1
|
Hernández-Martín N, Martínez MG, Bascuñana P, Fernández de la Rosa R, García-García L, Gómez F, Solas M, Martín ED, Pozo MA. Astrocytic Ca 2+ activation by chemogenetics mitigates the effect of kainic acid-induced excitotoxicity on the hippocampus. Glia 2024; 72:2217-2230. [PMID: 39188024 DOI: 10.1002/glia.24607] [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: 05/29/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
Astrocytes play a multifaceted role regulating brain glucose metabolism, ion homeostasis, neurotransmitters clearance, and water dynamics being essential in supporting synaptic function. Under different pathological conditions such as brain stroke, epilepsy, and neurodegenerative disorders, excitotoxicity plays a crucial role, however, the contribution of astrocytic activity in protecting neurons from excitotoxicity-induced damage is yet to be fully understood. In this work, we evaluated the effect of astrocytic activation by Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) on brain glucose metabolism in wild-type (WT) mice, and we investigated the effects of sustained astrocyte activation following an insult induced by intrahippocampal (iHPC) kainic acid (KA) injection using 2-deoxy-2-[18F]-fluoro-D-glucose (18F-FDG) positron emission tomography (PET) imaging, along with behavioral test, nuclear magnetic resonance (NMR) spectroscopy and histochemistry. Astrocytic Ca2+ activation increased the 18F-FDG uptake, but this effect was not found when the study was performed in knock out mice for type-2 inositol 1,4,5-trisphosphate receptor (Ip3r2-/-) nor in floxed mice to abolish glucose transporter 1 (GLUT1) expression in hippocampal astrocytes (GLUT1ΔGFAP). Sustained astrocyte activation after KA injection reversed the brain glucose hypometabolism, restored hippocampal function, prevented neuronal death, and increased hippocampal GABA levels. The findings of our study indicate that astrocytic GLUT1 function is crucial for regulating brain glucose metabolism. Astrocytic Ca2+ activation has been shown to promote adaptive changes that significantly contribute to mitigating the effects of KA-induced damage. This evidence suggests a protective role of activated astrocytes against KA-induced excitotoxicity.
Collapse
Affiliation(s)
- Nira Hernández-Martín
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | | | - Pablo Bascuñana
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Rubén Fernández de la Rosa
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Bioimac, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis García-García
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisca Gómez
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Maite Solas
- Facultad de Farmacia, Universidad de Navarra, Pamplona, Spain
| | | | - Miguel A Pozo
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
2
|
Han Y, Liu H, Li Y, Liu Z. B-Glycine as a marker for β cell imaging and β cell mass evaluation. Eur J Nucl Med Mol Imaging 2024; 51:2558-2568. [PMID: 38632133 DOI: 10.1007/s00259-024-06712-y] [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: 12/24/2023] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE β cell mass (BCM) and function are essential to the diagnosis and therapy of diabetes. Diabetic patients serve β cell loss is, and damage of β cells leads to severe insulin deficiency. Our understanding of the role of BCM in diabetes progression is extremely limited by lacking efficient methods to evaluate BCM in vivo. In vitro methods of labeling islets, including loading of contrast reagent or integration of exogenous biomarker, require artificial manipulation on islets, of which the clinical application is limited. Imaging methods targeting endogenous biomarkers may solve the above problems. However, traditional reagents targeting GLP-1R and VAMT2 result in a high background of adjacent tissues, complicating the identification of pancreatic signals. Here, we report a non-invasive and quantitative imaging technique by using radiolabeled glycine mimics ([18F]FBG, a boron-trifluoride derivative of glycine) to assay islet function and monitor BCM changes in living animals. METHODS Glycine derivatives, FBG, FBSa, 2Me-FBG, 3Me-FBG, were successfully synthesized and labeled with 18F. Specificity of glycine derivatives were characterized by in vitro experiment. PET imaging and biodistribution studies were performed in animal models carring GLYT over-expressed cells. In vivo evaluation of BCM with [18F]FBG were performed in STZ (streptozocin) induced T1D (type 1 diabetes) models. RESULTS GLYT responds to excess blood glycine levels and transports glycine into islet cells to maintain the activity of the glycine receptor (GLYR). Best PET imaging condition was 80 min after given a total of 240 ~ 250 nmol imaging reagent (a mixture of [18F]FBG and natural glycine) intravenously. [18F]FBG can detect both endogenous and exogenous islets clearly in vivo. When applied to STZ induced T1D mouse models, total uptake of [18F]FBG in the pancreas exhibited a linear correlation with survival BCM. CONCLUSION [18F]FBG targeting the endogenous glycine transporter (GLYT), which is highly expressed on islet cells, avoiding extra modification on islet cells. Meanwhile the highly restricted expression pattern of GLYT excluded the background in adjacent tissues. This [18F]FBG-based imaging technique provides a non-invasive method to quantify BCM in vivo, implying a new evaluation index for diabetic assessment.
Collapse
Affiliation(s)
- Yuxiang Han
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University, Beijing, 100871, China
| | - Hui Liu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University, Beijing, 100871, China
| | - Yimin Li
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University, Beijing, 100871, China
| | - Zhibo Liu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Peking University, Beijing, 100871, China.
- Center for Life Sciences, Peking University-Tsinghua University, Peking University, Beijing, 100871, China.
| |
Collapse
|
3
|
Zabegalov KN, Costa FV, Kolesnikova TO, de Abreu MS, Petersen EV, Yenkoyan KB, Kalueff AV. Can we gain translational insights into the functional roles of cerebral cortex from acortical rodent and naturally acortical zebrafish models? Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110964. [PMID: 38354895 DOI: 10.1016/j.pnpbp.2024.110964] [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: 11/12/2023] [Revised: 01/11/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Cerebral cortex is found only in mammals and is particularly prominent and developed in humans. Various rodent models with fully or partially ablated cortex are commonly used to probe the role of cortex in brain functions and its multiple subcortical projections, including pallium, thalamus and the limbic system. Various rodent models are traditionally used to study the role of cortex in brain functions. A small teleost fish, the zebrafish (Danio rerio), has gained popularity in neuroscience research, and albeit (like other fishes) lacking cortex, its brain performs well some key functions (e.g., memory, consciousness and motivation) with complex, context-specific and well-defined behaviors. Can rodent and zebrafish models help generate insights into the role of cortex in brain functions, and dissect its cortex-specific (vs. non-cortical) functions? To address this conceptual question, here we evaluate brain functionality in intact vs. decorticated rodents and further compare it in the zebrafish, a naturally occurring acortical species. Overall, comparing cortical and acortical rodent models with naturally acortical zebrafish reveals both distinct and overlapping contributions of neocortex and 'precortical' zebrafish telencephalic regions to higher brain functions. Albeit morphologically different, mammalian neocortex and fish pallium may possess more functional similarities than it is presently recognized, calling for further integrative research utilizing both cortical and decorticated/acortical vertebrate model organisms.
Collapse
Affiliation(s)
- Konstantin N Zabegalov
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan; Life Improvement by Future Technologies (LIFT) Center, LLC, Moscow, Russia
| | - Fabiano V Costa
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia
| | | | | | | | - Konstantin B Yenkoyan
- Neuroscience Laboratory, COBRAIN Center, Yerevan State Medical University named after M. Heratsi, Yerevan, Armenia; Department of Biochemistry, Yerevan State Medical University named after M. Heratsi, Yerevan, Armenia.
| | - Allan V Kalueff
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
| |
Collapse
|
4
|
Wei H, Jiang H, Zhou Y, Liu L, Ma W, Ni S, Zhou C, Ji X. Cerebral venous congestion alters CNS homeostatic plasticity, evoking tinnitus-like behavior. Cell Biosci 2024; 14:47. [PMID: 38594782 PMCID: PMC11003147 DOI: 10.1186/s13578-024-01221-9] [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: 07/11/2023] [Accepted: 03/19/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Brain function and neuronal activity depend on a constant supply of blood from the cerebral circulation. The cerebral venous system (CVS) contains approximately 70% of the total cerebral blood volume; similar to the cerebral arterial system, the CVS plays a prominent role in the maintenance of central nervous system (CNS) homeostasis. Impaired venous autoregulation, which can appear in forms such as cerebral venous congestion, may lead to metabolic abnormalities in the brain, causing severe cerebral functional defects and even chronic tinnitus. However, the role of cerebral venous congestion in the progression of tinnitus is underrecognized, and its pathophysiology is still incompletely understood. This study elucidated the specific pathogenetic role of cerebral venous congestion in the onset and persistence of tinnitus and the possible neurophysiological mechanisms. RESULTS We found that a rat model of cerebral venous congestion exhibited tinnitus-like behavioral manifestations at 14 days postoperatively; from that point onward, they showed signs of persistent tinnitus without significant hearing impairment. Subsequent neuroimaging and neurochemical findings showed CNS homeostatic plasticity disturbance in rats with cerebral venous congestion, reflected in increased neural metabolic activity, ultrastructural synaptic changes, upregulated synaptic efficacy, reduced inhibitory synaptic transmission (due to GABA deficiency), and elevated expression of neuroplasticity-related proteins in central auditory and extra-auditory pathways. CONCLUSION Collectively, our data suggest that alternations in CNS homeostatic plasticity may play a vital role in tinnitus pathology caused by cerebral venous congestion. These findings provide a new perspective on tinnitus related to cerebral venous congestion and may facilitate the development of precise interventions to interrupt its pathogenesis.
Collapse
Affiliation(s)
- Huimin Wei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Ministry of Science and Technology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Huimin Jiang
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Ministry of Science and Technology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Yifan Zhou
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Ministry of Science and Technology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Lu Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Ma
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Ministry of Science and Technology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China
| | - Shanshan Ni
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Ministry of Science and Technology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
- Department of Neurology, Wuqing Hospital of Traditional Chinese Medicine Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 301700, China.
| | - Chen Zhou
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Ministry of Science and Technology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
| | - Xunming Ji
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China.
- Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Ministry of Science and Technology, Capital Medical University, No.10 Xitoutiao, You An Men, Beijing, 100069, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
5
|
Ríos C, Aguirre-Aranda I, Avendaño-Estrada A, Ángel Ávila-Rodríguez M, Manjarrez-Marmolejo J, Franco-Pérez J, Islas-Cortez M, Ruiz-Diaz A, Méndez-Armenta M, Diaz-Ruiz A. Characterization of the anticonvulsant effect of dapsone on metabolic activity assessed by [ 18F]FDG -PET after kainic acid-induced status epilepticus in rats. Brain Res 2023; 1803:148227. [PMID: 36592802 DOI: 10.1016/j.brainres.2022.148227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Development of effective drugs for epilepsy are needed, as nearly 30 % of epileptic patients, are resistant to current treatments. This study is aimed to characterize the anticonvulsant effect of dapsone (DDS), in the kainic acid (KA)-induced Status Epilepticus (SE) by recording the brain metabolic activity with an [18F]FDG-PET analysis. METHODS Wistar rats received KA (10 mg/kg, i.p., single dose) to produce sustained seizures. [18F]FDG-PET and electroencephalographic (EEG) studies were then performed. DDS or vehicle were administered 30 min before KA. [18F]FDG uptake and EEG were evaluated at baseline, 2 and 25 h after KA injection. Likewise, caspase-8, 3 hippocampal activities and Fluoro-Jade B neuronal degeneration and Hematoxylin-eosin staining were measured 25 h after KA. RESULTS PET data evaluated at 2 h showed hyper-uptake of [18F]FDG in the control group, which was decreased by DDS. At 25 h, hypo-uptake was observed in the control group and higher values due to DDS effect. EEG spectral power was increased 2 h after KA administration in the control group during the generalized tonic-clonic seizures, which was reversed by DDS, correlated with [18F]FDG-PET uptake changes. The values of caspases-8 activity decreased 48 and 43 % vs control group in the groups treated with DDS (12.5 y 25 mg/kg respectively), likewise; caspase-3 activity diminished by 57 and 53 %. Fewer degenerated neurons were observed due to DDS treatments. CONCLUSIONS This study pinpoints the anticonvulsant therapeutic potential of DDS. Given its safety and effectiveness, DDS may be a viable alternative for patients with drug-resistant epilepsy.
Collapse
Affiliation(s)
- Camilo Ríos
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico; Laboratorio de Neurofarmacología Molecular, Universidad Autónoma Metropolitana Xochimilco,04960 Ciudad de México, Mexico
| | - Iñigo Aguirre-Aranda
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico
| | - Arturo Avendaño-Estrada
- Unidad Radiofarmacia-Ciclotrón, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico
| | - Miguel Ángel Ávila-Rodríguez
- Unidad Radiofarmacia-Ciclotrón, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510 Ciudad de México, Mexico
| | - Joaquín Manjarrez-Marmolejo
- Laboratorio de Fisiología de la Formación Reticular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269 Ciudad de México. Mexico
| | - Javier Franco-Pérez
- Laboratorio de Fisiología de la Formación Reticular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, 14269 Ciudad de México. Mexico
| | - Marcela Islas-Cortez
- Doctorado en Ciencias Químico Biológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Amairani Ruiz-Diaz
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico
| | - Marisela Méndez-Armenta
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico
| | - Araceli Diaz-Ruiz
- Departamento de Neuroquímica Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez,14269 Ciudad de México, Mexico.
| |
Collapse
|
6
|
Age-related reduction of hemispheric asymmetry by pigeons: A behavioral and FDG-PET imaging investigation of visual discrimination. Learn Behav 2022; 50:125-139. [DOI: 10.3758/s13420-021-00507-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 11/08/2022]
|
7
|
Wang C, Liu S, Liu F, Bhutta A, Patterson TA, Slikker W. Application of Nonhuman Primate Models in the Studies of Pediatric Anesthesia Neurotoxicity. Anesth Analg 2022; 134:1203-1214. [PMID: 35147575 DOI: 10.1213/ane.0000000000005926] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Numerous animal models have been used to study developmental neurotoxicity associated with short-term or prolonged exposure of common general anesthetics at clinically relevant concentrations. Pediatric anesthesia models using the nonhuman primate (NHP) may more accurately reflect the human condition because of their phylogenetic similarity to humans with regard to reproduction, development, neuroanatomy, and cognition. Although they are not as widely used as other animal models, the contribution of NHP models in the study of anesthetic-induced developmental neurotoxicity has been essential. In this review, we discuss how neonatal NHP animals have been used for modeling pediatric anesthetic exposure; how NHPs have addressed key data gaps and application of the NHP model for the studies of general anesthetic-induced developmental neurotoxicity. The appropriate application and evaluation of the NHP model in the study of general anesthetic-induced developmental neurotoxicity have played a key role in enhancing the understanding and awareness of the potential neurotoxicity associated with pediatric general anesthetics.
Collapse
Affiliation(s)
- Cheng Wang
- From the Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - Shuliang Liu
- From the Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - Fang Liu
- From the Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - Adnan Bhutta
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Tucker A Patterson
- Office of the Director, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| | - William Slikker
- Office of the Director, National Center for Toxicological Research/FDA, Jefferson, Arkansas
| |
Collapse
|
8
|
Jahreis I, Bascuñana P, Ross TL, Bankstahl JP, Bankstahl M. Choice of anesthesia and data analysis method strongly increases sensitivity of 18F-FDG PET imaging during experimental epileptogenesis. PLoS One 2021; 16:e0260482. [PMID: 34818362 PMCID: PMC8612569 DOI: 10.1371/journal.pone.0260482] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose Alterations in brain glucose metabolism detected by 2-deoxy-2-[18F]-fluoro-D-glucose (18F-FDG) positron emission tomography (PET) may serve as an early predictive biomarker and treatment target for epileptogenesis. Here, we aimed to investigate changes in cerebral glucose metabolism before induction of epileptogenesis, during epileptogenesis as well as during chronic epilepsy. As anesthesia is usually unavoidable for preclinical PET imaging and influences the distribution of the radiotracer, four different protocols were compared. Procedures We investigated 18F-FDG uptake phase in conscious rats followed by a static scan as well as dynamic scans under continuous isoflurane, medetomidine-midazolam-fentanyl (MMF), or propofol anesthesia. Furthermore, we applied different analysis approaches: atlas-based regional analysis, statistical parametric mapping, and kinetic analysis. Results At baseline and compared to uptake in conscious rats, isoflurane and propofol anesthesia resulted in decreased cortical 18F-FDG uptake while MMF anesthesia led to a globally decreased tracer uptake. During epileptogenesis, MMF anesthesia was clearly best distinctive for visualization of prominently increased glucometabolism in epilepsy-related brain areas. Kinetic modeling further increased sensitivity, particularly for continuous isoflurane anesthesia. During chronic epilepsy, hypometabolism affecting more or less the whole brain was detectable with all protocols. Conclusion This study reveals evaluation of anesthesia protocols for preclinical 18F-FDG PET imaging as a critical step in the study design. Together with an appropriate data analysis workflow, the chosen anesthesia protocol may uncover otherwise concealed disease-associated regional glucometabolic changes.
Collapse
Affiliation(s)
- Ina Jahreis
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias L. Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Jens P. Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- * E-mail:
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
9
|
Bandopadhyay R, Singh T, Ghoneim MM, Alshehri S, Angelopoulou E, Paudel YN, Piperi C, Ahmad J, Alhakamy NA, Alfaleh MA, Mishra A. Recent Developments in Diagnosis of Epilepsy: Scope of MicroRNA and Technological Advancements. BIOLOGY 2021; 10:1097. [PMID: 34827090 PMCID: PMC8615191 DOI: 10.3390/biology10111097] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/18/2022]
Abstract
Epilepsy is one of the most common neurological disorders, characterized by recurrent seizures, resulting from abnormally synchronized episodic neuronal discharges. Around 70 million people worldwide are suffering from epilepsy. The available antiepileptic medications are capable of controlling seizures in around 60-70% of patients, while the rest remain refractory. Poor seizure control is often associated with neuro-psychiatric comorbidities, mainly including memory impairment, depression, psychosis, neurodegeneration, motor impairment, neuroendocrine dysfunction, etc., resulting in poor prognosis. Effective treatment relies on early and correct detection of epileptic foci. Although there are currently a few well-established diagnostic techniques for epilepsy, they lack accuracy and cannot be applied to patients who are unsupportive or harbor metallic implants. Since a single test result from one of these techniques does not provide complete information about the epileptic foci, it is necessary to develop novel diagnostic tools. Herein, we provide a comprehensive overview of the current diagnostic tools of epilepsy, including electroencephalography (EEG) as well as structural and functional neuroimaging. We further discuss recent trends and advances in the diagnosis of epilepsy that will enable more effective diagnosis and clinical management of patients.
Collapse
Affiliation(s)
- Ritam Bandopadhyay
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA;
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.A.); (C.P.)
| | - Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia;
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.A.); (C.P.)
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia;
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
| | - Mohamed A. Alfaleh
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Awanish Mishra
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)—Guwahati, Changsari, Guwahati 781101, Assam, India
| |
Collapse
|
10
|
Mouly AM, Bouillot C, Costes N, Zimmer L, Ravel N, Litaudon P. PET Metabolic Imaging of Time-Dependent Reorganization of Olfactory Cued Fear Memory Networks in Rats. Cereb Cortex 2021; 32:2717-2728. [PMID: 34668524 DOI: 10.1093/cercor/bhab376] [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: 07/22/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022] Open
Abstract
Memory consolidation involves reorganization at both the synaptic and system levels. The latter involves gradual reorganization of the brain regions that support memory and has been mostly highlighted using hippocampal-dependent tasks. The standard memory consolidation model posits that the hippocampus becomes gradually less important over time in favor of neocortical regions. In contrast, this reorganization of circuits in amygdala-dependent tasks has been less investigated. Moreover, this question has been addressed using primarily lesion or cellular imaging approaches thus precluding the comparison of recent and remote memory networks in the same animals. To overcome this limitation, we used microPET imaging to characterize, in the same animals, the networks activated during the recall of a recent versus remote memory in an olfactory cued fear conditioning paradigm. The data highlighted the drastic difference between the extents of the two networks. Indeed, although the recall of a recent odor fear memory activates a large network of structures spanning from the prefrontal cortex to the cerebellum, significant activations during remote memory retrieval are limited to the piriform cortex. These results strongly support the view that amygdala-dependent memories also undergo system-level reorganization, and that sensory cortical areas might participate in the long-term storage of emotional memories.
Collapse
Affiliation(s)
- Anne-Marie Mouly
- Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Université Claude Bernard Lyon 1, Bron Cedex 69675, France
| | | | | | - Luc Zimmer
- Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Université Claude Bernard Lyon 1, Bron Cedex 69675, France.,CERMEP-Life Imaging, Bron Cedex 69677, France.,Hospices Civils de Lyon, Lyon 69002, France
| | - Nadine Ravel
- Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Université Claude Bernard Lyon 1, Bron Cedex 69675, France
| | - Philippe Litaudon
- Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Université Claude Bernard Lyon 1, Bron Cedex 69675, France
| |
Collapse
|
11
|
Characterization of metabolic activity induced by kainic acid in adult rat whole brain at the early stage: A 18FDG-PET study. Brain Res 2021; 1769:147621. [PMID: 34403661 DOI: 10.1016/j.brainres.2021.147621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/08/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Brain metabolic processes are not fully characterized in the kainic acid (KA)-induced Status Epilepticus (KASE). Thus, we evaluated the usefulness of 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) as an experimental strategy to evaluate in vivo, in a non-invasive way, the glucose consumption in several brain regions, in a semi-quantitative study to compare and to correlate with data from electroencephalography and histology studies. METHODS Sixteen male Wistar rats underwent FDG-PET scans at basal state and after KA injection. FDG-PET images were normalized to an MRI-based atlas and segmented to locate regions. Standardized uptake values (SUV) were obtained at several time points. EEGs and cell viability by histological analysis, were also evaluated. RESULTS FDG-PET data showed changes in regions such as: amygdala, hippocampus, accumbens, entorhinal cortex, motor cortex and hypothalamus. Remarkably, hippocampal hypermetabolism was found (mean SUV = 2.66 ± 0.057) 2 h after KA administration, while hypometabolism at 24 h (mean SUV = 1.83 ± 0.056) vs basal values (mean SUV = 2.19 ± 0.057). EEG showed increased spectral power values 2 h post-KA administration. Hippocampal viable-cell counting 24 h after KA was decreased, while Fluoro-Jade B-positive cells were increased, as compared to control rats, coinciding with the hypometabolism detected in the same region by semi-quantitative FDG-PET at 24 h after KASE. CONCLUSIONS PET is suitable to measure metabolic brain changes in the rat model of status epilepticus induced by KA (KASE) at the first 24 h, compared to that of EEG; PET data may also be sensitive to cell viability.
Collapse
|
12
|
Hoffman CE, Parker WE, Rapoport BI, Zhao M, Ma H, Schwartz TH. Innovations in the Neurosurgical Management of Epilepsy. World Neurosurg 2020; 139:775-788. [PMID: 32689698 DOI: 10.1016/j.wneu.2020.03.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/02/2020] [Indexed: 10/23/2022]
Abstract
Technical limitations and clinical challenges have historically limited the diagnostic tools and treatment methods available for surgical approaches to the management of epilepsy. By contrast, recent technological innovations in several areas hold significant promise in improving outcomes and decreasing morbidity. We review innovations in the neurosurgical management of epilepsy in several areas, including wireless recording and stimulation systems (particularly responsive neurostimulation [NeuroPace]), conformal electrodes for high-resolution electrocorticography, robot-assisted stereotactic surgery, optogenetics and optical imaging methods, novel positron emission tomography ligands, and new applications of focused ultrasonography. Investigation into genetic causes of and susceptibilities to epilepsy has introduced a new era of precision medicine, enabling the understanding of cell signaling mechanisms underlying epileptic activity as well as patient-specific molecularly targeted treatment options. We discuss the emerging path to individualized treatment plans, predicted outcomes, and improved selection of effective interventions, on the basis of these developments.
Collapse
Affiliation(s)
- Caitlin E Hoffman
- Department of Neurological Surgery, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York, USA.
| | - Whitney E Parker
- Department of Neurological Surgery, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Benjamin I Rapoport
- Department of Neurological Surgery, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Mingrui Zhao
- Department of Neurological Surgery, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Hongtao Ma
- Department of Neurological Surgery, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Theodore H Schwartz
- Department of Neurological Surgery, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
13
|
Lee YH, Lee HT, Chen CL, Chang CH, Hsu CY, Shyu WC. Role of FOXC1 in regulating APSCs self-renewal via STI-1/PrP C signaling. Am J Cancer Res 2019; 9:6443-6465. [PMID: 31588228 PMCID: PMC6771253 DOI: 10.7150/thno.35619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/20/2019] [Indexed: 01/03/2023] Open
Abstract
Forkhead box protein C1 (FOXC1) is known to regulate developmental processes in the skull and brain. Methods: The unique multipotent arachnoid-pia stem cells (APSCs) isolated from human and mouse arachnoid-pia membranes of meninges were grown as 3D spheres and displayed a capacity for self-renewal. Additionally, APSCs also expressed the surface antigens as mesenchymal stem cells. By applying the FOXC1 knockout mice and mouse brain explants, signaling cascade of FOXC1-STI-1-PrPC was investigated to demonstrate the molecular regulatory pathway for APSCs self-renewal. Moreover, APSCs implantation in stroke model was also verified whether neurogenic property of APSCs could repair the ischemic insult of the stroke brain. Results: Activated FOXC1 regulated the proliferation of APSCs in a cell cycle-dependent manner, whereas FOXC1-mediated APSCs self-renewal was abolished in FOXC1 knockout mice (FOXC1-/- mice). Moreover, upregulation of STI-1 regulated by FOXC1 enhanced cell survival and self-renewal of APSCs through autocrine signaling of cellular prion protein (PrPC). Mouse brain explants STI-1 rescues the cortical phenotype in vitro and induces neurogenesis in the FOXC1 -/- mouse brain. Furthermore, administration of APSCs in ischemic brain restored the neuroglial microenvironment and improved neurological dysfunction. Conclusion: We identified a novel role for FOXC1 in the direct regulation of the STI-1-PrPC signaling pathway to promote cell proliferation and self-renewal of APSCs.
Collapse
|
14
|
Mapping Changes of Whole Brain Blood Flow in Rats with Myocardial Ischemia/Reperfusion Injury Assessed by Positron Emission Tomography. Curr Med Sci 2019; 39:653-657. [DOI: 10.1007/s11596-019-2087-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/06/2019] [Indexed: 01/02/2023]
|
15
|
PET imaging of metabolic changes after neural stem cells and GABA progenitor cells transplantation in a rat model of temporal lobe epilepsy. Eur J Nucl Med Mol Imaging 2019; 46:2392-2397. [PMID: 31338549 DOI: 10.1007/s00259-019-04408-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/18/2019] [Indexed: 10/26/2022]
Abstract
PURPOSE Stem cell transplantation is promising for temporal lobe epilepsy (TLE) treatment. This study aimed to use PET imaging for the investigation of dynamic metabolic changes after transplantation of human neural stem cells (NSCs) and human GABA progenitor cells (GPCs) in a rat model of TLE. METHODS 18F-FDG PET imaging, video-electroencephalography (EEG), whole-cell patch-clamp recordings and immunostaining were performed after transplantation of NSCs and GPCs. RESULTS PET imaging demonstrated that glucose metabolism was gradually improved in the NSCs group, but decreased in GPCs and the control. Video-EEG manifested that seizures were suppressed after NSCs or GPCs transplantation; whole-cell patch-clamp confirmed increased inhibitory response of GPC-derived cells; immunostaining studies verified that the transplanted NSCs and GPCs could survive, migrate and differentiate into mature neuronal subtypes. CONCLUSION 18F-FDG PET imaging could be a distinguishing approach for evaluation of dynamic glycolytic metabolic changes after transplantation of NSCs and GPCs in TLE. Whole-cell patch-clamp provides evidence for functional maturation and integration of transplanted stem cells within host circuits.
Collapse
|
16
|
Leiter I, Bascuñana P, Bengel FM, Bankstahl JP, Bankstahl M. Attenuation of epileptogenesis by 2-deoxy-d-glucose is accompanied by increased cerebral glucose supply, microglial activation and reduced astrocytosis. Neurobiol Dis 2019; 130:104510. [PMID: 31212069 DOI: 10.1016/j.nbd.2019.104510] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 06/02/2019] [Accepted: 06/14/2019] [Indexed: 02/03/2023] Open
Abstract
RATIONALE Neuronal excitability and brain energy homeostasis are strongly interconnected and evidence suggests that both become altered during epileptogenesis. Pharmacologic modulation of cerebral glucose metabolism might therefore exert anti-epileptogenic effects. Here we provide mechanistic insights into effects of the glycolytic inhibitor 2-deoxy-d-glucose (2-DG) on experimental epileptogenesis by longitudinal 2-deoxy-2[18F]fluoro-d-glucose positron emission tomography ([18F]FDG PET) and histology. METHODS To imitate epileptogenesis, 6 Hz-corneal kindling was performed in male NMRI mice by twice daily electrical stimulation for 21 days. Kindling groups were treated i.p. 1 min after each stimulation with either 250 mg/kg 2-DG (CoKi_2-DG) or saline (CoKi_vehicle). A separate group of unstimulated mice was treated with 2-DG (2-DG_only). Dynamic 60-min [18F]FDG PET/CT scans were acquired at baseline and interictally on days 10 and 17 of kindling. [18F]FDG uptake (%injected dose/cc) was quantified in predefined regions of interest (ROI) using a MRI-based brain atlas, and kinetic modelling was performed to evaluate glucose net influx rate Ki and glucose metabolic rate MRGlu. Furthermore, statistical parametric mapping (SPM) analysis was applied on kinetic brain maps. For histological evaluation, brain sections were stained for glucose transporter 1 (GLUT1), astrocytes, microglia, as well as dying neurons. RESULTS Post-stimulation 2-DG treatment attenuated early kindling progression, indicated by a reduction of fully-kindled mice, and a lower overall percentage of type five seizures. While 2-DG treatment alone led to globally increased Ki and MRGlu values at day 17, kindling progression per se did not influence glucose turnover. Kindling accompanied by 2-DG treatment, however, resulted in regionally elevated [18F]FDG uptake as well as increased Ki at days 10 and 17 compared both to baseline and to the 2-DG_only group. In hippocampus and thalamus, higher MRGlu values were found in the CoKi_2-DG vs. the CoKi_vehicle group at day 17. t maps resulting from SPM analysis generally confirmed the results of the ROI analysis, and additionally revealed increased MRGlu restricted to the ventral hippocampus when comparing the CoKi_2-DG and the 2-DG_only group both at days 10 and, more distinct, day 17. Immunohistochemical staining showed an attenuated kindling-induced regional activation of astrocytes in the CoKi_2-DG group. Interestingly, 2-DG treatment alone (and also in combination with kindling, but not kindling alone) led to increased microglial activation scores, whereas neither staining of GLUT1 nor of dying neurons revealed any differences to untreated controls. CONCLUSIONS Post-stimulation treatment with 2-DG exerts disease-modifying effects in the mouse 6 Hz corneal kindling model. The observed local increase in glucose supply and turnover, the alleviation of astroglial activation and the activation of microglia by 2-DG might contribute separately or in combination to its positive interference with epileptogenesis.
Collapse
Affiliation(s)
- Ina Leiter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover and Center for Systems Neuroscience, Bünteweg 17, 30559 Hannover, Germany; Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Frank Michael Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Peter Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover and Center for Systems Neuroscience, Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
17
|
Salerno M, Ferrer E, Wei S, Li X, Gao W, Ouellette D, Balanoff A, Vaska P. Behavioral neuroimaging in birds using PET. J Neurosci Methods 2019; 317:157-164. [PMID: 30710608 DOI: 10.1016/j.jneumeth.2019.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 01/17/2019] [Accepted: 01/29/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Birds comprise the most diverse group of terrestrial vertebrates. This success likely is related to the evolution of powered flight over 75 mya. Modern approaches for studying brain function, however, have yet to be fully adapted and applied to birds, especially as they relate to specific behaviors including flight. New method: We have developed a comprehensive set of in vivo experimental methods utilizing PET imaging with F-18 labeled fluorodeoxyglucose (FDG) to study regional changes in metabolism specifically related to flight, yet applicable to other behaviors as well. It incorporates approaches for selection of species, behavioral/imaging paradigm, animal preparation, radiotracer injection route, image quantification, and image analysis via an enhanced brain atlas. We also carried out preliminary modeling studies to better understand tracer kinetics. RESULTS The methods were successful in identifying brain regions statistically associated with flight using only 8 animals. Peak brain uptake of FDG between birds and rodents is similar despite much higher blood glucose levels in birds. We also confirmed that brain uptake of FDG steadily decreases after the initial peak and provide evidence that it may be related to greater dephosphorylation of FDG phosphate than that observed in mammals. Comparison with existing methods: FDG PET has been used in only a few studies of the bird brain. We introduce a new species, more realistic flight behavior, paired (test/retest) design, and improved quantification and analysis approaches. CONCLUSIONS The proposed imaging protocol is non-invasive yet sensitive to regional metabolic changes in the bird brain related to behavior.
Collapse
Affiliation(s)
- Michael Salerno
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Elizabeth Ferrer
- Department of Anatomical Sciences, Stony Brook University, Stony Brook, NY, 11794-8081, USA
| | - Shouyi Wei
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Xiang Li
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Wenrong Gao
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - David Ouellette
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Amy Balanoff
- Johns Hopkins University, Center for Functional Anatomy and Evolution, Baltimore, MD, 21205, USA; American Museum of Natural History, Division of Paleontology, New York, NY, 10024, USA.
| | - Paul Vaska
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-5230, USA; Department of Radiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
18
|
Neuroimaging Biomarkers of Experimental Epileptogenesis and Refractory Epilepsy. Int J Mol Sci 2019; 20:ijms20010220. [PMID: 30626103 PMCID: PMC6337422 DOI: 10.3390/ijms20010220] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 12/31/2018] [Accepted: 01/03/2019] [Indexed: 11/17/2022] Open
Abstract
This article provides an overview of neuroimaging biomarkers in experimental epileptogenesis and refractory epilepsy. Neuroimaging represents a gold standard and clinically translatable technique to identify neuropathological changes in epileptogenesis and longitudinally monitor its progression after a precipitating injury. Neuroimaging studies, along with molecular studies from animal models, have greatly improved our understanding of the neuropathology of epilepsy, such as the hallmark hippocampus sclerosis. Animal models are effective for differentiating the different stages of epileptogenesis. Neuroimaging in experimental epilepsy provides unique information about anatomic, functional, and metabolic alterations linked to epileptogenesis. Recently, several in vivo biomarkers for epileptogenesis have been investigated for characterizing neuronal loss, inflammation, blood-brain barrier alterations, changes in neurotransmitter density, neurovascular coupling, cerebral blood flow and volume, network connectivity, and metabolic activity in the brain. Magnetic resonance imaging (MRI) is a sensitive method for detecting structural and functional changes in the brain, especially to identify region-specific neuronal damage patterns in epilepsy. Positron emission tomography (PET) and single-photon emission computerized tomography are helpful to elucidate key functional alterations, especially in areas of brain metabolism and molecular patterns, and can help monitor pathology of epileptic disorders. Multimodal procedures such as PET-MRI integrated systems are desired for refractory epilepsy. Validated biomarkers are warranted for early identification of people at risk for epilepsy and monitoring of the progression of medical interventions.
Collapse
|
19
|
Schöberl F, Irving S, Pradhan C, Bardins S, Trapp C, Schneider E, Kugler G, Bartenstein P, Dieterich M, Brandt T, Zwergal A. Prolonged allocentric navigation deficits indicate hippocampal damage in TGA. Neurology 2018; 92:e234-e243. [DOI: 10.1212/wnl.0000000000006779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/17/2018] [Indexed: 12/20/2022] Open
Abstract
ObjectiveTo investigate long-term recovery of allocentric and egocentric spatial orientation as a sensitive marker for hippocampal and extrahippocampal network function in transient global amnesia (TGA).MethodsA group of 18 patients with TGA performed an established real-space navigation paradigm, requiring allo- and egocentric spatial orientation abilities, 3 days (postacute stage) and 3 months (follow-up) after symptom onset. Visual exploration behavior and navigation strategy were documented by a gaze-controlled, head-fixed camera. Allo- and egocentric spatial orientation performance was compared to that of 12 age-matched healthy controls. Navigation-induced brain activations were measured using [18F]-fluorodeoxyglucose-PET in a subgroup of 8 patients in the postacute stage and compared to those of the controls.ResultsIn the postacute stage, the patients navigated worse and had higher error rates than controls in allocentric (p = 0.002), but not in egocentric, route planning (p = 0.30), despite complete recovery of verbal (p = 0.58) and figural memory (p = 0.11). Until follow-up, allocentric navigation deficits improved, but higher error rates and reduced use of shortcuts persisted (p < 0.0001). Patients still exhibited relatively more fixations of unique landmarks during follow-up (p = 0.05). PET measurements during the postacute stage showed increased navigation-induced brain activations in the right hippocampus, bilateral retrosplenial, parietal, and mesiofrontal cortices, and cerebellar dentate nucleus in patients compared to controls (p < 0.005).ConclusionsPatients with TGA show selective and prolonged deficits of allocentric spatial orientation. Activations in right hippocampal and extrahippocampal hubs of the cerebral navigation network functionally substitute for the deficit in creating and updating the internal cognitive map in TGA.
Collapse
|
20
|
Yu S, Xu H, Chi X, Wei L, Cheng Q, Yang Y, Zhou C, Ding F. 2-(4-Methoxyphenyl)ethyl-2-Acetamido-2-deoxy-β-d-pyranoside (A Salidroside Analog) Confers Neuroprotection with a Wide Therapeutic Window by Regulating Local Glucose Metabolism in a Rat Model of Cerebral Ischemic Injury. Neuroscience 2018; 391:60-72. [PMID: 30223020 DOI: 10.1016/j.neuroscience.2018.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/02/2023]
Abstract
2-(4-Methoxyphenyl)ethyl-2-acetamido-2-deoxy-β-d-pyranoside (salidroside analog-4g, SalA-4g), has shown neuroprotective prospects for the treatment of ischemic stroke. However, the dose-response and time window study for SalA-4g, and the mechanism of SalA-4g-mediated neuroprotection remain unclear. Here, we systematically investigated the therapeutic time window and dosage of SalA-4g in permanent focal cerebral ischemia in rats. SalA-4g dose-dependently improved stroke outcome. Either pre-treatment or post-treatment of SalA-4g exhibited notable neuroprotection, and maintained for up to 6 h after ischemia onset. Moreover, significant neurological functional recovery was found after SalA-4g administration in long-term functional assays. Further studies suggested that SalA-4g ameliorated neuronal cell death, elevated local glucose metabolism and enhanced the expression level of glucose transporter 1 and 3 in the ipsilateral cortex and striatum. We suggest that data of this study are critical in exploring the clinical application prospects of SalA-4g for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Shu Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Hui Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Xiaojing Chi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Li Wei
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Qiong Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Chun Zhou
- Department of Neurology, Affiliated Hospital of Nantong University, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, 20 Xisi Road, Nantong, JS 226001, PR China.
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China; Department of Neurology, Affiliated Hospital of Nantong University, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, 20 Xisi Road, Nantong, JS 226001, PR China.
| |
Collapse
|
21
|
Zhang X, Yin Q, Berridge M, Wang C. Application of molecular imaging technology in neurotoxicology research. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2018; 36:113-124. [PMID: 30199343 DOI: 10.1080/10590501.2018.1492200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Molecular imaging has been widely applied in preclinical research. Among these new molecular imaging modalities, microPET imaging can be utilized as a very powerful tool that can obtain the measurements of multiple biological processes in various organs repeatedly in a same subject. This review discusses how this new approach provides noninvasive biomarker for neurotoxicology research and summarizes microPET findings with multiple radiotracers on the variety of neurotoxicity induced by toxic agents in both the rodent and the nonhuman primate brain.
Collapse
Affiliation(s)
- Xuan Zhang
- a Division of Neurotoxicology , U.S. Food and Drug Administration, National Center for Toxicological Research , Jefferson , Arkansas , USA
| | - Qi Yin
- a Division of Neurotoxicology , U.S. Food and Drug Administration, National Center for Toxicological Research , Jefferson , Arkansas , USA
| | - Marc Berridge
- b 3D Imaging, LLC, University of Arkansas for Medical Sciences , Little Rock , Arkansas , USA
| | - Che Wang
- a Division of Neurotoxicology , U.S. Food and Drug Administration, National Center for Toxicological Research , Jefferson , Arkansas , USA
| |
Collapse
|
22
|
Non-imaged based method for matching brains in a common anatomical space for cellular imagery. J Neurosci Methods 2018; 304:136-145. [DOI: 10.1016/j.jneumeth.2018.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/06/2018] [Accepted: 04/07/2018] [Indexed: 11/18/2022]
|
23
|
Sperry MM, Kartha S, Granquist EJ, Winkelstein BA. Inter-subject FDG PET Brain Networks Exhibit Multi-scale Community Structure with Different Normalization Techniques. Ann Biomed Eng 2018; 46:1001-1012. [PMID: 29644496 PMCID: PMC5980783 DOI: 10.1007/s10439-018-2022-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/30/2018] [Indexed: 12/18/2022]
Abstract
Inter-subject networks are used to model correlations between brain regions and are particularly useful for metabolic imaging techniques, like 18F-2-deoxy-2-(18F)fluoro-D-glucose (FDG) positron emission tomography (PET). Since FDG PET typically produces a single image, correlations cannot be calculated over time. Little focus has been placed on the basic properties of inter-subject networks and if they are affected by group size and image normalization. FDG PET images were acquired from rats (n = 18), normalized by whole brain, visual cortex, or cerebellar FDG uptake, and used to construct correlation matrices. Group size effects on network stability were investigated by systematically adding rats and evaluating local network connectivity (node strength and clustering coefficient). Modularity and community structure were also evaluated in the differently normalized networks to assess meso-scale network relationships. Local network properties are stable regardless of normalization region for groups of at least 10. Whole brain-normalized networks are more modular than visual cortex- or cerebellum-normalized network (p < 0.00001); however, community structure is similar at network resolutions where modularity differs most between brain and randomized networks. Hierarchical analysis reveals consistent modules at different scales and clustering of spatially-proximate brain regions. Findings suggest inter-subject FDG PET networks are stable for reasonable group sizes and exhibit multi-scale modularity.
Collapse
Affiliation(s)
- Megan M Sperry
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sonia Kartha
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Eric J Granquist
- Oral & Maxillofacial Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- , 240 Skirkanich Hall, 210 S. 33rd St., Philadelphia, PA, 19104, USA.
| |
Collapse
|
24
|
Gold MEL, Norell MA, Budassi M, Vaska P, Schulz D. Rapid 18F-FDG Uptake in Brain of Awake, Behaving Rat and Anesthetized Chicken has Implications for Behavioral PET Studies in Species With High Metabolisms. Front Behav Neurosci 2018; 12:115. [PMID: 29922136 PMCID: PMC5996747 DOI: 10.3389/fnbeh.2018.00115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 05/18/2018] [Indexed: 11/30/2022] Open
Abstract
Brain-behavior studies using 18F-FDG PET aim to reveal brain regions that become active during behavior. In standard protocols, 18F-FDG is injected, the behavior is executed during 30–60 min of tracer uptake, and then the animal is anesthetized and scanned. Hence, the uptake of 18F-FDG is not itself observed and could, in fact, be complete in very little time. This has implications for behavioral studies because uptake is assumed to reflect concurrent behavior. Here, we utilized a new, miniature PET scanner termed RatCAP to measure uptake simultaneously with behavior. We employed a novel injection protocol in which we administered 18F-FDG (i.v.) four times over two 2 h to allow for repeated measurements and the correlation of changes in uptake and behavioral activity. Furthermore, using standard PET methods, we explored the effects of injection route on uptake time in chickens, a model for avians, for which PET studies are just beginning. We found that in the awake, behaving rat most of the 18F-FDG uptake occurred within minutes and overlapped to a large extent with 18F-FDG data taken from longer uptake periods. By contrast, behavior which occurred within minutes of the 18F-FDG infusion differed markedly from the behavior that occurred during later uptake periods. Accordingly, we found that changes in 18F-FDG uptake in the striatum, motor cortex and cerebellum relative to different reference regions significantly predicted changes in behavioral activity during the scan, if the time bins used for correlation were near the injection times of 18F-FDG. However, when morphine was also injected during the scan, which completely abolished behavioral activity for over 50 min, a large proportion of the variance in behavioral activity was also explained by the uptake data from the entire scan. In anesthetized chickens, tracer uptake was complete in about 80 min with s.c. injection, but 8 min with i.v. injection. In conclusion, uptake time needs to be taken into account to more accurately correlate PET and behavioral data in mammals and avians. Additionally, RatCAP together with multiple, successive injections of 18F-FDG may be useful to explore changes in uptake over time in relation to changes in behavior.
Collapse
Affiliation(s)
- Maria E L Gold
- Division of Paleontology, American Museum of Natural History, New York, NY, United States.,Department of Anatomical Sciences, Stony Brook University, Stony Brook, NY, United States.,Department of Biology, Suffolk University, Boston, MA, United States
| | - Mark A Norell
- Division of Paleontology, American Museum of Natural History, New York, NY, United States
| | - Michael Budassi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Paul Vaska
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, United States.,Department of Radiology, Stony Brook University, Stony Brook, NY, United States.,Biosciences Department, Brookhaven National Laboratory, Upton, NY, United States
| | - Daniela Schulz
- Department of Psychology, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
25
|
Li YY, Zhang B, Yu KW, Li C, Xie HY, Bao WQ, Kong YY, Jiao FY, Guan YH, Bai YL. Effects of constraint-induced movement therapy on brain glucose metabolism in a rat model of cerebral ischemia: a micro PET/CT study. Int J Neurosci 2018; 128:736-745. [PMID: 29251083 DOI: 10.1080/00207454.2017.1418343] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Ying-Ying Li
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Bei Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke-Wei Yu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ce Li
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong-Yu Xie
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei-Qi Bao
- Center, Department of Nuclear Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan-Yan Kong
- Center, Department of Nuclear Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Yang Jiao
- Center, Department of Nuclear Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Hui Guan
- Center, Department of Nuclear Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Long Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Aguirre-Benítez EL, Porras MG, Parra L, González-Ríos J, Garduño-Torres DF, Albores-García D, Avendaño A, Ávila-Rodríguez MA, Melo AI, Jiménez-Estrada I, Mendoza-Garrido ME, Toriz C, Diaz D, Ibarra-Coronado E, Mendoza-Ángeles K, Hernández-Falcón J. Disruption of behavior and brain metabolism in artificially reared rats. Dev Neurobiol 2017; 77:1413-1429. [DOI: 10.1002/dneu.22548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 10/05/2017] [Accepted: 10/16/2017] [Indexed: 12/22/2022]
Affiliation(s)
| | - Mercedes G. Porras
- Departamento de Fisiología, Facultad de Medicina; UNAM, México, AP 70250, Av. Universidad No. 3000, Col. Copilco Universidad, México, CDMX; 04510 México México
| | - Leticia Parra
- Departamento de Anatomía, Facultad de Medicina; UNAM; México Mexico
| | | | | | | | - Arturo Avendaño
- Unidad Radiofarmacia-Ciclotrón, Facultad de Medicina, UNAM; México Mexico
| | | | - Angel I. Melo
- Centro de Investigación en Reproducción Animal CINVESTAV-Universidad Autónoma de Tlaxcala, Apdo Postal 62. C.P. Tlaxcala, C.P; Tlaxcala 90000 México
| | - Ismael Jiménez-Estrada
- Departamento de Fisiología, Biofísica y Neurociencias; CINVESTAV, IPN Av. Instituto Politécnico Nacional 2508 Col. San Pedro Zacatenco, Del. Gustavo A. Madero, C.P, CDMX; México 07360 México
| | - Ma. Eugenia Mendoza-Garrido
- Departamento de Fisiología, Biofísica y Neurociencias; CINVESTAV, IPN Av. Instituto Politécnico Nacional 2508 Col. San Pedro Zacatenco, Del. Gustavo A. Madero, C.P, CDMX; México 07360 México
| | - César Toriz
- Departamento de Fisiología, Biofísica y Neurociencias; CINVESTAV, IPN Av. Instituto Politécnico Nacional 2508 Col. San Pedro Zacatenco, Del. Gustavo A. Madero, C.P, CDMX; México 07360 México
| | - Daniel Diaz
- Centro de Ciencias de la Complejidad (C3) UNAM; México México
| | - Elizabeth Ibarra-Coronado
- Departamento de Fisiología, Facultad de Medicina; UNAM, México, AP 70250, Av. Universidad No. 3000, Col. Copilco Universidad, México, CDMX; 04510 México México
| | - Karina Mendoza-Ángeles
- Departamento de Fisiología, Facultad de Medicina; UNAM, México, AP 70250, Av. Universidad No. 3000, Col. Copilco Universidad, México, CDMX; 04510 México México
| | - Jesús Hernández-Falcón
- Departamento de Fisiología, Facultad de Medicina; UNAM, México, AP 70250, Av. Universidad No. 3000, Col. Copilco Universidad, México, CDMX; 04510 México México
| |
Collapse
|
27
|
Neuroimaging in animal models of epilepsy. Neuroscience 2017; 358:277-299. [DOI: 10.1016/j.neuroscience.2017.06.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 02/06/2023]
|
28
|
Lee HT, Liu SP, Lin CH, Lee SW, Hsu CY, Sytwu HK, Hsieh CH, Shyu WC. A Crucial Role of CXCL14 for Promoting Regulatory T Cells Activation in Stroke. Theranostics 2017; 7:855-875. [PMID: 28382159 PMCID: PMC5381249 DOI: 10.7150/thno.17558] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/29/2016] [Indexed: 12/19/2022] Open
Abstract
Inflammatory processes have a detrimental role in the pathophysiology of ischemic stroke. However, little is known about the endogenous anti-inflammatory mechanisms in ischemic brain. Here, we identify CXCL14 as a critical mediator of these mechanisms. CXCL14 levels were upregulated in the ischemic brains of humans and rodents. Moreover, hypoxia inducible factor-1α (HIF-1α) drives hypoxia- or cerebral ischemia (CI)-dependent CXCL14 expression via directly binding to the CXCL14 promoter. Depletion of CXCL14 inhibited the accumulation of immature dendritic cells (iDC) or regulatory T cells (Treg) and increased the infarct volume, whereas the supplementation of CXCL14 had the opposite effects. CXCL14 promoted the adhesion, migration, and homing of circulating CD11c+ iDC to the ischemic tissue via the upregulation of the cellular prion protein (PrPC), PECAM-1, and MMPs. The accumulation of Treg in ischemic areas of the brain was mediated through a cooperative effect of CXCL14 and iDC-secreted IL-2-induced Treg differentiation. Interestingly, CXCL14 largely promoted IL-2-induced Treg differentiation. These findings indicate that CXCL14 is a critical immunomodulator involved in the stroke-induced inflammatory reaction. Passive CXCL14 supplementation provides a tractable path for clinical translation in the improvement of stroke-induced neuroinflammation.
Collapse
|
29
|
Zhang X, Liu F, Slikker W, Wang C, Paule MG. Minimally invasive biomarkers of general anesthetic-induced developmental neurotoxicity. Neurotoxicol Teratol 2016; 60:95-101. [PMID: 27784630 DOI: 10.1016/j.ntt.2016.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/29/2016] [Accepted: 10/21/2016] [Indexed: 12/22/2022]
Abstract
The association of general anesthesia with developmental neurotoxicity, while nearly impossible to study in pediatric populations, is clearly demonstrable in a variety of animal models from rodents to nonhuman primates. Nearly all general anesthetics tested have been shown to cause abnormal brain cell death in animals when administered during periods of rapid brain growth. The ability to repeatedly assess in the same subjects adverse effects induced by general anesthetics provides significant power to address the time course of important events associated with exposures. Minimally-invasive procedures provide the opportunity to bridge the preclinical/clinical gap by providing the means to more easily translate findings from the animal laboratory to the human clinic. Positron Emission Tomography or PET is a tool with great promise for realizing this goal. PET for small animals (microPET) is providing valuable data on the life cycle of general anesthetic induced neurotoxicity. PET radioligands (annexin V and DFNSH) targeting apoptotic processes have demonstrated that a single bout of general anesthesia effected during a vulnerable period of CNS development can result in prolonged apoptotic signals lasting for several weeks in the rat. A marker of cellular proliferation (FLT) has demonstrated in rodents that general anesthesia-induced inhibition of neural progenitor cell proliferation is evident when assessed a full 2weeks after exposure. Activated glia express Translocator Protein (TSPO) which can be used as a marker of presumed neuroinflammatory processes and a PET ligand for the TSPO (FEPPA) has been used to track this process in both rat and nonhuman primate models. It has been shown that single bouts of general anesthesia can result in elevated TSPO expression lasting for over a week. These examples demonstrate the utility of specific PET tracers to inform, in a minimally-invasive fashion, processes associated with general anesthesia-induced developmental neurotoxicity. The fact that PET procedures are also used clinically suggests an opportunity to confirm in humans what has been repeatedly observed in animals.
Collapse
|
30
|
Role of IGF1R(+) MSCs in modulating neuroplasticity via CXCR4 cross-interaction. Sci Rep 2016; 6:32595. [PMID: 27586516 PMCID: PMC5009335 DOI: 10.1038/srep32595] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/03/2016] [Indexed: 12/11/2022] Open
Abstract
To guide the use of human mesenchymal stem cells (MSCs) toward clinical applications, identifying pluripotent-like-markers for selecting MSCs that retain potent self-renewal-ability should be addressed. Here, an insulin-like growth factor 1 receptor (IGF1R)–expressing sub-population in human dental pulp MSCs (hDSCs), displayed multipotent properties. IGF1R expression could be maintained in hDSCs when they were cultured in 2% human cord blood serum (hUCS) in contrast to that in 10% fetal calf serum (FCS). Cytokine array showed that hUCS contained higher amount of several growth factors compared to FCS, including IGF-1 and platelet-derived growth factor (PDGF-BB). These cytokines modulates the signaling events in the hDSCs and potentially enhances engraftment upon transplantation. Specifically, a bidirectional cross-talk between IGF1R/IGF1 and CXCR4/SDF-1α signaling pathways in hDSCs, as revealed by interaction of the two receptors and synergistic activation of both signaling pathways. In rat stroke model, animals receiving IGF1R+ hDSCs transplantation, interaction between IGF1R and CXCR4 was demonstrated to promote neuroplasticity, therefore improving neurological function through increasing glucose metabolic activity, enhancing angiogenesis and anti-inflammatiory effects. Therefore, PDGF in hUCS-culture system contributed to the maintenance of the expression of IGF1R in hDSCs. Furthermore, implantation of IGF1R+ hDSCs exerted enhanced neuroplasticity via integrating inputs from both CXCR4 and IGF1R signaling pathways.
Collapse
|
31
|
Fu TM, Hong G, Zhou T, Schuhmann TG, Viveros RD, Lieber CM. Stable long-term chronic brain mapping at the single-neuron level. Nat Methods 2016; 13:875-82. [PMID: 27571550 DOI: 10.1038/nmeth.3969] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 07/26/2016] [Indexed: 12/22/2022]
Abstract
Stable in vivo mapping and modulation of the same neurons and brain circuits over extended periods is critical to both neuroscience and medicine. Current electrical implants offer single-neuron spatiotemporal resolution but are limited by such factors as relative shear motion and chronic immune responses during long-term recording. To overcome these limitations, we developed a chronic in vivo recording and stimulation platform based on flexible mesh electronics, and we demonstrated stable multiplexed local field potentials and single-unit recordings in mouse brains for at least 8 months without probe repositioning. Properties of acquired signals suggest robust tracking of the same neurons over this period. This recording and stimulation platform allowed us to evoke stable single-neuron responses to chronic electrical stimulation and to carry out longitudinal studies of brain aging in freely behaving mice. Such advantages could open up future studies in mapping and modulating changes associated with learning, aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Tian-Ming Fu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Guosong Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Tao Zhou
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Thomas G Schuhmann
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Robert D Viveros
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Charles M Lieber
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA.,John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
32
|
Rohleder C, Wiedermann D, Neumaier B, Drzezga A, Timmermann L, Graf R, Leweke FM, Endepols H. The Functional Networks of Prepulse Inhibition: Neuronal Connectivity Analysis Based on FDG-PET in Awake and Unrestrained Rats. Front Behav Neurosci 2016; 10:148. [PMID: 27493627 PMCID: PMC4954847 DOI: 10.3389/fnbeh.2016.00148] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/06/2016] [Indexed: 12/21/2022] Open
Abstract
Prepulse inhibition (PPI) is a neuropsychological process during which a weak sensory stimulus (“prepulse”) attenuates the motor response (“startle reaction”) to a subsequent strong startling stimulus. It is measured as a surrogate marker of sensorimotor gating in patients suffering from neuropsychological diseases such as schizophrenia, as well as in corresponding animal models. A variety of studies has shown that PPI of the acoustical startle reaction comprises three brain circuitries for: (i) startle mediation, (ii) PPI mediation, and (iii) modulation of PPI mediation. While anatomical connections and information flow in the startle and PPI mediation pathways are well known, spatial and temporal interactions of the numerous regions involved in PPI modulation are incompletely understood. We therefore combined [18F]fluoro-2-deoxyglucose positron-emission-tomography (FDG-PET) with PPI and resting state control paradigms in awake rats. A battery of subtractive, correlative as well as seed-based functional connectivity analyses revealed a default mode-like network (DMN) active during resting state only. Furthermore, two functional networks were observed during PPI: Metabolic activity in the lateral circuitry was positively correlated with PPI effectiveness and involved the auditory system and emotional regions. The medial network was negatively correlated with PPI effectiveness, i.e., associated with startle, and recruited a spatial/cognitive network. Our study provides evidence for two distinct neuronal networks, whose continuous interplay determines PPI effectiveness in rats, probably by either protecting the prepulse or facilitating startle processing. Discovering similar networks affected in neuropsychological disorders may help to better understand mechanisms of sensorimotor gating deficits and provide new perspectives for therapeutic strategies.
Collapse
Affiliation(s)
- Cathrin Rohleder
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany; Institute of Radiochemistry and Experimental Molecular Imaging, University Hospital of CologneCologne, Germany
| | - Dirk Wiedermann
- In-Vivo NMR Laboratory, Max-Planck-Institute for Metabolism Research Cologne, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, University Hospital of CologneCologne, Germany; Forschungszentrum Jülich GmbH, Institute of Neurosciences and Medicine, INM-5: Nuclear ChemistryJülich, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne Cologne, Germany
| | - Lars Timmermann
- Department of Neurology, University Hospital of Cologne Cologne, Germany
| | - Rudolf Graf
- Multimodal Imaging of Brain Metabolism, Max-Planck-Institute for Metabolism Research Cologne, Germany
| | - F Markus Leweke
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Heike Endepols
- Institute of Radiochemistry and Experimental Molecular Imaging, University Hospital of Cologne Cologne, Germany
| |
Collapse
|
33
|
Yi B, Hu S, Zuo C, Jiao F, Lv J, Chen D, Ma Y, Chen J, Mei L, Wang X, Huang Z, Wu H. Effects of long-term salicylate administration on synaptic ultrastructure and metabolic activity in the rat CNS. Sci Rep 2016; 6:24428. [PMID: 27068004 PMCID: PMC4828705 DOI: 10.1038/srep24428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 03/30/2016] [Indexed: 11/17/2022] Open
Abstract
Tinnitus is associated with neural hyperactivity in the central nervous system (CNS). Salicylate is a well-known ototoxic drug, and we induced tinnitus in rats using a model of long-term salicylate administration. The gap pre-pulse inhibition of acoustic startle test was used to infer tinnitus perception, and only rats in the chronic salicylate-treatment (14 days) group showed evidence of experiencing tinnitus. After small animal positron emission tomography scans were performed, we found that the metabolic activity of the inferior colliculus (IC), the auditory cortex (AC), and the hippocampus (HP) were significantly higher in the chronic treatment group compared with saline group (treated for 14 days), which was further supported by ultrastructural changes at the synapses. The alterations all returned to baseline 14 days after the cessation of salicylate-treatment (wash-out group), indicating that these changes were reversible. These findings indicate that long-term salicylate administration induces tinnitus, enhanced neural activity and synaptic ultrastructural changes in the IC, AC, and HP of rats due to neuroplasticity. Thus, an increased metabolic rate and synaptic transmission in specific areas of the CNS may contribute to the development of tinnitus.
Collapse
Affiliation(s)
- Bin Yi
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.,Laboratory of Auditory Neuroscience, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai China
| | - Shousen Hu
- Department of Otolaryngology Head and Neck Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province China
| | - Chuantao Zuo
- PET Center, Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai China
| | - Fangyang Jiao
- PET Center, Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai China
| | - Jingrong Lv
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China
| | - Dongye Chen
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.,Laboratory of Auditory Neuroscience, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai China
| | - Yufei Ma
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China
| | - Jianyong Chen
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.,Laboratory of Auditory Neuroscience, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai China
| | - Ling Mei
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.,Laboratory of Auditory Neuroscience, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai China
| | - Xueling Wang
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.,Laboratory of Auditory Neuroscience, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai China
| | - Zhiwu Huang
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.,Laboratory of Auditory Neuroscience, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai China
| | - Hao Wu
- Department of Otolaryngology Head and Neck Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai China.,Laboratory of Auditory Neuroscience, Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai China
| |
Collapse
|
34
|
Lin CH, Chiu L, Lee HT, Chiang CW, Liu SP, Hsu YH, Lin SZ, Hsu CY, Hsieh CH, Shyu WC. PACAP38/PAC1 signaling induces bone marrow-derived cells homing to ischemic brain. Stem Cells 2016; 33:1153-72. [PMID: 25523790 PMCID: PMC4409028 DOI: 10.1002/stem.1915] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 11/04/2014] [Accepted: 11/08/2014] [Indexed: 11/08/2022]
Abstract
Understanding stem cell homing, which is governed by environmental signals from the surrounding niche, is important for developing effective stem cell-based repair strategies. The molecular mechanism by which the brain under ischemic stress recruits bone marrow-derived cells (BMDCs) to the vascular niche remains poorly characterized. Here we report that hypoxia-inducible factor-1α (HIF-1α) activation upregulates pituitary adenylate cyclase-activating peptide 38 (PACAP38), which in turn activates PACAP type 1 receptor (PAC1) under hypoxia in vitro and cerebral ischemia in vivo. BMDCs homing to endothelial cells in the ischemic brain are mediated by HIF-1α activation of the PACAP38-PAC1 signaling cascade followed by upregulation of cellular prion protein and α6-integrin to enhance the ability of BMDCs to bind laminin in the vascular niche. Exogenous PACAP38 confers a similar effect in facilitating BMDCs homing into the ischemic brain, resulting in reduction of ischemic brain injury. These findings suggest a novel HIF-1α-activated PACAP38-PAC1 signaling process in initiating BMDCs homing into the ischemic brain for reducing brain injury and enhancing functional recovery after ischemic stroke. Stem Cells2015;33:1153–1172
Collapse
Affiliation(s)
- Chen-Huan Lin
- Center for Neuropsychiatry and Translational Medicine Research Center, China Medical University and Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Park GY, Lee EM, Seo MS, Seo YJ, Oh JS, Son WC, Kim KS, Kim JS, Kang JK, Kang KS. Preserved Hippocampal Glucose Metabolism on 18F-FDG PET after Transplantation of Human Umbilical Cord Blood-derived Mesenchymal Stem Cells in Chronic Epileptic Rats. J Korean Med Sci 2015; 30:1232-40. [PMID: 26339161 PMCID: PMC4553668 DOI: 10.3346/jkms.2015.30.9.1232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 05/18/2015] [Indexed: 11/20/2022] Open
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) may be a promising modality for treating medial temporal lobe epilepsy. (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET) is a noninvasive method for monitoring in vivo glucose metabolism. We evaluated the efficacy of hUCB-MSCs transplantation in chronic epileptic rats using FDG-PET. Rats with recurrent seizures were randomly assigned into three groups: the stem cell treatment (SCT) group received hUCB-MSCs transplantation into the right hippocampus, the sham control (ShC) group received same procedure with saline, and the positive control (PC) group consisted of treatment-negative epileptic rats. Normal rats received hUCB-MSCs transplantation acted as the negative control (NC). FDG-PET was performed at pre-treatment baseline and 1- and 8-week posttreatment. Hippocampal volume was evaluated and histological examination was done. In the SCT group, bilateral hippocampi at 8-week after transplantation showed significantly higher glucose metabolism (0.990 ± 0.032) than the ShC (0.873 ± 0.087; P < 0.001) and PC groups (0.858 ± 0.093; P < 0.001). Histological examination resulted that the transplanted hUCB-MSCs survived in the ipsilateral hippocampus and migrated to the contralateral hippocampus but did not differentiate. In spite of successful engraftment, seizure frequency among the groups was not significantly different. Transplanted hUCB-MSCs can engraft and migrate, thereby partially restoring bilateral hippocampal glucose metabolism. The results suggest encouraging effect of hUCB-MSCs on restoring epileptic networks.
Collapse
Affiliation(s)
- Ga Young Park
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- The Asan Institute for Life Science, Seoul, Korea
| | - Eun Mi Lee
- Department of Neurology, Ulsan University Hospital, Ulsan, Korea
| | - Min-Soo Seo
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Yoo-Jin Seo
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jungsu S. Oh
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Woo-Chan Son
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ki Soo Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Joong Koo Kang
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
36
|
Cerebral glucose metabolism changes in rat brain upon forepaw electrical stimulation at different frequencies. Neuroreport 2015; 26:197-205. [PMID: 25674904 DOI: 10.1097/wnr.0000000000000327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PET imaging techniques and statistical parametric mapping analysis have been developed to identify neuronal functional activation from brain imaging. The purpose of this study was to examine the efficacy of the glucose metabolism in rat brain during forepaw electrical stimulation with different frequencies (3, 10, or 20 Hz) compared with a nonstimulated group (control). Fluorine-18 fluorodeoxyglucose was injected after confirmation of the range of normal physiology. For quantitative analysis, we used cerebral metabolism rate of glucose consumption (CMRglc) responses in the primary somatosensory cortex (S1FL) and motor cortex (M1). On comparing CMRglc responses in the contralateral S1FL and M1 with those of the ipsilateral areas in intragroup analysis, a significant increase (P<0.05) was observed in two electrical stimulation groups (10 and 20 Hz) but not at the 3 Hz level. In intergroup analysis, the CMRglc responses in the contralateral region of interest were compared with those of the control group to validate which electrical frequency conditions were appropriate to induce neuronal functional activation. Among the stimulation groups, significant increases in CMRglc response were only observed at 10 Hz (P<0.05). Therefore, 10 Hz is the most suitable frequency to confirm changes in CMRglc in the S1FL and M1 of the rat brain, and also fluorine-18 fluorodeoxyglucose PET could be useful to investigate recovery and plasticity in neurological diseases associated with primary sensory-motor cortex function.
Collapse
|
37
|
Kolodziej A, Lippert M, Angenstein F, Neubert J, Pethe A, Grosser OS, Amthauer H, Schroeder UH, Reymann KG, Scheich H, Ohl FW, Goldschmidt J. SPECT-imaging of activity-dependent changes in regional cerebral blood flow induced by electrical and optogenetic self-stimulation in mice. Neuroimage 2014; 103:171-180. [DOI: 10.1016/j.neuroimage.2014.09.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/13/2014] [Accepted: 09/08/2014] [Indexed: 12/29/2022] Open
|
38
|
Dedeurwaerdere S, Shultz SR, Federico P, Engel J. Workshop on Neurobiology of Epilepsy appraisal: new systemic imaging technologies to study the brain in experimental models of epilepsy. Epilepsia 2014; 55:819-28. [PMID: 24836499 DOI: 10.1111/epi.12642] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2014] [Indexed: 12/14/2022]
Abstract
Modern functional neuroimaging provides opportunities to visualize activity of the entire brain, making it an indispensable diagnostic tool for epilepsy. Various forms of noninvasive functional neuroimaging are now also being performed as research tools in animal models of epilepsy and provide opportunities for parallel animal/human investigations into fundamental mechanisms of epilepsy and identification of epilepsy biomarkers. Recent animal studies of epilepsy using positron emission tomography, tractography, and functional magnetic resonance imaging were reviewed. Epilepsy is an abnormal emergent property of disturbances in neuronal networks which, even for epilepsies characterized by focal seizures, involve widely distributed systems, often in both hemispheres. Functional neuroimaging in animal models now provides opportunities to examine neuronal disturbances in the whole brain that underlie generalized and focal seizure generation as well as various types of epileptogenesis. Tremendous advances in understanding the contribution of specific properties of widely distributed neuronal networks to both normal and abnormal human behavior have been provided by current functional neuroimaging methodologies. Successful application of functional neuroimaging of the whole brain in the animal laboratory now permits investigations during epileptogenesis and correlation with deep brain electroencephalography (EEG) activity. With the continuing development of these techniques and analytical methods, the potential for future translational research on epilepsy is enormous. A PowerPoint slide summarizing this article is available for download in the Supporting Information section here.
Collapse
|
39
|
Thompson SJ, Millecamps M, Aliaga A, Seminowicz DA, Low LA, Bedell BJ, Stone LS, Schweinhardt P, Bushnell MC. Metabolic brain activity suggestive of persistent pain in a rat model of neuropathic pain. Neuroimage 2014; 91:344-52. [PMID: 24462776 DOI: 10.1016/j.neuroimage.2014.01.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/07/2013] [Accepted: 01/13/2014] [Indexed: 10/25/2022] Open
Abstract
Persistent pain is a central characteristic of neuropathic pain conditions in humans. Knowing whether rodent models of neuropathic pain produce persistent pain is therefore crucial to their translational applicability. We investigated the spared nerve injury (SNI) model of neuropathic pain and the formalin pain model in rats using positron emission tomography (PET) with the metabolic tracer [18F]fluorodeoxyglucose (FDG) to determine if there is ongoing brain activity suggestive of persistent pain. For the formalin model, under brief anesthesia we injected one hindpaw with 5% formalin and the FDG tracer into a tail vein. We then allowed the animals to awaken and observed pain behavior for 30min during the FDG uptake period. The rat was then anesthetized and placed in the scanner for static image acquisition, which took place between minutes 45 and 75 post-tracer injection. A single reference rat brain magnetic resonance image (MRI) was used to align the PET images with the Paxinos and Watson rat brain atlas. Increased glucose metabolism was observed in the somatosensory region associated with the injection site (S1 hindlimb contralateral), S1 jaw/upper lip and cingulate cortex. Decreases were observed in the prelimbic cortex and hippocampus. Second, SNI rats were scanned 3weeks post-surgery using the same scanning paradigm, and region-of-interest analyses revealed increased metabolic activity in the contralateral S1 hindlimb. Finally, a second cohort of SNI rats was scanned while anesthetized during the tracer uptake period, and the S1 hindlimb increase was not observed. Increased brain activity in the somatosensory cortex of SNI rats resembled the activity produced with the injection of formalin, suggesting that the SNI model may produce persistent pain. The lack of increased activity in S1 hindlimb with general anesthetic demonstrates that this effect can be blocked, as well as highlights the importance of investigating brain activity in awake and behaving rodents.
Collapse
Affiliation(s)
- Scott J Thompson
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3G 0G1, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2T5, Canada.
| | - Magali Millecamps
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3G 0G1, Canada; Faculty of Dentistry, McGill University, Montreal, QC H3A 2T5, Canada
| | - Antonio Aliaga
- Small Animal Imaging Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - David A Seminowicz
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Lucie A Low
- Division of Intramural Research, National Center for Complementary and Alternative Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barry J Bedell
- Small Animal Imaging Lab, McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada; Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Laura S Stone
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3G 0G1, Canada; Faculty of Dentistry, McGill University, Montreal, QC H3A 2T5, Canada; Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Petra Schweinhardt
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3G 0G1, Canada; Faculty of Dentistry, McGill University, Montreal, QC H3A 2T5, Canada; Department of Neurology & Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - M Catherine Bushnell
- Division of Intramural Research, National Center for Complementary and Alternative Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
40
|
Shih YYI, Yash TV, Rogers B, Duong TQ. FMRI of deep brain stimulation at the rat ventral posteromedial thalamus. Brain Stimul 2013; 7:190-3. [PMID: 24309153 DOI: 10.1016/j.brs.2013.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 09/25/2013] [Accepted: 11/01/2013] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Functional magnetic resonance imaging (fMRI) of deep brain stimulation (DBS) has potentials to reveal neuroanatomical connectivity of a specific brain region in vivo. OBJECTIVE This study aimed to demonstrate frequency and amplitude tunings of the thalamocortical tract using DBS fMRI at the rat ventral posteromedial thalamus. METHODS Blood oxygenation level dependent (BOLD) fMRI data were acquired in a total of twelve rats at a high-field 11.7 T MRI scanner with modulation of nine stimulus frequencies (1-40 Hz) and seven stimulus amplitudes (0.2-3.6 mA). RESULTS BOLD response in the barrel cortex peaked at 25 Hz. The response increased with stimulus amplitude and reached a plateau at 1 mA. Cortical spreading depolarization (CSD) was observed occasionally after DBS that carries >10% BOLD waves spanning the entire ipsilateral cortex. CONCLUSION fMRI is sensitive to the frequency effect of DBS and has potential to investigate the function of a particular neuroanatomical pathway.
Collapse
Affiliation(s)
- Yen-Yu Ian Shih
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Tiwari V Yash
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Bill Rogers
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Timothy Q Duong
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; South Texas Veterans Health Care System, Department of Veterans Affairs, USA.
| |
Collapse
|
41
|
Xi W, Su D, Nie B, Yu Y, Shan B, Chen Q, Tian M, Zhang H. 18F-FDG PET study reveals brain functional changes during attention in rats. J Nucl Med 2013; 54:1969-73. [PMID: 24115527 DOI: 10.2967/jnumed.113.123000] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Attentional impairments are seen in many clinical syndromes, including attention deficit hyperactivity disorder, schizophrenia, and Alzheimer disease. Understanding the mechanism of attention can be helpful for the diagnosis and treatment of these diseases. The aim of this study was to assess brain glucose metabolic changes in a rat model of attention. METHODS Small-animal PET studies were performed at 4 stages. Statistical parametric mapping was used for image analysis. RESULTS Increased (18)F-FDG uptake was found in the lateral hypothalamic area and left accumbens nucleus in the learning condition. Under the attentive condition, increased (18)F-FDG uptake was observed in the right retrosplenial cortex but (18)F-FDG uptake was decreased in the right medial geniculate nucleus. (18)F-FDG uptake change in the right retrosplenial cortex was negatively correlated with correct latency of behavior performance. CONCLUSION (18)F-FDG small-animal PET imaging provided novel findings on attention-related glucose metabolic changes, which were significantly correlated with the behavior performance in this rat model.
Collapse
Affiliation(s)
- Wang Xi
- Department of Nuclear Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Scott BB, Brody CD, Tank DW. Cellular resolution functional imaging in behaving rats using voluntary head restraint. Neuron 2013; 80:371-84. [PMID: 24055015 DOI: 10.1016/j.neuron.2013.08.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2013] [Indexed: 10/26/2022]
Abstract
High-throughput operant conditioning systems for rodents provide efficient training on sophisticated behavioral tasks. Combining these systems with technologies for cellular resolution functional imaging would provide a powerful approach to study neural dynamics during behavior. Here we describe an integrated two-photon microscope and behavioral apparatus that allows cellular resolution functional imaging of cortical regions during epochs of voluntary head restraint. Rats were trained to initiate periods of restraint up to 8 s in duration, which provided the mechanical stability necessary for in vivo imaging while allowing free movement between behavioral trials. A mechanical registration system repositioned the head to within a few microns, allowing the same neuronal populations to be imaged on each trial. In proof-of-principle experiments, calcium-dependent fluorescence transients were recorded from GCaMP-labeled cortical neurons. In contrast to previous methods for head restraint, this system can be incorporated into high-throughput operant conditioning systems.
Collapse
Affiliation(s)
- Benjamin B Scott
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Bezos Center for Neural Circuit Dynamics, Princeton University, Princeton, NJ 08544, USA
| | | | | |
Collapse
|
43
|
Simultaneous PET-MRI reveals brain function in activated and resting state on metabolic, hemodynamic and multiple temporal scales. Nat Med 2013; 19:1184-9. [PMID: 23975025 DOI: 10.1038/nm.3290] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 01/28/2013] [Indexed: 02/07/2023]
Abstract
Combined positron emission tomography (PET) and magnetic resonance imaging (MRI) is a new tool to study functional processes in the brain. Here we study brain function in response to a barrel-field stimulus simultaneously using PET, which traces changes in glucose metabolism on a slow time scale, and functional MRI (fMRI), which assesses fast vascular and oxygenation changes during activation. We found spatial and quantitative discrepancies between the PET and the fMRI activation data. The functional connectivity of the rat brain was assessed by both modalities: the fMRI approach determined a total of nine known neural networks, whereas the PET method identified seven glucose metabolism-related networks. These results demonstrate the feasibility of combined PET-MRI for the simultaneous study of the brain at activation and rest, revealing comprehensive and complementary information to further decode brain function and brain networks.
Collapse
|
44
|
Lee SD, Lai TW, Lin SZ, Lin CH, Hsu YH, Li CY, Wang HJ, Lee W, Su CY, Yu YL, Shyu WC. Role of stress-inducible protein-1 in recruitment of bone marrow derived cells into the ischemic brains. EMBO Mol Med 2013; 5:1227-46. [PMID: 23836498 PMCID: PMC3944463 DOI: 10.1002/emmm.201202258] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 05/15/2013] [Accepted: 05/17/2013] [Indexed: 12/21/2022] Open
Abstract
Stress-inducible protein-1 (STI-1) is the proposed ligand for the cellular prion protein (PrPC), which is thought to facilitate recovery following stroke. Whether STI-1 expression is affected by stroke and how its signalling facilitates recovery remain elusive. Brain slices from patients that died of ischemic stroke were collected for STI-1 immunohistochemistry. These findings were compared to results from cell cultures, mice with or without the PrPC knockout, and rats. Based on these findings, molecular and pharmacological interventions were administered to investigate the underlying mechanisms and to test the possibility for therapy in experimental stroke models. STI-1 was upregulated in the ischemic brains from humans and rodents. The increase in STI-1 expression in vivo was not cell-type specific, as it was found in neurons, glia and endothelial cells. Likewise, this increase in STI-1 expression can be mimicked by sublethal hypoxia in primary cortical cultures (PCCs) in vitro, and appear to have resulted from the direct binding of the hypoxia inducible factor-1α (HIF-1α) to the STI-1 promoter. Importantly, this STI-1 signalling promoted bone marrow derived cells (BMDCs) proliferation and migration in vitro and recruitment to the ischemic brain in vivo, and augmenting its signalling facilitated neurological recovery in part by recruiting BMDCs to the ischemic brain. Our results thus identified a novel mechanism by which ischemic insults can trigger a self-protective mechanism to facilitate recovery. This work identifies HIF-1α-mediated transcription of STI-1 and PrPc interaction as leading to BMDCs recruitment into ischemic brains following stroke in both patients and animal models of stroke, highlighting novel neuroprotective possibilities.
Collapse
Affiliation(s)
- Shin-Da Lee
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Low-frequency stimulation inhibits epileptogenesis by modulating the early network of the limbic system as evaluated in amygdala kindling model. Brain Struct Funct 2013; 219:1685-96. [DOI: 10.1007/s00429-013-0594-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 06/03/2013] [Indexed: 12/29/2022]
|
46
|
Yu YL, Chou RH, Shyu WC, Hsieh SC, Wu CS, Chiang SY, Chang WJ, Chen JN, Tseng YJ, Lin YH, Lee W, Yeh SP, Hsu JL, Yang CC, Hung SC, Hung MC. Smurf2-mediated degradation of EZH2 enhances neuron differentiation and improves functional recovery after ischaemic stroke. EMBO Mol Med 2013; 5:531-47. [PMID: 23526793 PMCID: PMC3628108 DOI: 10.1002/emmm.201201783] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 01/16/2013] [Accepted: 01/17/2013] [Indexed: 12/20/2022] Open
Abstract
EZH2 plays an important role in stem cell renewal and maintenance by inducing gene silencing via its histone methyltransferase activity. Previously, we showed that EZH2 downregulation enhances neuron differentiation of human mesenchymal stem cells (hMSCs); however, the underlying mechanisms of EZH2-regulated neuron differentiation are still unclear. Here, we identify Smurf2 as the E3 ubiquitin ligase responsible for the polyubiquitination and proteasome-mediated degradation of EZH2, which is required for neuron differentiation. A ChIP-on-chip screen combined with gene microarray analysis revealed that PPARγ was the only gene involved in neuron differentiation with significant changes in both its modification and expression status during differentiation. Moreover, knocking down PPARγ prevented cells from undergoing efficient neuron differentiation. In animal model, rats implanted with intracerebral EZH2-knocked-down hMSCs or hMSCs plus treatment with PPARγ agonist (rosiglitazone) showed better improvement than those without EZH2 knockdown or rosiglitazone treatment after a stroke. Together, our results support Smurf2 as a regulator of EZH2 turnover to facilitate PPARγ expression, which is specifically required for neuron differentiation, providing a molecular mechanism for clinical applications in the neurodegenerative diseases.
Collapse
Affiliation(s)
- Yung-Luen Yu
- Graduate Institute of Cancer Biology, Center for Molecular Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cherry SR. Fundamentals of Positron Emission Tomography and Applications in Preclinical Drug Development. J Clin Pharmacol 2013; 41:482-91. [PMID: 11361044 DOI: 10.1177/00912700122010357] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Positron emission tomography (PET) is a nuclear imaging technique that can dynamically image trace amounts of positron-labeled radiopharmaceuticals in vivo. Tracer concentrations can be determined quantitatively, and by application of appropriate tracer kinetic models, the rates of a wide range of different biological processes can be measured noninvasively in humans. PET has been used as a research tool for more than 25 years and has also found clinical applications, particularly in oncology, neurological disorders, and cardiovascular disease. Recently, there has been tremendous interest in applying PET technology to in vivo small-animal imaging. Significant improvements in the imaging technology now permit a wide range of PET studies in mice and rats, using compact, relatively low-cost, dedicated small-animal PET scanners. This article reviews the fundamental basis of PET imaging and discusses the development of small-animal PET scanners and their possible application in preclinical drug development.
Collapse
Affiliation(s)
- S R Cherry
- Crump Institute for Molecular Imaging and Department of Molecular and Medical Pharmacology, UCLA School of Medicine, Los Angeles, California, 90095-1770, USA
| |
Collapse
|
48
|
Mace E, Montaldo G, Osmanski BF, Cohen I, Fink M, Tanter M. Functional ultrasound imaging of the brain: theory and basic principles. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2013; 60:492-506. [PMID: 23475916 DOI: 10.1109/tuffc.2013.2592] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Hemodynamic changes in the brain are often used as surrogates of neuronal activity to infer the loci of brain activity. A major limitation of conventional Doppler ultrasound for the imaging of these changes is that it is not sensitive enough to detect the blood flow in small vessels where the major part of the hemodynamic response occurs. Here, we present a μDoppler ultrasound method able to detect and map the cerebral blood volume (CBV) over the entire brain with an important increase in sensitivity. This method is based on imaging the brain at an ultrafast frame rate (1 kHz) using compounded plane wave emissions. A theoretical model demonstrates that the gain in sensitivity of the μDoppler method is due to the combination of 1) the high signal-to-noise ratio of the gray scale images, resulting from the synthetic compounding of backscattered echoes; and 2) the extensive signal averaging enabled by the high temporal sampling of ultrafast frame rates. This μDoppler imaging is performed in vivo on trepanned rats without the use of contrast agents. The resulting images reveal detailed maps of the rat brain vascularization with an acquisition time as short as 320 ms per slice. This new method is the basis for a real-time functional ultrasound (fUS) imaging of the brain.
Collapse
Affiliation(s)
- Emilie Mace
- Institut Langevin, CNRS UMR7587, Inserm U979, Université Paris VII, Ecole Superieure de Physique et de Chimie Industrielles de Paris, Paris, France.
| | | | | | | | | | | |
Collapse
|
49
|
Using microPET imaging in quantitative verification of the acupuncture effect in ischemia stroke treatment. Sci Rep 2013; 3:1070. [PMID: 23323213 PMCID: PMC3545224 DOI: 10.1038/srep01070] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/02/2013] [Indexed: 11/27/2022] Open
Abstract
Acupuncture has been indispensable in Chinese medicine. However, its function still remains elusive. This paper studies the effect of acupuncture in ischemia stroke treatment using the Sprague Dawley rat animal model. We induced focal cerebral ischemia in rats using the middle cerebral artery occlusion (MCAO) procedure. For each rat in the real acupuncture group (n = 63), the sham acupoint treatment group (n = 62), and the blank control group (n = 30), we acquired 3-D fluorodeoxyglucose-microPET images at baseline, after MCAO, and after treatment, respectively. Then, we measured the changes of the injury-volume in the right hemisphere of these rats. The measurements showed that real acupuncture slightly reduced the injury-volume, sham acupoint treatment increased the injury-volume, and blank control had no obvious effect in reducing the injury-volume. Statistical tests also confirmed that acupuncture was more effective than random stimulus in improving the metabolic recovery after stroke.
Collapse
|
50
|
Zhang X, Newport GD, Paule MG, Liu F, Liu S, Berridge MS, Apana SM, Ali SF, Slikker W, Wang C. Quantitative Assessment of Acetyl-Carnitine Effects on Anesthetic-Induced Neuronal Death Using MicroPET/CT Imaging. ACTA ACUST UNITED AC 2013. [DOI: 10.4303/jdar/235653] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|