1
|
Haas S, Bravo F, Ionescu TM, Gonzalez-Menendez I, Quintanilla-Martinez L, Dunkel G, Kuebler L, Hahn A, Lanzenberger R, Weigelin B, Reischl G, Pichler BJ, Herfert K. Functional PET/MRI reveals active inhibition of neuronal activity during optogenetic activation of the nigrostriatal pathway. SCIENCE ADVANCES 2024; 10:eadn2776. [PMID: 39454014 PMCID: PMC11506239 DOI: 10.1126/sciadv.adn2776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/23/2024] [Indexed: 10/27/2024]
Abstract
The dopaminergic system is a central component of the brain's neurobiological framework, governing motor control and reward responses and playing an essential role in various brain disorders. Within this complex network, the nigrostriatal pathway represents a critical circuit for dopamine neurotransmission from the substantia nigra to the striatum. However, stand-alone functional magnetic resonance imaging is unable to study the intricate interplay between brain activation and its molecular underpinnings. In our study, the use of a functional [fluorine-18]2-fluor-2-deoxy-d-glucose positron emission tomography approach, simultaneously with blood oxygen level-dependent functional magnetic resonance imaging, provided an important insight that demonstrates an active suppression of the nigrostriatal activity during optogenetic stimulation. This result increases our understanding of the molecular mechanisms of brain function and provides an important perspective on how dopamine influences hemodynamic responses in the brain.
Collapse
Affiliation(s)
- Sabrina Haas
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Fernando Bravo
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Tudor M. Ionescu
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Gina Dunkel
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Laura Kuebler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Bettina Weigelin
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Bernd J. Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Kristina Herfert
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
2
|
Ouyang M, Detre JA, Hyland JL, Sindabizera KL, Kuschner ES, Edgar JC, Peng Y, Huang H. Spatiotemporal cerebral blood flow dynamics underlies emergence of the limbic-sensorimotor-association cortical gradient in human infancy. Nat Commun 2024; 15:8944. [PMID: 39414859 PMCID: PMC11484854 DOI: 10.1038/s41467-024-53354-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 10/02/2024] [Indexed: 10/18/2024] Open
Abstract
Infant cerebral blood flow (CBF) delivers nutrients and oxygen to fulfill brain energy consumption requirements for the fastest period of postnatal brain development across the lifespan. However, organizing principle of whole-brain CBF dynamics during infancy remains obscure. Leveraging a unique cohort of 100+ infants with high-resolution arterial spin labeled MRI, we find the emergence of the cortical hierarchy revealed by the highest-resolution infant CBF maps available to date. Infant CBF across cortical regions increases in a biphasic pattern featured by initial rapid and subsequently slower rate, and break-point ages increasing along the limbic-sensorimotor-association cortical gradient. Increases in CBF in sensorimotor cortices are associated with enhanced language and motor skills, and frontoparietal association cortices with cognitive skills. The study discovers emergence of the hierarchical limbic-sensorimotor-association cortical gradient in infancy and offers standardized reference of infant brain CBF and insight into the physiological basis of cortical specialization and real-world infant developmental functioning.
Collapse
Affiliation(s)
- Minhui Ouyang
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John A Detre
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica L Hyland
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kay L Sindabizera
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emily S Kuschner
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J Christopher Edgar
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yun Peng
- Department of Radiology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Hao Huang
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Rey-Bretal D, García-Varela L, Gómez-Lado N, Moscoso A, Piñeiro-Fiel M, Díaz-Platas L, Medin S, Fernández-Ferreiro A, Ruibal Á, Sobrino T, Silva-Rodríguez J, Aguiar P. Quantitative brain [ 18F]FDG PET beyond normal blood glucose levels. Neuroimage 2024; 300:120873. [PMID: 39341474 DOI: 10.1016/j.neuroimage.2024.120873] [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: 05/09/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction SUV measurements from static brain [18F]FDG PET acquisitions are a commonly used tool in preclinical research, providing a simple alternative for kinetic modelling, which requires complex and time-consuming dynamic acquisitions. However, SUV can be severely affected by the animal handling and preconditioning protocols, primarily by those that may induce changes in blood glucose levels (BGL). Here, we aimed at developing and investigating the feasibility of SUV-based approaches for a wide range of BGL far beyond normal values, and consequently, to develop and validate a new model to generate standardized and reproducible SUV measurements for any BGL. Material and methods We performed dynamic and static brain [18F]FDG PET acquisitions in 52 male Sprague-Dawley rats sorted into control (n = 10), non-fasting (n = 14), insulin-induced hypoglycemia (n = 12) and glucagon-induced hyperglycemia (n = 16) groups. Brain [18F]FDG PET images were cropped, aligned and co-registered to a standard template to calculate whole-brain and regional SUV. Cerebral Metabolic Rate of Glucose (CMRglc) was also estimated from 2-Tissue Compartment Model (2TCM) and Patlak plot for validation purposes. Results Our results showed that BGL=100±6 mg/dL can be considered a reproducible reference value for normoglycemia. Furthermore, we successfully established a 2nd-degree polynomial model (C1=0.66E-4, C2=-0.0408 and C3=7.298) relying exclusively on BGL measures at pre-[18F]FDG injection time, that characterizes more precisely the relationship between SUV and BGL for a wide range of BGL values (from 10 to 338 mg/dL). We confirmed the ability of this model to generate corrected SUV estimations that are highly correlated to CMRglc estimations (R2= 0.54 2TCM CMRgluc and R2= 0.49 Patlak CMRgluc). Besides, slight regional differences in SUV were found in animals from extreme BGL groups, showing that [18F]FDG uptake is mostly directed toward central regions of the brain when BGLs are significantly decreased. Conclusion Our study successfully established a non-linear model that relies exclusively on pre-scan BGL measurements to characterize the relationship between [18F]FDG SUV and BGL. The extensive validation confirmed its ability to generate SUV-based surrogates of CMRglu along a wide range of BGL and it holds the potential to be adopted as a standard protocol by the preclinical neuroimaging community using brain [18F]FDG PET imaging.
Collapse
Affiliation(s)
- David Rey-Bretal
- Molecular Imaging Biomarkers Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Biomarkers Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Lara García-Varela
- Molecular Imaging Biomarkers Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Biomarkers Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Noemí Gómez-Lado
- Molecular Imaging Biomarkers Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Biomarkers Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alexis Moscoso
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Psychiatry and Neurochemistry, Institute of Physiology and Neuroscience, University of Gothenburg, Gothenburg, Sweden
| | - Manuel Piñeiro-Fiel
- Molecular Imaging Biomarkers Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Biomarkers Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Lucía Díaz-Platas
- Galician PET Radiopharmacy Unit, GALARIA, University Clinical Hospital, Santiago de Compostela, Spain
| | - Santiago Medin
- Galician PET Radiopharmacy Unit, GALARIA, University Clinical Hospital, Santiago de Compostela, Spain
| | - Anxo Fernández-Ferreiro
- Pharmacy Department, University Clinical Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, Spain; FarmaCHUS Lab, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Álvaro Ruibal
- Molecular Imaging Biomarkers Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Biomarkers Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Jesús Silva-Rodríguez
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain; Reina Sofia Alzheimer Centre, CIEN Foundation, ISCIII, Madrid, Spain.
| | - Pablo Aguiar
- Molecular Imaging Biomarkers Group. Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain; Nuclear Medicine Department and Molecular Imaging Biomarkers Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Kroll T, Miranda A, Drechsel A, Beer S, Lang M, Drzezga A, Rosa-Neto P, Verhaeghe J, Elmenhorst D, Bauer A. Dynamic neuroreceptor positron emission tomography in non-anesthetized rats using point source based motion correction: A feasibility study with [ 11C]ABP688. J Cereb Blood Flow Metab 2024; 44:1852-1866. [PMID: 38684219 PMCID: PMC11504418 DOI: 10.1177/0271678x241239133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 05/02/2024]
Abstract
To prevent motion artifacts in small animal positron emission tomography (PET), animals are routinely scanned under anesthesia or physical restraint. Both may potentially alter metabolism and neurochemistry. This study investigates the feasibility of fully awake acquisition and subsequent absolute quantification of dynamic brain PET data via pharmacokinetic modelling in moving rats using the glutamate 5 receptor radioligand [11C]ABP688 and point source based motion correction. Five male rats underwent three dynamic [11C]ABP688 PET scans: two test-retest awake PET scans and one scan under anesthesia for comparison. Specific radioligand binding was determined via the simplified reference tissue model (reference: cerebellum) and outcome parameters BPND and R1 were evaluated in terms of stability and reproducibility. Test-retest measurements in awake animals gave reliable results with high correlations of BPND (y = 1.08 × -0.2, r = 0.99, p < 0.01) and an acceptable variability (mean over all investigated regions 15.7 ± 2.4%). Regional [11C]ABP688 BPNDs under awake and anesthetized conditions were comparable although in awake scans, absolute radioactive peak uptakes were lower and relative blood flow in terms of R1 was higher. Awake small animal PET with absolute quantification of neuroreceptor availability is technically feasible and reproducible thereby providing a suitable alternative whenever effects of anesthesia are undesirable, e.g. in sleep research.
Collapse
Affiliation(s)
- Tina Kroll
- Institute of Neurosciences and Medicine (INM-2), Forschungszentrum Jülich GmbH, Germany
| | - Alan Miranda
- Molecular Imaging Center Antwerp, University of Antwerp, Belgium
| | - Alexandra Drechsel
- Institute of Neurosciences and Medicine (INM-2), Forschungszentrum Jülich GmbH, Germany
| | - Simone Beer
- Institute of Neurosciences and Medicine (INM-2), Forschungszentrum Jülich GmbH, Germany
| | - Markus Lang
- Institute of Neurosciences and Medicine (INM-5), Forschungszentrum Jülich GmbH, Germany
| | - Alexander Drzezga
- Institute of Neurosciences and Medicine (INM-2), Forschungszentrum Jülich GmbH, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Belgium
| | - David Elmenhorst
- Institute of Neurosciences and Medicine (INM-2), Forschungszentrum Jülich GmbH, Germany
- Department of Nuclear Medicine, University Hospital Cologne, Germany
| | - Andreas Bauer
- Institute of Neurosciences and Medicine (INM-2), Forschungszentrum Jülich GmbH, Germany
| |
Collapse
|
5
|
Wang Y, Mo J, Sun Y, Yu H, Liu C, Liu Q, Fan Y, Wang S, Liu X, Jiang Y, Cai L. Establishment of a normal control model of children's brain 18-fluorodeoxyglucose positron emission tomography and analysis of the changing pattern in patients aged 0-14 years. Quant Imaging Med Surg 2024; 14:4703-4713. [PMID: 39022258 PMCID: PMC11250353 DOI: 10.21037/qims-23-1809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/23/2024] [Indexed: 07/20/2024]
Abstract
Background It is difficult to obtain 18-fluorodeoxyglucose positron emission tomography (18FDG-PET) data from normal children, and changes in brain metabolism in children due to growth and development are poorly understood. For the first time, we established a normal control model of brain 18FDG-PET in children and evaluated its feasibility. The association of PET with age in children aged 0-14 years was analyzed. This study aimed to establish a normal control model of brain 18FDG-PET in children for the first time and to verify its feasibility, and to analyze the trend of PET with age in children aged 0-14 years. Methods In this retrospective cohort study, the 18FDG-PET imaging data of patients with no epileptiform discharge involvement contralateral to the epileptogenic zone were consecutively collected from January 2015 to June 2022 according to strictly defined screening criteria. For the normal control data, the hemisphere contralateral to the epileptogenic zone was mirrored and spliced to form an intact brain. The cohort of children aged 0-14 years was divided into 14 groups according age by year. Subsequently, patients who underwent lesionectomy with clear hypometabolism that roughly coincided with the extent of surgical resection were examined. The PET scan was compared with the control model, and the ratio of overlapping parts (hypometabolic areas ∩ surgical resection area) to hypometabolic parts (ROH) was calculated. Multiple regression analysis was performed on the normal control model for every 3- to 4-year age interval. Results A total of 159 normal control models were established. Five patients were randomly selected to verify the reliability of each yearly model. The average ROH was 0.968. Metabolism increasing with age in the different brain regions was observed at ages 0-2~, 3-5~, and 6-10 years. No age-related metabolic increase or decrease was found in the 10- to 14-year-old group. The metabolism in the 7- to 8-year-old group was higher than that in the 13- to 14-year-old group. Conclusions With strict screening criteria, the method of mirroring the contralateral hemisphere of the epileptic zone and splicing it into a complete brain as a means of creating a normal control group is feasible. The method offers convenience to the studies that lack healthy pediatric controls. Children under 10 years of age (especially 0-6 years old) experience considerable metabolic changes year on year. After the age of 10 years, the changes in metabolism gradually decrease, and metabolism also slowly decreases. Our findings provide guidance the clinical interpretation of areas with hypometabolism and emphasize the importance of establishing a normal control model of the child's brain, which should not be replaced by an adult model.
Collapse
Affiliation(s)
- Yao Wang
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Jiajie Mo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Sun
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Hao Yu
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Chang Liu
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Qingzhu Liu
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Yan Fan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Shuang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xiaoyan Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Lixin Cai
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| |
Collapse
|
6
|
Tiefenbach J, Shannon L, Lobosky M, Johnson S, Chan HH, Byram N, Machado AG, Androjna C, Baker KB. A novel restrainer device for acquistion of brain images in awake rats. Neuroimage 2024; 289:120556. [PMID: 38423263 PMCID: PMC10935597 DOI: 10.1016/j.neuroimage.2024.120556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
Functional neuroimaging methods like fMRI and PET are vital in neuroscience research, but require that subjects remain still throughout the scan. In animal research, anesthetic agents are typically applied to facilitate the acquisition of high-quality data with minimal motion artifact. However, anesthesia can have profound effects on brain metabolism, selectively altering dynamic neural networks and confounding the acquired data. To overcome the challenge, we have developed a novel head fixation device designed to support awake rat brain imaging. A validation experiment demonstrated that the device effectively minimizes animal motion throughout the scan, with mean absolute displacement and mean relative displacement of 0.0256 (SD: 0.001) and 0.009 (SD: 0.002), across eight evaluated subjects throughout fMRI image acquisition (total scanning time per subject: 31 min, 12 s). Furthermore, the awake scans did not induce discernable stress to the animals, with stable physiological parameters throughout the scan (Mean HR: 344, Mean RR: 56, Mean SpO2: 94 %) and unaltered serum corticosterone levels (p = 0.159). In conclusion, the device presented in this paper offers an effective and safe method of acquiring functional brain images in rats, allowing researchers to minimize the confounding effects of anesthetic use.
Collapse
Affiliation(s)
- Jakov Tiefenbach
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH 44195, USA.
| | - Logan Shannon
- Engineering Core, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | - Mark Lobosky
- Small Animal Imaging Core, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | - Sadie Johnson
- Engineering Core, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | - Hugh H Chan
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | - Nicole Byram
- Cleveland Clinic Innovations, Cleveland Clinic, OH 44195, USA
| | - Andre G Machado
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | - Charlie Androjna
- Engineering Core, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| | - Kenneth B Baker
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, OH 44195, USA
| |
Collapse
|
7
|
Kepes Z, Arato V, Csikos C, Hegedus E, Esze R, Nagy T, Joszai I, Emri M, Kertesz I, Trencsenyi G. In Vivo Evaluation of Brain [ 18F]F-FDG Uptake Pattern Under Different Anaesthesia Protocols. In Vivo 2024; 38:587-597. [PMID: 38418149 PMCID: PMC10905451 DOI: 10.21873/invivo.13477] [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: 10/20/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 03/01/2024]
Abstract
BACKGROUND/AIM Since the use of anaesthetics has the drawback of altering radiotracer distribution, preclinical positron emission tomography (PET) imaging findings of anaesthetised animals must be carefully handled. This study aimed at assessing the cerebral [18F]F-FDG uptake pattern in healthy Wistar rats under four different anaesthesia protocols using microPET/magnetic resonance imaging (MRI) examinations. MATERIALS AND METHODS Post-injection of 15±1.2 MBq of [18F]F-FDG, either while awake or during the isoflurane-induced incubation phase was applied. Prior to microPET/MRI imaging, one group of the rats was subjected to forane-only anaesthesia while the other group was anaesthetised with the co-administration of forane and dexmedetomidine/Dexdor® Results: While as for the whole brain it was the addition of dexmedetomidine/Dexdor® to the anaesthesia protocol that generated the differences between the radiotracer concentrations of the investigated groups, regarding the cortex, the [18F]F-FDG accumulation was rather affected by the way of incubation. To ensure the most consistent and highest uptake, forane-induced anaesthesia coupled with an awake uptake condition seemed to be most suitable method of anaesthetisation for cerebral metabolic assessment. Diminished whole brain and cortical tracer accumulation detected upon dexmedetomidine/Dexdor® administration highlights the significance of the mechanism of action of different anaesthetics on radiotracer pharmacokinetics. CONCLUSION Overall, the standardization of PET protocols is of utmost importance to avoid the confounding factors derived from anaesthesia.
Collapse
Affiliation(s)
- Zita Kepes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary;
| | - Viktória Arato
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Csaba Csikos
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eva Hegedus
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Regina Esze
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Nagy
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Istvan Joszai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklos Emri
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Istvan Kertesz
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Trencsenyi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
8
|
Arias-Valcayo F, Galve P, Manuel Udías J, Vaquero JJ, Desco M, Herraiz JL. Optimizing Point Source Tracking in Awake Rat PET Imaging: A Comprehensive Study of Motion Detection and Best Correction Conditions. APPLIED SCIENCES 2023; 13:12329. [DOI: 10.3390/app132212329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Preclinical PET animal studies require immobilization of the animal, typically accomplished through the administration of anesthesia, which may affect the radiotracer biodistribution. The use of 18F point sources attached to the rat head is one of the most promising methods for motion compensation in awake rat PET studies. However, the presence of radioactive markers may degrade image quality. In this study, we aimed to investigate the most favorable conditions for preclinical PET studies using awake rats with attached point sources. Firstly, we investigate the optimal activity conditions for the markers and rat-injected tracer using Monte Carlo simulations to determine the parameters of maximum detectability without compromising image quality. Additionally, we scrutinize the impact of delayed window correction for random events on marker detectability and overall image quality within these studies. Secondly, we present a method designed to mitigate the influence of rapid rat movements, which resulted in a medium loss of events of around 30%, primarily observed during the initial phase of the data acquisition. We validated our study with PET acquisitions from an awake rat within the acceptable conditions of activity and motion compensation parameters. This acquisition revealed an 8% reduction in resolution compared to a sedated animal, along with a 6% decrease in signal-to-noise ratio (SNR). These outcomes affirm the viability of our method for conducting awake preclinical brain studies.
Collapse
Affiliation(s)
- Fernando Arias-Valcayo
- Grupo de Física Nuclear, Departamento de Estructura de la Materia, Física Térmica y Electrónica & Instituto de Física de Partículas y del Cosmos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Pablo Galve
- Grupo de Física Nuclear, Departamento de Estructura de la Materia, Física Térmica y Electrónica & Instituto de Física de Partículas y del Cosmos, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
- Paris Cardiovascular Research Center, Inserm UMR970, Université de Paris, 75015 Paris, France
| | - Jose Manuel Udías
- Grupo de Física Nuclear, Departamento de Estructura de la Materia, Física Térmica y Electrónica & Instituto de Física de Partículas y del Cosmos, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| | - Juan José Vaquero
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, 28911 Leganés, Spain
- Instituto de Investigación Sanitaria del Hospital Gregorio Marañón, Unidad de Medicina y Cirugía Experimental, 28009 Madrid, Spain
| | - Manuel Desco
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, 28911 Leganés, Spain
- Instituto de Investigación Sanitaria del Hospital Gregorio Marañón, Unidad de Medicina y Cirugía Experimental, 28009 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
- CIBER de Salud Mental Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Joaquín L. Herraiz
- Grupo de Física Nuclear, Departamento de Estructura de la Materia, Física Térmica y Electrónica & Instituto de Física de Partículas y del Cosmos, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Del Hospital Clínico San Carlos (IdISSC), Ciudad Universitaria, 28040 Madrid, Spain
| |
Collapse
|
9
|
Miranda A, Bertoglio D, De Weerdt C, Staelens S, Verhaeghe J. Isoflurane and ketamine-xylazine modify pharmacokinetics of [ 18F]SynVesT-1 in the mouse brain. J Cereb Blood Flow Metab 2023; 43:1612-1624. [PMID: 37113068 PMCID: PMC10414002 DOI: 10.1177/0271678x231173185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/26/2023] [Indexed: 04/29/2023]
Abstract
We investigated the effect of isoflurane and ketamine-xylazine anesthesia on the positron emission tomography (PET) tracer [18F]SynVesT-1 in the mouse brain. [18F]SynVesT-1 PET scans were performed in C57BL/6J mice in five conditions: isoflurane anesthesia (ANISO), ketamine-xylazine (ANKX), awake freely moving (AW), awake followed by isoflurane administration (AW/ANISO) or followed by ketamine-xylazine (AW/ANKX) 20 min post tracer injection. ANISO, ANKX and AW scans were also performed in mice administered with levetiracetam (LEV, 200 mg/kg) to assess non-displaceable binding. Metabolite analysis was performed in ANISO, ANKX and AW mice. Finally, in vivo autoradiography in ANISO, ANKX and AW mice at 30 min post-injection was performed for validation. Kinetic modeling, with a metabolite corrected image derived input function, was performed to calculate total and non-displaceable volume of distribution (VT(IDIF)). VT(IDIF) was higher in ANISO compared to AW (p < 0.0001) while VT(IDIF) in ANKX was lower compared with AW (p < 0.0001). Non-displaceable VT(IDIF) was significantly different between ANISO and AW, but not between ANKX and AW. Change in the TAC washout was observed after administration of either isoflurane or ketamine-xylazine. Observed changes in tracer kinetics and volume of distribution might be explained by physiological changes due to anesthesia, as well as by induced cellular effects.
Collapse
Affiliation(s)
- Alan Miranda
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Daniele Bertoglio
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Caro De Weerdt
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
10
|
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: 0.5] [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
|
11
|
Yu Q, Ouyang M, Detre J, Kang H, Hu D, Hong B, Fang F, Peng Y, Huang H. Infant brain regional cerebral blood flow increases supporting emergence of the default-mode network. eLife 2023; 12:e78397. [PMID: 36693116 PMCID: PMC9873253 DOI: 10.7554/elife.78397] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
Human infancy is characterized by most rapid regional cerebral blood flow (rCBF) increases across lifespan and emergence of a fundamental brain system default-mode network (DMN). However, how infant rCBF changes spatiotemporally across the brain and how the rCBF increase supports emergence of functional networks such as DMN remains unknown. Here, by acquiring cutting-edge multi-modal MRI including pseudo-continuous arterial-spin-labeled perfusion MRI and resting-state functional MRI of 48 infants cross-sectionally, we elucidated unprecedented 4D spatiotemporal infant rCBF framework and region-specific physiology-function coupling across infancy. We found that faster rCBF increases in the DMN than visual and sensorimotor networks. We also found strongly coupled increases of rCBF and network strength specifically in the DMN, suggesting faster local blood flow increase to meet extraneuronal metabolic demands in the DMN maturation. These results offer insights into the physiological mechanism of brain functional network emergence and have important implications in altered network maturation in brain disorders.
Collapse
Affiliation(s)
- Qinlin Yu
- Department of Radiology, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Radiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Minhui Ouyang
- Department of Radiology, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Radiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - John Detre
- Department of Radiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Neurology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Huiying Kang
- Department of Radiology, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Radiology, Beijing Children’s Hospital, Capital Medical UniversityBeijingChina
| | - Di Hu
- Department of Radiology, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Radiology, Beijing Children’s Hospital, Capital Medical UniversityBeijingChina
| | - Bo Hong
- Department of Biomedical Engineering, Tsinghua UniversityBeijingChina
| | - Fang Fang
- School of Psychological and Cognitive Sciences, Peking UniversityBeijingChina
| | - Yun Peng
- Department of Radiology, Beijing Children’s Hospital, Capital Medical UniversityBeijingChina
| | - Hao Huang
- Department of Radiology, Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Radiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
12
|
Dickie BR, Jin T, Wang P, Hinz R, Harris W, Boutin H, Parker GJ, Parkes LM, Matthews JC. Quantitative kinetic modelling and mapping of cerebral glucose transport and metabolism using glucoCESL MRI. J Cereb Blood Flow Metab 2022; 42:2066-2079. [PMID: 35748031 PMCID: PMC9580170 DOI: 10.1177/0271678x221108841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chemical-exchange spin-lock (CESL) MRI can map regional uptake and utilisation of glucose in the brain at high spatial resolution (i.e sub 0.2 mm3 voxels). We propose two quantitative kinetic models to describe glucose-induced changes in tissue R1ρ and apply them to glucoCESL MRI data acquired in tumour-bearing and healthy rats. When assuming glucose transport is saturable, the maximal transport capacity (Tmax) measured in normal tissue was 3.2 ± 0.6 µmol/min/mL, the half saturation constant (Kt) was 8.8 ± 2.2 mM, the metabolic rate of glucose consumption (MRglc) was 0.21 ± 0.13 µmol/min/mL, and the cerebral blood volume (vb) was 0.006 ± 0.005 mL/mL. Values in tumour were: Tmax = 7.1 ± 2.7 µmol/min/mL, Kt = 14 ± 1.7 mM, MRglc = 0.22 ± 0.09 µmol/min/mL, vb = 0.030 ± 0.035 mL/mL. Tmax and Kt were significantly higher in tumour tissue than normal tissue (p = 0.006 and p = 0.011, respectively). When assuming glucose uptake also occurs via free diffusion, the free diffusion rate (kd) was 0.061 ± 0.017 mL/min/mL in normal tissue and 0.12 ± 0.042 mL/min/mL in tumour. These parameter estimates agree well with literature values obtained using other approaches (e.g. NMR spectroscopy).
Collapse
Affiliation(s)
- Ben R Dickie
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Tao Jin
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ping Wang
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rainer Hinz
- Division of Informatics, Imaging, and Data Science, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - William Harris
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Geoff Jm Parker
- Bioxydyn Limited, Manchester, UK.,Centre for Medical Image Computing, Department of Medical Physics & Biomedical Engineering and Department of Neuroinflammation, University College London, London, UK
| | - Laura M Parkes
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Julian C Matthews
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
13
|
Chaney R, Garnier P, Quirié A, Martin A, Prigent-Tessier A, Marie C. Region-Dependent Increase of Cerebral Blood Flow During Electrically Induced Contraction of the Hindlimbs in Rats. Front Physiol 2022; 13:811118. [PMID: 35492591 PMCID: PMC9040888 DOI: 10.3389/fphys.2022.811118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/04/2022] [Indexed: 11/30/2022] Open
Abstract
Elevation of cerebral blood flow (CBF) may contribute to the cerebral benefits of the regular practice of physical exercise. Surprisingly, while electrically induced contraction of a large muscular mass is a potential substitute for physical exercise to improve cognition, its effect on CBF remains to be investigated. Therefore, the present study investigated CBF in the cortical area representing the hindlimb, the hippocampus and the prefrontal cortex in the same anesthetized rats subjected to either acute (30 min) or chronic (30 min for 7 days) electrically induced bilateral hindlimb contraction. While CBF in the cortical area representing the hindlimb was assessed from both laser doppler flowmetry (LDFCBF) and changes in p-eNOSSer1177 levels (p-eNOSCBF), CBF was evaluated only from changes in p-eNOSSer1177 levels in the hippocampus and the prefrontal cortex. The contribution of increased cardiac output and increased neuronal activity to CBF changes were examined. Stimulation was associated with tachycardia and no change in arterial blood pressure. It increased LDFCBF with a time- and intensity-dependent manner as well as p-eNOSCBF in the area representing the hindlimb. By contrast, p-eNOSCBF was unchanged in the two other regions. The augmentation of LDFCBF was partially reduced by atenolol (a ß1 receptor antagonist) and not reproduced by the administration of dobutamine (a ß1 receptor agonist). Levels of c-fos as a marker of neuronal activation selectively increased in the area representing the hindlimb. In conclusion, electrically induced bilateral hindlimb contraction selectively increased CBF in the cortical area representing the stimulated muscles as a result of neuronal hyperactivity and increased cardiac output. The absence of CBF changes in cognition-related brain regions does not support flow-dependent neuroplasticity in the pro-cognitive effect of electrically induced contraction of a large muscular mass.
Collapse
Affiliation(s)
- Remi Chaney
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| | - Philippe Garnier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France.,Département Génie Biologique, IUT, Dijon, France
| | - Aurore Quirié
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| | - Alain Martin
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences du Sport, Dijon, France
| | - Anne Prigent-Tessier
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| | - Christine Marie
- INSERM UMR1093-CAPS, Université Bourgogne Franche-Comté, UFR des Sciences de Santé, Dijon, France
| |
Collapse
|
14
|
Miranda A, Bertoglio D, Stroobants S, Staelens S, Verhaeghe J. Translation of Preclinical PET Imaging Findings: Challenges and Motion Correction to Overcome the Confounding Effect of Anesthetics. Front Med (Lausanne) 2021; 8:753977. [PMID: 34746189 PMCID: PMC8569248 DOI: 10.3389/fmed.2021.753977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
Preclinical brain positron emission tomography (PET) in animals is performed using anesthesia to avoid movement during the PET scan. In contrast, brain PET scans in humans are typically performed in the awake subject. Anesthesia is therefore one of the principal limitations in the translation of preclinical brain PET to the clinic. This review summarizes the available literature supporting the confounding effect of anesthesia on several PET tracers for neuroscience in preclinical small animal scans. In a second part, we present the state-of-the-art methodologies to circumvent this limitation to increase the translational significance of preclinical research, with an emphasis on motion correction methods. Several motion tracking systems compatible with preclinical scanners have been developed, each one with its advantages and limitations. These systems and the novel experimental setups they can bring to preclinical brain PET research are reviewed here. While technical advances have been made in this field, and practical implementations have been demonstrated, the technique should become more readily available to research centers to allow for a wider adoption of the motion correction technique for brain research.
Collapse
Affiliation(s)
- Alan Miranda
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Daniele Bertoglio
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
- University Hospital Antwerp, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| |
Collapse
|