151
|
Kim DJ, Jassar H, Lim M, Nascimento TD, DaSilva AF. Dopaminergic Regulation of Reward System Connectivity Underpins Pain and Emotional Suffering in Migraine. J Pain Res 2021; 14:631-643. [PMID: 33727857 PMCID: PMC7955762 DOI: 10.2147/jpr.s296540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/17/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose It has been suggested that reward system dysfunction may account for emotion and pain suffering in migraine. However, there is a lack of evidence whether the altered reward system connectivity is directly associated with clinical manifestations, including negative affect and ictal pain severity and, at the molecular level, the dopamine (DA) D2/D3 receptors (D2/3Rs) signaling implicated in encoding motivational and emotional cues. Patients and Methods We acquired resting-state functional MRI from interictal episodic migraine (EM) patients and age-matched healthy controls, as well as positron emission tomography (PET) with [11C]raclopride, a selective radiotracer for DA D2/3Rs, from a subset of these participants. The nucleus accumbens (NAc) was seeded to measure functional connectivity (FC) and DA D2/3Rs availability based on its essential involvement in pain-related aversive/reward functions. Associations of the brain measures with positive/negative affect and ictal pain severity were also assessed. Results Compared with controls, the EM group showed weaker right NAc connectivity with areas implicated in pain and emotional regulation, such as the amygdala, rostral anterior cingulate cortex, hippocampus, and thalamus; but showed stronger left NAc connectivity with the dorsolateral prefrontal cortex and lingual gyrus. Moreover, among the altered NAc connectivities, only right NAc-amygdala connectivity was inversely correlated with DA D2/3Rs availability in migraine patients (diagnostic group-by-D2/3Rs interaction p < 0.007). At a clinical level, such weaker NAc-amygdala connectivity was associated with lower interictal positive affect and greater ictal pain severity over the head and facial extension area (pain area and intensity number summation, PAINS). Conclusion Together, our findings suggest that altered reward system connectivity, specifically between the NAc and amygdala, might be affected by endogenous DA D2/3Rs signaling, and such process might be a neural mechanism that underlies emotional and pain suffering in episodic migraineurs.
Collapse
Affiliation(s)
- Dajung J Kim
- Headache and Orofacial Pain Effort (H.O.P.E.), Biologic and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Hassan Jassar
- Headache and Orofacial Pain Effort (H.O.P.E.), Biologic and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Manyoel Lim
- Headache and Orofacial Pain Effort (H.O.P.E.), Biologic and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Thiago D Nascimento
- Headache and Orofacial Pain Effort (H.O.P.E.), Biologic and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Alexandre F DaSilva
- Headache and Orofacial Pain Effort (H.O.P.E.), Biologic and Material Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| |
Collapse
|
152
|
Shigwedha PK, Yamada T, Hanaoka K, Ishii K, Kimura Y, Fukuoka Y. Improving contrast between gray and white matter of Logan graphical analysis' parametric images in positron emission tomography through least-squares cubic regression and principal component analysis. Biomed Phys Eng Express 2021; 7. [PMID: 33662939 DOI: 10.1088/2057-1976/abec18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/04/2021] [Indexed: 11/11/2022]
Abstract
Logan graphical analysis (LGA) is a method for in vivo quantification of tracer kinetics in positron emission tomography (PET). The shortcoming of LGA is the presence of a negative bias in the estimated parameters for noisy data. Various approaches have been proposed to address this issue. We recently applied an alternative regression method called least-squares cubic (LSC), which considers the errors in both the predictor and response variables to estimate the LGA slope. LSC reduced the bias in non-displaceable binding potential estimates while causing slight increases in the variance. In this study, we combined LSC with a principal component analysis (PCA) denoising technique to counteract the effects of variance on parametric image quality, which was assessed in terms of the contrast between gray and white matter. Tissue time-activity curves were denoised through PCA, prior to estimating the regression parameters using LSC. We refer to this approach as LSC-PCA. LSC-PCA was assessed against OLS-PCA (PCA with ordinary least-squares (OLS)), LSC, and conventional OLS-based LGA. Comparisons were made for simulated11C-carfentanil and11C Pittsburgh compound B (11C-PiB) data, and clinical11C-PiB PET images. PCA-based methods were compared over a range of principal components, varied by the percentage variance they account for in the data. The results showed reduced variances in distribution volume ratio estimates in the simulations for LSC-PCA compared to LSC, and lower bias compared to OLS-PCA and OLS. Contrasts were not significantly improved in clinical data, but they showed a significant improvement in simulation data |indicating a potential advantage of LSC-PCA over OLS-PCA. The effects of bias reintroduction when many principal components are used were also observed in OLS-PCA clinical images. We therefore encourage the use of LSC-PCA. LSC-PCA can allow the use of many principal components with minimal risk of bias, thereby strengthening the interpretation of PET parametric images.
Collapse
Affiliation(s)
- Paulus Kapundja Shigwedha
- Department of Electrical Engineering and Electronics, Graduate School of Engineering, Kogakuin University, 1-24-2 Nishi-shinjuku, Shinjuku-ku, Tokyo, 163-8677, JAPAN
| | - Takahiro Yamada
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, JAPAN
| | - Kohei Hanaoka
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, JAPAN
| | - Kazunari Ishii
- Department of Radiology, Kindai University Graduate School of Medical Sciences, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, JAPAN
| | - Yuichi Kimura
- Department of Computational Systems Biology, Kindai University Faculty of Biology Oriented Science and Technology Graduate School of Biology Oriented Science and Technology, 930 Nishitani, Kinokawa, Wakayama, 649-6493, JAPAN
| | - Yutaka Fukuoka
- Department of Electrical Engineering and Electronics, Graduate School of Engineering, Kogakuin University, 1-24-2 Nishi-shinjuku, Shinjuku-ku, Tokyo, 163-8677, JAPAN
| |
Collapse
|
153
|
Heeman F, Yaqub M, Lopes Alves I, Heurling K, Bullich S, Gispert JD, Boellaard R, Lammertsma AA. Simulating the effect of cerebral blood flow changes on regional quantification of [ 18F]flutemetamol and [ 18F]florbetaben studies. J Cereb Blood Flow Metab 2021; 41:579-589. [PMID: 32281514 PMCID: PMC7907983 DOI: 10.1177/0271678x20918029] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Global and regional changes in cerebral blood flow (CBF) can result in biased quantitative estimates of amyloid load by PET imaging. Therefore, the current simulation study assessed effects of these changes on amyloid quantification using a reference tissue approach for [18F]flutemetamol and [18F]florbetaben. Previously validated pharmacokinetic rate constants were used to simulate time-activity curves (TACs) corresponding to full dynamic and dual-time-window acquisition protocols. CBF changes were simulated by varying the tracer delivery (K1) from +25 to -25%. The standardized uptake value ratio (SUVr) was computed and TACs were fitted using reference Logan (RLogan) and the simplified reference tissue model (SRTM) to obtain the relative delivery rate (R1) and volume of distribution ratio (DVR). RLogan was least affected by CBF changes (χ2 = 583 p < 0.001, χ2 = 81 p < 0.001, for [18F]flutemetamol and [18F]florbetaben, respectively) and the extent of CBF sensitivity generally increased for higher levels of amyloid. Further, SRTM-derived R1 changes correlated well with simulated CBF changes (R2 > 0.95) and SUVr's sensitivity to CBF changes improved for later uptake-times, with the exception of [18F]flutemetamol cortical changes. In conclusion, RLogan is the preferred method for amyloid quantification of [18F]flutemetamol and [18F]florbetaben studies and SRTM could be additionally used for obtaining a CBF proxy.
Collapse
Affiliation(s)
- Fiona Heeman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Maqsood Yaqub
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Isadora Lopes Alves
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam, Netherlands
| | | | | | - Juan D Gispert
- Barcelonaβeta Brain Research Centre, Pasqual Maragall Foundation, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.,Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ronald Boellaard
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Adriaan A Lammertsma
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam, Netherlands
| | | |
Collapse
|
154
|
Verfaillie SC, Golla SS, Timmers T, Tuncel H, van der Weijden CW, Schober P, Schuit RC, van der Flier WM, Windhorst AD, Lammertsma AA, van Berckel BN, Boellaard R. Repeatability of parametric methods for [ 18F]florbetapir imaging in Alzheimer's disease and healthy controls: A test-retest study. J Cereb Blood Flow Metab 2021; 41:569-578. [PMID: 32321347 PMCID: PMC7907981 DOI: 10.1177/0271678x20915403] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Accumulation of amyloid beta (Aβ) is one of the pathological hallmarks of Alzheimer's disease (AD), which can be visualized using [18F]florbetapir positron emission tomography (PET). The aim of this study was to evaluate various parametric methods and to assess their test-retest (TRT) reliability. Two 90 min dynamic [18F]florbetapir PET scans, including arterial sampling, were acquired (n = 8 AD patient, n = 8 controls). The following parametric methods were used; (reference:cerebellum); Logan and spectral analysis (SA), receptor parametric mapping (RPM), simplified reference tissue model2 (SRTM2), reference Logan (rLogan) and standardized uptake value ratios (SUVr(50-70)). BPND+1, DVR, VT and SUVr were compared with corresponding estimates (VT or DVR) from the plasma input reversible two tissue compartmental (2T4k_VB) model with corresponding TRT values for 90-scan duration. RPM (r2 = 0.92; slope = 0.91), Logan (r2 = 0.95; slope = 0.84) and rLogan (r2 = 0.94; slope = 0.88), and SRTM2 (r2 = 0.91; slope = 0.83), SA (r2 = 0.91; slope = 0.88), SUVr (r2 = 0.84; slope = 1.16) correlated well with their 2T4k_VB counterparts. RPM (controls: 1%, AD: 3%), rLogan (controls: 1%, AD: 3%) and SUVr(50-70) (controls: 3%, AD: 8%) showed an excellent TRT reliability. In conclusion, most parametric methods showed excellent performance for [18F]florbetapir, but RPM and rLogan seem the methods of choice, combining the highest accuracy and best TRT reliability.
Collapse
Affiliation(s)
- Sander Cj Verfaillie
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands
| | - Sandeep Sv Golla
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands
| | - Tessa Timmers
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands.,Neurology & Alzheimer Center, Amsterdam Neuroscience, Amsterdam University Medical center location VUmc, The Netherlands
| | - Hayel Tuncel
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands
| | - Chris Wj van der Weijden
- Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Patrick Schober
- Department of Anaesthesiology, Amsterdam University Medical center location VUmc, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands
| | - Wiesje M van der Flier
- Neurology & Alzheimer Center, Amsterdam Neuroscience, Amsterdam University Medical center location VUmc, The Netherlands.,Epidemiology & Biostatistics, Amsterdam Neuroscience, Amsterdam University Medical center location VUmc, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands
| | - Bart Nm van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical center location VUmc, The Netherlands.,Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
155
|
Schoemaker D, Charidimou A, Zanon Zotin MC, Raposo N, Johnson KA, Sanchez JS, Greenberg SM, Viswanathan A. Association of Memory Impairment With Concomitant Tau Pathology in Patients With Cerebral Amyloid Angiopathy. Neurology 2021; 96:e1975-e1986. [PMID: 33627498 DOI: 10.1212/wnl.0000000000011745] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 01/13/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Relying on tau-PET imaging, this cross-sectional study explored whether memory impairment is linked to the presence of concomitant tau pathology in individuals with cerebral amyloid angiopathy (CAA). METHODS Forty-six patients with probable CAA underwent a neuropsychological examination and an MRI for quantification of structural markers of cerebral small vessel disease. A subset of these participants also completed a [11C]-Pittsburgh compound B (n = 39) and [18F]-flortaucipir (n = 40) PET for in vivo estimation of amyloid and tau burden, respectively. Participants were classified as amnestic or nonamnestic on the basis of neuropsychological performance. Statistical analyses were performed to examine differences in cognition, structural markers of cerebral small vessel disease, and amyloid- and tau-PET retention between participants with amnestic and those with nonamnestic CAA. RESULTS Patients with probable CAA with an amnestic presentation displayed a globally more severe profile of cognitive impairment, smaller hippocampal volume (p < 0.001), and increased tau-PET binding in regions susceptible to Alzheimer disease neurodegeneration (p = 0.003) compared to their nonamnestic counterparts. Amnestic and nonamnestic patients with CAA did not differ on any other MRI markers or on amyloid-PET binding. In a generalized linear model including all evaluated neuroimaging markers, tau-PET retention (β = -0.85, p = 0.001) and hippocampal volume (β = 0.64 p = 0.01) were the only significant predictors of memory performance. The cognitive profile of patients with CAA with an elevated tau-PET retention was distinctly characterized by a significantly lower performance on the memory domain (p = 0.004). CONCLUSIONS These results suggest that the presence of objective memory impairment in patients with probable CAA could serve as a marker for underlying tau pathology. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that tau-PET retention is related to the presence of objective memory impairment in patients with CAA.
Collapse
Affiliation(s)
- Dorothee Schoemaker
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston.
| | - Andreas Charidimou
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston
| | - Maria Clara Zanon Zotin
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston
| | - Nicolas Raposo
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston
| | - Keith A Johnson
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston
| | - Justin S Sanchez
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston
| | - Steven M Greenberg
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston
| | - Anand Viswanathan
- From the Departments of Psychiatry (D.S.), Neurology (A.C., M.C.Z.Z., K.A.J., J.S.S., S.M.G., A.V.), and Radiology (K.A.J., J.S.S.), Massachusetts General Hospital, Harvard Medical School, Boston; and Department of Neurology (N.R.), Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse (University Hospital Centre), France. M.C.Z.Z. is currently at the Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil; N.R. is at Toulouse Neuroimaging Center, Université de Toulouse, INSERM, UPS, France; and K.A.J. is at the Department of Neurology, Brigham and Women's Hospital, Boston
| |
Collapse
|
156
|
Validation of Parametric Methods for [ 11C]UCB-J PET Imaging Using Subcortical White Matter as Reference Tissue. Mol Imaging Biol 2021; 22:444-452. [PMID: 31209780 DOI: 10.1007/s11307-019-01387-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE The aim of this study was to evaluate different non-invasive methods for generating (R)-1-((3-([11C]methyl)pyridin-4-yl)methyl)-4-(3,4,5-trifluorophenyl)pyrrolidin-2-one) ([11C]UCB-J) parametric maps using white matter (centrum semi-ovale-SO) as reference tissue. PROCEDURES Ten healthy volunteers (8 M/2F; age 27.6 ± 10.0 years) underwent a 90-min dynamic [11C]UCB-J positron emission tomography (PET) scan with full arterial blood sampling and metabolite analysis before and after administration of a novel chemical entity with high affinity for presynaptic synaptic vesicle glycoprotein 2A (SV2A). A simplified reference tissue model (SRTM2), multilinear reference tissue model (MRTM2), and reference Logan graphical analysis (rLGA) were used to generate binding potential maps using SO as reference tissue (BPSO). Shorter dynamic acquisitions down to 50 min were also considered. In addition, standard uptake value ratios (SUVR) relative to SO were evaluated for three post-injection intervals (SUVRSO,40-70min, SUVRSO,50-80min, and SUVRSO,60-90min respectively). Regional parametric BPSO + 1 and SUVRSO were compared with regional distribution volume ratios of a 1-tissue compartment model (1TCM DVRSO) using Spearman correlation and Bland-Altman analysis. RESULTS For all methods, highly significant correlations were found between regional, parametric BPSO + 1 (r = [0.63;0.96]) or SUVRSO (r = [0.90;0.91]) estimates and regional 1TCM DVRSO. For a 90-min dynamic scan, parametric SRTM2 and MRTM2 values presented similar small bias and variability (- 3.0 ± 2.9 % for baseline SRTM2) and outperformed rLGA (- 10.0 ± 5.3 % for baseline rLGA). Reducing the dynamic acquisition to 60 min had limited impact on the bias and variability of parametric SRTM2 BPSO estimates (- 1.0 ± 9.9 % for baseline SRTM2) while a higher variability (- 1.83 ± 10.8 %) for baseline MRTM2 was observed for shorter acquisition times. Both SUVRSO,60-90min and SUVRSO,50-80min showed similar small bias and variability (- 2.8 ± 4.6 % bias for baseline SUVRSO,60-90min). CONCLUSION SRTM2 is the preferred method for a voxelwise analysis of dynamic [11C]UCB-J PET using SO as reference tissue, while reducing the dynamic acquisition to 60 min has limited impact on [11C]UCB-J BPSO parametric maps. For a static PET protocol, both SUVRSO,60-90min and SUVRSO,50-80min images are an excellent proxy for [11C]UCB-J BPSO parametric maps.
Collapse
|
157
|
A high-resolution in vivo atlas of the human brain's benzodiazepine binding site of GABA A receptors. Neuroimage 2021; 232:117878. [PMID: 33610745 PMCID: PMC8256681 DOI: 10.1016/j.neuroimage.2021.117878] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/15/2020] [Accepted: 02/12/2021] [Indexed: 12/29/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the human brain and plays a key role in several brain functions and neuropsychiatric disorders such as anxiety, epilepsy, and depression. For decades, several in vivo and ex vivo techniques have been used to highlight the mechanisms of the GABA system, however, no studies have currently combined the techniques to create a high-resolution multimodal view of the GABA system. Here, we present a quantitative high-resolution in vivo atlas of the human brain benzodiazepine receptor sites (BZR) located on postsynaptic ionotropic GABAA receptors (GABAA Rs), generated on the basis of in vivo [11C]flumazenil Positron Emission Tomography (PET) data. Next, based on ex vivo autoradiography data, we transform the PET-generated atlas from binding values into BZR protein density. Finally, we examine the brain regional association between BZR protein density and ex vivo mRNA expression for the 19 subunits in the GABAA R, including an estimation of the minimally required expression of mRNA levels for each subunit to translate into BZR protein. This represents the first publicly available quantitative high-resolution in vivo atlas of the spatial distribution of BZR densities in the healthy human brain. The atlas provides a unique neuroscientific tool as well as novel insights into the association between mRNA expression for individual subunits in the GABAA R and the BZR density at each location in the brain.
Collapse
|
158
|
Goodheart AE, Locascio JJ, Samore WR, Collins JA, Brickhouse M, Schultz A, Touroutoglou A, Johnson KA, Frosch MP, Growdon JH, Dickerson BC, Gomperts SN. 18F-AV-1451 positron emission tomography in neuropathological substrates of corticobasal syndrome. Brain 2021; 144:266-277. [PMID: 33578418 PMCID: PMC7880673 DOI: 10.1093/brain/awaa383] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 11/12/2022] Open
Abstract
Multiple neuropathological processes can manifest in life as a corticobasal syndrome. We sought to relate retention of the tau-PET tracer 18F-AV-1451 and structural magnetic resonance measures of regional atrophy to clinical features in clinically diagnosed and neuropathologically confirmed cases of corticobasal syndrome and to determine whether these vary with the underlying neuropathological changes. In this observational, cross-sectional study, 11 subjects (eight female and three male, median age 72 years) with corticobasal syndrome underwent structural MRI, tau-PET with 18F-AV-1451, amyloid-PET with 11C-Pittsburgh compound B, detailed clinical examinations and neuropsychological testing. Of the 11, three had evidence of high amyloid burden consistent with Alzheimer's disease while eight did not. Neuropathological evaluations were acquired in six cases. Mixed effects general linear models were used to compare 18F-AV-1451 retention and atrophy in amyloid-negative corticobasal syndrome cases to 32 age-matched healthy control subjects and to relate cortical and subcortical 18F-AV-1451 retention and atrophy to clinical features. Subjects without amyloid, including three with pathologically confirmed corticobasal degeneration, showed greater regional 18F-AV-1451 retention and associated regional atrophy in areas commonly associated with corticobasal degeneration pathology than healthy control subjects [retention was higher compared to healthy controls (P = 0.0011), driven especially by the precentral gyrus (P = 0.011) and pallidum (P < 0.0001), and greater atrophy was seen in subjects compared to control subjects (P = 0.0004)]. Both 18F-AV-1451 retention and atrophy were greater in the clinically more affected hemisphere [on average, retention was 0.173 standardized uptake value ratio units higher on the more affected side (95% confidence interval, CI 0.11-0.24, P < 0.0001), and volume was 0.719 lower on the more affected side (95% CI 0.35-1.08, P = 0.0001)]. 18F-AV-1451 retention was greater in subcortical than in cortical regions, P < 0.0001. In contrast to these findings, subjects with amyloid-positive corticobasal syndrome, including two neuropathologically confirmed cases of Alzheimer's disease, demonstrated greater and more widespread 18F-AV-1451 retention and regional atrophy than observed in the amyloid-negative cases. There was thalamic 18F-AV-1451 retention but minimal cortical and basal ganglia uptake in a single corticobasal syndrome subject without neuropathological evidence of tau pathology, likely representing non-specific signal. Asymmetric cortical and basal ganglia 18F-AV-1451 retention consonant with the clinical manifestations characterize corticobasal syndrome due to corticobasal degeneration, whereas the cortical retention in cases associated with Alzheimer's disease is greater and more diffuse.
Collapse
Affiliation(s)
- Anna E Goodheart
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph J Locascio
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Wesley R Samore
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica A Collins
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Michael Brickhouse
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Aaron Schultz
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew P Frosch
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - John H Growdon
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Bradford C Dickerson
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Stephen N Gomperts
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Massachusetts Alzheimer's Disease Research Center, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
159
|
Villemagne VL, Barkhof F, Garibotto V, Landau SM, Nordberg A, van Berckel BNM. Molecular Imaging Approaches in Dementia. Radiology 2021; 298:517-530. [PMID: 33464184 DOI: 10.1148/radiol.2020200028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The increasing prevalence of dementia worldwide places a high demand on healthcare providers to perform a diagnostic work-up in relatively early stages of the disease, given that the pathologic process usually begins decades before symptoms are evident. Structural imaging is recommended to rule out other disorders and can only provide diagnosis in a late stage with limited specificity. Where PET imaging previously focused on the spatial pattern of hypometabolism, the past decade has seen the development of novel tracers to demonstrate characteristic protein abnormalities. Molecular imaging using PET/SPECT is able to show amyloid and tau deposition in Alzheimer disease and dopamine depletion in parkinsonian disorders starting decades before symptom onset. Novel tracers for neuroinflammation and synaptic density are being developed to further unravel the molecular pathologic characteristics of dementia disorders. In this article, the authors review the current status of established and emerging PET tracers in a diagnostic setting and also their value as prognostic markers in research studies and outcome measures for clinical trials in Alzheimer disease.
Collapse
Affiliation(s)
- Victor L Villemagne
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Frederik Barkhof
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Valentina Garibotto
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Susan M Landau
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Agneta Nordberg
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| | - Bart N M van Berckel
- From the Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pa (V.L.V.); Department of Medicine, the University of Melbourne, Melbourne, Australia (V.L.V.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Amsterdam, the Netherlands (F.B., B.N.M.v.B.); UCL institutes of Neurology and Healthcare Engineering, London, England (F.B.); Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and Laboratory of Neuroimaging and Innovative Molecular Tracers, Geneva University, Geneva, Switzerland (V.G.); Helen Wills Neuroscience Institute, University of California, Berkeley, Calif (S.M.L.); Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, Calif (S.M.L.); Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden (A.N.); and Theme Aging, Karolinska University Hospital, Stockholm, Sweden (A.N.)
| |
Collapse
|
160
|
Hashimoto F, Ohba H, Ote K, Kakimoto A, Tsukada H, Ouchi Y. 4D deep image prior: dynamic PET image denoising using an unsupervised four-dimensional branch convolutional neural network. Phys Med Biol 2021; 66:015006. [PMID: 33227725 DOI: 10.1088/1361-6560/abcd1a] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although convolutional neural networks (CNNs) demonstrate the superior performance in denoising positron emission tomography (PET) images, a supervised training of the CNN requires a pair of large, high-quality PET image datasets. As an unsupervised learning method, a deep image prior (DIP) has recently been proposed; it can perform denoising with only the target image. In this study, we propose an innovative procedure for the DIP approach with a four-dimensional (4D) branch CNN architecture in end-to-end training to denoise dynamic PET images. Our proposed 4D CNN architecture can be applied to end-to-end dynamic PET image denoising by introducing a feature extractor and a reconstruction branch for each time frame of the dynamic PET image. In the proposed DIP method, it is not necessary to prepare high-quality and large patient-related PET images. Instead, a subject's own static PET image is used as additional information, dynamic PET images are treated as training labels, and denoised dynamic PET images are obtained from the CNN outputs. Both simulation with [18F]fluoro-2-deoxy-D-glucose (FDG) and preclinical data with [18F]FDG and [11C]raclopride were used to evaluate the proposed framework. The results showed that our 4D DIP framework quantitatively and qualitatively outperformed 3D DIP and other unsupervised denoising methods. The proposed 4D DIP framework thus provides a promising procedure for dynamic PET image denoising.
Collapse
Affiliation(s)
- Fumio Hashimoto
- Central Research Laboratory, Hamamatsu Photonics K. K., 5000 Hirakuchi, Hamakita-ku, Hamamatsu 434-8601, Japan
| | | | | | | | | | | |
Collapse
|
161
|
Krohn KA, Vera DR. Concepts for design and analysis of receptor radiopharmaceuticals: The Receptor-Binding Radiotracers series of meetings provided the foundation. Nucl Med Biol 2021; 92:5-23. [PMID: 32331709 PMCID: PMC8049838 DOI: 10.1016/j.nucmedbio.2020.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/08/2020] [Indexed: 12/14/2022]
Abstract
A symposium at George Washington University on Receptor-Binding Radiotracers in 1980 and three follow-up meetings held at University of California, San Diego provided a forum for debating the critical concepts involved in the new field of designing and evaluating radiotracers for imaging receptors and transporters. This review is intended to educate young investigators who may be relatively new to receptor radiopharmaceutical development. Our anticipated audience includes researchers in basic pharmacology, radiochemistry, imaging technology and kinetic data analysis and how these disciplines have worked together to build our understanding of the human biology of transporters and receptor signaling in health and disease. We have chosen to focus on radiochemical design of a useful imaging agent and how design is coupled to analysis of data collected from dynamic imaging with that agent. Some pharmacology may be required for designing the imaging agent and some imaging physics may be important in optimizing the quality of data that is collected. However, the key to a successful imaging agent is matching the radiotracer to the target receptor and to analysis of the time-course data that is used to parse delivery from specific binding and subsequent metabolism or degradation. Properly designed imaging agents are providing critical information about human biology in health and disease as well as pharmacodynamic response to drug interventions. The review emphasizes some of the ideas that were controversial at the 1980 conference and chronicles with literature examples how they have resolved over the four decades of using radiotracers to study transporters and receptors in human subjects. These examples show that there are situations where a very small KD, i.e. high affinity, has the potential to yield an image that reflects blood flow more than receptor density. The examples also show that by combining two studies, one with high specific activity and a second with low specific activity injections one can unravel the pseudo-first order rate B'max into the true second-order rate constant, k3, and the unoccupied receptor density. The final section describes how mathematical methods first presented to the receptor-imaging community in 1980 are now being used to provide confidence in the analysis of kinetic biodistribution studies. Our hope is that by bringing these concepts together in a single review, the next generation of scientists developing receptor imaging agents can be much more efficient than their pioneers in developing useful imaging methods.
Collapse
Affiliation(s)
- Kenneth A Krohn
- Center for Radiochemistry Research, Department of Diagnostic Radiology, Mail Code L104, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, United States of America.
| | - David R Vera
- UCSD Moores Cancer Center, Department of Radiology, Mail Code 0819, University of California, San Diego, CA 92037, United States of America
| |
Collapse
|
162
|
|
163
|
Pascali G, Panetta D, De Simone M, Burchielli S, Lucchesi V, Sanguinetti E, Zanoni S, Iozzo P, Saccomanni G, Manera C, Salvadori PA. Preliminary Investigation of a Novel 18F Radiopharmaceutical for Imaging CB2 Receptors in a SOD Mouse Model. Aust J Chem 2021. [DOI: 10.1071/ch20247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We successfully radiolabelled a novel prospective cannabinoid type 2 receptor ligand with 18F and tested its biodistribution in animal models by positron emission tomography (PET)/computed tomography (CT) imaging. The radiolabelling process was conducted on an alkyl mesylate fragment of the main naphthyridine core, using highly efficient microfluidic technology. No preliminary protection was needed, and the product was purified by semi-prep HPLC and SPE formulation, allowing the desired diastereomeric mixture to be obtained in 29% radiochemical yield and>95% radiochemically pure. SOD1G93A mice were used as model of overexpression of CB2 receptors; PET imaging revealed a significant increase of the tracer distribution volume in the brain of symptomatic subjects compared with the asymptomatic ones.
Collapse
|
164
|
Brody SL, Gunsten SP, Luehmann HP, Sultan DH, Hoelscher M, Heo GS, Pan J, Koenitzer JR, Lee EC, Huang T, Mpoy C, Guo S, Laforest R, Salter A, Russell TD, Shifren A, Combadiere C, Lavine KJ, Kreisel D, Humphreys BD, Rogers BE, Gierada DS, Byers DE, Gropler RJ, Chen DL, Atkinson JJ, Liu Y. Chemokine Receptor 2-targeted Molecular Imaging in Pulmonary Fibrosis. A Clinical Trial. Am J Respir Crit Care Med 2021; 203:78-89. [PMID: 32673071 PMCID: PMC7781144 DOI: 10.1164/rccm.202004-1132oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) is a progressive inflammatory lung disease without effective molecular markers of disease activity or treatment responses. Monocyte and interstitial macrophages that express the C-C motif CCR2 (chemokine receptor 2) are active in IPF and central to fibrosis.Objectives: To phenotype patients with IPF for potential targeted therapy, we developed 64Cu-DOTA-ECL1i, a radiotracer to noninvasively track CCR2+ monocytes and macrophages using positron emission tomography (PET).Methods: CCR2+ cells were investigated in mice with bleomycin- or radiation-induced fibrosis and in human subjects with IPF. The CCR2+ cell populations were localized relative to fibrotic regions in lung tissue and characterized using immunolocalization, single-cell mass cytometry, and Ccr2 RNA in situ hybridization and then correlated with parallel quantitation of lung uptake by 64Cu-DOTA-ECL1i PET.Measurements and Main Results: Mouse models established that increased 64Cu-DOTA-ECL1i PET uptake in the lung correlates with CCR2+ cell infiltration associated with fibrosis (n = 72). As therapeutic models, the inhibition of fibrosis by IL-1β blockade (n = 19) or antifibrotic pirfenidone (n = 18) reduced CCR2+ macrophage accumulation and uptake of the radiotracer in mouse lungs. In lung tissues from patients with IPF, CCR2+ cells concentrated in perifibrotic regions and correlated with radiotracer localization (n = 21). Human imaging revealed little lung uptake in healthy volunteers (n = 7), whereas subjects with IPF (n = 4) exhibited intensive signals in fibrotic zones.Conclusions: These findings support a role for imaging CCR2+ cells within the fibrogenic niche in IPF to provide a molecular target for personalized therapy and monitoring.Clinical trial registered with www.clinicaltrials.gov (NCT03492762).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christophe Combadiere
- INSERM, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Sorbonne Université, Paris, France
| | - Kory J. Lavine
- Department of Medicine
- Department of Developmental Biology
| | - Daniel Kreisel
- Department of Surgery, and
- Department of Immunology and Pathology, Washington University School of Medicine, Saint Louis, Missouri; and
| | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Viswanath V, Chitalia R, Pantel AR, Karp JS, Mankoff DA. Analysis of Four-Dimensional Data for Total Body PET Imaging. PET Clin 2021; 16:55-64. [PMID: 33218604 PMCID: PMC8722496 DOI: 10.1016/j.cpet.2020.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The high sensitivity and total-body coverage of total-body PET scanners will be valuable for a number of clinical and research applications outlined in this article.
Collapse
Affiliation(s)
- Varsha Viswanath
- Department of Radiology, University of Pennsylvania, John Morgan Building, 3620 Hamilton Walk, Room 150, Philadelphia, PA 19103, USA.
| | - Rhea Chitalia
- Department of Radiology, University of Pennsylvania, Richards Building, 3700 Hamilton Walk, Room D700, Philadelphia, PA 19103, USA
| | - Austin R Pantel
- Department of Radiology, University of Pennsylvania, Hospital of the University of Pennsylvania, 1 Donner Building, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
| | - Joel S Karp
- Department of Radiology, University of Pennsylvania, John Morgan Building, 3620 Hamilton Walk, Room 150, Philadelphia, PA 19103, USA
| | - David A Mankoff
- Department of Radiology, Abramson Cancer Center, University of Pennsylvania, Hospital of the University of Pennsylvania, 1 Donner Building, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
| |
Collapse
|
166
|
Thomsen MB, Ferreira SA, Schacht AC, Jacobsen J, Simonsen M, Betzer C, Jensen PH, Brooks DJ, Landau AM, Romero-Ramos M. PET imaging reveals early and progressive dopaminergic deficits after intra-striatal injection of preformed alpha-synuclein fibrils in rats. Neurobiol Dis 2020; 149:105229. [PMID: 33352233 DOI: 10.1016/j.nbd.2020.105229] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/24/2022] Open
Abstract
Alpha-synuclein (a-syn) can aggregate and form toxic oligomers and insoluble fibrils which are the main component of Lewy bodies. Intra-neuronal Lewy bodies are a major pathological characteristic of Parkinson's disease (PD). These fibrillar structures can act as seeds and accelerate the aggregation of monomeric a-syn. Indeed, recent studies show that injection of preformed a-syn fibrils (PFF) into the rodent brain can induce aggregation of the endogenous monomeric a-syn resulting in neuronal dysfunction and eventual cell death. We injected 8 μg of murine a-syn PFF, or soluble monomeric a-syn into the right striatum of rats. The animals were monitored behaviourally using the cylinder test, which measures paw asymmetry, and the corridor task that measures lateralized sensorimotor response to sugar treats. In vivo PET imaging was performed after 6, 13 and 22 weeks using [11C]DTBZ, a marker of the vesicular monoamine 2 transporter (VMAT2), and after 15 and 22 weeks using [11C]UCB-J, a marker of synaptic SV2A protein in nerve terminals. Histology was performed at the three time points using antibodies against dopaminergic markers, aggregated a-syn, and MHCII to evaluate the immune response. While the a-syn PFF injection caused only mild behavioural changes, [11C]DTBZ PET showed a significant and progressive decrease of VMAT2 binding in the ipsilateral striatum. This was accompanied by a small progressive decrease in [11C]UCB-J binding in the same area. In addition, our histological analysis revealed a gradual spread of misfolded a-syn pathology in areas anatomically connected to striatum that became bilateral with time. The striatal a-syn PFF injection resulted in a progressive unilateral degeneration of dopamine terminals, and an early and sustained presence of MHCII positive ramified microglia in the ipsilateral striatum and substantia nigra. Our study shows that striatal injections of a-syn fibrils induce progressive pathological synaptic dysfunction prior to cell death that can be detected in vivo with PET. We confirm that intrastriatal injection of a-syn PFFs provides a model of progressive a-syn pathology with loss of dopaminergic and synaptic function accompanied by neuroinflammation, as found in human PD.
Collapse
Affiliation(s)
- Majken B Thomsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Sara A Ferreira
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Anna C Schacht
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Jan Jacobsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Mette Simonsen
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark
| | - Cristine Betzer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Poul H Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Anne M Landau
- Department of Nuclear Medicine and PET Center, Institute of Clinical Medicine, Aarhus University and Hospital, Aarhus, Denmark; Translational Neuropsychiatry Unit, Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
167
|
Harrison TM, Du R, Klencklen G, Baker SL, Jagust WJ. Distinct effects of beta-amyloid and tau on cortical thickness in cognitively healthy older adults. Alzheimers Dement 2020; 17:1085-1096. [PMID: 33325068 PMCID: PMC8203764 DOI: 10.1002/alz.12249] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 10/22/2020] [Accepted: 11/01/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Published reports of associations between β-amyloid (Aβ) and cortical integrity conflict. Tau biomarkers may help elucidate the complex relationship between pathology and neurodegeneration in aging. METHODS We measured cortical thickness using magnetic resonance imaging, Aβ using Pittsburgh compound B positron emission tomography (PiB-PET), and tau using flortaucipir (FTP)-PET in 125 cognitively normal older adults. We examined relationships among PET measures, cortical thickness, and cognition. RESULTS Cortical thickness was reduced in PiB+/FTP+ participants compared to the PiB+/FTP- and PiB-/FTP- groups. Continuous PiB associations with cortical thickness were weak but positive in FTP- participants and negative in FTP+. FTP strongly negatively predicted thickness regardless of PiB status. FTP was associated with memory and cortical thickness, and mediated the association of PiB with memory. DISCUSSION Past findings linking Aβ and cortical thickness are likely weak due to opposing effects of Aβ on cortical thickness relative to tau burden. Tau, in contrast to Aβ, is strongly related to cortical thickness and memory.
Collapse
Affiliation(s)
- Theresa M Harrison
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, USA
| | - Richard Du
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, USA
| | - Giuliana Klencklen
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, USA
| | - Suzanne L Baker
- Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, USA.,Lawrence Berkeley National Laboratory, Berkeley, California, USA
| |
Collapse
|
168
|
Timmers T, Ossenkoppele R, Visser D, Tuncel H, Wolters EE, Verfaillie SCJ, van der Flier WM, Boellaard R, Golla SSV, van Berckel BNM. Test-retest repeatability of [ 18F]Flortaucipir PET in Alzheimer's disease and cognitively normal individuals. J Cereb Blood Flow Metab 2020; 40:2464-2474. [PMID: 31575335 PMCID: PMC7705644 DOI: 10.1177/0271678x19879226] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/18/2019] [Accepted: 08/08/2019] [Indexed: 11/29/2022]
Abstract
The aim of this study was to investigate the test-retest (TRT) repeatability of various parametric quantification methods for [18F]Flortaucipir positron emission tomography (PET). We included eight subjects with dementia or mild cognitive impairment due to Alzheimer's disease and six cognitively normal subjects. All underwent two 130-min dynamic [18F]Flortaucipir PET scans within 3 ± 1 weeks. Data were analyzed using reference region models receptor parametric mapping (RPM), simplified reference tissue method 2 (SRTM2) and reference logan (RLogan), as well as standardized uptake value ratios (SUVr, time intervals 40-60, 80-100 and 110-130 min post-injection) with cerebellar gray matter as reference region. We obtained distribution volume ratio or SUVr, first for all brain regions and then in three tau-specific regions-of-interest (ROIs). TRT repeatability (%) was defined as |retest-test|/(average (test + retest)) × 100. For all methods and across ROIs, TRT repeatability ranged from (median (IQR)) 0.84% (0.68-2.15) to 6.84% (2.99-11.50). TRT repeatability was good for all reference methods used, although semi-quantitative models (i.e. SUVr) performed marginally worse than quantitative models, for instance TRT repeatability of RPM: 1.98% (0.78-3.58) vs. SUVr80-100: 3.05% (1.28-5.52), p < 0.001. Furthermore, for SUVr80-100 and SUVr110-130, with higher average SUVr, more variation was observed. In conclusion, while TRT repeatability was good for all models used, quantitative methods performed slightly better than semi-quantitative methods.
Collapse
Affiliation(s)
- Tessa Timmers
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Denise Visser
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Hayel Tuncel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Emma E Wolters
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sander CJ Verfaillie
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sandeep SV Golla
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Bart NM van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
169
|
ADORA2A variation and adenosine A 1 receptor availability in the human brain with a focus on anxiety-related brain regions: modulation by ADORA1 variation. Transl Psychiatry 2020; 10:406. [PMID: 33235193 PMCID: PMC7686488 DOI: 10.1038/s41398-020-01085-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 10/11/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Adenosine, its interacting A1 and A2A receptors, and particularly the variant rs5751876 in the A2A gene ADORA2A have been shown to modulate anxiety, arousal, and sleep. In a pilot positron emission tomography (PET) study in healthy male subjects, we suggested an effect of rs5751876 on in vivo brain A1 receptor (A1AR) availability. As female sex and adenosinergic/dopaminergic interaction partners might have an impact on this rs5751876 effect on A1AR availability, we aimed to (1) further investigate the pilot male-based findings in an independent, newly recruited cohort including women and (2) analyze potential modulation of this rs5751876 effect by additional adenosinergic/dopaminergic gene variation. Healthy volunteers (32/11 males/females) underwent phenotypic characterization including self-reported sleep and A1AR-specific quantitative PET. Rs5751876 and 31 gene variants of adenosine A1, A2A, A2B, and A3 receptors, adenosine deaminase, and dopamine D2 receptor were genotyped. Multivariate analysis revealed an rs5751876 effect on A1AR availability (P = 0.047), post hoc confirmed in 30 of 31 brain regions (false discovery rate (FDR) corrected P values < 0.05), but statistically stronger in anxiety-related regions (e.g., amygdala, hippocampus). Additional effects of ADORA1 rs1874142 were identified; under its influence rs5751876 and rs5751876 × sleep had strengthened effects on A1AR availability (Pboth < 0.02; post hoc FDR-corrected Ps < 0.05 for 29/30 regions, respectively). Our results support the relationship between rs5751876 and A1AR availability. Additional impact of rs1874142, together with rs5751876 and sleep, might be involved in regulating arousal and thus the development of mental disorders like anxiety disorders. The interplay of further detected suggestive ADORA2A × DRD2 interaction, however, necessitates larger future samples more comparable to magnetic resonance imaging (MRI)-based samples.
Collapse
|
170
|
Brašić JR, Nandi A, Russell DS, Jennings D, Barret O, Mathur A, Slifer K, Sedlak T, Martin SD, Brinson Z, Vyas P, Seibyl JP, Berry-Kravis EM, Wong DF, Budimirovic DB. Reduced Expression of Cerebral Metabotropic Glutamate Receptor Subtype 5 in Men with Fragile X Syndrome. Brain Sci 2020; 10:E899. [PMID: 33255214 PMCID: PMC7760509 DOI: 10.3390/brainsci10120899] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/07/2020] [Accepted: 11/14/2020] [Indexed: 12/28/2022] Open
Abstract
Glutamatergic receptor expression is mostly unknown in adults with fragile X syndrome (FXS). Favorable behavioral effects of negative allosteric modulators (NAMs) of the metabotropic glutamate receptor subtype 5 (mGluR5) in fmr1 knockout (KO) mouse models have not been confirmed in humans with FXS. Measurement of cerebral mGluR5 expression in humans with FXS exposed to NAMs might help in that effort. We used positron emission tomography (PET) to measure the mGluR5 density as a proxy of mGluR5 expression in cortical and subcortical brain regions to confirm target engagement of NAMs for mGluR5s. The density and the distribution of mGluR5 were measured in two independent samples of men with FXS (N = 9) and typical development (TD) (N = 8). We showed the feasibility of this complex study including MRI and PET, meaning that this challenging protocol can be accomplished in men with FXS with an adequate preparation. Analysis of variance of estimated mGluR5 expression showed that mGluR5 expression was significantly reduced in cortical and subcortical regions of men with FXS in contrast to age-matched men with TD.
Collapse
Affiliation(s)
- James R. Brašić
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
| | - Ayon Nandi
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
| | - David S. Russell
- Clinical Research, Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Research Clinic, Invicro LLC, New Haven, CT 06510, USA
| | - Danna Jennings
- Clinical Research, Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Research Clinic, Invicro LLC, New Haven, CT 06510, USA
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Olivier Barret
- Clinical Research, Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
| | - Anil Mathur
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
| | - Keith Slifer
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Department of Behavioral Psychology, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Thomas Sedlak
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
- Department of Psychiatry and Behavioral Sciences-General Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Samuel D. Martin
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
- Department of Neuroscience, Zanvyl Krieger School of Arts and Sciences, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Zabecca Brinson
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
| | - Pankhuri Vyas
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
| | - John P. Seibyl
- Clinical Research, Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Research Clinic, Invicro LLC, New Haven, CT 06510, USA
| | - Elizabeth M. Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Dean F. Wong
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (A.M.); (T.S.); (S.D.M.); (Z.B.); (P.V.); (D.F.W.)
- Precision Radio-Theranostics Translational Laboratories, Mallinckrodt Institute of Radiology, School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Departments of Psychiatry and Neurogenetics, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
171
|
Okazawa H, Ikawa M, Tsujikawa T, Makino A, Mori T, Kiyono Y, Kosaka H. Noninvasive Measurement of [ 11C]PiB Distribution Volume Using Integrated PET/MRI. Diagnostics (Basel) 2020; 10:diagnostics10120993. [PMID: 33255169 PMCID: PMC7760725 DOI: 10.3390/diagnostics10120993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/18/2020] [Accepted: 11/22/2020] [Indexed: 11/16/2022] Open
Abstract
A noninvasive image-derived input function (IDIF) method using PET/MRI was applied to quantitative measurements of [11C] Pittsburgh compound-B (PiB) distribution volume (DV) and compared with other metrics. Fifty-three patients suspected of early dementia (71 ± 11 y) underwent 70 min [11C]PiB PET/MRI. Nineteen of them (68 ± 11 y) without head motion during the scan were enrolled in this study and compared with 16 age-matched healthy controls (CTL: 68 ± 11 y). The dynamic frames reconstructed from listmode PET data were used for DV calculation. IDIF with metabolite correction was applied to the Logan plot method, and DV was normalized into DV ratio (DVR) images using the cerebellar reference (DVRL). DVR and standardized uptake value ratio (SUVR) images were also calculated using the reference tissue graphical method (DVRr) and the 50–70 min static data with cerebellar reference, respectively. Cortical values were compared using the 3D-T1WI MRI segmentation. All patients were assigned to the early Alzheimer’s disease (eAD) group because of positive [11C]PiB accumulation. The correlations of regional values were better for DVRL vs. DVRr (r2 = 0.97) than for SUVR vs. DVRr (r2 = 0.88). However, all metrics clearly differentiated eAD from CTL with appropriate thresholds. Noninvasive quantitative [11C]PiB PET/MRI measurement provided equivalent DVRs with the two methods. SUVR images showed acceptable results despite inferior variability and image quality to DVR images.
Collapse
Affiliation(s)
- Hidehiko Okazawa
- Biomedical Imaging Research Center, University of Fukui, Eiheiji-cho 910-1193, Japan; (M.I.); (T.T.); (A.M.); (T.M.); (Y.K.)
- Correspondence: ; Tel.: +81-776-61-8491
| | - Masamichi Ikawa
- Biomedical Imaging Research Center, University of Fukui, Eiheiji-cho 910-1193, Japan; (M.I.); (T.T.); (A.M.); (T.M.); (Y.K.)
- Department of Advanced Medicine for Community Healthcare, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Tetsuya Tsujikawa
- Biomedical Imaging Research Center, University of Fukui, Eiheiji-cho 910-1193, Japan; (M.I.); (T.T.); (A.M.); (T.M.); (Y.K.)
| | - Akira Makino
- Biomedical Imaging Research Center, University of Fukui, Eiheiji-cho 910-1193, Japan; (M.I.); (T.T.); (A.M.); (T.M.); (Y.K.)
| | - Tetsuya Mori
- Biomedical Imaging Research Center, University of Fukui, Eiheiji-cho 910-1193, Japan; (M.I.); (T.T.); (A.M.); (T.M.); (Y.K.)
| | - Yasushi Kiyono
- Biomedical Imaging Research Center, University of Fukui, Eiheiji-cho 910-1193, Japan; (M.I.); (T.T.); (A.M.); (T.M.); (Y.K.)
| | - Hirotaka Kosaka
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
| |
Collapse
|
172
|
Regional Tau Effects on Prospective Cognitive Change in Cognitively Normal Older Adults. J Neurosci 2020; 41:366-375. [PMID: 33219003 DOI: 10.1523/jneurosci.2111-20.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/21/2020] [Accepted: 11/12/2020] [Indexed: 11/21/2022] Open
Abstract
Studies suggest that tau deposition starts in the anterolateral entorhinal cortex (EC) with normal aging, and that the presence of β-amyloid (Aβ) facilitates its spread to neocortex, which may reflect the beginning of Alzheimer's disease (AD). Functional connectivity between the anterolateral EC and the anterior-temporal (AT) memory network appears to drive higher tau deposition in AT than in the posterior-medial (PM) memory network. Here, we investigated whether this differential vulnerability to tau deposition may predict different cognitive consequences of EC, AT, and PM tau. Using 18F-flortaucipir (FTP) and 11C-Pittsburgh compound-B (PiB) positron emission tomography (PET) imaging, we measured tau and Aβ in 124 cognitively normal human older adults (74 females, 50 males) followed for an average of 2.8 years for prospective cognition. We found that higher FTP in all three regions was individually related to faster memory decline, and that the effects of AT and PM FTP, but not EC, were driven by Aβ+ individuals. Moreover, when we included all three FTP measures competitively in the same model, only AT FTP significantly predicted memory decline. Our data support a model whereby tau, facilitated by Aβ, transits from EC to cortical regions that are most closely associated with the anterolateral EC, which specifically affects memory in the initial stage of AD. Memory also appears to be affected by EC tau in the absence of Aβ, which may be less clinically consequential. These findings may provide clarification of differences between normal aging and AD, and elucidate the transition between the two stages.SIGNIFICANCE STATEMENT Tau and β-amyloid (Aβ) are hallmarks of Alzheimer's disease (AD) but are also found in cognitively normal people. It is unclear whether, and how, this early deposition of tau and Aβ may affect cognition in normal aging and the asymptomatic stage of AD. We show that tau deposition in the entorhinal cortex (EC), which is common in advanced age, predicts memory decline in older adults independent of Aβ, likely reflecting normal, age-related memory loss. In contrast, tau in anterior-temporal (AT) regions is most predictive of memory decline in Aβ+ individuals. These data support the idea that tau preferentially spreads to specific cortical regions, likely through functional connections, which plays a primary role in memory decline in the early stage of AD.
Collapse
|
173
|
Ibanez L, Bahena JA, Yang C, Dube U, Farias FHG, Budde JP, Bergmann K, Brenner-Webster C, Morris JC, Perrin RJ, Cairns NJ, O'Donnell J, Álvarez I, Diez-Fairen M, Aguilar M, Miller R, Davis AA, Pastor P, Kotzbauer P, Campbell MC, Perlmutter JS, Rhinn H, Harari O, Cruchaga C, Benitez BA. Functional genomic analyses uncover APOE-mediated regulation of brain and cerebrospinal fluid beta-amyloid levels in Parkinson disease. Acta Neuropathol Commun 2020; 8:196. [PMID: 33213513 PMCID: PMC7678051 DOI: 10.1186/s40478-020-01072-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 11/25/2022] Open
Abstract
Alpha-synuclein is the main protein component of Lewy bodies, the pathological hallmark of Parkinson's disease. However, genetic modifiers of cerebrospinal fluid (CSF) alpha-synuclein levels remain unknown. The use of CSF levels of amyloid beta1-42, total tau, and phosphorylated tau181 as quantitative traits in genetic studies have provided novel insights into Alzheimer's disease pathophysiology. A systematic study of the genomic architecture of CSF biomarkers in Parkinson's disease has not yet been conducted. Here, genome-wide association studies of CSF biomarker levels in a cohort of individuals with Parkinson's disease and controls (N = 1960) were performed. PD cases exhibited significantly lower CSF biomarker levels compared to controls. A SNP, proxy for APOE ε4, was associated with CSF amyloid beta1-42 levels (effect = - 0.5, p = 9.2 × 10-19). No genome-wide loci associated with CSF alpha-synuclein, total tau, or phosphorylated tau181 levels were identified in PD cohorts. Polygenic risk score constructed using the latest Parkinson's disease risk meta-analysis were associated with Parkinson's disease status (p = 0.035) and the genomic architecture of CSF amyloid beta1-42 (R2 = 2.29%; p = 2.5 × 10-11). Individuals with higher polygenic risk scores for PD risk presented with lower CSF amyloid beta1-42 levels (p = 7.3 × 10-04). Two-sample Mendelian Randomization revealed that CSF amyloid beta1-42 plays a role in Parkinson's disease (p = 1.4 × 10-05) and age at onset (p = 7.6 × 10-06), an effect mainly mediated by variants in the APOE locus. In a subset of PD samples, the APOE ε4 allele was associated with significantly lower levels of CSF amyloid beta1-42 (p = 3.8 × 10-06), higher mean cortical binding potentials (p = 5.8 × 10-08), and higher Braak amyloid beta score (p = 4.4 × 10-04). Together these results from high-throughput and hypothesis-free approaches converge on a genetic link between Parkinson's disease, CSF amyloid beta1-42, and APOE.
Collapse
Affiliation(s)
- Laura Ibanez
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - Jorge A Bahena
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - Chengran Yang
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - Umber Dube
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - Fabiana H G Farias
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - John P Budde
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - Kristy Bergmann
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - Carol Brenner-Webster
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
| | - John C Morris
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Richard J Perrin
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University, St. Louis, MO, 63110, USA
| | - Nigel J Cairns
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pathology and Immunology, Washington University, St. Louis, MO, 63110, USA
- College of Medicine and Health, University of Exeter, Exeter, Devon, UK
| | - John O'Donnell
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
| | - Ignacio Álvarez
- Memory Unit, Department of Neurology, University Hospital Mutua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
| | - Monica Diez-Fairen
- Memory Unit, Department of Neurology, University Hospital Mutua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
| | - Miquel Aguilar
- Memory Unit, Department of Neurology, University Hospital Mutua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
| | - Rebecca Miller
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
| | - Albert A Davis
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
| | - Pau Pastor
- Memory Unit, Department of Neurology, University Hospital Mutua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
- Fundació per a la Recerca Biomèdica i Social Mútua de Terrassa, University of Barcelona, Terrassa, Barcelona, Spain
| | - Paul Kotzbauer
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
| | - Meghan C Campbell
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
- Departments of Neuroscience and Radiology, Programs in Physical Therapy and Occupational Therapy, Washington University, St. Louis, MO, 63110, USA
| | - Joel S Perlmutter
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University, St. Louis, MO, 63110, USA
- Departments of Neuroscience and Radiology, Programs in Physical Therapy and Occupational Therapy, Washington University, St. Louis, MO, 63110, USA
| | - Herve Rhinn
- Department of Bioinformatics, Alector, INC, San Francisco, CA, 94080, USA
| | - Oscar Harari
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Carlos Cruchaga
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA
- Hope Center for Neurologic Disorders, Washington University, St. Louis, MO, 63110, USA
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bruno A Benitez
- Department of Psychiatry, BJC Institute of Health, Washington University School of Medicine, Box 8134, 425 S. Euclid Ave., St. Louis, MO, 63110, USA.
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, 63110, USA.
| |
Collapse
|
174
|
Bertoglio D, Verhaeghe J, Miranda A, Wyffels L, Stroobants S, Dominguez C, Munoz-Sanjuan I, Skinbjerg M, Liu L, Staelens S. Kinetic Modelling and Test-Retest Reproducibility for the Dopamine D 1R Radioligand [ 11C]SCH23390 in Healthy and Diseased Mice. Mol Imaging Biol 2020; 23:208-219. [PMID: 33179158 PMCID: PMC7910372 DOI: 10.1007/s11307-020-01561-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 01/25/2023]
Abstract
Purpose Our aim in this study was to compare different non-invasive pharmacokinetic models and assess test–retest reproducibility of the radioligand [11C]SCH23390 for the quantification of dopamine D1-like receptor (D1R) in both wild-type (WT) mice and heterozygous (HET) Q175DN mice as Huntington’s disease (HD) model. Procedures Adult WT (n = 9) and HET (n = 14) mice underwent a 90-min [11C]SCH23390 positron emission tomography (PET) scan followed by computed tomography (CT) to evaluate the pharmacokinetic modelling in healthy and diseased conditions. Additionally, 5 WT mice and 7 HET animals received a second [11C]SCH23390 PET scan for test–retest reproducibility. Parallel assessment of the simplified reference tissue model (SRTM), the multilinear reference tissue model (MRTM) and the Logan reference tissue model (Logan Ref) using the striatum as a receptor-rich region and the cerebellum as a receptor-free (reference) region was performed to define the most suitable method for regional- and voxel-based quantification of the binding potential (BPND). Finally, standardised uptake value ratio (SUVR-1) was assessed as a potential simplified measurement. Results For all models, we measured a significant decline in dopamine D1R density (e.g. SRTM = − 38.5 ± 5.0 %, p < 0.0001) in HET mice compared to WT littermates. Shortening the 90-min scan duration resulted in large underestimation of striatal BPND in both WT mice (SRTM 60 min: − 17.7 ± 2.8 %, p = 0.0078) and diseased HET (SRTM 60 min: − 13.1 ± 4.1 %, p = 0.0001). Striatal BPND measurements were very reproducible with an average test–retest variability below 5 % when using both MRTM and SRTM. Parametric BPND maps generated with SRTM were highly reliable, showing nearly perfect agreement to the regional analysis (r2 = 0.99, p < 0.0001). Finally, SRTM provided the most accurate estimate for relative tracer delivery R1 with both regional- and voxel-based analyses. SUVR-1 at different time intervals were not sufficiently reliable when compared to BPND (r2 < 0.66). Conclusions Ninety-minute acquisition and the use of SRTM for pharmacokinetic modelling is recommended. [11C]SCH23390 PET imaging demonstrates optimal characteristics for the study of dopamine D1R density in models of psychiatric and neurological disorders as exemplified in the Q175DN mouse model of HD. Supplementary Information The online version contains supplementary material available at 10.1007/s11307-020-01561-1.
Collapse
Affiliation(s)
- Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium
| | - Leonie Wyffels
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | | | | | | | - Longbin Liu
- CHDI Management/CHDI Foundation, Los Angeles, CA, USA
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
175
|
Yoon B, Baker SL, Korman D, Tennant VR, Harrison TM, Landau S, Jagust WJ. Conscientiousness is associated with less amyloid deposition in cognitively normal aging. Psychol Aging 2020; 35:993-999. [PMID: 33166168 DOI: 10.1037/pag0000582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Little is known about the association between personality and Alzheimer's disease (AD) biomarkers, and existing results are inconsistent. We aimed to determine whether personality was associated with β-amyloid (Aβ) accumulation in cognitively normal aging. One hundred twenty-nine participants were included in this cross-sectional study. Personality was measured with the Big Five Inventory (BFI) and brain Aβ deposition was assessed with [11C] Pittsburgh compound B (PiB)-positron emission tomography (PET) imaging. Conscientiousness scores had a negative association with global PiB distribution volume ratio (DVR) in all participants after adjusting for age, sex, education, and vascular risk factors (β[SE] = -0.19[0.09], 95% confidence interval [CI: -0.35, -0.02], p = .031), while agreeableness, extraversion, neuroticism, and openness had no association with global PiB DVR. Assuming the relative stability of personality traits, these findings suggest that conscientiousness may protect against Aβ accumulation in cognitively normal aging through mechanisms that are as yet unknown. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
|
176
|
Winer JR, Mander BA, Kumar S, Reed M, Baker SL, Jagust WJ, Walker MP. Sleep Disturbance Forecasts β-Amyloid Accumulation across Subsequent Years. Curr Biol 2020; 30:4291-4298.e3. [PMID: 32888482 PMCID: PMC7642104 DOI: 10.1016/j.cub.2020.08.017] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/08/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022]
Abstract
Experimental sleep-wake disruption in rodents and humans causally modulates β-amyloid (Aβ) dynamics (e.g., [1-3]). This leads to the hypothesis that, beyond cross-sectional associations, impaired sleep structure and physiology could represent prospective biomarkers of the speed with which Aβ accumulates over time. Here, we test the hypothesis that initial baseline measures of non-rapid eye movement (NREM) sleep slow-wave activity (SWA) and sleep quality (efficiency) provide future forecasting sensitivity to the rate of Aβ accumulation over subsequent years. A cohort of clinically normal older adults was assessed using objective sleep polysomnography in combination with longitudinal tracking of Aβ accumulation with [11C]PiB positron emission tomography (PET) imaging. Both the proportion of NREM SWA below 1 Hz and the measure of sleep efficiency predicted the speed (slope) of subsequent Aβ deposition over time, and these associations remained robust when taking into account additional cofactors of interest (e.g., age, sex, sleep apnea). Moreover, these measures were specific, such that no other macro- and microphysiological architecture metrics of sleep demonstrated such sensitivity. Our data support the proposal that objective sleep markers could be part of a set of biomarkers that statistically forecast the longitudinal trajectory of cortical Aβ deposition in the human brain. Sleep may therefore represent a potentially affordable, scalable, repeatable, and non-invasive tool for quantifying of Aβ pathological progression, prior to cognitive symptoms of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Joseph R Winer
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA
| | - Bryce A Mander
- Department of Psychiatry and Human Behavior, University of California, Irvine, 101 The City Drive, Orange, CA 92697, USA
| | - Samika Kumar
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA
| | - Mark Reed
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA
| | - Suzanne L Baker
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - William J Jagust
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, 132 Barker Hall, Berkeley, CA 94720, USA
| | - Matthew P Walker
- Center for Human Sleep Science, Department of Psychology, University of California, Berkeley, Berkeley Way West, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, 132 Barker Hall, Berkeley, CA 94720, USA.
| |
Collapse
|
177
|
Stær K, Iranzo A, Stokholm MG, Østergaard K, Serradell M, Otto M, Svendsen KB, Garrido A, Vilas D, Santamaria J, Møller A, Gaig C, Brooks DJ, Borghammer P, Tolosa E, Pavese N. Cortical cholinergic dysfunction correlates with microglial activation in the substantia innominata in REM sleep behavior disorder. Parkinsonism Relat Disord 2020; 81:89-93. [PMID: 33099132 DOI: 10.1016/j.parkreldis.2020.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/12/2020] [Accepted: 10/06/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION In vivo PET studies in patients with isolated REM sleep behavior disorder (iRBD) have shown presence of neuroinflammation (microglial activation) in the substantia nigra, and reduced cortical acetylcholinesterase activity, suggestive of cholinergic dysfunction, that was more widespread in patients with poorer cognitive performances. This study aimed to explore whether reduced cortical acetylcholinesterase activity in iRBD is linked to microglial activation in the substantia innominata (SI), the major source of cholinergic input to the cortex. METHODS We used 11C(R)-PK11195 and 11C-Donepezil PET to assess levels of activated microglia and cholinergic function, respectively, in 19 iRBD patients. 11C(R)-PK11195 binding potential (BPND) and 11C-Donepezil distribution volume ratio (DVR) values were correlated using the Pearson statistic. RESULTS We found that a lower cortical 11C-Donepezil DVR correlated with a higher 11C(R)-PK11195 BPND in the SI (r = -0.48, p = 0.04). At a voxel level, the strongest negative correlations were found in the frontal and temporal lobes. CONCLUSION Our results suggest that reduced cortical acetylcholinesterase activity observed in our iRBD patients could be linked to the occurrence of neuroinflammation in the SI. Early modulation of microglial activation might therefore preserve cortical cholinergic functions in these patients.
Collapse
Affiliation(s)
- Kristian Stær
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | - Alex Iranzo
- Department of Neurology, Hospital Clínic de Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, IDIBAPS, Universitat de Barcelona, Catalonia, Spain; Multidisciplinary Sleep Unit, Hospital Clinic, Barcelona, Spain
| | - Morten G Stokholm
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | | | - Mónica Serradell
- Department of Neurology, Hospital Clínic de Barcelona, Spain; Multidisciplinary Sleep Unit, Hospital Clinic, Barcelona, Spain
| | - Marit Otto
- Department of Clinical Neurophysiology, Aarhus University Hospital, Denmark
| | | | - Alicia Garrido
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, IDIBAPS, Universitat de Barcelona, Catalonia, Spain; Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Catalonia, Spain
| | - Dolores Vilas
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, IDIBAPS, Universitat de Barcelona, Catalonia, Spain; Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Catalonia, Spain
| | - Joan Santamaria
- Department of Neurology, Hospital Clínic de Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, IDIBAPS, Universitat de Barcelona, Catalonia, Spain; Multidisciplinary Sleep Unit, Hospital Clinic, Barcelona, Spain
| | - Arne Møller
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | - Carles Gaig
- Department of Neurology, Hospital Clínic de Barcelona, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, IDIBAPS, Universitat de Barcelona, Catalonia, Spain; Multidisciplinary Sleep Unit, Hospital Clinic, Barcelona, Spain
| | - David J Brooks
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark; Translational and Clinical Research Institute, Newcastle University, United Kingdom
| | - Per Borghammer
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | - Eduardo Tolosa
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Clínic, IDIBAPS, Universitat de Barcelona, Catalonia, Spain; Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Catalonia, Spain
| | - Nicola Pavese
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark; Translational and Clinical Research Institute, Newcastle University, United Kingdom.
| |
Collapse
|
178
|
Fu JF, Matarazzo M, McKenzie J, Neilson N, Vafai N, Dinelle K, Felicio AC, McKeown MJ, Stoessl AJ, Sossi V. Serotonergic System Impacts Levodopa Response in Early Parkinson's and Future Risk of Dyskinesia. Mov Disord 2020; 36:389-397. [PMID: 33090574 DOI: 10.1002/mds.28340] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/31/2020] [Accepted: 10/03/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The serotonergic system is known to contribute to levodopa-derived dopamine release in advanced Parkinson's disease. OBJECTIVE We investigated the role of the serotonergic system in determining response to treatment in early disease and risk for complications concurrently with dopaminergic alterations. METHODS Eighteen patients with early and stable Parkinson's disease underwent multitracer positron emission tomography using [11 C]dihydrotetrabenazine (vesicular monoamine transporter 2 marker), [11 C]methylphenidate (dopamine transporter marker), [11 C]-3-amino-4-(2-dimethylaminomethylphenylsulfanyl)-benzonitrile (DASB, serotonin transporter marker), and [11 C]raclopride (D2 marker) to investigate relationships between striatal dopaminergic and serotonergic alterations and levodopa-induced dopamine release, related to motor response to treatment and risk for dyskinesias, using a novel joint pattern analysis. RESULTS The joint pattern analysis revealed correlated spatial patterns conceptually related to abnormal dopamine turnover in the putamen (higher dopamine release associated with dopaminergic and serotonergic denervation); response to treatment significantly inversely correlated with turnover-related dopamine release (P < 10-5 ). Patterns identified without inclusion of the DASB data showed no correlation with clinical data, indicating an important contribution from the serotonergic system to a clinically relevant abnormal dopamine release in early disease. Subjects who experienced dyskinesia 3 years after baseline scans showed higher turnover-related dopamine release compared with subjects who remained stable (P < 0.01). CONCLUSIONS Joint analysis of dopaminergic and serotonergic data identified a turnover-related dopamine release component, strongly related to motor response to levodopa in early disease and contributing to higher risk for dyskinesia. These findings suggest that the contribution of the serotonergic system to dopamine release not only increases the risk for motor complications but also fails to provide sustained therapeutic advantage in early disease. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jessie F Fu
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michele Matarazzo
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada
| | - Jessamyn McKenzie
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada
| | - Nicole Neilson
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada
| | - Nasim Vafai
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada
| | - Katie Dinelle
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada
| | - Andre C Felicio
- Department of Neurology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Martin J McKeown
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada.,Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - A Jon Stoessl
- Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada.,Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada.,Djavad Mowafaghian Centre for Brain Health, Pacific Parkinson's Research Centre, University of British Columbia & Vancouver Coastal Health, Vancouver, British Columbia, Canada
| |
Collapse
|
179
|
Heeman F, Hendriks J, Lopes Alves I, Ossenkoppele R, Tolboom N, van Berckel BNM, Lammertsma AA, Yaqub M. [ 11C]PIB amyloid quantification: effect of reference region selection. EJNMMI Res 2020; 10:123. [PMID: 33074395 PMCID: PMC7572969 DOI: 10.1186/s13550-020-00714-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/24/2020] [Indexed: 11/24/2022] Open
Abstract
Background The standard reference region (RR) for amyloid-beta (Aβ) PET studies is the cerebellar grey matter (GMCB), while alternative RRs have mostly been utilized without prior validation against the gold standard. This study compared five commonly used RRs to gold standard plasma input-based quantification using the GMCB. Methods Thirteen subjects from a test–retest (TRT) study and 30 from a longitudinal study were retrospectively included (total: 17 Alzheimer’s disease, 13 mild cognitive impairment, 13 controls). Dynamic [11C]PiB PET (90 min) and T1-weighted MR scans were co-registered and time–activity curves were extracted for cortical target regions and the following RRs: GMCB, whole cerebellum (WCB), white matter brainstem/pons (WMBS), whole brainstem (WBS) and eroded subcortical white matter (WMES). A two-tissue reversible plasma input model (2T4k_Vb) with GMCB as RR, reference Logan and the simplified reference tissue model were used to derive distribution volume ratios (DVRs), and standardized uptake value (SUV) ratios were calculated for 40–60 min and 60–90 min intervals. Parameter variability was evaluated using TRT scans, and correlations and agreements with the gold standard (DVR from 2T4k_Vb with GMCB RR) were also assessed. Next, longitudinal changes in SUVs (both intervals) were assessed for each RR. Finally, the ability to discriminate between visually Aβ positive and Aβ negative scans was assessed. Results All RRs yielded stable TRT performance (max 5.1% variability), with WCB consistently showing lower variability. All approaches were able to discriminate between Aβ positive and Aβ negative scans, with highest effect sizes obtained for GMCB (range − 0.9 to − 0.7), followed by WCB (range − 0.8 to − 0.6). Furthermore, all approaches provided good correlations with the gold standard (r ≥ 0.78), while the highest bias (as assessed by the regression slope) was observed using WMES (range slope 0.52–0.67), followed by WBS (range slope 0.58–0.92) and WMBS (range slope 0.62–0.91). Finally, RR SUVs were stable across a period of 2.6 years for all except WBS and WMBS RRs (60–90 min interval). Conclusions GMCB and WCB are considered the best RRs for quantifying amyloid burden using [11C]PiB PET.
Collapse
Affiliation(s)
- Fiona Heeman
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Janine Hendriks
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Isadora Lopes Alves
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Rik Ossenkoppele
- Neurology and Alzheimer Center, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.,Clinical Memory Research Unit, Lund University, Malmö, Sweden
| | - Nelleke Tolboom
- Imaging Division, Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | |
Collapse
|
180
|
Mueller KD, Koscik RL, Du L, Bruno D, Jonaitis EM, Koscik AZ, Christian BT, Betthauser TJ, Chin NA, Hermann BP, Johnson SC. Proper names from story recall are associated with beta-amyloid in cognitively unimpaired adults at risk for Alzheimer's disease. Cortex 2020; 131:137-150. [PMID: 32861209 PMCID: PMC7530114 DOI: 10.1016/j.cortex.2020.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/11/2020] [Accepted: 07/21/2020] [Indexed: 12/29/2022]
Abstract
Due to advances in the early detection of Alzheimer's disease (AD) biomarkers including beta-amyloid (Aβ), neuropsychological measures that are sensitive to concurrent, subtle changes in cognition are critically needed. Story recall tasks have shown sensitivity to early memory declines in persons with mild cognitive impairment (MCI) and early stage dementia, as well as in persons with autosomal dominantly inherited AD up to 10 years prior to a dementia diagnosis. However, the evidence is inconclusive regarding relationships between evidence of Aβ and story recall measures. Because story recall tasks require the encoding and delayed retrieval of several lexical-semantic categories, such as proper names, verbs, and numerical expressions, and because lexical categories have been shown to be differentially impaired in persons with MCI, we focused on item-level analyses of lexical-semantic retrieval from a quintessential story recall task, Logical Memory from the Wechsler Memory Scale-Revised. Our objective was to investigate whether delayed recall of lexical categories (proper names, verbs and/or numerical expressions), as well as the traditional total score measure, was associated with "preclinical AD," or cognitively unimpaired adults with positive Aβ deposition on positron emission tomography (PET) neuroimaging using Pittsburgh Compound B (PiB). We developed an item-level scoring system, in which we parsed items into lexical categories and examined the immediate and delayed recall of these lexical categories from 217 cognitively unimpaired participants from the Wisconsin Registry for Alzheimer's Prevention (WRAP). We performed binary logistic regression models with story recall score as predictor and Aβ status (positive/negative) as the outcome. Using baseline Logical Memory data, proper names from delayed story recall were significantly associated with Aβ status, such that participants who recalled more proper names were less likely to be classified as PiB(+) (odds ratio = .58, p = .01). None of the other story recall variables, including total score, were associated with PiB status. Secondary analyses determined that immediate recall of proper names was not significantly associated with Aβ, suggesting a retrieval deficit rather than that of encoding. The present findings suggest that lexical semantic retrieval measures from existing story recall tasks may be sensitive to Aβ deposition, and may provide added utility to a widely-used, long-standing neuropsychological test for early detection of cognitive decline on the AD continuum.
Collapse
Affiliation(s)
- Kimberly D Mueller
- Department of Communication Sciences and Disorders, University of Wisconsin - Madison, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Rebecca L Koscik
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Lianlian Du
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Davide Bruno
- School of Natural Sciences and Psychology, Liverpool John Moores University, Liverpool, UK
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Audra Z Koscik
- Department of Communication Sciences and Disorders, University of Wisconsin - Madison, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin - Madison, Madison, WI, USA; Department of Medical Physics, University of Wisconsin - Madison, Madison, WI, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Medical Physics, University of Wisconsin - Madison, Madison, WI, USA
| | - Nathaniel A Chin
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bruce P Hermann
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Neurology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Veterans Hospital, Madison, WI, USA
| |
Collapse
|
181
|
Buchecker V, Waldron AM, van Dijk RM, Koska I, Brendel M, von Ungern-Sternberg B, Lindner S, Gildehaus FJ, Ziegler S, Bartenstein P, Potschka H. [ 18F]MPPF and [ 18F]FDG μPET imaging in rats: impact of transport and restraint stress. EJNMMI Res 2020; 10:112. [PMID: 32990819 PMCID: PMC7524912 DOI: 10.1186/s13550-020-00693-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
Background Stress exposure can significantly affect serotonergic signaling with a particular impact on 5-HT1A receptor expression. Positron emission tomography (PET) provides opportunities for molecular imaging of alterations in 5-HT1A receptor binding following stress exposure. Considering the possible role of 5-HT1A receptors in stress coping mechanisms, respective imaging approaches are of particular interest. Material and methods For twelve consecutive days, Sprague Dawley rats were exposed to daily transport with a 1 h stay in a laboratory or daily transport plus 1 h restraint in a narrow tube. Following, animals were subjected to μPET imaging with 2′-methoxyphenyl-(N-2′-pyridinyl)-p-[18F]fluoro-benzamidoethylpiperazine ([18F]MPPF) and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG). Behavioral and biochemical parameters were analyzed to obtain additional information. Results In rats with repeated transport, hippocampal [18F]MPPF binding exceeded that in the naive group, while no difference in [18F]FDG uptake was detected between the groups. A transient decline in body weight was observed in rats with transport or combined transport and restraint. Thereby, body weight development correlated with [18F]MPPF binding. Conclusions Mild-to-moderate stress associated with daily transport and exposure to a laboratory environment can trigger significant alterations in hippocampal binding of the 5-HT1A receptor ligand [18F]MPPF. This finding indicates that utmost care is necessary to control and report transport and associated handling procedures for animals used in μPET studies analyzing the serotonergic system in order to enhance the robustness of conclusions and allow replicability of findings. In view of earlier studies indicating that an increase in hippocampal 5-HT1A receptor expression may be associated with a resilience to stress, it would be of interest to further evaluate 5-HT1A receptor imaging approaches as a candidate biomarker for the vulnerability to stress.
Collapse
Affiliation(s)
- Verena Buchecker
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Königinstr. 16, 80539, Munich, Germany
| | - Ann-Marie Waldron
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Königinstr. 16, 80539, Munich, Germany
| | - R Maarten van Dijk
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Königinstr. 16, 80539, Munich, Germany
| | - Ines Koska
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Königinstr. 16, 80539, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University, Munich, Germany
| | | | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Franz Josef Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Königinstr. 16, 80539, Munich, Germany.
| |
Collapse
|
182
|
Oh SJ, Lee HJ, Jeong YJ, Nam KR, Kang KJ, Han SJ, Lee KC, Lee YJ, Choi JY. Evaluation of the neuroprotective effect of taurine in Alzheimer's disease using functional molecular imaging. Sci Rep 2020; 10:15551. [PMID: 32968166 PMCID: PMC7511343 DOI: 10.1038/s41598-020-72755-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/04/2020] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the leading cause of dementia, but therapeutic treatment options are limited. Taurine has been reported to have neuroprotective properties against dementia, including AD. The present study aimed to investigate the treatment effect of taurine in AD mice by functional molecular imaging. To elucidate glutamate alterations by taurine, taurine was administered to 5xFAD transgenic mice from 2 months of age, known to apear amyloid deposition. Then, we performed glutamate positron emission tomography (PET) imaging studies for three groups (wild-type, AD, and taurine-treated AD, n = 5 in each group). As a result, brain uptake in the taurine-treated AD group was 31-40% higher than that in the AD group (cortex: 40%, p < 0.05; striatum: 32%, p < 0.01; hippocampus: 36%, p < 0.01; thalamus: 31%, p > 0.05) and 3-14% lower than that in the WT group (cortex: 10%, p > 0.05; striatum: 15%, p > 0.05; hippocampus: 14%, p > 0.05; thalamus: 3%, p > 0.05). However, we did not observe differences in Aβ pathology between the taurine-treated AD and AD groups in immunohistochemistry experiments. Our results reveal that although taurine treatment did not completely recover the glutamate system, it significantly increased metabolic glutamate receptor type 5 brain uptake. Therefore, taurine has therapeutic potential against AD.
Collapse
Affiliation(s)
- Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Hae-June Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Ye Ji Jeong
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Sang Jin Han
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea.
| |
Collapse
|
183
|
Yamasaki T, Zhang X, Kumata K, Zhang Y, Deng X, Fujinaga M, Chen Z, Mori W, Hu K, Wakizaka H, Hatori A, Xie L, Ogawa M, Nengaki N, Van R, Shao Y, Sheffler DJ, Cosford NDP, Liang SH, Zhang MR. Identification and Development of a New Positron Emission Tomography Ligand 4-(2-Fluoro-4-[ 11C]methoxyphenyl)-5-((1-methyl-1 H-pyrazol-3-yl)methoxy)picolinamide for Imaging Metabotropic Glutamate Receptor Subtype 2 (mGlu 2). J Med Chem 2020; 63:11469-11483. [PMID: 32960052 DOI: 10.1021/acs.jmedchem.9b01991] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Metabotropic glutamate receptor 2 (mGlu2) is a known target for treating several central nervous system (CNS) disorders. To develop a viable positron emission tomography (PET) ligand for mGlu2, we identified new candidates 5a-i that are potent negative allosteric modulators (NAMs) of mGlu2. Among these candidates, 4-(2-fluoro-4-methoxyphenyl)-5-((1-methyl-1H-pyrazol-3-yl)methoxy)picolinamide (5i, also named as [11C]MG2-1812) exhibited high potency, high subtype selectivity, and favorable lipophilicity. Compound 5i was labeled with positron-emitting carbon-11 (11C) to obtain [11C]5i in high radiochemical yield and high molar activity by O-[11C]methylation of the phenol precursor 12 with [11C]CH3I. In vitro autoradiography with [11C]5i showed heterogeneous radioactive accumulation in the brain tissue sections, ranked in the order: cortex > striatum > hippocampus > cerebellum ≫ thalamus > pons. PET study of [11C]5i indicated in vivo specific binding of mGlu2 in the rat brain. Based on the [11C]5i scaffold, further optimization for new candidates is underway to identify a more suitable ligand for imaging mGlu2.
Collapse
Affiliation(s)
- Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Xiaofei Zhang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Xiaoyun Deng
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Masayuki Fujinaga
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Wakana Mori
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hidekatsu Wakizaka
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Akiko Hatori
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Masanao Ogawa
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan.,SHI Accelerator Service, Ltd., 1-17-6 Osaki, Shinagawa-ku, Tokyo 141-0032, Japan
| | - Nobuki Nengaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan.,SHI Accelerator Service, Ltd., 1-17-6 Osaki, Shinagawa-ku, Tokyo 141-0032, Japan
| | - Richard Van
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Douglas J Sheffler
- Cancer Metabolism and Signaling Networks Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Nicholas D P Cosford
- Cancer Metabolism and Signaling Networks Program and Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
184
|
Meyer M, Lamare F, Asselineau J, Foubert‐Samier A, Mazère J, Zanotti‐Fregonara P, Rizzo G, Delamarre A, Spampinato U, Rascol O, Pavy‐Le Traon A, Tison F, Fernandez P, Sibon I, Meissner WG. Brain
5‐HT1A
Receptor Binding in Multiple System Atrophy: An [
18
F
]‐
MPPF PET
Study. Mov Disord 2020; 36:246-251. [DOI: 10.1002/mds.28295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/05/2020] [Accepted: 08/26/2020] [Indexed: 11/09/2022] Open
Affiliation(s)
- Marie Meyer
- Service de Médecine Nucléaire, CHU de Bordeaux Bordeaux France
- Institut des Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS, UMR 5287, Bordeaux University Bordeaux France
| | - Frédéric Lamare
- Service de Médecine Nucléaire, CHU de Bordeaux Bordeaux France
- Institut des Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS, UMR 5287, Bordeaux University Bordeaux France
| | - Julien Asselineau
- Public Health Department, Clinical Epidemiology Unit Bordeaux University Hospital Bordeaux France
| | - Alexandra Foubert‐Samier
- Service de Neurologie, CHU Bordeaux Bordeaux France
- French Reference Centre for MSA University Hospital Bordeaux Bordeaux France
- Inserm, UMR1219, Bordeaux Population Health Research Center, Bordeaux University, ISPED Bordeaux France
- Univ. de Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, F‐33000, France Bordeaux France
| | - Joachim Mazère
- Service de Médecine Nucléaire, CHU de Bordeaux Bordeaux France
- Institut des Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS, UMR 5287, Bordeaux University Bordeaux France
| | - Paolo Zanotti‐Fregonara
- Service de Médecine Nucléaire, CHU de Bordeaux Bordeaux France
- Institut des Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS, UMR 5287, Bordeaux University Bordeaux France
| | - Gaia Rizzo
- Invicro and Division of Brain Sciences Imperial College London London UK
| | - Anna Delamarre
- French Reference Centre for MSA University Hospital Bordeaux Bordeaux France
- Univ. de Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, F‐33000, France Bordeaux France
| | | | - Olivier Rascol
- French Reference Centre for MSA University Hospital Toulouse Toulouse France
- Clinical Investigation Center CIC 1436 and Departments of Neurosciences and Clinical Pharmacology Inserm, Toulouse University and CHU Toulouse Toulouse France
| | - Anne Pavy‐Le Traon
- French Reference Centre for MSA University Hospital Toulouse Toulouse France
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm U 1048, Toulouse University Toulouse France
| | - François Tison
- Service de Neurologie, CHU Bordeaux Bordeaux France
- French Reference Centre for MSA University Hospital Bordeaux Bordeaux France
- Univ. de Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, F‐33000, France Bordeaux France
| | - Philippe Fernandez
- Service de Médecine Nucléaire, CHU de Bordeaux Bordeaux France
- Institut des Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS, UMR 5287, Bordeaux University Bordeaux France
| | - Igor Sibon
- Service de Neurologie, CHU Bordeaux Bordeaux France
| | - Wassilios G Meissner
- Service de Neurologie, CHU Bordeaux Bordeaux France
- French Reference Centre for MSA University Hospital Bordeaux Bordeaux France
- Univ. de Bordeaux, CNRS, IMN, UMR 5293, Bordeaux, F‐33000, France Bordeaux France
- Department of Medicine University of Otago, Christchurch, and New Zealand Brain Research Institute Christchurch New Zealand
| |
Collapse
|
185
|
de Boer L, Garzón B, Axelsson J, Riklund K, Nyberg L, Bäckman L, Guitart-Masip M. Corticostriatal White Matter Integrity and Dopamine D1 Receptor Availability Predict Age Differences in Prefrontal Value Signaling during Reward Learning. Cereb Cortex 2020; 30:5270-5280. [PMID: 32484215 PMCID: PMC7472214 DOI: 10.1093/cercor/bhaa104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 12/25/2022] Open
Abstract
Probabilistic reward learning reflects the ability to adapt choices based on probabilistic feedback. The dopaminergically innervated corticostriatal circuit in the brain plays an important role in supporting successful probabilistic reward learning. Several components of the corticostriatal circuit deteriorate with age, as it does probabilistic reward learning. We showed previously that D1 receptor availability in NAcc predicts the strength of anticipatory value signaling in vmPFC, a neural correlate of probabilistic learning that is attenuated in older participants and predicts probabilistic reward learning performance. We investigated how white matter integrity in the pathway between nucleus accumbens (NAcc) and ventromedial prefrontal cortex (vmPFC) relates to the strength of anticipatory value signaling in vmPFC in younger and older participants. We found that in a sample of 22 old and 23 young participants, fractional anisotropy in the pathway between NAcc and vmPFC predicted the strength of value signaling in vmPFC independently from D1 receptor availability in NAcc. These findings provide tentative evidence that integrity in the dopaminergic and white matter pathways of corticostriatal circuitry supports the expression of value signaling in vmPFC which supports reward learning, however, the limited sample size calls for independent replication. These and future findings could add to the improved understanding of how corticostriatal integrity contributes to reward learning ability.
Collapse
Affiliation(s)
- Lieke de Boer
- Neurobiology, Care Sciences and Society, Aging Research Center, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Benjamín Garzón
- Neurobiology, Care Sciences and Society, Aging Research Center, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Jan Axelsson
- Department of Radiation Sciences, Diagnostic Radiology, University Hospital, Umeå University, Umeå SE-901 87, Sweden.,Department of Integrative Medical Biology, Physiology, Umeå University, Umeå SE-901 87, Sweden
| | - Katrine Riklund
- Department of Radiation Sciences, Diagnostic Radiology, University Hospital, Umeå University, Umeå SE-901 87, Sweden.,Department of Integrative Medical Biology, Physiology, Umeå University, Umeå SE-901 87, Sweden
| | - Lars Nyberg
- Department of Radiation Sciences, Diagnostic Radiology, University Hospital, Umeå University, Umeå SE-901 87, Sweden.,Department of Integrative Medical Biology, Physiology, Umeå University, Umeå SE-901 87, Sweden.,Umeå Center for Functional Brain Imaging, Umeå University, Umeå 907 36, Sweden
| | - Lars Bäckman
- Neurobiology, Care Sciences and Society, Aging Research Center, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Marc Guitart-Masip
- Neurobiology, Care Sciences and Society, Aging Research Center, Karolinska Institutet, Stockholm 171 65, Sweden.,Max Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, London WC1B 5EH, UK
| |
Collapse
|
186
|
Wei W, Poirion E, Bodini B, Tonietto M, Durrleman S, Colliot O, Stankoff B, Ayache N. Predicting PET-derived myelin content from multisequence MRI for individual longitudinal analysis in multiple sclerosis. Neuroimage 2020; 223:117308. [PMID: 32889117 DOI: 10.1016/j.neuroimage.2020.117308] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/20/2020] [Accepted: 08/21/2020] [Indexed: 12/31/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating and inflammatory disease of the central nervous system (CNS). The demyelination process can be repaired by the generation of a new sheath of myelin around the axon, a process termed remyelination. In MS patients, the demyelination-remyelination cycles are highly dynamic. Over the years, magnetic resonance imaging (MRI) has been increasingly used in the diagnosis of MS and it is currently the most useful paraclinical tool to assess this diagnosis. However, conventional MRI pulse sequences are not specific for pathological mechanisms such as demyelination and remyelination. Recently, positron emission tomography (PET) with radiotracer [11C]PIB has become a promising tool to measure in-vivo myelin content changes which is essential to push forward our understanding of mechanisms involved in the pathology of MS, and to monitor individual patients in the context of clinical trials focused on repair therapies. However, PET imaging is invasive due to the injection of a radioactive tracer. Moreover, it is an expensive imaging test and not offered in the majority of medical centers in the world. In this work, by using multisequence MRI, we thus propose a method to predict the parametric map of [11C]PIB PET, from which we derived the myelin content changes in a longitudinal analysis of patients with MS. The method is based on the proposed conditional flexible self-attention GAN (CF-SAGAN) which is specifically adjusted for high-dimensional medical images and able to capture the relationships between the spatially separated lesional regions during the image synthesis process. Jointly applying the sketch-refinement process and the proposed attention regularization that focuses on the MS lesions, our approach is shown to outperform the state-of-the-art methods qualitatively and quantitatively. Specifically, our method demonstrated a superior performance for the prediction of myelin content at voxel-wise level. More important, our method for the prediction of myelin content changes in patients with MS shows similar clinical correlations to the PET-derived gold standard indicating the potential for clinical management of patients with MS.
Collapse
Affiliation(s)
- Wen Wei
- Université Côte d'Azur, Inria, Epione Project-Team, Sophia Antipolis, France; Inria, Aramis Project-Team, Paris, France; Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France.
| | - Emilie Poirion
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France
| | - Benedetta Bodini
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France; APHP, Hôpital Saint Antoine, Neurology Department, Paris, France
| | - Matteo Tonietto
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France
| | - Stanley Durrleman
- Inria, Aramis Project-Team, Paris, France; Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France
| | - Olivier Colliot
- Inria, Aramis Project-Team, Paris, France; Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France
| | - Bruno Stankoff
- Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013 Paris, France; APHP, Hôpital Saint Antoine, Neurology Department, Paris, France
| | - Nicholas Ayache
- Université Côte d'Azur, Inria, Epione Project-Team, Sophia Antipolis, France
| |
Collapse
|
187
|
Karalija N, Jonassson L, Johansson J, Papenberg G, Salami A, Andersson M, Riklund K, Nyberg L, Boraxbekk CJ. High long-term test-retest reliability for extrastriatal 11C-raclopride binding in healthy older adults. J Cereb Blood Flow Metab 2020; 40:1859-1868. [PMID: 31506011 PMCID: PMC7446562 DOI: 10.1177/0271678x19874770] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In vivo dopamine D2-receptor availability is frequently assessed with 11C-raclopride and positron emission tomography. Due to low signal-to-noise ratios for 11C-raclopride in areas with low D2 receptor densities, the ligand has been considered unreliable for measurements outside the dopamine-dense striatum. Intriguingly, recent studies show that extrastriatal 11C-raclopride binding potential (BPND) values are (i) reliably higher than in the cerebellum (where D2-receptor levels are negligible), (ii) correlate with behavior in the expected direction, and (iii) showed good test-retest reliability in a sample of younger adults. The present work demonstrates high seven-month test-retest reliability of striatal and extrastriatal 11C-raclopride BPND values in healthy, older adults (n = 27, age: 64-78 years). Mean 11C-raclopride BPND values were stable between test sessions in subcortical nuclei, and in frontal and temporal cortices (p > 0.05). Across all structures analyzed, intraclass correlation coefficients were high (0.85-0.96), absolute variability was low (mean: 4-8%), and coefficients of variance ranged between 9 and 25%. Furthermore, regional 11C-raclopride BPND values correlated with previously determined 18F-fallypride BPND values (ρ = 0.97 and 0.92 in correlations with and without striatal values, respectively, p < 0.01) and postmortem determined D2-receptor densities (including striatum: ρ = 0.92; p < 0.001; excluding striatum: ρ = 0.75; p = 0.067). These observations suggest that extrastriatal 11C-raclopride measurements represent a true D2 signal.
Collapse
Affiliation(s)
- Nina Karalija
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Lars Jonassson
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jarkko Johansson
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Goran Papenberg
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Alireza Salami
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden.,Wallenberg Centre for Molecular Medicine, Lund, Sweden
| | - Micael Andersson
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Katrine Riklund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Lars Nyberg
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Carl-Johan Boraxbekk
- Department of Radiation Sciences, Umeå University, Umeå, Sweden.,Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.,Danish Research Center for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark
| |
Collapse
|
188
|
Nørgaard M, Ganz M, Svarer C, Frokjaer VG, Greve DN, Strother SC, Knudsen GM. Different preprocessing strategies lead to different conclusions: A [ 11C]DASB-PET reproducibility study. J Cereb Blood Flow Metab 2020; 40:1902-1911. [PMID: 31575336 PMCID: PMC7446563 DOI: 10.1177/0271678x19880450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Positron emission tomography (PET) neuroimaging provides unique possibilities to study biological processes in vivo under basal and interventional conditions. For quantification of PET data, researchers commonly apply different arrays of sequential data analytic methods ("preprocessing pipeline"), but it is often unknown how the choice of preprocessing affects the final outcome. Here, we use an available data set from a double-blind, randomized, placebo-controlled [11C]DASB-PET study as a case to evaluate how the choice of preprocessing affects the outcome of the study. We tested the impact of 384 commonly used preprocessing strategies on a previously reported positive association between the change from baseline in neocortical serotonin transporter binding determined with [11C]DASB-PET, and change in depressive symptoms, following a pharmacological sex hormone manipulation intervention in 30 women. The two preprocessing steps that were most critical for the outcome were motion correction and kinetic modeling of the dynamic PET data. We found that 36% of the applied preprocessing strategies replicated the originally reported finding (p < 0.05). For preprocessing strategies with motion correction, the replication percentage was 72%, whereas it was 0% for strategies without motion correction. In conclusion, the choice of preprocessing strategy can have a major impact on a study outcome.
Collapse
Affiliation(s)
- Martin Nørgaard
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Melanie Ganz
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Claus Svarer
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Vibe G Frokjaer
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Douglas N Greve
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen C Strother
- Rotman Research Institute, Baycrest, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
189
|
Sanchez-Catasus CA, Bohnen NI, Yeh FC, D'Cruz N, Kanel P, Müller MLTM. Dopaminergic Nigrostriatal Connectivity in Early Parkinson Disease: In Vivo Neuroimaging Study of 11C-DTBZ PET Combined with Correlational Tractography. J Nucl Med 2020; 62:545-552. [PMID: 32859707 DOI: 10.2967/jnumed.120.248500] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/25/2020] [Indexed: 12/11/2022] Open
Abstract
Previous histopathologic and animal studies have shown axonal impairment and loss of connectivity of the nigrostriatal pathway in Parkinson disease (PD). However, there are conflicting reports from in vivo human studies. 11C-dihydrotetrabenazine (11C-DTBZ) is a vesicular monoamine type 2 transporter PET ligand that allows assessment of nigrostriatal presynaptic dopaminergic terminal integrity. Correlational tractography based on diffusion MRI can incorporate ligand-specific information provided by 11C-DTBZ PET into the fiber-tracking process. The purpose of this study was to assess the in vivo association between the integrity of the nigrostriatal tract (defined by correlational tractography) and the degree of striatal dopaminergic denervation based on 11C-DTBZ PET. Methods: The study involved 30 subjects with mild to moderate PD (23 men and 7 women; mean age, 66 ± 6.2 y; disease duration, 6.4 ± 4.0 y; Hoehn and Yahr stage, 2.1 ± 0.6; Movement Disorder Society [MDS]-revised Unified Parkinson Disease Rating Scale [UPDRS] [I-III] total score, 43.4 ± 17.8) and 30 control subjects (18 men and 12 women; mean age, 62 ± 10.3 y). 11C-DTBZ PET was performed using standard synthesis and acquisition protocols. Correlational tractography was performed to assess quantitative anisotropy (QA; a measure of tract integrity) of white matter fibers correlating with information derived from striatal 11C-DTBZ data using the DSI Studio toolbox. Scans were realigned according to least and most clinically affected cerebral hemispheres. Results: Nigrostriatal tracts were identified in both hemispheres of PD patients. Higher mean QA values along the identified tracts were significantly associated with higher striatal 11C-DTBZ distribution volume ratios (least affected: r = 0.57, P = 0.001; most affected: r = 0.44, P = 0.02). Lower mean QA values of the identified tract in the LA hemisphere associated with increased severity of bradykinesia sub-score derived from MDS-UPDRS part III (r = -0.42; P = 0.02). Cross-validation revealed the generalizability of these results. Conclusion: These findings suggest that impaired integrity of dopaminergic nigrostriatal nerve terminals is associated with nigrostriatal axonal dysfunction in mild to moderate PD. Assessment of nigrostriatal tract integrity may be suitable as a biomarker of early- or even prodromal-stage PD.
Collapse
Affiliation(s)
- Carlos A Sanchez-Catasus
- Division of Nuclear Medicine, Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan.,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan
| | - Nicolaas I Bohnen
- Division of Nuclear Medicine, Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan.,Department of Neurology, University of Michigan Health System, Ann Arbor, Michigan.,Neurology Service and GRECC, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Nicholas D'Cruz
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| | - Prabesh Kanel
- Division of Nuclear Medicine, Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan
| | - Martijn L T M Müller
- Division of Nuclear Medicine, Department of Radiology, University of Michigan Health System, Ann Arbor, Michigan .,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
190
|
Jonasson M, Nordeman P, Eriksson J, Wilking H, Wikström J, Takahashi K, Niwa T, Hosoya T, Watanabe Y, Antoni G, Sundström Poromaa I, Lubberink M, Comasco E. Quantification of aromatase binding in the female human brain using [
11
C]cetrozole positron emission tomography. J Neurosci Res 2020; 98:2208-2218. [DOI: 10.1002/jnr.24707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022]
Affiliation(s)
- My Jonasson
- Department of Surgical Sciences Uppsala University Uppsala Sweden
- Department of Medical Physics Uppsala University Hospital Uppsala Sweden
| | - Patrik Nordeman
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | | | - Johan Wikström
- Department of Surgical Sciences Uppsala University Uppsala Sweden
| | - Kayo Takahashi
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
| | - Takashi Niwa
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
| | - Takamitsu Hosoya
- RIKEN Center for Biosystems Dynamics Research Kobe Japan
- Institute of Biomaterials and Bioengineering Tokyo Medical and Dental University Tokyo Japan
| | | | - Gunnar Antoni
- Department of Medicinal Chemistry Uppsala University Uppsala Sweden
- PET centre Uppsala University Hospital Uppsala Sweden
| | | | - Mark Lubberink
- Department of Surgical Sciences Uppsala University Uppsala Sweden
- Department of Medical Physics Uppsala University Hospital Uppsala Sweden
| | - Erika Comasco
- Department of Neuroscience Science for Life Laboratory Uppsala University Uppsala Sweden
| |
Collapse
|
191
|
Abstract
Neuroimaging with positron emission tomography (PET) is the most powerful tool for understanding pharmacology, neurochemistry, and pathology in the living human brain. This technology combines high-resolution scanners to measure radioactivity throughout the human body with specific, targeted radioactive molecules, which allow measurements of a myriad of biological processes in vivo. While PET brain imaging has been active for almost 40 years, the pace of development for neuroimaging tools, known as radiotracers, and for quantitative analytical techniques has increased dramatically over the past decade. Accordingly, the fundamental questions that can be addressed with PET have expanded in basic neurobiology, psychiatry, neurology, and related therapeutic development. In this review, we introduce the field of human PET neuroimaging, some of its conceptual underpinnings, and motivating questions. We highlight some of the more recent advances in radiotracer development, quantitative modeling, and applications of PET to the study of the human brain.
Collapse
Affiliation(s)
- Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA;
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
192
|
Tjerkaski J, Cervenka S, Farde L, Matheson GJ. Kinfitr - an open-source tool for reproducible PET modelling: validation and evaluation of test-retest reliability. EJNMMI Res 2020; 10:77. [PMID: 32642865 PMCID: PMC7343683 DOI: 10.1186/s13550-020-00664-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/25/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In positron emission tomography (PET) imaging, binding is typically estimated by fitting pharmacokinetic models to the series of measurements of radioactivity in the target tissue following intravenous injection of a radioligand. However, there are multiple different models to choose from and numerous analytical decisions that must be made when modelling PET data. Therefore, it is important that analysis tools be adapted to the specific circumstances, and that analyses be documented in a transparent manner. Kinfitr, written in the open-source programming language R, is a tool developed for flexible and reproducible kinetic modelling of PET data, i.e. performing all steps using code which can be publicly shared in analysis notebooks. In this study, we compared outcomes obtained using kinfitr with those obtained using PMOD: a widely used commercial tool. RESULTS Using previously collected test-retest data obtained with four different radioligands, a total of six different kinetic models were fitted to time-activity curves derived from different brain regions. We observed good correspondence between the two kinetic modelling tools both for binding estimates and for microparameters. Likewise, no substantial differences were observed in the test-retest reliability estimates between the two tools. CONCLUSIONS In summary, we showed excellent agreement between the open-source R package kinfitr, and the widely used commercial application PMOD. We, therefore, conclude that kinfitr is a valid and reliable tool for kinetic modelling of PET data.
Collapse
Affiliation(s)
- Jonathan Tjerkaski
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden.
| | - Simon Cervenka
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Lars Farde
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| | - Granville James Matheson
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76, Stockholm, Sweden
| |
Collapse
|
193
|
DeCarli C, Villeneuve S, Maillard P, Harvey D, Singh B, Carmichael O, Fletcher E, Olichney J, Farias S, Jagust W, Reed B, Mungas D. Vascular Burden Score Impacts Cognition Independent of Amyloid PET and MRI Measures of Alzheimer's Disease and Vascular Brain Injury. J Alzheimers Dis 2020; 68:187-196. [PMID: 30775991 DOI: 10.3233/jad-180965] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND/OBJECTIVE To determine the impact of vascular burden on rates of decline in episodic memory and executive function. We hypothesize that greater vascular burden will have an additive negative impact on cognition after accounting for baseline cognitive impairment, positron emission tomography (PET) amyloid burden, and magnetic resonance imaging (MRI) measures. METHODS Individuals were followed an average of 5 years with serial cognitive assessments. Predictor variables include vascular burden score (VBS), quantitative brain MRI assessment, and amyloid imaging. Subjects consisted of 65 individuals, 53% of whom were male, aged 73.2±7.2 years on average with an average of 15.5±3.3 years of educational achievement. RESULTS Baseline cognitive impairment was significantly associated poorer episodic memory (p < 0.0001), smaller hippocampal volume (p < 0.0001), smaller brain volume (p = 0.0026), and greater global Pittsburg Imaging Compound B (PiB) index (p = 0.0008). Greater amyloid burden was associated with greater decline in episodic memory over time (β= -0.20±0.07, p < 0.005). VBS was significantly associated with the level of executive function performance (β= -0.14±0.05, p < 0.005) and there was a significant negative interaction between VBS, cognitive impairment, and PiB index (β= -0.065±0.03, p = 0.03). CONCLUSIONS Our results find a significant influence of VBS independent of standard MRI measures and cerebral amyloid burden on executive function. In addition, VBS reduced the amount of cerebral amyloid burden needed to result in cognitive impairment. We conclude that the systemic effects of vascular disease as reflected by the VBS independently influence cognitive ability.
Collapse
Affiliation(s)
- Charles DeCarli
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Pauline Maillard
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Danielle Harvey
- Division of Biostatistics, School of Medicine, University of California at Davis, Davis, CA, USA
| | - Baljeet Singh
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Owen Carmichael
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Evan Fletcher
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - John Olichney
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - Sarah Farias
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley CA, USA
| | - Bruce Reed
- Center for Scientific Review, Division of Neuroscience, Development and Aging, NIH, Bethesda, MD, USA
| | - Dan Mungas
- UC Davis Department of Neurology and Center for Neuroscience, Davis, CA, USA
| |
Collapse
|
194
|
Schmidt ME, Janssens L, Moechars D, Rombouts FJR, Timmers M, Barret O, Constantinescu CC, Madonia J, Russell DS, Sandiego CM, Kolb H. Clinical evaluation of [ 18F] JNJ-64326067, a novel candidate PET tracer for the detection of tau pathology in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2020; 47:3176-3185. [PMID: 32535652 PMCID: PMC7680304 DOI: 10.1007/s00259-020-04880-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/19/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The accumulation of misfolded tau is a common feature of several neurodegenerative disorders, with Alzheimer's disease (AD) being the most common. Earlier we identified JNJ-64326067, a novel isoquinoline derivative with high affinity and selectivity for tau aggregates from human AD brain. We report the dosimetry of [18F] JNJ-64326067 and results of a proof-of-concept study comparing subjects with probable Alzheimer's disease to age-matched healthy controls. METHODS [18F] JNJ-64326067 PET scans were acquired for 90 min and then from 120 to 180 min in 5 participants with [18F]-florbetapir PET amyloid positive probable AD (73 ± 9 years) and 5 [18F]-florbetapir PET amyloid negative healthy controls (71 ± 7 years). Whole-body [18F] JNJ-64326067 PET CT scans were acquired in six healthy subjects for 5.5 h in 3 scanning sessions. Brain PET scans were visually reviewed. Regional quantification included kinetic analysis of distribution volume ration (DVR) estimated by Logan graphical analysis over the entire scan and static analysis of SUVr in late frames. Both methods used ventral cerebellar cortex as a reference region. RESULTS One of the healthy controls had focal areas of PET signal in occipital and parietal cortex underlying the site of a gunshot injury as an adolescent; the other four healthy subjects had no tau brain signal. Four of the 5 AD participants had visually apparent retention of [18F] JNJ-64326067 in relevant cortical regions. One of the AD subjects was visually negative. Cortical signal in visually positive subjects approached steady state by 120 min. Temporal and frontal cortical SUVr/DVR values in visually positive AD subjects ranged from 1.21 to 3.09/1.2 to 2.18 and from 0.92 to 1.28/0.91 to 1.16 in healthy controls. Whole-body effective dose was estimated to be 0.0257 mSv/MBq for females and 0.0254 mSv/MBq for males. CONCLUSIONS [18F] JNJ-64326067 could be useful for detection and quantitation of tau aggregates.
Collapse
Affiliation(s)
- Mark E Schmidt
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium.
| | - Luc Janssens
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Diederik Moechars
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | | | - Maarten Timmers
- Janssen Research & Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Olivier Barret
- Invicro, a Konica Minolta company, New Haven, CT, USA.,Laboratory of Neurodegenerative Diseases, Molecular Imaging Research Center, French Atomic Energy Commission, Fontenay-aux-roses, France
| | | | - Jennifer Madonia
- Invicro, a Konica Minolta company, New Haven, CT, USA.,Biohaven Pharmaceuticals, New Haven, Connecticut, USA
| | | | | | | |
Collapse
|
195
|
Cselényi Z, Jucaite A, Kristensson C, Stenkrona P, Ewing P, Varrone A, Johnström P, Schou M, Vazquez-Romero A, Moein MM, Bolin M, Siikanen J, Grybäck P, Larsson B, Halldin C, Grime K, Eriksson UG, Farde L. Quantification and reliability of [ 11C]VC - 002 binding to muscarinic acetylcholine receptors in the human lung - a test-retest PET study in control subjects. EJNMMI Res 2020; 10:59. [PMID: 32495011 PMCID: PMC7270393 DOI: 10.1186/s13550-020-00634-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/22/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The radioligand [11C]VC-002 was introduced in a small initial study long ago for imaging of muscarinic acetylcholine receptors (mAChRs) in human lungs using positron emission tomography (PET). The objectives of the present study in control subjects were to advance the methodology for quantification of [11C]VC-002 binding in lung and to examine the reliability using a test-retest paradigm. This work constituted a self-standing preparatory step in a larger clinical trial aiming at estimating mAChR occupancy in the human lungs following inhalation of mAChR antagonists. METHODS PET measurements using [11C]VC-002 and the GE Discovery 710 PET/CT system were performed in seven control subjects at two separate occasions, 2-19 days apart. One subject discontinued the study after the first measurement. Radioligand binding to mAChRs in lung was quantified using an image-derived arterial input function. The total distribution volume (VT) values were obtained on a regional and voxel-by-voxel basis. Kinetic one-tissue and two-tissue compartment models (1TCM, 2TCM), analysis based on linearization of the compartment models (multilinear Logan) and image analysis by data-driven estimation of parametric images based on compartmental theory (DEPICT) were applied. The test-retest repeatability of VT estimates was evaluated by absolute variability (VAR) and intraclass correlation coefficients (ICCs). RESULTS The 1TCM was the statistically preferred model for description of [11C]VC-002 binding in the lungs. Low VAR (< 10%) across analysis methods indicated good reliability of the PET measurements. The VT estimates were stable after 60 min. CONCLUSIONS The kinetic behaviour and good repeatability of [11C]VC-002 as well as the novel lung image analysis methodology support its application in applied studies on drug-induced mAChR receptor occupancy and the pathophysiology of pulmonary disorders. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03097380, registered: 31 March 2017.
Collapse
Affiliation(s)
- Zsolt Cselényi
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden.
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.
| | - Aurelija Jucaite
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | | | - Per Stenkrona
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Pär Ewing
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Peter Johnström
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Magnus Schou
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Ana Vazquez-Romero
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Mohammad Mahdi Moein
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Martin Bolin
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jonathan Siikanen
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Pär Grybäck
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Bengt Larsson
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Ken Grime
- BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | | | - Lars Farde
- PET Science Centre, Precision Medicine, R&D, AstraZeneca, Stockholm, Sweden
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
196
|
Mitelman SA, Buchsbaum MS, Christian BT, Merrill BM, Buchsbaum BR, Mukherjee J, Lehrer DS. Positive association between cerebral grey matter metabolism and dopamine D 2/D 3 receptor availability in healthy and schizophrenia subjects: An 18F-fluorodeoxyglucose and 18F-fallypride positron emission tomography study. World J Biol Psychiatry 2020; 21:368-382. [PMID: 31552783 DOI: 10.1080/15622975.2019.1671609] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objectives: Overlapping decreases in extrastriatal dopamine D2/D3-receptor availability and glucose metabolism have been reported in subjects with schizophrenia. It remains unknown whether these findings are physiologically related or coincidental.Methods: To ascertain this, we used two consecutive 18F-fluorodeoxyglucose and 18F-fallypride positron emission tomography scans in 19 healthy and 25 unmedicated schizophrenia subjects. Matrices of correlations between 18F-fluorodeoxyglucose uptake and 18F-fallypride binding in voxels at the same xyz location and AFNI-generated regions of interest were evaluated in both diagnostic groups.Results:18F-fluorodeoxyglucose uptake and 18F-fallypride binding potential were predominantly positively correlated across the striatal and extrastriatal grey matter in both healthy and schizophrenia subjects. In comparison to healthy subjects, significantly weaker correlations in subjects with schizophrenia were confirmed in the right cingulate gyrus and thalamus, including the mediodorsal, lateral dorsal, anterior, and midline nuclei. Schizophrenia subjects showed decreased D2/D3-receptor availability in the hypothalamus, mamillary bodies, thalamus and several thalamic nuclei, and increased glucose uptake in three lobules of the cerebellar vermis.Conclusions: Dopaminergic system may be involved in modulation of grey matter metabolism and neurometabolic coupling in both healthy human brain and psychopathology. Hyperdopaminergic state in untreated schizophrenia may at least partly account for the corresponding decreases in grey matter metabolism.
Collapse
Affiliation(s)
- Serge A Mitelman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City,NY, USA.,Department of Psychiatry, Division of Child and Adolescent Psychiatry, Elmhurst Hospital Center, Elmhurst, IL, USA
| | - Monte S Buchsbaum
- Departments of Psychiatry and Radiology, University of California, San Diego, CA, USA.,Department of Psychiatry and Human Behavior, University of California, Irvine School of Medicine, Orange, CA, USA
| | - Bradley T Christian
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian M Merrill
- Department of Psychiatry, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Bradley R Buchsbaum
- The Rotman Research Institute, Baycrest Centre for Geriatric Care and Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Jogeshwar Mukherjee
- Department of Radiological Sciences, Preclinical Imaging, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Douglas S Lehrer
- Department of Psychiatry, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| |
Collapse
|
197
|
Arakawa R, Takano A, Halldin C. PET technology for drug development in psychiatry. Neuropsychopharmacol Rep 2020; 40:114-121. [PMID: 32463584 PMCID: PMC7722687 DOI: 10.1002/npr2.12084] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/05/2019] [Accepted: 10/18/2019] [Indexed: 12/14/2022] Open
Abstract
Positron emission tomography (PET) is a non‐invasive imaging method to measure the molecule in vivo. PET imaging can evaluate the central nervous system drugs as target engagement in the human brain. For antipsychotic drugs, adequate dopamine D2 receptor occupancy (“therapeutic window”) is reported to be from 65%‐70% to 80% to achieve the antipsychotic effect without extrapyramidal symptoms. For antidepressants, the clinical threshold of serotonin transporter (5‐HTT) occupancy is reported to be 70%‐80% although the relation between the side effect and 5‐HTT occupancy has not yet been established. Evaluation of norepinephrine transporter (NET) occupancy for antidepressant is ongoing as adequate PET radioligands for NET were developed recently. Measurement of the target occupancy has been a key element to evaluate the in vivo target engagement of the drugs. In order to evaluate new drug targets for disease conditions such as negative symptoms/cognitive impairment of schizophrenia and treatment‐resistant depression, new PET radioligands need to be developed concurrently with the drug development. PET imaging can evaluate the central nervous system drugs as target engagement in the human brain. The uptake of [11C]raclopride for dopamine D2 receptors decreased from (A) baseline to (B) antipsychotic administration conditions.![]()
Collapse
Affiliation(s)
- Ryosuke Arakawa
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Akihiro Takano
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.,Takeda Development Center Japan, Takeda Pharmaceutical Company Limited, Osaka, Japan
| | - Christer Halldin
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
198
|
Weng CC, Hsiao IT, Yang QF, Yao CH, Tai CY, Wu MF, Yen TC, Jang MK, Lin KJ. Characterization of 18F-PM-PBB3 ( 18F-APN-1607) Uptake in the rTg4510 Mouse Model of Tauopathy. Molecules 2020; 25:molecules25071750. [PMID: 32290239 PMCID: PMC7181044 DOI: 10.3390/molecules25071750] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/03/2022] Open
Abstract
Misfolding, aggregation, and cerebral accumulation of tau deposits are hallmark features of Alzheimer’s disease. Positron emission tomography study of tau can facilitate the development of anti-tau treatment. Here, we investigated a novel tau tracer 18F-PM-PBB3 (18F-APN-1607) in a mouse model of tauopathy. Dynamic PET scans were collected in groups of rTg4510 transgenic mice at 2–11 months of age. Associations between distribution volume ratios (DVR) and standardized uptake value ratios (SUVR) with cerebellum reference were used to determine the optimal scanning time and uptake pattern for each age. Immunohistochemistry staining of neurofibrillary tangles and autoradiography study was performed for ex vivo validation. An SUVR 40–70 min was most consistently correlated with DVR and was used in further analyses. Significant increased 18F-PM-PBB3 uptake in the brain cortex was found in six-month-old mice (+28.9%, p < 0.05), and increased further in the nine-month-old group (+38.8%, p < 0.01). The trend of increased SUVR value remained evident in the hippocampus and striatum regions except for cortex where uptake becomes slightly reduced in 11-month-old animals (+37.3%, p < 0.05). Radioactivity distributions from autoradiography correlate well to the presence of human tau (HT7 antibody) and hyperphosphorylated tau (antibody AT8) from the immunohistochemistry study of the adjacent brain sections. These findings supported that the 40–70 min 18F-PM-PBB3 PET scan with SUVR measurement can detect significantly increased tau deposits in a living rTg4510 transgenic mouse models as early as six-months-old. The result exhibited promising dynamic imaging capability of this novel tau tracer, and the above image characteristics should be considered in the design of longitudinal preclinical tau image studies.
Collapse
Affiliation(s)
- Chi-Chang Weng
- HARC and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 333, Taiwan; (C.C.-W.); (I.-T.H.); (Q.-F.Y.)
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ing-Tsung Hsiao
- HARC and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 333, Taiwan; (C.C.-W.); (I.-T.H.); (Q.-F.Y.)
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Qing-Fang Yang
- HARC and Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 333, Taiwan; (C.C.-W.); (I.-T.H.); (Q.-F.Y.)
| | - Cheng-Hsiang Yao
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chin-Yin Tai
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Meng-Fang Wu
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Tzu-Chen Yen
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Ming-Kuei Jang
- APRINOIA Therapeutics Inc., Taipei 11503, Taiwan; (C.-Y.T.); (M.-F.W.); (T.-C.Y.); (M.-K.J.)
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Center for Advanced Molecular Imaging and Translation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Correspondence:
| |
Collapse
|
199
|
Waninger S, Berka C, Stevanovic Karic M, Korszen S, Mozley PD, Henchcliffe C, Kang Y, Hesterman J, Mangoubi T, Verma A. Neurophysiological Biomarkers of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2020; 10:471-480. [PMID: 32116262 PMCID: PMC7242849 DOI: 10.3233/jpd-191844] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND There is a need for reliable and robust Parkinson's disease biomarkers that reflect severity and are sensitive to disease modifying investigational therapeutics. OBJECTIVE To demonstrate the utility of EEG as a reliable, quantitative biomarker with potential as a pharmacodynamic endpoint for use in clinical assessments of neuroprotective therapeutics for Parkison's disease. METHODS A multi modal study was performed including aquisition of resting state EEG data and dopamine transporter PET imaging from Parkinson's disease patients off medication and compared against age-matched controls. RESULTS Qualitative and test/retest analysis of the EEG data demonstrated the reliability of the methods. Source localization using low resolution brain electromagnetic tomography identified significant differences in Parkinson's patients versus control subjects in the anterior cingulate and temporal lobe, areas with established association to Parkinson's disease pathology. Changes in cortico-cortical and cortico-thalamic coupling were observed as excessive EEG beta coherence in Parkinson's disease patients, and correlated with UPDRS scores and dopamine transporter activity, supporting the potential for cortical EEG coherence to serve as a reliable measure of disease severity. Using machine learning approaches, an EEG discriminant function analysis classifier was identified that parallels the loss of dopamine synapses as measured by dopamine transporter PET. CONCLUSION Our results support the utility of EEG in characterizing alterations in neurophysiological oscillatory activity associated with Parkinson's disease and highlight potential as a reliable method for monitoring disease progression and as a pharmacodynamic endpoint for Parkinson's disease modification therapy.
Collapse
Affiliation(s)
- Shani Waninger
- Advanced Brain Monitoring Inc., Carlsbad, CA, USA,Correspondence to: Shani Waninger, Advanced Brain Monitoring, Inc., 2237 Faraday Avenue, Suite 100,
Carlsbad, CA 92008, USA. E-mail:
| | - Chris Berka
- Advanced Brain Monitoring Inc., Carlsbad, CA, USA
| | | | | | | | | | - Yeona Kang
- Weill Cornell Medical College, New York, NY, USA
| | | | | | | |
Collapse
|
200
|
Koopman T, Verburg N, Pouwels PJ, Wesseling P, Hoekstra OS, De Witt Hamer PC, Lammertsma AA, Yaqub M, Boellaard R. Quantitative parametric maps of O-(2-[ 18F]fluoroethyl)-L-tyrosine kinetics in diffuse glioma. J Cereb Blood Flow Metab 2020; 40:895-903. [PMID: 31122112 PMCID: PMC7074601 DOI: 10.1177/0271678x19851878] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Quantitative parametric images of O-(2-[18F]fluoroethyl)-L-tyrosine kinetics in diffuse gliomas could be used to improve glioma grading, tumour delineation or the assessment of the uptake distribution of this positron emission tomography tracer. In this study, several parametric images and tumour-to-normal maps were compared in terms of accuracy of region averages (when compared to results from nonlinear regression of a reversible two-tissue compartment plasma input model) and image noise using 90 min of dynamic scan data acquired in seven patients with diffuse glioma. We included plasma input methods (the basis function implementation of the single-tissue compartment model, spectral analysis and Logan graphical analysis) and reference tissue methods (basis function implementations of the simplified reference tissue model, variations of the multilinear reference tissue model and non-invasive Logan graphical analysis) as well as tumour-to-normal ratio maps at three intervals. (Non-invasive) Logan graphical analysis provided volume of distribution maps and distribution volume ratio maps with the lowest level of noise, while the basis function implementations provided the best accuracy. Tumour-to-normal ratio maps provided better results if later interval times were used, i.e. 60-90 min instead of 20-40 min, leading to lower bias (2.9% vs. 10.8%, respectively) and less noise (12.8% vs. 14.4%).
Collapse
Affiliation(s)
- Thomas Koopman
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Niels Verburg
- Neurosurgical Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Petra Jw Pouwels
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Otto S Hoekstra
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philip C De Witt Hamer
- Neurosurgical Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Brain Tumor Center Amsterdam, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|