1
|
Chauveau F, Winkeler A, Chalon S, Boutin H, Becker G. PET imaging of neuroinflammation: any credible alternatives to TSPO yet? Mol Psychiatry 2025; 30:213-228. [PMID: 38997465 DOI: 10.1038/s41380-024-02656-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Over the last decades, the role of neuroinflammation in neuropsychiatric conditions has attracted an exponentially growing interest. A key driver for this trend was the ability to image brain inflammation in vivo using PET radioligands targeting the Translocator Protein 18 kDa (TSPO), which is known to be expressed in activated microglia and astrocytes upon inflammatory events as well as constitutively in endothelial cells. TSPO is a mitochondrial protein that is expressed mostly by microglial cells upon activation but is also expressed by astrocytes in some conditions and constitutively by endothelial cells. Therefore, our current understanding of neuroinflammation dynamics is hampered by the lack of alternative targets available for PET imaging. We performed a systematic search and review on radiotracers developed for neuroinflammation PET imaging apart from TSPO. The following targets of interest were identified through literature screening (including previous narrative reviews): P2Y12R, P2X7R, CSF1R, COX (microglial targets), MAO-B, I2BS (astrocytic targets), CB2R & S1PRs (not specific of a single cell type). We determined the level of development and provided a scoping review for each target. Strikingly, astrocytic biomarker MAO-B has progressed in clinical investigations the furthest, while few radiotracers (notably targeting S1P1Rs, CSF1R) are being implemented in clinical investigations. Other targets such as CB2R and P2X7R have proven disappointing in clinical studies (e.g. poor signal, lack of changes in disease conditions, etc.). While astrocytic targets are promising, development of new biomarkers and tracers specific for microglial activation has proven challenging.
Collapse
Affiliation(s)
- Fabien Chauveau
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
| | - Alexandra Winkeler
- Université Paris-Saclay, Inserm, CNRS, CEA, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France
| | - Sylvie Chalon
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France
| | - Hervé Boutin
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France.
| | - Guillaume Becker
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
- Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail, 14 rue Pierre et Marie Curie, 94701, Maisons-Alfort, Cedex, France
| |
Collapse
|
2
|
Qiu L, Wang J, Tewari M, Rensing DT, Egan TM, Perlmutter JS, Tu Z. Synthesis and in vitro evaluation of novel compounds and discovery of a promising iodine-125 radioligand for purinergic P2X7 receptor (P2X7R). Bioorg Med Chem 2024; 118:118054. [PMID: 39740572 DOI: 10.1016/j.bmc.2024.118054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
The purinergic P2X ligand-gated ion channel 7 receptor (P2X7R) plays a critical role in various inflammatory processes and other diseases. Fast determination of compounds P2X7R binding potency and discovery of a promise PET radiotracer for imaging P2X7R require a P2X7R suitable radioligand for radioactive competitive binding assay. Herein, we designed and synthesized thirteen new P2X7R ligands and determined the in vitro binding potency. The fluorescence screening assay identified the iodide compound 1c with high potency and specificity toward P2X7R with an IC50 of 0.25 ± 0.05 nM. Therefore, 1c was 125I-labeled to afford [125I]1c with a good radiochemical yield (44 ± 12 %, n = 3) and high radiochemical purity (>95 %). Radioligand saturation binding assay showed that [125I]1c specifically bound to human P2X7R with high affinity (Kd = 1.68 nM and Bmax = 94 fmol/mg). A radioactive high throughput binding assay using [125I]1c for our new compounds demonstrated that the imidazole compounds 1b, 1c, and 1d exhibited high inhibition for >70 %, while the analogues of GSK314181A exhibited low inhibition for <35 %. In addition, our radioligand competitive binding assays using [125I]1c demonstrated that 1b, 1c, and 1d have high potency with IC50 values of 7.91 ± 0.22, 7.06 ± 1.68, and 7.16 ± 0.41 nM toward P2X7R, respectively.Together, compounds 1b, 1c, and 1d are highly potent for P2X7R, and [125I]1c has great potential to be a radioligand for screening P2X7R binding potency of the new compounds and investigating the P2X7R expression in animal models of human disease.
Collapse
Affiliation(s)
- Lin Qiu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Jinzhi Wang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Manju Tewari
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, United States
| | - Derek T Rensing
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Terrance M Egan
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, United States
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States; Neurology, Neuroscience, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|
3
|
Lehto J, Aarnio R, Tuisku J, Sucksdorff M, Koivumäki EM, Nylund M, Helin S, Rajander J, Danon J, Gilchrist J, Kassiou M, Oikonen V, Airas L. P2X 7-receptor binding in new-onset and secondary progressive MS - a [ 11C]SMW139 PET study. EJNMMI Res 2024; 14:123. [PMID: 39636350 PMCID: PMC11621262 DOI: 10.1186/s13550-024-01186-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND PET imaging of activated microglia has improved our understanding of the pathology behind disability progression in MS, and pro-inflammatory microglia at 'smoldering' lesion rims have been implicated as drivers of disability progression. The P2X 7R is upregulated in the cellular membranes of activated microglia. A single-tissue dual-input model was applied to quantify P2X 7R binding in the normal appearing white matter, perilesional areas and thalamus among progressive MS patients, healthy controls and newly diagnosed relapsing MS patients. RESULTS Overall, tracer uptake in the MS brain was not significantly higher compared to HCs. In the 3 mm perilesional rim of all T1 lesions, tracer binding was higher among relapsing patients compared to progressive patients. Tracer binding was higher in males compared to females. Disease duration correlated with tracer binding in the normal appearing white matter. Age correlated negatively with tracer binding in the perilesional rims. CONCLUSIONS Even as binding estimates obtained with the dual-input model were consistent with the expected distribution of P2X 7Rs in the MS brain, the small free fraction of the parent tracer may limit its accuracy and applicability, and binding estimates between subjects were highly variable. Conclusive evidence for the applicability of [11C]SMW139 to detect MS-related diffuse smoldering inflammation was not obtained.
Collapse
Affiliation(s)
- Jussi Lehto
- Turku PET Centre, Turku, Finland.
- Neurocenter, Turku University Hospital, Turku, Finland.
- InFLAMES Research Flagship, University of Turku, Turku, Finland.
| | | | | | - Marcus Sucksdorff
- Turku PET Centre, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | | | - Marjo Nylund
- Turku PET Centre, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | | | - Johan Rajander
- Turku PET Centre, Turku, Finland
- Åbo Akademi University, Turku, Finland
| | - Jonathan Danon
- School of Chemistry, The University of Sydney, Sydney, Australia
| | - Jayson Gilchrist
- School of Chemistry, The University of Sydney, Sydney, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, Australia
| | | | - Laura Airas
- Turku PET Centre, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| |
Collapse
|
4
|
Torres-Rico M, García-Calvo V, Gironda-Martínez A, Pascual-Guerra J, García AG, Maneu V. Targeting calciumopathy for neuroprotection: focus on calcium channels Cav1, Orai1 and P2X7. Cell Calcium 2024; 123:102928. [PMID: 39003871 DOI: 10.1016/j.ceca.2024.102928] [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: 05/30/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
As the uncontrolled entry of calcium ions (Ca2+) through plasmalemmal calcium channels is a cell death trigger, the conjecture is here raised that mitigating such an excess of Ca2+ entry should rescue from death the vulnerable neurons in neurodegenerative diseases (NDDs). However, this supposition has failed in some clinical trials (CTs). Thus, a recent CT tested whether isradipine, a blocker of the Cav1 subtype of voltage-operated calcium channels (VOCCs), exerted a benefit in patients with Parkinson's disease (PD); however, outcomes were negative. This is one more of the hundreds of CTs done under the principle of one-drug-one-target, that have failed in Alzheimer's disease (AD) and other NDDs during the last three decades. As there are myriad calcium channels to let Ca2+ ions gain the cell cytosol, it seems reasonable to predict that blockade of Ca2+ entry through a single channel may not be capable of preventing the Ca2+ flood of cells by the uncontrolled Ca2+ entry. Furthermore, as Ca2+ signaling is involved in the regulation of myriad functions in different cell types, it seems also reasonable to guess that a therapy should be more efficient by targeting different cells with various drugs. Here, we propose to mitigate Ca2+ entry by the simultaneous partial blockade of three quite different subtypes of plasmalemmal calcium channels that is, the Cav1 subtype of VOCCs, the Orai1 store-operated calcium channel (SOCC), and the purinergic P2X7 calcium channel. All three channels are expressed in both microglia and neurons. Thus, by targeting the three channels with a combination of three drug blockers we expect favorable changes in some of the pathogenic features of NDDs, namely (i) to mitigate Ca2+ entry into microglia; (ii) to decrease the Ca2+-dependent microglia activation; (iii) to decrease the sustained neuroinflammation; (iv) to decrease the uncontrolled Ca2+ entry into neurons; (v) to rescue vulnerable neurons from death; and (vi) to delay disease progression. In this review we discuss the arguments underlying our triad hypothesis in the sense that the combination of three repositioned medicines targeting Cav1, Orai1, and P2X7 calcium channels could boost neuroprotection and delay the progression of AD and other NDDs.
Collapse
Affiliation(s)
| | | | - Adrián Gironda-Martínez
- Instituto Fundación Teófilo Hernando, Madrid, Spain; Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Antonio G García
- Instituto Fundación Teófilo Hernando, Madrid, Spain; Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain; Facultad de Medicina, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Victoria Maneu
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, Alicante, Spain.
| |
Collapse
|
5
|
Liu X, Li Y, Huang L, Kuang Y, Wu X, Ma X, Zhao B, Lan J. Unlocking the therapeutic potential of P2X7 receptor: a comprehensive review of its role in neurodegenerative disorders. Front Pharmacol 2024; 15:1450704. [PMID: 39139642 PMCID: PMC11319138 DOI: 10.3389/fphar.2024.1450704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The P2X7 receptor (P2X7R), an ATP-gated ion channel, has emerged as a crucial player in neuroinflammation and a promising therapeutic target for neurodegenerative disorders. This review explores the current understanding of P2X7R's structure, activation, and physiological roles, focusing on its expression and function in microglial cells. The article examines the receptor's involvement in calcium signaling, microglial activation, and polarization, as well as its role in the pathogenesis of Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis. The review highlights the complex nature of P2X7R signaling, discussing its potential neuroprotective and neurotoxic effects depending on the disease stage and context. It also addresses the development of P2X7R antagonists and their progress in clinical trials, identifying key research gaps and future perspectives for P2X7R-targeted therapy development. By providing a comprehensive overview of the current state of knowledge and future directions, this review serves as a valuable resource for researchers and clinicians interested in exploring the therapeutic potential of targeting P2X7R for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Xiaoming Liu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yiwen Li
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Liting Huang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Yingyan Kuang
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiaoxiong Wu
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiangqiong Ma
- Henan Hospital of Integrated Chinese and Western Medicine, Zhengzhou, China
| | - Beibei Zhao
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Jiao Lan
- Shenzhen Baoan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| |
Collapse
|
6
|
Kong Y, Cao L, Wang J, Zhuang J, Xie F, Zuo C, Huang Q, Shi K, Rominger A, Li M, Wu P, Guan Y, Ni R. In vivo reactive astrocyte imaging using [ 18F]SMBT-1 in tauopathy and familial Alzheimer's disease mouse models: A multi-tracer study. J Neurol Sci 2024; 462:123079. [PMID: 38878650 DOI: 10.1016/j.jns.2024.123079] [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: 01/24/2024] [Revised: 05/13/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Reactive astrocytes play an important role in the development of Alzheimer's disease and primary tauopathies. Here, we aimed to investigate the relationships between reactive astrocytes. Microgliosis and glucose metabolism with Tau and amyloid beta pathology by using multi-tracer imaging in widely used tauopathy and familial Alzheimer's disease mouse models. RESULTS Positron emission tomography imaging using [18F]PM-PBB3 (tau), [18F]florbetapir (amyloid-beta), [18F]SMBT-1 (monoamine oxidase-B), [18F]DPA-714 (translocator protein) and [18F]fluorodeoxyglucose was carried out in 3- and 7-month-old rTg4510 tau mice, 5 × FAD familial Alzheimer's disease mice and wild-type mice. Immunofluorescence staining was performed to validate the pathological distribution in the mouse brain after in vivo imaging. We found increased regional levels of [18F]PM-PBB3, [18F]SMBT-1, and [18F]DPA-714 and hypoglucose metabolism in the brains of 7-month-old rTg4510 mice compared to age-matched wild-type mice. Increased [18F]SMBT-1 uptake was observed in the brains of 3, 7-month-old 5 × FAD mice, with elevated regional [18F]florbetapir and [18F]DPA-714 uptakes in the brains of 7-month-old 5 × FAD mice, compared to age-matched wild-type mice. Positive correlations were shown between [18F]SMBT-1 and [18F]PM-PBB3, [18F]DPA-714 and [18F]PM-PBB3 in rTg4510 mice, and between [18F]florbetapir and [18F]DPA-714 SUVRs in 5 × FAD mice. CONCLUSION In summary, these findings provide in vivo evidence that reactive astrocytes, microglial activation, and cerebral hypoglucose metabolism are associated with tau and amyloid pathology development in animal models of tauopathy and familial Alzheimer's disease.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Cao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China; Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Jiao Wang
- Lab of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Junyi Zhuang
- Lab of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Kuangyu Shi
- Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Axel Rominger
- Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Ming Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ruiqing Ni
- Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland; Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland; Inst. Biomedical Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
7
|
Kong Y, Cao L, Wang J, Zhuang J, Liu Y, Bi L, Qiu Y, Hou Y, Huang Q, Xie F, Yang Y, Shi K, Rominger A, Guan Y, Jin H, Ni R. Increased Cerebral Level of P2X7R in a Tauopathy Mouse Model by PET Using [ 18F]GSK1482160. ACS Chem Neurosci 2024; 15:2112-2120. [PMID: 38776461 PMCID: PMC11157487 DOI: 10.1021/acschemneuro.4c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Neuroinflammation plays an important role in Alzheimer's disease and primary tauopathies. The aim of the current study was to map [18F]GSK1482160 for imaging of purinergic P2X7R in Alzheimer's disease and primary tauopathy mouse models. Small animal PET was performed using [18F]GSK1482160 in widely used mouse models of Alzheimer's disease (APP/PS1, 5×FAD, and 3×Tg), 4-repeat tauopathy (rTg4510) mice, and age-matched wild-type mice. Increased uptake of [18F]GSK1482160 was observed in the brains of 7-month-old rTg4510 mice compared to wild-type mice and compared to 3-month-old rTg4510 mice. A positive correlation between hippocampal tau [18F]APN-1607 and [18F]GSK1482160 uptake was found in rTg4510 mice. No significant differences in the uptake of [18F]GSK1482160 was observed for APP/PS1 mice, 5×FAD mice, or 3×Tg mice. Immunofluorescence staining further indicated the distribution of P2X7Rs in the brains of 7-month-old rTg4510 mice with accumulation of tau inclusion. These findings provide in vivo imaging evidence for an increased level of P2X7R in the brains of tauopathy mice.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Lei Cao
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
- Institute
for Regenerative Medicine, University of
Zurich, Zurich 8952, Switzerland
| | - Jiao Wang
- Lab
of Molecular
Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Junyi Zhuang
- Lab
of Molecular
Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yongshan Liu
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Lei Bi
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Yifan Qiu
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Yuyi Hou
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Qi Huang
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Fang Xie
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Yunhao Yang
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Kuangyu Shi
- Department
of Nuclear Medicine, University Hospital,
Inselspital Bern, Bern 3010, Switzerland
| | - Axel Rominger
- Department
of Nuclear Medicine, University Hospital,
Inselspital Bern, Bern 3010, Switzerland
| | - Yihui Guan
- PET Center,
Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Hongjun Jin
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Ruiqing Ni
- Institute
for Regenerative Medicine, University of
Zurich, Zurich 8952, Switzerland
- Department
of Nuclear Medicine, University Hospital,
Inselspital Bern, Bern 3010, Switzerland
- Institute
for Biomedical Engineering, University of
Zurich & ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
8
|
Liu J, Liu TT, Mou L, Zhang Y, Chen X, Wang Q, Deng BL, Liu J. P2X7 receptor: a potential target for treating comorbid anxiety and depression. Purinergic Signal 2024:10.1007/s11302-024-10007-0. [PMID: 38642324 DOI: 10.1007/s11302-024-10007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024] Open
Abstract
In clinical practice, depression and anxiety frequently coexist, and they are both comorbid with somatic diseases. The P2X7R is an adenosine 5'-triphosphate (ATP)-gated non-selective cation channel that is widely expressed in immune-related cells. Under conditions of stress, chronic pain, and comorbid chronic physical illness, P2X7R activation in glial cells leads to neuroinflammation. This could contribute to the development of anxiety and depression-related emotional disturbances. Previous studies have shown that the P2X7 receptor (P2X7R) plays an important role in the pathogenesis of both anxiety and depression. Thus, the P2X7R may play a role in the comorbidity of anxiety and depression. Positron emission tomography can be used to assess the degree and location of neuroinflammation by monitoring functional and expression-related changes in P2X7R, which can facilitate clinical diagnoses and guide the treatment of patients with anxiety and depression. Moreover, single nucleotide polymorphisms (SNPs) in the P2X7R gene are associated with susceptibility to different types of psychiatric disorders. Thus, evaluating the SNPs of the P2X7R gene could enable personalized mood disorder diagnoses and treatments. If the P2X7R were set as a therapeutic target, selective P2X7R antagonists may modulate P2X7R function, thereby altering the balance of intra- and extra-cellular ATP. This could have therapeutic implications for treating anxiety and depression, as well as for pain management. According to in vitro and in vivo studies, the P2X7R plays an important role in anxiety and depression. In this review, we consider the potential of the P2X7R as a therapeutic target for comorbid anxiety and depression, and discuss the potential diagnostic and therapeutic value of this receptor.
Collapse
Affiliation(s)
- Jun Liu
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
- Department of Geriatric Neurology, Qinglongchang Ward, Chengdu Sixth People's Hospital, Chengdu, China
| | - Ting-Ting Liu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Mou
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yuwen Zhang
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Xiang Chen
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Qi Wang
- Department of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Bin-Lu Deng
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Liu
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China.
- Department of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China.
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
9
|
Zheng H, Liu Q, Zhou S, Luo H, Zhang W. Role and therapeutic targets of P2X7 receptors in neurodegenerative diseases. Front Immunol 2024; 15:1345625. [PMID: 38370420 PMCID: PMC10869479 DOI: 10.3389/fimmu.2024.1345625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
The P2X7 receptor (P2X7R), a non-selective cation channel modulated by adenosine triphosphate (ATP), localizes to microglia, astrocytes, oligodendrocytes, and neurons in the central nervous system, with the most incredible abundance in microglia. P2X7R partake in various signaling pathways, engaging in the immune response, the release of neurotransmitters, oxidative stress, cell division, and programmed cell death. When neurodegenerative diseases result in neuronal apoptosis and necrosis, ATP activates the P2X7R. This activation induces the release of biologically active molecules such as pro-inflammatory cytokines, chemokines, proteases, reactive oxygen species, and excitotoxic glutamate/ATP. Subsequently, this leads to neuroinflammation, which exacerbates neuronal involvement. The P2X7R is essential in the development of neurodegenerative diseases. This implies that it has potential as a drug target and could be treated using P2X7R antagonists that are able to cross the blood-brain barrier. This review will comprehensively and objectively discuss recent research breakthroughs on P2X7R genes, their structural features, functional properties, signaling pathways, and their roles in neurodegenerative diseases and possible therapies.
Collapse
Affiliation(s)
- Huiyong Zheng
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiang Liu
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Siwei Zhou
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongliang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wenjun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Zhang J, Gao L, Zhang Y, Wang H, Sun S, Wu L. Involvement of microglial P2X7 receptor in pain modulation. CNS Neurosci Ther 2024; 30:e14496. [PMID: 37950524 PMCID: PMC10805404 DOI: 10.1111/cns.14496] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/23/2023] [Accepted: 10/02/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Pain is a rapid response mechanism that compels organisms to retreat from the harmful stimuli and triggers a repair response. Nonetheless, when pain persists for extended periods, it can lead to adverse changes into in the individual's brain, negatively impacting their emotional state and overall quality of life. Microglia, the resident immune cells in the central nervous system (CNS), play a pivotal role in regulating a variety of pain-related disorders. Specifically, recent studies have shed light on the central role that microglial purinergic ligand-gated ion channel 7 receptor (P2X7R) plays in regulating pain. In this respect, the P2X7R on microglial membranes represents a potential therapeutic target. AIMS To expound on the intricate link between microglial P2X7R and pain, offering insights into potential avenues for future research. METHODS We reviewed 140 literature and summarized the important role of microglial P2X7R in regulating pain, including the structure and function of P2X7R, the relationship between P2X7R and microglial polarization, P2X7R-related signaling pathways, and the effects of P2X7R antagonists on pain regulation. RESULTS P2X7R activation is related to M1 polarization of microglia, while suppressing P2X7R can transfer microglia from M1 into M2 phenotype. And targeting the P2X7R-mediated signaling pathways helps to explore new therapy for pain alleviation. P2X7R antagonists also hold potential for translational and clinical applications in pain management. CONCLUSIONS Microglial P2X7R holds promise as a potential novel pharmacological target for clinical treatments due to its distinctive structure, function, and the development of antagonists.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Lei Gao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Yaoyuan Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Haozhen Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Shukai Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Li‐an Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of StomatologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
11
|
Ottoy J, De Picker L, Kang MS. Microglial Positron Emission Tomography Imaging In Vivo : Positron Emission Tomography Radioligands: Utility in Research and Clinical Practice. ADVANCES IN NEUROBIOLOGY 2024; 37:579-589. [PMID: 39207714 DOI: 10.1007/978-3-031-55529-9_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS) play a key role in regulating and maintaining homeostasis in the brain. However, the CNS is also vulnerable to infections and inflammatory processes. In response to CNS perturbations, microglia become reactive, notably with expression of the translocator protein (TSPO), primarily on their outer mitochondrial membrane. Despite TSPO being commonly used as a marker for microglia, it is also present in other cell types such as astrocytes. Positron emission tomography (PET) ligands that target the TSPO enable the noninvasive detection and quantification of glial reactivity. While some limitations were raised, TSPO PET remains an attractive biomarker of CNS infection and inflammation. This book chapter delves into the development and application of microglial PET imaging with a focus on the TSPO PET. First, we provide an overview of the evolution of TSPO PET radioligands from first-generation to second-generation ligands and their applications in studying neuroinflammation (or CNS inflammation). Subsequently, we discuss the limitations and challenges associated with TSPO PET. Then we go on to explore non-TSPO targets for microglial PET imaging. Finally, we conclude with future directions for research and clinical practice in this field.
Collapse
Affiliation(s)
- Julie Ottoy
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium
- University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Min Su Kang
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
12
|
Kalita M, Park JH, Kuo RC, Hayee S, Marsango S, Straniero V, Alam IS, Rivera-Rodriguez A, Pandrala M, Carlson ML, Reyes ST, Jackson IM, Suigo L, Luo A, Nagy SC, Valoti E, Milligan G, Habte F, Shen B, James ML. PET Imaging of Innate Immune Activation Using 11C Radiotracers Targeting GPR84. JACS AU 2023; 3:3297-3310. [PMID: 38155640 PMCID: PMC10751761 DOI: 10.1021/jacsau.3c00435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 12/30/2023]
Abstract
Chronic innate immune activation is a key hallmark of many neurological diseases and is known to result in the upregulation of GPR84 in myeloid cells (macrophages, microglia, and monocytes). As such, GPR84 can potentially serve as a sensor of proinflammatory innate immune responses. To assess the utility of GPR84 as an imaging biomarker, we synthesized 11C-MGX-10S and 11C-MGX-11Svia carbon-11 alkylation for use as positron emission tomography (PET) tracers targeting this receptor. In vitro experiments demonstrated significantly higher binding of both radiotracers to hGPR84-HEK293 cells than that of parental control HEK293 cells. Co-incubation with the GPR84 antagonist GLPG1205 reduced the binding of both radiotracers by >90%, demonstrating their high specificity for GPR84 in vitro. In vivo assessment of each radiotracer via PET imaging of healthy mice illustrated the superior brain uptake and pharmacokinetics of 11C-MGX-10S compared to 11C-MGX-11S. Subsequent use of 11C-MGX-10S to image a well-established mouse model of systemic and neuro-inflammation revealed a high PET signal in affected tissues, including the brain, liver, lung, and spleen. In vivo specificity of 11C-MGX-10S for GPR84 was confirmed by the administration of GLPG1205 followed by radiotracer injection. When compared with 11C-DPA-713-an existing radiotracer used to image innate immune activation in clinical research studies-11C-MGX-10S has multiple advantages, including its higher binding signal in inflamed tissues in the CNS and periphery and low background signal in healthy saline-treated subjects. The pronounced uptake of 11C-MGX-10S during inflammation, its high specificity for GPR84, and suitable pharmacokinetics strongly support further investigation of 11C-MGX-10S for imaging GPR84-positive myeloid cells associated with innate immune activation in animal models of inflammatory diseases and human neuropathology.
Collapse
Affiliation(s)
- Mausam Kalita
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Jun Hyung Park
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Renesmee Chenting Kuo
- Department
of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Samira Hayee
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Sara Marsango
- Centre
for Translational Pharmacology, School of Molecular Biosciences, College
of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland,
U.K.
| | - Valentina Straniero
- Department
of Pharmaceutical Sciences, University of
Milan, via Luigi Mangiagalli
25, 20133 Milano, Italy
| | - Israt S. Alam
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | | | - Mallesh Pandrala
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Mackenzie L. Carlson
- Department
of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, United States
| | - Samantha T. Reyes
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Isaac M. Jackson
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Lorenzo Suigo
- Department
of Pharmaceutical Sciences, University of
Milan, via Luigi Mangiagalli
25, 20133 Milano, Italy
| | - Audrey Luo
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Sydney C. Nagy
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Ermanno Valoti
- Department
of Pharmaceutical Sciences, University of
Milan, via Luigi Mangiagalli
25, 20133 Milano, Italy
| | - Graeme Milligan
- Centre
for Translational Pharmacology, School of Molecular Biosciences, College
of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland,
U.K.
| | - Frezghi Habte
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Bin Shen
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
| | - Michelle L. James
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
- Department
of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
13
|
Yin Y, Wei L, Caseley EA, Lopez‐Charcas O, Wei Y, Li D, Muench SP, Roger S, Wang L, Jiang L. Leveraging the ATP-P2X7 receptor signalling axis to alleviate traumatic CNS damage and related complications. Med Res Rev 2023; 43:1346-1373. [PMID: 36924449 PMCID: PMC10947395 DOI: 10.1002/med.21952] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 11/11/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
The P2X7 receptor is an exceptional member of the P2X purinergic receptor family, with its activation requiring high concentrations of extracellular adenosine 5'-triphosphate (ATP) that are often associated with tissue damage and inflammation. In the central nervous system (CNS), it is highly expressed in glial cells, particularly in microglia. In this review, we discuss the role and mechanisms of the P2X7 receptor in mediating neuroinflammation and other pathogenic events in a variety of traumatic CNS damage conditions, which lead to loss of neurological and cognitive functions. We raise the perspective on the steady progress in developing CNS-penetrant P2X7 receptor-specific antagonists that leverage the ATP-P2X7 receptor signaling axis as a potential therapeutic strategy to alleviate traumatic CNS damage and related complications.
Collapse
Affiliation(s)
- Yaling Yin
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Linyu Wei
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Emily A. Caseley
- Faculty of Biological Sciences, School of Biomedical SciencesUniversity of LeedsLeedsUK
| | - Osbaldo Lopez‐Charcas
- EA4245, Transplantation, Immunology and Inflammation, Faculty of MedicineUniversity of ToursToursFrance
| | - Yingjuan Wei
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Dongliang Li
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
- Sanquan College of Xinxiang Medical UniversityXinxiangChina
| | - Steve P. Muench
- Faculty of Biological Sciences, School of Biomedical SciencesUniversity of LeedsLeedsUK
| | - Sebastian Roger
- EA4245, Transplantation, Immunology and Inflammation, Faculty of MedicineUniversity of ToursToursFrance
| | - Lu Wang
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
| | - Lin‐Hua Jiang
- Sino‐UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangChina
- Faculty of Biological Sciences, School of Biomedical SciencesUniversity of LeedsLeedsUK
| |
Collapse
|
14
|
Neumann KD, Broshek DK, Newman BT, Druzgal TJ, Kundu BK, Resch JE. Concussion: Beyond the Cascade. Cells 2023; 12:2128. [PMID: 37681861 PMCID: PMC10487087 DOI: 10.3390/cells12172128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Sport concussion affects millions of athletes each year at all levels of sport. Increasing evidence demonstrates clinical and physiological recovery are becoming more divergent definitions, as evidenced by several studies examining blood-based biomarkers of inflammation and imaging studies of the central nervous system (CNS). Recent studies have shown elevated microglial activation in the CNS in active and retired American football players, as well as in active collegiate athletes who were diagnosed with a concussion and returned to sport. These data are supportive of discordance in clinical symptomology and the inflammatory response in the CNS upon symptom resolution. In this review, we will summarize recent advances in the understanding of the inflammatory response associated with sport concussion and broader mild traumatic brain injury, as well as provide an outlook for important research questions to better align clinical and physiological recovery.
Collapse
Affiliation(s)
- Kiel D. Neumann
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Donna K. Broshek
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, Charlottesville, VA 22903, USA;
| | - Benjamin T. Newman
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - T. Jason Druzgal
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - Bijoy K. Kundu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (B.T.N.); (T.J.D.); (B.K.K.)
| | - Jacob E. Resch
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
15
|
Miao J, Ma H, Yang Y, Liao Y, Lin C, Zheng J, Yu M, Lan J. Microglia in Alzheimer's disease: pathogenesis, mechanisms, and therapeutic potentials. Front Aging Neurosci 2023; 15:1201982. [PMID: 37396657 PMCID: PMC10309009 DOI: 10.3389/fnagi.2023.1201982] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by protein aggregation in the brain. Recent studies have revealed the critical role of microglia in AD pathogenesis. This review provides a comprehensive summary of the current understanding of microglial involvement in AD, focusing on genetic determinants, phenotypic state, phagocytic capacity, neuroinflammatory response, and impact on synaptic plasticity and neuronal regulation. Furthermore, recent developments in drug discovery targeting microglia in AD are reviewed, highlighting potential avenues for therapeutic intervention. This review emphasizes the essential role of microglia in AD and provides insights into potential treatments.
Collapse
Affiliation(s)
- Jifei Miao
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Haixia Ma
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yang Yang
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yuanpin Liao
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Cui Lin
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Juanxia Zheng
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Muli Yu
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Jiao Lan
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
16
|
Sluyter R, Adriouch S, Fuller SJ, Nicke A, Sophocleous RA, Watson D. Animal Models for the Investigation of P2X7 Receptors. Int J Mol Sci 2023; 24:ijms24098225. [PMID: 37175933 PMCID: PMC10179175 DOI: 10.3390/ijms24098225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The P2X7 receptor is a trimeric ligand-gated cation channel activated by extracellular adenosine 5'-triphosphate. The study of animals has greatly advanced the investigation of P2X7 and helped to establish the numerous physiological and pathophysiological roles of this receptor in human health and disease. Following a short overview of the P2X7 distribution, roles and functional properties, this article discusses how animal models have contributed to the generation of P2X7-specific antibodies and nanobodies (including biologics), recombinant receptors and radioligands to study P2X7 as well as to the pharmacokinetic testing of P2X7 antagonists. This article then outlines how mouse and rat models have been used to study P2X7. These sections include discussions on preclinical disease models, polymorphic P2X7 variants, P2X7 knockout mice (including bone marrow chimeras and conditional knockouts), P2X7 reporter mice, humanized P2X7 mice and P2X7 knockout rats. Finally, this article reviews the limited number of studies involving guinea pigs, rabbits, monkeys (rhesus macaques), dogs, cats, zebrafish, and other fish species (seabream, ayu sweetfish, rainbow trout and Japanese flounder) to study P2X7.
Collapse
Affiliation(s)
- Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Sahil Adriouch
- UniRouen, INSERM, U1234, Pathophysiology, Autoimmunity, and Immunotherapy, (PANTHER), Univ Rouen Normandie, University of Rouen, F-76000 Rouen, France
| | - Stephen J Fuller
- Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Nepean Hospital, Kingswood, NSW 2750, Australia
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, 80336 Munich, Germany
| | - Reece A Sophocleous
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
17
|
Singh P, Singh D, Srivastava P, Mishra G, Tiwari AK. Evaluation of advanced, pathophysiologic new targets for imaging of CNS. Drug Dev Res 2023; 84:484-513. [PMID: 36779375 DOI: 10.1002/ddr.22040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/12/2022] [Accepted: 12/31/2022] [Indexed: 02/14/2023]
Abstract
The inadequate information about the in vivo pathological, physiological, and neurological impairments, as well as the absence of in vivo tools for assessing brain penetrance and the efficiency of newly designed drugs, has hampered the development of new techniques for the treatment for variety of new central nervous system (CNS) diseases. The searching sites such as Science Direct and PubMed were used to find out the numerous distinct tracers across 16 CNS targets including tau, synaptic vesicle glycoprotein, the adenosine 2A receptor, the phosphodiesterase enzyme PDE10A, and the purinoceptor, among others. Among the most encouraging are [18 F]FIMX for mGluR imaging, [11 C]Martinostat for Histone deacetylase, [18 F]MNI-444 for adenosine 2A imaging, [11 C]ER176 for translocator protein, and [18 F]MK-6240 for tau imaging. We also reviewed the findings for each tracer's features and potential for application in CNS pathophysiology and therapeutic evaluation investigations, including target specificity, binding efficacy, and pharmacokinetic factors. This review aims to present a current evaluation of modern positron emission tomography tracers for CNS targets, with a focus on recent advances for targets that have newly emerged for imaging in humans.
Collapse
Affiliation(s)
- Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Deepika Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| | - Pooja Srivastava
- Division of Cyclotron and Radiopharmaceuticals Sciences, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Gauri Mishra
- Department of Zoology, Swami Shraddhananad College, University of Delhi, Alipur, Delhi, India
| | - Anjani K Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
18
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
Spotlight on P2X7 Receptor PET Imaging: A Bright Target or a Failing Star? Int J Mol Sci 2023; 24:ijms24021374. [PMID: 36674884 PMCID: PMC9861945 DOI: 10.3390/ijms24021374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/23/2022] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
The homotrimeric P2X7 receptor (P2X7R) is expressed by virtually all cells of the innate and adaptive immune system and plays a crucial role in various pathophysiological processes such as autoimmune and neurodegenerative diseases, inflammation, neuropathic pain and cancer. Consequently, the P2X7R is considered a promising target for therapy and diagnosis. As the development of tracers comes hand-in-hand with the development of potent and selective receptor ligands, there is a rising number of PET tracers available in preclinical and clinical studies. This review analyzes the development of P2X7R positron emission tomography (PET) tracers and their potential in various PET imaging applications.
Collapse
|
20
|
Mikkelsen JD, Aripaka SS, Kaad S, Pazarlar BA, Pinborg L, Finsen B, Varrone A, Bang-Andersen B, Bastlund JF. Characterization of the Novel P2X7 Receptor Radioligand [ 3H]JNJ-64413739 in Human Brain Tissue. ACS Chem Neurosci 2022; 14:111-118. [PMID: 36535632 PMCID: PMC9817075 DOI: 10.1021/acschemneuro.2c00561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Radioligands targeting microglia cells have been developed to identify and determine neuroinflammation in the living brain. One recently discovered ligand is JNJ-64413739 that binds selectively to the purinergic receptor P2X7R. The expression of P2X7R is increased under inflammation; hence, the ligand is considered useful in the detection of neuroinflammation in the brain. [18F]JNJ-64413739 has been evaluated in healthy subjects with positron emission tomography; however, the in vitro binding properties of the ligand in human brain tissue have not been investigated. Therefore, the purpose of this study was to measure Bmax and Kd of [3H]JNJ-64413739 using autoradiography on human cortical tissue sections resected from a total of 48 patients with treatment-resistant epilepsy. Correlations between the specific binding of [3H]JNJ-64413739 with age, sex, and duration of disease were explored. Finally, to examine the relationship between P2X7R and TSPO availability, specific binding of [3H]JNJ-64413739 and [123I]CLINDE was examined in the same tissue. The binding was measured in both cortical gray and subcortical white matter. Saturation revealed a Kd (5 nM) value similar between gray and white matter but a larger Bmax in the white than in the gray matter. The binding was completely displaced by the cold ligand and structurally different P2X7R ligands. The variability in saturable binding among the samples was found to be 38% in gray and white matter but was not correlated to either age, sex, or the duration of the disease. Interestingly, there was no significant correlation between [3H]JNJ-64413739 and [123I]CLINDE binding. These data demonstrate that [3H]JNJ-64413739 is a suitable radioligand for evaluating the distribution and expression of the P2X7R in the human brain.
Collapse
Affiliation(s)
- Jens D. Mikkelsen
- Neurobiology
Research Unit, University Hospital Rigshospitalet, Copenhagen 2100, Denmark,Institute
of Neuroscience, University of Copenhagen, Copenhagen 2200, Denmark,Department
of Molecular Medicine, University of Southern
Denmark, Odense 5000, Denmark,. Tel.: +45 3545 6701
| | - Sanjay S. Aripaka
- Neurobiology
Research Unit, University Hospital Rigshospitalet, Copenhagen 2100, Denmark
| | - Sif Kaad
- Neurobiology
Research Unit, University Hospital Rigshospitalet, Copenhagen 2100, Denmark
| | - Burcu A. Pazarlar
- Neurobiology
Research Unit, University Hospital Rigshospitalet, Copenhagen 2100, Denmark,Physiology
Department, Faculty of Medicine, Izmir Katip
Celebi University, Izmir 35330, Turkey
| | - Lars Pinborg
- Neurobiology
Research Unit, University Hospital Rigshospitalet, Copenhagen 2100, Denmark,Epilepsy
Clinic, Department of Neurology, Copenhagen
University Hospital, Rigshospitalet, Copenhagen 2100, Denmark
| | - Bente Finsen
- Department
of Molecular Medicine, University of Southern
Denmark, Odense 5000, Denmark
| | | | | | | |
Collapse
|
21
|
Huang G, Qiu Y, Bi L, Wei H, Li G, Li Z, Ye P, Yang M, Shen Y, Liu H, Wang L, Jin H. PET Imaging of P2X7 Receptor (P2X7R) for Neuroinflammation with Improved Radiosynthesis of Tracer [18F]4A in Mice and Non-human Primates. ACS Chem Neurosci 2022; 13:3464-3476. [PMID: 36441909 DOI: 10.1021/acschemneuro.2c00506] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The P2X7 receptor (P2X7R) is a key neuroinflammation target in a variety of neurodegenerative diseases. Improved radiosynthesis was developed according to the previously reported P2X7R antagonist GSK1482160. Biodistribution, radiometabolite, and dynamic positron emission tomography/computed tomography-magnetic resonance imaging (PET/CT-MRI) of the lipopolysaccharide (LPS) rat model and the transgenic mouse model of Alzheimer's disease (AD) revealed a stable, low uptake of [18F]4A in the brain of healthy rats but a higher standardized uptake value ratio (SUVR) in LPS-treated rats (1.316 ± 0.062, n = 3) than in sham (1.093 ± 0.029, n = 3). There were higher area under curves (AUCs) in the neocortex (25.12 ± 1.11 vs 18.94 ± 1.47), hippocampus (22.50 ± 3.41 vs 15.90 ± 1.59), and basal ganglia (22.26 ± 0.81 vs 15.32 ± 1.76) of AD mice (n = 3) than the controls (n = 3) (p < 0.05). Furthermore, 50 min dynamic PET in healthy nonhuman primates (NHPs) indicated [18F]4A could penetrate the blood-brain barrier (BBB). In conclusion, [18F]4A from this study is a potent P2X7R PET tracer that warrants further neuroinflammation quantification in human studies.
Collapse
Affiliation(s)
- Guolong Huang
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Yifan Qiu
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Lei Bi
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou510630, China
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou510630, China
| | - Zhijun Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Peizhen Ye
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Min Yang
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Yanfang Shen
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Hao Liu
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou510630, China
| | - Hongjun Jin
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai519000, Guangdong, China
| |
Collapse
|
22
|
Boyle AJ, Murrell E, Tong J, Schifani C, Narvaez A, Wuest M, West F, Wuest F, Vasdev N. PET Imaging of Fructose Metabolism in a Rodent Model of Neuroinflammation with 6-[ 18F]fluoro-6-deoxy-D-fructose. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238529. [PMID: 36500626 PMCID: PMC9736258 DOI: 10.3390/molecules27238529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Fluorine-18 labeled 6-fluoro-6-deoxy-D-fructose (6-[18F]FDF) targets the fructose-preferred facilitative hexose transporter GLUT5, which is expressed predominantly in brain microglia and activated in response to inflammatory stimuli. We hypothesize that 6-[18F]FDF will specifically image microglia following neuroinflammatory insult. 6-[18F]FDF and, for comparison, [18F]FDG were evaluated in unilateral intra-striatal lipopolysaccharide (LPS)-injected male and female rats (50 µg/animal) by longitudinal dynamic PET imaging in vivo. In LPS-injected rats, increased accumulation of 6-[18F]FDF was observed at 48 h post-LPS injection, with plateaued uptake (60-120 min) that was significantly higher in the ipsilateral vs. contralateral striatum (0.985 ± 0.047 and 0.819 ± 0.033 SUV, respectively; p = 0.002, n = 4M/3F). The ipsilateral-contralateral difference in striatal 6-[18F]FDF uptake expressed as binding potential (BPSRTM) peaked at 48 h (0.19 ± 0.11) and was significantly decreased at one and two weeks. In contrast, increased [18F]FDG uptake in the ipsilateral striatum was highest at one week post-LPS injection (BPSRTM = 0.25 ± 0.06, n = 4M). Iba-1 and GFAP immunohistochemistry confirmed LPS-induced activation of microglia and astrocytes, respectively, in ipsilateral striatum. This proof-of-concept study revealed an early response of 6-[18F]FDF to neuroinflammatory stimuli in rat brain. 6-[18F]FDF represents a potential PET radiotracer for imaging microglial GLUT5 density in brain with applications in neuroinflammatory and neurodegenerative diseases.
Collapse
Affiliation(s)
- Amanda J. Boyle
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Correspondence: (A.J.B.); (N.V.); Tel.: +1-416-535-8501 (ext. 30884) (A.J.B.); +1-416-535-8501 (ext. 30988) (N.V.)
| | - Emily Murrell
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Junchao Tong
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Christin Schifani
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Andrea Narvaez
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Melinda Wuest
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
| | - Frederick West
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Frank Wuest
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2N4, Canada
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Correspondence: (A.J.B.); (N.V.); Tel.: +1-416-535-8501 (ext. 30884) (A.J.B.); +1-416-535-8501 (ext. 30988) (N.V.)
| |
Collapse
|
23
|
Reagan AM, Christensen KE, Graham LC, Bedwell AA, Eldridge K, Speedy R, Figueiredo LL, Persohn SC, Bottiglieri T, Nho K, Sasner M, Territo PR, Rozen R, Howell GR. The 677C > T variant in methylenetetrahydrofolate reductase causes morphological and functional cerebrovascular deficits in mice. J Cereb Blood Flow Metab 2022; 42:2333-2350. [PMID: 36050860 PMCID: PMC9670012 DOI: 10.1177/0271678x221122644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 02/03/2023]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) particularly Alzheimer's disease and related dementias (ADRDs) are increasing; however, mechanisms driving cerebrovascular decline are poorly understood. Methylenetetrahydrofolate reductase (MTHFR) is a critical enzyme in the folate and methionine cycles. Variants in MTHFR, notably 677 C > T, are associated with dementias, but no mouse model existed to identify mechanisms by which MTHFR677C > T increases risk. Therefore, MODEL-AD created a novel knock-in (KI) strain carrying the Mthfr677C > T allele on the C57BL/6J background (Mthfr677C > T) to characterize morphology and function perturbed by the variant. Consistent with human clinical data, Mthfr677C > T mice have reduced enzyme activity in the liver and elevated plasma homocysteine levels. MTHFR enzyme activity is also reduced in the Mthfr677C > T brain. Mice showed reduced tissue perfusion in numerous brain regions by PET/CT as well as significantly reduced vascular density, pericyte number and increased GFAP-expressing astrocytes in frontal cortex. Electron microscopy revealed cerebrovascular damage including endothelial and pericyte apoptosis, reduced luminal size, and increased astrocyte and microglial presence in the microenvironment. Collectively, these data support a mechanism by which variations in MTHFR perturb cerebrovascular health laying the foundation to incorporate our new Mthfr677C > T mouse model in studies examining genetic susceptibility for cerebrovascular dysfunction in ADRDs.
Collapse
Affiliation(s)
| | - Karen E Christensen
- Departments of Human Genetics and Pediatrics, McGill University,
Research Institute of the Health Center, Montreal, QC, Canada
| | | | - Amanda A Bedwell
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kierra Eldridge
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rachael Speedy
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lucas L Figueiredo
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Scott C Persohn
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor
Scott & White Research Institute, Dallas, TX, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Indiana Alzheimer’s Disease Research
Center, Department of Radiology and Imaging Sciences, Indiana University School
of Medicine, Indianapolis, IN, USA
| | | | - Paul R Territo
- Department of Medicine, Division of Clinical Pharmacology,
Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rima Rozen
- Departments of Human Genetics and Pediatrics, McGill University,
Research Institute of the Health Center, Montreal, QC, Canada
| | - Gareth R Howell
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences, Tufts University School
of Medicine, Boston, MA, USA
- Graduate School of Biomedical Sciences and Engineering,
University of Maine, Orono, ME, USA
| |
Collapse
|
24
|
Huang J. Novel brain PET imaging agents: Strategies for imaging neuroinflammation in Alzheimer’s disease and mild cognitive impairment. Front Immunol 2022; 13:1010946. [PMID: 36211392 PMCID: PMC9537554 DOI: 10.3389/fimmu.2022.1010946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disease with a concealed onset and continuous deterioration. Mild cognitive impairment (MCI) is the prodromal stage of AD. Molecule-based imaging with positron emission tomography (PET) is critical in tracking pathophysiological changes among AD and MCI patients. PET with novel targets is a promising approach for diagnostic imaging, particularly in AD patients. Our present review overviews the current status and applications of in vivo molecular imaging toward neuroinflammation. Although radiotracers can remarkably diagnose AD and MCI patients, a variety of limitations prevent the recommendation of a single technique. Recent studies examining neuroinflammation PET imaging suggest an alternative approach to evaluate disease progression. This review concludes that PET imaging towards neuroinflammation is considered a promising approach to deciphering the enigma of the pathophysiological process of AD and MCI.
Collapse
|
25
|
Huang G, Lu X, Qiu Y, Bi L, Ye P, Yang M, Shen Y, Jin H, Han J. Hetero-aryl bromide precursor fluorine-18 radiosynthesis and preclinical evaluation of a novel positron emission tomography (PET) tracer [ 18F]GSK1482160. Bioorg Med Chem 2022; 73:116996. [PMID: 36126443 DOI: 10.1016/j.bmc.2022.116996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/26/2022]
Abstract
The purinergic P2X7 receptor (P2X7R), an ATP gated ion channel, is an important therapeutic target for various inflammatory immune and neurodegenerative diseases. A novel P2X7R targeting radiotracer GSK1482160 was radiosynthesized by hetero-aryl bromides precursor 10 with [18F]Et4NF, 20-30 % radiochemical yield, > 68 GBq/μmol specific activity, >98 % radiochemical purity. Evaluation in healthy male Sprague-Dawley rats revealed that [18F]GSK1482160 ([18F]11) was stably retained 87.81 %, 72.45 %, and 56.32 % in brain, blood and liver respectively 60-min post-injection. Ex-vivo biodistribution of [18F]11 proved that it was able to target the P2X7R in vivo and there was no defluorination in the major organs. PET/MRI imaging and autoradiography revealed that [18F]11 was able to penetrate the blood-brain barrier (BBB) and to be a promising P2X7R PET radioligand for clinical translation.
Collapse
Affiliation(s)
- Guolong Huang
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai 200032, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xiaolei Lu
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai 200032, China
| | - Yifan Qiu
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Lei Bi
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Peizhen Ye
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Min Yang
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yanfang Shen
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Hongjun Jin
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Junbin Han
- Institute of Radiation Medicine, Fudan University, Xietu Road 2094, Shanghai 200032, China.
| |
Collapse
|
26
|
Genetzakis E, Gilchrist J, Kassiou M, Figtree GA. Development and clinical translation of P2X7 receptor antagonists: A potential therapeutic target in coronary artery disease? Pharmacol Ther 2022; 237:108228. [DOI: 10.1016/j.pharmthera.2022.108228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/17/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022]
|
27
|
Zucker E, Burd I. P2X7 receptor as a potential therapeutic target for perinatal brain injury associated with preterm birth. Exp Neurol 2022; 357:114207. [PMID: 35985555 DOI: 10.1016/j.expneurol.2022.114207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 01/09/2023]
Abstract
Inflammation-induced preterm birth is the leading cause of perinatal mortality and long-term sequelae in surviving children. IL-1β is a major contributor to inflammation-induced preterm labor and its sequelae. It has recently been demonstrated that the cytokine storm and its progression depend on IL-1β release into circulation and that the P2X7 receptor (P2X7R) is the key player of the ATP-driven NLRP3/caspase-1 activation, necessary for the cleavage of pro-IL-1β to its mature form as well as its subsequent secretion. Being a key component to the inflammatory cascade, P2X7R illuminates a new therapeutic avenue to halt progression of inflammation prior to perinatal brain injury. In this review, we summarize the basic role of the P2X7 receptor in the inflammatory signaling cascade and the possibility of it being used as a therapeutic target in perinatal brain injury. We discuss the antagonists and agonists of the receptor as well as its role in other inflammatory diseases, showing the importance of discovering the functions of the receptor.
Collapse
Affiliation(s)
- Emily Zucker
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
28
|
Oblak AL, Kotredes KP, Pandey RS, Reagan AM, Ingraham C, Perkins B, Lloyd C, Baker D, Lin PB, Soni DM, Tsai AP, Persohn SA, Bedwell AA, Eldridge K, Speedy R, Meyer JA, Peters JS, Figueiredo LL, Sasner M, Territo PR, Sukoff Rizzo SJ, Carter GW, Lamb BT, Howell GR. Plcg2M28L Interacts With High Fat/High Sugar Diet to Accelerate Alzheimer's Disease-Relevant Phenotypes in Mice. Front Aging Neurosci 2022; 14:886575. [PMID: 35813947 PMCID: PMC9263289 DOI: 10.3389/fnagi.2022.886575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Obesity is recognized as a significant risk factor for Alzheimer's disease (AD). Studies have supported the notion that obesity accelerates AD-related pathophysiology in mouse models of AD. The majority of studies, to date, have focused on the use of early-onset AD models. Here, we evaluate the impact of genetic risk factors on late-onset AD (LOAD) in mice fed with a high fat/high sugar diet (HFD). We focused on three mouse models created through the IU/JAX/PITT MODEL-AD Center. These included a combined risk model with APOE4 and a variant in triggering receptor expressed on myeloid cells 2 (Trem2R47H ). We have termed this model, LOAD1. Additional variants including the M28L variant in phospholipase C Gamma 2 (Plcg2M28L ) and the 677C > T variant in methylenetetrahydrofolate reductase (Mthfr 677C > T ) were engineered by CRISPR onto LOAD1 to generate LOAD1.Plcg2M28L and LOAD1.Mthfr 677C > T . At 2 months of age, animals were placed on an HFD that induces obesity or a control diet (CD), until 12 months of age. Throughout the study, blood was collected to assess the levels of cholesterol and glucose. Positron emission tomography/computed tomography (PET/CT) was completed prior to sacrifice to image for glucose utilization and brain perfusion. After the completion of the study, blood and brains were collected for analysis. As expected, animals fed a HFD, showed a significant increase in body weight compared to those fed a CD. Glucose increased as a function of HFD in females only with cholesterol increasing in both sexes. Interestingly, LOAD1.Plcg2M28L demonstrated an increase in microglia density and alterations in regional brain glucose and perfusion on HFD. These changes were not observed in LOAD1 or LOAD1.Mthfr 677C > T animals fed with HFD. Furthermore, LOAD1.Plcg2M28L but not LOAD1.Mthfr 677C > T or LOAD1 animals showed transcriptomics correlations with human AD modules. Our results show that HFD affects the brain in a genotype-specific manner. Further insight into this process may have significant implications for the development of lifestyle interventions for the treatment of AD.
Collapse
Affiliation(s)
- Adrian L. Oblak
- Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | | - Ravi S. Pandey
- The Jackson Laboratory, Bar Harbor, ME, United States
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | | - Cynthia Ingraham
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Bridget Perkins
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Christopher Lloyd
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Deborah Baker
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Peter B. Lin
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Disha M. Soni
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Andy P. Tsai
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Scott A. Persohn
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Amanda A. Bedwell
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Kierra Eldridge
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Rachael Speedy
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Jill A. Meyer
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Johnathan S. Peters
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Lucas L. Figueiredo
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | | - Paul R. Territo
- Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Stacey J. Sukoff Rizzo
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Bruce T. Lamb
- Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | | |
Collapse
|
29
|
Therapeutic potentials and structure-activity relationship of 1,3-benzodioxole N-carbamothioyl carboxamide derivatives as selective and potent antagonists of P2X4 and P2X7 receptors. Eur J Med Chem 2022; 238:114491. [DOI: 10.1016/j.ejmech.2022.114491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/21/2022] [Indexed: 11/19/2022]
|
30
|
Cui WW, Wang SY, Zhang YQ, Wang Y, Fan YZ, Guo CR, Li XH, Lei YT, Wang WH, Yang XN, Hattori M, Li CZ, Wang J, Yu Y. P2X3-selective mechanism of Gefapixant, a drug candidate for the treatment of refractory chronic cough. Comput Struct Biotechnol J 2022; 20:1642-1653. [PMID: 35465163 PMCID: PMC9014320 DOI: 10.1016/j.csbj.2022.03.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/16/2022] [Accepted: 03/27/2022] [Indexed: 11/25/2022] Open
Abstract
The mechanism by which Gefapixant/AF-219 selectively acts on the P2X3 receptor is unclear. The negative allosteric site of AF-219 at P2X3 is also a potent allosteric site for other P2X subtypes. The selectivity of AF-219 for P2X3 is determined by the accessibility of binding site and the internal shape of this pocket. The finding will provide new perspectives for drug design against P2X3-mediated diseases such as RCC.
Gefapixant/AF-219, a selective inhibitor of the P2X3 receptor, is the first new drug other than dextromethorphan to be approved for the treatment of refractory chronic cough (RCC) in nearly 60 years. To date, seven P2X subtypes (P2X1-7) activated by extracellular ATP have been cloned, and subtype selectivity of P2X inhibitors is a prerequisite for reducing side effects. We previously identified the site and mechanism of action of Gefapixant/AF-219 on the P2X3 receptor, which occupies a pocket consisting of the left flipper (LF) and lower body (LB) domains. However, the mechanism by which AF-219 selectively acts on the P2X3 receptor is unknown. Here, we combined mutagenesis, chimera construction, molecular simulations, covalent occupation and chemical synthesis, and find that the negative allosteric site of AF-219 at P2X3 is also present in other P2X subtypes, at least for P2X1, P2X2 and P2X4. By constructing each chimera of AF-219 sensitive P2X3 and insensitive P2X2 subtypes, the insensitive P2X2 subtype was made to acquire the inhibitory properties of AF-219 and AF-353, an analog of AF-219 with higher affinity. Our results suggest that the selectivity of AF-219/AF-353 for P2X3 over the other P2X subtypes is determined by a combination of the accessibility of P2X3 binding site and the internal shape of this pocket, a finding that could provide new perspectives for drug design against P2X3-mediated diseases such as RCC, idiopathic pulmonary fibrosis, hypertension and overactive bladder disorder.
Collapse
|
31
|
Brumberg J, Varrone A. New PET radiopharmaceuticals for imaging CNS diseases. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
32
|
Chen Z, Haider A, Chen J, Xiao Z, Gobbi L, Honer M, Grether U, Arnold SE, Josephson L, Liang SH. The Repertoire of Small-Molecule PET Probes for Neuroinflammation Imaging: Challenges and Opportunities beyond TSPO. J Med Chem 2021; 64:17656-17689. [PMID: 34905377 PMCID: PMC9094091 DOI: 10.1021/acs.jmedchem.1c01571] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Neuroinflammation is an adaptive response of the central nervous system to diverse potentially injurious stimuli, which is closely associated with neurodegeneration and typically characterized by activation of microglia and astrocytes. As a noninvasive and translational molecular imaging tool, positron emission tomography (PET) could provide a better understanding of neuroinflammation and its role in neurodegenerative diseases. Ligands to translator protein (TSPO), a putative marker of neuroinflammation, have been the most commonly studied in this context, but they suffer from serious limitations. Herein we present a repertoire of different structural chemotypes and novel PET ligand design for classical and emerging neuroinflammatory targets beyond TSPO. We believe that this Perspective will support multidisciplinary collaborations in academic and industrial institutions working on neuroinflammation and facilitate the progress of neuroinflammation PET probe development for clinical use.
Collapse
Affiliation(s)
- Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Jiahui Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Zhiwei Xiao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Luca Gobbi
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Michael Honer
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Uwe Grether
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Steven E. Arnold
- Department of Neurology and the Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, Massachusetts 02129, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| |
Collapse
|
33
|
van der Wildt B, Janssen B, Pekošak A, Stéen EJL, Schuit RC, Kooijman EJM, Beaino W, Vugts DJ, Windhorst AD. Novel Thienopyrimidine-Based PET Tracers for P2Y 12 Receptor Imaging in the Brain. ACS Chem Neurosci 2021; 12:4465-4474. [PMID: 34757711 PMCID: PMC8640995 DOI: 10.1021/acschemneuro.1c00641] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
The P2Y12 receptor (P2Y12R) is uniquely expressed
on microglia in the brain, and its expression level directly depends
on the microglial activation state. Therefore, P2Y12R provides
a promising imaging marker for distinguishing the pro- and anti-inflammatory
microglial phenotypes, both of which play crucial roles in neuroinflammatory
diseases. In this study, three P2Y12R antagonists were
selected from the literature, radiolabeled with carbon-11 or fluorine-18,
and evaluated in healthy Wistar rats. Brain imaging was performed
with and without blocking of efflux transporters P-glycoprotein and breast cancer resistance protein using tariquidar.
Low brain uptake in healthy rats was observed for all tracers at baseline
conditions, whereas blocking of efflux transporters resulted in a
strong (6–7 fold) increase in brain uptake for both of them.
Binding of the most promising tracer, [18F]3, was further evaluated by in vitro autoradiography on rat brain
sections, ex vivo metabolite studies, and in vivo P2Y12R blocking studies. In vitro binding of [18F]3 on rat brain sections indicated high P2Y12R targeting
with approximately 70% selective and specific binding. At 60 min post-injection,
over 95% of radioactivity in the brain accounted for an intact tracer.
In blood plasma, still 40% intact tracer was found, and formed metabolites
did not enter the brain. A moderate P2Y12R blocking effect
was observed in vivo by positron emission tomography (PET) imaging
with [18F]3 (p = 0.04). To
conclude, three potential P2Y12R PET tracers were obtained
and analyzed for P2Y12R targeting in the brain. Unfortunately,
the brain uptake appeared low. Future work will focus on the design
of P2Y12R inhibitors with improved physicochemical characteristics
to reduce efflux transport and increase brain penetration.
Collapse
Affiliation(s)
- Berend van der Wildt
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Bieneke Janssen
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Aleksandra Pekošak
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - E. Johanna L. Stéen
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Robert C. Schuit
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Esther J. M. Kooijman
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Wissam Beaino
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Danielle J. Vugts
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Albert D. Windhorst
- Department of Radiology & Nuclear Medicine, Neuroscience Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
34
|
Nutma E, Ceyzériat K, Amor S, Tsartsalis S, Millet P, Owen DR, Papadopoulos V, Tournier BB. Cellular sources of TSPO expression in healthy and diseased brain. Eur J Nucl Med Mol Imaging 2021; 49:146-163. [PMID: 33433698 PMCID: PMC8712293 DOI: 10.1007/s00259-020-05166-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022]
Abstract
The 18 kDa translocator protein (TSPO) is a highly conserved protein located in the outer mitochondrial membrane. TSPO binding, as measured with positron emission tomography (PET), is considered an in vivo marker of neuroinflammation. Indeed, TSPO expression is altered in neurodegenerative, neuroinflammatory, and neuropsychiatric diseases. In PET studies, the TSPO signal is often viewed as a marker of microglial cell activity. However, there is little evidence in support of a microglia-specific TSPO expression. This review describes the cellular sources and functions of TSPO in animal models of disease and human studies, in health, and in central nervous system diseases. A discussion of methods of analysis and of quantification of TSPO is also presented. Overall, it appears that the alterations of TSPO binding, their cellular underpinnings, and the functional significance of such alterations depend on many factors, notably the pathology or the animal model under study, the disease stage, and the involved brain regions. Thus, further studies are needed to fully determine how changes in TSPO binding occur at the cellular level with the ultimate goal of revealing potential therapeutic pathways.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Division of Nuclear medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
- Division of Radiation Oncology, Department of Oncology, University Hospitals of Geneva, Geneva, Switzerland
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - David R Owen
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
35
|
Kotredes KP, Oblak A, Pandey RS, Lin PBC, Garceau D, Williams H, Uyar A, O’Rourke R, O’Rourke S, Ingraham C, Bednarczyk D, Belanger M, Cope Z, Foley KE, Logsdon BA, Mangravite LM, Sukoff Rizzo SJ, Territo PR, Carter GW, Sasner M, Lamb BT, Howell GR. Uncovering Disease Mechanisms in a Novel Mouse Model Expressing Humanized APOEε4 and Trem2*R47H. Front Aging Neurosci 2021; 13:735524. [PMID: 34707490 PMCID: PMC8544520 DOI: 10.3389/fnagi.2021.735524] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Late-onset Alzheimer's disease (AD; LOAD) is the most common human neurodegenerative disease, however, the availability and efficacy of disease-modifying interventions is severely lacking. Despite exceptional efforts to understand disease progression via legacy amyloidogenic transgene mouse models, focus on disease translation with innovative mouse strains that better model the complexity of human AD is required to accelerate the development of future treatment modalities. LOAD within the human population is a polygenic and environmentally influenced disease with many risk factors acting in concert to produce disease processes parallel to those often muted by the early and aggressive aggregate formation in popular mouse strains. In addition to extracellular deposits of amyloid plaques and inclusions of the microtubule-associated protein tau, AD is also defined by synaptic/neuronal loss, vascular deficits, and neuroinflammation. These underlying processes need to be better defined, how the disease progresses with age, and compared to human-relevant outcomes. To create more translatable mouse models, MODEL-AD (Model Organism Development and Evaluation for Late-onset AD) groups are identifying and integrating disease-relevant, humanized gene sequences from public databases beginning with APOEε4 and Trem2*R47H, two of the most powerful risk factors present in human LOAD populations. Mice expressing endogenous, humanized APOEε4 and Trem2*R47H gene sequences were extensively aged and assayed using a multi-disciplined phenotyping approach associated with and relative to human AD pathology. Robust analytical pipelines measured behavioral, transcriptomic, metabolic, and neuropathological phenotypes in cross-sectional cohorts for progression of disease hallmarks at all life stages. In vivo PET/MRI neuroimaging revealed regional alterations in glycolytic metabolism and vascular perfusion. Transcriptional profiling by RNA-Seq of brain hemispheres identified sex and age as the main sources of variation between genotypes including age-specific enrichment of AD-related processes. Similarly, age was the strongest determinant of behavioral change. In the absence of mouse amyloid plaque formation, many of the hallmarks of AD were not observed in this strain. However, as a sensitized baseline model with many additional alleles and environmental modifications already appended, the dataset from this initial MODEL-AD strain serves an important role in establishing the individual effects and interaction between two strong genetic risk factors for LOAD in a mouse host.
Collapse
Affiliation(s)
| | - Adrian Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana University Bloomington, Indianapolis, IN, United States
| | | | - Peter Bor-Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana University Bloomington, Indianapolis, IN, United States
| | - Dylan Garceau
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Asli Uyar
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Rita O’Rourke
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Cynthia Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana University Bloomington, Indianapolis, IN, United States
| | | | | | - Zackary Cope
- Department of Medicine—Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kate E. Foley
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | | | - Stacey J. Sukoff Rizzo
- Department of Medicine—Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Paul R. Territo
- Stark Neurosciences Research Institute, School of Medicine, Indiana University Bloomington, Indianapolis, IN, United States
| | | | | | - Bruce T. Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana University Bloomington, Indianapolis, IN, United States
| | | |
Collapse
|
36
|
Zhou R, Ji B, Kong Y, Qin L, Ren W, Guan Y, Ni R. PET Imaging of Neuroinflammation in Alzheimer's Disease. Front Immunol 2021; 12:739130. [PMID: 34603323 PMCID: PMC8481830 DOI: 10.3389/fimmu.2021.739130] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation play an important role in Alzheimer's disease pathogenesis. Advances in molecular imaging using positron emission tomography have provided insights into the time course of neuroinflammation and its relation with Alzheimer's disease central pathologies in patients and in animal disease models. Recent single-cell sequencing and transcriptomics indicate dynamic disease-associated microglia and astrocyte profiles in Alzheimer's disease. Mitochondrial 18-kDa translocator protein is the most widely investigated target for neuroinflammation imaging. New generation of translocator protein tracers with improved performance have been developed and evaluated along with tau and amyloid imaging for assessing the disease progression in Alzheimer's disease continuum. Given that translocator protein is not exclusively expressed in glia, alternative targets are under rapid development, such as monoamine oxidase B, matrix metalloproteinases, colony-stimulating factor 1 receptor, imidazoline-2 binding sites, cyclooxygenase, cannabinoid-2 receptor, purinergic P2X7 receptor, P2Y12 receptor, the fractalkine receptor, triggering receptor expressed on myeloid cells 2, and receptor for advanced glycation end products. Promising targets should demonstrate a higher specificity for cellular locations with exclusive expression in microglia or astrocyte and activation status (pro- or anti-inflammatory) with highly specific ligand to enable in vivo brain imaging. In this review, we summarised recent advances in the development of neuroinflammation imaging tracers and provided an outlook for promising targets in the future.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Nephrology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yanyan Kong
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Limei Qin
- Inner Mongolia Baicaotang Qin Chinese Mongolia Hospital, Hohhot, China
| | - Wuwei Ren
- School of Information Science and Technology, Shanghaitech University, Shanghai, China
| | - Yihui Guan
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, University of Zurich & Eidgenössische Technische Hochschule Zürich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
37
|
P2X7 receptor blockade reduces tau induced toxicity, therapeutic implications in tauopathies. Prog Neurobiol 2021; 208:102173. [PMID: 34516970 DOI: 10.1016/j.pneurobio.2021.102173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 01/28/2023]
Abstract
Tauopathies are neurodegenerative diseases characterized by the presence of aberrant intraneuronal aggregates of hyperphosphorylated Tau protein. Recent studies suggest that associated chronic neuroinflammation may contribute to the pathological Tau dissemination. However, the underlying molecular mechanisms remain unknown. Since purinergic P2X7 receptors (P2X7) can sense the rise of extracellular ATP levels associated with neuroinflammation, its involvement in neurodegeneration-associated inflammation was suggested. We found a P2X7 upregulation in patients diagnosed with different tauopathies and in a tauopathy mouse model, P301S mice. In vivo pharmacological or genetic blockade of P2X7 reverted microglial activation in P301S mice leading to a reduction in microglial migratory, secretory, and proliferative capacities, and promoting phagocytic function. Furthermore, it reduced the intraneuronal phosphorylated Tau levels in a GSK3-dependent way and increased extracellular phosphorylated Tau levels by reducing the expression of ectoenzyme TNAP. Accordingly, pharmacological or genetic blockade of P2X7 improved the cellular survival, motor and memory deficits and anxiolytic profile in P301S mice. Contrary, P2X7 overexpression caused a significant worsening of Tau-induced toxicity and aggravated the deteriorated motor and memory deficits in P301S mice. Our results indicate that P2X7 plays a deleterious role in tauopathies and suggest that its blockade may be a promising approach to treat Tauopathies.
Collapse
|
38
|
Mertens N, Schmidt ME, Hijzen A, Van Weehaeghe D, Ravenstijn P, Depre M, de Hoon J, Van Laere K, Koole M. Minimally invasive quantification of cerebral P2X7R occupancy using dynamic [ 18F]JNJ-64413739 PET and MRA-driven image derived input function. Sci Rep 2021; 11:16172. [PMID: 34373571 PMCID: PMC8352986 DOI: 10.1038/s41598-021-95715-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 07/29/2021] [Indexed: 01/21/2023] Open
Abstract
[18F]JNJ-64413739 has been evaluated as PET-ligand for in vivo quantification of purinergic receptor subtype 7 receptor (P2X7R) using Logan graphical analysis with a metabolite-corrected arterial plasma input function. In the context of a P2X7R PET dose occupancy study, we evaluated a minimally invasive approach by limiting arterial sampling to baseline conditions. Meanwhile, post dose distribution volumes (VT) under blocking conditions were estimated by combining baseline blood to plasma ratios and metabolite fractions with an MR angiography driven image derived input function (IDIF). Regional postdose VT,IDIF values were compared with corresponding VT,AIF estimates using a arterial input function (AIF), in terms of absolute values, test–retest reliability and receptor occupancy. Compared to an invasive AIF approach, postdose VT,IDIF values and corresponding receptor occupancies showed only limited bias (Bland–Altman analysis: 0.06 ± 0.27 and 3.1% ± 6.4%) while demonstrating a high correlation (Spearman ρ = 0.78 and ρ = 0.98 respectively). In terms of test–retest reliability, regional intraclass correlation coefficients were 0.98 ± 0.02 for VT,IDIF compared to 0.97 ± 0.01 for VT,AIF. These results confirmed that a postdose IDIF, guided by MR angiography and using baseline blood and metabolite data, can be considered for accurate [18F]JNJ-64413739 PET quantification in a repeated PET study design, thus avoiding multiple invasive arterial sampling and increasing dosing flexibility.
Collapse
Affiliation(s)
- Nathalie Mertens
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | | | - Anja Hijzen
- Janssen Research and Development, Beerse, Belgium
| | - Donatienne Van Weehaeghe
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | | | - Marleen Depre
- Center for Clinical Pharmacology, University Hospital and KU Leuven, Leuven, Belgium
| | - Jan de Hoon
- Center for Clinical Pharmacology, University Hospital and KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University Hospital and KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
39
|
Beaino W, Janssen B, Vugts DJ, de Vries HE, Windhorst AD. Towards PET imaging of the dynamic phenotypes of microglia. Clin Exp Immunol 2021; 206:282-300. [PMID: 34331705 PMCID: PMC8561701 DOI: 10.1111/cei.13649] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023] Open
Abstract
There is increasing evidence showing the heterogeneity of microglia activation in neuroinflammatory and neurodegenerative diseases. It has been hypothesized that pro‐inflammatory microglia are detrimental and contribute to disease progression, while anti‐inflammatory microglia play a role in damage repair and remission. The development of therapeutics targeting the deleterious glial activity and modulating it into a regenerative phenotype relies heavily upon a clearer understanding of the microglia dynamics during disease progression and the ability to monitor therapeutic outcome in vivo. To that end, molecular imaging techniques are required to assess microglia dynamics and study their role in disease progression as well as to evaluate the outcome of therapeutic interventions. Positron emission tomography (PET) is such a molecular imaging technique, and provides unique capabilities for non‐invasive quantification of neuroinflammation and has the potential to discriminate between microglia phenotypes and define their role in the disease process. However, several obstacles limit the possibility for selective in vivo imaging of microglia phenotypes mainly related to the poor characterization of specific targets that distinguish the two ends of the microglia activation spectrum and lack of suitable tracers. PET tracers targeting translocator protein 18 kDa (TSPO) have been extensively explored, but despite the success in evaluating neuroinflammation they failed to discriminate between microglia activation statuses. In this review, we highlight the current knowledge on the microglia phenotypes in the major neuroinflammatory and neurodegenerative diseases. We also discuss the current and emerging PET imaging targets, the tracers and their potential in discriminating between the pro‐ and anti‐inflammatory microglia activation states.
Collapse
Affiliation(s)
- Wissam Beaino
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Bieneke Janssen
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Danielle J Vugts
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| |
Collapse
|
40
|
Zarrinmayeh H, Territo PR. Purinergic Receptors of the Central Nervous System: Biology, PET Ligands, and Their Applications. Mol Imaging 2021; 19:1536012120927609. [PMID: 32539522 PMCID: PMC7297484 DOI: 10.1177/1536012120927609] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purinergic receptors play important roles in central nervous system (CNS). These receptors are involved in cellular neuroinflammatory responses that regulate functions of neurons, microglial and astrocytes. Based on their endogenous ligands, purinergic receptors are classified into P1 or adenosine, P2X and P2Y receptors. During brain injury or under pathological conditions, rapid diffusion of extracellular adenosine triphosphate (ATP) or uridine triphosphate (UTP) from the damaged cells, promote microglial activation that result in the changes in expression of several of these receptors in the brain. Imaging of the purinergic receptors with selective Positron Emission Tomography (PET) radioligands has advanced our understanding of the functional roles of some of these receptors in healthy and diseased brains. In this review, we have accumulated a list of currently available PET radioligands of the purinergic receptors that are used to elucidate the receptor functions and participations in CNS disorders. We have also reviewed receptors lacking radiotracer, laying the foundation for future discoveries of novel PET radioligands to reveal these receptors roles in CNS disorders.
Collapse
Affiliation(s)
- Hamideh Zarrinmayeh
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul R Territo
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
41
|
Drill M, Jones NC, Hunn M, O'Brien TJ, Monif M. Antagonism of the ATP-gated P2X7 receptor: a potential therapeutic strategy for cancer. Purinergic Signal 2021; 17:215-227. [PMID: 33728582 PMCID: PMC8155177 DOI: 10.1007/s11302-021-09776-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
The P2X receptor 7 (P2X7R) is a plasma membrane receptor sensing extracellular ATP associated with a wide variety of cellular functions. It is most commonly expressed on immune cells and is highly upregulated in a number of human cancers where it can play a trophic role in tumorigenesis. Activation of this receptor leads to the formation of a non-selective cation channel, which has been associated with several cellular functions mediated by the PI3K/Akt pathway and protein kinases. Due to its broad range of functions, the receptor represents a potential therapeutic target for a number of cancers. This review describes the range of mechanisms associated with P2X7R activation in cancer settings and highlights the potential of targeted inhibition of P2X7R as a therapy. It also describes in detail a number of key P2X7R antagonists currently in pre-clinical and clinical development, including oxidised ATP, Brilliant Blue G (BBG), KN-62, KN-04, A740003, A438079, GSK1482160, CE-224535, JNJ-54175446, JNJ-55308942, and AZ10606120. Lastly, it summarises the in vivo studies and clinical trials associated with the use and development of these P2X7R antagonists in different disease contexts.
Collapse
Affiliation(s)
- Matthew Drill
- Department of Neuroscience, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Melbourne University, Parkville, VIC, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Nigel C Jones
- Department of Neuroscience, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Martin Hunn
- Department of Neuroscience, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurosurgery, Alfred Hospital, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Mastura Monif
- Department of Neuroscience, Faculty of Medicine, Nursing and Health Sciences, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Department of Physiology, Melbourne University, Parkville, VIC, Australia.
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia.
- Department of Neurology, Melbourne Health, Parkville, VIC, Australia.
| |
Collapse
|
42
|
Merighi S, Poloni TE, Terrazzan A, Moretti E, Gessi S, Ferrari D. Alzheimer and Purinergic Signaling: Just a Matter of Inflammation? Cells 2021; 10:cells10051267. [PMID: 34065393 PMCID: PMC8161210 DOI: 10.3390/cells10051267] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a widespread neurodegenerative pathology responsible for about 70% of all cases of dementia. Adenosine is an endogenous nucleoside that affects neurodegeneration by activating four membrane G protein-coupled receptor subtypes, namely P1 receptors. One of them, the A2A subtype, is particularly expressed in the brain at the striatal and hippocampal levels and appears as the most promising target to counteract neurological damage and adenosine-dependent neuroinflammation. Extracellular nucleotides (ATP, ADP, UTP, UDP, etc.) are also released from the cell or are synthesized extracellularly. They activate P2X and P2Y membrane receptors, eliciting a variety of physiological but also pathological responses. Among the latter, the chronic inflammation underlying AD is mainly caused by the P2X7 receptor subtype. In this review we offer an overview of the scientific evidence linking P1 and P2 mediated purinergic signaling to AD development. We will also discuss potential strategies to exploit this knowledge for drug development.
Collapse
Affiliation(s)
- Stefania Merighi
- Department of Translational Medicine and for Romagna, University of Ferrara, 44100 Ferrara, Italy; (S.M.); (A.T.); (E.M.)
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation & ASP Golgi-Redaelli, Abbiategrasso, 20081 Milan, Italy;
| | - Anna Terrazzan
- Department of Translational Medicine and for Romagna, University of Ferrara, 44100 Ferrara, Italy; (S.M.); (A.T.); (E.M.)
| | - Eva Moretti
- Department of Translational Medicine and for Romagna, University of Ferrara, 44100 Ferrara, Italy; (S.M.); (A.T.); (E.M.)
| | - Stefania Gessi
- Department of Translational Medicine and for Romagna, University of Ferrara, 44100 Ferrara, Italy; (S.M.); (A.T.); (E.M.)
- Correspondence: (S.G.); (D.F.)
| | - Davide Ferrari
- Department of Life Science and Biotechnology, University of Ferrara, 44100 Ferrara, Italy
- Correspondence: (S.G.); (D.F.)
| |
Collapse
|
43
|
Abstract
The purine nucleotide ATP is a fundamental unit in cellular energy metabolism. Extracellular ATP and its metabolites are also ligands for a family of receptors, known as purinergic receptors, which are expressed ubiquitously in almost every cell type. In the immune system, extracellular ATP and its signals regulate the migration and activation of immune cells to orchestrate the induction and resolution of inflammation. In this review, we provide an overview of purinergic receptors and their downstream signaling related to macrophage activation. We also discuss the roles of purinergic signaling for macrophage functions in physiological and pathological conditions.
Collapse
Affiliation(s)
- Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine
| | - Naoki Takemura
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Tatsuya Saitoh
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
44
|
Territo PR, Zarrinmayeh H. P2X 7 Receptors in Neurodegeneration: Potential Therapeutic Applications From Basic to Clinical Approaches. Front Cell Neurosci 2021; 15:617036. [PMID: 33889073 PMCID: PMC8055960 DOI: 10.3389/fncel.2021.617036] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/02/2021] [Indexed: 12/27/2022] Open
Abstract
Purinergic receptors play important roles in central nervous system (CNS), where the bulk of these receptors are implicated in neuroinflammatory responses and regulation of cellular function of neurons, microglial and astrocytes. Within the P2X receptor family, P2X7 receptor is generally known for its inactivity in normal conditions and activation by moderately high concentrations (>100 μM) of extracellular adenosine 5′-triphosphate (ATP) released from injured cells as a result of brain injury or pathological conditions. Activation of P2X7R contributes to the activation and proliferation of microglia and directly contribute to neurodegeneration by provoking microglia-mediated neuronal death, glutamate-mediated excitotoxicity, and NLRP3 inflammasome activation that results in initiation, maturity and release of the pro-inflammatory cytokines and generation of reactive oxygen and nitrogen species. These components of the inflammatory response play important roles in many neural pathologies and neurodegeneration disorders. In CNS, expression of P2X7R on microglia, astrocytes, and oligodendrocytes are upregulated under neuroinflammatory conditions. Several in vivo studies have demonstrated beneficial effects of the P2X7 receptor antagonists in animal model systems of neurodegenerative diseases. A number of specific and selective P2X7 receptor antagonists have been developed, but only few of them have shown efficient brain permeability. Finding potent and selective P2X7 receptor inhibitors which are also CNS penetrable and display acceptable pharmacokinetics (PK) has presented challenges for both academic researchers and pharmaceutical companies. In this review, we discuss the role of P2X7 receptor function in neurodegenerative diseases, the pharmacological inhibition of the receptor, and PET radiopharmaceuticals which permit non-invasive monitoring of the P2X7 receptor contribution to neuroinflammation associated with neurodegeneration.
Collapse
Affiliation(s)
- Paul R Territo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hamideh Zarrinmayeh
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
45
|
Imaging Inflammation with Positron Emission Tomography. Biomedicines 2021; 9:biomedicines9020212. [PMID: 33669804 PMCID: PMC7922638 DOI: 10.3390/biomedicines9020212] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/28/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
The impact of inflammation on the outcome of many medical conditions such as cardiovascular diseases, neurological disorders, infections, cancer, and autoimmune diseases has been widely acknowledged. However, in contrast to neurological, oncologic, and cardiovascular disorders, imaging plays a minor role in research and management of inflammation. Imaging can provide insights into individual and temporospatial biology and grade of inflammation which can be of diagnostic, therapeutic, and prognostic value. There is therefore an urgent need to evaluate and understand current approaches and potential applications for imaging of inflammation. This review discusses radiotracers for positron emission tomography (PET) that have been used to image inflammation in cardiovascular diseases and other inflammatory conditions with a special emphasis on radiotracers that have already been successfully applied in clinical settings.
Collapse
|
46
|
Schubert JJ, Veronese M, Fryer TD, Manavaki R, Kitzbichler MG, Nettis MA, Mondelli V, Pariante CM, Bullmore ET, Turkheimer FE. A Modest Increase in 11C-PK11195-Positron Emission Tomography TSPO Binding in Depression Is Not Associated With Serum C-Reactive Protein or Body Mass Index. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 6:716-724. [PMID: 33515765 PMCID: PMC8264953 DOI: 10.1016/j.bpsc.2020.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/27/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023]
Abstract
Background Immune mechanisms have been implicated in the pathogenesis of depression. Translocator protein (TSPO)–targeted positron emission tomography (PET) has been used to assess neuroinflammation in major depressive disorder. We aimed to 1) test the hypothesis of significant case-control differences in TSPO binding in the anterior cingulate cortex, prefrontal cortex, and insula regions; and 2) explore the relationship between cerebral TSPO binding and peripheral blood C-reactive protein (CRP) concentration. Methods A total of 51 depressed subjects with Hamilton Depression Rating Scale score >13 (median 17; interquartile range, 16–22) and 25 healthy control subjects underwent dynamic brain 11C-PK11195 PET and peripheral blood immune marker characterization. Depressed subjects were divided into high CRP (>3 mg/L; n = 20) and low CRP (<3 mg/L; n = 31). Results Across the three regions, TSPO binding was significantly increased in depressed versus control subjects (η2p = .09; F1,71 = 6.97, p = .01), which was not influenced by body mass index. The case-control difference was greatest in the anterior cingulate cortex (d = 0.49; t74 = 2.00, p = .03) and not significant in the prefrontal cortex or insula (d = 0.27 and d = 0.36, respectively). Following CRP stratification, significantly higher TSPO binding was observed in low-CRP depression compared with controls (d = 0.53; t54 = 1.96, p = .03). These effect sizes are comparable to prior major depressive disorder case-control TSPO PET data. No significant correlations were observed between TSPO and CRP measures. Conclusions Consistent with previous findings, there is a modest increase in TSPO binding in depressed patients compared with healthy control subjects. The lack of a significant correlation between brain TSPO binding and blood CRP concentration or body mass index poses questions about the interactions between central and peripheral immune responses in the pathogenesis of depression.
Collapse
Affiliation(s)
- Julia J Schubert
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - Tim D Fryer
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom; Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Roido Manavaki
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Manfred G Kitzbichler
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Maria A Nettis
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; National Institute for Health and Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; National Institute for Health and Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; National Institute for Health and Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom; Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| |
Collapse
|
47
|
|
48
|
Meyer JH, Cervenka S, Kim MJ, Kreisl WC, Henter ID, Innis RB. Neuroinflammation in psychiatric disorders: PET imaging and promising new targets. Lancet Psychiatry 2020; 7:1064-1074. [PMID: 33098761 PMCID: PMC7893630 DOI: 10.1016/s2215-0366(20)30255-8] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/14/2023]
Abstract
Neuroinflammation is a multifaceted physiological and pathophysiological response of the brain to injury and disease. Given imaging findings of 18 kDa translocator protein (TSPO) and the development of radioligands for other inflammatory targets, PET imaging of neuroinflammation is at a particularly promising stage. This Review critically evaluates PET imaging results of inflammation in psychiatric disorders, including major depressive disorder, schizophrenia and psychosis disorders, substance use, and obsessive-compulsive disorder. We also consider promising new targets that can be measured in the brain, such as monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, colony stimulating factor 1 receptor, and the purinergic P2X7 receptor. Thus far, the most compelling TSPO imaging results have arguably been found in major depressive disorder, for which consistent increases have been observed, and in schizophrenia and psychosis, for which patients show reduced TSPO levels. This pattern highlights the importance of validating brain biomarkers of neuroinflammation for each condition separately before moving on to patient stratification and treatment monitoring trials.
Collapse
Affiliation(s)
- Jeffrey H Meyer
- Campbell Family Mental Health Research Institute, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm, Sweden
| | - Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
49
|
Cipollina G, Davari Serej A, Di Nolfi G, Gazzano A, Marsala A, Spatafora MG, Peviani M. Heterogeneity of Neuroinflammatory Responses in Amyotrophic Lateral Sclerosis: A Challenge or an Opportunity? Int J Mol Sci 2020; 21:E7923. [PMID: 33113845 PMCID: PMC7662281 DOI: 10.3390/ijms21217923] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex pathology: (i) the neurodegeneration is chronic and progressive; it starts focally in specific central nervous system (CNS) areas and spreads to different districts; (ii) multiple cell types further than motor neurons (i.e., glial/immune system cells) are actively involved in the disease; (iii) both neurosupportive and neurotoxic neuroinflammatory responses were identified. Microglia cells (a key player of neuroinflammation in the CNS) attracted great interest as potential target cell population that could be modulated to counteract disease progression, at least in preclinical ALS models. However, the heterogeneous/multifaceted microglia cell responses occurring in different CNS districts during the disease represent a hurdle for clinical translation of single-drug therapies. To address this issue, over the past ten years, several studies attempted to dissect the complexity of microglia responses in ALS. In this review, we shall summarize these results highlighting how the heterogeneous signature displayed by ALS microglia reflects not only the extent of neuronal demise in different regions of the CNS, but also variable engagement in the attempts to cope with the neuronal damage. We shall discuss novel avenues opened by the advent of single-cell and spatial transcriptomics technologies, underlining the potential for discovery of novel therapeutic targets, as well as more specific diagnostic/prognostic not-invasive markers of neuroinflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marco Peviani
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; (G.C.); (A.D.S.); (G.D.N.); (A.G.); (A.M.); (M.G.S.)
| |
Collapse
|
50
|
Beaino W, Janssen B, Kooijman E, Vos R, Schuit RC, O'Brien-Brown J, Kassiou M, van Het Hof B, Vugts DJ, de Vries HE, Windhorst AD. PET imaging of P2X 7R in the experimental autoimmune encephalomyelitis model of multiple sclerosis using [ 11C]SMW139. J Neuroinflammation 2020; 17:300. [PMID: 33054803 PMCID: PMC7556947 DOI: 10.1186/s12974-020-01962-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Non-invasive imaging of the activation status of microglia and the ability to identify a pro- or anti-inflammatory environment can provide valuable insights not only into pathogenesis of neuro-inflammatory and neurodegenerative diseases but also the monitoring of the efficacy of immunomodulatory therapies. P2X7R is highly expressed on pro-inflammatory microglia and [11C]SMW139, a specific P2X7R tracer for positron emission tomography imaging, showed good pharmacokinetics, stability, and brain permeability in vivo. Our objective was to evaluate the potential of [11C]SMW139 for PET imaging of neuroinflammation in vivo in the experimental autoimmune encephalomyelitis (EAE) model. METHODS We induced EAE in Lewis rats by immunization with MBP 69-88 in complete Freund's adjuvant (CFA). We determined the affinity of [11C]SMW139 to human and rat P2X7R using saturation binding assay. Using this tracer, PET imaging was performed at the peak of disease and in the recovery phase. In vivo blocking experiments were conducted to validate the specific brain uptake of the tracer. Immunohistochemistry staining and autoradiography were performed to evaluate the level of neuroinflammation and validate the specific binding of [11C]SMW139. RESULTS [11C]SMW139 showed good affinity for the rat P2X7R with a Kd of 20.6 ± 1.7 nM. The uptake of [11C]SMW139 was significantly higher in EAE animals at the peak of disease compared to the recovery phase but not in CFA control animals. The amplitude of increase of [11C]SMW139 uptake showed significant positive correlation with clinical scores mainly in the spinal cord (Pearson = 0.75, Spearman = 0.76; p < 0.0001). Treating EAE animals with P2X7R antagonist JNJ-47965567 blocked the uptake of [11C]SMW139 in the spinal cord, cerebellum, and brain stem, demonstrating specific accumulation of the tracer. P-glycoprotein blocking with tariquidar (30 mg/kg) did not affect tracer penetration in the brain showing that [11C]SMW139 is not a Pgp substrate. CONCLUSION Our data shows that [11C]SMW139 is a promising PET tracer for imaging neuroinflammation and evaluating the dynamics of pro-inflammatory microglia in the brain. This can provide crucial insights into the role of microglia in disease progression and enables the development of novel treatment strategies aimed at modulating the immune response in order to promote neuroprotection.
Collapse
Affiliation(s)
- Wissam Beaino
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands.
| | - Bieneke Janssen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands.,Present address: Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Esther Kooijman
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Ricardo Vos
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Robert C Schuit
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | | | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW, Australia
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, AUMC MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Danielle J Vugts
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, AUMC MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| |
Collapse
|