1
|
Nandi A, Nakano M, Brašić JR, Brinson ZS, Kitzmiller K, Mathur A, Mohamed M, Roberts J, Wong DF, Kuwabara H. Improved Quantification of MicroPET/CT Imaging Using CT-derived Scaling Factors. Mol Imaging Biol 2024; 26:1016-1026. [PMID: 39313673 DOI: 10.1007/s11307-024-01947-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE Combined micro-PET/CT scanners are widely employed to investigate models of brain disorders in rodents using PET-based coregistration. We examined if CT-based coregistration could improve estimates of brain dimensions and consequently estimates of nondisplaceable binding potential (BPND) in rodent PET studies. PROCEDURES PET and CT scans were acquired on 5 female and 5 male CD-1 mice with 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB), a radiotracer for the metabotropic glutamate receptor subtype 5 (mGluR5). In the proposed PET/CT (PTCT) approach, the tracer-specific standard volume was dimension-customized to each animal using the scaling factors from CT-to-standard CT coregistration to simplify PET-to-standard PET coregistration (i.e., 3 CT- and 6 PET-derived parameters). For comparison, conventional PET-based coregistration was performed with 9 (PT9) or 12 (PT12) parameters. PET frames were transferred to the standard space by the three approaches (PTCT, PT9, and PT12) to obtain regional time-activity curves (TACs) and BPND in 14 standard volumes of interest (VOIs). Lastly, CT images of the animals were transferred to the standard space by CT-based parameters from PTCT and with the scaling factors replaced with those from PET-based PT9 to evaluate agreement of the skull to the standard CT. RESULTS The PET-based approaches showed various degrees of underestimations of scaling factors in the posterior-anterior-direction compared to PTCT, which resulted in negatively proportional overestimation of radioactivity in the cerebellum (reference region) up to 20%, and proportional, more prominent underestimation of BPND in target regions down to -50%. The skulls of individual animals agreed with the standard skull for scaling factors from PTCT but not for the scaling factors from PT9, which suggested inaccuracy of the latter. CONCLUSIONS The results indicated that conventional PET-based coregistration approaches could yield biased estimates of BPND in proportion to errors of brain dimensions when applied to tracers for which the cerebellum serves as reference region. The proposed PTCT provides evidence of a quantitative improvement over PET-based approaches for brain studies using micro-PET/CT scanners.
Collapse
Affiliation(s)
- Ayon Nandi
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
| | - Masayoshi Nakano
- Clinical Science Division, R&D, Janssen Pharmaceutical K.K, Tokyo, Japan
| | - James Robert Brašić
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
- Department of Psychiatry, New York City Health + Hospitals/Bellevue, New York, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Zabecca S Brinson
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kelly Kitzmiller
- Radiation Oncology, University of Maryland Medical Center, Baltimore, MD, USA
| | - Anil Mathur
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
| | | | - Joshua Roberts
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dean F Wong
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA
- Lab of CNS Neuropsychopharmacology And Multimodal Imaging (CNAMI), Mallinckrodt Institute of Radiology, Washington University in St Louis, St Louis, MO, USA
| | - Hiroto Kuwabara
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, JHOC Room 3243, 601 N. Caroline St, Baltimore, 21287, MD, USA.
| |
Collapse
|
2
|
Kikuchi T, Okamura T, Zhang MR. Numerical simulation method for the assessment of the effect of molar activity on the pharmacokinetics of radioligands in small animals. EJNMMI Radiopharm Chem 2024; 9:78. [PMID: 39570519 PMCID: PMC11582259 DOI: 10.1186/s41181-024-00308-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND It is well recognized that the molar activity of a radioligand is an important pharmacokinetic parameter, especially in positron emission tomography (PET) of small animals. Occupation of a significant number of binding sites by radioligand molecules results in low radioligand accumulation in a target region (mass effect). Nevertheless, small-animal PET studies have often been performed without consideration of the molar activity or molar dose of radioligands. A simulation study would therefore help to assess the importance of the mass effect in small-animal PET. Here, we introduce a new compartmental model-based numerical method, which runs on commonly used spreadsheet software, to simulate the effect of molar activity or molar dose on the pharmacokinetics of radioligands. RESULTS Assuming a two-tissue compartmental model, time-concentration curves of a radioligand were generated using four simulation methods and the well-known Runge-Kutta numerical method. The values were compared with theoretical values obtained under an ultra-high molar activity condition (pseudo-first-order binding kinetics), a steady-state condition and an equilibrium condition (second-order binding kinetics). For all conditions, the simulation method using the simplest calculation yielded values closest to the theoretical values and comparable with those obtained using the Runge-Kutta method. To satisfy a maximum occupancy less than 5%, simulations showed that a molar activity greater than 150 GBq/μmol is required for a model radioligand when 20 MBq is administered to a 250 g rat and when the concentration of binding sites in target regions is greater than 1.25 nM. CONCLUSIONS The simulation method used in this study is based on a very simple calculation and runs on widely used spreadsheet software. Therefore, simulation of radioligand pharmacokinetics using this method can be performed on a personal computer and help to assess the importance of the mass effect in small-animal PET. This simulation method also enables the generation of a model time-activity curve for the evaluation of kinetic analysis methods.
Collapse
Affiliation(s)
- Tatsuya Kikuchi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | - Toshimitsu Okamura
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
3
|
Fayez H, Selim A, Shamma R, Rashed H. Intranasal Radioiodinated Ferulic Acid Polymeric Micelles as the First Nuclear Medicine Imaging Probe for ETRA Brain Receptor. Curr Radiopharm 2024; 17:209-217. [PMID: 38213167 DOI: 10.2174/0118744710269885231113070356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/17/2023] [Accepted: 09/27/2023] [Indexed: 01/13/2024]
Abstract
INTRODUCTION The aim of this work was to prepare a selective nuclear medicine imaging probe for the Endothelin 1 receptor A in the brain. MATERIAL AND METHODS Ferulic acid (an ETRA antagonist) was radiolabeled using 131I by direct electrophilic substitution method. The radiolabeled ferulic acid was formulated as polymeric micelles to allow intranasal brain delivery. Biodistribution was studied in Swiss albino mice by comparing brain uptake of 131I-ferulic acid after IN administration of 131I-ferulic acid polymeric micelles, IN administration of 131I-ferulic acid solution and IV administration of 131I-ferulic acid solution. RESULTS Successful radiolabeling was achieved with an RCY of 98 % using 200 μg of ferulic acid and 60 μg of CAT as oxidizing agents at pH 6, room temperature and 30 min reaction time. 131I-ferulic acid polymeric micelles were successfully formulated with the particle size of 21.63 nm and polydispersity index of 0.168. Radioactivity uptake in the brain and brain/blood uptake ratio for I.N 131I-ferulic acid polymeric micelles were greater than the two other routes at all periods. CONCLUSION Our results provide 131I-ferulic acid polymeric micelles as a hopeful nuclear medicine tracer for ETRA brain receptor.
Collapse
Affiliation(s)
- Hend Fayez
- Department of Labeled Compounds, Hot Laboratories Center, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Adli Selim
- Department of Labeled Compounds, Hot Laboratories Center, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Rehab Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11561, Egypt
| | - Hassan Rashed
- Department of Labeled Compounds, Hot Laboratories Center, Egyptian Atomic Energy Authority, Cairo, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Sinai University, Kantara, Egypt
| |
Collapse
|
4
|
Nandi A, Nakano M, Brašić JR, Brinson ZS, Kitzmiller K, Mathur A, Mohamed M, Roberts J, Wong DF, Kuwabara H. Improved Quantification of MicroPET/CT Imaging Using CT-derived Scaling Factors. RESEARCH SQUARE 2023:rs.3.rs-3612275. [PMID: 38077018 PMCID: PMC10705595 DOI: 10.21203/rs.3.rs-3612275/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Purpose Combined micro-PET/CT scanners are widely employed to investigate models of brain disorders in rodents using PET-based coregistration. We examined if CT-based coregistration could improve estimates of brain dimensions and consequently estimates of nondisplaceable binding potential (BPND) in rodent PET studies. Procedures PET and CT scans were acquired on 5 female and 5 male CD-1 mice with PET and CT scans were acquired on 5 female and 5 male CD-1 mice with 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB), a radiotracer for the metabotropic glutamate receptor subtype 5 (mGluR5). In the proposed PET/CT (PTCT) approach, the tracer-specific standard volume was dimension-customized to each animal using the scaling factors from CT-to-standard CT coregistration to simplify PET-to-standard PET coregistration (i.e., 3 CT- and 6 PET-derived parameters). For comparison, conventional PET-based coregistration was performed with 9 (PT9) or 12 (PT12) parameters. PET frames were transferred to the standard space by the three approaches (PTCT, PT9, and PT12) to obtain regional time-activity curves (TACs) and BPND in 14 standard volumes of interest (VOIs). Lastly, CT images of the animals were transferred to the standard space by CT-based parameters from PTCT and with the scaling factors replaced with those from PET-based PT9 to evaluate agreement of the skull to the standard CT. Results The PET-based approaches showed various degrees of underestimations of scaling factors in the posterior-anterior-direction compared to PTCT, which resulted in negatively proportional overestimation of radioactivity in the cerebellum (reference region) up to 20%, and proportional, more prominent underestimation of BPND in target regions down to -50%. The skulls of individual animals agreed with the standard skull for scaling factors from PTCT but not for the scaling factors from PT9, which suggested inaccuracy of the latter. Conclusions The results indicated that conventional PET-based coregistration approaches could yield biased estimates of BPND due to erroneous estimates of brain dimensions when applied to tracers for which the cerebellum serves as reference region. The proposed PTCT provides evidence of a quantitative improvement over PET-based approaches for brain studies using micro-PET/CT scanners.
Collapse
Affiliation(s)
- Ayon Nandi
- Johns Hopkins School of Medicine: The Johns Hopkins University School of Medicine
| | | | | | | | | | - Anil Mathur
- Johns Hopkins School of Medicine: The Johns Hopkins University School of Medicine
| | | | - Joshua Roberts
- Johns Hopkins School of Medicine: The Johns Hopkins University School of Medicine
| | - Dean F Wong
- Washington University in St Louis School of Medicine Mallinckrodt Institute of Radiology
| | - Hiroto Kuwabara
- Johns Hopkins School of Medicine: The Johns Hopkins University School of Medicine
| |
Collapse
|
5
|
Skovbjerg G, Roostalu U, Salinas CG, Skytte JL, Perens J, Clemmensen C, Elster L, Frich CK, Hansen HH, Hecksher-Sørensen J. Uncovering CNS access of lipidated exendin-4 analogues by quantitative whole-brain 3D light sheet imaging. Neuropharmacology 2023:109637. [PMID: 37391028 DOI: 10.1016/j.neuropharm.2023.109637] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 07/02/2023]
Abstract
Peptide-based drug development for CNS disorders is challenged by poor blood-brain barrier (BBB) penetrability of peptides. While acylation protractions (lipidation) have been successfully applied to increase circulating half-life of therapeutic peptides, little is known about the CNS accessibility of lipidated peptide drugs. Light-sheet fluorescence microscopy (LSFM) has emerged as a powerful method to visualize whole-brain 3D distribution of fluorescently labelled therapeutic peptides at single-cell resolution. Here, we applied LSFM to map CNS distribution of the clinically relevant GLP-1 receptor agonist (GLP-1RA) exendin-4 (Ex4) and lipidated analogues following peripheral administration. Mice received an intravenous dose (100 nmol/kg) of IR800 fluorophore-labelled Ex4 (Ex4), Ex4 acylated with a C16-monoacid (Ex4_C16MA) or C18-diacid (Ex4_C18DA). Other mice were administered C16MA-acylated exendin 9-39 (Ex9-39_C16MA), a selective GLP-1R antagonist, serving as negative control for GLP-1R mediated agonist internalization. Two hours post-dosing, brain distribution of Ex4 and analogues was predominantly restricted to the circumventricular organs, notably area postrema and nucleus of the solitary tract. Ex4_C16MA and Ex9-39_C16MA also distributed to the paraventricular hypothalamic nucleus and medial habenula. Notably, Ex4_C18DA was detected in deeper-lying brain structures such as dorsomedial/ventromedial hypothalamic nuclei and the dentate gyrus. Similar CNS distribution maps of Ex4-C16MA and Ex9-39_C16MA suggest that brain access of lipidated Ex4 analogues is independent on GLP-1 receptor internalization. The cerebrovasculature was devoid of specific labelling, hence not supporting a direct role of GLP-1 RAs in BBB function. In conclusion, peptide lipidation increases CNS accessibility of Ex4. Our fully automated LSFM pipeline is suitable for mapping whole-brain distribution of fluorescently labelled drugs.
Collapse
Affiliation(s)
- Grethe Skovbjerg
- Gubra ApS, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Urmas Roostalu
- Gubra ApS, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | | | - Jacob L Skytte
- Gubra ApS, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | - Johanna Perens
- Gubra ApS, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Denmark
| | - Lisbeth Elster
- Gubra ApS, Hørsholm Kongevej 11B, 2970, Hørsholm, Denmark
| | | | | | | |
Collapse
|
6
|
Yu X, Gao Z, Gao M, Qiao M. Bibliometric Analysis on GABA-A Receptors Research Based on CiteSpace and VOSviewer. J Pain Res 2023; 16:2101-2114. [PMID: 37361426 PMCID: PMC10289248 DOI: 10.2147/jpr.s409380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/11/2023] [Indexed: 06/28/2023] Open
Abstract
Background GABA-A receptors are the primary mediators of brain inhibitory neurotransmission. In the past years, many studies focused on this channel to decipher the pathogenesis of related diseases but lacked bibliometric analysis research. This study aims to explore the research status and identify the research trends of GABA-A receptor channels. Methods Publications related to GABA-A receptor channels were retrieved from the Web of Science Core Collection from 2012 to 2022. After screening, the VOSviewer 1.6.18 and Citespace 5.8 R3 were used for bibliometric analysis from journals, countries, institutions, authors, co-cited references and keywords. Results We included 12,124 publications in the field of GABA-A receptor channels for analysis. The data shows that although there was a slight decrease in annual publications from 2012 to 2021, it remained at a relatively high level. Most publications were in the domain of neuroscience. Additionally, the United States was the most prolific country, followed by China. Univ Toronto was the most productive institution, and James M Cook led essential findings in this field. Furthermore, brain activation, GABAAR subunits expression, modulation mechanism in pain and anxiety behaviors and GABA and dopamine were paid attention to by researchers. And top research frontiers were molecular docking, autoimmune encephalitic series, obesity, sex difference, diagnosis and management, EEG and KCC2. Conclusion Taken together, academic attention on GABA-A receptor channels was never neglected since 2012. Our analysis identified key information, such as core countries, institutions and authors in this field. Molecular docking, autoimmune encephalitic series, obesity, sex difference, diagnosis and management, EEG and KCC2 will be the future research direction.
Collapse
Affiliation(s)
- Xufeng Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People’s Republic of China
| | - Zhan Gao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People’s Republic of China
| | - Mingzhou Gao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People’s Republic of China
| | - Mingqi Qiao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, People’s Republic of China
| |
Collapse
|
7
|
Wang X, Wang T, Fan X, Zhang Z, Wang Y, Li Z. A Molecular Toolbox of Positron Emission Tomography Tracers for General Anesthesia Mechanism Research. J Med Chem 2023; 66:6463-6497. [PMID: 37145921 DOI: 10.1021/acs.jmedchem.2c01965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
With appropriate radiotracers, positron emission tomography (PET) allows direct or indirect monitoring of the spatial and temporal distribution of anesthetics, neurotransmitters, and biomarkers, making it an indispensable tool for studying the general anesthesia mechanism. In this Perspective, PET tracers that have been recruited in general anesthesia research are introduced in the following order: 1) 11C/18F-labeled anesthetics, i.e., PET tracers made from inhaled and intravenous anesthetics; 2) PET tracers targeting anesthesia-related receptors, e.g., neurotransmitters and voltage-gated ion channels; and 3) PET tracers for studying anesthesia-related neurophysiological effects and neurotoxicity. The radiosynthesis, pharmacodynamics, and pharmacokinetics of the above PET tracers are mainly discussed to provide a practical molecular toolbox for radiochemists, anesthesiologists, and those who are interested in general anesthesia.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Tao Wang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaowei Fan
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhao Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zijing Li
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
8
|
Müller L, Power Guerra N, Schildt A, Lindner T, Stenzel J, Behrangi N, Bergner C, Alberts T, Bühler D, Kurth J, Krause BJ, Janowitz D, Teipel S, Vollmar B, Kuhla A. [ 18F]GE-180-PET and Post Mortem Marker Characteristics of Long-Term High-Fat-Diet-Induced Chronic Neuroinflammation in Mice. Biomolecules 2023; 13:biom13050769. [PMID: 37238638 DOI: 10.3390/biom13050769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/14/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity is characterized by immoderate fat accumulation leading to an elevated risk of neurodegenerative disorders, along with a host of metabolic disturbances. Chronic neuroinflammation is a main factor linking obesity and the propensity for neurodegenerative disorders. To determine the cerebrometabolic effects of diet-induced obesity (DIO) in female mice fed a long-term (24 weeks) high-fat diet (HFD, 60% fat) compared to a group on a control diet (CD, 20% fat), we used in vivo PET imaging with the radiotracer [18F]FDG as a marker for brain glucose metabolism. In addition, we determined the effects of DIO on cerebral neuroinflammation using translocator protein 18 kDa (TSPO)-sensitive PET imaging with [18F]GE-180. Finally, we performed complementary post mortem histological and biochemical analyses of TSPO and further microglial (Iba1, TMEM119) and astroglial (GFAP) markers as well as cerebral expression analyses of cytokines (e.g., Interleukin (IL)-1β). We showed the development of a peripheral DIO phenotype, characterized by increased body weight, visceral fat, free triglycerides and leptin in plasma, as well as increased fasted blood glucose levels. Furthermore, we found obesity-associated hypermetabolic changes in brain glucose metabolism in the HFD group. Our main findings with respect to neuroinflammation were that neither [18F]GE-180 PET nor histological analyses of brain samples seem fit to detect the predicted cerebral inflammation response, despite clear evidence of perturbed brain metabolism along with elevated IL-1β expression. These results could be interpreted as a metabolically activated state in brain-resident immune cells due to a long-term HFD.
Collapse
Affiliation(s)
- Luisa Müller
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Department of Psychosomatic Medicine and Psychotherapy, Rostock University Medical Centre, 18147 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| | - Nicole Power Guerra
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Institute of Anatomy, Rostock University Medical Centre, 18057 Rostock, Germany
- Smell & Taste Clinic, Department of Otorhinolaryngology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01034 Dresden, Germany
| | - Anna Schildt
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Tobias Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Jan Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Newshan Behrangi
- Institute of Anatomy and Cell Biology, Medical University of Bonn, 53115 Bonn, Germany
| | - Carina Bergner
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Teresa Alberts
- Institute of Anatomy and Cell Biology, Medical University of Bonn, 53115 Bonn, Germany
| | - Daniel Bühler
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Jens Kurth
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Bernd Joachim Krause
- Department of Clinic and Polyclinic for Nuclear Medicine, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Deborah Janowitz
- Department of Psychiatry, University of Greifswald, 17475 Greifswald, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, Rostock University Medical Centre, 18147 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, 18147 Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| | - Angela Kuhla
- Rudolf-Zenker-Institute for Experimental Surgery, Rostock University Medical Centre, 18057 Rostock, Germany
- Centre for Transdisciplinary Neurosciences Rostock (CTNR), Rostock University Medical Centre, 18147 Rostock, Germany
| |
Collapse
|
9
|
Robertson N, Sempere L, Kenyon E, Mallet C, Smith K, Hix J, Halim A, Fan J, Moore A. Omniparticle Contrast Agent for Multimodal Imaging: Synthesis and Characterization in an Animal Model. Mol Imaging Biol 2023; 25:401-412. [PMID: 36071300 PMCID: PMC9989039 DOI: 10.1007/s11307-022-01770-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Individual imaging modalities have certain advantages, but each suffers from drawbacks that other modalities may overcome. The goal of this study was to create a novel contrast agent suitable for various imaging modalities that after a single administration can bridge and strengthen the collaboration between the research fields as well as enrich the information obtained from any one modality. PROCEDURES The contrast agent platform is based on dextran-coated iron oxide nanoparticles (for MRI and MPI) and synthesized using a modified co-precipitation method, followed by a series of conjugation steps with a fluorophore (for fluorescence and photoacoustic imaging), thyroxine (for CT imaging), and chelators for radioisotope labeling (for PET imaging). The fully conjugated agent was then tested in vitro in cell uptake, viability, and phantom studies and in vivo in a model of intraductal injection and in a tumor model. RESULTS The agent was synthesized, characterized, and tested in vitro where it showed the ability to produce a signal on MRI/MPI/FL/PA/CT and PET images. Studies in cells showed the expected concentration-dependent uptake of the agent without noticeable toxicity. In vivo studies demonstrated localization of the agent to the ductal tree in mice after intraductal injection with different degrees of resolution, with CT being the best for this particular application. In a model of injected labeled tumor cells, the agent produced a signal with all modalities and showed persistence in tumor cells confirmed by histology. CONCLUSIONS A fully functional omniparticle contrast agent was synthesized and tested in vitro and in vivo in two animal models. Results shown here point to the generation of a potent signal in all modalities tested without detrimental toxicity. Future use of this agent includes its exploration in various models of human disease including image-guided diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Neil Robertson
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Radiology, College of Human Medicine, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Lorenzo Sempere
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Radiology, College of Human Medicine, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Elizabeth Kenyon
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Christiane Mallet
- Department of Radiology, College of Human Medicine, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI, 48824, USA
| | - Kylie Smith
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
| | - Jeremy Hix
- Department of Radiology, College of Human Medicine, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI, 48824, USA
| | - Alan Halim
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Jinda Fan
- Department of Radiology, College of Human Medicine, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA
| | - Anna Moore
- Precision Health Program, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA.
- Department of Radiology, College of Human Medicine, Michigan State University, 766 Service Road, East Lansing, MI, 48824, USA.
- Department of Chemistry, College of Natural Sciences, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
10
|
Zheng C, Toyonaga T, Chen B, Nicholson L, Mennie W, Liu M, Spurrier J, Deluca K, Strittmatter SM, Carson RE, Huang Y, Cai Z. Decreased synaptic vesicle glycoprotein 2A binding in a rodent model of familial Alzheimer's disease detected by [ 18F]SDM-16. Front Neurol 2023; 14:1045644. [PMID: 36846134 PMCID: PMC9945093 DOI: 10.3389/fneur.2023.1045644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Synapse loss is one of the hallmarks of Alzheimer's disease (AD) and is associated with cognitive decline. In this study, we tested [18F]SDM-16, a novel metabolically stable SV2A PET imaging probe, in the transgenic APPswe/PS1dE9 (APP/PS1) mouse model of AD and age-matched wild-type (WT) mice at 12 months of age. Methods Based on previous preclinical PET imaging studies using [11C]UCB-J and [18F]SynVesT-1 in the same strain animals, we used the simplified reference tissue model (SRTM), with brain stem as the pseudo reference region to calculate distribution volume ratios (DVRs). Results To simplify and streamline the quantitative analysis, we compared the standardized uptake value ratios (SUVRs) from different imaging windows to DVRs and found that the averaged SUVRs from 60-90 min post-injection (p.i.) are most consistent with the DVRs. Thus, we used averaged SUVRs from 60-90 min for group comparisons and found statistically significant differences in the tracer uptake in different brain regions, e.g., hippocampus (p = 0.001), striatum (p = 0.002), thalamus (p = 0.003), and cingulate cortex (p = 0.0003). Conclusions In conclusion, [18F]SDM-16 was used to detect decreased SV2A levels in the brain of APP/PS1 AD mouse model at one year old. Our data suggest that [18F]SDM-16 has similar statistical power in detecting the synapse loss in APP/PS1 mice as [11C]UCB-J and [18F]SynVesT-1, albeit later imaging window (60-90 min p.i.) is needed when SUVR is used as a surrogate for DVR for [18F]SDM-16 due to its slower brain kinetics.
Collapse
Affiliation(s)
- Chao Zheng
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Baosheng Chen
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - LaShae Nicholson
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - William Mennie
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Michael Liu
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Joshua Spurrier
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Kristin Deluca
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Stephen M. Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Departments of Neuroscience and Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Richard E. Carson
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
11
|
Képes Z, Barkóczi A, Szabó JP, Kálmán-Szabó I, Arató V, Jószai I, Deák Á, Kertész I, Hajdu I, Trencsényi G. In Vivo Preclinical Assessment of β-Amyloid-Affine [ 11C]C-PIB Accumulation in Aluminium-Induced Alzheimer's Disease-Resembling Hypercholesterinaemic Rat Model. Int J Mol Sci 2022; 23:ijms232213950. [PMID: 36430429 PMCID: PMC9695619 DOI: 10.3390/ijms232213950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Aluminum (Al) excess and hypercholesterinaemia are established risks of Alzheimer's disease (AD). The aim of this study was to establish an AD-resembling hypercholesterinaemic animal model-with the involvement of 8 week and 48 week-old Fischer-344 rats-by Al administration for the safe and rapid verification of β-amyloid-targeted positron emission tomography (PET) radiopharmaceuticals. Measurement of lipid parameters and β-amyloid-affine [11C]C-Pittsburgh Compound B ([11C]C-PIB) PET examinations were performed. Compared with the control, the significantly elevated cholesterol and LDL levels of the rats receiving the cholesterol-rich diet support the development of hypercholesterinaemia (p ≤ 0.01). In the older cohort, a notably increased age-related radiopharmaceutical accumulation was registered compared to in the young (p ≤ 0.05; p ≤ 0.01). A monotherapy-induced slight elevation of mean standardised uptake values (SUVmean) was statistically not significant; however, adult rats administered a combined diet expressed remarkable SUVmean increment compared to the adult control (SUVmean: from 0.78 ± 0.16 to 1.99 ± 0.28). One and two months after restoration to normal diet, the cerebral [11C]C-PIB accumulation of AD-mimicking animals decreased by half and a third, respectively, to the baseline value. The proposed in vivo Al-induced AD-resembling animal system seems to be adequate for the understanding of AD neuropathology and future drug testing and radiopharmaceutical development.
Collapse
Affiliation(s)
- Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Correspondence:
| | - Alexandra Barkóczi
- Department of Urology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Judit P. Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Ibolya Kálmán-Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Viktória Arató
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - István Jószai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Ádám Deák
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - István Kertész
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - István Hajdu
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| |
Collapse
|
12
|
Sharma S, Kalidindi T, Joshi S, Digwal CS, Panchal P, Burnazi E, Lee SG, Pillarsetty N, Chiosis G. Synthesis of 124I-labeled epichaperome probes and assessment in visualizing pathologic protein-protein interaction networks in tumor bearing mice. STAR Protoc 2022; 3:101318. [PMID: 35496791 PMCID: PMC9046997 DOI: 10.1016/j.xpro.2022.101318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epichaperomes are disease-associated pathologic scaffolds composed of tightly bound chaperones and co-chaperones. They provide opportunities for precision medicine where aberrant protein-protein interaction networks, rather than a single protein, are detected and targeted. This protocol describes the synthesis and characterization of two 124I-labeled epichaperome probes, [124I]-PU-H71 and [124I]-PU-AD, both which have translated to clinical studies. It shows specific steps in the use of these reagents to image and quantify epichaperome-positivity in tumor bearing mice through positron emission tomography. For complete details on the use and execution of this protocol, please refer to Bolaender et al. (2021), Inda et al. (2020), and Pillarsetty et al. (2019).
Collapse
Affiliation(s)
- Sahil Sharma
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA
| | - Teja Kalidindi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Suhasini Joshi
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA
| | - Chander S. Digwal
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA
| | - Palak Panchal
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA
| | - Eva Burnazi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sang Gyu Lee
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Gabriela Chiosis
- Program in Chemical Biology, Sloan Kettering Institute, New York, NY 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
13
|
Ren W, Ji B, Guan Y, Cao L, Ni R. Recent Technical Advances in Accelerating the Clinical Translation of Small Animal Brain Imaging: Hybrid Imaging, Deep Learning, and Transcriptomics. Front Med (Lausanne) 2022; 9:771982. [PMID: 35402436 PMCID: PMC8987112 DOI: 10.3389/fmed.2022.771982] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/16/2022] [Indexed: 12/26/2022] Open
Abstract
Small animal models play a fundamental role in brain research by deepening the understanding of the physiological functions and mechanisms underlying brain disorders and are thus essential in the development of therapeutic and diagnostic imaging tracers targeting the central nervous system. Advances in structural, functional, and molecular imaging using MRI, PET, fluorescence imaging, and optoacoustic imaging have enabled the interrogation of the rodent brain across a large temporal and spatial resolution scale in a non-invasively manner. However, there are still several major gaps in translating from preclinical brain imaging to the clinical setting. The hindering factors include the following: (1) intrinsic differences between biological species regarding brain size, cell type, protein expression level, and metabolism level and (2) imaging technical barriers regarding the interpretation of image contrast and limited spatiotemporal resolution. To mitigate these factors, single-cell transcriptomics and measures to identify the cellular source of PET tracers have been developed. Meanwhile, hybrid imaging techniques that provide highly complementary anatomical and molecular information are emerging. Furthermore, deep learning-based image analysis has been developed to enhance the quantification and optimization of the imaging protocol. In this mini-review, we summarize the recent developments in small animal neuroimaging toward improved translational power, with a focus on technical improvement including hybrid imaging, data processing, transcriptomics, awake animal imaging, and on-chip pharmacokinetics. We also discuss outstanding challenges in standardization and considerations toward increasing translational power and propose future outlooks.
Collapse
Affiliation(s)
- Wuwei Ren
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Engineering Research Center of Energy Efficient and Custom AI IC, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Cao
- Shanghai Changes Tech, Ltd., Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, ETH Zürich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Dopamine transporter silencing in the rat: systems-level alterations in striato-cerebellar and prefrontal-midbrain circuits. Mol Psychiatry 2022; 27:2329-2339. [PMID: 35246636 PMCID: PMC9126810 DOI: 10.1038/s41380-022-01471-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/15/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
Silencing of dopamine transporter (DAT), a main controlling factor of dopaminergic signaling, results in biochemical and behavioral features characteristic for neuropsychiatric diseases with presumed hyperdopaminergia including schizophrenia, attention deficit hyperactivity disorder (ADHD), bipolar disorder, and obsessive-compulsive disorder (OCD). Investigation of DAT silencing thus provides a transdiagnostic approach towards a systems-level understanding of common underlying pathways. Using a high-field multimodal imaging approach and a highly sensitive cryogenic coil, we integrated structural, functional and metabolic investigations in tandem with behavioral assessments on a newly developed preclinical rat model, comparing DAT homozygous knockout (DAT-KO, N = 14), heterozygous knockout (N = 8) and wild-type male rats (N = 14). We identified spatially distributed structural and functional brain alterations encompassing motor, limbic and associative loops that demonstrated strong behavioral relevance and were highly consistent across imaging modalities. DAT-KO rats manifested pronounced volume loss in the dorsal striatum, negatively correlating with cerebellar volume increase. These alterations were associated with hyperlocomotion, repetitive behavior and loss of efficient functional small-world organization. Further, prefrontal and midbrain regions manifested opposite changes in functional connectivity and local network topology. These prefrontal disturbances were corroborated by elevated myo-inositol levels and increased volume. To conclude, our imaging genetics approach provides multimodal evidence for prefrontal-midbrain decoupling and striato-cerebellar neuroplastic compensation as two key features of constitutive DAT blockade, proposing them as transdiagnostic mechanisms of hyperdopaminergia. Thus, our study connects developmental DAT blockade to systems-level brain changes, underlying impaired action inhibition control and resulting in motor hyperactivity and compulsive-like features relevant for ADHD, schizophrenia and OCD.
Collapse
|
15
|
Compensating Positron Range Effects of Ga-68 in Preclinical PET Imaging by Using Convolutional Neural Network: A Monte Carlo Simulation Study. Diagnostics (Basel) 2021; 11:diagnostics11122275. [PMID: 34943511 PMCID: PMC8700176 DOI: 10.3390/diagnostics11122275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/11/2021] [Accepted: 12/01/2021] [Indexed: 11/17/2022] Open
Abstract
This study aimed to investigate the feasibility of positron range correction based on three different convolutional neural network (CNN) models in preclinical PET imaging of Ga-68. The first model (CNN1) was originally designed for super-resolution recovery, while the second model (CNN2) and the third model (CNN3) were originally designed for pseudo CT synthesis from MRI. A preclinical PET scanner and 30 phantom configurations were modeled in Monte Carlo simulations, where each phantom configuration was simulated twice, once for Ga-68 (CNN input images) and once for back-to-back 511-keV gamma rays (CNN output images) with a 20 min emission scan duration. The Euclidean distance was used as the loss function to minimize the difference between CNN input and output images. According to our results, CNN3 outperformed CNN1 and CNN2 qualitatively and quantitatively. With regard to qualitative observation, it was found that boundaries in Ga-68 images became sharper after correction. As for quantitative analysis, the recovery coefficient (RC) and spill-over ratio (SOR) were increased after correction, while no substantial increase in coefficient of variation of RC (CVRC) or coefficient of variation of SOR (CVSOR) was observed. Overall, CNN3 should be a good candidate architecture for positron range correction in Ga-68 preclinical PET imaging.
Collapse
|
16
|
Amirrashedi M, Sarkar S, Mamizadeh H, Ghadiri H, Ghafarian P, Zaidi H, Ay MR. Leveraging deep neural networks to improve numerical and perceptual image quality in low-dose preclinical PET imaging. Comput Med Imaging Graph 2021; 94:102010. [PMID: 34784505 DOI: 10.1016/j.compmedimag.2021.102010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/24/2023]
Abstract
The amount of radiotracer injected into laboratory animals is still the most daunting challenge facing translational PET studies. Since low-dose imaging is characterized by a higher level of noise, the quality of the reconstructed images leaves much to be desired. Being the most ubiquitous techniques in denoising applications, edge-aware denoising filters, and reconstruction-based techniques have drawn significant attention in low-count applications. However, for the last few years, much of the credit has gone to deep-learning (DL) methods, which provide more robust solutions to handle various conditions. Albeit being extensively explored in clinical studies, to the best of our knowledge, there is a lack of studies exploring the feasibility of DL-based image denoising in low-count small animal PET imaging. Therefore, herein, we investigated different DL frameworks to map low-dose small animal PET images to their full-dose equivalent with quality and visual similarity on a par with those of standard acquisition. The performance of the DL model was also compared to other well-established filters, including Gaussian smoothing, nonlocal means, and anisotropic diffusion. Visual inspection and quantitative assessment based on quality metrics proved the superior performance of the DL methods in low-count small animal PET studies, paving the way for a more detailed exploration of DL-assisted algorithms in this domain.
Collapse
Affiliation(s)
- Mahsa Amirrashedi
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran.
| | - Saeed Sarkar
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hojjat Mamizadeh
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Ghadiri
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran.
| | - Pardis Ghafarian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran; PET/CT and Cyclotron Center, Masih Daneshvari Hospital, Shahid Beheshti University of Medical, Tehran, Iran.
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva CH-1211, Switzerland; Geneva University Neurocenter, Geneva University, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| | - Mohammad Reza Ay
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Filip T, Mairinger S, Neddens J, Sauberer M, Flunkert S, Stanek J, Wanek T, Okamura N, Langer O, Hutter-Paier B, Kuntner C. Characterization of an APP/tau rat model of Alzheimer's disease by positron emission tomography and immunofluorescent labeling. Alzheimers Res Ther 2021; 13:175. [PMID: 34656177 PMCID: PMC8522096 DOI: 10.1186/s13195-021-00916-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/05/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND To better understand the etiology and pathomechanisms of Alzheimer's disease, several transgenic animal models that overexpress human tau or human amyloid-beta (Aβ) have been developed. In the present study, we generated a novel transgenic rat model by cross-breeding amyloid precursor protein (APP) rats with tau rats. We characterized this model by performing positron emission tomography scans combined with immunofluorescent labeling and cerebrospinal fluid analyses. METHODS APP/Tau rats were generated by cross-breeding male McGill-R-Thy1-APP transgenic rats with female hTau-40/P301L transgenic rats. APP/Tau double transgenic rats and non-transgenic (ntg) littermates aged 7, 13, and 21 months were subjected to dynamic [11C] PiB scan and dynamic [18F]THK-5317 scans. For regional brain analysis, a template was generated from anatomical MR images of selected animals, which was co-registered with the PET images. Regional analysis was performed by application of the simplified reference tissue model ([11C]PiB data), whereas [18F]THK-5317 data were analyzed using a 2-tissue compartment model and Logan graphical analysis. In addition, immunofluorescent labeling (tau, amyloid) and cerebrospinal fluid analyses were performed. RESULTS [11C]PiB binding potential (BPND) and [18F]THK-5317 volume of distribution (VT) showed an increase with age in several brain regions in the APP/Tau group but not in the ntg control group. Immunohistochemical analysis of brain slices of PET-scanned animals revealed a positive correlation between Aβ labeling and [11C]PiB regional BPND. Tau staining yielded a trend towards higher levels in the cortex and hippocampus of APP/Tau rats compared with ntg littermates, but without reaching statistical significance. No correlation was found between tau immunofluorescence labeling results and the respective [18F]THK-5317 VT values. CONCLUSIONS We thoroughly characterized a novel APP/Tau rat model using combined PET imaging and immunofluorescence analysis. We observed an age-related increase in [11C]PiB and [18F]THK-5317 binding in several brain regions in the APP/Tau group but not in the ntg group. Although we were able to reveal a positive correlation between amyloid labeling and [11C]PiB regional brain uptake, we observed relatively low human tau and amyloid fibril expression levels and a somewhat unstable brain pathology which questions the utility of this animal model for further studies.
Collapse
Affiliation(s)
- Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Research, Medical University Vienna, Vienna, Austria
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Joerg Neddens
- Neuropharmacology, QPS Austria GmbH, Grambach, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Claudia Kuntner
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444, Seibersdorf, Austria.
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Shi XF, Ji B, Kong Y, Guan Y, Ni R. Multimodal Contrast Agents for Optoacoustic Brain Imaging in Small Animals. Front Bioeng Biotechnol 2021; 9:746815. [PMID: 34650961 PMCID: PMC8505530 DOI: 10.3389/fbioe.2021.746815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
Optoacoustic (photoacoustic) imaging has demonstrated versatile applications in biomedical research, visualizing the disease pathophysiology and monitoring the treatment effect in an animal model, as well as toward applications in the clinical setting. Given the complex disease mechanism, multimodal imaging provides important etiological insights with different molecular, structural, and functional readouts in vivo. Various multimodal optoacoustic molecular imaging approaches have been applied in preclinical brain imaging studies, including optoacoustic/fluorescence imaging, optoacoustic imaging/magnetic resonance imaging (MRI), optoacoustic imaging/MRI/Raman, optoacoustic imaging/positron emission tomography, and optoacoustic/computed tomography. There is a rapid development in molecular imaging contrast agents employing a multimodal imaging strategy for pathological targets involved in brain diseases. Many chemical dyes for optoacoustic imaging have fluorescence properties and have been applied in hybrid optoacoustic/fluorescence imaging. Nanoparticles are widely used as hybrid contrast agents for their capability to incorporate different imaging components, tunable spectrum, and photostability. In this review, we summarize contrast agents including chemical dyes and nanoparticles applied in multimodal optoacoustic brain imaging integrated with other modalities in small animals, and provide outlook for further research.
Collapse
Affiliation(s)
- Xue-feng Shi
- Department of Respiratory Medicine, Qinghai Provincial People’s Hospital, Xining, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
A novel antioxidant ergothioneine PET radioligand for in vivo imaging applications. Sci Rep 2021; 11:18450. [PMID: 34531467 PMCID: PMC8446031 DOI: 10.1038/s41598-021-97925-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 09/01/2021] [Indexed: 11/09/2022] Open
Abstract
Ergothioneine (ERGO) is a rare amino acid mostly found in fungi, including mushrooms, with recognized antioxidant activity to protect tissues from damage by reactive oxygen species (ROS) components. Prior to this publication, the biodistribution of ERGO has been performed solely in vitro using extracted tissues. The aim of this study was to develop a feasible chemistry for the synthesis of an ERGO PET radioligand, [11C]ERGO, to facilitate in vivo study. The radioligand probe was synthesized with identical structure to ERGO by employing an orthogonal protection/deprotection approach. [11C]methylation of the precursor was performed via [11C]CH3OTf to provide [11C]ERGO radioligand. The [11C]ERGO was isolated by RP-HPLC with a molar activity of 690 TBq/mmol. To demonstrate the biodistribution of the radioligand, we administered approximately 37 MBq/0.1 mL in 5XFAD mice, a mouse model of Alzheimer's disease via the tail vein. The distribution of ERGO in the brain was monitored using 90-min dynamic PET scans. The delivery and specific retention of [11C]ERGO in an LPS-mediated neuroinflammation mouse model was also demonstrated. For the pharmacokinetic study, the concentration of the compound in the serum started to decrease 10 min after injection while starting to distribute in other peripheral tissues. In particular, a significant amount of the compound was found in the eyes and small intestine. The radioligand was also distributed in several regions of the brain of 5XFAD mice, and the signal remained strong 30 min post-injection. This is the first time the biodistribution of this antioxidant and rare amino acid has been demonstrated in a preclinical mouse model in a highly sensitive and non-invasive manner.
Collapse
|
20
|
Placzek MS. Imaging Kappa Opioid Receptors in the Living Brain with Positron Emission Tomography. Handb Exp Pharmacol 2021; 271:547-577. [PMID: 34363128 DOI: 10.1007/164_2021_498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Kappa opioid receptor (KOR) neuroimaging using positron emission tomography (PET) has been immensely successful in all phases of discovery and validation in relation to radiotracer development from preclinical imaging to human imaging. There are now several KOR-specific PET radiotracers that can be utilized for neuroimaging, including agonist and antagonist ligands, as well as C-11 and F-18 variants. These technologies will increase KOR PET utilization by imaging centers around the world and have provided a foundation for future studies. In this chapter, I review the advances in KOR radiotracer discovery, focusing on ligands that have been translated into human imaging, and highlight key attributes unique to each KOR PET radiotracer. The utilization of these radiotracers in KOR PET neuroimaging can be subdivided into three major investigational classes: the first, measurement of KOR density; the second, measurement of KOR drug occupancy; the third, detecting changes in endogenous dynorphin following activation or deactivation. Given the involvement of the KOR/dynorphin system in a number of brain disorders including, but not limited to, pain, itch, mood disorders and addiction, measuring KOR density in the living brain will offer insight into the chronic effects of these disorders on KOR tone in humans. Notably, KOR PET has been successful at measuring drug occupancy in the human brain to guide dose selection for maximal therapeutic efficacy while avoiding harmful side effects. Lastly, we discuss the potential of KOR PET to detect changes in endogenous dynorphin in the human brain, to elucidate neural mechanisms and offer critical insight into disease-modifying therapeutics. We conclude with comments on other translational neuroimaging modalities such as MRI that could be used to study KOR-dynorphin tone in the living human brain.
Collapse
Affiliation(s)
- Michael S Placzek
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA. .,Department of Radiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Uccelli NA, Codagnone MG, Traetta ME, Levanovich N, Rosato Siri MV, Urrutia L, Falasco G, Vázquez S, Pasquini JM, Reinés AG. Neurobiological substrates underlying corpus callosum hypoconnectivity and brain metabolic patterns in the valproic acid rat model of autism spectrum disorder. J Neurochem 2021; 159:128-144. [PMID: 34081798 DOI: 10.1111/jnc.15444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 12/26/2022]
Abstract
Atypical connectivity between brain regions and altered structure of the corpus callosum (CC) in imaging studies supports the long-distance hypoconnectivity hypothesis proposed for autism spectrum disorder (ASD). The aim of this study was to unveil the CC ultrastructural and cellular changes employing the valproic acid (VPA) rat model of ASD. Male Wistar rats were exposed to VPA (450 mg/kg i.p.) or saline (control) during gestation (embryonic day 10.5), and maturation, exploration, and social behavior were subsequently tested. Myelin content, ultrastructure, and oligodendroglial lineage were studied in the CC at post-natal days 15 (infant) and 36 (juvenile). As a functional outcome, brain metabolic activity was determined by positron emission tomography. Concomitantly with behavioral deficits in juvenile VPA rats, the CC showed reduced myelin basic protein, conserved total number of axons, reduced percentage of myelinated axons, and aberrant and less compact arrangements of myelin sheath ultrastructure. Mature oligodendrocytes decreased and oligodendrocyte precursors increased in the absence of astrogliosis or microgliosis. In medial prefrontal and somatosensory cortices of juvenile VPA rats, myelin ultrastructure and oligodendroglial lineage were preserved. VPA animals exhibited global brain hypometabolism and local hypermetabolism in brain regions relevant for ASD. In turn, the CC of infant VPA rats showed reduced myelin content but preserved oligodendroglial lineage. Our findings indicate that CC hypomyelination is established during infancy and prior to oligodendroglial pattern alterations, which suggests that axon-oligodendroglia communication could be compromised in VPA animals. Thus, CC hypomyelination may underlie white matter alterations and contribute to atypical patterns of connectivity and metabolism found in ASD.
Collapse
Affiliation(s)
- Nonthué Alejandra Uccelli
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN) Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martín Gabriel Codagnone
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN) Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Evelyn Traetta
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN) Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nadia Levanovich
- Fundación para la lucha contra las enfermedades neurológicas de la infancia (FLENI), Centro de Imágenes Moleculares (CIM), Escobar, Argentina
| | - María Victoria Rosato Siri
- CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológica (IQUIFIB) Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Leandro Urrutia
- Fundación para la lucha contra las enfermedades neurológicas de la infancia (FLENI), Centro de Imágenes Moleculares (CIM), Escobar, Argentina
| | - Germán Falasco
- Fundación para la lucha contra las enfermedades neurológicas de la infancia (FLENI), Centro de Imágenes Moleculares (CIM), Escobar, Argentina
| | - Silvia Vázquez
- Fundación para la lucha contra las enfermedades neurológicas de la infancia (FLENI), Centro de Imágenes Moleculares (CIM), Escobar, Argentina
| | - Juana María Pasquini
- CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológica (IQUIFIB) Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Gabriela Reinés
- CONICET-Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN) Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Walker M, Kuebler L, Goehring CM, Pichler BJ, Herfert K. Imaging SERT Availability in a Rat Model of L-DOPA-Induced Dyskinesia. Mol Imaging Biol 2021; 22:634-642. [PMID: 31392531 DOI: 10.1007/s11307-019-01418-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The development of L-DOPA-induced dyskinesia (LID) is one of the most severe side effects of chronic L-DOPA treatment in Parkinson's disease patients. [11C]DASB positron emission tomography (PET) provides a prominent tool to visualize and quantify serotonin transporter (SERT) pathology in vivo in patients and in animal models. To evaluate the effect of chronic L-DOPA treatment on SERT availability in an animal model of LID, we performed a longitudinal PET study. PROCEDURES Rats received a unilateral 6-hydroxydopamine (6-OHDA) lesion, and striatal and extrastriatal SERT expression levels were studied with [11C]DASB, a marker of SERT availability, before and after daily treatment with L-DOPA. Dyskinesias were evaluated at different time points over a period of 21 days. RESULTS [11C]DASB binding was found to be decreased after 6-OHDA lesions in the striatum, cortex, and hippocampus 5 weeks after 6-OHDA injection in the lesioned hemisphere of the rat brain. Chronic L-DOPA priming resulted in a relative preservation of SERT availability in the lesioned and healthy hemisphere compared to baseline measurements. CONCLUSIONS Our longitudinal PET data support a preservation of SERT availability after the induction of L-DOPA-induced dyskinesia, which is in line with previous reports in dyskinetic PD patients.
Collapse
Affiliation(s)
- Michael Walker
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Laura Kuebler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Chris Marc Goehring
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany.
| |
Collapse
|
23
|
Lothmann K, Amunts K, Herold C. The Neurotransmitter Receptor Architecture of the Mouse Olfactory System. Front Neuroanat 2021; 15:632549. [PMID: 33967704 PMCID: PMC8102831 DOI: 10.3389/fnana.2021.632549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
The uptake, transmission and processing of sensory olfactory information is modulated by inhibitory and excitatory receptors in the olfactory system. Previous studies have focused on the function of individual receptors in distinct brain areas, but the receptor architecture of the whole system remains unclear. Here, we analyzed the receptor profiles of the whole olfactory system of adult male mice. We examined the distribution patterns of glutamatergic (AMPA, kainate, mGlu2/3, and NMDA), GABAergic (GABAA, GABAA(BZ), and GABAB), dopaminergic (D1/5) and noradrenergic (α1 and α2) neurotransmitter receptors by quantitative in vitro receptor autoradiography combined with an analysis of the cyto- and myelo-architecture. We observed that each subarea of the olfactory system is characterized by individual densities of distinct neurotransmitter receptor types, leading to a region- and layer-specific receptor profile. Thereby, the investigated receptors in the respective areas and strata showed a heterogeneous expression. Generally, we detected high densities of mGlu2/3Rs, GABAA(BZ)Rs and GABABRs. Noradrenergic receptors revealed a highly heterogenic distribution, while the dopaminergic receptor D1/5 displayed low concentrations, except in the olfactory tubercle and the dorsal endopiriform nucleus. The similarities and dissimilarities of the area-specific multireceptor profiles were analyzed by a hierarchical cluster analysis. A three-cluster solution was found that divided the areas into the (1) olfactory relay stations (main and accessory olfactory bulb), (2) the olfactory cortex (anterior olfactory cortex, dorsal peduncular cortex, taenia tecta, piriform cortex, endopiriform nucleus, entorhinal cortex, orbitofrontal cortex) and the (3) olfactory tubercle, constituting its own cluster. The multimodal receptor-architectonic analysis of each component of the olfactory system provides new insights into its neurochemical organization and future possibilities for pharmaceutic targeting.
Collapse
Affiliation(s)
- Kimberley Lothmann
- C. & O. Vogt-Institute of Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Katrin Amunts
- C. & O. Vogt-Institute of Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.,Institute of Neuroscience and Medicine INM-1, Research Centre Jülich, Jülich, Germany
| | - Christina Herold
- C. & O. Vogt-Institute of Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
24
|
D'Elia A, Schiavi S, Soluri A, Massari R, Soluri A, Trezza V. Role of Nuclear Imaging to Understand the Neural Substrates of Brain Disorders in Laboratory Animals: Current Status and Future Prospects. Front Behav Neurosci 2020; 14:596509. [PMID: 33362486 PMCID: PMC7759612 DOI: 10.3389/fnbeh.2020.596509] [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: 08/20/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Molecular imaging, which allows the real-time visualization, characterization and measurement of biological processes, is becoming increasingly used in neuroscience research. Scintigraphy techniques such as single photon emission computed tomography (SPECT) and positron emission tomography (PET) provide qualitative and quantitative measurement of brain activity in both physiological and pathological states. Laboratory animals, and rodents in particular, are essential in neuroscience research, providing plenty of models of brain disorders. The development of innovative high-resolution small animal imaging systems together with their radiotracers pave the way to the study of brain functioning and neurotransmitter release during behavioral tasks in rodents. The assessment of local changes in the release of neurotransmitters associated with the performance of a given behavioral task is a turning point for the development of new potential drugs for psychiatric and neurological disorders. This review addresses the role of SPECT and PET small animal imaging systems for a better understanding of brain functioning in health and disease states. Brain imaging in rodent models faces a series of challenges since it acts within the boundaries of current imaging in terms of sensitivity and spatial resolution. Several topics are discussed, including technical considerations regarding the strengths and weaknesses of both technologies. Moreover, the application of some of the radioligands developed for small animal nuclear imaging studies is discussed. Then, we examine the changes in metabolic and neurotransmitter activity in various brain areas during task-induced neural activation with special regard to the imaging of opioid, dopaminergic and cannabinoid receptors. Finally, we discuss the current status providing future perspectives on the most innovative imaging techniques in small laboratory animals. The challenges and solutions discussed here might be useful to better understand brain functioning allowing the translation of preclinical results into clinical applications.
Collapse
Affiliation(s)
- Annunziata D'Elia
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| | - Sara Schiavi
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| | - Andrea Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Roberto Massari
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Alessandro Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (CNR), Rome, Italy
| | - Viviana Trezza
- Section of Biomedical Sciences and Technologies, Department of Science, University “Roma Tre”, Rome, Italy
| |
Collapse
|
25
|
Watanabe H, Matsushita N, Shimizu Y, Iikuni S, Nakamoto Y, Togashi K, Ono M. Synthesis and characterization of a novel 18F-labeled 2,5-diarylnicotinamide derivative targeting orexin 2 receptor. MEDCHEMCOMM 2019; 10:2126-2130. [PMID: 32904113 PMCID: PMC7451066 DOI: 10.1039/c9md00397e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 10/25/2019] [Indexed: 11/21/2022]
Abstract
Orexin 2 receptor (OX2R) is thought to play an important role in the arousal-promoting function, but its distribution and function in the pathophysiology of orexin-mediated disorders remains to be fully elucidated. In the present study, we synthesized and characterized a novel 18F-labeled 2,5-diarylnicotinamide (DAN) derivative as a potential positron emission tomography (PET) probe for in vivo imaging of OX2R. In in vitro binding experiments, [18F]DAN-1 selectively bound to OX2R. In a biodistribution study using normal mice, [18F]DAN-1 displayed moderate brain uptake (2.10% ID per g at 10 min post-injection). In addition, the radioactivity in the mouse brain at 30 min post-injection was significantly decreased by co-injection with nonradioactive DAN-1, but high nonspecific binding was observed. These results suggested that further structural modifications of [18F]DAN-1 are needed to use it for imaging OX2R in the brain.
Collapse
Affiliation(s)
- Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis , Graduate School of Pharmaceutical Sciences , Kyoto University , 46-29 Yoshida Shimoadachi-cho, Sakyo-ku , Kyoto 606-8501 , Japan . ; ; ; Tel: +81 75 753 4566
| | - Naoki Matsushita
- Department of Patho-Functional Bioanalysis , Graduate School of Pharmaceutical Sciences , Kyoto University , 46-29 Yoshida Shimoadachi-cho, Sakyo-ku , Kyoto 606-8501 , Japan . ; ; ; Tel: +81 75 753 4566
| | - Yoichi Shimizu
- Department of Patho-Functional Bioanalysis , Graduate School of Pharmaceutical Sciences , Kyoto University , 46-29 Yoshida Shimoadachi-cho, Sakyo-ku , Kyoto 606-8501 , Japan . ; ; ; Tel: +81 75 753 4566
- Department of Diagnostic Imaging and Nuclear Medicine , Graduate School of Medicine , Kyoto University , 54 Shogoin Kawahara-cho, Sakyo-ku , Kyoto 606-8507 , Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis , Graduate School of Pharmaceutical Sciences , Kyoto University , 46-29 Yoshida Shimoadachi-cho, Sakyo-ku , Kyoto 606-8501 , Japan . ; ; ; Tel: +81 75 753 4566
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine , Graduate School of Medicine , Kyoto University , 54 Shogoin Kawahara-cho, Sakyo-ku , Kyoto 606-8507 , Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine , Graduate School of Medicine , Kyoto University , 54 Shogoin Kawahara-cho, Sakyo-ku , Kyoto 606-8507 , Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis , Graduate School of Pharmaceutical Sciences , Kyoto University , 46-29 Yoshida Shimoadachi-cho, Sakyo-ku , Kyoto 606-8501 , Japan . ; ; ; Tel: +81 75 753 4566
| |
Collapse
|