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
|
Laroy M, Van Laere K, Vandenbulcke M, Emsell L, Bouckaert F. Molecular Positron Emission Tomography and Single-Photon Emission Computed Tomography Imaging for Understanding the Neurobiological Mechanisms of Electroconvulsive Therapy: A Scoping Review. J ECT 2024:00124509-990000000-00238. [PMID: 39714318 DOI: 10.1097/yct.0000000000001094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
ABSTRACT Electroconvulsive therapy (ECT) effectively treats severe psychiatric disorders such as depression, mania, catatonia, and schizophrenia. Although its exact mechanism remains unclear, ECT is thought to induce neurochemical and neuroendocrine changes. Positron emission tomography (PET) and single-photon emission computed tomography (SPECT) have provided vital insights into ECT's neurobiological effects. This scoping review investigates the role of molecular imaging in understanding these effects. A systematic search across PubMed, EMBASE, Web of Science, Cochrane, and Scopus databases yielded 857 unique records, from which 45 peer-reviewed articles in English with longitudinal PET or SPECT measures in ECT patients were included. The review identifies 2 main research directions: ECT's impact on brain activity and neurotransmitters. Initial research assessed regional cerebral blood flow and regional glucose metabolism during ictal (during ECT), postictal (within 24 hours), short-term (within a week), and long-term (beyond a week) follow-up as markers of brain activity. Initial findings showed an anterior-posterior regional cerebral blood flow gradient during the ictal phase, with subsequent normalization of hypoperfusion in frontal and parietal regions, and persistent long-term effects. Later, research shifted to the monoamine hypothesis of depression, examining ECT's impact on serotonin and dopamine systems via PET imaging. Results on receptor availability post-ECT were mixed, showing both reductions and no significant changes, indicating variable effects. This scoping review further highlights the need to explore new targets, tailor methodologies for patient populations, and foster multicenter studies. Although SPECT has been valuable, advances in PET imaging now make it preferable, offering unparalleled insights into ECT's molecular and neurobiological mechanisms.
Collapse
Affiliation(s)
- Maarten Laroy
- From the Neuropsychiatry, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- Department of Nuclear Medicine and Molecular Imaging, University Hospitals Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
3
|
Emvalomenos GM, Kang JWM, Jupp B, Mychasiuk R, Keay KA, Henderson LA. Recent developments and challenges in positron emission tomography imaging of gliosis in chronic neuropathic pain. Pain 2024; 165:2184-2199. [PMID: 38713812 DOI: 10.1097/j.pain.0000000000003247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/05/2024] [Indexed: 05/09/2024]
Abstract
ABSTRACT Understanding the mechanisms that underpin the transition from acute to chronic pain is critical for the development of more effective and targeted treatments. There is growing interest in the contribution of glial cells to this process, with cross-sectional preclinical studies demonstrating specific changes in these cell types capturing targeted timepoints from the acute phase and the chronic phase. In vivo longitudinal assessment of the development and evolution of these changes in experimental animals and humans has presented a significant challenge. Recent technological advances in preclinical and clinical positron emission tomography, including the development of specific radiotracers for gliosis, offer great promise for the field. These advances now permit tracking of glial changes over time and provide the ability to relate these changes to pain-relevant symptomology, comorbid psychiatric conditions, and treatment outcomes at both a group and an individual level. In this article, we summarize evidence for gliosis in the transition from acute to chronic pain and provide an overview of the specific radiotracers available to measure this process, highlighting their potential, particularly when combined with ex vivo / in vitro techniques, to understand the pathophysiology of chronic neuropathic pain. These complementary investigations can be used to bridge the existing gap in the field concerning the contribution of gliosis to neuropathic pain and identify potential targets for interventions.
Collapse
Affiliation(s)
- Gaelle M Emvalomenos
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - James W M Kang
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Bianca Jupp
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Kevin A Keay
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Luke A Henderson
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| |
Collapse
|
4
|
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
|
5
|
Lee N, Choi JY, Ryu YH. The development status of PET radiotracers for evaluating neuroinflammation. Nucl Med Mol Imaging 2024; 58:160-176. [PMID: 38932754 PMCID: PMC11196502 DOI: 10.1007/s13139-023-00831-4] [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: 10/11/2023] [Revised: 11/16/2023] [Accepted: 12/05/2023] [Indexed: 06/28/2024] Open
Abstract
Neuroinflammation is associated with the pathophysiologies of neurodegenerative and psychiatric disorders. Evaluating neuroinflammation using positron emission tomography (PET) plays an important role in the early diagnosis and determination of proper treatment of brain diseases. To quantify neuroinflammatory responses in vivo, many PET tracers have been developed using translocator proteins, imidazole-2 binding site, cyclooxygenase, monoamine oxidase-B, adenosine, cannabinoid, purinergic P2X7, and CSF-1 receptors as biomarkers. In this review, we introduce the latest developments in PET tracers that can image neuroinflammation, focusing on clinical trials, and further consider their current implications.
Collapse
Affiliation(s)
- Namhun Lee
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
6
|
Martynov A, Farber B, Bomko T, Beckles DL, Kleyn I. Molecular Modeling, Synthesis, and Antihyperglycemic Activity of the New Benzimidazole Derivatives - Imidazoline Receptor Agonists. Drug Des Devel Ther 2024; 18:1035-1052. [PMID: 38585255 PMCID: PMC10999201 DOI: 10.2147/dddt.s447289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/28/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction The paper presents the results of a study on the first synthesized benzimidazole derivatives obtained from labile nature carboxylic acids. The synthesis conditions of these substances were studied, their structure was proved, and some components were found to have sugar-reducing activity on the model of alloxan diabetes in rats. Methods The study used molecular modeling methods such as docking based on the evolutionary model (igemdock), RP_HPLC method to monitor the synthesis reaction, and 1H NMR and 13C NMR, and other methods of organic chemistry to confirm the structures of synthesized substances. Results & Discussion The docking showed that the ursodeoxycholic acid benzimidazole derivatives have high tropics to all imidazoline receptor carriers (PDB ID: 2XCG, 2bk3, 3p0c, 1QH4). The ursodeoxycholic acid benzimidazole derivative and arginine and histidine benzimidazole derivatives showed the highest sugar-lowering activity in the experiment on alloxan-diabetic rats. For these derivatives, the difference in glucose levels of treated rats was significant against untreated control. Therefore, the new derivatives of benzimidazole and labile natural organic acids can be used to create new classes of imidazoline receptor inhibitors for the treatment of diabetes mellitus and hypertension.
Collapse
Affiliation(s)
- Artur Martynov
- Laboratory and Clinical department of Molecular Immunopharmacology, SI “ I. Mechnikov Institute of Microbiology and Immunology of National Academy of Medical Sciences of Ukraine, Kharkiv, Ukraine
| | | | - Tatyana Bomko
- Laboratory and Clinical department of Molecular Immunopharmacology, SI “ I. Mechnikov Institute of Microbiology and Immunology of National Academy of Medical Sciences of Ukraine, Kharkiv, Ukraine
| | | | - Ilya Kleyn
- SUNY Downstate Medical Center / University Hospital of Brooklyn, New York, NY, USA
| |
Collapse
|
7
|
Fontana IC, Kumar A, Okamura N, Nordberg A. Multitracer Approach to Understanding the Complexity of Reactive Astrogliosis in Alzheimer's Brains. ACS Chem Neurosci 2024; 15:328-336. [PMID: 38133820 PMCID: PMC10797624 DOI: 10.1021/acschemneuro.3c00646] [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: 10/07/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
A monoamine oxidase B (MAO-B) selective positron emission tomography (PET) tracer [11C]-deuterium-l-deprenyl holds promise for imaging reactive astrogliosis in neurodegenerative diseases, such as Alzheimer's disease (AD). Two novel PET tracers ([11C]-BU99008 and [18F]-SMBT-1) have recently been developed to assess the complexity of reactive astrogliosis in the AD continuum. We have investigated the binding properties of SMBT-1, l-deprenyl, and BU99008 in AD and cognitively normal control (CN) brains. Competition binding assays with [3H]-l-deprenyl and [3H]-BU99008 versus unlabeled SMBT-1 in postmortem AD and CN temporal and frontal cortex brains demonstrated that SMBT-1 interacted with [3H]-deprenyl at a single binding site (nM range) and with [3H]-BU99008 at multiple binding sites (from nM to μM). Autoradiography studies on large frozen postmortem AD and CN hemisphere brain sections demonstrated that 1 μM SMBT-1 almost completely displaced the [3H]-l-deprenyl binding (>90%), while SMBT-1 only partly displaced the [3H]-BU99008 binding (50-60% displacement) in cortical regions. In conclusion, SMBT-1, l-deprenyl, and BU99008 interact at the same MAO-B binding site, while BU99008 shows an additional independent binding site in AD and CN brains. The high translational power of our studies in human AD and CN brains suggests that the multitracer approach with SMBT-1, l-deprenyl, and BU99008 could be useful for imaging reactive astrogliosis.
Collapse
Affiliation(s)
- Igor C. Fontana
- Division
of Clinical Geriatrics, Center for Alzheimer Research, Department
of Neurobiology, Care Sciences and Society, Karolinska Institutet, S-141 83 Stockholm, Sweden
| | - Amit Kumar
- Division
of Clinical Geriatrics, Center for Alzheimer Research, Department
of Neurobiology, Care Sciences and Society, Karolinska Institutet, S-141 83 Stockholm, Sweden
| | - Nobuyuki Okamura
- Department
of Pharmacology, Tohoku Medical and Pharmaceutical
University, Sendai 983-8536, Japan
| | - Agneta Nordberg
- Division
of Clinical Geriatrics, Center for Alzheimer Research, Department
of Neurobiology, Care Sciences and Society, Karolinska Institutet, S-141 83 Stockholm, Sweden
- Theme
Inflammation and Aging, Karolinska University
Hospital, S-141 57 Stockholm, Sweden
| |
Collapse
|
8
|
Ozolmez N, Silindir-Gunay M, Volkan-Salanci B. An overview: Radiotracers and nano-radiopharmaceuticals for diagnosis of Parkinson's disease. Appl Radiat Isot 2024; 203:111110. [PMID: 37989065 DOI: 10.1016/j.apradiso.2023.111110] [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: 07/25/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023]
Abstract
Parkinson's disease (PD) is a widespread progressive neurodegenerative disease. Clinical diagnosis approaches are insufficient to provide an early and accurate diagnosis before a substantial of loss of dopaminergic neurons. PET and SPECT can be used for accurate and early diagnosis of PD by using target-specific radiotracers. Additionally, the importance of BBB penetrating targeted nanosystems has increased in recent years. This article reviews targeted radiopharmaceuticals used in clinics and novel nanocarriers for research purposes of PD imaging.
Collapse
Affiliation(s)
- Nur Ozolmez
- Hacettepe University, Faculty of Pharmacy, Department of Radiopharmacy, Ankara, Turkey.
| | - Mine Silindir-Gunay
- Hacettepe University, Faculty of Pharmacy, Department of Radiopharmacy, Ankara, Turkey.
| | - Bilge Volkan-Salanci
- Hacettepe University, Faculty of Medicine, Department of Nuclear Medicine, Ankara, Turkey.
| |
Collapse
|
9
|
Bagán A, Abás S, Palà-Pujadas J, Irisarri A, Griñán-Ferré C, Pallàs M, Muneta-Arrate I, Muguruza C, Callado LF, Pérez B, Molins E, Morales-García JÁ, Escolano C. Exploring the reactivity of bicyclic α-iminophosphonates to access new imidazoline I 2 receptor ligands. Bioorg Chem 2023; 142:106935. [PMID: 39492364 DOI: 10.1016/j.bioorg.2023.106935] [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: 07/02/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/05/2024]
Abstract
Recent studies pointed out the modulation of imidazoline I2 receptors (I2-IR) by selective ligands as a putative strategy to face neurodegenerative diseases. Foregoing the classical 2-imidazoline/imidazole-containing I2-IR ligands, we report a family of bicyclic α-iminophosphonates endowed with high affinity and selectivity upon I2-IR and we advanced a representative compound B06 in preclinical phases. In this paper, we describe the synthetic possibilities of bicyclic α-iminophosphonates by exploring its ambivalent reactivity, leading to unprecedented molecules that showed promising activities as I2-IR ligands in human brain tissues and good BBB permeation capabilities. After in silico ADME prediction studies, we assessed the neuroprotective properties of selected compounds and beneficial effect in an in vitro model of Alzheimeŕs and Parkinson's disease. Along with their neuroprotective effect, compounds showed a potent anti-inflammatory response when evaluated in a neuroinflammation cellular model. Moreover, this is the first time that the neuroprotective effects of imidazoline I2-IR ligands in a transgenic Alzheimer's disease Caenorhabditis elegans strain are investigated. Using a thrashing assay, we found a significant cognition improvement in this in vivo model after treatment with the new bicyclic α-phosphoprolines. Therefore, our results confirmed the need of exploring structurally new I2-IR ligands and their potential for therapeutic strategies in neurodegeneration.
Collapse
Affiliation(s)
- Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Sònia Abás
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Judith Palà-Pujadas
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Alba Irisarri
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31. E-08028, Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31. E-08028, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Mercè Pallàs
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31. E-08028, Barcelona, Spain
| | - Itziar Muneta-Arrate
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Carolina Muguruza
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Spain; Biocruces Bizkaia Health Research Institute Bizkaia, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology. Autonomous University of Barcelona, E-08193 Cerdanyola, Spain
| | - Elies Molins
- Institut de Ciència de Materials de Barcelona (CSIC), Campus UAB, E-08193 Cerdanyola, Spain
| | - José Á Morales-García
- Department of Cell Biology. School of Medicine, Complutense University (UCM), 28040 Madrid, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain.
| |
Collapse
|
10
|
Bagán A, Rodriguez-Arévalo S, Taboada-Jara T, Griñán-Ferré C, Pallàs M, Brocos-Mosquera I, Callado LF, Morales-García JA, Pérez B, Diaz C, Fernández-Godino R, Genilloud O, Beljkas M, Oljacic S, Nikolic K, Escolano C. Preclinical Evaluation of an Imidazole-Linked Heterocycle for Alzheimer's Disease. Pharmaceutics 2023; 15:2381. [PMID: 37896141 PMCID: PMC10610545 DOI: 10.3390/pharmaceutics15102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 10/29/2023] Open
Abstract
Humanity is facing a vast prevalence of neurodegenerative diseases, with Alzheimer's disease (AD) being the most dominant, without efficacious drugs, and with only a few therapeutic targets identified. In this scenario, we aim to find molecular entities that modulate imidazoline I2 receptors (I2-IRs) that have been pointed out as relevant targets in AD. In this work, we explored structural modifications of well-established I2-IR ligands, giving access to derivatives with an imidazole-linked heterocycle as a common key feature. We report the synthesis, the affinity in human I2-IRs, the brain penetration capabilities, the in silico ADMET studies, and the three-dimensional quantitative structure-activity relationship (3D-QSAR) studies of this new bunch of I2-IR ligands. Selected compounds showed neuroprotective properties and beneficial effects in an in vitro model of Parkinson's disease, rescued the human dopaminergic cell line SH-SY5Y from death after treatment with 6-hydroxydopamine, and showed crucial anti-inflammatory effects in a cellular model of neuroinflammation. After a preliminary pharmacokinetic study, we explored the action of our representative 2-(benzo[b]thiophen-2-yl)-1H-imidazole LSL33 in a mouse model of AD (5xFAD). Oral administration of LSL33 at 2 mg/Kg for 4 weeks ameliorated 5XFAD cognitive impairment and synaptic plasticity, as well as reduced neuroinflammation markers. In summary, this new I2-IR ligand that promoted beneficial effects in a well-established AD mouse model should be considered a promising therapeutic strategy for neurodegeneration.
Collapse
Affiliation(s)
- Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.B.); (S.R.-A.)
| | - Sergio Rodriguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.B.); (S.R.-A.)
| | - Teresa Taboada-Jara
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (T.T.-J.); (C.G.-F.); (M.P.)
| | - Christian Griñán-Ferré
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (T.T.-J.); (C.G.-F.); (M.P.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Mercè Pallàs
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (T.T.-J.); (C.G.-F.); (M.P.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), National Institute of Health Carlos III, 28029 Madrid, Spain
| | - Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain; (I.B.-M.); (L.F.C.)
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, 28029 Madrid, Spain
| | - Luis F. Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain; (I.B.-M.); (L.F.C.)
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, 28029 Madrid, Spain
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - José A. Morales-García
- Department of Cell Biology, School of Medicine, Complutense University (UCM), 28040 Madrid, Spain;
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Autonomous University of Barcelona, 08193 Cerdanyola, Spain;
| | - Caridad Diaz
- Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (R.F.-G.); (O.G.)
| | - Rosario Fernández-Godino
- Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (R.F.-G.); (O.G.)
| | - Olga Genilloud
- Fundación MEDINA Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (R.F.-G.); (O.G.)
| | - Milan Beljkas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.); (K.N.)
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.); (K.N.)
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.); (K.N.)
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.B.); (S.R.-A.)
| |
Collapse
|
11
|
Morito T, Harada R, Iwata R, Ishikawa Y, Okamura N, Kudo Y, Furumoto S, Yanai K, Tashiro M. Evaluation of 18F labeled glial fibrillary acidic protein binding nanobody and its brain shuttle peptide fusion proteins using a neuroinflammation rat model. PLoS One 2023; 18:e0287047. [PMID: 37315033 DOI: 10.1371/journal.pone.0287047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 05/27/2023] [Indexed: 06/16/2023] Open
Abstract
Astrogliosis is a crucial feature of neuroinflammation and is characterized by the significant upregulation of glial fibrillary acidic protein (GFAP) expression. Hence, visualizing GFAP in the living brain of patients with damaged central nervous system using positron emission tomography (PET) is of great importance, and it is expected to depict neuroinflammation more directly than existing neuroinflammation imaging markers. However, no PET radiotracers for GFAP are currently available. Therefore, neuroimaging with antibody-like affinity proteins could be a viable strategy for visualizing imaging targets that small molecules rarely recognize, such as GFAP, while we need to overcome the challenges of slow clearance and low brain permeability. The E9 nanobody, a small-affinity protein with high affinity and selectivity for GFAP, was utilized in this study. E9 was engineered by fusing a brain shuttle peptide that facilitates blood-brain barrier permeation via two different types of linker domains: E9-GS-ApoE (EGA) and E9-EAK-ApoE (EEA). E9, EGA and EEA were radiolabeled with fluorine-18 using cell-free protein radiosynthesis. In vitro autoradiography showed that all radiolabeled proteins exhibited a significant difference in neuroinflammation in the brain sections created from a rat model constructed by injecting lipopolysaccharide (LPS) into the unilateral striatum of wildtype rats, and an excess competitor displaced their binding. However, exploratory in vivo PET imaging and ex vivo biodistribution studies in the rat model failed to distinguish neuroinflammatory lesions within 3 h of 18F-EEA intravenous injection. This study contributes to a better understanding of the characteristics of small-affinity proteins fused with a brain shuttle peptide for further research into the use of protein molecules as PET tracers for imaging neuropathology.
Collapse
Affiliation(s)
- Takahiro Morito
- Division of Cyclotron Nuclear Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Ren Iwata
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan
| | - Yoichi Ishikawa
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Yukitsuka Kudo
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Shozo Furumoto
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Miyagi, Japan
| | - Kazuhiko Yanai
- Division of Cyclotron Nuclear Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Manabu Tashiro
- Division of Cyclotron Nuclear Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
12
|
Parker CA, Nutt DJ, Tyacke RJ. Imidazoline-I2 PET Tracers in Neuroimaging. Int J Mol Sci 2023; 24:9787. [PMID: 37372936 DOI: 10.3390/ijms24129787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Targeting neuroinflammation, and in particular, microglial activation and astrocytosis, is a current area of the focus of new treatment interventions for a number of neurodegenerative disorders. Probing the roles of microglia and astrocytes in human disease requires the development of useful tools, such as PET imaging tools that are specific for the cell type(s) of interest. This review concentrates on the recent advances in the development of Imidazoline2 binding site (I2BS) PET tracers, which are purported to target astrocytes, and hence could represent key clinical imaging tools for targeting astrocytes in neurodegenerative disease. Five PET tracers for the I2BS are described in this review, with only one (11C-BU99008) being currently validated to GMP for clinical use, and data reported from healthy volunteers, Alzheimer's disease patients, and Parkinson's disease patients. The clinical data utilising 11C-BU99008 have revealed the potential early involvement of astrogliosis in neurodegeneration that might precede the activation of microglia, which, if confirmed, could provide a vital new means for potentially targeting neurodegeneration earlier in the disease course.
Collapse
Affiliation(s)
- Christine A Parker
- Neuropsychopharmacology Unit, Division of Psychiatry, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
- GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | - David J Nutt
- Neuropsychopharmacology Unit, Division of Psychiatry, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Robin J Tyacke
- Neuropsychopharmacology Unit, Division of Psychiatry, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
13
|
Fontana IC, Scarpa M, Malarte ML, Rocha FM, Ausellé-Bosch S, Bluma M, Bucci M, Chiotis K, Kumar A, Nordberg A. Astrocyte Signature in Alzheimer's Disease Continuum through a Multi-PET Tracer Imaging Perspective. Cells 2023; 12:1469. [PMID: 37296589 PMCID: PMC10253101 DOI: 10.3390/cells12111469] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/02/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Reactive astrogliosis is an early event in the continuum of Alzheimer's disease (AD). Current advances in positron emission tomography (PET) imaging provide ways of assessing reactive astrogliosis in the living brain. In this review, we revisit clinical PET imaging and in vitro findings using the multi-tracer approach, and point out that reactive astrogliosis precedes the deposition of Aβ plaques, tau pathology, and neurodegeneration in AD. Furthermore, considering the current view of reactive astrogliosis heterogeneity-more than one subtype of astrocyte involved-in AD, we discuss how astrocytic body fluid biomarkers might fit into trajectories different from that of astrocytic PET imaging. Future research focusing on the development of innovative astrocytic PET radiotracers and fluid biomarkers may provide further insights into the heterogeneity of reactive astrogliosis and improve the detection of AD in its early stages.
Collapse
Affiliation(s)
- Igor C. Fontana
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Miriam Scarpa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Mona-Lisa Malarte
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Filipa M. Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
- Instituto de Ciência Biomédicas Abel Salazar da Universidade do Porto, 4050-313 Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, 4200-465 Porto, Portugal
| | - Sira Ausellé-Bosch
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
- Faculty of Health and Life Sciences, Pompeu Fabra University, 08003 Barcelona, Spain
| | - Marina Bluma
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Marco Bucci
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Konstantinos Chiotis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52 Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, 141 57 Stockholm, Sweden
| |
Collapse
|
14
|
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
|
15
|
Kumar A, Fontana IC, Nordberg A. Reactive astrogliosis: A friend or foe in the pathogenesis of Alzheimer's disease. J Neurochem 2023; 164:309-324. [PMID: 34931315 DOI: 10.1111/jnc.15565] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/28/2022]
Abstract
Astrocytes are highly efficient homeostatic glial cells playing a crucial role in optimal brain functioning and homeostasis. Astrocytes respond to changes in brain homoeostasis following central nervous system (CNS) injury/diseased state by a specific defence mechanism called reactive astrogliosis. Recent studies have implicated and placed reactive astrogliosis in the centre of pathophysiology of Alzheimer's disease (AD) and other neurodegenerative disorders. The AD biomarker field is evolving rapidly with new findings providing strong evidence which supports the notion that a reactive astrogliosis is an early event in the time course of AD progression which may precede other pathological hallmarks of AD. Clinical/translational in vivo PET and in vitro postmortem brain imaging studies demonstrated 'a first and second wave' of reactive astrogliosis in AD with distinct close-loop relationships with other pathological biomarkers at different stages of the disease. At the end stages, reactive astrocytes are found to be associated, or in proximity, with amyloid plaque and tau pathological deposits in postmortem AD brains. Several new PET-tracers, which are being in pipeline and validated at a very fast pace for mapping and visualising reactive astrogliosis in the brain, will provide further invaluable mechanistic insights into AD and other non-AD dementia pathologies. The complementary roles of microglia and astrocyte activation in AD progression, along with the clinical value of new fluid astrocytes biomarkers in the context of existing biomarkers, are the latest avenue that needs further exploration.
Collapse
Affiliation(s)
- Amit Kumar
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Igor C Fontana
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
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
|
17
|
Mohamed MA, Zeng Z, Gennaro M, Lao-Kaim NP, Myers JFM, Calsolaro V, Femminella GD, Tyacke RJ, Martin-Bastida A, Gunn RN, Nutt DJ, Edison P, Piccini P, Roussakis AA. Astrogliosis in aging and Parkinson’s disease dementia: a new clinical study with 11C-BU99008 PET. Brain Commun 2022; 4:fcac199. [PMID: 36072646 PMCID: PMC9445175 DOI: 10.1093/braincomms/fcac199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/20/2022] [Accepted: 08/16/2022] [Indexed: 11/14/2022] Open
Abstract
The role of astrogliosis in the pathology of brain aging and neurodegenerative diseases has recently drawn great attention. Imidazoline-2 binding sites represent a possible target to map the distribution of reactive astrocytes. In this study, we use 11C-BU99008, an imidazoline-2 binding sites-specific PET radioligand, to image reactive astrocytes in vivo in healthy controls and patients with established Parkinson’s disease dementia. Eighteen healthy controls (age: 45–78 years) and six patients with Parkinson’s disease dementia (age: 64–77 years) had one 11C-BU99008 PET-CT scan with arterial input function. All subjects underwent one 3 T MRI brain scan to facilitate the analysis of the PET data and to capture individual cerebral atrophy. Regional 11C-BU99008 volumes of distribution were calculated for each subject by the two-tissue compartmental modelling. Positive correlations between 11C-BU99008 volumes of distribution values and age were found for all tested regions across the brain within healthy controls (P < 0.05); furthermore, multiple regression indicated that aging affects 11C-BU99008 volumes of distribution values in a region-specific manner. Independent samples t-test indicated that there was no significant group difference in 11C-BU99008 volumes of distribution values between Parkinson’s disease dementia (n = 6; mean age = 71.97 ± 4.66 years) and older healthy controls (n = 9; mean age = 71.90 ± 5.51 years). Our data set shows that astrogliosis is common with aging in a region-specific manner. However, in this set-up, 11C-BU99008 PET cannot differentiate patients with Parkinson’s disease dementia from healthy controls of similar age.
Collapse
Affiliation(s)
- Mohamed A Mohamed
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Zhou Zeng
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
- Xiangya Hospital of Central South University , Changsha, Hunan , P.R. China
| | - Marta Gennaro
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Nicholas P Lao-Kaim
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Jim F M Myers
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Valeria Calsolaro
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Grazia Daniela Femminella
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
- Department of Translational Medical Sciences, University of Naples Federico II , Naples , Italy
| | - Robin J Tyacke
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Antonio Martin-Bastida
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
- Department of Neurology and Neurosciences, Clinica Universidad de Navarra , Pamplona-Madrid , Spain
| | - Roger N Gunn
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - David J Nutt
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | - Paola Piccini
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital , London , UK
| | | |
Collapse
|
18
|
Staging of Alzheimer's disease: past, present, and future perspectives. Trends Mol Med 2022; 28:726-741. [PMID: 35717526 DOI: 10.1016/j.molmed.2022.05.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 01/01/2023]
Abstract
For many years Alzheimer's disease (AD) was associated with the dementia stage of the disease, the tail end of a pathophysiological process that lasts approximately two decades. Whereas early disease staging assessments focused on progressive deterioration of clinical functioning, brain imaging with positron emission tomography (PET) and cerebrospinal fluid (CSF) biomarker studies highlighted the long preclinical phase of AD in which a cascade of detectable biological abnormalities precede cognitive decline. The recent proliferation of imaging and fluid biomarkers of AD pathophysiology provide an opportunity for the identification of several biological stages in the preclinical phase of AD. We discuss the use of clinical and biomarker information in past, present, and future staging of AD. We highlight potential applications of PET, CSF, and plasma biomarkers for staging AD severity in vivo.
Collapse
|
19
|
Bagán A, Morales-García JA, Griñán-Ferré C, Díaz C, Pérez del Palacio J, Ramos MC, Vicente F, Pérez B, Brea J, Loza MI, Pallàs M, Escolano C. Insights into the Pharmacokinetics and In Vitro Cell-Based Studies of the Imidazoline I 2 Receptor Ligand B06. Int J Mol Sci 2022; 23:ijms23105408. [PMID: 35628219 PMCID: PMC9141032 DOI: 10.3390/ijms23105408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
The impact of neurodegenerative diseases (ND) is becoming unbearable for humankind due to their vast prevalence and the lack of efficacious treatments. In this scenario, we focused on imidazoline I2 receptors (I2-IR) that are widely distributed in the brain and are altered in patients with brain disorders. We took the challenge of modulating I2-IR by developing structurally new molecules, in particular, a family of bicyclic α-iminophosphonates, endowed with high affinity and selectivity to these receptors. Treatment of two murine models, one for age-related cognitive decline and the other for Alzheimer's disease (AD), with representative compound B06 ameliorated their cognitive impairment and improved their behavioural condition. Furthermore, B06 revealed beneficial in vitro ADME-Tox properties. The pharmacokinetics (PK) and metabolic profile are reported to de-risk B06 for progressing in the preclinical development. To further characterize the pharmacological properties of B06, we assessed its neuroprotective properties and beneficial effect in an in vitro model of Parkinson's disease (PD). B06 rescued the human dopaminergic cell line SH-SY5Y from death after treatment with 6-hydroxydopamine (6-OHDA) and showed a crucial anti-inflammatory effect in a cellular model of neuroinflammation. This research reveals B06 as a putative candidate for advancing in the difficult path of drug discovery and supports the modulation of I2-IR as a fresh approach for the therapy of ND.
Collapse
Affiliation(s)
- Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain;
| | - José A. Morales-García
- The Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Department of Cell Biology, School of Medicine, Complutense University (UCM), 28040 Madrid, Spain;
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (C.G.-F.); (M.P.)
| | - Caridad Díaz
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - José Pérez del Palacio
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - Maria C. Ramos
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - Francisca Vicente
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Avda. del Conocimiento 34, 18016 Armilla, Spain; (C.D.); (J.P.d.P.); (M.C.R.); (F.V.)
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Autonomous University of Barcelona, 08193 Barcelona, Spain;
| | - José Brea
- Innopharma Screening Platform, BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15706 Santiago de Compostela, Spain; (J.B.); (M.I.L.)
| | - María Isabel Loza
- Innopharma Screening Platform, BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15706 Santiago de Compostela, Spain; (J.B.); (M.I.L.)
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, University of Barcelona, 08028 Barcelona, Spain; (C.G.-F.); (M.P.)
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain;
- Correspondence:
| |
Collapse
|
20
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
21
|
Garland EF, Hartnell IJ, Boche D. Microglia and Astrocyte Function and Communication: What Do We Know in Humans? Front Neurosci 2022; 16:824888. [PMID: 35250459 PMCID: PMC8888691 DOI: 10.3389/fnins.2022.824888] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia and astrocytes play essential roles in the central nervous system contributing to many functions including homeostasis, immune response, blood-brain barrier maintenance and synaptic support. Evidence has emerged from experimental models of glial communication that microglia and astrocytes influence and coordinate each other and their effects on the brain environment. However, due to the difference in glial cells between humans and rodents, it is essential to confirm the relevance of these findings in human brains. Here, we aim to review the current knowledge on microglia-astrocyte crosstalk in humans, exploring novel methodological techniques used in health and disease conditions. This will include an in-depth look at cell culture and iPSCs, post-mortem studies, imaging and fluid biomarkers, genetics and transcriptomic data. In this review, we will discuss the advantages and limitations of these methods, highlighting the understanding these methods have brought the field on these cells communicative abilities, and the knowledge gaps that remain.
Collapse
Affiliation(s)
| | | | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
22
|
Harada R, Furumoto S, Kudo Y, Yanai K, Villemagne VL, Okamura N. Imaging of Reactive Astrogliosis by Positron Emission Tomography. Front Neurosci 2022; 16:807435. [PMID: 35210989 PMCID: PMC8862631 DOI: 10.3389/fnins.2022.807435] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Many neurodegenerative diseases are neuropathologically characterized by neuronal loss, gliosis, and the deposition of misfolded proteins such as β-amyloid (Aβ) plaques and tau tangles in Alzheimer’s disease (AD). In postmortem AD brains, reactive astrocytes and activated microglia are observed surrounding Aβ plaques and tau tangles. These activated glial cells secrete pro-inflammatory cytokines and reactive oxygen species, which may contribute to neurodegeneration. Therefore, in vivo imaging of glial response by positron emission tomography (PET) combined with Aβ and tau PET would provide new insights to better understand the disease process, as well as aid in the differential diagnosis, and monitoring glial response disease-specific therapeutics. There are two promising targets proposed for imaging reactive astrogliosis: monoamine oxidase-B (MAO-B) and imidazoline2 binding site (I2BS), which are predominantly expressed in the mitochondrial membranes of astrocytes and are upregulated in various neurodegenerative conditions. PET tracers targeting these two MAO-B and I2BS have been evaluated in humans. [18F]THK-5351, which was originally designed to target tau aggregates in AD, showed high affinity for MAO-B and clearly visualized reactive astrocytes in progressive supranuclear palsy (PSP). However, the lack of selectivity of [18F]THK-5351 binding to both MAO-B and tau, severely limits its clinical utility as a biomarker. Recently, [18F]SMBT-1 was developed as a selective and reversible MAO-B PET tracer via compound optimization of [18F]THK-5351. In this review, we summarize the strategy underlying molecular imaging of reactive astrogliosis and clinical studies using MAO-B and I2BS PET tracers.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
- *Correspondence: Ryuichi Harada,
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Department of New Therapeutics Innovation for Alzheimer’s and Dementia, Institute of Development and Aging, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Victor L. Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Nobuyuki Okamura,
| |
Collapse
|
23
|
Harada R, Shimizu Y, Du Y, Ishikawa Y, Iwata R, Kudo Y, Yanai K, Okamura N, Furumoto S. The Role of Chirality of [ 18F]SMBT-1 in Imaging of Monoamine Oxidase-B. ACS Chem Neurosci 2022; 13:322-329. [PMID: 35049267 DOI: 10.1021/acschemneuro.1c00655] [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: 02/02/2023] Open
Abstract
(S)-(2-Methylpyrid-5-yl)-6-[(3-[18F]fluoro-2-hydroxy)propoxy]quinoline ([18F]SMBT-1) was recently developed as a novel class of selective and reversible monoamine oxidase-B (MAO-B) tracers for in vivo imaging of reactive astrogliosis via positron emission tomography. To investigate the effect of the chirality of [18F]SMBT-1 on tracer performance, we synthesized (S)-[18F]6 ([18F]SMBT-1) and (R)-[18F]6 and compared their binding properties, pharmacokinetics, and metabolism. (S)-6 showed higher binding affinity to MAO-B and lower binding affinity to MAO-A than (R)-6, demonstrating a higher selectivity ratio (MAO-B/MAO-A). A pharmacokinetic study in mice demonstrated that both (S)-[18F]6 and (R)-[18F]6 showed sufficient initial brain uptake. However, (S)-[18F]6 was cleared significantly faster from the body. An abundant sulfoconjugate metabolite M2 was observed in plasma for (S)-[18F]6 but not for (R)-[18F]6. In vitro sulfation assays confirmed that (S)-6 was more reactive than (R)-6, consistent with the in vivo findings. Mefenamic acid, a selective sulfotransferase 1A1 (SULT1A1) inhibitor, strongly inhibited the in vitro sulfation of (S)-6 by mouse liver fractions, human liver cytosol fractions, and human recombinant SULT1A1 enzyme. Genetic polymorphisms of SULT1A1 did not affect the sulfation of (S)-6 in vitro. In conclusion, (S)-[18F]6 had a more favorable binding affinity and binding selectivity for MAO-B than (R)-[18F]6. Additionally, (S)-[18F]6 also possessed better pharmacological and metabolic properties than (R)-[18F]6. These results suggest that (S)-[18F]6 ([18F]SMBT-1) is a promising candidate for application in the imaging of MAO-B in vivo.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Graduate School of Medicine, Tohoku University, 2-1 Seiryomachi, Aobaku, Sendai 980-8575, Japan
- Department of New Therapeutics Innovation for Alzheimer’s and Dementia, Institute of Development Aging and Cancer (IDAC), Tohoku University, 2-1 Seiryomachi, Aobaku, Sendai 980-8575, Japan
| | - Yuki Shimizu
- Cyclotron and Radioisotope Center (CYRIC), Tohoku University, 6-3 Aoba, Aramaki, Aobaku, Sendai 980-8578, Japan
| | - Yiqing Du
- Department of Pharmacology, Graduate School of Medicine, Tohoku University, 2-1 Seiryomachi, Aobaku, Sendai 980-8575, Japan
| | - Yoichi Ishikawa
- Cyclotron and Radioisotope Center (CYRIC), Tohoku University, 6-3 Aoba, Aramaki, Aobaku, Sendai 980-8578, Japan
| | - Ren Iwata
- Cyclotron and Radioisotope Center (CYRIC), Tohoku University, 6-3 Aoba, Aramaki, Aobaku, Sendai 980-8578, Japan
| | - Yukitsuka Kudo
- Department of New Therapeutics Innovation for Alzheimer’s and Dementia, Institute of Development Aging and Cancer (IDAC), Tohoku University, 2-1 Seiryomachi, Aobaku, Sendai 980-8575, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Graduate School of Medicine, Tohoku University, 2-1 Seiryomachi, Aobaku, Sendai 980-8575, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyaginoku, Sendai 983-8536, Japan
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center (CYRIC), Tohoku University, 6-3 Aoba, Aramaki, Aobaku, Sendai 980-8578, Japan
| |
Collapse
|
24
|
Relationship between astrocyte reactivity, using novel 11C-BU99008 PET, and glucose metabolism, grey matter volume and amyloid load in cognitively impaired individuals. Mol Psychiatry 2022; 27:2019-2029. [PMID: 35125495 PMCID: PMC9126819 DOI: 10.1038/s41380-021-01429-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/10/2021] [Accepted: 12/23/2021] [Indexed: 12/01/2022]
Abstract
Post mortem neuropathology suggests that astrocyte reactivity may play a significant role in neurodegeneration in Alzheimer's disease. We explored this in vivo using multimodal PET and MRI imaging. Twenty subjects (11 older, cognitively impaired patients and 9 age-matched healthy controls) underwent brain scanning using the novel reactive astrocyte PET tracer 11C-BU99008, 18F-FDG and 18F-florbetaben PET, and T1-weighted MRI. Differences between cognitively impaired patients and healthy controls in regional and voxel-wise levels of astrocyte reactivity, glucose metabolism, grey matter volume and amyloid load were explored, and their relationship to each other was assessed using Biological Parametric Mapping (BPM). Amyloid beta (Aβ)-positive patients showed greater 11C-BU99008 uptake compared to controls, except in the temporal lobe, whilst further increased 11C-BU99008 uptake was observed in Mild Cognitive Impairment subjects compared to those with Alzheimer's disease in the frontal, temporal and cingulate cortices. BPM correlations revealed that regions which showed reduced 11C-BU99008 uptake in Aβ-positive patients compared to controls, such as the temporal lobe, also showed reduced 18F-FDG uptake and grey matter volume, although the correlations with 18F-FDG uptake were not replicated in the ROI analysis. BPM analysis also revealed a regionally-dynamic relationship between astrocyte reactivity and amyloid uptake: increased amyloid load in cortical association areas of the temporal lobe and cingulate cortices was associated with reduced 11C-BU99008 uptake, whilst increased amyloid uptake in primary motor and sensory areas (in which amyloid deposition occurs later) was associated with increased 11C-BU99008 uptake. These novel observations add to the hypothesis that while astrocyte reactivity may be triggered by early Aβ-deposition, sustained pro-inflammatory astrocyte reactivity with greater amyloid deposition may lead to astrocyte dystrophy and amyloid-associated neuropathology such as grey matter atrophy and glucose hypometabolism, although the evidence for glucose hypometabolism here is less strong.
Collapse
|
25
|
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
|
26
|
Martins D, Giacomel A, Williams SCR, Turkheimer F, Dipasquale O, Veronese M. Imaging transcriptomics: Convergent cellular, transcriptomic, and molecular neuroimaging signatures in the healthy adult human brain. Cell Rep 2021; 37:110173. [PMID: 34965413 DOI: 10.1016/j.celrep.2021.110173] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/30/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
The integration of transcriptomic and neuroimaging data, "imaging transcriptomics," has recently emerged to generate hypotheses about potential biological pathways underlying regional variability in neuroimaging features. However, the validity of this approach is yet to be examined in depth. Here, we sought to bridge this gap by performing transcriptomic decoding of the regional distribution of well-known molecular markers spanning different elements of the biology of the healthy human brain. Imaging transcriptomics identifies biological and cell pathways that are consistent with the known biology of a wide range of molecular neuroimaging markers. The extent to which it can capture patterns of gene expression that align well with elements of the biology of the neuroinflammatory axis, at least in healthy controls without a proinflammatory challenge, is inconclusive. Imaging transcriptomics might constitute an interesting approach to improve our understanding of the biological pathways underlying regional variability in a wide range of neuroimaging phenotypes.
Collapse
Affiliation(s)
- Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK.
| | - Alessio Giacomel
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Steven C R Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK; Department of Information Engineering, University of Padua, Via Gradenigo, 6/b, 35131 Padova, Italy.
| | | |
Collapse
|
27
|
Liu Y, Jiang H, Qin X, Tian M, Zhang H. PET imaging of reactive astrocytes in neurological disorders. Eur J Nucl Med Mol Imaging 2021; 49:1275-1287. [PMID: 34873637 PMCID: PMC8921128 DOI: 10.1007/s00259-021-05640-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/24/2021] [Indexed: 12/17/2022]
Abstract
The reactive astrocytes manifest molecular, structural, and functional remodeling in injury, infection, or diseases of the CNS, which play a critical role in the pathological mechanism of neurological diseases. A growing need exists for dependable approach to better characterize the activation of astrocyte in vivo. As an advanced molecular imaging technology, positron emission tomography (PET) has the potential for visualizing biological activities at the cellular levels. In the review, we summarized the PET visualization strategies for reactive astrocytes and discussed the applications of astrocyte PET imaging in neurological diseases. Future studies are needed to pay more attention to the development of specific imaging agents for astrocytes and further improve our exploration of reactive astrocytes in various diseases.
Collapse
Affiliation(s)
- Yu Liu
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Han Jiang
- PET-CT Center, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xiyi Qin
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China.
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China. .,Institute of Nuclear Medicine and Molecular Imaging of Zhejiang University, Hangzhou, China. .,Key Laboratory of Medical Molecular Imaging of Zhejiang Province, Hangzhou, China. .,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China. .,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, China.
| |
Collapse
|
28
|
Positron emission tomography in multiple sclerosis - straight to the target. Nat Rev Neurol 2021; 17:663-675. [PMID: 34545219 DOI: 10.1038/s41582-021-00537-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Following the impressive progress in the treatment of relapsing-remitting multiple sclerosis (MS), the major challenge ahead is the development of treatments to prevent or delay the irreversible accumulation of clinical disability in progressive forms of the disease. The substrate of clinical progression is neuro-axonal degeneration, and a deep understanding of the mechanisms that underlie this process is a precondition for the development of therapies for progressive MS. PET imaging involves the use of radiolabelled compounds that bind to specific cellular and metabolic targets, thereby enabling direct in vivo measurement of several pathological processes. This approach can provide key insights into the clinical relevance of these processes and their chronological sequence during the disease course. In this Review, we focus on the contribution that PET is making to our understanding of extraneuronal and intraneuronal mechanisms that are involved in the pathogenesis of irreversible neuro-axonal damage in MS. We consider the major challenges with the use of PET in MS and the steps necessary to realize clinical benefits of the technique. In addition, we discuss the potential of emerging PET tracers and future applications of existing compounds to facilitate the identification of effective neuroprotective treatments for patients with MS.
Collapse
|
29
|
Calsolaro V, Matthews PM, Donat CK, Livingston NR, Femminella GD, Guedes SS, Myers J, Fan Z, Tyacke RJ, Venkataraman AV, Perneczky R, Gunn R, Rabiner EA, Gentleman S, Parker CA, Murphy PS, Wren PB, Hinz R, Sastre M, Nutt DJ, Edison P. Astrocyte reactivity with late-onset cognitive impairment assessed in vivo using 11C-BU99008 PET and its relationship with amyloid load. Mol Psychiatry 2021; 26:5848-5855. [PMID: 34267329 PMCID: PMC8758500 DOI: 10.1038/s41380-021-01193-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 05/16/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
11C-BU99008 is a novel positron emission tomography (PET) tracer that enables selective imaging of astrocyte reactivity in vivo. To explore astrocyte reactivity associated with Alzheimer's disease, 11 older, cognitively impaired (CI) subjects and 9 age-matched healthy controls (HC) underwent 3T magnetic resonance imaging (MRI), 18F-florbetaben and 11C-BU99008 PET. The 8 amyloid (Aβ)-positive CI subjects had higher 11C-BU99008 uptake relative to HC across the whole brain, but particularly in frontal, temporal, medial temporal and occipital lobes. Biological parametric mapping demonstrated a positive voxel-wise neuroanatomical correlation between 11C-BU99008 and 18F-florbetaben. Autoradiography using 3H-BU99008 with post-mortem Alzheimer's brains confirmed through visual assessment that increased 3H-BU99008 binding localised with the astrocyte protein glial fibrillary acid protein and was not displaced by PiB or florbetaben. This proof-of-concept study provides direct evidence that 11C-BU99008 can measure in vivo astrocyte reactivity in people with late-life cognitive impairment and Alzheimer's disease. Our results confirm that increased astrocyte reactivity is found particularly in cortical regions with high Aβ load. Future studies now can explore how clinical expression of disease varies with astrocyte reactivity.
Collapse
Affiliation(s)
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Cornelius K Donat
- Department of Brain Sciences, Imperial College London, London, UK
- Centre for Blast Injury Studies, Imperial College London, London, UK
| | | | | | | | - Jim Myers
- Department of Brain Sciences, Imperial College London, London, UK
| | - Zhen Fan
- Department of Brain Sciences, Imperial College London, London, UK
| | - Robin J Tyacke
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- German Centre for Neurodegenerative Disorders (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Roger Gunn
- Department of Brain Sciences, Imperial College London, London, UK
- Invicro, London, UK
| | | | - Steve Gentleman
- Department of Brain Sciences, Imperial College London, London, UK
| | - Christine A Parker
- Department of Brain Sciences, Imperial College London, London, UK
- GlaxoSmithKline, Stevenage, UK
| | | | | | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, London, UK
| | - David J Nutt
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, London, UK.
- Cardiff University, Cardiff, Wales, United Kingdom.
| |
Collapse
|
30
|
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
|
31
|
Benzofuranyl-2-imidazoles as imidazoline I 2 receptor ligands for Alzheimer's disease. Eur J Med Chem 2021; 222:113540. [PMID: 34118720 DOI: 10.1016/j.ejmech.2021.113540] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 12/29/2022]
Abstract
Recent findings unveil the pharmacological modulation of imidazoline I2 receptors (I2-IR) as a novel strategy to face unmet medical neurodegenerative diseases. In this work, we report the chemical characterization, three-dimensional quantitative structure-activity relationship (3D-QSAR) and ADMET in silico of a family of benzofuranyl-2-imidazoles that exhibit affinity against human brain I2-IR and most of them have been predicted to be brain permeable. Acute treatment in mice with 2-(2-benzofuranyl)-2-imidazole, known as LSL60101 (garsevil), showed non-warning properties in the ADMET studies and an optimal pharmacokinetic profile. Moreover, LSL60101 induced hypothermia in mice while decreased pro-apoptotic FADD protein in the hippocampus. In vivo studies in the familial Alzheimer's disease 5xFAD murine model with the representative compound, revealed significant decreases in the protein expression levels of antioxidant enzymes superoxide dismutase and glutathione peroxidase in hippocampus. Overall, LSL60101 plays a neuroprotective role by reducing apoptosis and modulating oxidative stress.
Collapse
|
32
|
Is Chelation Therapy a Potential Treatment for Parkinson's Disease? Int J Mol Sci 2021; 22:ijms22073338. [PMID: 33805195 PMCID: PMC8036775 DOI: 10.3390/ijms22073338] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/25/2021] [Accepted: 03/06/2021] [Indexed: 12/14/2022] Open
Abstract
Iron loading in some brain regions occurs in Parkinson’s Disease (PD), and it has been considered that its removal by iron chelators could be an appropriate therapeutic approach. Since neuroinflammation with microgliosis is also a common feature of PD, it is possible that iron is sequestered within cells as a result of the “anaemia of chronic disease” and remains unavailable to the chelator. In this review, the extent of neuroinflammation in PD is discussed together with the role played by glia cells, specifically microglia and astrocytes, in controlling iron metabolism during inflammation, together with the results of MRI studies. The current use of chelators in clinical medicine is presented together with a discussion of two clinical trials of PD patients where an iron chelator was administered and showed encouraging results. It is proposed that the use of anti-inflammatory drugs combined with an iron chelator might be a better approach to increase chelator efficacy.
Collapse
|
33
|
Goud NS, Bhattacharya A, Joshi RK, Nagaraj C, Bharath RD, Kumar P. Carbon-11: Radiochemistry and Target-Based PET Molecular Imaging Applications in Oncology, Cardiology, and Neurology. J Med Chem 2021; 64:1223-1259. [PMID: 33499603 DOI: 10.1021/acs.jmedchem.0c01053] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The positron emission tomography (PET) molecular imaging technique has gained its universal value as a remarkable tool for medical diagnosis and biomedical research. Carbon-11 is one of the promising radiotracers that can report target-specific information related to its pharmacology and physiology to understand the disease status. Currently, many of the available carbon-11 (t1/2 = 20.4 min) PET radiotracers are heterocyclic derivatives that have been synthesized using carbon-11 inserted different functional groups obtained from primary and secondary carbon-11 precursors. A spectrum of carbon-11 PET radiotracers has been developed against many of the upregulated and emerging targets for the diagnosis, prognosis, prediction, and therapy in the fields of oncology, cardiology, and neurology. This review focuses on the carbon-11 radiochemistry and various target-specific PET molecular imaging agents used in tumor, heart, brain, and neuroinflammatory disease imaging along with its associated pathology.
Collapse
Affiliation(s)
- Nerella Sridhar Goud
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Ahana Bhattacharya
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Raman Kumar Joshi
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Chandana Nagaraj
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Rose Dawn Bharath
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Pardeep Kumar
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| |
Collapse
|
34
|
Sarkar S, Biswas SC. Astrocyte subtype-specific approach to Alzheimer's disease treatment. Neurochem Int 2021; 145:104956. [PMID: 33503465 DOI: 10.1016/j.neuint.2021.104956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 01/01/2021] [Accepted: 01/05/2021] [Indexed: 01/08/2023]
Abstract
Astrocytes respond to any pathological condition in the central nervous system (CNS) including Alzheimer's disease (AD), and this response is called astrocyte reactivity. Astrocyte reaction to a CNS insult is a highly heterogeneous phenomenon in which the astrocytes undergo a set of morphological, molecular and functional changes with a characteristic secretome profile. Such astrocytes are termed as 'reactive astrocytes'. Controversies regarding the reactive astrocytes abound. Recently, a continuum of reactive astrocyte profiles with distinct transcriptional states has been identified. Among them, disease-associated astrocytes (DAA) were uniquely present in AD mice and expressed a signature set of genes implicated in complement cascade, endocytosis and aging. Earlier, two stimulus-specific reactive astrocyte subtypes with their unique transcriptomic signatures were identified using mouse models of neuroinflammation and ischemia and termed as A1 astrocytes (detrimental) and A2 astrocytes (beneficial) respectively. Interestingly, although most of the A1 signature genes were also detected in DAA, as opposed to A2 astrocyte signatures, some of the A1 specific genes were expressed in other astrocyte subtypes, indicating that these nomenclature-based signatures are not very specific. In this review, we elaborate the disparate functions and cytokine profiles of reactive astrocyte subtypes in AD and tried to distinguish them by designating neurotoxic astrocytes as A1-like and neuroprotective ones as A2-like without directly referring to the A1/A2 original nomenclature. We have also focused on the dual nature from a functional perspective of some cytokines depending on AD-stage, highlighting a number of them as major candidates in AD therapy. Therefore, we suggest that promoting subtype-specific beneficial roles, inhibiting subtype-specific detrimental roles or targeting subtype-specific cytokines constitute a novel therapeutic approach to AD treatment.
Collapse
Affiliation(s)
- Sukanya Sarkar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700 032, India
| | - Subhas C Biswas
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700 032, India.
| |
Collapse
|
35
|
Astroglial tracer BU99008 detects multiple binding sites in Alzheimer's disease brain. Mol Psychiatry 2021; 26:5833-5847. [PMID: 33888872 PMCID: PMC8758481 DOI: 10.1038/s41380-021-01101-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/17/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022]
Abstract
With reactive astrogliosis being established as one of the hallmarks of Alzheimer's disease (AD), there is high interest in developing novel positron emission tomography (PET) tracers to detect early astrocyte reactivity. BU99008, a novel astrocytic PET ligand targeting imidazoline-2 binding sites (I2BS) on astrocytes, might be a suitable candidate. Here we demonstrate for the first time that BU99008 could visualise reactive astrogliosis in postmortem AD brains and propose a multiple binding site [Super-high-affinity (SH), High-affinity (HA) and Low-affinity (LA)] model for BU99008, I2BS specific ligands (2-BFI and BU224) and deprenyl in AD and control (CN) brains. The proportion (%) and affinities of these sites varied significantly between the BU99008, 2-BFI, BU224 and deprenyl in AD and CN brains. Regional binding studies demonstrated significantly higher 3H-BU99008 binding in AD brain regions compared to CN. Comparative autoradiography studies reinforced these findings, showing higher specific binding for 3H-BU99008 than 3H-Deprenyl in sporadic AD brain compared to CN, implying that they might have different targets. The data clearly shows that BU99008 could detect I2BS expressing reactive astrocytes with good selectivity and specificity and hence be a potential attractive clinical astrocytic PET tracer for gaining further insight into the role of reactive astrogliosis in AD.
Collapse
|
36
|
Novel PET Biomarkers to Disentangle Molecular Pathways across Age-Related Neurodegenerative Diseases. Cells 2020; 9:cells9122581. [PMID: 33276490 PMCID: PMC7761606 DOI: 10.3390/cells9122581] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
There is a need to disentangle the etiological puzzle of age-related neurodegenerative diseases, whose clinical phenotypes arise from known, and as yet unknown, pathways that can act distinctly or in concert. Enhanced sub-phenotyping and the identification of in vivo biomarker-driven signature profiles could improve the stratification of patients into clinical trials and, potentially, help to drive the treatment landscape towards the precision medicine paradigm. The rapidly growing field of neuroimaging offers valuable tools to investigate disease pathophysiology and molecular pathways in humans, with the potential to capture the whole disease course starting from preclinical stages. Positron emission tomography (PET) combines the advantages of a versatile imaging technique with the ability to quantify, to nanomolar sensitivity, molecular targets in vivo. This review will discuss current research and available imaging biomarkers evaluating dysregulation of the main molecular pathways across age-related neurodegenerative diseases. The molecular pathways focused on in this review involve mitochondrial dysfunction and energy dysregulation; neuroinflammation; protein misfolding; aggregation and the concepts of pathobiology, synaptic dysfunction, neurotransmitter dysregulation and dysfunction of the glymphatic system. The use of PET imaging to dissect these molecular pathways and the potential to aid sub-phenotyping will be discussed, with a focus on novel PET biomarkers.
Collapse
|
37
|
Vasilopoulou F, Griñán-Ferré C, Rodríguez-Arévalo S, Bagán A, Abás S, Escolano C, Pallàs M. I 2 imidazoline receptor modulation protects aged SAMP8 mice against cognitive decline by suppressing the calcineurin pathway. GeroScience 2020; 43:965-983. [PMID: 33128688 PMCID: PMC8110656 DOI: 10.1007/s11357-020-00281-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 09/28/2020] [Indexed: 12/26/2022] Open
Abstract
Brain aging and dementia are current problems that must be solved. The levels of imidazoline 2 receptors (I2-IRs) are increased in the brain in Alzheimer's disease (AD) and other neurodegenerative diseases. We tested the action of the specific and selective I2-IR ligand B06 in a mouse model of accelerated aging and AD, the senescence-accelerated mouse prone 8 (SAMP8) model. Oral administration of B06 for 4 weeks improved SAMP8 mouse behavior and cognition and reduced AD hallmarks, oxidative stress, and apoptotic and neuroinflammation markers. Likewise, B06 regulated glial excitatory amino acid transporter 2 and N-methyl-D aspartate 2A and 2B receptor subunit protein levels. Calcineurin (CaN) is a phosphatase that controls the phosphorylation levels of cAMP response element-binding (CREB), apoptotic mediator BCL-2-associated agonist of cell death (BAD) and GSK3β, among other molecules. Interestingly, B06 was able to reduce the levels of the CaN active form (CaN A). Likewise, CREB phosphorylation, BAD gene expression, and other factors were modified after B06 treatment. Moreover, phosphorylation of a target of CaN, nuclear factor of activated T-cells, cytoplasmic 1 (NFATC1), was increased in B06-treated mice, impeding the transcription of genes related to neuroinflammation and neural plasticity. In summary, this I2 imidazoline ligand can exert its beneficial effects on age-related conditions by modulating CaN pathway action and affecting several molecular pathways, playing a neuroprotective role in SAMP8 mice.
Collapse
Affiliation(s)
- Foteini Vasilopoulou
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Sergio Rodríguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Sònia Abás
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Neurociencies, University of Barcelona, Av. Joan XXIII, 27-31, E-08028, Barcelona, Spain.
| |
Collapse
|
38
|
Bousquet P, Hudson A, García-Sevilla JA, Li JX. Imidazoline Receptor System: The Past, the Present, and the Future. Pharmacol Rev 2020; 72:50-79. [PMID: 31819014 DOI: 10.1124/pr.118.016311] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Imidazoline receptors historically referred to a family of nonadrenergic binding sites that recognize compounds with an imidazoline moiety, although this has proven to be an oversimplification. For example, none of the proposed endogenous ligands for imidazoline receptors contain an imidazoline moiety but they are diverse in their chemical structure. Three receptor subtypes (I1, I2, and I3) have been proposed and the understanding of each has seen differing progress over the decades. I1 receptors partially mediate the central hypotensive effects of clonidine-like drugs. Moxonidine and rilmenidine have better therapeutic profiles (fewer side effects) than clonidine as antihypertensive drugs, thought to be due to their higher I1/α 2-adrenoceptor selectivity. Newer I1 receptor agonists such as LNP599 [3-chloro-2-methyl-phenyl)-(4-methyl-4,5-dihydro-3H-pyrrol-2-yl)-amine hydrochloride] have little to no activity on α 2-adrenoceptors and demonstrate promising therapeutic potential for hypertension and metabolic syndrome. I2 receptors associate with several distinct proteins, but the identities of these proteins remain elusive. I2 receptor agonists have demonstrated various centrally mediated effects including antinociception and neuroprotection. A new I2 receptor agonist, CR4056 [2-phenyl-6-(1H-imidazol-1yl) quinazoline], demonstrated clear analgesic activity in a recently completed phase II clinical trial and holds great promise as a novel I2 receptor-based first-in-class nonopioid analgesic. The understanding of I3 receptors is relatively limited. Existing data suggest that I3 receptors may represent a binding site at the Kir6.2-subtype ATP-sensitive potassium channels in pancreatic β-cells and may be involved in insulin secretion. Despite the elusive nature of their molecular identities, recent progress on drug discovery targeting imidazoline receptors (I1 and I2) demonstrates the exciting potential of these compounds to elicit neuroprotection and to treat various disorders such as hypertension, metabolic syndrome, and chronic pain.
Collapse
Affiliation(s)
- Pascal Bousquet
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| | - Alan Hudson
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| | - Jesús A García-Sevilla
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| | - Jun-Xu Li
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| |
Collapse
|
39
|
Zeng Z, Roussakis AA, Lao-Kaim NP, Piccini P. Astrocytes in Parkinson's disease: from preclinical assays to in vivo imaging and therapeutic probes. Neurobiol Aging 2020; 95:264-270. [PMID: 32905922 DOI: 10.1016/j.neurobiolaging.2020.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/30/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is increasingly thought to be associated with glial pathology. Recently, research in neurodegenerative disorders has applied a greater focus to better understanding the role of astrocytes in the disease pathophysiology. In this article, we review results from the latest preclinical and clinical work, including functional imaging studies on astrocytes in PD and highlight key molecules that may prove valuable as biomarkers. We discuss how astrocytes may contribute to the initiation and progression of PD. We additionally present trials of investigational medicinal products and the current background for the design of future clinical trials.
Collapse
Affiliation(s)
- Zhou Zeng
- Department of Brain Sciences, Imperial College London, Neurology Imaging Unit, London, UK; Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | | - Nicholas P Lao-Kaim
- Department of Brain Sciences, Imperial College London, Neurology Imaging Unit, London, UK
| | - Paola Piccini
- Department of Brain Sciences, Imperial College London, Neurology Imaging Unit, London, UK.
| |
Collapse
|
40
|
Belloli S, Morari M, Murtaj V, Valtorta S, Moresco RM, Gilardi MC. Translation Imaging in Parkinson's Disease: Focus on Neuroinflammation. Front Aging Neurosci 2020; 12:152. [PMID: 32581765 PMCID: PMC7289967 DOI: 10.3389/fnagi.2020.00152] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the appearance of α-synuclein insoluble aggregates known as Lewy bodies. Neurodegeneration is accompanied by neuroinflammation mediated by cytokines and chemokines produced by the activated microglia. Several studies demonstrated that such an inflammatory process is an early event, and contributes to oxidative stress and mitochondrial dysfunctions. α-synuclein fibrillization and aggregation activate microglia and contribute to disease onset and progression. Mutations in different genes exacerbate the inflammatory phenotype in the monogenic compared to sporadic forms of PD. Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) with selected radiopharmaceuticals allow in vivo imaging of molecular modifications in the brain of living subjects. Several publications showed a reduction of dopaminergic terminals and dopamine (DA) content in the basal ganglia, starting from the early stages of the disease. Moreover, non-dopaminergic neuronal pathways are also affected, as shown by in vivo studies with serotonergic and glutamatergic radiotracers. The role played by the immune system during illness progression could be investigated with PET ligands that target the microglia/macrophage Translocator protein (TSPO) receptor. These agents have been used in PD patients and rodent models, although often without attempting correlations with other molecular or functional parameters. For example, neurodegeneration and brain plasticity can be monitored using the metabolic marker 2-Deoxy-2-[18F]fluoroglucose ([18F]-FDG), while oxidative stress can be probed using the copper-labeled diacetyl-bis(N-methyl-thiosemicarbazone) ([Cu]-ATSM) radioligand, whose striatal-specific binding ratio in PD patients seems to correlate with a disease rating scale and motor scores. Also, structural and functional modifications during disease progression may be evaluated by Magnetic Resonance Imaging (MRI), using different parameters as iron content or cerebral volume. In this review article, we propose an overview of in vivo clinical and non-clinical imaging research on neuroinflammation as an emerging marker of early PD. We also discuss how multimodal-imaging approaches could provide more insights into the role of the inflammatory process and related events in PD development.
Collapse
Affiliation(s)
- Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Michele Morari
- Section of Pharmacology, Department of Medical Sciences, National Institute for Neuroscience, University of Ferrara, Ferrara, Italy
| | - Valentina Murtaj
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Maria Carla Gilardi
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
41
|
Abás S, Rodríguez-Arévalo S, Bagán A, Griñán-Ferré C, Vasilopoulou F, Brocos-Mosquera I, Muguruza C, Pérez B, Molins E, Luque FJ, Pérez-Lozano P, de Jonghe S, Daelemans D, Naesens L, Brea J, Loza MI, Hernández-Hernández E, García-Sevilla JA, García-Fuster MJ, Radan M, Djikic T, Nikolic K, Pallàs M, Callado LF, Escolano C. Bicyclic α-Iminophosphonates as High Affinity Imidazoline I2 Receptor Ligands for Alzheimer’s Disease. J Med Chem 2020; 63:3610-3633. [DOI: 10.1021/acs.jmedchem.9b02080] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sònia Abás
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Sergio Rodríguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Andrea Bagán
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Christian Griñán-Ferré
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Foteini Vasilopoulou
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Iria Brocos-Mosquera
- Department of Pharmacology, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Carolina Muguruza
- Department of Pharmacology, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Belén Pérez
- Department of Pharmacology, Therapeutic and Toxicology, Autonomous University of Barcelona, E-08193 Barcelona, Spain
| | - Elies Molins
- Institut de Ciència de Materials de Barcelona (CSIC), Campus UAB, E-08193 Cerdanyola, Spain
| | - F. Javier Luque
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona, E-08921 Santa Coloma de Gramanet, Spain
| | - Pilar Pérez-Lozano
- Unit of Pharmaceutical Technology, Pharmacy and Pharmaceutical Technology, and Physical Chemistry Department, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Steven de Jonghe
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Dirk Daelemans
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Lieve Naesens
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - José Brea
- Innopharma screening platform, BioFarma research group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - M. Isabel Loza
- Innopharma screening platform, BioFarma research group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Elena Hernández-Hernández
- IUNICS University of the Balearic Islands (UIB), and Health Research Institute of the Balearic Islands (IdISBa), Cra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Jesús A. García-Sevilla
- IUNICS University of the Balearic Islands (UIB), and Health Research Institute of the Balearic Islands (IdISBa), Cra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - M. Julia García-Fuster
- IUNICS University of the Balearic Islands (UIB), and Health Research Institute of the Balearic Islands (IdISBa), Cra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Milica Radan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Teodora Djikic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Mercè Pallàs
- Pharmacology Section, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institut de Neurociències, University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| | - Luis F. Callado
- Department of Pharmacology, and Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, University of the Basque Country, UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, E-08028 Barcelona, Spain
| |
Collapse
|
42
|
McCluskey SP, Plisson C, Rabiner EA, Howes O. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. Eur J Nucl Med Mol Imaging 2020; 47:451-489. [PMID: 31541283 PMCID: PMC6974496 DOI: 10.1007/s00259-019-04488-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE A limit on developing new treatments for a number of central nervous system (CNS) disorders has been the inadequate understanding of the in vivo pathophysiology underlying neurological and psychiatric disorders and the lack of in vivo tools to determine brain penetrance, target engagement, and relevant molecular activity of novel drugs. Molecular neuroimaging provides the tools to address this. This article aims to provide a state-of-the-art review of new PET tracers for CNS targets, focusing on developments in the last 5 years for targets recently available for in-human imaging. METHODS We provide an overview of the criteria used to evaluate PET tracers. We then used the National Institute of Mental Health Research Priorities list to identify the key CNS targets. We conducted a PubMed search (search period 1st of January 2013 to 31st of December 2018), which yielded 40 new PET tracers across 16 CNS targets which met our selectivity criteria. For each tracer, we summarised the evidence of its properties and potential for use in studies of CNS pathophysiology and drug evaluation, including its target selectivity and affinity, inter and intra-subject variability, and pharmacokinetic parameters. We also consider its potential limitations and missing characterisation data, but not specific applications in drug development. Where multiple tracers were present for a target, we provide a comparison of their properties. RESULTS AND CONCLUSIONS Our review shows that multiple new tracers have been developed for proteinopathy targets, particularly tau, as well as the purinoceptor P2X7, phosphodiesterase enzyme PDE10A, and synaptic vesicle glycoprotein 2A (SV2A), amongst others. Some of the most promising of these include 18F-MK-6240 for tau imaging, 11C-UCB-J for imaging SV2A, 11C-CURB and 11C-MK-3168 for characterisation of fatty acid amide hydrolase, 18F-FIMX for metabotropic glutamate receptor 1, and 18F-MNI-444 for imaging adenosine 2A. Our review also identifies recurrent issues within the field. Many of the tracers discussed lack in vivo blocking data, reducing confidence in selectivity. Additionally, late-stage identification of substantial off-target sites for multiple tracers highlights incomplete pre-clinical characterisation prior to translation, as well as human disease state studies carried out without confirmation of test-retest reproducibility.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| | - Christophe Plisson
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
43
|
Wilson H, Dervenoulas G, Pagano G, Tyacke RJ, Polychronis S, Myers J, Gunn RN, Rabiner EA, Nutt D, Politis M. Imidazoline 2 binding sites reflecting astroglia pathology in Parkinson's disease: an in vivo11C-BU99008 PET study. Brain 2019; 142:3116-3128. [PMID: 31504212 DOI: 10.1093/brain/awz260] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/06/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Astroglia are multifunctional cells that regulate neuroinflammation and maintain homeostasis within the brain. Astroglial α-synuclein-positive cytoplasmic accumulations have been shown post-mortem in patients with Parkinson's disease and therefore astroglia may play an important role in the initiation and progression of Parkinson's disease. Imidazoline 2 binding sites are expressed on activated astroglia in the cortex, hippocampus, basal ganglia and brainstem; therefore, by measuring imidazoline 2 binding site levels we can indirectly evaluate astrogliosis in patients with Parkinson's disease. Here, we aimed to evaluate the role of astroglia activation in vivo in patients with Parkinson's disease using 11C-BU99008 PET, a novel radioligand with high specificity and selectivity for imidazoline 2 binding sites. Twenty-two patients with Parkinson's disease and 14 healthy control subjects underwent 3 T MRI and a 120-min 11C-BU99008 PET scan with volume of distribution (VT) estimated using a two-tissue compartmental model with a metabolite corrected arterial plasma input function. Parkinson's disease patients were stratified into early (n = 8) and moderate/advanced (n = 14) groups according to disease stage. In early Parkinson's disease, increased 11C-BU99008 VT uptake was observed in frontal (P = 0.022), temporal (P = 0.02), parietal (P = 0.026) and occipital (P = 0.047) cortical regions compared with healthy controls. The greatest 11C-BU99008 VT increase in patients with early Parkinson's disease was observed in the brainstem (52%; P = 0.018). In patients with moderate/advanced Parkinson's disease, loss of 11C-BU99008 VT was observed across frontal (P = 0.002), temporal (P < 0.001), parietal (P = 0.039), occipital (P = 0.024), and insula (P < 0.001) cortices; and in the subcortical regions of caudate (P < 0.001), putamen (P < 0.001) and thalamus (P < 0.001); and in the brainstem (P = 0.018) compared with healthy controls. In patients with Parkinson's disease, loss of 11C-BU99008 VT in cortical regions, striatum, thalamus and brainstem correlated with longer disease duration (P < 0.05) and higher disease burden scores, measured with Movement Disorder Society Unified Parkinson's Disease Rating Scale (P < 0.05). In the subgroup of patients with moderate/advanced Parkinson's disease, loss of 11C-BU99008 VT in the frontal (r = 0.79; P = 0.001), temporal (r = 0.74; P = 0.002) and parietal (r = 0.89; P < 0.001) cortex correlated with global cognitive impairment. This study demonstrates in vivo the role of astroglia in the initiation and progression of Parkinson's disease. Reactive astroglia observed early in Parkinson's disease could reflect a neuroprotective compensatory mechanisms and pro-inflammatory upregulation in response to α-synuclein accumulation. However, as the disease progresses and significant neurodegeneration occurs, astroglia lose their reactive function and such loss in the cortex has clinical relevance in the development of cognitive impairment.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - George Dervenoulas
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Gennaro Pagano
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Robin J Tyacke
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, Burlington Danes Building, Hammersmith Hospital campus, 160 Du Cane Road, London, UK
| | - Sotirios Polychronis
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Jim Myers
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, Burlington Danes Building, Hammersmith Hospital campus, 160 Du Cane Road, London, UK
| | - Roger N Gunn
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
- Invicro LLC, Centre for Imaging Sciences, Hammersmith Hospital, London, UK
| | - Eugenii A Rabiner
- Invicro LLC, Centre for Imaging Sciences, Hammersmith Hospital, London, UK
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - David Nutt
- Neuropsychopharmacology Unit, Centre for Academic Psychiatry, Division of Brain Sciences, Imperial College London, Burlington Danes Building, Hammersmith Hospital campus, 160 Du Cane Road, London, UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
44
|
Chronic inflammation in multiple sclerosis - seeing what was always there. Nat Rev Neurol 2019; 15:582-593. [PMID: 31420598 DOI: 10.1038/s41582-019-0240-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2019] [Indexed: 12/18/2022]
Abstract
Activation of innate immune cells and other compartmentalized inflammatory cells in the brains and spinal cords of people with relapsing-remitting multiple sclerosis (MS) and progressive MS has been well described histopathologically. However, conventional clinical MRI is largely insensitive to this inflammatory activity. The past two decades have seen the introduction of quantitative dynamic MRI scanning with contrast agents that are sensitive to the reduction in blood-brain barrier integrity associated with inflammation and to the trafficking of inflammatory myeloid cells. New MRI imaging sequences provide improved contrast for better detection of grey matter lesions. Quantitative lesion volume measures and magnetic resonance susceptibility imaging are sensitive to the activity of macrophages in the rims of white matter lesions. PET and magnetic resonance spectroscopy methods can also be used to detect contributions from innate immune activation in the brain and spinal cord. Some of these advanced research imaging methods for visualization of chronic inflammation are practical for relatively routine clinical applications. Observations made with the use of these techniques suggest ways of stratifying patients with MS to improve their care. The imaging methods also provide new tools to support the development of therapies for chronic inflammation in MS.
Collapse
|
45
|
Prospects and challenges of imaging neuroinflammation beyond TSPO in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2019; 46:2831-2847. [PMID: 31396666 PMCID: PMC6879435 DOI: 10.1007/s00259-019-04462-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation, as defined by the activation of microglia and astrocytes, has emerged in the last years as a key element of the pathogenesis of neurodegenerative diseases based on genetic findings and preclinical and human studies. This has raised the need for new methodologies to assess and follow glial activation in patients, prompting the development of PET ligands for molecular imaging of glial cells and novel structural MRI and DTI tools leading to a multimodal approach. The present review describes the recent advancements in microglia and astrocyte biology in the context of health, ageing, and Alzheimer's disease, the most common dementia worldwide. The review further delves in molecular imaging discussing the challenges associated with past and present targets, including conflicting findings, and finally, presenting novel methodologies currently explored to improve our in vivo knowledge of the neuroinflammatory patterns in Alzheimer's disease. With glial cell activation as a potential therapeutic target in neurodegenerative diseases, the translational research between cell biologists, chemists, physicists, radiologists, and neurologists should be strengthened.
Collapse
|
46
|
Moriguchi S, Wilson AA, Miler L, Rusjan PM, Vasdev N, Kish SJ, Rajkowska G, Wang J, Bagby M, Mizrahi R, Varughese B, Houle S, Meyer JH. Monoamine Oxidase B Total Distribution Volume in the Prefrontal Cortex of Major Depressive Disorder: An [11C]SL25.1188 Positron Emission Tomography Study. JAMA Psychiatry 2019; 76:634-641. [PMID: 30840042 PMCID: PMC6551845 DOI: 10.1001/jamapsychiatry.2019.0044] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
IMPORTANCE Monoamine oxidase B (MAO-B) is an important, high-density enzyme in the brain that generates oxidative stress by hydrogen peroxide production, alters mitochondrial function, and metabolizes nonserotonergic monoamines. Recent advances in positron emission tomography radioligand development for MAO-B in humans enable highly quantitative measurement of MAO-B distribution volume (MAO-B VT), an index of MAO-B density. To date, this is the first investigation of MAO-B in the brain of major depressive disorder that evaluates regions beyond the raphe and amygdala. OBJECTIVE To investigate whether MAO-B VT is elevated in the prefrontal cortex in major depressive episodes (MDEs) of major depressive disorder. DESIGN, SETTING, AND PARTICIPANTS This case-control study was performed at a tertiary care psychiatric hospital from April 1, 2014, to August 30, 2018. Twenty patients with MDEs without current psychiatric comorbidities and 20 age-matched controls underwent carbon 11-labeled [11C]SL25.1188 positron emission tomography scanning to measure MAO-B VT. All participants were drug and medication free, nonsmoking, and otherwise healthy. MAIN OUTCOMES AND MEASURES The MAO-B VT in the prefrontal cortex (PFC). The second main outcome was to evaluate the association between MAO-B VT in the PFC and duration of major depressive disorder illness. RESULTS Twenty patients with MDEs (mean [SD] age, 34.2 [13.2] years; 11 women) and 20 healthy controls (mean [SD] age, 33.7 [13.1] years; 10 women) were recruited. Patients with MDEs had significantly greater MAO-B VT in the PFC (mean, 26%; analysis of variance, F1,38 = 19.6, P < .001). In individuals with MDEs, duration of illness covaried positively with MAO-B VT in the PFC (analysis of covariance, F1,18 = 15.2, P = .001), as well as most other cortex regions and the thalamus. CONCLUSIONS AND RELEVANCE Fifty percent (10 of 20) of patients with MDEs had MAO-B VT values in the PFC exceeding those of healthy controls. Greater MAO-B VT is an index of MAO-B overexpression, which may contribute to pathologies of mitochondrial dysfunction, elevated synthesis of neurotoxic products, and increased metabolism of nonserotonergic monoamines. Hence, this study identifies a common pathological marker associated with downstream consequences poorly targeted by the common selective serotonin reuptake inhibitor treatments. It is also recommended that the highly selective MAO-B inhibitor medications that are compatible for use with other antidepressants and have low risk for hypertensive crisis should be developed or repurposed as adjunctive treatment for MDEs.
Collapse
Affiliation(s)
- Sho Moriguchi
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Alan A. Wilson
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Laura Miler
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Pablo M. Rusjan
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Neil Vasdev
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Stephen J. Kish
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Grazyna Rajkowska
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson
| | - Junming Wang
- Department of Pathology, University of Mississippi Medical Center, Jackson
| | - Michael Bagby
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Romina Mizrahi
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Ben Varughese
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sylvain Houle
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey H. Meyer
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Carter SF, Herholz K, Rosa-Neto P, Pellerin L, Nordberg A, Zimmer ER. Astrocyte Biomarkers in Alzheimer's Disease. Trends Mol Med 2019; 25:77-95. [PMID: 30611668 DOI: 10.1016/j.molmed.2018.11.006] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023]
Abstract
Astrocytic contributions to Alzheimer's disease (AD) progression were, until recently, largely overlooked. Astrocytes are integral to normal brain function and astrocyte reactivity is an early feature of AD, potentially providing a promising target for preclinical diagnosis and treatment. Several in vivo AD biomarkers already exist, but presently there is a paucity of specific and sensitive in vivo astrocyte biomarkers that can accurately measure preclinical AD. Measuring monoamine oxidase-B with neuroimaging and glial fibrillary acidic protein from bodily fluids are biomarkers that are currently available. Developing novel, more specific, and sensitive astrocyte biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial astrocytic functions in response to AD pathology. This review discusses astrocyte biomarkers in the context of AD.
Collapse
Affiliation(s)
- Stephen F Carter
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Karl Herholz
- Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, Canada; Douglas Hospital Research Centre, Montreal, Canada; Montreal Neurological Institute, Montreal, Canada
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, Lausanne, Switzerland; Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, Bordeaux Cedex 33760, France
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Eduardo R Zimmer
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute (BraIns) of Rio Grande do Sul, Porto Alegre, Brazil; Website: www.zimmer-lab.org.
| |
Collapse
|
48
|
Longitudinal association between astrocyte function and glucose metabolism in autosomal dominant Alzheimer's disease. Eur J Nucl Med Mol Imaging 2018; 46:348-356. [PMID: 30515545 PMCID: PMC6333721 DOI: 10.1007/s00259-018-4217-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE The spatial resolution of 18F-fluorodeoxyglucose PET does not allow the specific cellular origin of its signal to be determined, but it is commonly accepted that transport and trapping of 18F-fluorodeoxyglucose reflects neuronal glucose metabolism. The main frameworks for the diagnosis of Alzheimer's disease suggest that hypometabolism measured with 18F-fluorodeoxyglucose PET is a biomarker of neuronal injury and neurodegeneration. There is preclinical evidence to suggest that astrocytes contribute, at least partially, to the in vivo 18F-fluorodeoxyglucose PET signal. However, due to a paucity of PET tracers for imaging astrocytic processes, the relationship between astrocyte function and glucose metabolism in human brain is not fully understood. The aim of this study was to investigate the longitudinal association between astrocyte function and glucose metabolism in Alzheimer's disease. METHODS The current investigation combined longitudinal PET data from patients with autosomal dominant Alzheimer's disease, including data on astrocyte function (11C-deuterium-L-deprenyl binding) and glucose metabolism (18F-fluorodeoxyglucose uptake). Research participants included 7 presymptomatic and 4 symptomatic mutation carriers (age 44.9 ± 9.8 years and 58.0 ± 3.7 years, respectively) and 16 noncarriers (age 51.1 ± 14.2 years). Eight carriers and eight noncarriers underwent longitudinal follow-up PET imaging at an average of 2.8 ± 0.2 and 3.0 ± 0.5 years from baseline, respectively. RESULTS Longitudinal decline in astrocyte function as measured using 11C-deuterium-L-deprenyl PET was significantly associated with progressive hypometabolism (18F-fluorodeoxyglucose uptake) in mutation carriers; no significant association was observed in noncarriers. CONCLUSION The emerging data shift the accepted wisdom that decreases in cerebral metabolism measured with 18F-fluorodeoxyglucose solely reflect neuronal injury, and places astrocytes more centrally in the development of Alzheimer's disease.
Collapse
|
49
|
Molecular Imaging in Huntington's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 142:289-333. [PMID: 30409256 DOI: 10.1016/bs.irn.2018.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huntington's disease (HD) is a rare monogenic neurodegenerative disorder caused by a trinucleotide CAG repeat expansion in the huntingtin gene resulting in the formation of intranuclear inclusions of mutated huntingtin. The accumulation of mutated huntingtin leads to loss of GABAergic medium spiny neurons (MSNs); subsequently resulting in the development of chorea, cognitive dysfunction and psychiatric symptoms. Premanifest HD gene expansion carriers, provide a unique cohort to examine very early molecular changes, occurring before the development of overt symptoms, to elucidate disease pathophysiology and identify reliable biomarkers of HD progression. Positron emission tomography (PET) is a non-invasive molecular imaging technique allowing the evaluation of specific molecular targets in vivo. Selective PET radioligands provide invaluable tools to investigate the role of the dopaminergic system, brain metabolism, microglial activation, phosphodiesterase 10A, and cannabinoid, GABA, adenosine and opioid receptors in HD. PET has been employed to monitor disease progression aiming to identify a reliable biomarker to predict phenoconversion from premanifest to manifest HD.
Collapse
|
50
|
Edison P, Donat CK, Sastre M. In vivo Imaging of Glial Activation in Alzheimer's Disease. Front Neurol 2018; 9:625. [PMID: 30131755 PMCID: PMC6090997 DOI: 10.3389/fneur.2018.00625] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by memory loss and decline of cognitive function, associated with progressive neurodegeneration. While neuropathological processes like amyloid plaques and tau neurofibrillary tangles have been linked to neuronal death in AD, the precise role of glial activation on disease progression is still debated. It was suggested that neuroinflammation could occur well ahead of amyloid deposition and may be responsible for clearing amyloid, having a neuroprotective effect; however, later in the disease, glial activation could become deleterious, contributing to neuronal toxicity. Recent genetic and preclinical studies suggest that the different activation states of microglia and astrocytes are complex, not as polarized as previously thought, and that the heterogeneity in their phenotype can switch during disease progression. In the last few years, novel imaging techniques e.g., new radiotracers for assessing glia activation using positron emission tomography and advanced magnetic resonance imaging technologies have emerged, allowing the correlation of neuro-inflammatory markers with cognitive decline, brain function and brain pathology in vivo. Here we review all new imaging technology in AD patients and animal models that has the potential to serve for early diagnosis of the disease, to monitor disease progression and to test the efficacy and the most effective time window for potential anti-inflammatory treatments.
Collapse
Affiliation(s)
- Paul Edison
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Cornelius K Donat
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Magdalena Sastre
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|