1
|
Ohshima M, Moriguchi T, Enmi JI, Kawashima H, Koshino K, Zeniya T, Tsuji M, Iida H. [ 123I]CLINDE SPECT as a neuroinflammation imaging approach in a rat model of stroke. Exp Neurol 2024; 378:114843. [PMID: 38823675 DOI: 10.1016/j.expneurol.2024.114843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
Poststroke neuroinflammation exacerbates disease progression. [11C]PK11195-positron emission tomography (PET) imaging has been used to visualize neuroinflammation; however, its short half-life of 20 min limits its clinical use. [123I]CLINDE has a longer half-life (13h); therefore, [123I]CLINDE-single-photon emission computed tomography (SPECT) imaging is potentially more practical than [11C]PK11195-PET imaging in clinical settings. The objectives of this study were to 1) validate neuroinflammation imaging using [123I]CLINDE and 2) investigate the mechanisms underlying stroke in association with neuroinflammation using multimodal techniques, including magnetic resonance imaging (MRI), gas-PET, and histological analysis, in a rat model of ischemic stroke, that is, permanent middle cerebral artery occlusion (pMCAo). At 6 days post-pMCAo, [123I]CLINDE-SPECT considerably corresponded to the immunohistochemical images stained with the CD68 antibody (a marker for microglia/microphages), comparable to the level observed in [11C]PK11195-PET images. In addition, the [123I]CLINDE-SPECT images corresponded well with autoradiography images. Rats with severe infarcts, as defined by MRI, exhibited marked neuroinflammation in the peri-infarct area and less neuroinflammation in the ischemic core, accompanied by a substantial reduction in the cerebral metabolic rate of oxygen (CMRO2) in 15O-gas-PET. Rats with moderate-to-mild infarcts exhibited neuroinflammation in the ischemic core, where CMRO2 levels were mildly reduced. This study demonstrates that [123I]CLINDE-SPECT imaging is suitable for neuroinflammation imaging and that the distribution of neuroinflammation varies depending on the severity of infarction.
Collapse
Affiliation(s)
- Makiko Ohshima
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Visionsgatan 4, Solna 171 64, Sweden
| | - Tetsuaki Moriguchi
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Institute of Physics, University of Tsukuba, Ibaraki 305-8571, Japan
| | - Jun-Ichiro Enmi
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Center for Information and Neural Networks (CiNet), Advanced ICT Research Institute, National Institute of Information and Communications Technology (NICT), 1-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hidekazu Kawashima
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Radioisotope Research Center, Kyoto Pharmaceutical University, 1 Misasagi-Shichono-cho, Yamashina-ku, Kyoto 607-8412, Japan
| | - Kazuhiro Koshino
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Department of Systems and Informatics, Hokkaido Information University, 59-2 Nishi-nopporo, Ebetsu, Hokkaido, Japan
| | - Tsutomu Zeniya
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Graduate School of Science and Technology, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori 036-8561, Japan
| | - Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto 605-8501, Japan.
| | - Hidehiro Iida
- Department of Investigative Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka 564-8565, Japan; Faculty of Medicine, University of Turku, and Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520 Turku, Finland
| |
Collapse
|
2
|
Blum N, Mirian C, Maier AD, Mathiesen TI, Vilhardt F, Haslund-Vinding JL. Translocator protein (TSPO) expression in neoplastic cells and tumor-associated macrophages in meningiomas. J Neuropathol Exp Neurol 2023; 82:1020-1032. [PMID: 37952221 DOI: 10.1093/jnen/nlad093] [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] [Indexed: 11/14/2023] Open
Abstract
Meningiomas are the most common primary intracranial tumors and show extensive infiltration of macrophages. The mitochondrial membrane protein translocator protein (TSPO) has been used as an in vivo marker of microglia and macrophage activation to visualize neuroinflammation. However, it is unknown which cell types express TSPO in meningiomas. Immunohistochemistry of 38 WHO grade 1-3 meningiomas was subjected to segmentation and deep learning classification of TSPO expression to either Iba1-positive tumor-associated macrophages (TAMs) or all other (mainly neoplastic) cells. A possible association between clinical data and TSPO expression intensities was also investigated. TAMs accounted for 15.9%-26% of all cells in the meningioma tissue. Mean fluorescence intensity of TSPO was significantly higher in TAMs (p < 0.0001), but the mass of neoplastic cells in the tumors exceeded that of TAMs. Thus, the summed fluorescence intensity of TSPO in meningioma cells was 64.1% higher than in TAMs (p = 0.0003). We observed no correlation between TSPO expression intensity and WHO grade. These results indicate that both macrophage-lineage and neoplastic cells in meningiomas express TSPO and that the SPECT-TSPO signal in meningiomas mainly reflects the latter; TSPO is expressed equally in parenchymal activated and resting macrophage/microglia lineage cells.
Collapse
Affiliation(s)
- Nadja Blum
- Department of Neurosurgery, Rigshospitalet, Copenhagen, Denmark
| | | | - Andrea Daniela Maier
- Department of Neurosurgery, Rigshospitalet, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | | | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark
| | | |
Collapse
|
3
|
Van Camp N, Lavisse S, Roost P, Gubinelli F, Hillmer A, Boutin H. TSPO imaging in animal models of brain diseases. Eur J Nucl Med Mol Imaging 2021; 49:77-109. [PMID: 34245328 PMCID: PMC8712305 DOI: 10.1007/s00259-021-05379-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/25/2021] [Indexed: 12/19/2022]
Abstract
Over the last 30 years, the 18-kDa TSPO protein has been considered as the PET imaging biomarker of reference to measure increased neuroinflammation. Generally assumed to image activated microglia, TSPO has also been detected in endothelial cells and activated astrocytes. Here, we provide an exhaustive overview of the recent literature on the TSPO-PET imaging (i) in the search and development of new TSPO tracers and (ii) in the understanding of acute and chronic neuroinflammation in animal models of neurological disorders. Generally, studies testing new TSPO radiotracers against the prototypic [11C]-R-PK11195 or more recent competitors use models of acute focal neuroinflammation (e.g. stroke or lipopolysaccharide injection). These studies have led to the development of over 60 new tracers during the last 15 years. These studies highlighted that interpretation of TSPO-PET is easier in acute models of focal lesions, whereas in chronic models with lower or diffuse microglial activation, such as models of Alzheimer's disease or Parkinson's disease, TSPO quantification for detection of neuroinflammation is more challenging, mirroring what is observed in clinic. Moreover, technical limitations of preclinical scanners provide a drawback when studying modest neuroinflammation in small brains (e.g. in mice). Overall, this review underlines the value of TSPO imaging to study the time course or response to treatment of neuroinflammation in acute or chronic models of diseases. As such, TSPO remains the gold standard biomarker reference for neuroinflammation, waiting for new radioligands for other, more specific targets for neuroinflammatory processes and/or immune cells to emerge.
Collapse
Affiliation(s)
- Nadja Van Camp
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Sonia Lavisse
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Pauline Roost
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Francesco Gubinelli
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France
| | - Ansel Hillmer
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, CT, USA
| | - Hervé Boutin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Brain and Mental Health, University of Manchester, M13 9PL, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, 27 Palatine Road, M20 3LJ, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
4
|
Zak A, Lemaire L, Chalon S, Chicheri G, Marzag H, Bodard S, Sérrière S, Routier S, Buron F, Vercouillie J. [ 18 F]-labeled positron emission tomography ligand for the histamine H4 receptor. J Labelled Comp Radiopharm 2021; 64:363-372. [PMID: 34089268 DOI: 10.1002/jlcr.3929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/18/2021] [Accepted: 05/28/2021] [Indexed: 11/10/2022]
Abstract
We synthesized 5-[18 F]-fluoro-1H-indol-2-yl)(4-methyl-1-piperazinyl)methanone ([18 F]5) via a Suzuki approach starting from a protected pinacol borane precursor followed by acidic hydrolysis of the t-Boc protecting group. The non-optimized radiochemical yield was 5.7 ± 1.35%, radiochemical purity was over 99%, and molar activity was 100.7 ± 34.5 GBq/μmol (n = 3). [18 F]5 was stable in rat plasma for at least 4 h and was evaluated by μPET imaging and biodistribution using a unilateral quinolinic acid rat model of neuroinflammation. The time-activity curve showed that [18 F]5 entered the brain immediately after intravenous injection and then left it progressively with a very low level reached from 30 min after injection. The biodistribution study showed no difference in the accumulation of [18 F]5 between the lesioned and intact side of the brain and between control rats and animals pretreated with a saturating dose of JNJ-7777120 as a specific H4R antagonist. Hence, despite its in vitro nanomolar affinity for H4R, and its ability to cross the blood-brain barrier in rats, [18 F]5 does not appear suitable to image in vivo the receptor by PET.
Collapse
Affiliation(s)
- Agnieszka Zak
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans, France
| | - Lucas Lemaire
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans, France
| | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Gabrielle Chicheri
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CERRP, Université de Tours, Tours, France
| | - Hamid Marzag
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans, France
| | - Sylvie Bodard
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sophie Sérrière
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvain Routier
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans, France
| | - Frédéric Buron
- Institut de Chimie Organique et Analytique, Université d'Orléans, UMR CNRS 7311, Orléans, France
| | - Johnny Vercouillie
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
- CERRP, Université de Tours, Tours, France
- INSERM CIC 1415, University Hospital, Tours, France
| |
Collapse
|
5
|
Ceyzériat K, Gloria Y, Tsartsalis S, Fossey C, Cailly T, Fabis F, Millet P, Tournier BB. Alterations in dopamine system and in its connectivity with serotonin in a rat model of Alzheimer's disease. Brain Commun 2021; 3:fcab029. [PMID: 34286270 PMCID: PMC8287930 DOI: 10.1093/braincomms/fcab029] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/20/2022] Open
Abstract
Dopamine pathways alterations are reported in Alzheimer’s disease. However, it is
difficult in humans to establish when these deficits appear and their impact in the course
of Alzheimer’s disease. In the TgF344-Alzheimer’s disease rat model at the age of
6 months, we showed a reduction in in vivo release of striatal dopamine
due to serotonin 5HT2A-receptor blockade, in the absence of alterations in
5HT2A-receptor binding, suggesting a reduction in
5HT2A-receptor-dopamine system connectivity. In addition, a functional
hypersensitivity of postsynaptic dopamine D2-receptors and
D2-autoreceptors was also reported without any change in D2-receptor
density and in the absence of amyloid plaques or overexpression of the 18 kDa translocator
protein (an inflammatory marker) in areas of the dopamine system. Citalopram, a selective
serotonin reuptake inhibitor, induced functional
5HT2A-receptor−D2-receptor connectivity changes but had no effect on
D2-autoreceptor hypersensitivity. In older rats, dopamine cell bodies
overexpressed translocator protein and dopamine projection sites accumulated amyloid.
Interestingly, the 5HT2A-receptor density is decreased in the accumbens
subdivisions and the substantia nigra pars compacta. This reduction in the striatum is
related to the astrocytic expression of 5HT2A-receptor. Our results indicate
that both serotonin/dopamine connectivity and dopamine signalling pathways are
dysregulated and potentially represent novel early diagnostic and therapeutic avenues.
Collapse
Affiliation(s)
- Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, 1206 Geneva, Switzerland.,Division of Nuclear medicine, Diagnostic Department, University Hospitals and Geneva University of Geneva, 1206 Geneva, Switzerland.,Division of Radiation Oncology, Department of Oncology, University Hospitals of Geneva, 1206 Geneva, Switzerland
| | - Yesica Gloria
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, 1206 Geneva, Switzerland
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, 1206 Geneva, Switzerland
| | - Christine Fossey
- Normandie University, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Thomas Cailly
- Normandie University, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France.,Department of Nuclear Medicine, CHU Cote de Nacre, 14000 Caen, France.,Normandie University, UNICAEN, IMOGERE, 14000 Caen, France
| | - Frédéric Fabis
- Normandie University, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, 1206 Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, 1206 Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| |
Collapse
|
6
|
Tournier BB, Tsartsalis S, Ceyzériat K, Medina Z, Fraser BH, Grégoire MC, Kövari E, Millet P. Fluorescence-activated cell sorting to reveal the cell origin of radioligand binding. J Cereb Blood Flow Metab 2020; 40:1242-1255. [PMID: 31242048 PMCID: PMC7238369 DOI: 10.1177/0271678x19860408] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many studies have explored the role of TSPO (18 kDa translocator protein) as a marker of neuroinflammation using single-photon emission computed tomography (SPECT) or positron emission tomography (PET). In vivo imaging does not allow to determine the cells in which TSPO is altered. We propose a methodology based on fluorescence-activated cell sorting to sort different cell types of radioligand-treated tissues. We compared left/right hippocampus of rats in response to a unilateral injection of lipopolysaccharide (LPS), ciliary neurotrophic factor (CNTF) or saline. We finally applied this methodology in human samples (Alzheimer's disease patients and controls). Our data show that the pattern of TSPO overexpression differs across animal models of acute neuroinflammation. LPS induces a microglial expansion and an increase in microglial TSPO binding. CNTF is associated with an increase in TSPO binding in microglia and astrocytes in association with an increase in the number of microglial binding sites per cell. In humans, we show that the increase in CLINDE binding in Alzheimer's disease concerns microglia and astrocytes in the presence of a microglial expansion. Thus, the cellular basis of TSPO overexpression is condition dependent, and alterations in TSPO binding found in PET/SPECT imaging studies cannot be attributed to particular cell types indiscriminately.
Collapse
Affiliation(s)
- Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland.,Division of Nuclear medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Zadith Medina
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Ben H Fraser
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Marie-Claude Grégoire
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Enikö Kövari
- Division of Geriatric Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| |
Collapse
|
7
|
Nutma E, Stephenson JA, Gorter RP, de Bruin J, Boucherie DM, Donat CK, Breur M, van der Valk P, Matthews PM, Owen DR, Amor S. A quantitative neuropathological assessment of translocator protein expression in multiple sclerosis. Brain 2020; 142:3440-3455. [PMID: 31578541 PMCID: PMC6821167 DOI: 10.1093/brain/awz287] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/11/2019] [Accepted: 07/25/2019] [Indexed: 01/09/2023] Open
Abstract
The 18 kDa translocator protein (TSPO) is increasingly used to study brain and spinal cord inflammation in degenerative diseases of the CNS such as multiple sclerosis. The enhanced TSPO PET signal that arises during disease is widely considered to reflect activated pathogenic microglia, although quantitative neuropathological data to support this interpretation have not been available. With the increasing interest in the role of chronic microglial activation in multiple sclerosis, characterising the cellular neuropathology associated with TSPO expression is of clear importance for understanding the cellular and pathological processes on which TSPO PET imaging is reporting. Here we have studied the cellular expression of TSPO and specific binding of two TSPO targeting radioligands (3H-PK11195 and 3H-PBR28) in tissue sections from 42 multiple sclerosis cases and 12 age-matched controls. Markers of homeostatic and reactive microglia, astrocytes, and lymphocytes were used to investigate the phenotypes of cells expressing TSPO. There was an approximate 20-fold increase in cells double positive for TSPO and HLA-DR in active lesions and in the rim of chronic active lesion, relative to normal appearing white matter. TSPO was uniformly expressed across myeloid cells irrespective of their phenotype, rather than being preferentially associated with pro-inflammatory microglia or macrophages. TSPO+ astrocytes were increased up to 7-fold compared to normal-appearing white matter across all lesion subtypes and accounted for 25% of the TSPO+ cells in these lesions. To relate TSPO protein expression to ligand binding, specific binding of the TSPO ligands 3H-PK11195 and 3H-PBR28 was determined in the same lesions. TSPO radioligand binding was increased up to seven times for 3H-PBR28 and up to two times for 3H-PK11195 in active lesions and the centre of chronic active lesions and a strong correlation was found between the radioligand binding signal for both tracers and the number of TSPO+ cells across all of the tissues examined. In summary, in multiple sclerosis, TSPO expression arises from microglia of different phenotypes, rather than being restricted to microglia which express classical pro-inflammatory markers. While the majority of cells expressing TSPO in active lesions or chronic active rims are microglia/macrophages, our findings also emphasize the significant contribution of activated astrocytes, as well as smaller contributions from endothelial cells. These observations establish a quantitative framework for interpretation of TSPO in multiple sclerosis and highlight the need for neuropathological characterization of TSPO expression for the interpretation of TSPO PET in other neurodegenerative disorders.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC, Location VUmc, The Netherlands
| | - Jodie A Stephenson
- Department of Pathology, Amsterdam UMC, Location VUmc, The Netherlands.,Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Rianne P Gorter
- Department of Pathology, Amsterdam UMC, Location VUmc, The Netherlands
| | - Joy de Bruin
- Department of Pathology, Amsterdam UMC, Location VUmc, The Netherlands
| | | | | | - Marjolein Breur
- Department of Pathology, Amsterdam UMC, Location VUmc, The Netherlands
| | - Paul van der Valk
- Department of Pathology, Amsterdam UMC, Location VUmc, The Netherlands
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, UK.,UK Dementia Research Institute, Imperial College London, UK
| | - David R Owen
- Department of Brain Sciences, Imperial College London, UK
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, Location VUmc, The Netherlands.,Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| |
Collapse
|
8
|
Recent Developments in TSPO PET Imaging as A Biomarker of Neuroinflammation in Neurodegenerative Disorders. Int J Mol Sci 2019; 20:ijms20133161. [PMID: 31261683 PMCID: PMC6650818 DOI: 10.3390/ijms20133161] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is an inflammatory response in the brain and spinal cord, which can involve the activation of microglia and astrocytes. It is a common feature of many central nervous system disorders, including a range of neurodegenerative disorders. An overlap between activated microglia, pro-inflammatory cytokines and translocator protein (TSPO) ligand binding was shown in early animal studies of neurodegeneration. These findings have been translated in clinical studies, where increases in TSPO positron emission tomography (PET) signal occur in disease-relevant areas across a broad spectrum of neurodegenerative diseases. While this supports the use of TSPO PET as a biomarker to monitor response in clinical trials of novel neurodegenerative therapeutics, the clinical utility of current TSPO PET radioligands has been hampered by the lack of high affinity binding to a prevalent form of polymorphic TSPO (A147T) compared to wild type TSPO. This review details recent developments in exploration of ligand-sensitivity to A147T TSPO that have yielded ligands with improved clinical utility. In addition to developing a non-discriminating TSPO ligand, the final frontier of TSPO biomarker research requires developing an understanding of the cellular and functional interpretation of the TSPO PET signal. Recent insights resulting from single cell analysis of microglial phenotypes are reviewed.
Collapse
|
9
|
Mravec B, Horvathova L, Cernackova A. Hypothalamic Inflammation at a Crossroad of Somatic Diseases. Cell Mol Neurobiol 2019; 39:11-29. [PMID: 30377908 PMCID: PMC11469881 DOI: 10.1007/s10571-018-0631-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/24/2018] [Indexed: 02/08/2023]
Abstract
Various hypothalamic nuclei function as central parts of regulators that maintain homeostasis of the organism. Recently, findings have shown that inflammation in the hypothalamus may significantly affect activity of these homeostats and consequently participate in the development of various somatic diseases such as obesity, diabetes, hypertension, and cachexia. In addition, hypothalamic inflammation may also affect aging and lifespan. Identification of the causes and mechanisms involved in the development of hypothalamic inflammation creates not only a basis for better understanding of the etiopathogenesis of somatic diseases, but for the development of new therapeutic approaches for their treatment, as well.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 813 72, Bratislava, Slovakia.
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia.
| | - Lubica Horvathova
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alena Cernackova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 813 72, Bratislava, Slovakia
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
10
|
TSPO in diverse CNS pathologies and psychiatric disease: A critical review and a way forward. Pharmacol Ther 2018; 194:44-58. [PMID: 30189290 DOI: 10.1016/j.pharmthera.2018.09.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The use of Translocator Protein 18 kDa (TSPO) as a clinical neuroimaging biomarker of brain injury and neuroinflammation has increased exponentially in the last decade. There has been a furious pace in the development of new radiotracers for TSPO positron emission tomography (PET) imaging and its use has now been extensively described in many neurological and mental disorders. This fast pace of research and the ever-increasing number of new laboratories entering the field often times lack an appreciation of the historical perspective of the field and introduce dogmatic, but unproven facts, related to the underlying neurobiology of the TSPO response to brain injury and neuroinflammation. Paradoxically, while in neurodegenerative disorders and in all types of CNS pathologies brain TSPO levels increase, a new observation in psychiatric disorders such as schizophrenia is decreased brain levels of TSPO measured by PET. The neurobiological bases for this new finding is currently not known, but rigorous experimental design using multiple experimental approaches and careful interpretation of results is critically important to provide the methodological and/or biological underpinnings to this new observation. This review provides a perspective of the early history of validating TSPO as a biomarker of brain injury and neuroinflammation and a critical analysis of controversial topics in the literature related to the cellular sources of the TSPO response. The latter is important in order to provide the correct interpretation of PET studies in neurodegenerative and psychiatric disorders. Furthermore, this review proposes some yet to be explored explanations to new findings in psychiatric disorders and new approaches to quantitatively assess the glial sources of the TSPO response in order to move the field forward.
Collapse
|
11
|
Oleoylethanolamide treatment reduces neurobehavioral deficits and brain pathology in a mouse model of Gulf War Illness. Sci Rep 2018; 8:12921. [PMID: 30150699 PMCID: PMC6110778 DOI: 10.1038/s41598-018-31242-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/15/2018] [Indexed: 12/12/2022] Open
Abstract
There are nearly 250,000 Gulf War (GW) veterans who suffer from Gulf War Illness (GWI), a multi-symptom condition that remains untreatable. The main objective was to determine if targeting peroxisomal function could be of therapeutic value in GWI. We performed a pilot study that showed accumulation of very long chain fatty acids (VLCFA), which are metabolized in peroxisomes, in plasma from veterans with GWI. We then examined if targeting peroxisomal β-oxidation with oleoylethanolamide (OEA) restores these lipids to the normal levels and mitigates neuroinflammation and neurobehavioral deficits in a well-established mouse model of GWI. In GWI mice, treatment with OEA corresponded with cognitive benefits and reduced fatigue and disinhibition-like behavior in GWI mice. Biochemical and molecular analysis of the brain tissue showed reduced astroglia and microglia staining, decreased levels of chemokines and cytokines, and decreased NFκB phosphorylation. Treatment with OEA reduced accumulation of peroxisome specific VLCFA in the brains of GWI mice. These studies further support the translational value of targeting peroxisomes. We expect that OEA may be a potential therapy for treating neurobehavioral symptoms and the underlying lipid dysfunction and neuroinflammation associated with GWI. Oleoylethanolamide is available as a dietary supplement, making it appealing for human translational studies.
Collapse
|
12
|
Valotassiou V, Malamitsi J, Papatriantafyllou J, Dardiotis E, Tsougos I, Psimadas D, Alexiou S, Hadjigeorgiou G, Georgoulias P. SPECT and PET imaging in Alzheimer’s disease. Ann Nucl Med 2018; 32:583-593. [PMID: 30128693 DOI: 10.1007/s12149-018-1292-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Varvara Valotassiou
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece.
| | - Julia Malamitsi
- Medical Physics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Ioannis Tsougos
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - Dimitrios Psimadas
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - Sotiria Alexiou
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| | - George Hadjigeorgiou
- Neurology Department, University Hospital of Larissa, Thessaly, Greece
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Greece
| | - Panagiotis Georgoulias
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, 41110, Larissa, Thessaly, Greece
| |
Collapse
|
13
|
Foucault-Fruchard L, Doméné A, Page G, Windsor M, Emond P, Rodrigues N, Dollé F, Damont A, Buron F, Routier S, Chalon S, Antier D. Neuroprotective effect of the alpha 7 nicotinic receptor agonist PHA 543613 in an in vivo excitotoxic adult rat model. Neuroscience 2017; 356:52-63. [PMID: 28527955 DOI: 10.1016/j.neuroscience.2017.05.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/03/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022]
Abstract
Neuroinflammation is a key component of the pathophysiology of neurodegenerative diseases. The link between nicotine intake and positive outcome has been established, suggesting a role played by nicotinic receptors (nAChRs), especially α7nAChRs. The objective of this study was to evaluate the potential dose effects of PHA 543613 on neuron survival and striatal microglial activation in a rat model of brain excitotoxicity. A preliminary study was performed in vitro to confirm PHA 543613 agonist properties on α7nAChRs. Rats were lesioned in the right striatum with quinolinic acid (QA) and received either vehicle or PHA 543613 at 6 or 12mg/kg twice a day until sacrifice at Day 4 post-lesion. We first compared the translocator protein quantitative autoradiography in QA-lesioned brains with [3H]DPA-714 and [3H]PK-11195. The effects of PHA 543613 on microglial activation and neuronal survival were then evaluated through [3H]DPA-714 binding and immunofluorescence staining (Ox-42, NeuN) on adjacent brain sections. We demonstrated that [3H]DPA-714 provides a better signal-to-noise ratio than [3H]PK-11195. Furthermore, we showed that repeated PHA 543613 administration at a dose of 12mg/kg to QA-lesioned rats significantly protected neurons and reduced the intensity of microglial activation. This study reinforces the hypothesis that α7nAChR agonists can provide beneficial effects in the treatment of neurodegenerative diseases through potential modulation of microglial activation.
Collapse
Affiliation(s)
- Laura Foucault-Fruchard
- UMR INSERM U930, Université François Rabelais, Tours, France; CHRU de Tours, Hôpital Bretonneau, Tours, France.
| | - Aurélie Doméné
- UMR INSERM U930, Université François Rabelais, Tours, France.
| | - Guylène Page
- EA3808 - CiMoTheMA, Université de Poitiers, Poitiers, France.
| | | | - Patrick Emond
- UMR INSERM U930, Université François Rabelais, Tours, France.
| | - Nuno Rodrigues
- UMR CNRS 7311, Institut de Chimie Organique et Analytique, Université d'Orléans, Orléans, France.
| | - Frédéric Dollé
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France.
| | | | - Frédéric Buron
- UMR CNRS 7311, Institut de Chimie Organique et Analytique, Université d'Orléans, Orléans, France.
| | - Sylvain Routier
- UMR CNRS 7311, Institut de Chimie Organique et Analytique, Université d'Orléans, Orléans, France.
| | - Sylvie Chalon
- UMR INSERM U930, Université François Rabelais, Tours, France.
| | - Daniel Antier
- UMR INSERM U930, Université François Rabelais, Tours, France; CHRU de Tours, Hôpital Bretonneau, Tours, France.
| |
Collapse
|
14
|
Feng L, Jensen P, Thomsen G, Dyssegaard A, Svarer C, Knudsen LV, Møller K, Thomsen C, Mikkelsen JD, Guilloteau D, Knudsen GM, Pinborg LH. The Variability of Translocator Protein Signal in Brain and Blood of Genotyped Healthy Humans Using In Vivo 123I-CLINDE SPECT Imaging: A Test–Retest Study. J Nucl Med 2016; 58:989-995. [DOI: 10.2967/jnumed.116.183202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 10/21/2016] [Indexed: 11/16/2022] Open
|
15
|
Poutiainen P, Jaronen M, Quintana FJ, Brownell AL. Precision Medicine in Multiple Sclerosis: Future of PET Imaging of Inflammation and Reactive Astrocytes. Front Mol Neurosci 2016; 9:85. [PMID: 27695400 PMCID: PMC5023680 DOI: 10.3389/fnmol.2016.00085] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 08/30/2016] [Indexed: 12/29/2022] Open
Abstract
Non-invasive molecular imaging techniques can enhance diagnosis to achieve successful treatment, as well as reveal underlying pathogenic mechanisms in disorders such as multiple sclerosis (MS). The cooperation of advanced multimodal imaging techniques and increased knowledge of the MS disease mechanism allows both monitoring of neuronal network and therapeutic outcome as well as the tools to discover novel therapeutic targets. Diverse imaging modalities provide reliable diagnostic and prognostic platforms to better achieve precision medicine. Traditionally, magnetic resonance imaging (MRI) has been considered the golden standard in MS research and diagnosis. However, positron emission tomography (PET) imaging can provide functional information of molecular biology in detail even prior to anatomic changes, allowing close follow up of disease progression and treatment response. The recent findings support three major neuroinflammation components in MS: astrogliosis, cytokine elevation, and significant changes in specific proteins, which offer a great variety of specific targets for imaging purposes. Regardless of the fact that imaging of astrocyte function is still a young field and in need for development of suitable imaging ligands, recent studies have shown that inflammation and astrocyte activation are related to progression of MS. MS is a complex disease, which requires understanding of disease mechanisms for successful treatment. PET is a precise non-invasive imaging method for biochemical functions and has potential to enhance early and accurate diagnosis for precision therapy of MS. In this review we focus on modulation of different receptor systems and inflammatory aspect of MS, especially on activation of glial cells, and summarize the recent findings of PET imaging in MS and present the most potent targets for new biomarkers with the main focus on experimental MS research.
Collapse
Affiliation(s)
- Pekka Poutiainen
- Athinoula A Martinos Biomedical Imaging Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, MA, USA
| | - Merja Jaronen
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical SchoolBoston, MA, USA
| | - Francisco J. Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical SchoolBoston, MA, USA
| | - Anna-Liisa Brownell
- Athinoula A Martinos Biomedical Imaging Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, MA, USA
| |
Collapse
|
16
|
Cacheux F, Médran-Navarrete V, Dollé F, Marguet F, Puech F, Damont A. Synthesis and in vitro characterization of novel fluorinated derivatives of the translocator protein 18 kDa ligand CfO-DPA-714. Eur J Med Chem 2016; 125:346-359. [PMID: 27688189 DOI: 10.1016/j.ejmech.2016.09.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 11/30/2022]
Abstract
The translocator protein 18 kDa (TSPO) is today a validated target for a number of therapeutic applications, but also a well-recognized diagnostic/imaging biomarker for the evaluation of inflammatory related-disease state and progression, prompting the development of specific and dedicated TSPO ligands worldwide. For this purpose, pyrazolo[1,5-a]pyrimidine acetamides constitute a unique class of high affinity and selectivity TSPO ligands; it includes DPA-714, a fluorine-containing derivative that has also been labelled with the positron-emitter fluorine-18, and is nowadays widely used as a Positron Emission Tomography imaging probe. Recently, to prevent defluorination issues encountered in vivo with this tracer, a first series of analogues was reported where the oxygen atom bridging the phenyl ring of the core structure and the fluorinated moiety was replaced with a more robust linkage. Among this new series, CfO-DPA-714 was discovered as a highly promising TSPO ligand. Herein, a novel series of fluorinated analogues of the latter molecule were synthesized and in vitro characterized, where the pharmacomodulation at the amide position of the molecule was explored. Thirteen compounds were thus prepared from a common key-ester intermediate (synthesized in 7 steps from 4-iodobenzoate - 11% overall yield) and a set of commercially available amines and obtained with moderate to good yields (23-81%) and high purities (>95%). With one exception, all derivatives displayed nanomolar to subnanomolar affinity for the TSPO and also high selectivity versus the CBR (Ki (CBR)/Ki (TSPO) > 103). Within this series, three compounds showed better Ki values (0.25, 0.26 and 0.30 nM) than that of DPA-714 (0.91 nM) and CfO-DPA-714 (0.37 nM), and favorable lipophilicity for brain penetration (3.6 < logD7.4 < 4.4). Among these three compounds, the N-methyl-N-propyl amide analogue (9) exhibited similar metabolic stability when compared to CfO-DPA-714 in mouse, rat and human microsomes. Therefore, the latter compound stands out as a promising candidate for drug development or for use as a PET probe, once fluorine-18-labelled, for in vivo neuroinflammation imaging.
Collapse
Affiliation(s)
- Fanny Cacheux
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France; Inserm/CEA/Université Paris Sud, UMR 1023, ERL 9218 CNRS, IMIV, Université Paris-Saclay, Orsay, France
| | - Vincent Médran-Navarrete
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France; Inserm/CEA/Université Paris Sud, UMR 1023, ERL 9218 CNRS, IMIV, Université Paris-Saclay, Orsay, France
| | - Frédéric Dollé
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France; Inserm/CEA/Université Paris Sud, UMR 1023, ERL 9218 CNRS, IMIV, Université Paris-Saclay, Orsay, France
| | | | | | - Annelaure Damont
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France; Inserm/CEA/Université Paris Sud, UMR 1023, ERL 9218 CNRS, IMIV, Université Paris-Saclay, Orsay, France.
| |
Collapse
|
17
|
Changes in Binding of [(123)I]CLINDE, a High-Affinity Translocator Protein 18 kDa (TSPO) Selective Radioligand in a Rat Model of Traumatic Brain Injury. Neuromolecular Med 2016; 18:158-69. [PMID: 26969181 DOI: 10.1007/s12017-016-8385-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/02/2016] [Indexed: 01/01/2023]
Abstract
After traumatic brain injury (TBI), secondary injuries develop, including neuroinflammatory processes that contribute to long-lasting impairments. These secondary injuries represent potential targets for treatment and diagnostics. The translocator protein 18 kDa (TSPO) is expressed in activated microglia cells and upregulated in response to brain injury and therefore a potential biomarker of the neuroinflammatory processes. Second-generation radioligands of TSPO, such as [(123)I]CLINDE, have a higher signal-to-noise ratio as the prototype ligand PK11195. [(123)I]CLINDE has been employed in human studies using single-photon emission computed tomography to image the neuroinflammatory response after stroke. In this study, we used the same tracer in a rat model of TBI to determine changes in TSPO expression. Adult Sprague-Dawley rats were subjected to moderate controlled cortical impact injury and sacrificed at 6, 24, 72 h and 28 days post surgery. TSPO expression was assessed in brain sections employing [(123)I]CLINDE in vitro autoradiography. From 24 h to 28 days post surgery, injured animals exhibited a marked and time-dependent increase in [(123)I]CLINDE binding in the ipsilateral motor, somatosensory and parietal cortex, as well as in the hippocampus and thalamus. Interestingly, binding was also significantly elevated in the contralateral M1 motor cortex following TBI. Craniotomy without TBI caused a less marked increase in [(123)I]CLINDE binding, restricted to the ipsilateral hemisphere. Radioligand binding was consistent with an increase in TSPO mRNA expression and CD11b immunoreactivity at the contusion site. This study demonstrates the applicability of [(123)I]CLINDE for detailed regional and quantitative assessment of glial activity in experimental models of TBI.
Collapse
|
18
|
Radiopharmaceuticals for PET imaging of neuroinflammation. MEDECINE NUCLEAIRE-IMAGERIE FONCTIONNELLE ET METABOLIQUE 2016. [DOI: 10.1016/j.mednuc.2016.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
19
|
Insight into the Molecular Imaging of Alzheimer's Disease. Int J Biomed Imaging 2016; 2016:7462014. [PMID: 26880871 PMCID: PMC4736963 DOI: 10.1155/2016/7462014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/16/2015] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease is a complex neurodegenerative disease affecting millions of individuals worldwide. Earlier it was diagnosed only via clinical assessments and confirmed by postmortem brain histopathology. The development of validated biomarkers for Alzheimer's disease has given impetus to improve diagnostics and accelerate the development of new therapies. Functional imaging like positron emission tomography (PET), single photon emission computed tomography (SPECT), functional magnetic resonance imaging (fMRI), and proton magnetic resonance spectroscopy provides a means of detecting and characterising the regional changes in brain blood flow, metabolism, and receptor binding sites that are associated with Alzheimer's disease. Multimodal neuroimaging techniques have indicated changes in brain structure and metabolic activity, and an array of neurochemical variations that are associated with neurodegenerative diseases. Radiotracer-based PET and SPECT potentially provide sensitive, accurate methods for the early detection of disease. This paper presents a review of neuroimaging modalities like PET, SPECT, and selected imaging biomarkers/tracers used for the early diagnosis of AD. Neuroimaging with such biomarkers and tracers could achieve a much higher diagnostic accuracy for AD and related disorders in the future.
Collapse
|
20
|
Lee DE, Yue X, Ibrahim WG, Lentz MR, Peterson KL, Jagoda EM, Kassiou M, Maric D, Reid WC, Hammoud DA. Lack of neuroinflammation in the HIV-1 transgenic rat: an [(18)F]-DPA714 PET imaging study. J Neuroinflammation 2015; 12:171. [PMID: 26377670 PMCID: PMC4574011 DOI: 10.1186/s12974-015-0390-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/02/2015] [Indexed: 11/29/2022] Open
Abstract
Background HIV-associated neuroinflammation is believed to be a major contributing factor in the development of HIV-associated neurocognitive disorders (HAND). In this study, we used micropositron emission tomography (PET) imaging to quantify neuroinflammation in HIV-1 transgenic rat (Tg), a small animal model of HIV, known to develop neurological and behavioral problems. Methods Dynamic [18F]DPA-714 PET imaging was performed in Tg and age-matched wild-type (WT) rats in three age groups: 3-, 9-, and 16-month-old animals. As a positive control for neuroinflammation, we performed unilateral intrastriatal injection of quinolinic acid (QA) in a separate group of WT rats. To confirm our findings, we performed multiplex immunofluorescent staining for Iba1 and we measured cytokine/chemokine levels in brain lysates of Tg and WT rats at different ages. Results [18F]DPA-714 uptake in HIV-1 Tg rat brains was generally higher than in age-matched WT rats but this was not statistically significant in any age group. [18F]DPA-714 uptake in the QA-lesioned rats was significantly higher ipsilateral to the lesion compared to contralateral side indicating neuroinflammatory changes. Iba1 immunofluorescence showed no significant differences in microglial activation between the Tg and WT rats, while the QA-lesioned rats showed significant activation. Finally, cytokine/chemokine levels in brain lysates of the Tg rats and WT rats were not significantly different. Conclusion Microglial activation might not be the primary mechanism for neuropathology in the HIV-1 Tg rats. Although [18F]DPA-714 is a good biomarker of neuroinflammation, it cannot be reliably used as an in vivo biomarker of neurodegeneration in the HIV-1 Tg rat. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0390-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dianne E Lee
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wael G Ibrahim
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Margaret R Lentz
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Kristin L Peterson
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Elaine M Jagoda
- Molecular Imaging Program (MIP), National Cancer Institute (NCI), Bethesda, MD, USA
| | - Michael Kassiou
- Chemistry Department, The University of Sydney, Sydney, Australia
| | - Dragan Maric
- Division of Intermural Research (DIR), National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - William C Reid
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, National Institutes of Health/Clinical Center, 10 Center Drive, Room 1C368, Bethesda, MD, 20814-9692, USA.
| |
Collapse
|
21
|
Lu FM, Yuan Z. PET/SPECT molecular imaging in clinical neuroscience: recent advances in the investigation of CNS diseases. Quant Imaging Med Surg 2015; 5:433-47. [PMID: 26029646 PMCID: PMC4426104 DOI: 10.3978/j.issn.2223-4292.2015.03.16] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/23/2015] [Indexed: 01/10/2023]
Abstract
Molecular imaging is an attractive technology widely used in clinical practice that greatly enhances our understanding of the pathophysiology and treatment in central nervous system (CNS) diseases. It is a novel multidisciplinary technique that can be defined as real-time visualization, in vivo characterization and qualification of biological processes at the molecular and cellular level. It involves the imaging modalities and the corresponding imaging agents. Nowadays, molecular imaging in neuroscience has provided tremendous insights into disturbed human brain function. Among all of the molecular imaging modalities, positron emission tomography (PET) and single photon emission computed tomography (SPECT) have occupied a particular position that visualize and measure the physiological processes using high-affinity and high-specificity molecular radioactive tracers as imaging probes in intact living brain. In this review, we will put emphasis on the PET/SPECT applications in Alzheimer's disease (AD) and Parkinson's disease (PD) as major CNS disorders. We will first give an overview of the main classical molecular neuroimaging modalities. Then, the major clinical applications of PET and SPECT along with molecular probes in the fields of psychiatry and neurology will be discussed.
Collapse
Affiliation(s)
- Feng-Mei Lu
- Bioimaging Core, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Zhen Yuan
- Bioimaging Core, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| |
Collapse
|
22
|
Feng L, Svarer C, Thomsen G, de Nijs R, Larsen VA, Jensen P, Adamsen D, Dyssegaard A, Fischer W, Meden P, Krieger D, Møller K, Knudsen GM, Pinborg LH. In Vivo Quantification of Cerebral Translocator Protein Binding in Humans Using 6-Chloro-2-(4′-123I-Iodophenyl)-3-(N,N-Diethyl)-Imidazo[1,2-a]Pyridine-3-Acetamide SPECT. J Nucl Med 2014; 55:1966-72. [DOI: 10.2967/jnumed.114.143727] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
23
|
Liu G, Middleton RJ, Hatty CR, Kam WW, Chan R, Pham T, Harrison‐Brown M, Dodson E, Veale K, Banati RB. The 18 kDa translocator protein, microglia and neuroinflammation. Brain Pathol 2014; 24:631-53. [PMID: 25345894 PMCID: PMC8029074 DOI: 10.1111/bpa.12196] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is expressed in the injured brain. It has become known as an imaging marker of "neuroinflammation" indicating active disease, and is best interpreted as a nondiagnostic biomarker and disease staging tool that refers to histopathology rather than disease etiology. The therapeutic potential of TSPO as a drug target is mostly based on the understanding that it is an outer mitochondrial membrane protein required for the translocation of cholesterol, which thus regulates the rate of steroid synthesis. This pivotal role together with the evolutionary conservation of TSPO has underpinned the belief that any loss or mutation of TSPO should be associated with significant physiological deficits or be outright incompatible with life. However, against prediction, full Tspo knockout mice are viable and across their lifespan do not show the phenotype expected if cholesterol transport and steroid synthesis were significantly impaired. Thus, the "translocation" function of TSPO remains to be better substantiated. Here, we discuss the literature before and after the introduction of the new nomenclature for TSPO and review some of the newer findings. In light of the controversy surrounding the function of TSPO, we emphasize the continued importance of identifying compounds with confirmed selectivity and suggest that TSPO expression is analyzed within specific disease contexts rather than merely equated with the reified concept of "neuroinflammation."
Collapse
Affiliation(s)
- Guo‐Jun Liu
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ryan J. Middleton
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Claire R. Hatty
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Winnie Wai‐Ying Kam
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ronald Chan
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Tien Pham
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Meredith Harrison‐Brown
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Eoin Dodson
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Kelly Veale
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Richard B. Banati
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
- National Imaging Facility and Ramaciotti Brain Imaging CentreSydneyNSWAustralia
| |
Collapse
|
24
|
Callaghan PD, Wimberley CA, Rahardjo GL, Berghofer PJ, Pham TQ, Jackson T, Zahra D, Bourdier T, Wyatt N, Greguric I, Howell NR, Siegele R, Pastuovic Z, Mattner F, Loc’h C, Gregoire MC, Katsifis A. Comparison of in vivo binding properties of the 18-kDa translocator protein (TSPO) ligands [18F]PBR102 and [18F]PBR111 in a model of excitotoxin-induced neuroinflammation. Eur J Nucl Med Mol Imaging 2014; 42:138-51. [DOI: 10.1007/s00259-014-2895-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 08/13/2014] [Indexed: 12/22/2022]
|
25
|
Arlicot N, Tronel C, Bodard S, Garreau L, de la Crompe B, Vandevelde I, Guilloteau D, Antier D, Chalon S. Translocator Protein (18 kDa) Mapping with [
125
I]-CLINDE in the Quinolinic Acid Rat Model of Excitotoxicity: A Longitudinal Comparison with Microglial Activation, Astrogliosis, and Neuronal Death. Mol Imaging 2014. [DOI: 10.2310/7290.2013.00075] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Nicolas Arlicot
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Claire Tronel
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Sylvie Bodard
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Lucette Garreau
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Brice de la Crompe
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Inge Vandevelde
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Denis Guilloteau
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Daniel Antier
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| | - Sylvie Chalon
- From Inserm, U930; Université François-Rabelais de Tours, UMR-U930; CHRU de Tours, Hôpital Bretonneau, Pôle Santé Publique – Produits de Santé; and CHRU de Tours, Hôpital Bretonneau, Service de Médecine Nucléaire In Vitro, Tours, France
| |
Collapse
|
26
|
Tronel C, Page G, Bodard S, Chalon S, Antier D. The specific PKR inhibitor C16 prevents apoptosis and IL-1β production in an acute excitotoxic rat model with a neuroinflammatory component. Neurochem Int 2013; 64:73-83. [PMID: 24211709 DOI: 10.1016/j.neuint.2013.10.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/10/2013] [Accepted: 10/22/2013] [Indexed: 12/20/2022]
Abstract
The double-stranded RNA-dependent protein kinase (PKR), an apoptotic inducer, regulates much pro-inflammatory cytokine production. The purpose of this study was to evaluate in vivo the effects of the specific PKR inhibitor C16 in the striatum in an acute excitotoxic rat model with an important neuroinflammatory component. Inflammation was induced by unilateral striatal injection of quinolinic acid (QA) in 10-week-old normotensive rats. Animals were separated into groups receiving either vehicle or C16 for both sham and QA rats. The effects were assessed in ipsi- and contralateral striata by immunoblotting for PKR activation, by Luminex assay for cytokine levels and by immunofluorescent staining for cleaved caspase-3 to detect neuronal apoptosis. The highest dose of C16 (600μg/kg; C16-2) in QA rats reduced expression of the active catalytic domain of the PKR vs. that in vehicle-injected QA rats. A robust increase of IL-1β levels on the contralateral side of QA rats was prevented by C16-2 (97% inhibition). Macroscopic and microscopic observation of cerebral tissue (Hematoxylin & Eosin staining) revealed that tissue integrity was more preserved with C16-2 treatment than its vehicle in QA rats. Furthermore, C16-2 treatment decreased by 47% the neuronal loss and by 37% the number of positive cleaved caspase-3 neurons induced by QA injection. In conclusion, C16 prevented not only the PKR-induced neuronal loss but also the inflammatory response in this acute excitotoxic in vivo model, highlighting its promising neuroprotective properties to rescue acute brain lesions.
Collapse
Affiliation(s)
- C Tronel
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France.
| | - G Page
- EA 3808, University of Poitiers, "Molecular Targets and Therapeutics of Alzheimer's Disease (CiMoTheMA)", 6 rue de la Milétrie, BP 199, 86034 Poitiers, France
| | - S Bodard
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France
| | - S Chalon
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France
| | - D Antier
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France
| |
Collapse
|
27
|
Motbey CP, Clemens KJ, Apetz N, Winstock AR, Ramsey J, Li KM, Wyatt N, Callaghan PD, Bowen MT, Cornish JL, McGregor IS. High levels of intravenous mephedrone (4-methylmethcathinone) self-administration in rats: neural consequences and comparison with methamphetamine. J Psychopharmacol 2013; 27:823-36. [PMID: 23739178 DOI: 10.1177/0269881113490325] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mephedrone (MMC) is a relatively new recreational drug that has rapidly increased in popularity in recent years. This study explored the characteristics of intravenous MMC self-administration in the rat, with methamphetamine (METH) used as a comparator drug. Male Sprague-Dawley rats were trained to nose poke for intravenous MMC or METH in daily 2 h sessions over a 10 d acquisition period. Dose-response functions were then established under fixed- and progressive-ratio (FR and PR) schedules over three subsequent weeks of testing. Brains were analyzed ex vivo for striatal serotonin (5-HT) and dopamine (DA) levels and metabolites, while autoradiography assessed changes in the regional density of 5-HT and serotonin transporter (SERT) and DA transporter (DAT) and induction of the inflammation marker translocator protein (TSPO). Results showed that MMC was readily and vigorously self-administered via the intravenous route. Under a FR1 schedule, peak responding for MMC was obtained at 0.1 mg/kg/infusion, versus 0.01 mg/kg/infusion for METH. Break points under a PR schedule peaked at 1 mg/kg/infusion MMC versus 0.3 mg/kg/infusion for METH. Final intakes of MMC were 31.3 mg/kg/d compared to 4 mg/kg/d for METH. Rats self-administering MMC, but not METH, gained weight at a slower rate than control rats. METH, but not MMC, self-administration elevated TSPO receptor density in the nucleus accumbens and hippocampus, while MMC, but not METH, self-administration decreased striatal 5-hydroxyindolacetic acid (5-HIAA) concentrations. In summary, MMC supported high levels of self-administration, matching or exceeding those previously reported with other drugs of abuse.
Collapse
Affiliation(s)
- Craig P Motbey
- School of Psychology, University of Sydney, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Martins AF, Morfin JF, Kubíčková A, Kubíček V, Buron F, Suzenet F, Salerno M, Lazar AN, Duyckaerts C, Arlicot N, Guilloteau D, Geraldes CFGC, Tóth É. PiB-Conjugated, Metal-Based Imaging Probes: Multimodal Approaches for the Visualization of β-Amyloid Plaques. ACS Med Chem Lett 2013; 4:436-40. [PMID: 24900692 DOI: 10.1021/ml400042w] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/14/2013] [Indexed: 01/19/2023] Open
Abstract
In an effort toward the visualization of β-amyloid plaques by in vivo imaging techniques, we have conjugated an optimized derivative of the Pittsburgh compound B (PiB), a well-established marker of Aβ plaques, to DO3A-monoamide that is capable of forming stable, noncharged complexes with different trivalent metal ions including Gd(3+) for MRI and (111)In(3+) for SPECT applications. Proton relaxivity measurements evidenced binding of Gd(DO3A-PiB) to the amyloid peptide Aβ1-40 and to human serum albumin, resulting in a two- and four-fold relaxivity increase, respectively. Ex vivo immunohistochemical studies showed that the DO3A-PiB complexes selectively target Aβ plaques on Alzheimer's disease human brain tissue. Ex vivo biodistribution data obtained for the (111)In-analogue pointed to a moderate blood-brain barrier (BBB) penetration in adult male Swiss mice (without amyloid deposits) with 0.36% ID/g in the cortex at 2 min postinjection.
Collapse
Affiliation(s)
- André F. Martins
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
- Department of
Life Sciences,
Center of Neurosciences and Cell Biology (CNC), and Coimbra Chemistry
Center, University of Coimbra, Portugal
| | - Jean-François Morfin
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
| | - Anna Kubíčková
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
- Department of Analytical Chemistry, Charles University in Prague, Albertov 2030, 12840
Prague, Czech Republic
| | - Vojtěch Kubíček
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
| | - Frédéric Buron
- Institut de Chimie Organique
et Analytique, UMR 7311 CNRS/Université d’Orléans, rue de Chartres, 45067 Orléans, France
| | - Franck Suzenet
- Institut de Chimie Organique
et Analytique, UMR 7311 CNRS/Université d’Orléans, rue de Chartres, 45067 Orléans, France
| | - Milena Salerno
- Laboratoire CSPBAT, CNRS UMR
7244, UFR-SMBH, Université Paris 13, 74 rue Marcel Cachin, 93017 Bobigny, France
| | - Adina N. Lazar
- Centre de Recherche de l’Institut
du Cerveau et de la Moelle, CNRS UMR7225, INSERM, UMR975 and UPMC, Hôpital de la Pitié-Salpêtrière 47, Bd de l’Hôpital 75013 Paris, France
| | - Charles Duyckaerts
- Centre de Recherche de l’Institut
du Cerveau et de la Moelle, CNRS UMR7225, INSERM, UMR975 and UPMC, Hôpital de la Pitié-Salpêtrière 47, Bd de l’Hôpital 75013 Paris, France
| | - Nicolas Arlicot
- Inserm, U930, Université François Rabelais de Tours, CHRU de Tours, 37044
Tours Cedex 9, France
| | - Denis Guilloteau
- Inserm, U930, Université François Rabelais de Tours, CHRU de Tours, 37044
Tours Cedex 9, France
| | - Carlos F. G. C. Geraldes
- Department of
Life Sciences,
Center of Neurosciences and Cell Biology (CNC), and Coimbra Chemistry
Center, University of Coimbra, Portugal
| | - Éva Tóth
- Centre de Biophysique Moléculaire, CNRS, Rue Charles Sadron, 45071 Orléans Cedex
2, France
| |
Collapse
|
29
|
Oxidative stress is related to the deleterious effects of heme oxygenase-1 in an in vivo neuroinflammatory rat model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:264935. [PMID: 23533686 PMCID: PMC3606782 DOI: 10.1155/2013/264935] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 01/21/2013] [Accepted: 01/22/2013] [Indexed: 01/08/2023]
Abstract
Heme oxygenase-1 (HO-1) induction is associated with beneficial or deleterious effects depending on the experimental conditions adopted and the neurodegenerative rodent models used. The present study aimed first to evaluate the effects of cerebral HO-1 induction in an in vivo rat model of neuroinflammation by intrastriatal injection of quinolinic acid (QA) and secondly to explore the role played by reactive oxygen species (ROS) and free iron (Fe2+) derived from heme catabolism promoted by HO-1. Chronic I.P. treatment with the HO-1 inductor and substrate hemin was responsible for a significant dose-related increase of cerebral HO-1 production. Brain tissue loss, microglial activation, and neuronal death were significantly higher in rats receiving QA plus hemin (H-QA) versus QA and controls. Significant increase of ROS production in H-QA rat brain was inhibited by the specific HO-1 inhibitor ZnPP which supports the idea that ROS level augmentation in hemin-treated animals is a direct consequence of HO-1 induction. The cerebral tissue loss and ROS level in hemin-treated rats receiving the iron chelator deferoxamine were significantly decreased, demonstrating the involvement of Fe2+in brain ROS production. Therefore, the deleterious effects of HO-1 expression in this in vivo neuroinflammatory model were linked to a hyperproduction of ROS, itself promoted by free iron liberation.
Collapse
|
30
|
Benadiba M, Luurtsema G, Wichert-Ana L, Buchpigel CA, Filho GB. New Molecular Targets for PET and SPECT Imaging in Neurodegenerative Diseases. BRAZILIAN JOURNAL OF PSYCHIATRY 2012; 34 Suppl 2:S125-36. [DOI: 10.1016/j.rbp.2012.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 02/29/2012] [Indexed: 01/23/2023]
|
31
|
Motbey CP, Karanges E, Li KM, Wilkinson S, Winstock AR, Ramsay J, Hicks C, Kendig MD, Wyatt N, Callaghan PD, McGregor IS. Mephedrone in adolescent rats: residual memory impairment and acute but not lasting 5-HT depletion. PLoS One 2012; 7:e45473. [PMID: 23029034 PMCID: PMC3445542 DOI: 10.1371/journal.pone.0045473] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 08/17/2012] [Indexed: 11/19/2022] Open
Abstract
Mephedrone (4-methylmethcathinone, MMC) is a popular recreational drug, yet its potential harms are yet to be fully established. The current study examined the impact of single or repeated MMC exposure on various neurochemical and behavioral measures in rats. In Experiment 1 male adolescent Wistar rats received single or repeated (once a day for 10 days) injections of MMC (30 mg/kg) or the comparator drug methamphetamine (METH, 2.5 mg/kg). Both MMC and METH caused robust hyperactivity in the 1 h following injection although this effect did not tend to sensitize with repeated treatment. Striatal dopamine (DA) levels were increased 1 h following either METH or MMC while striatal and hippocampal serotonin (5-HT) levels were decreased 1 h following MMC but not METH. MMC caused greater increases in 5-HT metabolism and greater reductions in DA metabolism in rats that had been previously exposed to MMC. Autoradiographic analysis showed no signs of neuroinflammation ([(125)I]CLINDE ligand used as a marker for translocator protein (TSPO) expression) with repeated exposure to either MMC or METH. In Experiment 2, rats received repeated MMC (7.5, 15 or 30 mg/kg once a day for 10 days) and were examined for residual behavioral effects following treatment. Repeated high (30 mg/kg) dose MMC produced impaired novel object recognition 5 weeks after drug treatment. However, no residual changes in 5-HT or DA tissue levels were observed at 7 weeks post-treatment. Overall these results show that MMC causes acute but not lasting changes in DA and 5-HT tissue concentrations. MMC can also cause long-term memory impairment. Future studies of cognitive function in MMC users are clearly warranted.
Collapse
Affiliation(s)
- Craig P. Motbey
- School of Psychology, University of Sydney, New South Wales, Australia
| | - Emily Karanges
- School of Psychology, University of Sydney, New South Wales, Australia
| | - Kong M. Li
- Department of Pharmacology, University of Sydney, New South Wales, Australia
| | - Shane Wilkinson
- School of Chemistry, University of Sydney, New South Wales, Australia
| | - Adam R. Winstock
- Institute of Psychiatry, Kings College, University of London, United Kingdom
| | - John Ramsay
- TICTAC Communications Ltd., St George’s College, University of London, United Kingdom
| | - Callum Hicks
- School of Psychology, University of Sydney, New South Wales, Australia
| | - Michael D. Kendig
- School of Psychology, University of Sydney, New South Wales, Australia
| | - Naomi Wyatt
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, Sydney, New South Wales, Australia
| | - Paul D. Callaghan
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, Sydney, New South Wales, Australia
| | - Iain S. McGregor
- School of Psychology, University of Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
32
|
James ML, Gambhir SS. A molecular imaging primer: modalities, imaging agents, and applications. Physiol Rev 2012; 92:897-965. [PMID: 22535898 DOI: 10.1152/physrev.00049.2010] [Citation(s) in RCA: 742] [Impact Index Per Article: 57.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Molecular imaging is revolutionizing the way we study the inner workings of the human body, diagnose diseases, approach drug design, and assess therapies. The field as a whole is making possible the visualization of complex biochemical processes involved in normal physiology and disease states, in real time, in living cells, tissues, and intact subjects. In this review, we focus specifically on molecular imaging of intact living subjects. We provide a basic primer for those who are new to molecular imaging, and a resource for those involved in the field. We begin by describing classical molecular imaging techniques together with their key strengths and limitations, after which we introduce some of the latest emerging imaging modalities. We provide an overview of the main classes of molecular imaging agents (i.e., small molecules, peptides, aptamers, engineered proteins, and nanoparticles) and cite examples of how molecular imaging is being applied in oncology, neuroscience, cardiology, gene therapy, cell tracking, and theranostics (therapy combined with diagnostics). A step-by-step guide to answering biological and/or clinical questions using the tools of molecular imaging is also provided. We conclude by discussing the grand challenges of the field, its future directions, and enormous potential for further impacting how we approach research and medicine.
Collapse
Affiliation(s)
- Michelle L James
- Molecular Imaging Program, Department of Radiology, Stanford University, Palo Alto, CA 94305, USA
| | | |
Collapse
|
33
|
Jacobs AH, Tavitian B. Noninvasive molecular imaging of neuroinflammation. J Cereb Blood Flow Metab 2012; 32:1393-415. [PMID: 22549622 PMCID: PMC3390799 DOI: 10.1038/jcbfm.2012.53] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 03/05/2012] [Accepted: 03/23/2012] [Indexed: 12/23/2022]
Abstract
Inflammation is a highly dynamic and complex adaptive process to preserve and restore tissue homeostasis. Originally viewed as an immune-privileged organ, the central nervous system (CNS) is now recognized to have a constant interplay with the innate and the adaptive immune systems, where resident microglia and infiltrating immune cells from the periphery have important roles. Common diseases of the CNS, such as stroke, multiple sclerosis (MS), and neurodegeneration, elicit a neuroinflammatory response with the goal to limit the extent of the disease and to support repair and regeneration. However, various disease mechanisms lead to neuroinflammation (NI) contributing to the disease process itself. Molecular imaging is the method of choice to try to decipher key aspects of the dynamic interplay of various inducers, sensors, transducers, and effectors of the orchestrated inflammatory response in vivo in animal models and patients. Here, we review the basic principles of NI with emphasis on microglia and common neurologic disease mechanisms, the molecular targets which are being used and explored for imaging, and molecular imaging of NI in frequent neurologic diseases, such as stroke, MS, neurodegeneration, epilepsy, encephalitis, and gliomas.
Collapse
Affiliation(s)
- Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI) at the Westfalian Wilhelms-University of Münster (WWU), Münster, Germany.
| | | |
Collapse
|
34
|
Maia S, Arlicot N, Vierron E, Bodard S, Vergote J, Guilloteau D, Chalon S. Longitudinal and parallel monitoring of neuroinflammation and neurodegeneration in a 6-hydroxydopamine rat model of Parkinson's disease. Synapse 2012; 66:573-83. [DOI: 10.1002/syn.21543] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 01/24/2012] [Indexed: 12/20/2022]
|
35
|
Collins LM, Toulouse A, Connor TJ, Nolan YM. Contributions of central and systemic inflammation to the pathophysiology of Parkinson's disease. Neuropharmacology 2012; 62:2154-68. [PMID: 22361232 DOI: 10.1016/j.neuropharm.2012.01.028] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 01/13/2012] [Accepted: 01/31/2012] [Indexed: 12/19/2022]
Abstract
Idiopathic Parkinson's disease (PD) represents a complex interaction between the inherent vulnerability of the nigrostriatal dopaminergic system, a possible genetic predisposition, and exposure to environmental toxins including inflammatory triggers. Evidence now suggests that chronic neuroinflammation is consistently associated with the pathophysiology of PD. Activation of microglia and increased levels of pro-inflammatory mediators such as TNF-α, IL-1β and IL-6, reactive oxygen species and eicosanoids has been reported after post-mortem analysis of the substantia nigra from PD patients and in animal models of PD. It is hypothesised that chronically activated microglia secrete high levels of pro-inflammatory mediators which damage neurons and further activate microglia, resulting in a feed forward cycle promoting further inflammation and neurodegeneration. Moreover, nigrostriatal dopaminergic neurons are more vulnerable to pro-inflammatory and oxidative mediators than other cell types because of their low intracellular glutathione concentration. Systemic inflammation has also been suggested to contribute to neurodegeneration in PD, as lymphocyte infiltration has been observed in brains of PD patients and in animal models of PD, substantiating the current theory of a fundamental role of inflammation in neurodegeneration. We will examine the current evidence in the literature which offers insight into the premise that both central and systemic inflammation may contribute to neurodegeneration in PD. We will discuss the emerging possibility of the use of diagnostic tools such as imaging technologies for PD patients. Finally, we will present the immunomodulatory therapeutic strategies that are now under investigation and in clinical trials as potential neuroprotective drugs for PD.
Collapse
Affiliation(s)
- Louise M Collins
- Department of Anatomy and Neuroscience, University College Cork, Biosciences Institute, Western Road, Cork, Ireland
| | | | | | | |
Collapse
|
36
|
Mattner F, Bandin DL, Staykova M, Berghofer P, Gregoire MC, Ballantyne P, Quinlivan M, Fordham S, Pham T, Willenborg DO, Katsifis A. Evaluation of [¹²³I]-CLINDE as a potent SPECT radiotracer to assess the degree of astroglia activation in cuprizone-induced neuroinflammation. Eur J Nucl Med Mol Imaging 2011; 38:1516-28. [PMID: 21484375 DOI: 10.1007/s00259-011-1784-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 03/01/2011] [Indexed: 10/18/2022]
Abstract
PURPOSE The purpose of this study was to assess the feasibility and sensitivity of the high-affinity translocator protein (TSPO) ligand [(123)I]-CLINDE in imaging TSPO changes in vivo and characterise and compare astroglial and TSPO changes in the cuprizone model of demyelination and remyelination in C57BL/6 mice. METHODS C57BL/6 mice were fed with cuprizone for 4 weeks to induce demyelination followed by 2-4 weeks of standard diet (remyelination). Groups of mice were followed by in vivo single photon emission computed tomography (SPECT)/CT imaging using [(123)I]-CLINDE and uptake correlated with biodistribution, autoradiography, immunohistochemistry, immunofluorescence and real-time polymerase chain reaction (RT-PCR). RESULTS The uptake of [(123)I]-CLINDE in the brain as measured by SPECT imaging over the course of treatment reflects the extent of the physiological response, with significant increases observed during demyelination followed by a decrease in uptake during remyelination. This was confirmed by autoradiography and biodistribution studies. A positive correlation between TSPO expression and astrogliosis was found and both activated astrocytes and microglial cells expressed TSPO. [(123)I]-CLINDE uptake reflects astrogliosis in brain structures such as corpus callosum, caudate putamen, medium septum and olfactory tubercle as confirmed by both in vitro and in vivo results. CONCLUSION The dynamics in the cuprizone-induced astroglial and TSPO changes, observed by SPECT imaging, were confirmed by immunofluorescence, RT-PCR and autoradiography. The highly specific TSPO radioiodinated ligand CLINDE can be used as an in vivo marker for early detection and monitoring of a variety of neuropathological conditions using noninvasive brain imaging techniques.
Collapse
Affiliation(s)
- Filomena Mattner
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Rd, Lucas Heights, Sydney, NSW 2234, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Evaluation of prion deposits and microglial activation in scrapie-infected mice using molecular imaging probes. Mol Imaging Biol 2011; 12:576-82. [PMID: 20376566 DOI: 10.1007/s11307-010-0321-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE A characteristic of prion diseases which affect both animals and humans is the aggregation of PrP amyloid fibrils in the brain, associated with a chronic inflammatory response dominated by microglial activation. In this study, we hypothesised that specific ligands of the 18-kDa translocator protein (TSPO) would be effective in the evaluation of microglial activation related to PrP(sc) deposits in prion disease. PROCEDURES Chronological studies using in vitro autoradiography were carried out with [(3)H]-PK11195 and [(125)I]-IMPY on frozen cerebral sections from scrapie-infected mice and controls. Accumulation of prion deposits was confirmed by histoblot staining with prion protein-specific monoclonal antibody. Ex vivo autoradiographic studies were carried out with [(125)I]-CLINDE and [(125)I]-IMPY at the terminal stage of infection. RESULTS Chronological studies using in vitro autoradiography showed that PrP(sc) deposits were co-localised with activated microglia as early as 60 days post-inoculation. Progressive levels of PrP(sc) and TSPO staining were successively observed in the hippocampus, cortex and left thalamus of infected mouse brain sections in the course of the disease and were correlated with the signals obtained by histoblot staining. Significant TSPO labelling was also observed ex vivo in the cortex, hippocampus and thalamus of scrapie-infected mice. In parallel, [(125)I]-IMPY showed labelling in the same cerebral regions but with high background staining. CONCLUSIONS These findings indicate the ability of [(125)I]-IMPY and [(125)I]-CLINDE to evaluate prion deposits and microglial activation in vitro and ex vivo in scrapie-infected mice at different stages of the disease.
Collapse
|
38
|
Arlicot N, Petit E, Katsifis A, Toutain J, Divoux D, Bodard S, Roussel S, Guilloteau D, Bernaudin M, Chalon S. Detection and quantification of remote microglial activation in rodent models of focal ischaemia using the TSPO radioligand CLINDE. Eur J Nucl Med Mol Imaging 2010; 37:2371-80. [PMID: 20814674 DOI: 10.1007/s00259-010-1598-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 08/09/2010] [Indexed: 01/09/2023]
Abstract
PURPOSE Neuroinflammation is involved in stroke pathophysiology and might be imaged using radioligands targeting the 18 kDa translocator protein (TSPO). METHODS We studied microglial reaction in brain areas remote from the primary lesion site in two rodent models of focal cerebral ischaemia (permanent or transient) using [125I]-CLINDE, a promising TSPO single photon emission computed tomography radioligand. RESULTS In a mouse model of permanent middle cerebral artery occlusion (MCAO), ex vivo autoradiographic studies demonstrated, besides in the ischaemic territory, accumulation of [125I]-CLINDE in the ipsilateral thalamus with a binding that progressed up to 3 weeks after MCAO. [125I]-CLINDE binding markedly decreased in animals pre-injected with either unlabelled CLINDE or PK11195, while no change was observed with flumazenil pre-treatment, demonstrating TSPO specificity. In rats subjected to transient MCAO, [125I]-CLINDE binding in the ipsilateral thalamus and substantia nigra pars reticulata (SNr) was significantly higher than that in contralateral tissue. Moreover, [125I]-CLINDE binding in the thalamus and SNr was quantitatively correlated to the ischaemic volume assessed by MRI in the cortex and striatum, respectively. CONCLUSION Clinical consequences of secondary neuronal degeneration in stroke might be better treated thanks to the discrimination of neuronal processes using in vivo molecular imaging and potent TSPO radioligands like CLINDE to guide therapeutic interventions.
Collapse
Affiliation(s)
- Nicolas Arlicot
- UMR Inserm U 930, CNRS ERL 3106, Université François Rabelais de Tours, CHRU de Tours, Tours, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Venneti S, Wiley CA, Kofler J. Imaging microglial activation during neuroinflammation and Alzheimer's disease. J Neuroimmune Pharmacol 2009; 4:227-43. [PMID: 19052878 PMCID: PMC2682630 DOI: 10.1007/s11481-008-9142-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 11/17/2008] [Indexed: 01/07/2023]
Abstract
Microglial activation is an important pathogenic component of neurodegenerative disease processes. This state of increased inflammation is associated not only with neurotoxic consequences but also neuroprotective effects, e.g., phagocytosis and clearance of amyloid in Alzheimer's disease. In addition, activation of microglia appears to be one of the major mechanisms of amyloid clearance following active or passive immunotherapy. Imaging techniques may provide a minimally invasive tool to elucidate the complexities and dynamics of microglial function and dysfunction in aging and neurodegenerative diseases. Imaging microglia in vivo in live subjects by confocal or two/multiphoton microscopy offers the advantage of studying these cells over time in their native environment. Imaging microglia in human subjects by positron emission tomography scanning with translocator protein-18 kDa ligands can offer a measure of the inflammatory process and a means of detecting progression of disease and efficacy of therapeutics over time.
Collapse
Affiliation(s)
- Sriram Venneti
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, 3400 Spruce St, 6.093 Founders Building, Philadelphia, PA 19104, USA e-mail:
| | - Clayton A. Wiley
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, A-506, Pittsburgh, PA 15213, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, A-506, Pittsburgh, PA 15213, USA
| |
Collapse
|