51
|
Bettcher BM, Tansey MG, Dorothée G, Heneka MT. Peripheral and central immune system crosstalk in Alzheimer disease - a research prospectus. Nat Rev Neurol 2021; 17:689-701. [PMID: 34522039 PMCID: PMC8439173 DOI: 10.1038/s41582-021-00549-x] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Dysregulation of the immune system is a cardinal feature of Alzheimer disease (AD), and a considerable body of evidence indicates pathological alterations in central and peripheral immune responses that change over time. Considering AD as a systemic immune process raises important questions about how communication between the peripheral and central compartments occurs and whether this crosstalk represents a therapeutic target. We established a whitepaper workgroup to delineate the current status of the field and to outline a research prospectus for advancing our understanding of peripheral-central immune crosstalk in AD. To guide the prospectus, we begin with an overview of seminal clinical observations that suggest a role for peripheral immune dysregulation and peripheral-central immune communication in AD, followed by formative animal data that provide insights into possible mechanisms for these clinical findings. We then present a roadmap that defines important next steps needed to overcome conceptual and methodological challenges, opportunities for future interdisciplinary research, and suggestions for translating promising mechanistic studies into therapeutic interventions.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Behavioral Neurology Section, Department of Neurology, University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Malú G Tansey
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
| | - Guillaume Dorothée
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Team "Immune System and Neuroinflammation", Hôpital Saint-Antoine, Paris, France
| | - Michael T Heneka
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
52
|
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: 56] [Impact Index Per Article: 14.0] [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
|
53
|
Tournier BB, Tsartsalis S, Ceyzériat K, Fraser BH, Grégoire MC, Kövari E, Millet P. Astrocytic TSPO Upregulation Appears Before Microglial TSPO in Alzheimer's Disease. J Alzheimers Dis 2021; 77:1043-1056. [PMID: 32804124 PMCID: PMC7683091 DOI: 10.3233/jad-200136] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background: In vivo PET/SPECT imaging of neuroinflammation is primarily based on the estimation of the 18 kDa-translocator-protein (TSPO). However, TSPO is expressed by different cell types which complicates the interpretation. Objective: The present study evaluates the cellular origin of TSPO alterations in Alzheimer’s disease (AD). Methods: The TSPO cell origin was evaluated by combining radioactive imaging approaches using the TSPO radiotracer [125I]CLINDE and fluorescence-activated cell sorting, in a rat model of AD (TgF344-AD) and in AD subjects. Results: In the hippocampus of TgF344-AD rats, TSPO overexpression not only concerns glial cells but the increase is visible at 12 and 24 months in astrocytes and only at 24 months in microglia. In the temporal cortex of AD subjects, TSPO upregulation involved only glial cells. However, the mechanism of this upregulation appears different with an increase in the number of TSPO binding sites per cell without cell proliferation in the rat, and a microglial cell population expansion with a constant number of binding sites per cell in human AD. Conclusion: These data indicate an earlier astrocyte intervention than microglia and that TSPO in AD probably is an exclusive marker of glial activity without interference from other TSPO-expressing cells. This observation indicates that the interpretation of TSPO imaging depends on the stage of the pathology, and highlights the particular role of astrocytes.
Collapse
Affiliation(s)
- Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland.,Division of Nuclear medicine, University Hospitals of Geneva, Switzerland
| | - Ben H Fraser
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW, Australia
| | - Marie-Claude Grégoire
- ANSTO LifeSciences, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Sydney, NSW, Australia
| | - Enikö Kövari
- Division of Geriatric Psychiatry, Department of Mental Health and Psychiatry, University Hospitals of Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Switzerland
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Switzerland
| |
Collapse
|
54
|
Keskin E, Can EY, Aydın HA, Işık E, Özgen U, Şimşek K, Cengil O, Başar C, Kalaycı M. The preventative effect of of Ro5-4864 (peripheral benzodiazepine receptor agonist) on spinal epidural fibrosis after laminectomy in a rat model. Neurol Res 2021; 43:1107-1115. [PMID: 34461817 DOI: 10.1080/01616412.2021.1949689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To investigate the histopathological effects of a peripheral benzodiazepine receptor agonist (Ro5-4864) on epidural fibrosis (EF) in an experimental study model (post-laminectomy) in rats. METHODS A total of 32 albino Wistar rats were randomly divided into four equal groups (n = 8). In Group 1, no treatment was applied after laminectomy (control group). In Group 2, hemostasis was achieved after Laminectomy, and the surgical procedure was terminated by placing a 2-mm absorbable gelatin sponge dipped in saline into the epidural space. In Group 3, low-dose (4 mg/kg) Ro5-4864 was administered 30 minutes before the surgery. In Group 4, high-dose (8 mg/kg) Ro5-4864 was administered 30 minutes before the surgery. A histopathological examination was performed to evaluate arachnoidal invasion and EF. RESULTS Our data revealed the EF was significantly reduced in rats treated with high-dose Ro5-4864 (Group 4) compared to the control and saline-soaked Spongostan groups (p = 0.000 and p = 0.006, respectively). There was no significant difference between the groups treated with high- and low-dose Ro5-4864. Arachnoidal invasion was not seen in any of the rats in the high-dose R05-4864 group. However, the arachnoidal invasion results did not significantly differ between the study groups (p = 0.052 = 0.05). CONCLUSIONS Our study showed that Ro5-4864 could be effective in reducing EF in rats after.
Collapse
Affiliation(s)
- Emrah Keskin
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Emine Yılmaz Can
- Department of Pharmacology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Hasan Ali Aydın
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Emre Işık
- Department of Pathology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Utku Özgen
- Department of Neurosurgery, Atatürk State Hospital, Zonguldak, Turkey
| | - Kenan Şimşek
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Osman Cengil
- Department of Experimental Animal Research Laboratory, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Cansu Başar
- Insurance Information and Monitoring Center, Istanbul, Turkey
| | - Murat Kalaycı
- Department of Neurosurgery, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
55
|
Wang J, Beecher K. TSPO: an emerging role in appetite for a therapeutically promising biomarker. Open Biol 2021; 11:210173. [PMID: 34343461 PMCID: PMC8331234 DOI: 10.1098/rsob.210173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
There is accumulating evidence that an obesogenic Western diet causes neuroinflammatory damage to the brain, which then promotes further appetitive behaviour. Neuroinflammation has been extensively studied by analysing the translocator protein of 18 kDa (TSPO), a protein that is upregulated in the inflamed brain following a damaging stimulus. As a result, there is a rich supply of TSPO-specific agonists, antagonists and positron emission tomography ligands. One TSPO ligand, etifoxine, is also currently used clinically for the treatment of anxiety with a minimal side-effect profile. Despite the neuroinflammatory pathogenesis of diet-induced obesity, and the translational potential of targeting TSPO, there is sparse literature characterizing the effect of TSPO on appetite. Therefore, in this review, the influence of TSPO on appetite is discussed. Three putative mechanisms for TSPO's appetite-modulatory effect are then characterized: the TSPO–allopregnanolone–GABAAR signalling axis, glucosensing in tanycytes and association with the synaptic protein RIM-BP1. We highlight that, in addition to its plethora of functions, TSPO is a regulator of appetite. This review ultimately suggests that the appetite-modulating function of TSPO should be further explored due to its potential therapeutic promise.
Collapse
Affiliation(s)
- Joshua Wang
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kate Beecher
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
56
|
Resolving the cellular specificity of TSPO imaging in a rat model of peripherally-induced neuroinflammation. Brain Behav Immun 2021; 96:154-167. [PMID: 34052363 PMCID: PMC8323128 DOI: 10.1016/j.bbi.2021.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/09/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022] Open
Abstract
The increased expression of 18 kDa Translocator protein (TSPO) is one of the few available biomarkers of neuroinflammation that can be assessed in humans in vivo by positron emission tomography (PET). TSPO PET imaging of the central nervous system (CNS) has been widely undertaken, but to date no clear consensus has been reached about its utility in brain disorders. One reason for this could be because the interpretation of TSPO PET signal remains challenging, given the cellular heterogeneity and ubiquity of TSPO in the brain. The aim of the current study was to ascertain if TSPO PET imaging can be used to detect neuroinflammation induced by a peripheral treatment with a low dose of the endotoxin, lipopolysaccharide (LPS), in a rat model (ip LPS), and investigate the origin of TSPO signal changes in terms of their cellular sources and regional distribution. An initial pilot study utilising both [18F]DPA-714 and [11C]PK11195 TSPO radiotracers demonstrated [18F]DPA-714 to exhibit a significantly higher lesion-related signal in the intracerebral LPS rat model (ic LPS) than [11C]PK11195. Subsequently, [18F]DPA-714 was selected for use in the ip LPS study. Twenty-four hours after ip LPS, there was an increased uptake of [18F]DPA-714 across the whole brain. Further analyses of regions of interest, using immunohistochemistry and RNAscope Multiplex fluorescence V2 in situ hybridization technology, showed TSPO expression in microglia, monocyte derived-macrophages, astrocytes, neurons and endothelial cells. The expression of TSPO was significantly increased after ip LPS in a region-dependent manner: with increased microglia, monocyte-derived macrophages and astrocytes in the substantia nigra, in contrast to the hippocampus where TSPO was mostly confined to microglia and astrocytes. In summary, our data demonstrate the robust detection of peripherally-induced neuroinflammation in the CNS utilising the TSPO PET radiotracer, [18F]DPA-714, and importantly, confirm that the resultant increase in TSPO signal increase arises mostly from a combination of microglia, astrocytes and monocyte-derived macrophages.
Collapse
|
57
|
Beaino W, Janssen B, Vugts DJ, de Vries HE, Windhorst AD. Towards PET imaging of the dynamic phenotypes of microglia. Clin Exp Immunol 2021; 206:282-300. [PMID: 34331705 PMCID: PMC8561701 DOI: 10.1111/cei.13649] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023] Open
Abstract
There is increasing evidence showing the heterogeneity of microglia activation in neuroinflammatory and neurodegenerative diseases. It has been hypothesized that pro‐inflammatory microglia are detrimental and contribute to disease progression, while anti‐inflammatory microglia play a role in damage repair and remission. The development of therapeutics targeting the deleterious glial activity and modulating it into a regenerative phenotype relies heavily upon a clearer understanding of the microglia dynamics during disease progression and the ability to monitor therapeutic outcome in vivo. To that end, molecular imaging techniques are required to assess microglia dynamics and study their role in disease progression as well as to evaluate the outcome of therapeutic interventions. Positron emission tomography (PET) is such a molecular imaging technique, and provides unique capabilities for non‐invasive quantification of neuroinflammation and has the potential to discriminate between microglia phenotypes and define their role in the disease process. However, several obstacles limit the possibility for selective in vivo imaging of microglia phenotypes mainly related to the poor characterization of specific targets that distinguish the two ends of the microglia activation spectrum and lack of suitable tracers. PET tracers targeting translocator protein 18 kDa (TSPO) have been extensively explored, but despite the success in evaluating neuroinflammation they failed to discriminate between microglia activation statuses. In this review, we highlight the current knowledge on the microglia phenotypes in the major neuroinflammatory and neurodegenerative diseases. We also discuss the current and emerging PET imaging targets, the tracers and their potential in discriminating between the pro‐ and anti‐inflammatory microglia activation states.
Collapse
Affiliation(s)
- Wissam Beaino
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Bieneke Janssen
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Danielle J Vugts
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| |
Collapse
|
58
|
Viejo L, Noori A, Merrill E, Das S, Hyman BT, Serrano-Pozo A. Systematic review of human post-mortem immunohistochemical studies and bioinformatics analyses unveil the complexity of astrocyte reaction in Alzheimer's disease. Neuropathol Appl Neurobiol 2021; 48:e12753. [PMID: 34297416 PMCID: PMC8766893 DOI: 10.1111/nan.12753] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 12/24/2022]
Abstract
AIMS Reactive astrocytes in Alzheimer's disease (AD) have traditionally been demonstrated by increased glial fibrillary acidic protein (GFAP) immunoreactivity; however, astrocyte reaction is a complex and heterogeneous phenomenon involving multiple astrocyte functions beyond cytoskeletal remodelling. To better understand astrocyte reaction in AD, we conducted a systematic review of astrocyte immunohistochemical studies in post-mortem AD brains followed by bioinformatics analyses on the extracted reactive astrocyte markers. METHODS NCBI PubMed, APA PsycInfo and WoS-SCIE databases were interrogated for original English research articles with the search terms 'Alzheimer's disease' AND 'astrocytes.' Bioinformatics analyses included protein-protein interaction network analysis, pathway enrichment, and transcription factor enrichment, as well as comparison with public human -omics datasets. RESULTS A total of 306 articles meeting eligibility criteria rendered 196 proteins, most of which were reported to be upregulated in AD vs control brains. Besides cytoskeletal remodelling (e.g., GFAP), bioinformatics analyses revealed a wide range of functional alterations including neuroinflammation (e.g., IL6, MAPK1/3/8 and TNF), oxidative stress and antioxidant defence (e.g., MT1A/2A, NFE2L2, NOS1/2/3, PRDX6 and SOD1/2), lipid metabolism (e.g., APOE, CLU and LRP1), proteostasis (e.g., cathepsins, CRYAB and HSPB1/2/6/8), extracellular matrix organisation (e.g., CD44, MMP1/3 and SERPINA3), and neurotransmission (e.g., CHRNA7, GABA, GLUL, GRM5, MAOB and SLC1A2), among others. CTCF and ESR1 emerged as potential transcription factors driving these changes. Comparison with published -omics datasets validated our results, demonstrating a significant overlap with reported transcriptomic and proteomic changes in AD brains and/or CSF. CONCLUSIONS Our systematic review of the neuropathological literature reveals the complexity of AD reactive astrogliosis. We have shared these findings as an online resource available at www.astrocyteatlas.org.
Collapse
Affiliation(s)
- Lucía Viejo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Departamento de Farmacología y Terapéutica, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ayush Noori
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Harvard College, Cambridge, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA
| | - Emily Merrill
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,MIND Data Science Lab, Cambridge, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,MassGeneral Institute for Neurodegenerative Disease (MIND), Charlestown, MA, USA.,Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.,Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
59
|
Giordani A, Menziani MC, Moresco RM, Matarrese M, Paolino M, Saletti M, Giuliani G, Anzini M, Cappelli A. Exploring Translocator Protein (TSPO) Medicinal Chemistry: An Approach for Targeting Radionuclides and Boron Atoms to Mitochondria. J Med Chem 2021; 64:9649-9676. [PMID: 34254805 DOI: 10.1021/acs.jmedchem.1c00379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Translocator protein 18 kDa [TSPO or peripheral-type benzodiazepine receptor (PBR)] was identified in the search of binding sites for benzodiazepine anxiolytic drugs in peripheral regions. In these areas, binding sites for TSPO ligands were recognized in steroid-producing tissues. TSPO plays an important role in many cellular functions, and its coding sequence is highly conserved across species. TSPO is located predominantly on the membrane of mitochondria and is overexpressed in several solid cancers. TSPO basal expression in the CNS is low, but it becomes high in neurodegenerative conditions. Thus, TSPO constitutes not only as an outstanding drug target but also as a valuable marker for the diagnosis of a number of diseases. The aim of the present article is to show the lesson we have learned from our activity in TSPO medicinal chemistry and in approaching the targeted delivery to mitochondria by means of TSPO ligands.
Collapse
Affiliation(s)
- Antonio Giordani
- Rottapharm Biotech S.p.A., Via Valosa di Sopra 9, 20900 Monza, Italy
| | - Maria Cristina Menziani
- Dipartimento di Scienze Chimiche e Geologiche, Università di Modena e Reggio Emilia, Via Campi 103, 41121 Modena, Italy
| | - Rosa Maria Moresco
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Mario Matarrese
- Department of Medicine and Surgery, University of Milan-Bicocca, Nuclear Medicine Department, San Raffaele Scientific Institute, IBFM-CNR, Via Olgettina 60, 20132 Milano, Italy
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Germano Giuliani
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Maurizio Anzini
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia (Dipartimento di Eccellenza 2018-2022), Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
60
|
Zhang H, Wang H, Gao F, Yang J, Xu Y, Fu Y, Cai M, Zhang X, Yang Q, Tong K, Hu Y, Chen H, Ma C, He W, Zhang J. TSPO deficiency accelerates amyloid pathology and neuroinflammation by impairing microglial phagocytosis. Neurobiol Aging 2021; 106:292-303. [PMID: 34340010 DOI: 10.1016/j.neurobiolaging.2021.06.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 06/08/2021] [Accepted: 06/25/2021] [Indexed: 12/25/2022]
Abstract
Increasing evidence has placed inflammation and immune dysfunction at the center of the pathogenesis of Alzheimer's disease (AD). The mitochondrial protein translocator protein (18 kDa) (TSPO) is highly upregulated in microglia and astrocytes in response to inflammatory stimulation. However, the biological action of TSPO in the pathogenesis of AD has not been determined to date. In this study, we showed that TSPO expression was upregulated in brain tissues from AD patients and AD model mice. APP/PS1 mice lacking TSPO generated significantly higher levels of Aβ1-40 and Aβ1-42 peptides and more Aβ plaques, as well as enhanced microglial activation, in the brain. TSPO-deficient microglia cultured in vitro showed a significant decrease in the ability to phagocytose Aβ peptides or latex beads and generated more proinflammatory cytokines (TNF-α and IL-1β) in response to Aβ peptides. Our findings suggest that TSPO has protective functions against neuroinflammation and Aβ pathogenesis in AD. TSPO may be a potential drug target for the development of drugs that have therapeutic or preventive effects in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Han Zhang
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Huaishan Wang
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Fei Gao
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Jia Yang
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Yi Xu
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Yi Fu
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Menghua Cai
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Xue Zhang
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Qi Yang
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Kexin Tong
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Yu Hu
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Hui Chen
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei He
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China.
| | - Jianmin Zhang
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China.
| |
Collapse
|
61
|
Zhang PF, Hu H, Tan L, Yu JT. Microglia Biomarkers in Alzheimer's Disease. Mol Neurobiol 2021; 58:3388-3404. [PMID: 33713018 DOI: 10.1007/s12035-021-02348-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Early detection and clinical diagnosis of Alzheimer's disease (AD) have become an extremely important link in the prevention and treatment of AD. Because of the occult onset, the diagnosis and treatment of AD based on clinical symptoms are increasingly challenged by current severe situations. Therefore, molecular diagnosis models based on early AD pathological markers have received more attention. Among the possible pathological mechanisms, microglia which are necessary for normal brain function are highly expected and have been continuously studied in various models. Several AD biomarkers already exist, but currently there is a paucity of specific and sensitive microglia biomarkers which can accurately measure preclinical AD. Bringing microglia biomarkers into the molecular diagnostic system which is based on fluid and neuroimaging will play an important role in future scientific research and clinical practice. Furthermore, developing novel, more specific, and sensitive microglia biomarkers will make it possible to pharmaceutically target chemical pathways that preserve beneficial microglial functions in response to AD pathology. This review discusses microglia biomarkers in the context of AD.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
62
|
Kim B, Kim H, Kim S, Hwang YR. A brief review of non-invasive brain imaging technologies and the near-infrared optical bioimaging. Appl Microsc 2021; 51:9. [PMID: 34170436 PMCID: PMC8227874 DOI: 10.1186/s42649-021-00058-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Brain disorders seriously affect life quality. Therefore, non-invasive neuroimaging has received attention to monitoring and early diagnosing neural disorders to prevent their progress to a severe level. This short review briefly describes the current MRI and PET/CT techniques developed for non-invasive neuroimaging and the future direction of optical imaging techniques to achieve higher resolution and specificity using the second near-infrared (NIR-II) region of wavelength with organic molecules.
Collapse
Affiliation(s)
- Beomsue Kim
- Neural Circuit Research Group, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea.
| | - Hongmin Kim
- Neural Circuit Research Group, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Songhui Kim
- Neural Circuit Research Group, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| | - Young-Ran Hwang
- Neural Circuit Research Group, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, South Korea
| |
Collapse
|
63
|
Xiong B, Jin G, Xu Y, You W, Luo Y, Fang M, Chen B, Huang H, Yang J, Lin X, Yu C. Identification of Koumine as a Translocator Protein 18 kDa Positive Allosteric Modulator for the Treatment of Inflammatory and Neuropathic Pain. Front Pharmacol 2021; 12:692917. [PMID: 34248642 PMCID: PMC8264504 DOI: 10.3389/fphar.2021.692917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/07/2021] [Indexed: 01/08/2023] Open
Abstract
Koumine is an alkaloid that displays notable activity against inflammatory and neuropathic pain, but its therapeutic target and molecular mechanism still need further study. Translocator protein 18 kDa (TSPO) is a vital therapeutic target for pain treatment, and recent research implies that there may be allostery in TSPO. Our previous competitive binding assay hint that koumine may function as a TSPO positive allosteric modulator (PAM). Here, for the first time, we report the pharmacological characterization of koumine as a TSPO PAM. The results imply that koumine might be a high-affinity ligand of TSPO and that it likely acts as a PAM since it could delay the dissociation of 3H-PK11195 from TSPO. Importantly, the allostery was retained in vivo, as koumine augmented Ro5-4864-mediated analgesic and anti-inflammatory effects in several acute and chronic inflammatory and neuropathic pain models. Moreover, the positive allosteric modulatory effect of koumine on TSPO was further demonstrated in cell proliferation assays in T98G human glioblastoma cells. In summary, we have identified and characterized koumine as a TSPO PAM for the treatment of inflammatory and neuropathic pain. Our data lay a solid foundation for the use of the clinical candidate koumine to treat inflammatory and neuropathic pain, further demonstrate the allostery in TSPO, and provide the first proof of principle that TSPO PAM may be a novel avenue for the discovery of analgesics.
Collapse
Affiliation(s)
- Bojun Xiong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Guilin Jin
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Ying Xu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Wenbing You
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Yufei Luo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Menghan Fang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bing Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Huihui Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
64
|
Microglia in Neurodegenerative Events-An Initiator or a Significant Other? Int J Mol Sci 2021; 22:ijms22115818. [PMID: 34072307 PMCID: PMC8199265 DOI: 10.3390/ijms22115818] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
A change in microglia structure, signaling, or function is commonly associated with neurodegeneration. This is evident in the patient population, animal models, and targeted in vitro assays. While there is a clear association, it is not evident that microglia serve as an initiator of neurodegeneration. Rather, the dynamics imply a close interaction between the various cell types and structures in the brain that orchestrate the injury and repair responses. Communication between microglia and neurons contributes to the physiological phenotype of microglia maintaining cells in a surveillance state and allows the cells to respond to events occurring in their environment. Interactions between microglia and astrocytes is not as well characterized, nor are interactions with other members of the neurovascular unit; however, given the influence of systemic factors on neuroinflammation and disease progression, such interactions likely represent significant contributes to any neurodegenerative process. In addition, they offer multiple target sites/processes by which environmental exposures could contribute to neurodegenerative disease. Thus, microglia at least play a role as a significant other with an equal partnership; however, claiming a role as an initiator of neurodegeneration remains somewhat controversial.
Collapse
|
65
|
Morgan DG, Mielke MM. Knowledge gaps in Alzheimer's disease immune biomarker research. Alzheimers Dement 2021; 17:2030-2042. [PMID: 33984178 DOI: 10.1002/alz.12342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 11/09/2022]
Abstract
Considerable evidence has accumulated implicating a role for immune mechanisms in moderating the pathology in Alzheimer's disease dementia. However, the appropriate therapeutic target, the appropriate direction of manipulation, and the stage of disease at which to begin treatment remain unanswered questions. Part of the challenge derives from the absence of any selective pressure to develop a coordinated beneficial immune response to severe neural injury in adults. Thus, immune responses to the prevailing stimuli are likely to contain both beneficial and detrimental components. Knowledge gaps include: (1) how a biomarker change relates to the underlying biology, (2) the degree to which pathological stage group differences reflect a response to pathology versus trait differences among individuals regulating risk of developing pathology, (3) the degree to which biomarker levels are predictive of subsequent changes in pathology and/or cognition, and (4) experimental manipulations in model systems to determine whether differences in immune biomarkers are causally related to pathology.
Collapse
Affiliation(s)
- David G Morgan
- Alzheimer's Alliance, Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Michelle M Mielke
- Division of Epidemiology, Department of Health Sciences Research, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
66
|
Di Cataldo V, Debatisse J, Piraquive J, Géloën A, Grandin C, Verset M, Taborik F, Labaronne E, Loizon E, Millon A, Mury P, Pialoux V, Serusclat A, Lamberton F, Ibarrola D, Lavenne F, Le Bars D, Troalen T, Confais J, Crola Da Silva C, Mechtouff L, Contamin H, Fayad ZA, Canet-Soulas E. Cortical inflammation and brain signs of high-risk atherosclerosis in a non-human primate model. Brain Commun 2021; 3:fcab064. [PMID: 33937770 PMCID: PMC8063585 DOI: 10.1093/braincomms/fcab064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 11/14/2022] Open
Abstract
Atherosclerosis is a chronic systemic inflammatory disease, inducing cardiovascular and cerebrovascular acute events. A role of neuroinflammation is suspected, but not yet investigated in the gyrencephalic brain and the related activity at blood−brain interfaces is unknown. A non-human primate model of advanced atherosclerosis was first established using longitudinal blood samples, multimodal imaging and gene analysis in aged animals. Non-human primate carotid lesions were compared with human carotid endarterectomy samples. During the whole-body imaging session, imaging of neuroinflammation and choroid plexus function was performed. Advanced plaques were present in multiple sites, premature deaths occurred and downstream lesions (myocardial fibrosis, lacunar stroke) were present in this model. Vascular lesions were similar to in humans: high plaque activity on PET and MRI imaging and systemic inflammation (high plasma C-reactive protein levels: 42 ± 14 µg/ml). We also found the same gene association (metabolic, inflammatory and anti-inflammatory markers) as in patients with similar histological features. Metabolic imaging localized abnormal brain glucose metabolism in the frontal cortex. It corresponded to cortical neuro-inflammation (PET imaging) that correlated with C-reactive protein level. Multimodal imaging also revealed pronounced choroid plexus function impairment in aging atherosclerotic non-human primates. In conclusion, multimodal whole-body inflammation exploration at the vascular level and blood−brain interfaces identified high-risk aging atherosclerosis. These results open the way for systemic and central inflammation targeting in atherosclerosis in the new era of immunotherapy.
Collapse
Affiliation(s)
- Vanessa Di Cataldo
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Justine Debatisse
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France.,Siemens-Healthcare SAS, Saint-Denis, France
| | | | - Alain Géloën
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | - Emmanuel Labaronne
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Emmanuelle Loizon
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Antoine Millon
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Pauline Mury
- LIBM Laboratory, Univ Lyon, Université Lyon 1, Lyon, France
| | | | - André Serusclat
- Radiology Department, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
| | | | | | | | | | | | | | - Claire Crola Da Silva
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| | - Laura Mechtouff
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France.,Stroke Department, Hospices Civils de Lyon, Lyon, France
| | | | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, Univ Lyon, INSERM U1060, INRAE 1397, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
67
|
Knight AC, Varlow C, Zi T, Liang SH, Josephson L, Schmidt K, Patel S, Vasdev N. In Vitro Evaluation of [ 3H]CPPC as a Tool Radioligand for CSF-1R. ACS Chem Neurosci 2021; 12:998-1006. [PMID: 33667059 DOI: 10.1021/acschemneuro.0c00802] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Microglia play a role in several central nervous system (CNS) diseases and are a highly sought target for positron emission tomography (PET) imaging and therapeutic intervention. 5-Cyano-N-(4-(4-[11C]methylpiperazin-1-yl)-2-(piperidin-1-yl)phenyl)furan-2-carboxamide ([11C]CPPC) is a radiopharmaceutical designed to selectively target microglia via macrophage colony stimulating factor-1 receptor (CSF-1R) in the CNS. Herein, we report the first preclinical evaluation of [3H]CPPC using radioligand binding methods for the evaluation of putative CSF-1R inhibitors in rodent models of neuroinflammation. The distribution of [3H]CPPC by autoradiography did not align with 18 kDa translocator protein (TSPO) distribution using [3H]PBR28 and IBA-1 staining for microglia. In the CNS, [3H]CPPC had considerable nonspecific binding, as indicated by a low displacement of the tritiated ligand by unlabeled CPPC and the known CSF1R inhibitors BLZ-945 and PLX3397. Spleen was identified as a tissue that provided an adequate signal-to-noise ratio to enable screening with [3H]CPPC and a library of 20 novel PLX3397 derivatives. However, unlabeled CPPC lacked selectivity and showed off-target binding to a substantial number of kinase targets (204 out of 403 tested) at a concentration relevant to in vitro radioligand binding assays (10 μM). These findings suggest that, while [3H]CPPC may have utility as a radioligand tool for the evaluation of peripheral targets and screening of CSF-1R inhibitors, it may have limited utility as an in vivo CNS imaging probe on the basis of the current evaluation.
Collapse
Affiliation(s)
- Ashley C. Knight
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Cassis Varlow
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tong Zi
- Codiak Biosciences, Cambridge, Massachusetts 02140, United States
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
- MedChem Imaging, Inc., Boston, Massachusetts 02210, United States
| | - Karl Schmidt
- Codiak Biosciences, Cambridge, Massachusetts 02140, United States
| | - Shil Patel
- Codiak Biosciences, Cambridge, Massachusetts 02140, United States
| | - Neil Vasdev
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health (CAMH), Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
- MedChem Imaging, Inc., Boston, Massachusetts 02210, United States
| |
Collapse
|
68
|
Rivière G, Jaipuria G, Andreas LB, Leonov A, Giller K, Becker S, Zweckstetter M. Membrane-embedded TSPO: an NMR view. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2021; 50:173-180. [PMID: 33354729 PMCID: PMC8071791 DOI: 10.1007/s00249-020-01487-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/19/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022]
Abstract
Translocator Protein (18 kDa) (TSPO) is a mitochondrial transmembrane protein commonly used as a biomarker for neuroinflammation and is also a potential therapeutic target in neurodegenerative diseases. Despite intensive research efforts, the function of TSPO is still largely enigmatic. Deciphering TSPO structure in the native lipid environment is essential to gain insight into its cellular activities and to design improved diagnostic and therapeutic ligands. Here, we discuss the influence of lipid composition on the structure of mammalian TSPO embedded into lipid bilayers on the basis of solid-state NMR experiments. We further highlight that cholesterol can influence both the tertiary and quaternary TSPO structure and also influence TSPO localization in mitochondria-associated endoplasmic reticulum membranes.
Collapse
Affiliation(s)
- Gwladys Rivière
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Garima Jaipuria
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | - Loren B Andreas
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Andrei Leonov
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Karin Giller
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Stefan Becker
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Markus Zweckstetter
- Senior Research Group of Translational Structural Biology in Dementia, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany.
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
69
|
Escartin C, Galea E, Lakatos A, O'Callaghan JP, Petzold GC, Serrano-Pozo A, Steinhäuser C, Volterra A, Carmignoto G, Agarwal A, Allen NJ, Araque A, Barbeito L, Barzilai A, Bergles DE, Bonvento G, Butt AM, Chen WT, Cohen-Salmon M, Cunningham C, Deneen B, De Strooper B, Díaz-Castro B, Farina C, Freeman M, Gallo V, Goldman JE, Goldman SA, Götz M, Gutiérrez A, Haydon PG, Heiland DH, Hol EM, Holt MG, Iino M, Kastanenka KV, Kettenmann H, Khakh BS, Koizumi S, Lee CJ, Liddelow SA, MacVicar BA, Magistretti P, Messing A, Mishra A, Molofsky AV, Murai KK, Norris CM, Okada S, Oliet SHR, Oliveira JF, Panatier A, Parpura V, Pekna M, Pekny M, Pellerin L, Perea G, Pérez-Nievas BG, Pfrieger FW, Poskanzer KE, Quintana FJ, Ransohoff RM, Riquelme-Perez M, Robel S, Rose CR, Rothstein JD, Rouach N, Rowitch DH, Semyanov A, Sirko S, Sontheimer H, Swanson RA, Vitorica J, Wanner IB, Wood LB, Wu J, Zheng B, Zimmer ER, Zorec R, Sofroniew MV, Verkhratsky A. Reactive astrocyte nomenclature, definitions, and future directions. Nat Neurosci 2021; 24:312-325. [PMID: 33589835 PMCID: PMC8007081 DOI: 10.1038/s41593-020-00783-4] [Citation(s) in RCA: 1321] [Impact Index Per Article: 330.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022]
Abstract
Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.
Collapse
Affiliation(s)
- Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| | - Elena Galea
- Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
- ICREA, Barcelona, Spain.
| | - András Lakatos
- John van Geest Centre for Brain Repair and Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - James P O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Alberto Serrano-Pozo
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andrea Volterra
- Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - Giorgio Carmignoto
- Neuroscience Institute, Italian National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Amit Agarwal
- The Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Nicola J Allen
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, La Jolla, California, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Ari Barzilai
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences and Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv Tel Aviv, Israel
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Arthur M Butt
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Wei-Ting Chen
- Center for Brain and Disease Research, VIB and University of Leuven, Leuven, Belgium
| | - Martine Cohen-Salmon
- 'Physiology and Physiopathology of the Gliovascular Unit' Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, Unité Mixte de Recherche 7241 CNRS, Unité1050 INSERM, PSL Research University, Paris, France
| | - Colm Cunningham
- Trinity Biomedical Sciences Institute & Trinity College Institute of Neuroscience, School of Biochemistry & Immunology, Trinity College Dublin, Dublin, Republic of Ireland
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Bart De Strooper
- Center for Brain and Disease Research, VIB and University of Leuven, Leuven, Belgium
- UK Dementia Research Institute at the University College London, London, UK
| | - Blanca Díaz-Castro
- UK Dementia Research Institute at the University of Edinburgh, Centre for Discovery Brain Sciences, Edinburgh, UK
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe) and Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | | | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington DC, USA
| | - James E Goldman
- Department of Pathology & Cell Biology, Columbia University, New York, New York, USA
| | - Steven A Goldman
- University of Rochester Medical Center, Rochester, New York, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Science and Rigshospitalet, Kobenhavn N, Denmark
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet & Institute of Stem Cell Research, Helmholtz Center Munich, Munich, Germany
- Synergy, Excellence Cluster of Systems Neurology, Biomedical Center, Munich, Germany
| | - Antonia Gutiérrez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Dieter H Heiland
- Microenvironment and Immunology Research Laboratory, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurosurgery, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Matthew G Holt
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Masamitsu Iino
- Division of Cellular and Molecular Pharmacology, Nihon University School of Medicine, Tokyo, Japan
| | - Ksenia V Kastanenka
- Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Helmut Kettenmann
- Cellular Neurosciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science 55, Expo-ro, Yuseong-gu, Daejeon, Korea
| | - Shane A Liddelow
- Neuroscience Institute, Department of Neuroscience and Physiology, Department of Ophthalmology, NYU School of Medicine, New York, USA
| | - Brian A MacVicar
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pierre Magistretti
- Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Centre de Neurosciences Psychiatriques, University of Lausanne and CHUV, Site de Cery, Prilly-Lausanne, Lausanne, Switzerland
| | - Albee Messing
- Waisman Center and School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Anusha Mishra
- Department of Neurology Jungers Center for Neurosciences Research and Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Anna V Molofsky
- Departments of Psychiatry/Weill Institute for Neuroscience University of California, San Francisco, California, USA
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Seiji Okada
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Stéphane H R Oliet
- Université de Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux, France
| | - João F Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's -PT Government Associate Laboratory, Braga/Guimarães, Portugal
- IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence Laboratory, Campus of IPCA, Barcelos, Portugal
| | - Aude Panatier
- Université de Bordeaux, Inserm, Neurocentre Magendie, U1215, Bordeaux, France
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Luc Pellerin
- INSERM U1082, Université de Poitiers, Poitiers, France
| | - Gertrudis Perea
- Department of Functional and Systems Neurobiology, Cajal Institute, CSIC, Madrid, Spain
| | - Beatriz G Pérez-Nievas
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Frank W Pfrieger
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Kira E Poskanzer
- Department of Biochemistry & Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, California, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School. Associate Member, The Broad Institute, Boston, Massachusetts, USA
| | | | - Miriam Riquelme-Perez
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Stefanie Robel
- Fralin Biomedical Research Institute at Virginia Tech Carilion, School of Neuroscience Virginia Tech, Riverside Circle, Roanoke, Virginia, USA
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University, Düsseldorf, Germany
| | - Jeffrey D Rothstein
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, Paris, France
| | - David H Rowitch
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Swetlana Sirko
- Physiological Genomics, Biomedical Center, LMU Munich, Munich, Germany
- Institute for Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Harald Sontheimer
- Virginia Tech School of Neuroscience and Center for Glial Biology in Health, Disease and Cancer, Virginia Tech at the Fralin Biomedical Research Institute, Roanoke, Virginia, USA
| | - Raymond A Swanson
- Dept. of Neurology, University of California San Francisco and San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
| | - Javier Vitorica
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Dept. Bioquímica y Biología Molecular, Instituto de Biomedicina de Sevilla, Universidad de Sevilla, Hospital Virgen del Rocío/CSIC, Sevilla, Spain
| | - Ina-Beate Wanner
- Semel Institute for Neuroscience & Human Behavior, IDDRC, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, and Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Jiaqian Wu
- The Vivian L. Smith Department of Neurosurgery, Center for Stem Cell and Regenerative Medicine, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, McGovern Medical School, UTHealth, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Binhai Zheng
- Department of Neurosciences, UC San Diego School of Medicine, La Jolla; VA San Diego Research Service, San Diego, CA, USA
| | - Eduardo R Zimmer
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Robert Zorec
- Laboratory of Neuroendocrinology, Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
- Celica Biomedical, 1000, Ljubljana, Slovenia
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
70
|
De Carvalho LM, Wiers CE, Sun H, Wang G, Volkow ND. Increased transcription of TSPO, HDAC2, and HDAC6 in the amygdala of males with alcohol use disorder. Brain Behav 2021; 11:e01961. [PMID: 33216461 PMCID: PMC7882159 DOI: 10.1002/brb3.1961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Repeated exposure to high doses of alcohol triggers neuroinflammatory processes that contribute to craving and mood dysfunction in alcohol use disorder (AUD). The upregulation of the translocator protein (TSPO) is considered a biomarker of neuroinflammation, and TSPO ligands have been used as neuroimaging biomarkers of neuroinflammation. Epigenetic mechanisms are also implicated in neuroinflammatory responses to alcohol, and elevated expression of HDAC2 and HDAC6 has been reported in the brain of animals exposed to chronic alcohol. METHODS The present study examined the transcriptional regulation of TSPO, HDAC2, and HDAC6 in human postmortem brain tissue from males previously diagnosed with AUD (n = 11) compared to age-matched nondependent males (n = 13) in four brain regions relevant to AUD: prefrontal cortex (PFC), nucleus accumbens (NAc), hippocampus (HPP), and amygdala (AMY). RESULTS Translocator protein mRNA levels in AMY and PFC and HDAC2 and HDAC6 mRNA levels in AMY were upregulated in AUD compared to controls. In AMY, TSPO mRNA levels were positively associated with HDAC2 and HDAC6 mRNA levels, suggesting a possible regulation of TSPO by HDAC2 and HDAC6 in this brain region. In contrast, there were no group differences for TSPO, HDAC2, and HDAC6 in NAc and HPP. CONCLUSION Our study is the first to find upregulated TSPO mRNA levels in AMY and PFC in postmortem brains from AUD consistent with neuroinflammation, and in the amygdala, they implicate epigenetic regulation of TSPO by HDAC2 and HDAC6.
Collapse
Affiliation(s)
- Luana Martins De Carvalho
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
- Center for Alcohol Research in Epigenetics, Department of PsychiatryUniversity of Illinois at ChicagoChicagoILUSA
| | - Corinde E. Wiers
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
- Department of PsychiatryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Hui Sun
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
| | - Gene‐Jack Wang
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMDUSA
- National Institute on Drug AbuseNational Institutes of HealthRockvilleMDUSA
| |
Collapse
|
71
|
Lan N, Liu Y, Juan Z, Zhang R, Ma B, Xie K, Sun L, Feng H, Sun M, Liu J. The TSPO-specific Ligand PK11195 Protects Against LPS-Induced Cognitive Dysfunction by Inhibiting Cellular Autophagy. Front Pharmacol 2021; 11:615543. [PMID: 33708121 PMCID: PMC7941270 DOI: 10.3389/fphar.2020.615543] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/16/2020] [Indexed: 11/13/2022] Open
Abstract
Perioperative neurocognitive disorders (PND) is a common postoperative neurological complication. Neuroinflammation is a major cause that leads to PND. Autophagy, an intracellular process of lysosomal degradation, plays an important role in the development and maintenance of nervous system. PK11195 is a classic translocator protein (TSPO) ligand, which can improve the cognitive function of rats. In this study, we evaluate the protective effect of PK11195 on the learning and memory of rats. A rat model of lipopolysaccharide (LPS)-induced cognitive dysfunction was established by intraperitoneal injection of LPS. Morris Water Maze (MWM), Western blot, qRT-PCR, confocal microscopy and transmission electron microscopy (TEM) were used to study the role of TSPO-specific ligand PK11195 in LPS-activated mitochondrial autophagy in rat hippocampus. We found that PK11195 ameliorated LPS-induced learning and memory impairment, as indicated by decreased escape latencies, swimming distances and increased target quadrant platform crossing times and swimming times during MWM tests. TSPO, ATG7, ATG5, LC3B and p62 protein and mRNA expression increased in the hippocampus of PND model rats. The hippocampal microglia of PND model rats also have severe mitochondrial damage, and a large number of autophagosomes and phagocytic vesicles can be seen. PK11195 pretreatment significantly decreased the expression of TSPO, ATG7, ATG5, LC3B and p62 protein and mRNA, as well as mitochondrial damage. These findings suggested that PK11195 may alleviate the damage of LPS-induced cognitive dysfunction of rats by inhibiting microglia activation and autophagy.
Collapse
Affiliation(s)
- Nannan Lan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Yongxin Liu
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Zhaodong Juan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Rui Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Baoyu Ma
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Keliang Xie
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Lina Sun
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Hao Feng
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Meng Sun
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Jianfeng Liu
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China
| |
Collapse
|
72
|
Toppala S, Ekblad LL, Tuisku J, Helin S, Johansson JJ, Laine H, Löyttyniemi E, Marjamäki P, Blennow K, Zetterberg H, Jula A, Viitanen M, Rinne JO. Association of Early β-Amyloid Accumulation and Neuroinflammation Measured With [ 11C]PBR28 in Elderly Individuals Without Dementia. Neurology 2021; 96:e1608-e1619. [PMID: 33514647 PMCID: PMC8032368 DOI: 10.1212/wnl.0000000000011612] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
Objective To examine whether early β–amyloid (Aβ) accumulation and metabolic risk factors are associated with neuroinflammation in elderly individuals without dementia. Methods We examined 54 volunteers (mean age 70.0 years, 56% women, 51% APOE ɛ4 carriers) with the translocator protein (TSPO) tracer [11C]PBR28 to assess neuroinflammation and with [11C] Pittsburgh compound B (PiB) to assess cerebral Aβ accumulation. [11C]PBR28 and [11C]PiB standardized uptake value ratios (SUVRs) were quantified in 6 regions of interests by using the cerebellar cortex as a pseudo-reference and reference region, respectively. Fasting venous glucose, insulin, and high-sensitivity C-reactive protein (hs-CRP) values were determined. Homeostatic model assessment of insulin resistance (HOMA-IR) was calculated. A subset of individuals (n = 11) underwent CSF sampling, and Aβ40, Aβ42, total tau, phospho-tau, soluble TREM2, and YKL-40 levels were measured. Results Among the whole study group, no significant association was found between [11C]PiB and [11C]PBR28 SUVR composite scores (slope 0.02, p = 0.30). However, higher [11C]PiB binding was associated with higher [11C]PBR28 binding among amyloid-negative ([11C]PiB composite score ≤1.5) (TSPO genotype–, age- and sex-adjusted slope 0.26, p = 0.008) but not among amyloid-positive (slope −0.004, p = 0.88) participants. Higher CSF soluble TREM2 (rs = 0.72, p = 0.01) and YKL-40 (rs = 0.63, p = 0.04) concentrations were associated with a higher [11C]PBR28 composite score. Higher body mass index, HOMA-IR, and hs-CRP were associated with higher [11C]PBR28 binding in brain regions where Aβ accumulation is first detected in Alzheimer disease. Conclusions While there was no association between amyloid and neuroinflammation in the overall study group, neuroinflammation was associated with amyloid among the subgroup at early stages of amyloid pathology.
Collapse
Affiliation(s)
- Sini Toppala
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland.
| | - Laura L Ekblad
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Jouni Tuisku
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Semi Helin
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Jarkko J Johansson
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Hanna Laine
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Eliisa Löyttyniemi
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Päivi Marjamäki
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Kaj Blennow
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Henrik Zetterberg
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Antti Jula
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Matti Viitanen
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| | - Juha O Rinne
- From the Turku PET Centre (S.T., L.L.E., J.T., S.H., J.J., P.M., J.O.R.) and Department of Biostatistics (E.L.), University of Turku; Kuopio City Home Care (S.T.), Rehabilitation and Medical Services for Elderly, Kuopio, Finland; Amsterdam Alzheimer Center (L.L.E.), Amsterdam UMC, the Netherlands; Department of Radiation Sciences (J.J.), Umeå University, Sweden; City of Turku (H.L.), Welfare Division, Turku City Hospital, Turku, Finland; Department of Medicine (H.L.), University of Turku, Turku University Hospital, Finland; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease (H.Z.), UCL Institute of Neurology, Queen Square; UK Dementia Research Institute at UCL (H.Z.), London; National Institute for Health and Welfare (A.J.); Department of Geriatrics (M.V.), Turku City Hospital; University of Turku (M.V.), Finland; Division of Clinical Geriatrics (M.V.), NVS, Karolinska Institutet, Stockholm, Sweden; and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital, Finland
| |
Collapse
|
73
|
Rathitharan G, Truong J, Tong J, McCluskey T, Meyer JH, Mizrahi R, Warsh J, Rusjan P, Kennedy JL, Houle S, Kish SJ, Boileau I. Microglia imaging in methamphetamine use disorder: a positron emission tomography study with the 18 kDa translocator protein radioligand [F-18]FEPPA. Addict Biol 2021; 26:e12876. [PMID: 32017280 DOI: 10.1111/adb.12876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/08/2020] [Accepted: 01/12/2020] [Indexed: 01/19/2023]
Abstract
Activation of brain microglial cells, microgliosis, has been linked to methamphetamine (MA)-seeking behavior, suggesting that microglia could be a new therapeutic target for MA use disorder. Animal data show marked brain microglial activation following acute high-dose MA, but microglial status in human MA users is uncertain, with one positron emission tomography (PET) investigation reporting massively and globally increased translocator protein 18 kDa (TSPO; [C-11](R)-PK11195) binding, a biomarker for microgliosis, in MA users. Our aim was to measure binding of a second-generation TSPO radioligand, [F-18]FEPPA, in brain of human chronic MA users. Regional total volume of distribution (VT ) of [F-18]FEPPA was estimated with a two-tissue compartment model with arterial plasma input function for 10 regions of interest in 11 actively using MA users and 26 controls. A RM-ANOVA corrected for TSPO rs6971 polymorphism was employed to test significance. There was no main effect of group on [F-18]FEPPA VT (P = .81). No significant correlations between [F-18]FEPPA VT and MA use duration, weekly dosage, blood MA concentrations, regional brain volumes, and self-reported craving were observed. Our preliminary findings, consistent with our earlier postmortem data, do not suggest substantial brain microgliosis in MA use disorder but do not rule out microglia as a therapeutic target in MA addiction. Absence of increased [F-18]FEPPA TSPO binding might be related to insufficient MA dose or blunting of microglial response following repeated MA exposure, as suggested by some animal data.
Collapse
Affiliation(s)
- Gausiha Rathitharan
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Jennifer Truong
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Junchao Tong
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
| | - Tina McCluskey
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
| | - Jeffrey H. Meyer
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell 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
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Romina Mizrahi
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell 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
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Jerry Warsh
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell 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
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Pablo Rusjan
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Pharmacology and Toxicology University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - James L. Kennedy
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Sylvain Houle
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell 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 Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell 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
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| | - Isabelle Boileau
- Research Imaging Centre Centre for Addiction and Mental Health Toronto Ontario Canada
- Campbell Mental Health Research Institute Centre for Addiction and Mental Health Toronto Ontario Canada
- Department of Psychiatry University of Toronto Toronto Ontario Canada
- Institute of Medical Sciences University of Toronto Toronto Ontario Canada
| |
Collapse
|
74
|
Neuronal activity increases translocator protein (TSPO) levels. Mol Psychiatry 2021; 26:2025-2037. [PMID: 32398717 PMCID: PMC8440208 DOI: 10.1038/s41380-020-0745-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022]
Abstract
The mitochondrial protein, translocator protein (TSPO), is a widely used biomarker of neuroinflammation, but its non-selective cellular expression pattern implies roles beyond inflammatory processes. In the present study, we investigated whether neuronal activity modifies TSPO levels in the adult central nervous system. First, we used single-cell RNA sequencing to generate a cellular landscape of basal TSPO gene expression in the hippocampus of adult (12 weeks old) C57BL6/N mice, followed by confocal laser scanning microscopy to verify TSPO protein in neuronal and non-neuronal cell populations. We then quantified TSPO mRNA and protein levels after stimulating neuronal activity with distinct stimuli, including designer receptors exclusively activated by designer drugs (DREADDs), exposure to a novel environment and acute treatment with the psychostimulant drug, amphetamine. Single-cell RNA sequencing demonstrated a non-selective and multi-cellular gene expression pattern of TSPO at basal conditions in the adult mouse hippocampus. Confocal laser scanning microscopy confirmed that TSPO protein is present in neuronal and non-neuronal (astrocytes, microglia, vascular endothelial cells) cells of cortical (medial prefrontal cortex) and subcortical (hippocampus) brain regions. Stimulating neuronal activity through chemogenetic (DREADDs), physiological (novel environment exposure) or psychopharmacological (amphetamine treatment) approaches led to consistent increases in TSPO gene and protein levels in neurons, but not in microglia or astrocytes. Taken together, our findings show that neuronal activity has the potential to modify TSPO levels in the adult central nervous system. These findings challenge the general assumption that altered TSPO expression or binding unequivocally mirrors ongoing neuroinflammation and emphasize the need to consider non-inflammatory interpretations in some physiological or pathological contexts.
Collapse
|
75
|
Kong Y, Liu K, Hua T, Zhang C, Sun B, Guan Y. PET Imaging of Neutrophils Infiltration in Alzheimer's Disease Transgenic Mice. Front Neurol 2020; 11:523798. [PMID: 33362678 PMCID: PMC7758535 DOI: 10.3389/fneur.2020.523798] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 10/29/2020] [Indexed: 12/04/2022] Open
Abstract
Neutrophils are important components in the innate immune system. Neutrophil hyperactivation is regarded as a characteristic of Alzheimer's disease (AD). But in vivo imaging tools observing neutrophil activity in AD dynamically is lacking. This study aimed to identify neutrophil infiltration in AD transgenic mice. We used the AD triple-mutant transgenic mouse model and identified the genotype with RT-PCR. Behavioral experiments including an open-field test, a Morris water maze, and a Y-maze test were performed to evaluate the status of this AD model. 18F-AV45, 18F-PM-PBB3, 68Ga-PEG-cFLFLFK, and 18F-DPA714 were synthesized according to previous reports. We employed microPET to detect tracer uptake in the AD model and the control mice at different stages. Western blotting was used to observe the expression of functional proteins. We proved the successful establishment of AD models by RT-PCR, behavioral tests, and 18F-AV45 and 18F-PM-PBB3 PET imaging. We found an increased neutrophil accumulation in the brains of the AD mice through 68Ga-PEG-cFLFLFK PET imaging and Western blot assay. Our studies also demonstrated an elevated level of CAP37, which is produced by neutrophils, in the AD brain, and treatment with CAP37 promoted the expression of Iba1, iNOS, and COX-2 in BV2 cultures. Furthermore, our 18F-DPA714 PET imaging studies verified the raised activation of microglia in the brain of transgenic AD mice. Collectively, our findings indicate the increased activity of neutrophils in the brain and heart of AD model mice, 68Ga-PEG-cFLFLFK PET imaging represents a sensitive method to observe the status of neutrophils in AD, and infiltrated neutrophils can induce the activation of microglia by releasing CAP37 and blocking the activity of neutrophils may be beneficial for the control of AD progression.
Collapse
Affiliation(s)
- Yanyan Kong
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Kawai Liu
- Department of Mathematics, The Shanghai SMIC Private School, Shanghai, China
| | - Tao Hua
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
76
|
Systematic review and meta-analysis of human transcriptomics reveals neuroinflammation, deficient energy metabolism, and proteostasis failure across neurodegeneration. Neurobiol Dis 2020; 149:105225. [PMID: 33347974 PMCID: PMC7856076 DOI: 10.1016/j.nbd.2020.105225] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), Lewy body diseases (LBD), and the amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD) spectrum are defined by the accumulation of specific misfolded protein aggregates. However, the mechanisms by which each proteinopathy leads to neurodegeneration remain elusive. We hypothesized that there is a common "pan-neurodegenerative" gene expression signature driving pathophysiology across these clinically and pathologically diverse proteinopathies. To test this hypothesis, we performed a systematic review of human CNS transcriptomics datasets from AD, LBD, and ALS-FTD patients and age-matched controls in the Gene Expression Omnibus (GEO) and ArrayExpress databases, followed by consistent processing of each dataset, meta-analysis, pathway enrichment, and overlap analyses. After applying pre-specified eligibility criteria and stringent data pre-processing, a total of 2600 samples from 26 AD, 21 LBD, and 13 ALS-FTD datasets were included in the meta-analysis. The pan-neurodegenerative gene signature is characterized by an upregulation of innate immunity, cytoskeleton, and transcription and RNA processing genes, and a downregulation of the mitochondrial electron transport chain. Pathway enrichment analyses also revealed the upregulation of neuroinflammation (including Toll-like receptor, TNF, and NFκB signaling) and phagocytosis, and the downregulation of mitochondrial oxidative phosphorylation, lysosomal acidification, and ubiquitin-proteasome pathways. Our findings suggest that neuroinflammation and a failure in both neuronal energy metabolism and protein degradation systems are consistent features underlying neurodegenerative diseases, despite differences in the extent of neuronal loss and brain regions involved.
Collapse
|
77
|
Kreisl WC, Kim MJ, Coughlin JM, Henter ID, Owen DR, Innis RB. PET imaging of neuroinflammation in neurological disorders. Lancet Neurol 2020; 19:940-950. [PMID: 33098803 PMCID: PMC7912433 DOI: 10.1016/s1474-4422(20)30346-x] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/06/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022]
Abstract
A growing need exists for reliable in-vivo measurement of neuroinflammation to better characterise the inflammatory processes underlying various diseases and to inform the development of novel therapeutics that target deleterious glial activity. PET is well suited to quantify neuroinflammation and has the potential to discriminate components of the neuroimmune response. However, there are several obstacles to the reliable quantification of neuroinflammation by PET imaging. Despite these challenges, PET studies have consistently identified associations between neuroimmune responses and pathophysiology in brain disorders such as Alzheimer's disease. Tissue studies have also begun to clarify the meaning of changes in PET signal in some diseases. Furthermore, although PET imaging of neuroinflammation does not have an established clinical application, novel targets are under investigation and a small but growing number of studies have suggested that this imaging modality could have a role in drug development. Future studies are needed to further improve our knowledge of the cellular mechanisms that underlie changes in PET signal, how immune response contributes to neurological disease, and how it might be therapeutically modified.
Collapse
Affiliation(s)
- William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jennifer M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - David R Owen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
78
|
Microglia Implicated in Tauopathy in the Striatum of Neurodegenerative Disease Patients from Genotype to Phenotype. Int J Mol Sci 2020; 21:ijms21176047. [PMID: 32842621 PMCID: PMC7503242 DOI: 10.3390/ijms21176047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
We found interactions between dopamine and oxidative damage in the striatum involved in advanced neurodegeneration, which probably change the microglial phenotype. We observed possible microglia dystrophy in the striatum of neurodegenerative brains. To investigate the interactions between oxidative damage and microglial phenotype, we quantified myeloperoxidase (MPO), poly (ADP-Ribose) (PAR), and triggering receptors expressed on myeloid cell 2 (TREM2) using enzyme-linked immunosorbent assay (ELISA). To test the correlations of microglia dystrophy and tauopathy, we quantified translocator protein (TSPO) and tau fibrils using autoradiography. We chose the caudate and putamen of Lewy body diseases (LBDs) (Parkinson’s disease, Parkinson’s disease dementia, and Dementia with Lewy body), Alzheimer’s disease (AD), and control brains and genotyped for TSPO, TREM2, and bridging integrator 1 (BIN1) genes using single nucleotide polymorphisms (SNP) assays. TREM2 gene variants were absent across all samples. However, associations between TSPO and BIN1 gene polymorphisms and TSPO, MPO, TREM2, and PAR level variations were found. PAR levels reduced significantly in the caudate of LBDs. TSPO density and tau fibrils decreased remarkably in the striatum of LBDs but increased in AD. Oxidative damage, induced by misfolded tau proteins and dopamine metabolism, causes microglia dystrophy or senescence during the late stage of LBDs. Consequently, microglia dysfunction conversely reduces tau propagation. The G allele of the BIN1 gene is a potential risk factor for tauopathy.
Collapse
|
79
|
In Vivo TSPO Signal and Neuroinflammation in Alzheimer's Disease. Cells 2020; 9:cells9091941. [PMID: 32839410 PMCID: PMC7565089 DOI: 10.3390/cells9091941] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022] Open
Abstract
In the last decade, positron emission tomography (PET) and single-photon emission computed tomography (SPECT) in in vivo imaging has attempted to demonstrate the presence of neuroinflammatory reactions by measuring the 18 kDa translocator protein (TSPO) expression in many diseases of the central nervous system. We focus on two pathological conditions for which neuropathological studies have shown the presence of neuroinflammation, which translates in opposite in vivo expression of TSPO. Alzheimer's disease has been the most widely assessed with more than forty preclinical and clinical studies, showing overall that TSPO is upregulated in this condition, despite differences in the topography of this increase, its time-course and the associated cell types. In the case of schizophrenia, a reduction of TSPO has instead been observed, though the evidence remains scarce and contradictory. This review focuses on the key characteristics of TSPO as a biomarker of neuroinflammation in vivo, namely, on the cellular origin of the variations in its expression, on its possible biological/pathological role and on its variations across disease phases.
Collapse
|
80
|
Riban V, Meunier J, Buttigieg D, Villard V, Verleye M. In Vitro and In Vivo Neuroprotective Effects of Etifoxine in β-Amyloidinduced Toxicity Models. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:227-240. [DOI: 10.2174/1871527319666200601151007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 11/22/2022]
Abstract
Aim:
The aim of this study is to examine the effect of etifoxine on β-amyloid-induced toxicity
models.
Background:
Etifoxine is an anxiolytic compound with a dual mechanism of action; it is a positive allosteric
modulator of GABAergic receptors as well as a ligand for the 18 kDa mitochondrial Translocator
Protein (TSPO). TSPO has recently raised interest in Alzheimer’s Disease (AD), and experimental studies
have shown that some TSPO ligands could induce neuroprotective effects in animal models.
Objective:
In this study, we examined the potential protective effect of etifoxine in an in vitro and an
in vivo model of amyloid beta (Aβ)-induced toxicity in its oligomeric form, which is a crucial factor in
AD pathologic mechanisms.
Method:
Neuronal cultures were intoxicated with Aβ1-42, and the effects of etifoxine on oxidative
stress, Tau-hyperphosphorylation and synaptic loss were quantified. In a mice model, behavioral deficits
induced by intracerebroventricular administration of Aβ25-35 were measured in a spatial memory
test, the spontaneous alternation and in a contextual memory test, the passive avoidance test.
Results:
In neuronal cultures intoxicated with Aβ1-42, etifoxine dose-dependently decreased oxidative
stress (methionine sulfoxide positive neurons), tau-hyperphosphorylation and synaptic loss (ratio
PSD95/synaptophysin). In a mice model, memory impairments were fully alleviated by etifoxine administered
at anxiolytic doses (12.5-50mg/kg). In addition, markers of oxidative stress and apoptosis
were decreased in the hippocampus of these animals.
Conclusion:
Our results have shown that in these two models, etifoxine could fully prevent neurotoxicity
and pathological changes induced by Aβ. These results confirm that TSPO ligands could offer an
interesting therapeutic approach to Alzheimer’s disease.
Collapse
Affiliation(s)
- Veronique Riban
- Pharmacology Department, Biocodex, 3 Chemin d’Armancourt, 60200 Compiegne, France
| | - Johann Meunier
- Amylgen, 2196 Boulevard de la Lironde, 34980 Montferrier sur Lez, France
| | | | - Vanessa Villard
- Amylgen, 2196 Boulevard de la Lironde, 34980 Montferrier sur Lez, France
| | - Marc Verleye
- Pharmacology Department, Biocodex, 3 Chemin d’Armancourt, 60200 Compiegne, France
| |
Collapse
|
81
|
Ismail R, Parbo P, Madsen LS, Hansen AK, Hansen KV, Schaldemose JL, Kjeldsen PL, Stokholm MG, Gottrup H, Eskildsen SF, Brooks DJ. The relationships between neuroinflammation, beta-amyloid and tau deposition in Alzheimer's disease: a longitudinal PET study. J Neuroinflammation 2020; 17:151. [PMID: 32375809 PMCID: PMC7203856 DOI: 10.1186/s12974-020-01820-6] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 04/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The aim of this longitudinal study was to assess with positron emission tomography (PET) the relationship between levels of inflammation and the loads of aggregated β-amyloid and tau at baseline and again after 2 years in prodromal Alzheimer's disease. METHODS Forty-three subjects with mild cognitive impairment (MCI) had serial 11C-PK11195 PET over 2 years to measure inflammation changes, and 11C-PiB PET to determine β-amyloid fibril load; 22 also had serial 18F-Flortaucipir PET to determine tau tangle load. Cortical surface statistical mapping was used to localise areas showing significant changes in tracer binding over time and to interrogate correlations between tracer binding of the tracers at baseline and after 2 years. RESULTS Those MCI subjects with high 11C-PiB uptake at baseline (classified as prodromal Alzheimer's disease) had raised inflammation levels which significantly declined across cortical regions over 2 years although their β-amyloid levels continued to rise. Those MCI cases who had low/normal 11C-PiB uptake at baseline but their levels then rose over 2 years were classified as prodromal AD with low Thal phase 1-2 amyloid deposition at baseline. They showed levels of cortical inflammation which correlated with their rising β-amyloid load. Those MCI cases with baseline low 11C-PiB uptake that remained stable were classified as non-AD, and they showed no correlated inflammation levels. Finally, MCI cases which showed both high 11C-PiB and 18F-Flortaucipir uptake at baseline (MCI due to AD) showed a further rise in their tau tangle load over 2 years with a correlated rise in levels of inflammation. CONCLUSIONS Our baseline and 2-year imaging findings are compatible with a biphasic trajectory of inflammation in Alzheimer's disease: MCI cases with low baseline but subsequently rising β-amyloid load show correlated levels of microglial activation which then later decline when the β-amyloid load approaches AD levels. Later, as tau tangles form in β-amyloid positive MCI cases with prodromal AD, the rising tau load is associated with higher levels of inflammation.
Collapse
Affiliation(s)
- Rola Ismail
- Department of Clinical Medicine, PET-Centre, Aarhus University, Aarhus, Denmark.
| | - Peter Parbo
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, DK-8200, Aarhus N, Denmark
| | | | - Allan K Hansen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, DK-8200, Aarhus N, Denmark
| | - Kim V Hansen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, DK-8200, Aarhus N, Denmark
| | - Jeppe L Schaldemose
- Department of Clinical Medicine, PET-Centre, Aarhus University, Aarhus, Denmark
| | - Pernille L Kjeldsen
- Department of Clinical Medicine, PET-Centre, Aarhus University, Aarhus, Denmark
| | - Morten G Stokholm
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, DK-8200, Aarhus N, Denmark
| | - Hanne Gottrup
- Dept. of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Simon F Eskildsen
- Centre of Functionally Integrative Neuroscience (CFIN), Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Clinical Medicine, PET-Centre, Aarhus University, Aarhus, Denmark
- Institute of Neuroscience, University of Newcastle upon Tyne, Tyne, UK
- Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
82
|
Malpetti M, Kievit RA, Passamonti L, Jones PS, Tsvetanov KA, Rittman T, Mak E, Nicastro N, Bevan-Jones WR, Su L, Hong YT, Fryer TD, Aigbirhio FI, O’Brien JT, Rowe JB. Microglial activation and tau burden predict cognitive decline in Alzheimer's disease. Brain 2020; 143:1588-1602. [PMID: 32380523 PMCID: PMC7241955 DOI: 10.1093/brain/awaa088] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 02/07/2020] [Indexed: 11/12/2022] Open
Abstract
Tau pathology, neuroinflammation, and neurodegeneration are key aspects of Alzheimer's disease. Understanding whether these features predict cognitive decline, alone or in combination, is crucial to develop new prognostic measures and enhanced stratification for clinical trials. Here, we studied how baseline assessments of in vivo tau pathology (measured by 18F-AV-1451 PET), neuroinflammation (measured by 11C-PK11195 PET) and brain atrophy (derived from structural MRI) predicted longitudinal cognitive changes in patients with Alzheimer's disease pathology. Twenty-six patients (n = 12 with clinically probable Alzheimer's dementia and n = 14 with amyloid-positive mild cognitive impairment) and 29 healthy control subjects underwent baseline assessment with 18F-AV-1451 PET, 11C-PK11195 PET, and structural MRI. Cognition was examined annually over the subsequent 3 years using the revised Addenbrooke's Cognitive Examination. Regional grey matter volumes, and regional binding of 18F-AV-1451 and 11C-PK11195 were derived from 15 temporo-parietal regions characteristically affected by Alzheimer's disease pathology. A principal component analysis was used on each imaging modality separately, to identify the main spatial distributions of pathology. A latent growth curve model was applied across the whole sample on longitudinal cognitive scores to estimate the rate of annual decline in each participant. We regressed the individuals' estimated rate of cognitive decline on the neuroimaging components and examined univariable predictive models with single-modality predictors, and a multi-modality predictive model, to identify the independent and combined prognostic value of the different neuroimaging markers. Principal component analysis identified a single component for the grey matter atrophy, while two components were found for each PET ligand: one weighted to the anterior temporal lobe, and another weighted to posterior temporo-parietal regions. Across the whole-sample, the single-modality models indicated significant correlations between the rate of cognitive decline and the first component of each imaging modality. In patients, both stepwise backward elimination and Bayesian model selection revealed an optimal predictive model that included both components of 18F-AV-1451 and the first (i.e. anterior temporal) component for 11C-PK11195. However, the MRI-derived atrophy component and demographic variables were excluded from the optimal predictive model of cognitive decline. We conclude that temporo-parietal tau pathology and anterior temporal neuroinflammation predict cognitive decline in patients with symptomatic Alzheimer's disease pathology. This indicates the added value of PET biomarkers in predicting cognitive decline in Alzheimer's disease, over and above MRI measures of brain atrophy and demographic data. Our findings also support the strategy for targeting tau and neuroinflammation in disease-modifying therapy against Alzheimer's disease.
Collapse
Affiliation(s)
- Maura Malpetti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Rogier A Kievit
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Institute of Molecular Bioimaging and Physiology, National Research Council, Milano, Italy
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Kamen A Tsvetanov
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Nicolas Nicastro
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, Geneva University Hospitals, Switzerland
| | | | - Li Su
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Trust, Cambridge, UK
| |
Collapse
|
83
|
Fu Y, Wang D, Wang H, Cai M, Li C, Zhang X, Chen H, Hu Y, Zhang X, Ying M, He W, Zhang J. TSPO deficiency induces mitochondrial dysfunction, leading to hypoxia, angiogenesis, and a growth-promoting metabolic shift toward glycolysis in glioblastoma. Neuro Oncol 2020; 22:240-252. [PMID: 31563962 PMCID: PMC7442372 DOI: 10.1093/neuonc/noz183] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The ligands of mitochondrial translocator protein (TSPO) have been widely used as diagnostic biomarkers for glioma. However, the true biological actions of TSPO in vivo and its role in glioma tumorigenesis remain elusive. METHODS TSPO knockout xenograft and spontaneous mouse glioma models were employed to assess the roles of TSPO in the pathogenesis of glioma. A Seahorse Extracellular Flux Analyzer was used to evaluate mitochondrial oxidative phosphorylation and glycolysis in TSPO knockout and wild-type glioma cells. RESULTS TSPO deficiency promoted glioma cell proliferation in vitro in mouse GL261 cells and patient-derived stem cell-like GBM1B cells. TSPO knockout increased glioma growth and angiogenesis in intracranial xenografts and a mouse spontaneous glioma model. Loss of TSPO resulted in a greater number of fragmented mitochondria, increased glucose uptake and lactic acid conversion, decreased oxidative phosphorylation, and increased glycolysis. CONCLUSION TSPO serves as a key regulator of glioma growth and malignancy by controlling the metabolic balance between mitochondrial oxidative phosphorylation and glycolysis.1. TSPO deficiency promotes glioma growth and angiogenesis.2. TSPO regulates the balance between mitochondrial oxidative phosphorylation and glycolysis.
Collapse
Affiliation(s)
- Yi Fu
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Dongdong Wang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Huaishan Wang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Menghua Cai
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Chao Li
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Xue Zhang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Hui Chen
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Yu Hu
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingyao Ying
- Hugo W. Moser Research Institute at Kennedy Krieger, and Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Wei He
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| | - Jianmin Zhang
- Department of Immunology, Research Center on Pediatric Development and Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, China
| |
Collapse
|
84
|
Metaxas A, Thygesen C, Briting SRR, Landau AM, Darvesh S, Finsen B. Increased Inflammation and Unchanged Density of Synaptic Vesicle Glycoprotein 2A (SV2A) in the Postmortem Frontal Cortex of Alzheimer's Disease Patients. Front Cell Neurosci 2019; 13:538. [PMID: 31866830 PMCID: PMC6906198 DOI: 10.3389/fncel.2019.00538] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/20/2019] [Indexed: 01/29/2023] Open
Abstract
Sections from the middle frontal gyrus (Brodmann area 46) of autopsy-confirmed Alzheimer's disease (AD) patients and non-demented subjects were examined for the prevalence of hallmark AD pathology, including amyloid-β (Aβ) plaques, phosphorylated tau (pTau) tangles, neuroinflammation and synaptic loss (n = 7 subjects/group). Dense-core deposits of Aβ were present in all AD patients (7/7) and some non-demented subjects (3/7), as evidenced by 6E10 immunohistochemistry. Levels of Aβ immunoreactivity were higher in AD vs. non-AD cases. For pTau, AT8-positive neurofibrillary tangles and threads were exclusively observed in AD patient tissue. Levels of [3H]PK11195 binding to the translocator protein (TSPO), a marker of inflammatory processes, were elevated in the gray matter of AD patients compared to non-demented subjects. Levels of [3H]UCB-J binding to synaptic vesicle glycoprotein 2A (SV2A), a marker of synaptic density, were not different between groups. In AD patients, pTau immunoreactivity was positively correlated with [3H]PK11195, and negatively correlated with [3H]UCB-J binding levels. No correlation was observed between Aβ immunoreactivity and markers of neuroinflammation or synaptic density. These data demonstrate a close interplay between tau pathology, inflammation and SV2A density in AD, and provide useful information on the ability of neuroimaging biomarkers to diagnose AD dementia.
Collapse
Affiliation(s)
- Athanasios Metaxas
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Camilla Thygesen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sanne R R Briting
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET-Center, Aarhus University, Aarhus, Denmark
| | - Sultan Darvesh
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.,Department of Medicine, Neurology, and Geriatric Medicine, Dalhousie University, Halifax, NS, Canada
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|