .049, 95% CI 0.001-1.21), though these comparisons missed the corrected threshold for statistical significance (α = .025). Exploratory analyses demonstrated that unmedicated MDD subjects had the highest level of TSPO binding, followed by medicated MDD subjects, who did not differ from HC. TSPO binding correlated with interleukin-5 in cerebrospinal fluid but with no other central inflammatory markers. CONCLUSIONS This study found a trend towards increased TSPO binding in the brains of MDD subjects, and post hoc analysis extended these findings by demonstrating that this abnormality is significant in unmedicated (but not medicated) MDD subjects.
Collapse
Affiliation(s)
- Erica M. Richards
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | | | - Masahiro Fujita
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | - Laura Newman
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | - Cristan Farmer
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | - Elizabeth D. Ballard
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | - Rodrigo Machado-Vieira
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center, Houston, TX USA
| | - Peixiong Yuan
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | - Mark J. Niciu
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ioline D. Henter
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | | | | | - Hartmuth Kolb
- Janssen Research and Development, LLC, Titusville, NJ USA
| | - Robert B. Innis
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| | - Carlos A. Zarate Jr
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Building 10, CRC Room 6-5340, 10 Center Drive, Bethesda, MD 20892 USA
| |
Collapse
|
510
|
Xie W, Mallin BM, Richards JE. Development of brain functional connectivity and its relation to infant sustained attention in the first year of life. Dev Sci 2018; 22:e12703. [PMID: 29968370 DOI: 10.1111/desc.12703] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/24/2018] [Indexed: 11/28/2022]
Abstract
The study of brain functional connectivity is crucial to understanding the neural mechanisms underlying the improved behavioral performance and amplified ERP responses observed during infant sustained attention. Previous investigations on the development of functional brain connectivity during infancy are primarily confined to the use of functional and structural MRI techniques. The current study examined the relation between infant sustained attention and brain functional connectivity and their development during infancy with high-density EEG recordings. Fifty-nine infants were tested at 6 (N = 15), 8 (N =14), 10 (N = 17), and 12 (N = 13) months. Infant sustained attention was defined by measuring infant heart rate changes during infants' looking. Functional connectivity was estimated from the electrodes on the scalp and with reconstructed cortical source activities in brain regions. It was found that infant sustained attention was accompanied by attenuated functional connectivity in the dorsal attention and default mode networks in the alpha band. Graph theory analyses showed that there was an increase in path length and a decrease in clustering coefficient during infant sustained attention. The functional connectivity within the visual, somatosensory, dorsal attention, and ventral attention networks and graph theory measures of path length and clustering coefficient were found to increase with age. These findings suggest that infant sustained attention is accompanied by distinct patterns of brain functional connectivity. The current findings also suggest the rapid development of functional connectivity in brain networks during infancy.
Collapse
Affiliation(s)
- Wanze Xie
- Department of Psychology, University of South Carolina, Columbia, South Carolina.,Institute for Mind and Brain, University of South Carolina, Columbia, South Carolina.,Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - John E Richards
- Department of Psychology, University of South Carolina, Columbia, South Carolina.,Institute for Mind and Brain, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
511
|
Chiotis K, Saint-Aubert L, Rodriguez-Vieitez E, Leuzy A, Almkvist O, Savitcheva I, Jonasson M, Lubberink M, Wall A, Antoni G, Nordberg A. Longitudinal changes of tau PET imaging in relation to hypometabolism in prodromal and Alzheimer's disease dementia. Mol Psychiatry 2018; 23:1666-1673. [PMID: 28507319 DOI: 10.1038/mp.2017.108] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/14/2017] [Accepted: 04/04/2017] [Indexed: 12/17/2022]
Abstract
The development of tau-specific positron emission tomography (PET) tracers allows imaging in vivo the regional load of tau pathology in Alzheimer's disease (AD) and other tauopathies. Eighteen patients with baseline investigations enroled in a 17-month follow-up study, including 16 with AD (10 had mild cognitive impairment and a positive amyloid PET scan, that is, prodromal AD, and six had AD dementia) and two with corticobasal syndrome. All patients underwent PET scans with [18F]THK5317 (tau deposition) and [18F]FDG (glucose metabolism) at baseline and follow-up, neuropsychological assessment at baseline and follow-up and a scan with [11C]PIB (amyloid-β deposition) at baseline only. At a group level, patients with AD (prodromal or dementia) showed unchanged [18F]THK5317 retention over time, in contrast to significant decreases in [18F]FDG uptake in temporoparietal areas. The pattern of changes in [18F]THK5317 retention was heterogeneous across all patients, with qualitative differences both between the two AD groups (prodromal and dementia) and among individual patients. High [18F]THK5317 retention was significantly associated over time with low episodic memory encoding scores, while low [18F]FDG uptake was significantly associated over time with both low global cognition and episodic memory encoding scores. Both patients with corticobasal syndrome had a negative [11C]PIB scan, high [18F]THK5317 retention with a different regional distribution from that in AD, and a homogeneous pattern of increased [18F]THK5317 retention in the basal ganglia over time. These findings highlight the heterogeneous propagation of tau pathology among patients with symptomatic AD, in contrast to the homogeneous changes seen in glucose metabolism, which better tracked clinical progression.
Collapse
Affiliation(s)
- K Chiotis
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - L Saint-Aubert
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - E Rodriguez-Vieitez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - A Leuzy
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - O Almkvist
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Department of Psychology, Stockholm University, Stockholm, Sweden
| | - I Savitcheva
- Department of Radiology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - M Jonasson
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,Department of Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - M Lubberink
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,Department of Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - A Wall
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - G Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - A Nordberg
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden. .,Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
512
|
Mutlu U, Ikram MK, Roshchupkin GV, Bonnemaijer PWM, Colijn JM, Vingerling JR, Niessen WJ, Ikram MA, Klaver CCW, Vernooij MW. Thinner retinal layers are associated with changes in the visual pathway: A population-based study. Hum Brain Mapp 2018; 39:4290-4301. [PMID: 29935103 DOI: 10.1002/hbm.24246] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/26/2018] [Accepted: 05/29/2018] [Indexed: 01/23/2023] Open
Abstract
Increasing evidence shows that thinner retinal nerve fiber layer (RNFL) and ganglion cell layer (GCL), assessed on optical coherence tomography (OCT), are reflecting global brain atrophy. Yet, little is known on the relation of these layers with specific brain regions. Using voxel-based analysis, we aimed to unravel specific brain regions associated with these retinal layers. We included 2,235 persons (mean age: 67.3 years, 55% women) from the Rotterdam Study (2007-2012) who had gradable retinal OCT images and brain magnetic resonance imaging (MRI) scans, including diffusion tensor (DT) imaging. Thicknesses of peripapillary RNFL and perimacular GCL were measured using an automated segmentation algorithm. Voxel-based morphometry protocols were applied to process DT-MRI data. We investigated the association between retinal layer thickness with voxel-wise gray matter density and white matter microstructure by performing linear regression models. We found that thinner RNFL and GCL were associated with lower gray matter density in the visual cortex, and with lower fractional anisotropy and higher mean diffusivity in white matter tracts that are part of the optic radiation. Furthermore, thinner GCL was associated with lower gray matter density of the thalamus. Thinner RNFL and GCL are associated with gray and white matter changes in the visual pathway suggesting that retinal thinning on OCT may be specifically associated with changes in the visual pathway rather than with changes in the global brain. These findings may serve as a basis for understanding visual symptoms in elderly patients, patients with Alzheimer's disease, or patients with posterior cortical atrophy.
Collapse
Affiliation(s)
- Unal Mutlu
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Mohammad K Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Gennady V Roshchupkin
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Pieter W M Bonnemaijer
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Johanna M Colijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Johannes R Vingerling
- Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wiro J Niessen
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Imaging Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Mohammad A Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Caroline C W Klaver
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
513
|
van Holst RJ, Sescousse G, Janssen LK, Janssen M, Berry AS, Jagust WJ, Cools R. Increased Striatal Dopamine Synthesis Capacity in Gambling Addiction. Biol Psychiatry 2018; 83:1036-1043. [PMID: 28728675 PMCID: PMC6698370 DOI: 10.1016/j.biopsych.2017.06.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/15/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The hypothesis that dopamine plays an important role in the pathophysiology of pathological gambling is pervasive. However, there is little to no direct evidence for a categorical difference between pathological gamblers and healthy control subjects in terms of dopamine transmission in a drug-free state. Here we provide evidence for this hypothesis by comparing dopamine synthesis capacity in the dorsal and ventral parts of the striatum in 13 pathological gamblers and 15 healthy control subjects. METHODS This was achieved using [18F]fluoro-levo-dihydroxyphenylalanine dynamic positron emission tomography scans and striatal regions of interest that were hand-drawn based on visual inspection of individual structural magnetic resonance imaging scans. RESULTS Our results show that dopamine synthesis capacity was increased in pathological gamblers compared with healthy control subjects. Dopamine synthesis was 16% higher in the caudate body, 17% higher in the dorsal putamen, and 17% higher in the ventral striatum in pathological gamblers compared with control subjects. Moreover, dopamine synthesis capacity in the dorsal putamen and caudate head was positively correlated with gambling distortions in pathological gamblers. CONCLUSIONS Taken together, these results provide empirical evidence for increased striatal dopamine synthesis in pathological gambling.
Collapse
|
514
|
Does inflammation precede tau aggregation in early Alzheimer's disease? A PET study. Neurobiol Dis 2018; 117:211-216. [PMID: 29902557 DOI: 10.1016/j.nbd.2018.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 05/12/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Our aim was to assess with positron emission tomography (PET) the temporal and spatial inter-relationships between levels of cortical microglial activation and the aggregated amyloid-β and tau load in mild cognitive impairment (MCI) and early Alzheimer's disease (AD). METHODS Six clinically probable AD and 20 MCI subjects had inflammation (11C-(R)-PK11195), amyloid (11C-PiB) and tau (18F-flortaucipir) PET, magnetic resonance imaging (MRI) and a neuropsychological assessment. Parametric images of tracer binding were interrogated at a voxel level and by region of interest analyses. RESULTS 55% of MCI and 83% of AD subjects had a high amyloid-β load. We have previously reported that clusters of correlated amyloid and inflammation levels are present in cortex. Here we found no correlation between levels of inflammation (11C-(R)-PK11195 BPND) and tau (18F-flortaucipir SUVR) or MMSE scores in high amyloid-β cases. INTERPRETATION While correlated levels of amyloid-β and inflammation can be seen in MCI, we did not detect an association between levels of cortical tau tangles and inflammation in our series of high amyloid-β cases. High levels of inflammation could be seen in amyloid-β positive MCI cases where 18F-flortaucipir signals were low suggesting microglial activation precedes tau tangle formation. Inflammation levels were higher in high amyloid-β MCI than in early AD cases, compatible with it initially playing a protective role.
Collapse
|
515
|
Lohith TG, Bennacef I, Vandenberghe R, Vandenbulcke M, Salinas CA, Declercq R, Reynders T, Telan-Choing NF, Riffel K, Celen S, Serdons K, Bormans G, Tsai K, Walji A, Hostetler ED, Evelhoch JL, Van Laere K, Forman M, Stoch A, Sur C, Struyk A. Brain Imaging of Alzheimer Dementia Patients and Elderly Controls with 18F-MK-6240, a PET Tracer Targeting Neurofibrillary Tangles. J Nucl Med 2018; 60:107-114. [PMID: 29880509 DOI: 10.2967/jnumed.118.208215] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/14/2018] [Indexed: 11/16/2022] Open
Abstract
18F-MK-6240 (18F-labeled 6-(fluoro)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine) is a highly selective, subnanomolar-affinity PET tracer for imaging neurofibrillary tangles (NFTs). Plasma kinetics, brain uptake, and preliminary quantitative analysis of 18F-MK-6240 in healthy elderly (HE) subjects, subjects with clinically probable Alzheimer disease (AD), and subjects with amnestic mild cognitive impairment were characterized in a study that is, to our knowledge, the first to be performed on humans. Methods: Dynamic PET scans of up to 150 min were performed on 4 cognitively normal HE subjects, 4 AD subjects, and 2 amnestic mild cognitive impairment subjects after a bolus injection of 152-169 MBq of 18F-MK-6240 to evaluate tracer kinetics and distribution in brain. Regional SUV ratio (SUVR) and distribution volume ratio were determined using the cerebellar cortex as a reference region. Total distribution volume was assessed by compartmental modeling using radiometabolite-corrected input function in a subgroup of 6 subjects. Results: 18F-MK-6240 had rapid brain uptake with a peak SUV of 3-5, followed by a uniformly quick washout from all brain regions in HE subjects; slower clearance was observed in regions commonly associated with NFT deposition in AD subjects. In AD subjects, SUVR between 60 and 90 min after injection was high (approximately 2-4) in regions associated with NFT deposition, whereas in HE subjects, SUVR was approximately 1 across all brain regions, suggesting high tracer selectivity for binding NFTs in vivo. 18F-MK-6240 total distribution volume was approximately 2- to 3-fold higher in neocortical and medial temporal brain regions of AD subjects than in HE subjects and stabilized by 60 min in both groups. Distribution volume ratio estimated by the Logan reference tissue model or compartmental modeling correlated well (R 2 > 0.9) to SUVR from 60 to 90 min for AD subjects. Conclusion: 18F-MK-6240 exhibited favorable kinetics and high binding levels to brain regions with a plausible pattern for NFT deposition in AD subjects. In comparison, negligible tracer binding was observed in HE subjects. This pilot study suggests that simplified ratio methods such as SUVR can be used to quantify NFT binding. These results support further clinical development of 18F-MK-6240 for potential application in longitudinal studies.
Collapse
Affiliation(s)
- Talakad G Lohith
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Idriss Bennacef
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Rik Vandenberghe
- Neurology Department, University Hospitals Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Alzheimer Research Centre, KU Leuven, Leuven Institute for Neuroscience and Disease, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Neurology Department, University Hospitals Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Alzheimer Research Centre, KU Leuven, Leuven Institute for Neuroscience and Disease, Leuven, Belgium
| | | | - Ruben Declercq
- Translational Pharmacology Europe, Merck Sharp & Dohme Corp., Brussels, Belgium
| | - Tom Reynders
- Translational Pharmacology Europe, Merck Sharp & Dohme Corp., Brussels, Belgium
| | | | - Kerry Riffel
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Sofie Celen
- Radiopharmaceutical Research, KU Leuven, Leuven, Belgium
| | - Kim Serdons
- Division of Nuclear Medicine, University Hospitals Leuven and UZ Leuven, Leuven, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, KU Leuven, Leuven, Belgium
| | - Kuenhi Tsai
- Early Clinical Statistics, Merck & Co., Inc., West Point, Pennsylvania
| | - Abbas Walji
- Discovery Chemistry, Merck & Co., Inc., West Point, Pennsylvania; and
| | - Eric D Hostetler
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | | | - Koen Van Laere
- Division of Nuclear Medicine, University Hospitals Leuven and UZ Leuven, Leuven, Belgium
| | - Mark Forman
- Translational Pharmacology, Merck & Co., Inc., North Wales, Pennsylvania
| | - Aubrey Stoch
- Translational Pharmacology, Merck & Co., Inc., North Wales, Pennsylvania
| | - Cyrille Sur
- Translational Biomarkers, Merck & Co., Inc., West Point, Pennsylvania
| | - Arie Struyk
- Translational Pharmacology, Merck & Co., Inc., North Wales, Pennsylvania
| |
Collapse
|
516
|
A longitudinal study on deep brain stimulation of the medial forebrain bundle for treatment-resistant depression. Transl Psychiatry 2018; 8:111. [PMID: 29867109 PMCID: PMC5986795 DOI: 10.1038/s41398-018-0160-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/15/2018] [Indexed: 11/25/2022] Open
Abstract
Deep brain stimulation (DBS) to the superolateral branch of the medial forebrain bundle (MFB) has been reported to lead to rapid antidepressant effects. In this longitudinal study, we expand upon the initial results we reported at 26 weeks (Fenoy et al., 2016), showing sustained antidepressant effects of MFB DBS on six patients with treatment-resistant depression (TRD) over 1 year. The Montgomery-Åsberg Depression Rating Scale (MADRS) was used as the primary assessment tool. Deterministic fiber tracking was used to individually map the target area; analysis was performed to compare modulated fiber tracts between patients. Intraoperatively, upon stimulation at target, responders reported immediate increases in energy and motivation. An insertional effect was seen during the 4-week sham stimulation phase from baseline (28% mean MADRS reduction, p = 0.02). However, after 1 week of initiating stimulation, three of six patients had a > 50% decrease in MADRS scores relative to baseline (43% mean MADRS reduction, p = 0.005). One patient withdrew from study participation. At 52 weeks, four of remaining five patients have > 70% decrease in MADRS scores relative to baseline (73% mean MADRS reduction, p = 0.007). Evaluation of modulated fiber tracts reveals significant common orbitofrontal connectivity to the target region in all responders. Neuropsychological testing and 18F-fluoro-deoxyglucose-positron emission tomography cerebral metabolism evaluations performed at baseline and at 52 weeks showed minimal changes and verified safety. This longitudinal evaluation of MFB DBS demonstrated rapid antidepressant effects, as initially reported by Schlaepfer et al. (2013), and supports the use of DBS for TRD.
Collapse
|
517
|
Thordardottir S, Rodriguez-Vieitez E, Almkvist O, Ferreira D, Saint-Aubert L, Kinhult-Ståhlbom A, Thonberg H, Schöll M, Westman E, Wall A, Eriksdotter M, Zetterberg H, Blennow K, Nordberg A, Graff C. Reduced penetrance of the PSEN1 H163Y autosomal dominant Alzheimer mutation: a 22-year follow-up study. ALZHEIMERS RESEARCH & THERAPY 2018; 10:45. [PMID: 29747683 PMCID: PMC5944151 DOI: 10.1186/s13195-018-0374-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/17/2018] [Indexed: 12/14/2022]
Abstract
Background The range of onset ages within some PSEN1 families is wide, and a few cases of reduced penetrance of PSEN1 mutations have been reported. However, published data on reduced penetrance have been limited to clinical histories, often collected retrospectively and lacking biomarker information. We present a case of reduced penetrance of the PSEN1 H163Y mutation in a carrier prospectively followed for 22 years. Methods Two brothers (A and B), both carriers of the H163Y mutation, were followed between 1995 and 2017. They underwent repeated clinical evaluations, neuropsychological assessments, and cerebrospinal fluid analyses, as well as brain imaging examinations with structural magnetic resonance, [18F]fluorodeoxyglucose positron emission tomography, and [11C]Pittsburgh compound B positron emission tomography. Results Brother A was followed between 44 and 64 years of age. Cognitive symptoms due to Alzheimer’s disease set in at the age of 54. Gradual worsening of symptoms resulted in admittance to a nursing home owing to dependence on others for all activities of daily living. He showed a curvilinear decline in cognitive function on neuropsychological tests, and changes on magnetic resonance imaging, positron emission tomography, and biomarkers in the cerebrospinal fluid supported a clinical diagnosis of Alzheimer’s disease. Brother A died at the age of 64 and fulfilled the criteria for definitive Alzheimer’s disease according to neuropathological examination results. Brother B was followed between the ages of 43 and 65 and showed no cognitive deterioration on repeated neuropsychological test occasions. In addition, no biomarker evidence of Alzheimer’s disease pathology was detected, either on imaging examinations or in cerebrospinal fluid. Conclusions The average (SD) age of symptom onset for PSEN1 H163Y is 51 ± 7 years according to previous studies. However, we present a case of a biomarker-verified reduction in penetrance in a mutation carrier who was still symptom-free at the age of 65. This suggests that other genetic, epigenetic, and/or environmental factors modify the onset age. Electronic supplementary material The online version of this article (10.1186/s13195-018-0374-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Steinunn Thordardottir
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, 141 57, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden
| | - Elena Rodriguez-Vieitez
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, 141 57, Huddinge, Sweden
| | - Ove Almkvist
- Theme Aging, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden.,Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, 141 57, Huddinge, Sweden.,Department of Psychology, Stockholm University, 106 91, Stockholm, Sweden
| | - Daniel Ferreira
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, 141 57, Huddinge, Sweden
| | - Laure Saint-Aubert
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, 141 57, Huddinge, Sweden.,Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Anne Kinhult-Ståhlbom
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, 141 57, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden
| | - Håkan Thonberg
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, 141 57, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, 413 45, Gothenburg, Sweden.,Clinical Memory Research Unit, Lund University, 212 24, Malmö, Sweden
| | - Eric Westman
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, 141 57, Huddinge, Sweden
| | - Anders Wall
- Uppsala University, Department of Surgical Sciences, Section of Nuclear Medicine & PET, 751 85, Uppsala, Sweden
| | - Maria Eriksdotter
- Theme Aging, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden.,Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, 141 57, Huddinge, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 431 80, Mölndal, Sweden.,UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.,UK Dementia Research Institute at UCL, London, WC1N 3BG, UK.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Mölndal, Sweden
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, 431 80, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Mölndal, Sweden
| | - Agneta Nordberg
- Theme Aging, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden.,Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Translational Alzheimer Neurobiology, 141 57, Huddinge, Sweden
| | - Caroline Graff
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, 141 57, Huddinge, Sweden. .,Theme Aging, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden.
| |
Collapse
|
518
|
Leurquin-Sterk G, Ceccarini J, Crunelle CL, Weerasekera A, de Laat B, Himmelreich U, Bormans G, Van Laere K. Cerebral dopaminergic and glutamatergic transmission relate to different subjective responses of acute alcohol intake: an in vivo multimodal imaging study. Addict Biol 2018; 23:931-944. [PMID: 28884874 DOI: 10.1111/adb.12542] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/14/2017] [Accepted: 07/03/2017] [Indexed: 12/21/2022]
Abstract
Converging preclinical evidence links extrastriatal dopamine release and glutamatergic transmission via the metabotropic glutamate receptor 5 (mGluR5) to the rewarding properties of alcohol. To date, human evidence is lacking on how and where in the brain these processes occur. Mesocorticolimbic dopamine release upon intravenous alcohol administration and mGluR5 availability were measured in 11 moderate social drinkers by single-session [18 F]fallypride and [18 F]FPEB positron emission tomography, respectively. Additionally, baseline and postalcohol glutamate and glutamine levels in the anterior cingulate cortex (ACC) were measured by using proton-magnetic resonance spectroscopy. To investigate differences in reward domains linked to both neurotransmitters, regional imaging data were related to subjective alcohol responses. Alcohol induced significant [18 F]fallypride displacement in the prefrontal cortex (PFC), temporal and parietal cortices and thalamus (P < 0.05, corrected for multiple comparisons). Dopamine release in the ACC and orbitofrontal and ventromedial PFCs were correlated with subjective 'liking' and 'wanting' effects (P < 0.05). In contrast, baseline mGluR5 availability was positively correlated with the 'high' effect of alcohol in dorsolateral, ventrolateral and ventromedial PFCs and in the medial temporal lobe, thalamus and caudate nucleus (P < 0.05). Although neither proton-magnetic resonance spectroscopy glutamate nor glutamine levels were affected by alcohol, baseline ACC glutamate levels were negatively associated with the alcohol 'liking' effect (P < 0.003). These data reveal new mechanistic understanding and differential neurobiological underpinnings of the effects of acute alcohol consumption on human behavior. Specifically, prefrontal dopamine release may encode alcohol 'liking' and 'wanting' effects in specific areas underlying value processing and motivation, whereas mGluR5 availability in distinct prefrontal-temporal-subcortical regions is more related to the alcohol 'high' effect.
Collapse
Affiliation(s)
- Gil Leurquin-Sterk
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
| | - Jenny Ceccarini
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
| | - Cleo Lina Crunelle
- Toxicological Center, University of Antwerp, Department of Psychiatry; University Hospital Brussels; Belgium
| | | | - Bart de Laat
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
- MoSAIC, Molecular Small Animal Imaging Center; Belgium
| | - Uwe Himmelreich
- Department of Imaging and Pathology; Biomedical MRI/MoSAIC; Belgium
| | | | - Koen Van Laere
- Department of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology; University Hospitals Leuven; Belgium
- MoSAIC, Molecular Small Animal Imaging Center; Belgium
| |
Collapse
|
519
|
Xie W, Mallin BM, Richards JE. Development of infant sustained attention and its relation to EEG oscillations: an EEG and cortical source analysis study. Dev Sci 2018; 21:e12562. [PMID: 28382759 PMCID: PMC5628078 DOI: 10.1111/desc.12562] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/06/2017] [Indexed: 01/25/2023]
Abstract
The current study examined the relation between infant sustained attention and infant EEG oscillations. Fifty-nine infants were tested at 6 (N = 15), 8 (N = 17), 10 (N = 14), and 12 (N = 13) months. Three attention phases, stimulus orienting, sustained attention, and attention termination, were defined based on infants' heart rate changes. Frequency analysis using simultaneously recorded EEG focused on infant theta (2-6 Hz), alpha (6-9 Hz), and beta (9-14 Hz) rhythms. Cortical source analysis of EEG oscillations was conducted with realistic infant MRI models. Theta synchronization was found over fontal pole, temporal, and parietal electrodes during infant sustained attention for 10 and 12 months. Alpha desynchronization was found over frontal, central and parietal electrodes during sustained attention. This alpha effect started to emerge at 10 months and became well established by 12 months. No difference was found for the beta rhythm between different attention phases. The theta synchronization effect was localized to the orbital frontal, temporal pole, and ventral temporal areas. The alpha desynchronization effect was localized to the brain regions composing the default mode network including the posterior cingulate cortex and precuneus, medial prefrontal cortex, and inferior parietal gyrus. The alpha desynchronization effect was also localized to the pre- and post-central gyri. The present study demonstrates a connection between infant sustained attention and EEG oscillatory activities.
Collapse
Affiliation(s)
- Wanze Xie
- Department of Psychology, University of South Carolina
- Institute for Mind and Brain, University of South Carolina
| | | | - John E. Richards
- Department of Psychology, University of South Carolina
- Institute for Mind and Brain, University of South Carolina
| |
Collapse
|
520
|
Gao Y, Schilling KG, Stepniewska I, Plassard AJ, Choe AS, Li X, Landman BA, Anderson AW. Tests of cortical parcellation based on white matter connectivity using diffusion tensor imaging. Neuroimage 2018; 170:321-331. [PMID: 28235566 PMCID: PMC5568504 DOI: 10.1016/j.neuroimage.2017.02.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/23/2017] [Accepted: 02/18/2017] [Indexed: 10/20/2022] Open
Abstract
The cerebral cortex is conventionally divided into a number of domains based on cytoarchitectural features. Diffusion tensor imaging (DTI) enables noninvasive parcellation of the cortex based on white matter connectivity patterns. However, the correspondence between DTI-connectivity-based and cytoarchitectural parcellation has not been systematically established. In this study, we compared histological parcellation of New World monkey neocortex to DTI- connectivity-based classification and clustering in the same brains. First, we used supervised classification to parcellate parieto-frontal cortex based on DTI tractograms and the cytoarchitectural prior (obtained using Nissl staining). We performed both within and across sample classification, showing reasonable classification performance in both conditions. Second, we used unsupervised clustering to parcellate the cortex and compared the clusters to the cytoarchitectonic standard. We then explored the similarities and differences with several post-hoc analyses, highlighting underlying principles that drive the DTI-connectivity-based parcellation. The differences in parcellation between DTI-connectivity and Nissl histology probably represent both DTI's bias toward easily-tracked bundles and true differences between cytoarchitectural and connectivity defined domains. DTI tractograms appear to cluster more according to functional networks, rather than mapping directly onto cytoarchitectonic domains. Our results show that caution should be used when DTI-tractography classification, based on data from another brain, is used as a surrogate for cytoarchitectural parcellation.
Collapse
Affiliation(s)
- Yurui Gao
- Institute of Imaging Science, Vanderbilt University, United States; Department of Biomedical Engineering, Vanderbilt University, United States
| | - Kurt G Schilling
- Institute of Imaging Science, Vanderbilt University, United States; Department of Biomedical Engineering, Vanderbilt University, United States
| | | | - Andrew J Plassard
- Department of Computer Science, Vanderbilt University, United States
| | - Ann S Choe
- Institute of Imaging Science, Vanderbilt University, United States; Department of Biomedical Engineering, Vanderbilt University, United States
| | - Xia Li
- Institute of Imaging Science, Vanderbilt University, United States; Department of Radiology and Radiological Science, Vanderbilt University, United States
| | - Bennett A Landman
- Institute of Imaging Science, Vanderbilt University, United States; Department of Biomedical Engineering, Vanderbilt University, United States; Department of Computer Science, Vanderbilt University, United States; Department of Electrical Engineering, Vanderbilt University, United States
| | - Adam W Anderson
- Institute of Imaging Science, Vanderbilt University, United States; Department of Biomedical Engineering, Vanderbilt University, United States; Department of Radiology and Radiological Science, Vanderbilt University, United States
| |
Collapse
|
521
|
Evaluating accuracy of striatal, pallidal, and thalamic segmentation methods: Comparing automated approaches to manual delineation. Neuroimage 2018; 170:182-198. [DOI: 10.1016/j.neuroimage.2017.02.069] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 02/03/2017] [Accepted: 02/24/2017] [Indexed: 12/16/2022] Open
|
522
|
Conen S, Matthews JC, Patel NK, Anton-Rodriguez J, Talbot PS. Acute and chronic changes in brain activity with deep brain stimulation for refractory depression. J Psychopharmacol 2018; 32:430-440. [PMID: 29228889 DOI: 10.1177/0269881117742668] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Deep brain stimulation is a potential option for patients with treatment-refractory depression. Deep brain stimulation benefits have been reported when targeting either the subgenual cingulate or ventral anterior capsule/nucleus accumbens. However, not all patients respond and optimum stimulation-site is uncertain. We compared deep brain stimulation of the subgenual cingulate and ventral anterior capsule/nucleus accumbens separately and combined in the same seven treatment-refractory depression patients, and investigated regional cerebral blood flow changes associated with acute and chronic deep brain stimulation. Deep brain stimulation-response was defined as reduction in Montgomery-Asberg Depression Rating Scale score from baseline of ≥50%, and remission as a Montgomery-Asberg Depression Rating Scale score ≤8. Changes in regional cerebral blood flow were assessed using [15O]water positron emission tomography. Remitters had higher relative regional cerebral blood flow in the prefrontal cortex at baseline and all subsequent time-points compared to non-remitters and non-responders, with prefrontal cortex regional cerebral blood flow generally increasing with chronic deep brain stimulation. These effects were consistent regardless of stimulation-site. Overall, no significant regional cerebral blood flow changes were apparent when deep brain stimulation was acutely interrupted. Deep brain stimulation improved treatment-refractory depression severity in the majority of patients, with consistent changes in local and distant brain regions regardless of target stimulation. Remission of depression was reached in patients with higher baseline prefrontal regional cerebral blood flow. Because of the small sample size these results are preliminary and further evaluation is necessary to determine whether prefrontal cortex regional cerebral blood flow could be a predictive biomarker of treatment response.
Collapse
Affiliation(s)
- Silke Conen
- 1 Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester Academic Health Science Centre, UK
| | - Julian C Matthews
- 2 Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester Academic Health Science Centre, UK
| | - Nikunj K Patel
- 3 Department of Neurosurgery, Frenchay Hospital, Bristol, UK
| | - José Anton-Rodriguez
- 2 Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester Academic Health Science Centre, UK
| | - Peter S Talbot
- 1 Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester Academic Health Science Centre, UK
| |
Collapse
|
523
|
Huizinga W, Poot D, Vernooij M, Roshchupkin G, Bron E, Ikram M, Rueckert D, Niessen W, Klein S. A spatio-temporal reference model of the aging brain. Neuroimage 2018; 169:11-22. [DOI: 10.1016/j.neuroimage.2017.10.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/12/2017] [Accepted: 10/19/2017] [Indexed: 01/27/2023] Open
|
524
|
Postnov A, Schmidt ME, Pemberton DJ, de Hoon J, van Hecken A, van den Boer M, Zannikos P, van der Ark P, Palmer JA, Rassnick S, Celen S, Bormans G, van Laere K. Fatty Acid Amide Hydrolase Inhibition by JNJ-42165279: A Multiple-Ascending Dose and a Positron Emission Tomography Study in Healthy Volunteers. Clin Transl Sci 2018; 11:397-404. [PMID: 29575526 PMCID: PMC6039207 DOI: 10.1111/cts.12548] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/02/2018] [Indexed: 01/01/2023] Open
Abstract
Inhibition of fatty acid amide hydrolase (FAAH) potentiates endocannabinoid activity and is hypothesized to have therapeutic potential for mood and anxiety disorders and pain. The clinical profile of JNJ-42165279, an oral selective FAAH inhibitor, was assessed by investigating the pharmacokinetics, pharmacodynamics, safety, and binding to FAAH in the brain of healthy human volunteers. Concentrations of JNJ-42165279 (plasma, cerebrospinal fluid (CSF), urine) and fatty acid amides (FAA; plasma, CSF), and FAAH activity in leukocytes was determined in a phase I multiple ascending dose study. A positron emission tomography study with the FAAH tracer [11 C]MK3168 was conducted to determine brain FAAH occupancy after single and multiple doses of JNJ-42165279. JNJ-42165279 administration resulted in an increase in plasma and CSF FAA. Significant blocking of brain FAAH binding of [11 C]MK3168 was observed after pretreatment with JNJ-42165279. JNJ-42165279 produces potent central and peripheral FAAH inhibition. Saturation of brain FAAH occupancy occurred with doses ≥10 mg of JNJ-42165279. No safety concerns were identified.
Collapse
Affiliation(s)
- Andrey Postnov
- Division of Nuclear Medicine, Department of Imaging and Pathology, University Hospital and KU Leuven, Leuven, Belgium.,MEPhI National Research Nuclear University, Moscow, Russia
| | - Mark E Schmidt
- Neuroscience Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Darrel J Pemberton
- Neuroscience Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jan de Hoon
- Department of Pharmaceutical and Pharmacological Sciences, Center for Clinical Pharmacology, University Hospitals of Leuven, Leuven, Belgium
| | - Anne van Hecken
- Department of Pharmaceutical and Pharmacological Sciences, Center for Clinical Pharmacology, University Hospitals of Leuven, Leuven, Belgium
| | | | - Peter Zannikos
- Janssen Research & Development, Titusville, New Jersey, USA
| | | | - James A Palmer
- Janssen Research & Development, San Diego, California, USA
| | - Stef Rassnick
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Sofie Celen
- Laboratory of Radiopharmaceutical Research of the KU Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory of Radiopharmaceutical Research of the KU Leuven, Leuven, Belgium
| | - Koen van Laere
- Division of Nuclear Medicine, Department of Imaging and Pathology, University Hospital and KU Leuven, Leuven, Belgium
| |
Collapse
|
525
|
van Bergen JMG, Li X, Quevenco FC, Gietl AF, Treyer V, Meyer R, Buck A, Kaufmann PA, Nitsch RM, van Zijl PCM, Hock C, Unschuld PG. Simultaneous quantitative susceptibility mapping and Flutemetamol-PET suggests local correlation of iron and β-amyloid as an indicator of cognitive performance at high age. Neuroimage 2018; 174:308-316. [PMID: 29548847 DOI: 10.1016/j.neuroimage.2018.03.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 02/17/2018] [Accepted: 03/10/2018] [Indexed: 12/11/2022] Open
Abstract
The accumulation of β-amyloid plaques is a hallmark of Alzheimer's disease (AD), and recently published data suggest that increased brain iron burden may reflect pathologies that synergistically contribute to the development of cognitive dysfunction. While preclinical disease stages are considered most promising for therapeutic intervention, the link between emerging AD-pathology and earliest clinical symptoms remains largely unclear. In the current study we therefore investigated local correlations between iron and β-amyloid plaques, and their possible association with cognitive performance in healthy older adults. 116 older adults (mean age 75 ± 7.4 years) received neuropsychological testing to calculate a composite cognitive score of performance in episodic memory, executive functioning, attention, language and communication. All participants were scanned on a combined PET-MRI instrument and were administered T1-sequences for anatomical mapping, quantitative susceptibility mapping (QSM) for assessing iron, and 18F-Flutemetamol-PET for estimating β-amyloid plaque load. Biological parametric mapping (BPM) was used to generate masks indicating voxels with significant (p < 0.05) correlation between susceptibility and 18F-Flutemetamol-SUVR. We found a bilateral pattern of clusters characterized by a statistical relationship between magnetic susceptibility and 18F-Flutemetamol-SUVR, indicating local correlations between iron and β-amyloid plaque deposition. For two bilateral clusters, located in the frontal and temporal cortex, significant relationships (p<0.05) between local β-amyloid and the composite cognitive performance score could be observed. No relationship between whole-cortex β-amyloid plaque load and cognitive performance was observable. Our data suggest that the local correlation of β-amyloid plaque load and iron deposition may provide relevant information regarding cognitive performance of healthy older adults. Further studies are needed to clarify pathological correlates of the local interaction of β-amyloid, iron and other causes of altered magnetic susceptibility.
Collapse
Affiliation(s)
- J M G van Bergen
- Institute for Regenerative Medicine, University of Zurich, Switzerland.
| | - X Li
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - F C Quevenco
- Institute for Regenerative Medicine, University of Zurich, Switzerland
| | - A F Gietl
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - V Treyer
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - R Meyer
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - A Buck
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - P A Kaufmann
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - R M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - P C M van Zijl
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - C Hock
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| | - P G Unschuld
- Institute for Regenerative Medicine, University of Zurich, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Switzerland
| |
Collapse
|
526
|
Unterrainer M, Mahler C, Vomacka L, Lindner S, Havla J, Brendel M, Böning G, Ertl-Wagner B, Kümpfel T, Milenkovic VM, Rupprecht R, Kerschensteiner M, Bartenstein P, Albert NL. TSPO PET with [ 18F]GE-180 sensitively detects focal neuroinflammation in patients with relapsing-remitting multiple sclerosis. Eur J Nucl Med Mol Imaging 2018. [PMID: 29523925 DOI: 10.1007/s00259-018-3974-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE Expression of the translocator protein (TSPO) is upregulated in activated macrophages/microglia and is considered to be a marker of neuroinflammation. We investigated the novel TSPO ligand [18F]GE-180 in patients with relapsing-remitting multiple sclerosis (RRMS) to determine the feasibility of [18F]GE-180 PET imaging in RRMS patients and to assess its ability to detect active inflammatory lesions in comparison with the current gold standard, contrast-enhanced magnetic resonance imaging (MRI). METHODS Nineteen RRMS patients were prospectively included in this study. All patients underwent TSPO genotyping and were classified as high-affinity, medium-affinity or low-affinity binders (HAB/MAB/LAB). PET scans were performed after administration of 189 ± 12 MBq [18F]GE-180, and 60-90 min summation images were used for visual analysis and assessment of standardized uptake values (SUV). The frontal nonaffected cortex served as a pseudoreference region (PRR) for evaluation of SUV ratios (SUVR). PET data were correlated with MRI signal abnormalities, i.e. T2 hyperintensity or contrast enhancement (CE). When available, previous MRI data were used to follow the temporal evolution of individual lesions. RESULTS Focal lesions were identified as hot spots by visual inspection. Such lesions were detected in 17 of the 19 patients and overall 89 [18F]GE-180-positive lesions were found. TSPO genotyping revealed 11 patients with HAB status, 5 with MAB status and 3 with LAB status. There were no associations between underlying binding status (HAB, MAB and LAB) and the signal intensity in either lesions (SUVR 1.87 ± 0.43, 1.95 ± 0.48 and 1.86 ± 0.80, respectively; p = 0.280) or the PRR (SUV 0.36 ± 0.03, 0.40 ± 0.06 and 0.37 ± 0.03, respectively; p = 0.990). Of the 89 [18F]GE-180-positive lesions, 70 showed CE on MRI, while the remainder presented as T2 lesions without CE. SUVR were significantly higher in lesions with CE than in those without (2.00 ± 0.53 vs. 1.60 ± 0.15; p = 0.001). Notably, of 19 [18F]GE-180-positive lesions without CE, 8 previously showed CE, indicating that [18F]GE-180 imaging may be able to detect lesional activity that is sustained beyond the blood-brain barrier breakdown. CONCLUSION [18F]GE-180 PET can detect areas of focal macrophage/microglia activation in patients with RRMS in lesions with and without CE on MRI. Therefore, [18F]GE-180 PET imaging is a sensitive and quantitative approach to the detection of active MS lesions. It may provide information beyond contrast-enhanced MRI and is readily applicable to all patients. [18F]GE-180 PET imaging is therefore a promising new tool for the assessment of focal inflammatory activity in MS.
Collapse
Affiliation(s)
- Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - C Mahler
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.,Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - L Vomacka
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - S Lindner
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - J Havla
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.,Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - M Brendel
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - G Böning
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians University Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - B Ertl-Wagner
- Institute of Clinical Radiology, University Hospital, LMU Munich, Munich, Germany
| | - T Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.,Biomedical Center (BMC), LMU Munich, Munich, Germany
| | - V M Milenkovic
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - R Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - M Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.,Biomedical Center (BMC), LMU Munich, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians University Munich, Marchioninistr. 15, 81377, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians University Munich, Marchioninistr. 15, 81377, Munich, Germany. .,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
527
|
Fierstra J, van Niftrik C, Warnock G, Wegener S, Piccirelli M, Pangalu A, Esposito G, Valavanis A, Buck A, Luft A, Bozinov O, Regli L. Staging Hemodynamic Failure With Blood Oxygen-Level–Dependent Functional Magnetic Resonance Imaging Cerebrovascular Reactivity. Stroke 2018; 49:621-629. [DOI: 10.1161/strokeaha.117.020010] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/03/2017] [Accepted: 11/29/2017] [Indexed: 12/20/2022]
Abstract
Background and Purpose—
Increased stroke risk correlates with hemodynamic failure, which can be assessed with (
15
O-)H
2
O positron emission tomography (PET) cerebral blood flow (CBF) measurements. This gold standard technique, however, is not established for routine clinical imaging. Standardized blood oxygen-level–dependent (BOLD) functional magnetic resonance imaging+CO
2
is a noninvasive and potentially widely applicable tool to assess whole-brain quantitative cerebrovascular reactivity (CVR). We examined the agreement between the 2 imaging modalities and hypothesized that quantitative CVR can be a surrogate imaging marker to assess hemodynamic failure.
Methods—
Nineteen data sets of subjects with chronic cerebrovascular steno-occlusive disease (age, 60±11 years; 4 women) and unilaterally impaired perfusion reserve on Diamox-challenged (
15
O-)H
2
O PET were studied and compared with a standardized BOLD functional magnetic resonance imaging+CO
2
examination within 6 weeks (8±19 days). Agreement between quantitative CBF- and CVR-based perfusion reserve was assessed. Hemodynamic failure was staged according to PET findings: stage 0: normal CBF, normal perfusion reserve; stage I: normal CBF, decreased perfusion reserve; and stage II: decreased CBF, decreased perfusion reserve. The BOLD CVR data set of the same subjects was then matched to the corresponding stage of hemodynamic failure.
Results—
PET-based stage I versus stage II could also be clearly separated with BOLD CVR measurements (CVR for stage I 0.11 versus CVR for stage II −0.03;
P
<0.01). Hemispheric and middle cerebral artery territory difference analyses (ie, affected versus unaffected side) showed a significant correlation for CVR impairment in the affected hemisphere and middle cerebral artery territory (
P
<0.01,
R
2
=0.47 and
P
=0.02,
R
2
= 0.25, respectively).
Conclusions—
BOLD CVR corresponded well to CBF perfusion reserve measurements obtained with (
15
O-)H
2
O-PET, especially for detecting hemodynamic failure in the affected hemisphere and middle cerebral artery territory and for identifying hemodynamic failure stage II. BOLD CVR may, therefore, be considered for prospective studies assessing stroke risk in patients with chronic cerebrovascular steno-occlusive disease, in particular because it can potentially be implemented in routine clinical imaging.
Collapse
Affiliation(s)
- Jorn Fierstra
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Christiaan van Niftrik
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Geoffrey Warnock
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Susanne Wegener
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Marco Piccirelli
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Athina Pangalu
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Giuseppe Esposito
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Antonios Valavanis
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Alfred Buck
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Andreas Luft
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Oliver Bozinov
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| | - Luca Regli
- From the Departments of Neurosurgery (J.F., C.v.N., G.E., O.B., L.R.), Neuroradiology (M.P., A.V.), Neurology (S.W., A.L.), Pharmacology and Toxicology (G.W.), and Nuclear Medicine (A.B.), Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Switzerland
| |
Collapse
|
528
|
Dadar M, Maranzano J, Ducharme S, Carmichael OT, Decarli C, Collins DL. Validation of T1w-based segmentations of white matter hyperintensity volumes in large-scale datasets of aging. Hum Brain Mapp 2018; 39:1093-1107. [PMID: 29181872 PMCID: PMC6866430 DOI: 10.1002/hbm.23894] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Fluid-attenuated Inversion Recovery (FLAIR) and dual T2w and proton density (PD) magnetic resonance images (MRIs) are considered to be the optimum sequences for detecting white matter hyperintensities (WMHs) in aging and Alzheimer's disease populations. However, many existing large multisite studies forgo their acquisition in favor of other MRI sequences due to economic and time constraints. METHODS In this article, we have investigated whether FLAIR and T2w/PD sequences are necessary to detect WMHs in Alzheimer's and aging studies, compared to using only T1w images. Using a previously validated automated tool based on a Random Forests classifier, WMHs were segmented for the baseline visits of subjects from ADC, ADNI1, and ADNI2/GO studies with and without T2w/PD and FLAIR information. The obtained WMH loads (WMHLs) in different lobes were then correlated with manually segmented WMHLs, each other, age, cognitive, and clinical measures to assess the strength of the correlations with and without using T2w/PD and FLAIR information. RESULTS The WMHLs obtained from T1w-Only segmentations correlated with the manual WMHLs (ADNI1: r = .743, p < .001, ADNI2/GO: r = .904, p < .001), segmentations obtained from T1w + T2w + PD for ADNI1 (r = .888, p < .001) and T1w + FLAIR for ADNI2/GO (r = .969, p < .001), age (ADNI1: r = .391, p < .001, ADNI2/GO: r = .466, p < .001), and ADAS13 (ADNI1: r = .227, p < .001, ADNI2/GO: r = .190, p < 0.001), and NPI (ADNI1: r = .290, p < .001, ADNI2/GO: r = 0.144, p < .001), controlling for age. CONCLUSION Our results suggest that while T2w/PD and FLAIR provide more accurate estimates of the true WMHLs, T1w-Only segmentations can still provide estimates that hold strong correlations with the actual WMHLs, age, and performance on various cognitive/clinical scales, giving added value to datasets where T2w/PD or FLAIR are not available.
Collapse
Affiliation(s)
- Mahsa Dadar
- NeuroImaging and Surgical Tools LaboratoryMontreal Neurological Institute, McGill UniversityMontrealQuebecCanada
| | | | - Simon Ducharme
- Montreal Neurological Institute, McGill UniversityMontrealQuebecCanada
| | | | | | - D. Louis Collins
- NeuroImaging and Surgical Tools LaboratoryMontreal Neurological Institute, McGill UniversityMontrealQuebecCanada
| | | |
Collapse
|
529
|
Smit M, Vállez García D, de Jong BM, Zoons E, Booij J, Dierckx RA, Willemsen AT, de Vries EF, Bartels AL, Tijssen MA. Relationships between Serotonin Transporter Binding in the Raphe Nuclei, Basal Ganglia, and Hippocampus with Clinical Symptoms in Cervical Dystonia: A [ 11C]DASB Positron Emission Tomography Study. Front Neurol 2018. [PMID: 29541052 PMCID: PMC5835525 DOI: 10.3389/fneur.2018.00088] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Alterations of the central serotonergic system have been implicated in the pathophysiology of dystonia. In this molecular imaging study, we assessed whether altered presynaptic serotonin transporter (SERT) binding contributes to the pathophysiology of cervical dystonia (CD), concerning both motor and non-motor symptoms (NMS). Methods We assessed the non-displaceable binding potential (BPND) using the selective SERT tracer [11C]DASB and positron emission tomography (PET) in 14 CD patients and 12 age- and gender-matched controls. Severity of motor symptoms was scored using the Toronto Western Spasmodic Torticollis Rating Scale and Clinical Global Impression jerks/tremor scale. NMS for depressive symptoms, anxiety, fatigue, and sleep disturbances were assessed with quantitative rating scales. The relationship between SERT binding and clinical patient characteristics was analyzed with the Spearman’s rho test and multiple regression. Results When comparing the CD patients with controls, no significant differences in BPND were found. Higher BPND in the dorsal raphe nucleus was statistically significantly correlated (p < 0.001) with motor symptom severity (rs = 0.65), pain (rs = 0.73), and sleep disturbances (rs = 0.73), with motor symptom severity being the most important predictor of SERT binding. Furthermore, fatigue was negatively associated with the BPND in the medial raphe nucleus (rs = −0.61, p = 0.045), and sleep disorders were positively associated with the BPND in the caudate nucleus (rs = 0.58, p = 0.03) and the hippocampus (rs = 0.56, p = 0.02). Conclusion Motor symptoms, as well as pain, sleep disturbances, and fatigue in CD showed a significant relationship with SERT binding in the raphe nuclei. Moreover, fatigue showed a significant relationship with the medial raphe nucleus and sleep disorders with the caudate nucleus and hippocampus. These findings suggest that an altered serotonergic signaling in different brain areas in CD is related to different motor as well as NMS, which will further stimulate research on the role of serotonin in the pathogenesis of dystonia.
Collapse
Affiliation(s)
- Marenka Smit
- Department of Neurology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Bauke M de Jong
- Department of Neurology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Evelien Zoons
- Department of Neurology, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Jan Booij
- Department of Nuclear Medicine and Molecular Imaging, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Rudi A Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Antoon T Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Erik F de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Anna L Bartels
- Department of Neurology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands.,Department of Neurology, Ommelander Hospital Group, Groningen, Netherlands
| | - Marina A Tijssen
- Department of Neurology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| |
Collapse
|
530
|
Meijboom R, Steketee RME, Ham LS, van der Lugt A, van Swieten JC, Smits M. Differential Hemispheric Predilection of Microstructural White Matter and Functional Connectivity Abnormalities between Respectively Semantic and Behavioral Variant Frontotemporal Dementia. J Alzheimers Dis 2018; 56:789-804. [PMID: 28059782 DOI: 10.3233/jad-160564] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Semantic dementia (SD) and behavioral variant frontotemporal dementia (bvFTD), subtypes of frontotemporal dementia, are characterized by distinct clinical symptoms and neuroimaging features, with predominant left temporal grey matter (GM) atrophy in SD and bilateral or right frontal GM atrophy in bvFTD. Such differential hemispheric predilection may also be reflected by other neuroimaging features, such as brain connectivity. This study investigated white matter (WM) microstructure and functional connectivity differences between SD and bvFTD, focusing on the hemispheric predilection of these differences. Eight SD and 12 bvFTD patients, and 17 controls underwent diffusion tensor imaging and resting state functional MRI at 3T. Whole-brain WM microstructure was assessed to determine distinct WM tracts affected in SD and bvFTD. For these tracts, diffusivity measures and lateralization indices were calculated. Functional connectivity was established for GM regions affected in early stage SD or bvFTD. Results of a direct comparison between SD and bvFTD are reported. Whole-brain WM microstructure abnormalities were more pronounced in the left hemisphere in SD and bilaterally- with a slight predilection for the right- in bvFTD. Lateralization of tract-specific abnormalities was seen in SD only, toward the left hemisphere. Functional connectivity of disease-specific regions was mainly decreased bilaterally in SD and in the right hemisphere in bvFTD. SD and bvFTD show WM microstructure and functional connectivity abnormalities in different regions, that are respectively more pronounced in the left hemisphere in SD and in the right hemisphere in bvFTD. This indicates differential hemispheric predilection of brain connectivity abnormalities between SD and bvFTD.
Collapse
Affiliation(s)
- Rozanna Meijboom
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Centre Rotterdam, The Netherlands
| | - Rebecca M E Steketee
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Centre Rotterdam, The Netherlands
| | - Leontine S Ham
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Centre Rotterdam, The Netherlands
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Centre Rotterdam, The Netherlands
| | - John C van Swieten
- Department of Neurology, Erasmus MC - University Medical Centre Rotterdam, The Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC - University Medical Centre Rotterdam, The Netherlands
| |
Collapse
|
531
|
Schramm G, Holler M, Rezaei A, Vunckx K, Knoll F, Bredies K, Boada F, Nuyts J. Evaluation of Parallel Level Sets and Bowsher's Method as Segmentation-Free Anatomical Priors for Time-of-Flight PET Reconstruction. IEEE TRANSACTIONS ON MEDICAL IMAGING 2018; 37:590-603. [PMID: 29408787 PMCID: PMC5821901 DOI: 10.1109/tmi.2017.2767940] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
In this article, we evaluate Parallel Level Sets (PLS) and Bowsher's method as segmentation-free anatomical priors for regularized brain positron emission tomography (PET) reconstruction. We derive the proximity operators for two PLS priors and use the EM-TV algorithm in combination with the first order primal-dual algorithm by Chambolle and Pock to solve the non-smooth optimization problem for PET reconstruction with PLS regularization. In addition, we compare the performance of two PLS versions against the symmetric and asymmetric Bowsher priors with quadratic and relative difference penalty function. For this aim, we first evaluate reconstructions of 30 noise realizations of simulated PET data derived from a real hybrid positron emission tomography/magnetic resonance imaging (PET/MR) acquisition in terms of regional bias and noise. Second, we evaluate reconstructions of a real brain PET/MR data set acquired on a GE Signa time-of-flight PET/MR in a similar way. The reconstructions of simulated and real 3D PET/MR data show that all priors were superior to post-smoothed maximum likelihood expectation maximization with ordered subsets (OSEM) in terms of bias-noise characteristics in different regions of interest where the PET uptake follows anatomical boundaries. Our implementation of the asymmetric Bowsher prior showed slightly superior performance compared with the two versions of PLS and the symmetric Bowsher prior. At very high regularization weights, all investigated anatomical priors suffer from the transfer of non-shared gradients.
Collapse
|
532
|
Selvaraj S, Walker C, Arnone D, Cao B, Faulkner P, Cowen PJ, Roiser JP, Howes O. Effect of Citalopram on Emotion Processing in Humans: A Combined 5-HT 1A [ 11C]CUMI-101 PET and Functional MRI Study. Neuropsychopharmacology 2018; 43:655-664. [PMID: 28776580 PMCID: PMC5693328 DOI: 10.1038/npp.2017.166] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/18/2017] [Accepted: 08/01/2017] [Indexed: 12/21/2022]
Abstract
A subset of patients started on a selective serotonin reuptake inhibitor (SSRI) initially experience increased anxiety, which can lead to early discontinuation before therapeutic effects are manifest. The neural basis of this early SSRI effect is not known. Presynaptic dorsal raphe neuron (DRN) 5-HT1A receptors are known to have a critical role in affect processing. Thus we investigated the effect of acute citalopram on emotional processing and the relationship between DRN 5-HT1A receptor availability and amygdala reactivity. Thirteen (mean age 48±9 years) healthy male subjects received either a saline or citalopram infusion intravenously (10 mg over 30 min) on separate occasions in a single-blind, random order, crossover design. On each occasion, participants underwent a block design face-emotion processing task during fMRI known to activate the amygdala. Ten subjects also completed a positron emission tomography (PET) scan to quantify DRN 5-HT1A availability using [11C]CUMI-101. Citalopram infusion when compared with saline resulted in a significantly increased bilateral amygdala responses to fearful vs neutral faces (left p=0.025; right p=0.038 FWE-corrected). DRN [11C]CUMI-101 availability significantly positively correlated with the effect of citalopram on the left amygdala response to fearful faces (Z=2.51, p=0.027) and right amygdala response to happy faces (Z=2.33, p=0.032). Our findings indicate that the initial effect of SSRI treatment is to alter processing of aversive stimuli and that this is linked to DRN 5-HT1A receptors in line with evidence that 5-HT1A receptors have a role in mediating emotional processing.
Collapse
Affiliation(s)
- Sudhakar Selvaraj
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA,Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, London, UK,Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Biomedical and Behavioral Sciences Building (BBSB), 1941 East Road, Suite 3208 Houston, TX 77054, USA, Tel: +1 713 486 2500, Fax: +1 713 486 2553, E-mail:
| | - Chris Walker
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Danilo Arnone
- Institute of Psychiatry, King’s College London, Centre for Affective Disorders, London, UK,IoPPN, King’s College London, Institute of Psychiatry, Psychosis Studies, London, UK
| | - Bo Cao
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul Faulkner
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Philip J Cowen
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Jonathan P Roiser
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Oliver Howes
- Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, London, UK,IoPPN, King’s College London, Institute of Psychiatry, Psychosis Studies, London, UK,Institute of Clinical Sciences, Imperial College, Hammersmith Hospital, London, UK
| |
Collapse
|
533
|
Hanseeuw B, Dricot L, Lhommel R, Quenon L, Ivanoiu A. Patients with Amyloid-Negative Mild Cognitive Impairment have Cortical Hypometabolism but the Hippocampus is Preserved. J Alzheimers Dis 2018; 53:651-60. [PMID: 27232217 DOI: 10.3233/jad-160204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Patients with mild cognitive impairment (MCI) are at risk for Alzheimer's dementia but the presence of amyloid (Aβ) strongly increases this risk. In clinical settings, when Aβ status is not available, different neurodegenerative markers are used to characterize MCI. The accuracy of these markers to discriminate between Aβ-and Aβ+ MCI is not yet determined. OBJECTIVE To compare different markers of neurodegeneration in Aβ-and Aβ+ MCI, with an Aβ-elderly control (EC) group. METHODS Patients with MCI (n = 39) and EC (n = 28) underwent MRI, 18F-FDG PET, and Aβ PET (18F-flutemetamol). We compared FDG and MRI biomarker values in cortical and hippocampal regions of interest, and using voxel-wise surface maps. We computed ROC curves discriminating between the three groups for each biomarker. RESULTS All biomarker values were reduced in Aβ+ MCI compared to EC (p < 0.001). Aβ-MCI had low cortical metabolism (p = 0.002), but hippocampal volume, cortical thickness, and hippocampal metabolism were not significantly different between Aβ-MCI and EC (p > 0.40). Cortical metabolism best discriminated between MCI and EC (AUC = 0.92/0.86, Aβ+/Aβ-) while hippocampal volume best discriminated between Aβ-MCI and Aβ+ MCI (AUC = 0.79). CONCLUSIONS Cortical hypometabolism was observed in both Aβ-MCI and Aβ+ MCI whereas hippocampal atrophy was mostly found in Aβ+ MCI. For MCI patients without available Aβ information, hippocampal atrophy is thus more informative about Aβ status than cortical hypometabolism.
Collapse
Affiliation(s)
- Bernard Hanseeuw
- Neurology Department, Saint-Luc University Hospital, Brussels, Belgium.,Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.,Neurology Department, Massachusetts General Hospital and the Martinos Center for Biomedical Imaging, Boston, MA, USA
| | - Laurence Dricot
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Renaud Lhommel
- Nuclear Medicine Department, Saint-Luc University Hospital, Brussels, Belgium
| | - Lisa Quenon
- Neurology Department, Saint-Luc University Hospital, Brussels, Belgium.,Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Adrian Ivanoiu
- Neurology Department, Saint-Luc University Hospital, Brussels, Belgium.,Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
534
|
Leurquin-Sterk G, Ceccarini J, Crunelle CL, de Laat B, Verbeek J, Deman S, Neels H, Bormans G, Peuskens H, Van Laere K. Lower Limbic Metabotropic Glutamate Receptor 5 Availability in Alcohol Dependence. J Nucl Med 2018; 59:682-690. [PMID: 29348321 DOI: 10.2967/jnumed.117.199422] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/30/2017] [Indexed: 01/18/2023] Open
Abstract
Animal studies suggest an important role for the metabotropic glutamate receptor subtype 5 (mGlu5) in the pathophysiology of alcohol dependence, but direct human evidence is lacking. The goal of this study was to investigate cerebral mGlu5 availability in alcohol-dependent subjects versus controls using 18F-3-fluoro-5-[(pyridin-3-yl)ethynyl]benzonitrile (18F-FPEB) PET. Methods: Dynamic 90-min 18F-FPEB scans combined with arterial blood sampling were acquired for 16 recently abstinent alcohol-dependent subjects and 32 age-matched controls. Regional mGlu5 availability was quantified by the 18F-FPEB total distribution volume using both a voxel-by-voxel and a volume-of-interest analysis with partial-volume effect correction. Alcohol consumption within the last 3 mo was assessed by questionnaires and by hair ethyl glucuronide analysis. Craving was assessed using the Desire for Alcohol Questionnaire. Results: mGlu5 availability was lower in mainly limbic regions of alcohol-dependent subjects than in controls (P < 0.05, familywise error-corrected), ranging from 14% in the posterior cingulate cortex to 36% in the caudate nucleus. Lower mGlu5 availability was associated with higher hair ethyl glucuronide levels for most regions and was related to a lower level of craving specifically in the middle frontal gyrus, cingulate cortex, and inferolateral temporal lobe. Conclusion: These findings provide human in vivo evidence that limbic mGlu5 has a role in the pathophysiology of alcohol dependence, possibly involved in a compensatory mechanism helping to reduce craving during abstinence.
Collapse
Affiliation(s)
- Gil Leurquin-Sterk
- Division of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jenny Ceccarini
- Division of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Cleo L Crunelle
- Toxicological Center, University of Antwerp, Wilrijk, Belgium.,Department of Psychiatry, University Hospital Brussels, Brussels, Belgium
| | - Bart de Laat
- Division of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,MoSAIC: Molecular Small Animal Imaging Center, KU Leuven, Leuven, Belgium
| | - Jef Verbeek
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Stephanie Deman
- Genomics Core, UZ Leuven, and Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Hugo Neels
- Toxicological Center, University of Antwerp, Wilrijk, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmacy, KU Leuven, Leuven, Belgium; and
| | - Hendrik Peuskens
- University Psychiatric Center, KU Leuven, Kortenberg, and Kliniek Broeders Alexianen, Tienen, Belgium
| | - Koen Van Laere
- Division of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,MoSAIC: Molecular Small Animal Imaging Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
535
|
Brendel M, Schönecker S, Höglinger G, Lindner S, Havla J, Blautzik J, Sauerbeck J, Rohrer G, Zach C, Vettermann F, Lang AE, Golbe L, Nübling G, Bartenstein P, Furukawa K, Ishiki A, Bötzel K, Danek A, Okamura N, Levin J, Rominger A. [ 18F]-THK5351 PET Correlates with Topology and Symptom Severity in Progressive Supranuclear Palsy. Front Aging Neurosci 2018; 9:440. [PMID: 29387005 PMCID: PMC5776329 DOI: 10.3389/fnagi.2017.00440] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/20/2017] [Indexed: 11/16/2022] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative movement disorder characterized by deposition of fibrillar aggregates of 4R tau-protein in neurons and glial cells of the brain. These deposits are a key neuropathological finding, allowing a diagnosis of “definite PSP,” which is usually established post mortem. To date criteria for clinical diagnosis of PSP in vivo do not include biomarkers of tau pathology. For intervention trials, it is increasingly important to (i) establish biomarkers for an early diagnosis and (ii) to develop biomarkers that correlate with disease progression of PSP. [18F]-THK5351 is a novel PET-ligand that may afford in vivo visualization and quantification of tau-related alterations. We investigated binding characteristics of [18F]-THK5351 in patients with clinically diagnosed PSP and correlate tracer uptake with clinical findings. Eleven patients (68.4 ± 7.4 year; N = 6 female) with probable PSP according to current clinical criteria and nine healthy controls (71.7 ± 7.2 year; N = 4 female) underwent [18F]-THK5351 PET scanning. Voxel-wise statistical parametric comparison and volume-of-interest based quantification of standardized-uptake-values (SUV) were conducted using the cerebellar cortex as reference region. We correlated disease severity as measured with the help of the PSP Rating Scale (PSPRS) as well as several other clinical parameters with the individual PET findings. By voxel-wise mapping of [18F]-THK5351 uptake in the patient group we delineated typical distribution patterns that fit to known tau topology for PSP post mortem. Quantitative analysis indicated the strongest discrimination between PSP patients and healthy controls based on tracer uptake in the midbrain (+35%; p = 3.01E-7; Cohen's d: 4.0), followed by the globus pallidus, frontal cortex, and medulla oblongata. Midbrain [18F]-THK5351 uptake correlated well with clinical severity as measured by PSPRS (R = 0.66; p = 0.026). OCT and MRI delineated PSP patients from healthy controls by use of established discrimination thresholds but only OCT did as well correlate with clinical severity (R = 0.79; p = 0.024). Regional [18F]-THK5351 binding patterns correlated well with the established post mortem distribution of lesions in PSP and with clinical severity. The contribution of possible MAO-B binding to the [18F]-THK5351 signal needs to be further evaluated, but nevertheless [18F]-THK5351 PET may still serve as valuable biomarker for diagnosis of PSP.
Collapse
Affiliation(s)
- Matthias Brendel
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sonja Schönecker
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Technical University of Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Joachim Havla
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Janusch Blautzik
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julia Sauerbeck
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Guido Rohrer
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Zach
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Franziska Vettermann
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anthony E Lang
- Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson's Disease, and Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| | - Lawrence Golbe
- Department of Neurology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Georg Nübling
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katsutoshi Furukawa
- Division of Community Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Aiko Ishiki
- Department of Geriatrics and Gerontology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Kai Bötzel
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
536
|
Rodriguez-Vieitez E, Nordberg A. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol 2018; 1750:231-251. [PMID: 29512077 DOI: 10.1007/978-1-4939-7704-8_16] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The recent progress in the development of in vivo biomarkers is rapidly changing how neurodegenerative diseases are conceptualized and diagnosed, and how clinical trials are designed today. Alzheimer's disease (AD)-the most common neurodegenerative disorder-is characterized by a complex neuropathology involving the deposition of extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles (NFT) of hyperphosphorylated tau proteins, accompanied by the activation of glial cells-astrocytes and microglia-and neuroinflammatory responses, leading to neurodegeneration and cognitive dysfunction. An increasing diversity of positron emission tomography (PET) imaging radiotracers are available to selectively target the different pathophysiological processes of AD. Along with the success of Aβ PET and the more recent tau PET imaging, there is also a great interest to develop PET tracers to image glial activation and neuroinflammation. While most research to date has focused on imaging microgliosis, recent studies using 11C-deuterium-L-deprenyl (11C-DED) PET imaging suggest that astrocytosis may be present from very early stages of disease development in AD. This chapter provides a detailed description of the practical approach used for the analysis of 11C-DED PET imaging data in a multitracer PET paradigm including 11C-Pittsburgh compound B (11C-PiB) and 18F-fluorodeoxyglucose (18F-FDG). The multitracer PET approach allows investigating the comparative regional and temporal patterns of in vivo brain astrocytosis, fibrillar Aβ deposition, and glucose metabolism in patients at different stages of disease progression. This chapter attempts to stimulate further research in the field, including the development of novel PET tracers that may allow visualizing different aspects of the complex astrocytic and microglial responses in neurodegenerative diseases. Progress in the field will contribute to the incorporation of PET imaging of glial activation and neuroinflammation as biomarkers with clinical application, and motivate further investigation on glial cells as therapeutic targets in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Division of Translational Alzheimer Neurobiology, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Agneta Nordberg
- Division of Translational Alzheimer Neurobiology, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
537
|
Holmes SE, Hinz R, Conen S, Gregory CJ, Matthews JC, Anton-Rodriguez JM, Gerhard A, Talbot PS. Elevated Translocator Protein in Anterior Cingulate in Major Depression and a Role for Inflammation in Suicidal Thinking: A Positron Emission Tomography Study. Biol Psychiatry 2018; 83:61-69. [PMID: 28939116 DOI: 10.1016/j.biopsych.2017.08.005] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 08/04/2017] [Accepted: 08/06/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Major depressive disorder is associated with raised peripheral inflammatory markers. Mounting evidence also suggests that inflammation is involved in suicidal behavior. However, the involvement of inflammation in the brains of individuals with depression, and its association with suicidal ideation, needs further clarification. Translocator protein (TSPO), which is upregulated in activated glia (predominantly microglia), can be measured as an indication of neuroinflammation in vivo using positron emission tomography and TSPO-specific radioligands. METHODS We used [11C](R)-PK11195 positron emission tomography to compare TSPO availability in the anterior cingulate cortex (ACC), prefrontal cortex, and insula between 14 medication-free patients in a major depressive episode of at least moderate severity and 13 matched healthy control subjects. In a post hoc analysis, we also compared TSPO availability between patients with and without suicidal thoughts. RESULTS Multivariate analysis of variance indicated significantly higher TSPO in patients compared with control subjects (p = .005). The elevation was of large effect size and significant in the ACC (p = .022, Cohen's d = 0.95), with smaller nonsignificant elevations in the prefrontal cortex (p = .342, Cohen's d = 0.38) and insula (p = .466, Cohen's d = 0.29). TSPO was not elevated in patients without suicidal thinking but was significantly increased in those with suicidal thoughts compared with those without, most robustly in the ACC (p = .008) and insula (p = .023). CONCLUSIONS We confirm evidence for increased TSPO availability, suggestive of predominantly microglial activation, in the ACC during a moderate to severe major depressive episode. Our findings provide further incentive for evaluating anti-inflammatory therapies in major depressive disorder.
Collapse
Affiliation(s)
- Sophie E Holmes
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Rainer Hinz
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Silke Conen
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Catherine J Gregory
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Julian C Matthews
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jose M Anton-Rodriguez
- Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom; Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Peter S Talbot
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom; Greater Manchester Mental Health NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom.
| |
Collapse
|
538
|
Kobylecki C, Haense C, Harris JM, Stopford CL, Segobin SH, Jones M, Richardson AMT, Gerhard A, Anton-Rodriguez J, Thompson JC, Herholz K, Snowden JS. Functional neuroanatomical associations of working memory in early-onset Alzheimer's disease. Int J Geriatr Psychiatry 2018; 33:176-184. [PMID: 28299814 DOI: 10.1002/gps.4703] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/08/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To characterize metabolic correlates of working memory impairment in clinically defined subtypes of early-onset Alzheimer's disease. BACKGROUND Established models of working memory suggest a key role for frontal lobe function, yet the association in Alzheimer's disease between working memory impairment and visuospatial and language symptoms suggests that temporoparietal neocortical dysfunction may be responsible. METHODS Twenty-four patients with predominantly early-onset Alzheimer's disease were clinically classified into groups with predominantly amnestic, multidomain or visual deficits. Patients underwent neuropsychological evaluation focused on the domains of episodic and working memory, T1-weighted magnetic resonance imaging and brain fluorodeoxyglucose positron emission tomography. Fluorodeoxyglucose positron emission tomography data were analysed by using a region-of-interest approach. RESULTS Patients with multidomain and visual presentations performed more poorly on tests of working memory compared with amnestic Alzheimer's disease. Working memory performance correlated with glucose metabolism in left-sided temporoparietal, but not frontal neocortex. Carriers of the apolipoprotein E4 gene showed poorer episodic memory and better working memory performance compared with noncarriers. CONCLUSIONS Our findings support the hypothesis that working memory changes in early-onset Alzheimer's disease are related to temporoparietal rather than frontal hypometabolism and show dissociation from episodic memory performance. They further support the concept of subtypes of Alzheimer's disease with distinct cognitive profiles due to prominent neocortical dysfunction early in the disease course. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Christopher Kobylecki
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal NHS Foundation Trust, Salford, UK
| | - Cathleen Haense
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Jennifer M Harris
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal NHS Foundation Trust, Salford, UK
| | - Cheryl L Stopford
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal NHS Foundation Trust, Salford, UK
| | - Shailendra H Segobin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,INSERM-EPHE-Université de Caen de Basse-Normandie, Caen, France
| | - Matthew Jones
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal NHS Foundation Trust, Salford, UK
| | - Anna M T Richardson
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Medical School, University of Manchester, Manchester, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal NHS Foundation Trust, Salford, UK
| | - José Anton-Rodriguez
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jennifer C Thompson
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal NHS Foundation Trust, Salford, UK
| | - Karl Herholz
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Julie S Snowden
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|
539
|
Quantification of Tau Load in Alzheimer's Disease Clinical Trials Using Positron Emission Tomography. Methods Mol Biol 2018; 1750:221-229. [PMID: 29512076 DOI: 10.1007/978-1-4939-7704-8_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease is a neurodegenerative condition that is neuropathologically characterized by the presence of amyloid-β plaques and neurofibrillary tangles consisting of tau. Recently, several positron emission tomography (PET) tracers have been developed that yielded promising initial results. In this chapter, we discuss how tau PET can be used in the context in clinical trials. We argue that simplified reference tissue models based on dynamic data acquisition are most suitable for accurately measuring changes in tau pathology in trials tailored to reduce cerebral tau load. Therefore, we discuss the importance of tracer kinetic modeling and describe in detail how a reliable measurement of specific binding can be obtained.
Collapse
|
540
|
Park CH, Chun JW, Cho H, Kim DJ. Discriminating Pathological and Non-pathological Internet Gamers Using Sparse Neuroanatomical Features. Front Psychiatry 2018; 9:291. [PMID: 30008681 PMCID: PMC6033968 DOI: 10.3389/fpsyt.2018.00291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/12/2018] [Indexed: 11/24/2022] Open
Abstract
Internet gaming disorder (IGD) is often diagnosed on the basis of nine underlying criteria from the latest version of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5). Here, we examined whether such symptom-based categorization could be translated into computation-based classification. Structural MRI (sMRI) and diffusion-weighted MRI (dMRI) data were acquired in 38 gamers diagnosed with IGD, 68 normal gamers diagnosed as not having IGD, and 37 healthy non-gamers. We generated 108 features of gray matter (GM) and white matter (WM) structure from the MRI data. When regularized logistic regression was applied to the 108 neuroanatomical features to select important ones for the distinction between the groups, the disordered and normal gamers were represented in terms of 43 and 21 features, respectively, in relation to the healthy non-gamers, whereas the disordered gamers were represented in terms of 11 features in relation to the normal gamers. In support vector machines (SVM) using the sparse neuroanatomical features as predictors, the disordered and normal gamers were discriminated successfully, with accuracy exceeding 98%, from the healthy non-gamers, but the classification between the disordered and normal gamers was relatively challenging. These findings suggest that pathological and non-pathological gamers as categorized with the criteria from the DSM-5 could be represented by sparse neuroanatomical features, especially in the context of discriminating those from non-gaming healthy individuals.
Collapse
Affiliation(s)
- Chang-Hyun Park
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Ji-Won Chun
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul, South Korea
| | - Hyun Cho
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul, South Korea.,Department of Psychology, Korea University, Seoul, South Korea
| | - Dai-Jin Kim
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
541
|
Leuzy A, Rodriguez-Vieitez E, Saint-Aubert L, Chiotis K, Almkvist O, Savitcheva I, Jonasson M, Lubberink M, Wall A, Antoni G, Nordberg A. Longitudinal uncoupling of cerebral perfusion, glucose metabolism, and tau deposition in Alzheimer's disease. Alzheimers Dement 2017; 14:652-663. [PMID: 29268078 DOI: 10.1016/j.jalz.2017.11.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 12/01/2022]
Abstract
INTRODUCTION Cross-sectional findings using the tau tracer [18F]THK5317 (THK5317) have shown that [18F]fluorodeoxyglucose (FDG) positron emission tomography (PET) data can be approximated using perfusion measures (early-frame standardized uptake value ratio; ratio of tracer delivery in target to reference regions). In this way, a single PET study can provide both functional and molecular information. METHODS We included 16 patients with Alzheimer's disease who completed follow-up THK5317 and FDG studies 17 months after baseline investigations. Linear mixed-effects models and annual percentage change maps were used to examine longitudinal change. RESULTS Limited spatial overlap was observed between areas showing declines in THK5317 perfusion measures and FDG. Minimal overlap was seen between areas showing functional change and those showing increased retention of THK5317. DISCUSSION Our findings suggest a spatiotemporal offset between functional changes and tau pathology and a partial uncoupling between perfusion and metabolism, possibly as a function of Alzheimer's disease severity.
Collapse
Affiliation(s)
- Antoine Leuzy
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Elena Rodriguez-Vieitez
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Laure Saint-Aubert
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ove Almkvist
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden; Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Irina Savitcheva
- Department of Radiology, Karolinska University Hospital, Huddinge, Sweden
| | - My Jonasson
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Mark Lubberink
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Anders Wall
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Gunnar Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Agneta Nordberg
- Division of Translational Alzheimer Neurobiology, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Department of Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
542
|
Schreiner SJ, Kirchner T, Narkhede A, Wyss M, Van Bergen JMG, Steininger SC, Gietl A, Leh SE, Treyer V, Buck A, Pruessmann KP, Nitsch RM, Hock C, Henning A, Brickman AM, Unschuld PG. Brain amyloid burden and cerebrovascular disease are synergistically associated with neurometabolism in cognitively unimpaired older adults. Neurobiol Aging 2017; 63:152-161. [PMID: 29310864 DOI: 10.1016/j.neurobiolaging.2017.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/16/2017] [Accepted: 12/04/2017] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of cognitive dysfunction in older adults. The pathological hallmarks of AD such as beta amyloid (Aβ) aggregation and neurometabolic change, as indicated by altered myo-inositol (mI) and N-acetylaspartate (NAA) levels, typically precede the onset of cognitive dysfunction by years. Furthermore, cerebrovascular disease occurs early in AD, but the interplay between vascular and neurometabolic brain change is largely unknown. Thirty cognitively normal older adults (age = 70 ± 5.6 years, Mini-Mental State Examination = 29.2 ± 1) received 11-C-Pittsburgh Compound B positron emission tomography for estimating Aβ-plaque density, 7 Tesla fluid-attenuated inversion recovery magnetic resonance imaging for quantifying white matter hyperintensity volume as a marker of small vessel cerebrovascular disease and high-resolution magnetic resonance spectroscopic imaging at 7 Tesla, based on free induction decay acquisition localized by outer volume suppression to investigate tissue-specific neurometabolism in the posterior cingulate and precuneus. Aβ (β = 0.45, p = 0.018) and white matter hyperintensities (β = 0.40, p = 0.046) were independently and interactively (β = -0.49, p = 0.026) associated with a higher ratio of mI over NAA (mI/NAA) in the posterior cingulate and precuneus gray matter but not in the white matter. Our data suggest that cerebrovascular disease and Aβ burden are synergistically associated with AD-related gray matter neurometabolism in older adults.
Collapse
Affiliation(s)
- Simon J Schreiner
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Thomas Kirchner
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Atul Narkhede
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, College of Physicians and Surgeons, Columbia University, NY
| | - Michael Wyss
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Jiri M G Van Bergen
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Stephanie C Steininger
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Anton Gietl
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Sandra E Leh
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Klaas P Pruessmann
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| | - Anke Henning
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland; Max Planck Institute for Biological Cybernetics, Tubingen, Germany
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, College of Physicians and Surgeons, Columbia University, NY.
| | - Paul G Unschuld
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Hospital for Psychogeriatric Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
543
|
Höller Y, Tadzic A, Thomschewski AC, Höller P, Leis S, Tomasi SO, Hofer C, Bathke A, Nardone R, Trinka E. Factors Affecting Volume Changes of the Somatosensory Cortex in Patients with Spinal Cord Injury: To Be Considered for Future Neuroprosthetic Design. Front Neurol 2017; 8:662. [PMID: 29321758 PMCID: PMC5732216 DOI: 10.3389/fneur.2017.00662] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/23/2017] [Indexed: 01/10/2023] Open
Abstract
Spinal cord injury (SCI) leads to severe chronic disability, but also to secondary adaptive changes upstream to the injury in the brain which are most likely induced due to the lack of afferent information. These neuroplastic changes are a potential target for innovative therapies such as neuroprostheses, e.g., by stimulation in order to evoke sensation or in order to suppress phantom limb pain. Diverging results on gray matter atrophy have been reported in patients with SCI. Detectability of atrophy seems to depend on the selection of the regions of interest, while whole-brain approaches are not sensitive enough. In this study, we discussed previous research approaches and analyzed differential atrophic changes in incomplete SCI using manual segmentation of the somatosensory cortex. Patients with incomplete SCI (ASIA C-D), with cervical (N = 5) and thoracic (N = 6) injury were included. Time since injury was ≤12 months in 7 patients, and 144, 152, 216, and 312 months in the other patients. Age at the injury was ≤26 years in 4 patients and ≥50 years in 7 patients. A sample of 12 healthy controls was included in the study. In contrast to all previous studies that used voxel-based morphometry, we performed manual segmentation of the somatosensory cortex in the postcentral gyrus from structural magnetic resonance images and normalized the calculated volumes against the sum of volumes of an automated whole-head segmentation. Volumes were smaller in patients than in controls (p = 0.011), and as a tendency, female patients had smaller volumes than male patients (p = 0.017, uncorrected). No effects of duration (subacute vs. chronic), level of lesion (cervical vs. thoracic), region (left vs. right S1), and age at onset (≤26 vs. ≥50 years) was found. Our results demonstrate volume loss of S1 in incomplete SCI and encourage further research with larger sample sizes on volumetric changes in the acute and chronic stage of SCI, in order to document the moderating effect of type and location of injury on neuroplastic changes. A better understanding of neuroplastic changes in the sensorimotor cortex after SCI and its interaction with sex is needed in order to develop efficient rehabilitative interventions and neuroprosthetic technologies.
Collapse
Affiliation(s)
- Yvonne Höller
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Arijan Tadzic
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Aljoscha C. Thomschewski
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University Salzburg, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Peter Höller
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University Salzburg, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Stefan Leis
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University Salzburg, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| | - Santino Ottavio Tomasi
- Department of Neurosurgery, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Christoph Hofer
- Wavelab, Department of Computer Sciences, University of Salzburg, Salzburg, Austria
| | - Arne Bathke
- Department of Mathematics, University of Salzburg, Salzburg, Austria
| | - Raffaele Nardone
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University Salzburg, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
- Department of Neurology, Franz Tappeiner Hospital, Merano, Italy
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Medical Centre and Centre for Cognitive Neuroscience, Paracelsus Medical University Salzburg, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
544
|
Golla SSV, Adriaanse SM, Yaqub M, Windhorst AD, Lammertsma AA, van Berckel BNM, Boellaard R. Model selection criteria for dynamic brain PET studies. EJNMMI Phys 2017; 4:30. [PMID: 29209862 PMCID: PMC5716967 DOI: 10.1186/s40658-017-0197-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 11/23/2017] [Indexed: 12/04/2022] Open
Abstract
Background Several criteria exist to identify the optimal model for quantification of tracer kinetics. The purpose of this study was to evaluate the correspondence in kinetic model preference identification for brain PET studies among five model selection criteria: Akaike Information Criterion (AIC), AIC unbiased (AICC), model selection criterion (MSC), Schwartz Criterion (SC), and F-test. Materials and Methods Six tracers were evaluated: [11C]FMZ, [11C]GMOM, [11C]PK11195, [11C]Raclopride, [18F]FDG, and [11C]PHT, including data from five subjects per tracer. Time activity curves (TACs) were analysed using six plasma input models: reversible single-tissue model (1T2k), irreversible two-tissue model (2T3k), and reversible two-tissue model (2T4k), all with and without blood volume fraction parameter (VB). For each tracer and criterion, the percentage of TACs preferring a certain model was calculated. Results For all radiotracers, strong agreement was seen across the model selection criteria. The F-test was considered as the reference, as it is a frequently used hypothesis test. The F-test confirmed the AIC preferred model in 87% of all cases. The strongest (but minimal) disagreement across regional TACs was found when comparing AIC with AICC. Despite these regional discrepancies, same preferred kinetic model was obtained using all criteria, with an exception of one FMZ subject. Conclusion In conclusion, all five model selection criteria resulted in similar conclusions with only minor differences that did not affect overall model selection. Electronic supplementary material The online version of this article (10.1186/s40658-017-0197-0) 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, P.O. 7057, 1007, MB, Amsterdam, The Netherlands.
| | - Sofie M Adriaanse
- Department of Radiology and Nuclear Medicine, VU University Medical Center, P.O. 7057, 1007, MB, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, VU University Medical Center, P.O. 7057, 1007, MB, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, P.O. 7057, 1007, MB, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, P.O. 7057, 1007, MB, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, VU University Medical Center, P.O. 7057, 1007, MB, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, P.O. 7057, 1007, MB, Amsterdam, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
545
|
Urchs S, Armoza J, Benhajali Y, St-Aubin J, Orban P, Bellec P. MIST: A multi-resolution parcellation of functional brain networks. ACTA ACUST UNITED AC 2017. [DOI: 10.12688/mniopenres.12767.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Functional brain connectomics investigates functional connectivity between distinct brain parcels. There is an increasing interest to investigate connectivity across several levels of spatial resolution, from networks down to localized areas. Here we present the Multiresolution Intrinsic Segmentation Template (MIST), a multi-resolution parcellation of the cortical, subcortical and cerebellar gray matter. We provide annotated functional parcellations at nine resolutions from 7 to 444 functional parcels. The MIST parcellations compare well with prior work in terms of homogeneity and generalizability. We found that parcels at higher resolutions largely fell within the boundaries of larger parcels at lower resolutions. This allowed us to provide an overlap based pseudo-hierarchical decomposition tree that relates parcels across resolutions in a meaningful way. We provide an interactive web interface to explore the MIST parcellations and also made it accessible in the neuroimaging library nilearn. We believe that the MIST parcellation will facilitate future investigations of the multiresolution basis of brain function.
Collapse
|
546
|
Disrupted superior collicular activity may reveal cervical dystonia disease pathomechanisms. Sci Rep 2017; 7:16753. [PMID: 29196716 PMCID: PMC5711841 DOI: 10.1038/s41598-017-17074-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/21/2017] [Indexed: 11/13/2022] Open
Abstract
Cervical dystonia is a common neurological movement disorder characterised by muscle contractions causing abnormal movements and postures affecting the head and neck. The neural networks underpinning this condition are incompletely understood. While animal models suggest a role for the superior colliculus in its pathophysiology, this link has yet to be established in humans. The present experiment was designed to test the hypothesis that disrupted superior collicular processing is evident in affected patients and in relatives harbouring a disease-specific endophenotype (abnormal temporal discrimination). The study participants were 16 cervical dystonia patients, 16 unaffected first-degree relatives with abnormal temporal discrimination, 16 unaffected first-degree relatives with normal temporal discrimination and 16 healthy controls. The response of participant’s superior colliculi to looming stimuli was assessed by functional magnetic resonance imaging. Cervical dystonia patients and relatives with abnormal temporal discrimination demonstrated (i) significantly reduced superior collicular activation for whole brain and region of interest analysis; (ii) a statistically significant negative correlation between temporal discrimination threshold and superior collicular peak values. Our results support the hypothesis that disrupted superior collicular processing is involved in the pathogenesis of cervical dystonia. These findings, which align with animal models of cervical dystonia, shed new light on pathomechanisms in humans.
Collapse
|
547
|
Abstract
We have recently witnessed an explosion of large-scale initiatives and projects addressing mapping, modeling, simulation and atlasing of the human brain, including the BRAIN Initiative, the Human Brain Project, the Human Connectome Project (HCP), the Big Brain, the Blue Brain Project, the Allen Brain Atlas, the Brainnetome, among others. Besides these large and international initiatives, there are numerous mid-size and small brain atlas-related projects. My contribution to these global efforts has been to create adult human brain atlases in health and disease, and to develop atlas-based applications. For over two decades with my R&D lab I developed 35 brain atlases, licensed to 67 companies and made available in about 100 countries. This paper has two objectives. First, it provides an overview of the state of the art in brain atlasing. Second, as it is already 20 years from the release of our first brain atlas, I summarise my past and present efforts, share my experience in atlas creation, validation and commercialisation, compare with the state of the art, and propose future directions.
Collapse
Affiliation(s)
- Wieslaw L Nowinski
- John Paull II Center for Virtual Anatomy and Surgical Simulation, University of Cardinal Stefan Wyszynski in Warsaw, Poland
| |
Collapse
|
548
|
Rentiya Z, Khan NS, Ergun E, Ying SH, Desmond JE. Distinct cerebellar regions related to motor and cognitive performance in SCA6 patients. Neuropsychologia 2017; 107:25-30. [PMID: 29100951 PMCID: PMC5705404 DOI: 10.1016/j.neuropsychologia.2017.10.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 10/10/2017] [Accepted: 10/30/2017] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To demonstrate a correlation between anatomic regional changes in Spinocerebellar Ataxia type 6 (SCA6) patients and measures of cognitive performance on neuropsychological tests. METHODS Neurocognitive testing was conducted on 24 SCA6 and 28 control subjects. For each cognitive test, SCA6 patients were compared against the controls using Student's t-test. For the cerebellar patients, using voxel based morphometry, correlations between cerebellar gray matter volume at each voxel and performance on the neuropsychological exams were calculated using the Pearson correlation coefficient implemented in SPM8. RESULTS Compared to controls, SCA6 patients exhibited significantly impaired performance on the following cognitive tests: Rey-Auditory Verbal Learning Test Trial V, Controlled Oral Word Association phonemic test and semantic-verb test, Rey-Osterrieth Complex Figure copy test as well as immediate and delayed visuo-spatial memory recall test, Trail Making Test (TMT) Part A and Part B, Stroop Color Task completion time, Stroop Color-Word Task score, and Grooved Pegboard Test (GPT) Dominant and Non-Dominant Hand time. Correlations of gray matter density with cognitive test performance were determined for all SCA6 subjects. Using a p-value threshold of 0.001 and family-wise small volume error correction, significant correlations were found for GPT Non-Dominant, GPT Dominant, TMT Part A, and TMT Part B. CONCLUSION Different regional patterns of cerebellar involvement were found for the motoric GPT task and the executive version of the TMT. The results for the GPT strongly indicated that the integrity of medial superior hemispheric regions was associated with motor task performance, whereas executive cognitive function was localized in distinctly different inferior regions. This is the first VBM study to differentiate cognitive and motor contributions of the cerebellum.
Collapse
Affiliation(s)
- Zubir Rentiya
- Johns Hopkins University School of Medicine, Department Radiology, Neurology, Ophthalmology, United States.
| | - Noore-Sabah Khan
- Johns Hopkins University School of Medicine, Department Radiology, Neurology, Ophthalmology, United States.
| | - Ezgi Ergun
- Johns Hopkins Whiting School of Engineering, Department of Electrical and Computer Engineering, United States.
| | - Sarah H Ying
- Johns Hopkins University School of Medicine, Department Radiology, Neurology, Ophthalmology, United States.
| | - John E Desmond
- Johns Hopkins University School of Medicine, Department of Neurology, Neuroscience, Cognitive Science, United States.
| |
Collapse
|
549
|
Wu Z, Guo Y, Park SH, Gao Y, Dong P, Lee SW, Shen D. Robust brain ROI segmentation by deformation regression and deformable shape model. Med Image Anal 2017; 43:198-213. [PMID: 29149715 DOI: 10.1016/j.media.2017.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 09/11/2017] [Accepted: 11/01/2017] [Indexed: 01/18/2023]
Abstract
We propose a robust and efficient learning-based deformable model for segmenting regions of interest (ROIs) from structural MR brain images. Different from the conventional deformable-model-based methods that deform a shape model locally around the initialization location, we learn an image-based regressor to guide the deformable model to fit for the target ROI. Specifically, given any voxel in a new image, the image-based regressor can predict the displacement vector from this voxel towards the boundary of target ROI, which can be used to guide the deformable segmentation. By predicting the displacement vector maps for the whole image, our deformable model is able to use multiple non-boundary predictions to jointly determine and iteratively converge the initial shape model to the target ROI boundary, which is more robust to the local prediction error and initialization. In addition, by introducing the prior shape model, our segmentation avoids the isolated segmentations as often occurred in the previous multi-atlas-based methods. In order to learn an image-based regressor for displacement vector prediction, we adopt the following novel strategies in the learning procedure: (1) a joint classification and regression random forest is proposed to learn an image-based regressor together with an ROI classifier in a multi-task manner; (2) high-level context features are extracted from intermediate (estimated) displacement vector and classification maps to enforce the relationship between predicted displacement vectors at neighboring voxels. To validate our method, we compare it with the state-of-the-art multi-atlas-based methods and other learning-based methods on three public brain MR datasets. The results consistently show that our method is better in terms of both segmentation accuracy and computational efficiency.
Collapse
Affiliation(s)
- Zhengwang Wu
- IDEA Lab, BRIC, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Yanrong Guo
- IDEA Lab, BRIC, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Sang Hyun Park
- Department of Robotics Engineering, DGIST, Republic of Korea
| | - Yaozong Gao
- IDEA Lab, BRIC, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Pei Dong
- IDEA Lab, BRIC, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Seong-Whan Lee
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Dinggang Shen
- IDEA Lab, BRIC, UNC-Chapel Hill, Chapel Hill, NC, USA; Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
550
|
Yamagishi T, Li Y, Fermin ASR, Kanai R, Takagishi H, Matsumoto Y, Kiyonari T, Sakagami M. Behavioural Differences and Neural Substrates of Altruistic and Spiteful Punishment. Sci Rep 2017; 7:14654. [PMID: 29116134 PMCID: PMC5676782 DOI: 10.1038/s41598-017-15188-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/20/2017] [Indexed: 02/01/2023] Open
Abstract
Altruistic punishment following social norm violations promotes human cooperation. However, experimental evidence indicates that some forms of punishment are spiteful rather than altruistic. Using two types of punishment games and seven non-strategic games, we identified strong behavioural differences between altruistic and spiteful punishers. Altruistic punishers who rejected unfair offers in the ultimatum game and punished norm violators in the third-party punishment game behaved pro-socially in various non-strategic games. Spiteful punishers who rejected unfair offers in the ultimatum game but did not punish norm violators in the third-party punishment game behaved selfishly in non-strategic games. In addition, the left caudate nucleus was larger in spiteful punishers than in altruistic punishers. These findings are in contrast to the previous assumption that altruistic punishers derive pleasure from enforcement of fairness norms, and suggest that spiteful punishers derive pleasure from seeing the target experience negative consequences.
Collapse
Affiliation(s)
- Toshio Yamagishi
- Graduate School of International Corporate Strategy, Hitotsubashi University, Tokyo, Japan. .,Brain Science Institute, Tamagawa University, Tokyo, Japan.
| | - Yang Li
- Brain Science Institute, Tamagawa University, Tokyo, Japan.,Department of Psychology, University of Melbourne, Melbourne, Australia
| | | | - Ryota Kanai
- Department of Neuroinformatics, Araya Inc., Tokyo, Japan
| | | | | | - Toko Kiyonari
- School of Social Informatics, Aoyama Gakuin University, Kanagawa, Japan
| | | |
Collapse
|