1
|
Tournier BB, Mansouri Z, Salimi Y, Ceyzériat K, Mathoux G, Richard-Lepouriel H, Zullino D, Bois F, Zaidi H, Garibotto V, Tsartsalis S, Millet P. Radiation dosimetry of the 18 kDa translocator protein ligand [ 18F]PBR111 in humans. Nucl Med Biol 2025; 144-145:109011. [PMID: 40179687 DOI: 10.1016/j.nucmedbio.2025.109011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/12/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE The 18 kDa translocator protein (TSPO) is a mitochondrial protein that becomes overexpressed during neuroinflammatory conditions, such as in Alzheimer's disease or multiple sclerosis. TSPO is of interest because it serves as a marker for microglial and astrocytic activity, measurable via in vivo positron emission tomography (PET) molecular imaging. [18F]PBR111 is a second-generation TSPO PET radioligand with high signal specificity but a sensitivity to TSPO polymorphism, in comparison with first-generation ligands. This study focused on the biodistribution and dosimetry of [18F]PBR111 in healthy humans. METHOD Six volunteers (three males, three females) were administered approximately 200 MBq of [18F]PBR111. Organs such as the lungs and liver showed the highest initial radioactivity level, while the bone marrow and bladder accumulated activity over time, likely reflecting ligand defluorination and elimination. RESULTS Dosimetry findings revealed a total effective dose of 16.17 μSv/MBq, equivalent to 3.04 mSv per examination. Compared to animal models, human dosimetry showed lower radiation exposure, highlighting discrepancies in predictive models. Organ-specific dose comparisons with other TSPO ligands ([18F]PBR06, [18F]FEPPA, [18F]FEDAA1106) revealed similar distribution patterns. This study underscores the clinical viability of [18F]PBR111 for TSPO imaging, providing critical data for optimizing its safe use in research and clinical settings. CONCLUSION The findings support its potential for studying neuroinflammatory and systemic diseases. The trial registration number is NCT06398392.
Collapse
Affiliation(s)
- Benjamin B Tournier
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Zahra Mansouri
- Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
| | - Yazdan Salimi
- Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
| | | | - Gregory Mathoux
- Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
| | - Hélène Richard-Lepouriel
- Department of Psychiatry, Mood and Anxiety Disorders Unit, University Hospitals of Geneva, Geneva, Switzerland
| | - Daniel Zullino
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; University Hospitals of Geneva, Geneva, Switzerland
| | - Frédéric Bois
- Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
| | - Habib Zaidi
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
| | - Valentina Garibotto
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diagnostic Department, Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
| | - Stergios Tsartsalis
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, Mood and Anxiety Disorders Unit, University Hospitals of Geneva, Geneva, Switzerland
| | - Philippe Millet
- Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland.
| |
Collapse
|
2
|
Nakagawa K, Kodama K, Nagata W, Takahashi S, Satoh Y, Ishizuka T. Molecular hydrogen inhibits neuroinflammation and ameliorates depressive-like behaviors and short-term cognitive impairment in senescence-accelerated mouse prone 8 mice. Behav Brain Res 2025; 478:115330. [PMID: 39522774 DOI: 10.1016/j.bbr.2024.115330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/11/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND AIMS Neuroinflammation, a low-grade chronic inflammation of the central nervous system, is linked to age-related neuropsychiatric disorders such as senile depression and Alzheimer's disease. Recent studies have explored controlling neuroinflammation as a novel treatment strategy. Molecular hydrogen shows anti-inflammatory effects. However, its impacts on neuroinflammation and age-related neuropsychiatric disorders remain unelucidated. We investigated molecular hydrogen's effects on microglial activation, neuroinflammation, depressive-like behavior, and short-term cognitive decline in senescence-accelerated mouse-prone 8 (SAMP8) mice. METHODS Six-week-old SAMP8 or senescence-accelerated mouse-resistant 1 (SAMR1) mice received hydrogen-rich jelly (HRJ) or placebo jelly (PJ) from six weeks of age for 26-28 weeks. Depressive-like behavior was assessed using tail suspension and forced swimming tests, while cognitive function was evaluated using the Y-maze and object recognition tests. Brain tissues were used for immunohistochemical studies or to measure pro-inflammatory cytokine levels via enzyme-linked immunosorbent assay (ELISA). RESULTS HRJ intake reduced immobility time in both tail suspension and forced swimming tests and enhanced visual cognitive and spatial working memory in SAMP8 mice. Additionally, HRJ intake suppressed the 8-hydroxy-2'-deoxyguanosine (8-OHdG), Iba1, and cleaved caspase 3 expression levels in the medial prefrontal cortex and hippocampal dentate gyrus. Furthermore, HRJ intake significantly lowered IL-6 levels in brain tissues of SAMP8 mice. CONCLUSIONS These findings suggest that molecular hydrogen treatment may regulate neuroinflammation induced by activated microglia and improve depressive-like behavior and short-term cognitive impairment in SAMP8 mice.
Collapse
Affiliation(s)
- Keiichi Nakagawa
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Kayoko Kodama
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Wataru Nagata
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Sayaka Takahashi
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Yasushi Satoh
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan
| | - Toshiaki Ishizuka
- Department of Pharmacology, National Defense Medical College, Tokorozawa, Saitama 359-0042, Japan.
| |
Collapse
|
3
|
Ogata A, Ikenuma H, Yasuno F, Nihashi T, Hattori S, Sato Y, Ichise M, Ito K, Kato T, Kimura Y. First-in-Human Study of [ 11C]NCGG401 for Imaging Colony-Stimulating Factor 1 Receptors in the Brain. J Nucl Med 2025; 66:302-308. [PMID: 39746757 DOI: 10.2967/jnumed.124.268699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/14/2024] [Indexed: 01/04/2025] Open
Abstract
Microglia, the immune cells in the brain, play a significant role in the pathophysiology of neurodegenerative diseases. To visualize these cells in the living brain, we developed a PET ligand, [11C]NCGG401 (4-{2-[((1R,2R)-2-hydroxycyclohexyl)(methyl)amino]benzothiazol-6-yloxy}-N-methylpicolinamide, NCGG401), that targets colony-stimulating factor 1 receptor (CSF1R). In this study, we present the first-in-human evaluation of [11C]NCGG401 to assess its safety profile and then to evaluate its kinetics to quantify CSF1R in the human brain. Methods: Head to upper thigh PET scans were conducted in 3 healthy men to estimate the effective dose of [11C]NCGG401. Brain PET scans were performed on 6 healthy men, combined with arterial blood sampling and metabolite analyses. Compartmental and graphical models were used to quantify CSF1R in the human brain. [11C]NCGG401 PET data were indirectly compared with regional CSF1R protein levels after death that were reported in a proteomics study. In addition, the results of this study were directly compared with the PET imaging of 18-kDa translocator protein using [11C]DPA-713 (N,N-diethyl-2-[2-(4-methoxyphenyl)-5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-yl]acetamide, DPA-713). Results: The administration of [11C]NCGG401 did not result in severe adverse events. The effective doses per injected activity were 5.1 ± 0.2 µSv/MBq for men and 6.1 ± 0.3 µSv/MBq for women. [11C]NCGG401 demonstrated good brain permeability, with peak uptake reaching an SUV of 3. Regional total distribution volumes were reliably quantified using the 2-tissue compartment model and a Logan plot with 60 min of scan data. The resulting parametric images reflected the known distribution of CSF1R in the brain. Furthermore, regional total distribution volume values of [11C]NCGG401 showed good correlation with regional CSF1R protein levels. The [11C]NCGG401 images showed regional distributions different from those of [11C]DPA-713. Conclusion: [11C]NCGG401 images appear to reflect regional microglia-specific distributions of CSF1R in the brain, consistent with the findings of a CSF1R proteomics study by others. However, ultimate confirmation of specific CSF1R binding should be validated by evaluating, in suitable preclinical or human experiments, pharmacologic blockade of its binding in the brain in vivo.
Collapse
Affiliation(s)
- Aya Ogata
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Pharmacy, Faculty of Pharmacy, Gifu University of Medical Science, Kani, Japan
| | - Hiroshi Ikenuma
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Fumihiko Yasuno
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
- National Center for Geriatrics and Gerontology, National Hospital for Geriatric Medicine, Obu, Japan; and
| | - Takashi Nihashi
- National Center for Geriatrics and Gerontology, National Hospital for Geriatric Medicine, Obu, Japan; and
| | - Saori Hattori
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yayoi Sato
- Innovation Center for Translational Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Masanori Ichise
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kengo Ito
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yasuyuki Kimura
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan;
| |
Collapse
|
4
|
Eiff B, Bullmore ET, Clatworthy MR, Fryer TD, Pariante CM, Mondelli V, Maccioni L, Hadjikhani N, Loggia ML, Moskowitz MA, Bruner E, Veronese M, Turkheimer FE, Schubert JJ. Extra-axial inflammatory signal and its relationship to peripheral and central immunity in depression. Brain 2025; 148:635-646. [PMID: 39657983 PMCID: PMC11788198 DOI: 10.1093/brain/awae343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/13/2024] [Accepted: 10/06/2024] [Indexed: 12/12/2024] Open
Abstract
Although both central and peripheral inflammation have been observed consistently in depression, the relationship between the two remains obscure. Extra-axial immune cells may play a role in mediating the connection between central and peripheral immunity. This study investigates the potential roles of calvarial bone marrow and parameningeal spaces in mediating interactions between central and peripheral immunity in depression. PET was used to measure regional TSPO expression in the skull and parameninges as a marker of inflammatory activity. This measure was correlated with brain TSPO expression and peripheral cytokine concentrations in a cohort enriched for heightened peripheral and central immunity comprising 51 individuals with depression and 25 healthy controls. The findings reveal a complex relationship between regional skull TSPO expression and both peripheral and central immunity. Facial and parietal skull bone TSPO expression showed significant associations with both peripheral and central immunity. TSPO expression in the confluence of sinuses was also linked to both central and peripheral immune markers. Group-dependent elevations in TSPO expression within the occipital skull bone marrow were also found to be significantly associated with central inflammation. Significant associations between immune activity within the skull, parameninges, parenchyma and periphery highlight the role of the skull bone marrow and venous sinuses as pivotal sites for peripheral and central immune interactions.
Collapse
Affiliation(s)
- Brandi Eiff
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge CB21 5EF, UK
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK
- Cambridge University Hospitals NHS Foundation Trust, and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
| | - Lucia Maccioni
- Department of Information Engineering, University of Padova, 35131 Padova, Italy
| | - Nouchine Hadjikhani
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Michael A Moskowitz
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Emiliano Bruner
- Department of Paleobiology, Museo Nacional de Ciencias Naturales (CSIC), 28006 Madrid, Spain
- Alzheimer Center Reina Sofía, CIEN Foundation, ISCIII, 28031 Madrid, Spain
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Information Engineering, University of Padova, 35131 Padova, Italy
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
| | - Julia J Schubert
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London SE5 8AF, UK
| |
Collapse
|
5
|
Rogeau A, Boer AJ, Guedj E, Sala A, Sommer IE, Veronese M, van der Weijden-Germann M, Fraioli F. EANM perspective on clinical PET and SPECT imaging in schizophrenia-spectrum disorders: a systematic review of longitudinal studies. Eur J Nucl Med Mol Imaging 2025; 52:876-899. [PMID: 39576337 PMCID: PMC11754335 DOI: 10.1007/s00259-024-06987-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/08/2024] [Indexed: 01/03/2025]
Abstract
PURPOSE There is a need for biomarkers in psychiatry to improve diagnosis, prognosis and management, and with confirmed value in follow-up care. Radionuclide imaging, given its molecular imaging characteristics, is well-positioned for translation to the clinic. This systematic review lays the groundwork for integrating PET and SPECT imaging in the clinical management of schizophrenia-spectrum disorders. METHODS Systematic search of PubMed, Embase, Web of Science and Cochrane library databases was conducted from the earliest date available until February 2024. The focus was on longitudinal studies evaluating PET or SPECT imaging in individuals with a schizophrenia-spectrum or another psychotic disorders. Quality assessment was done using the Newcastle-Ottawa Scale (NOS), NIH scale for before-after studies and Cochrane Risk of Bias tool version 2 (Cochrane RoB2). Studies were further categorised into three groups: preclinical and diagnosis, predicting disease course or personalising treatment. RESULTS Fifty-six studies were included in the systematic review investigating in total 1329 patients over a median of 3 months. Over two-thirds used PET tracers, whereas the remaining studies employed SPECT tracers. The most frequently investigated system was dopaminergic transmission, followed by cerebral metabolism and blood flow. [18F]FDOPA demonstrated large effect size in predicting conversion of subjects at risk and treatment response. Additionally, treatment dosage could be optimised to reduce side effects using [123I]IBZM or [11C]raclopride. CONCLUSION Molecular imaging holds significant promise for real-life application in schizophrenia, with two particularly encouraging avenues being the prediction of conversion/response to antipsychotic medication and the improved management of antipsychotic dosage. Further longitudinal studies and clinical trials will be essential for validating both the clinical effectiveness and economic sustainability, as well as for exploring new applications.
Collapse
Affiliation(s)
- Antoine Rogeau
- Department of Nuclear Medicine, Lille University Hospital, Lille, France.
| | - Anne Jetske Boer
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eric Guedj
- Department of Nuclear Medicine, Aix Marseille Univ, APHM, CNRS, Centrale Marseille, Institut Fresnel, Hôpital de La Timone, CERIMED, Marseille, France
| | - Arianna Sala
- Coma Science Group, GIGA-Consciousness, University Hospital of Liège, Liège, Belgium
| | - Iris E Sommer
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mattia Veronese
- Department of Information Engineering, University of Padua, Padua, Italy
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - Francesco Fraioli
- Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
6
|
Emvalomenos GM, Kang JW, Salberg S, Li C, Jupp B, Long M, Haskali MB, Kellapatha S, Davanzo OII, Lim H, Mychasiuk R, Keay KA, Henderson LA. Evidence for glial reactivity using positron-emission tomography imaging of translocator Protein-18 kD [TSPO] in both sham and nerve-injured rats in a preclinical model of orofacial neuropathic pain. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2025; 17:100175. [PMID: 39758133 PMCID: PMC11699482 DOI: 10.1016/j.ynpai.2024.100175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 12/08/2024] [Accepted: 12/08/2024] [Indexed: 01/07/2025]
Abstract
Chronic neuropathic pain is a debilitating condition that results from damage to the nervous system. Current treatments are largely ineffective, with limited understanding of the underlying mechanisms hindering development of effective treatments. Preclinical models of neuropathic pain have revealed that non-neural changes are important for the development of neuropathic pain, although these data are derived almost exclusively from post-mortem histological analyses. Although these static snapshots have provided valuable data, they cannot provide insights into non-neural cell changes that could be also assessed in human patients with chronic pain. In this study we used translocator protein 18 kDa (TSPO) PET imaging with [18F]PBR06 to visualise in-vivo, the activity of macrophages and microglia in a rodent preclinical model of trigeminal neuropathic pain. Using chronic constriction injury of the infraorbital nerve (ION-CCI) we compared temporal changes in TSPO binding in male rats, prior to, and up to 28 days after ION-CCI compared with both sham-injured and naïve counterparts. Unexpectedly, we found significant increases in TSPO signal in both ION-CCI and sham-injured rats within the trigeminal ganglion, spinal trigeminal nucleus and paratrigeminal nucleus during the initial phase following surgery and/or nerve injury. This increased TSPO binding returned to control levels by day 28. Qualitative histological appraisal of macrophage accumulation and glial reactivity in both ION-CCI and sham-injured rats indicated macrophage accumulation in the trigeminal ganglion and microglial reactivity in the brainstem trigeminal complex. These findings show, glial changes in the peripheral nerve and brain in both nerve-injured and sham-injured rats in a preclinical model of neuropathic pain which provides a platform for translation into human patients.
Collapse
Affiliation(s)
- Gaelle M. Emvalomenos
- School of Medical Sciences [Neuroscience], and the Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - James W.M. Kang
- School of Medical Sciences [Neuroscience], and the Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Crystal Li
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Bianca Jupp
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Matthew Long
- The Radiopharmaceutical Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Mohammad B. Haskali
- The Radiopharmaceutical Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Sunil Kellapatha
- The Radiopharmaceutical Research Laboratory, The Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - OIivia I. Davanzo
- School of Medical Sciences [Neuroscience], and the Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - Hyunsol Lim
- School of Medical Sciences [Neuroscience], and the Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Kevin A. Keay
- School of Medical Sciences [Neuroscience], and the Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - Luke A. Henderson
- School of Medical Sciences [Neuroscience], and the Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| |
Collapse
|
7
|
Chauveau F, Winkeler A, Chalon S, Boutin H, Becker G. PET imaging of neuroinflammation: any credible alternatives to TSPO yet? Mol Psychiatry 2025; 30:213-228. [PMID: 38997465 DOI: 10.1038/s41380-024-02656-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Over the last decades, the role of neuroinflammation in neuropsychiatric conditions has attracted an exponentially growing interest. A key driver for this trend was the ability to image brain inflammation in vivo using PET radioligands targeting the Translocator Protein 18 kDa (TSPO), which is known to be expressed in activated microglia and astrocytes upon inflammatory events as well as constitutively in endothelial cells. TSPO is a mitochondrial protein that is expressed mostly by microglial cells upon activation but is also expressed by astrocytes in some conditions and constitutively by endothelial cells. Therefore, our current understanding of neuroinflammation dynamics is hampered by the lack of alternative targets available for PET imaging. We performed a systematic search and review on radiotracers developed for neuroinflammation PET imaging apart from TSPO. The following targets of interest were identified through literature screening (including previous narrative reviews): P2Y12R, P2X7R, CSF1R, COX (microglial targets), MAO-B, I2BS (astrocytic targets), CB2R & S1PRs (not specific of a single cell type). We determined the level of development and provided a scoping review for each target. Strikingly, astrocytic biomarker MAO-B has progressed in clinical investigations the furthest, while few radiotracers (notably targeting S1P1Rs, CSF1R) are being implemented in clinical investigations. Other targets such as CB2R and P2X7R have proven disappointing in clinical studies (e.g. poor signal, lack of changes in disease conditions, etc.). While astrocytic targets are promising, development of new biomarkers and tracers specific for microglial activation has proven challenging.
Collapse
Affiliation(s)
- Fabien Chauveau
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
| | - Alexandra Winkeler
- Université Paris-Saclay, Inserm, CNRS, CEA, BioMaps, Service Hospitalier Frédéric Joliot, 4 place du général Leclerc, 91401, Orsay, France
| | - Sylvie Chalon
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France
| | - Hervé Boutin
- UMR 1253 iBrain, Université de Tours - INSERM, Bâtiment Planiol, UFR de Médecine, 10 Boulevard Tonnellé, 37032, Tours, Cedex 01, France.
| | - Guillaume Becker
- Université Claude Bernard Lyon 1, Centre de Recherche en Neurosciences de Lyon, Inserm U1028, CNRS UMR5292, BIORAN, Groupement Hospitalier Est - CERMEP, 59 boulevard Pinel, 69677, Bron, Cedex, France
- Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail, 14 rue Pierre et Marie Curie, 94701, Maisons-Alfort, Cedex, France
| |
Collapse
|
8
|
Garland EF, Antony H, Kulagowska L, Scott T, Rogien C, Bottlaender M, Nicoll JAR, Boche D. The microglial translocator protein (TSPO) in Alzheimer's disease reflects a phagocytic phenotype. Acta Neuropathol 2024; 148:62. [PMID: 39540994 PMCID: PMC11564344 DOI: 10.1007/s00401-024-02822-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Translocator protein (TSPO) is a mitochondrial protein expressed by microglia, ligands for which are used as a marker of neuroinflammation in PET studies of Alzheimer's disease (AD). We previously showed increasing TSPO load in the cerebral cortex with AD progression, consistent with TSPO PET scan findings. Here, we aim to characterise the microglial phenotype associated with TSPO expression to aid interpretation of the signal generated by TSPO ligands in patients. Human post-mortem sections of temporal lobe (TL) and cerebellum (Cb) from cases classified by Braak group (0-II, III-IV, V-VI; each n = 10) were fluorescently double labelled for TSPO and microglial markers: Iba1, HLA-DR, CD68, MSR-A and CD64. Quantification was performed on scanned images using QuPath software to assess the microglial phenotype of TSPO. Qualitative analysis was also performed for TSPO with GFAP (astrocytes), CD31 (endothelial cells) and CD163 (perivascular macrophages) to characterise the cellular profile of TSPO. The percentage of CD68+TSPO+ double-labelled cells was significantly higher than for other microglial markers in both brain regions and in all Braak stages, followed by MSR-A+TSPO+ microglia. Iba1+TSPO+ cells were more numerous in the cerebellum than the temporal lobe, while CD64+TSPO+ cells were more numerous in the temporal lobe. No differences were observed for the other microglial markers. TSPO expression was also detected in endothelial cells, but not detected in astrocytes nor in perivascular macrophages. Our data suggest that TSPO is mainly related to a phagocytic profile of microglia (CD68+) in human AD, potentially highlighting the ongoing neurodegeneration.
Collapse
Affiliation(s)
- Emma F Garland
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Henrike Antony
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Laura Kulagowska
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Thomas Scott
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Charlotte Rogien
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Michel Bottlaender
- Paris-Saclay University, CEA, CNRS, Service Hospitalier Frederic Joliot, Orsay, Inserm, BioMaps, France
- UNIACT Neurospin, CEA, Gif-Sur-Yvette, France
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Trust, Southampton, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.
| |
Collapse
|
9
|
Cumbers GA, Harvey-Latham ED, Kassiou M, Werry EL, Danon JJ. Emerging TSPO-PET Radiotracers for Imaging Neuroinflammation: A Critical Analysis. Semin Nucl Med 2024; 54:856-874. [PMID: 39477764 DOI: 10.1053/j.semnuclmed.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 11/19/2024]
Abstract
The translocator protein (TSPO) is a biomarker for imaging neuroinflammation via Positron Emission Tomography (PET) across a broad range of CNS conditions. Most clinically used PET ligands targeting TSPO have limitations, including high lipophilicity and off-target binding or poor binding to a mutated TSPO isoform present in up to 30% of the population. Research efforts over the past decade have focused on development of improved TSPO PET radiotracers that overcome these limitations. This review provides a critical analysis of the development and validation of these so-called "third-generation" radiotracers in clinical and preclinical settings. We also offer our perspective on the future directions of TSPO PET imaging, including recommendations for overcoming current challenges and capitalizing on emerging opportunities in molecular imaging for neuroinflammatory diseases.
Collapse
Affiliation(s)
- Grace A Cumbers
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia
| | - Edward D Harvey-Latham
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia
| | - Michael Kassiou
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia.
| | - Eryn L Werry
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia; Central Clinical School, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Jonathan J Danon
- School of Chemistry, Faculty of Science, The University of Sydney, New South Wales, Australia
| |
Collapse
|
10
|
Emvalomenos GM, Kang JWM, Jupp B, Mychasiuk R, Keay KA, Henderson LA. Recent developments and challenges in positron emission tomography imaging of gliosis in chronic neuropathic pain. Pain 2024; 165:2184-2199. [PMID: 38713812 DOI: 10.1097/j.pain.0000000000003247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/05/2024] [Indexed: 05/09/2024]
Abstract
ABSTRACT Understanding the mechanisms that underpin the transition from acute to chronic pain is critical for the development of more effective and targeted treatments. There is growing interest in the contribution of glial cells to this process, with cross-sectional preclinical studies demonstrating specific changes in these cell types capturing targeted timepoints from the acute phase and the chronic phase. In vivo longitudinal assessment of the development and evolution of these changes in experimental animals and humans has presented a significant challenge. Recent technological advances in preclinical and clinical positron emission tomography, including the development of specific radiotracers for gliosis, offer great promise for the field. These advances now permit tracking of glial changes over time and provide the ability to relate these changes to pain-relevant symptomology, comorbid psychiatric conditions, and treatment outcomes at both a group and an individual level. In this article, we summarize evidence for gliosis in the transition from acute to chronic pain and provide an overview of the specific radiotracers available to measure this process, highlighting their potential, particularly when combined with ex vivo / in vitro techniques, to understand the pathophysiology of chronic neuropathic pain. These complementary investigations can be used to bridge the existing gap in the field concerning the contribution of gliosis to neuropathic pain and identify potential targets for interventions.
Collapse
Affiliation(s)
- Gaelle M Emvalomenos
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - James W M Kang
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Bianca Jupp
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Kevin A Keay
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Luke A Henderson
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| |
Collapse
|
11
|
Liu J, Jiang J, He C, Zhou L, Zhang Y, Zhao S, Yang Z. Platycodin D and voluntary running synergistically ameliorate memory deficits in 5 × FAD mice via mediating neuromodulation and neuroinflammation. Front Aging Neurosci 2024; 16:1451766. [PMID: 39385832 PMCID: PMC11461226 DOI: 10.3389/fnagi.2024.1451766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Alzheimer's disease (AD) is the leading cause of dementia, and currently, no effective treatments are available to reverse or halt its progression in clinical practice. Although a plethora of studies have highlighted the benefits of physical exercise in combating AD, elder individuals often have limited exercise capacity. Therefore, mild physical exercise and nutritional interventions represent potential strategies for preventing and mitigating neurodegenerative diseases. Our research, along with other studies, have demonstrated that platycodin D (PD) or its metabolite, platycodigenin, derived from the medicinal plant Platycodon grandiflorus, exerts neuroprotective effects against amyloid β (Aβ)-induced neuroinflammation. However, the combined effects of PD and physical exercise on alleviating AD have yet to be explored. The current study aimed to investigate whether combined therapy could synergistically ameliorate memory deficits and AD pathology in 5 × FAD mice. Methods Five-month-old 5 × FAD mice were randomly assigned to four groups, and received either PD (5 mg/kg/day, p.o.), voluntary running, or a combination of both for 47 days. Nest building test, locomotion test, and Morris water maze test were used to evaluate the cognitive function. Immunohistochemical and ELISA analysis was performed to determine Aβ build-up, microglia and astrocytes hyperactivation, and survival neurons in the hippocampus and perirhinal cortex. Real-time quantitative PCR analysis was used to assess the polarization of microglia and astrocytes. HPLC analysis was performed to measure monoamine neurotransmitters in the hippocampus. Results and discussion The combination of PD and voluntary running synergistically restored nest-building behavior, alleviated recognition and spatial memory deficits, and showed superior effects compared to monotherapy. In addition, the PD and voluntary running combination reduced Aβ build-up, decreased hyperactivation of microglia and astrocytes in the hippocampus and perirhinal cortex, promoted the polarization of inflammatory M1 microglia and reactive astrocytes toward beneficial phenotypes, and lowered systemic circulating pro-inflammatory cytokines while increasing anti-inflammatory cytokines in 5 × FAD mice. Furthermore, combined therapy effectively protected neurons and increased levels of 5-hydroxytryptamine (5-HT) and dopamine (DA) in the hippocampus of 5 × FAD mice. In conclusion, the combination of PD and voluntary running holds great potential as a treatment for AD, offering promise for delaying onset or progression of AD.
Collapse
Affiliation(s)
- Junxin Liu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Jiahui Jiang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Chuantong He
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Longjian Zhou
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Yi Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Shuai Zhao
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Zhiyou Yang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
12
|
Fairley LH, Lai KO, Grimm A, Eckert A, Barron AM. The mitochondrial translocator protein (TSPO) in Alzheimer's disease: Therapeutic and immunomodulatory functions. Biochimie 2024; 224:120-131. [PMID: 38971458 DOI: 10.1016/j.biochi.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The translocator protein (TSPO) has been widely investigated as a PET-imaging biomarker of neuroinflammation and, more recently, as a therapeutic target for the treatment of neurodegenerative disease. TSPO ligands have been shown to exert neuroprotective effects in vivo and in vitro models of Alzheimer's disease (AD), by reducing toxic beta amyloid peptides, and attenuating brain atrophy. Recent transcriptomic and proteomic analyses, and the generation of TSPO-KO mice, have enabled new insights into the mechanistic function of TSPO in AD. Using a multi-omics approach in both TSPO-KO- and TSPO ligand-treated mice, we have demonstrated a key role for TSPO in microglial respiratory metabolism and phagocytosis in AD. In this review, we discuss emerging evidence for therapeutic and immunomodulatory functions of TSPO in AD, and new tools for studying TSPO in the brain.
Collapse
Affiliation(s)
- Lauren H Fairley
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Kei Onn Lai
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore
| | - Amandine Grimm
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anne Eckert
- Transfaculty Research Platform, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 308232, Singapore.
| |
Collapse
|
13
|
Nuszkiewicz J, Kukulska-Pawluczuk B, Piec K, Jarek DJ, Motolko K, Szewczyk-Golec K, Woźniak A. Intersecting Pathways: The Role of Metabolic Dysregulation, Gastrointestinal Microbiome, and Inflammation in Acute Ischemic Stroke Pathogenesis and Outcomes. J Clin Med 2024; 13:4258. [PMID: 39064298 PMCID: PMC11278353 DOI: 10.3390/jcm13144258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/13/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
Acute ischemic stroke (AIS) remains a major cause of mortality and long-term disability worldwide, driven by complex and multifaceted etiological factors. Metabolic dysregulation, gastrointestinal microbiome alterations, and systemic inflammation are emerging as significant contributors to AIS pathogenesis. This review addresses the critical need to understand how these factors interact to influence AIS risk and outcomes. We aim to elucidate the roles of dysregulated adipokines in obesity, the impact of gut microbiota disruptions, and the neuroinflammatory cascade initiated by lipopolysaccharides (LPS) in AIS. Dysregulated adipokines in obesity exacerbate inflammatory responses, increasing AIS risk and severity. Disruptions in the gut microbiota and subsequent LPS-induced neuroinflammation further link systemic inflammation to AIS. Advances in neuroimaging and biomarker development have improved diagnostic precision. Here, we highlight the need for a multifaceted approach to AIS management, integrating metabolic, microbiota, and inflammatory insights. Potential therapeutic strategies targeting these pathways could significantly improve AIS prevention and treatment. Future research should focus on further elucidating these pathways and developing targeted interventions to mitigate the impacts of metabolic dysregulation, microbiome imbalances, and inflammation on AIS.
Collapse
Affiliation(s)
- Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Beata Kukulska-Pawluczuk
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Katarzyna Piec
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Dorian Julian Jarek
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Karina Motolko
- Student Research Club of Neurology, Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland;
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| |
Collapse
|
14
|
Caradonna E, Nemni R, Bifone A, Gandolfo P, Costantino L, Giordano L, Mormone E, Macula A, Cuomo M, Difruscolo R, Vanoli C, Vanoli E, Ferrara F. The Brain-Gut Axis, an Important Player in Alzheimer and Parkinson Disease: A Narrative Review. J Clin Med 2024; 13:4130. [PMID: 39064171 PMCID: PMC11278248 DOI: 10.3390/jcm13144130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are severe age-related disorders with complex and multifactorial causes. Recent research suggests a critical link between neurodegeneration and the gut microbiome, via the gut-brain communication pathway. This review examines the role of trimethylamine N-oxide (TMAO), a gut microbiota-derived metabolite, in the development of AD and PD, and investigates its interaction with microRNAs (miRNAs) along this bidirectional pathway. TMAO, which is produced from dietary metabolites like choline and carnitine, has been linked to increased neuroinflammation, protein misfolding, and cognitive decline. In AD, elevated TMAO levels are associated with amyloid-beta and tau pathologies, blood-brain barrier disruption, and neuronal death. TMAO can cross the blood-brain barrier and promote the aggregation of amyloid and tau proteins. Similarly, TMAO affects alpha-synuclein conformation and aggregation, a hallmark of PD. TMAO also activates pro-inflammatory pathways such as NF-kB signaling, exacerbating neuroinflammation further. Moreover, TMAO modulates the expression of various miRNAs that are involved in neurodegenerative processes. Thus, the gut microbiome-miRNA-brain axis represents a newly discovered mechanistic link between gut dysbiosis and neurodegeneration. MiRNAs regulate the key pathways involved in neuroinflammation, oxidative stress, and neuronal death, contributing to disease progression. As a direct consequence, specific miRNA signatures may serve as potential biomarkers for the early detection and monitoring of AD and PD progression. This review aims to elucidate the complex interrelationships between the gut microbiota, trimethylamine-N-oxide (TMAO), microRNAs (miRNAs), and the central nervous system, and the implications of these connections in neurodegenerative diseases. In this context, an overview of the current neuroradiology techniques available for studying neuroinflammation and of the animal models used to investigate these intricate pathologies will also be provided. In summary, a bulk of evidence supports the concept that modulating the gut-brain communication pathway through dietary changes, the manipulation of the microbiome, and/or miRNA-based therapies may offer novel approaches for implementing the treatment of debilitating neurological disorders.
Collapse
Affiliation(s)
- Eugenio Caradonna
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (E.C.); (F.F.)
| | - Raffaello Nemni
- Unit of Neurology, Centro Diagnostico Italiano S.p.A., Milan Fondazione Crespi Spano, 20011 Milan, Italy;
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
| | - Angelo Bifone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Patrizia Gandolfo
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
| | - Lucy Costantino
- Laboratory of Medical Genetics, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (L.C.); (L.G.)
| | - Luca Giordano
- Laboratory of Medical Genetics, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (L.C.); (L.G.)
| | - Elisabetta Mormone
- Institute for Stem-Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Anna Macula
- Centro Ricerche Bracco, Bracco Imaging S.p.A., Colleretto Giacosa, 10010 Turin, Italy;
- Department of Physics, University of Torino, 10124 Torino, Italy
| | - Mariarosa Cuomo
- Nuclear Medicine Unit, Imaging Department, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (P.G.); (M.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | | | - Camilla Vanoli
- Department of Clinical Psychology, Antioch University Los Angeles, Culver City, CA 90230, USA
| | - Emilio Vanoli
- School of Nursing, Cardiovascular Diseases, University of Pavia, 27100 Pavia, Italy;
| | - Fulvio Ferrara
- Integrated Laboratory Medicine Services, Centro Diagnostico Italiano S.p.A., 20011 Milan, Italy; (E.C.); (F.F.)
| |
Collapse
|
15
|
Pumo A, Legeay S. The dichotomous activities of microglia: A potential driver for phenotypic heterogeneity in Alzheimer's disease. Brain Res 2024; 1832:148817. [PMID: 38395249 DOI: 10.1016/j.brainres.2024.148817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/28/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, characterized by two defining neuropathological hallmarks: amyloid plaques composed of Aβ aggregates and neurofibrillary pathology. Recent research suggests that microglia have both beneficial and detrimental effects in the development of AD. A new theory proposes that microglia play a beneficial role in the early stages of the disease but become harmful in later stages. Further investigations are needed to gain a comprehensive understanding of this shift in microglia's function. This transition is likely influenced by specific conditions, including spatial, temporal, and transcriptional factors, which ultimately lead to the deterioration of microglial functionality. Additionally, recent studies have also highlighted the potential influence of microglia diversity on the various manifestations of AD. By deciphering the multiple states of microglia and the phenotypic heterogeneity in AD, significant progress can be made towards personalized medicine and better treatment outcomes for individuals affected by AD.
Collapse
Affiliation(s)
- Anna Pumo
- Université d'Angers, Faculté de Santé, Département Pharmacie, 16, Boulevard Daviers, Angers 49045, France.
| | - Samuel Legeay
- Université d'Angers, Faculté de Santé, Département Pharmacie, 16, Boulevard Daviers, Angers 49045, France; Univ Angers, Inserm, CNRS, MINT, SFR ICAT, Angers F-49000, France
| |
Collapse
|
16
|
Gouilly D, Vrillon A, Bertrand E, Goubeaud M, Catala H, Germain J, Ainaoui N, Rafiq M, Nogueira L, Mouton-Liger F, Planton M, Salabert AS, Hitzel A, Méligne D, Jasse L, Sarton B, Silva S, Lemesle B, Péran P, Payoux P, Thalamas C, Paquet C, Pariente J. Translocator protein (TSPO) genotype does not change cerebrospinal fluid levels of glial activation, axonal and synaptic damage markers in early Alzheimer's disease. Neuroimage Clin 2024; 43:103626. [PMID: 38850834 PMCID: PMC11201347 DOI: 10.1016/j.nicl.2024.103626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND PET imaging of the translocator protein (TSPO) is used to assess in vivo brain inflammation. One of the main methodological issues with this method is the allelic dependence of the radiotracer affinity. In Alzheimer's disease (AD), previous studies have shown similar clinical and patho-biological profiles between TSPO genetic subgroups. However, there is no evidence regarding the effect of the TSPO genotype on cerebrospinal-fluid biomarkers of glial activation, and synaptic and axonal damage. METHOD We performed a trans-sectional study in early AD to compare cerebrospinal-fluid levels of GFAP, YKL-40, sTREM2, IL-6, IL-10, NfL and neurogranin between TSPO genetic subgroups. RESULTS We recruited 33 patients with early AD including 16 (48%) high affinity binders, 13 (39%) mixed affinity binders, and 4/33 (12%) low affinity binders. No difference was observed in terms of demographics, and cerebrospinal fluid levels of each biomarker for the different subgroups. CONCLUSION TSPO genotype is not associated with a change in glial activation, synaptic and axonal damage in early AD. Further studies with larger numbers of participants will be needed to confirm that the inclusion of specific TSPO genetic subgroups does not introduce selection bias in studies and trials of AD that combine TSPO imaging with cerebrospinal fluid biomarkers.
Collapse
Affiliation(s)
- Dominique Gouilly
- Department of Cognitive Neurology, Epilepsy, Sleep and Movement Disorders, CHU Toulouse Purpan, Toulouse, France.
| | - Agathe Vrillon
- Université de Paris, Cognitive Neurology Center, GHU Nord, APHP, Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMRS11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Elsa Bertrand
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC 1436), Toulouse, France
| | - Marie Goubeaud
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC 1436), Toulouse, France
| | - Hélène Catala
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC 1436), Toulouse, France
| | - Johanne Germain
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC 1436), Toulouse, France
| | - Nadéra Ainaoui
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC 1436), Toulouse, France
| | - Marie Rafiq
- Department of Cognitive Neurology, Epilepsy, Sleep and Movement Disorders, CHU Toulouse Purpan, Toulouse, France; Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France
| | - Leonor Nogueira
- Laboratory of Cell Biology and Cytology, CHU Toulouse Purpan, Toulouse, France
| | - François Mouton-Liger
- Université de Paris, Inserm UMRS11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Mélanie Planton
- Department of Cognitive Neurology, Epilepsy, Sleep and Movement Disorders, CHU Toulouse Purpan, Toulouse, France; Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France
| | - Anne-Sophie Salabert
- Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, Toulouse, France
| | - Anne Hitzel
- Department of Nuclear Medicine, CHU Toulouse Purpan, Toulouse, France
| | - Déborah Méligne
- Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France
| | - Laurence Jasse
- Department of Cognitive Neurology, Epilepsy, Sleep and Movement Disorders, CHU Toulouse Purpan, Toulouse, France
| | - Benjamine Sarton
- Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France; Critical Care Unit, CHU Toulouse Purpan, Toulouse, France
| | - Stein Silva
- Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France; Critical Care Unit, CHU Toulouse Purpan, Toulouse, France
| | - Béatrice Lemesle
- Department of Cognitive Neurology, Epilepsy, Sleep and Movement Disorders, CHU Toulouse Purpan, Toulouse, France
| | - Patrice Péran
- Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France
| | - Pierre Payoux
- Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, Toulouse, France
| | - Claire Thalamas
- Center of Clinical Investigation, CHU Toulouse Purpan (CIC 1436), Toulouse, France
| | - Claire Paquet
- Université de Paris, Cognitive Neurology Center, GHU Nord, APHP, Hospital Lariboisière Fernand Widal, Paris, France; Université de Paris, Inserm UMRS11-44 Therapeutic Optimization in Neuropsychopharmacology, Paris, France
| | - Jérémie Pariente
- Department of Cognitive Neurology, Epilepsy, Sleep and Movement Disorders, CHU Toulouse Purpan, Toulouse, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC 1436), Toulouse, France; Toulouse Neuroimaging Center, UMR 1214, Inserm/UPS, Toulouse, France
| |
Collapse
|
17
|
Hector M, Langmann T, Wolf A. Translocator protein (18 kDa) (Tspo) in the retina and implications for ocular diseases. Prog Retin Eye Res 2024; 100:101249. [PMID: 38430990 DOI: 10.1016/j.preteyeres.2024.101249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Translocator protein (18 kDa) (Tspo), formerly known as peripheral benzodiazepine receptor is a highly conserved transmembrane protein primarily located in the outer mitochondrial membrane. In the central nervous system (CNS), especially in glia cells, Tspo is upregulated upon inflammation. Consequently, Tspo was used as a tool for diagnostic in vivo imaging of neuroinflammation in the brain and as a potential therapeutic target. Several synthetic Tspo ligands have been explored as immunomodulatory and neuroprotective therapy approaches. Although the function of Tspo and how its ligands exert these beneficial effects is not fully clear, it became a research topic of interest, especially in ocular diseases in the past few years. This review summarizes state-of-the-art knowledge of Tspo expression and its proposed functions in different cells of the retina including microglia, retinal pigment epithelium and Müller cells. Tspo is involved in cytokine signaling, oxidative stress and reactive oxygen species production, calcium signaling, neurosteroid synthesis, energy metabolism, and cholesterol efflux. We also highlight recent developments in preclinical models targeting Tspo and summarize the relevance of Tspo biology for ocular and retinal diseases. We conclude that glial upregulation of Tspo in different ocular pathologies and the use of Tspo ligands as promising therapeutic approaches in preclinical studies underline the importance of Tspo as a potential disease-modifying protein.
Collapse
Affiliation(s)
- Mandy Hector
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Anne Wolf
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
18
|
Jaisa-Aad M, Muñoz-Castro C, Healey MA, Hyman BT, Serrano-Pozo A. Characterization of monoamine oxidase-B (MAO-B) as a biomarker of reactive astrogliosis in Alzheimer's disease and related dementias. Acta Neuropathol 2024; 147:66. [PMID: 38568475 PMCID: PMC10991006 DOI: 10.1007/s00401-024-02712-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/03/2024] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Reactive astrogliosis accompanies the two neuropathological hallmarks of Alzheimer's disease (AD)-Aβ plaques and neurofibrillary tangles-and parallels neurodegeneration in AD and AD-related dementias (ADRD). Thus, there is growing interest in developing imaging and fluid biomarkers of reactive astrogliosis for AD/ADRD diagnosis and prognostication. Monoamine oxidase-B (MAO-B) is emerging as a target for PET imaging radiotracers of reactive astrogliosis. However, a thorough characterization of MAO-B expression in postmortem control and AD/ADRD brains is lacking. We sought to: (1) identify the primary cell type(s) expressing MAO-B in control and AD brains; (2) quantify MAO-B immunoreactivity in multiple brain regions of control and AD donors as a proxy for PET radiotracer uptake; (3) correlate MAO-B level with local AD neuropathological changes, reactive glia, and cortical atrophy; (4) determine whether the MAOB rs1799836 SNP genotype impacts MAO-B expression level; (5) compare MAO-B immunoreactivity across AD/ADRD, including Lewy body diseases (LBD) and frontotemporal lobar degenerations with tau (FTLD-Tau) and TDP-43 (FTLD-TDP). We found that MAO-B is mainly expressed by subpial and perivascular cortical astrocytes as well as by fibrous white matter astrocytes in control brains, whereas in AD brains, MAO-B is significantly upregulated by both cortical reactive astrocytes and white matter astrocytes across temporal, frontal, and occipital lobes. By contrast, MAO-B expression level was unchanged and lowest in cerebellum. Cortical MAO-B expression was independently associated with cortical atrophy and local measures of reactive astrocytes and microglia, and significantly increased in reactive astrocytes surrounding Thioflavin-S+ dense-core Aβ plaques. MAO-B expression was not affected by the MAOB rs1799836 SNP genotype. MAO-B expression was also significantly increased in the frontal cortex and white matter of donors with corticobasal degeneration, Pick's disease, and FTLD-TDP, but not in LBD or progressive supranuclear palsy. These findings support ongoing efforts to develop MAO-B-based PET radiotracers to image reactive astrogliosis in AD/ADRD.
Collapse
Affiliation(s)
- Methasit Jaisa-Aad
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Clara Muñoz-Castro
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Molly A Healey
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- MassGeneral Institute for Neurodegenerative Disease, 114 16th St., Charlestown, MA, 02129, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Etebar F, Harkin DG, White AR, Dando SJ. Non-invasive in vivo imaging of brain and retinal microglia in neurodegenerative diseases. Front Cell Neurosci 2024; 18:1355557. [PMID: 38348116 PMCID: PMC10859418 DOI: 10.3389/fncel.2024.1355557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Microglia play crucial roles in immune responses and contribute to fundamental biological processes within the central nervous system (CNS). In neurodegenerative diseases, microglia undergo functional changes and can have both protective and pathogenic roles. Microglia in the retina, as an extension of the CNS, have also been shown to be affected in many neurological diseases. While our understanding of how microglia contribute to pathological conditions is incomplete, non-invasive in vivo imaging of brain and retinal microglia in living subjects could provide valuable insights into their role in the neurodegenerative diseases and open new avenues for diagnostic biomarkers. This mini-review provides an overview of the current brain and retinal imaging tools for studying microglia in vivo. We focus on microglia targets, the advantages and limitations of in vivo microglia imaging approaches, and applications for evaluating the pathogenesis of neurological conditions, such as Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Fazeleh Etebar
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Damien G. Harkin
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Anthony R. White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Samantha J. Dando
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Centre for Vision and Eye Research, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| |
Collapse
|
20
|
Ottoy J, De Picker L, Kang MS. Microglial Positron Emission Tomography Imaging In Vivo : Positron Emission Tomography Radioligands: Utility in Research and Clinical Practice. ADVANCES IN NEUROBIOLOGY 2024; 37:579-589. [PMID: 39207714 DOI: 10.1007/978-3-031-55529-9_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS) play a key role in regulating and maintaining homeostasis in the brain. However, the CNS is also vulnerable to infections and inflammatory processes. In response to CNS perturbations, microglia become reactive, notably with expression of the translocator protein (TSPO), primarily on their outer mitochondrial membrane. Despite TSPO being commonly used as a marker for microglia, it is also present in other cell types such as astrocytes. Positron emission tomography (PET) ligands that target the TSPO enable the noninvasive detection and quantification of glial reactivity. While some limitations were raised, TSPO PET remains an attractive biomarker of CNS infection and inflammation. This book chapter delves into the development and application of microglial PET imaging with a focus on the TSPO PET. First, we provide an overview of the evolution of TSPO PET radioligands from first-generation to second-generation ligands and their applications in studying neuroinflammation (or CNS inflammation). Subsequently, we discuss the limitations and challenges associated with TSPO PET. Then we go on to explore non-TSPO targets for microglial PET imaging. Finally, we conclude with future directions for research and clinical practice in this field.
Collapse
Affiliation(s)
- Julie Ottoy
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium
- University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Min Su Kang
- Dr. Sandra E. Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Zhang S, Deng Z, Qiu Y, Lu G, Wu J, Huang H. FGIN-1-27 Mitigates Radiation-induced Mitochondrial Hyperfunction and Cellular Hyperactivation in Cultured Astrocytes. Neuroscience 2023; 535:23-35. [PMID: 37913861 DOI: 10.1016/j.neuroscience.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
Radiation-induced brain injury (RBI) poses a significant challenge in the context of radiotherapy for intracranial tumors, necessitating a comprehensive understanding of the cellular and molecular mechanisms involved. While prior investigations have underscored the role of astrocyte activation and excessive vascular endothelial growth factor production in microvascular damage associated with RBI, there remains a scarcity of studies examining the impact of radiation on astrocytes, particularly regarding organelles such as mitochondria. Thus, our study aimed to elucidate alterations in astrocyte and mitochondrial functionality following radiation exposure, with a specific focus on evaluating the potential ameliorative effects of translocator protein 18 kDa(TSPO) ligands. In this study, cultured astrocytes were subjected to X-ray irradiation, and their cellular states and mitochondrial functions were examined and compared to control cells. Our findings revealed that radiation-induced astrocytic hyperactivation, transforming them into the neurotoxic A1-type, concomitant with reduced cell proliferation. Additionally, radiation triggered mitochondrial hyperfunction, heightened the mitochondrial membrane potential, and increased oxidative metabolite production. However, following treatment with FGIN-1-27, a TSPO ligand, we observed a restoration of mitochondrial function and a reduction in oxidative metabolite production. Moreover, this intervention mitigated astrocyte hyperactivity, decreased the number of A1-type astrocytes, and restored cell proliferative capacity. In conclusion, our study has unveiled additional manifestations of radiation-induced astrocyte dysfunction and validated that TSPO ligands may serve as a promising therapeutic strategy to mitigate this dysfunction. It has potential clinical implications for the treatment of RBI.
Collapse
Affiliation(s)
- Shifeng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Zhezhi Deng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yuemin Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Gengxin Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Junyu Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Haiwei Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China.
| |
Collapse
|
22
|
Carlson ML, Jackson IM, Azevedo EC, Reyes ST, Alam IS, Kellow R, Castillo JB, Nagy SC, Sharma R, Brewer M, Cleland J, Shen B, James ML. Development and Initial Assessment of [ 18F]OP-801: a Novel Hydroxyl Dendrimer PET Tracer for Preclinical Imaging of Innate Immune Activation in the Whole Body and Brain. Mol Imaging Biol 2023; 25:1063-1072. [PMID: 37735280 DOI: 10.1007/s11307-023-01850-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 07/25/2023] [Accepted: 08/09/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE Innate immune activation plays a critical role in the onset and progression of many diseases. While positron emission tomography (PET) imaging provides a non-invasive means to visualize and quantify such immune responses, most available tracers are not specific for innate immune cells. To address this need, we developed [18F]OP-801 by radiolabeling a novel hydroxyl dendrimer that is selectively taken up by reactive macrophages/microglia and evaluated its ability to detect innate immune activation in mice following lipopolysaccharide (LPS) challenge. PROCEDURES OP-801 was radiolabeled in two steps: [18F]fluorination of a tosyl precursor to yield [18F]3-fluoropropylazide, followed by a copper-catalyzed click reaction. After purification and stability testing, [18F]OP-801 (150-250 μCi) was intravenously injected into female C57BL/6 mice 24 h after intraperitoneal administration of LPS (10 mg/kg, n=14) or saline (n=6). Upon completing dynamic PET/CT imaging, mice were perfused, and radioactivity was measured in tissues of interest via gamma counting or autoradiography. RESULTS [18F]OP-801 was produced with >95% radiochemical purity, 12-52 μCi/μg specific activity, and 4.3±1.5% decay-corrected yield. Ex vivo metabolite analysis of plasma samples (n=4) demonstrated high stability in mice (97±3% intact tracer >120 min post-injection). PET/CT images of mice following LPS challenge revealed higher signal in organs known to be inflamed in this context, including the liver, lung, and spleen. Gamma counting confirmed PET findings, showing significantly elevated signal in the same tissues compared to saline-injected mice: the liver (p=0.009), lung (p=0.030), and spleen (p=0.004). Brain PET/CT images (summed 50-60 min) revealed linearly increasing [18F]OP-801 uptake in the whole brain that significantly correlated with murine sepsis score (r=0.85, p<0.0001). Specifically, tracer uptake was significantly higher in the brain stem, cortex, olfactory bulb, white matter, and ventricles of LPS-treated mice compared to saline-treated mice (p<0.05). CONCLUSION [18F]OP-801 is a promising new PET tracer for sensitive and specific detection of activated macrophages and microglia that warrants further investigation.
Collapse
Affiliation(s)
| | - Isaac M Jackson
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - E Carmen Azevedo
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - Samantha T Reyes
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - Israt S Alam
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - Rowaid Kellow
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - Jessa B Castillo
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - Sydney C Nagy
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - Rishi Sharma
- Ashvattha Therapeutics, Inc., Redwood City, CA, USA
| | | | | | - Bin Shen
- Department of Radiology at Stanford University, Stanford, CA, USA
| | - Michelle L James
- Department of Radiology at Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences at Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
De Picker LJ, Morrens M, Branchi I, Haarman BCM, Terada T, Kang MS, Boche D, Tremblay ME, Leroy C, Bottlaender M, Ottoy J. TSPO PET brain inflammation imaging: A transdiagnostic systematic review and meta-analysis of 156 case-control studies. Brain Behav Immun 2023; 113:415-431. [PMID: 37543251 DOI: 10.1016/j.bbi.2023.07.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
INTRODUCTION The 18-kDa translocator protein (TSPO) is increasingly recognized as a molecular target for PET imaging of inflammatory responses in various central nervous system (CNS) disorders. However, the reported sensitivity and specificity of TSPO PET to identify brain inflammatory processes appears to vary greatly across disorders, disease stages, and applied quantification methods. To advance TSPO PET as a potential biomarker to evaluate brain inflammation and anti-inflammatory therapies, a better understanding of its applicability across disorders is needed. We conducted a transdiagnostic systematic review and meta-analysis of all in vivo human TSPO PET imaging case-control studies in the CNS. Specifically, we investigated the direction, strength, and heterogeneity associated with the TSPO PET signal across disorders in pre-specified brain regions, and explored the demographic and methodological sources of heterogeneity. METHODS We searched for English peer-reviewed articles that reported in vivo human case-control TSPO PET differences. We extracted the demographic details, TSPO PET outcomes, and technical variables of the PET procedure. A random-effects meta-analysis was applied to estimate case-control standardized mean differences (SMD) of the TSPO PET signal in the lobar/whole-brain cortical grey matter (cGM), thalamus, and cortico-limbic circuitry between different illness categories. Heterogeneity was evaluated with the I2 statistic and explored using subgroup and meta-regression analyses for radioligand generation, PET quantification method, age, sex, and publication year. Significance was set at the False Discovery Rate (FDR)-corrected P < 0.05. RESULTS 156 individual case-control studies were included in the systematic review, incorporating data for 2381 healthy controls and 2626 patients. 139 studies documented meta-analysable data and were grouped into 11 illness categories. Across all the illness categories, we observed a significantly higher TSPO PET signal in cases compared to controls for the cGM (n = 121 studies, SMD = 0.358, PFDR < 0.001, I2 = 68%), with a significant difference between the illness categories (P = 0.004). cGM increases were only significant for Alzheimer's disease (SMD = 0.693, PFDR < 0.001, I2 = 64%) and other neurodegenerative disorders (SMD = 0.929, PFDR < 0.001, I2 = 73%). Cortico-limbic increases (n = 97 studies, SMD = 0.541, P < 0.001, I2 = 67%) were most prominent for Alzheimer's disease, mild cognitive impairment, other neurodegenerative disorders, mood disorders and multiple sclerosis. Thalamic involvement (n = 79 studies, SMD = 0.393, P < 0.001, I2 = 71%) was observed for Alzheimer's disease, other neurodegenerative disorders, multiple sclerosis, and chronic pain and functional disorders (all PFDR < 0.05). Main outcomes for systemic immunological disorders, viral infections, substance use disorders, schizophrenia and traumatic brain injury were not significant. We identified multiple sources of between-study variance to the TSPO PET signal including a strong transdiagnostic effect of the quantification method (explaining 25% of between-study variance; VT-based SMD = 0.000 versus reference tissue-based studies SMD = 0.630; F = 20.49, df = 1;103, P < 0.001), patient age (9% of variance), and radioligand generation (5% of variance). CONCLUSION This study is the first overarching transdiagnostic meta-analysis of case-control TSPO PET findings in humans across several brain regions. We observed robust increases in the TSPO signal for specific types of disorders, which were widespread or focal depending on illness category. We also found a large and transdiagnostic horizontal (positive) shift of the effect estimates of reference tissue-based compared to VT-based studies. Our results can support future studies to optimize experimental design and power calculations, by taking into account the type of disorder, brain region-of-interest, radioligand, and quantification method.
Collapse
Affiliation(s)
- Livia J De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium.
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy
| | - Bartholomeus C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tatsuhiro Terada
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Min Su Kang
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, UK
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, BC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Claire Leroy
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France; Université Paris-Saclay, UNIACT, Neurospin, CEA, Gif-sur-Yvette, France
| | - Julie Ottoy
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Palmer JM, Huentelman M, Ryan L. More than just risk for Alzheimer's disease: APOE ε4's impact on the aging brain. Trends Neurosci 2023; 46:750-763. [PMID: 37460334 DOI: 10.1016/j.tins.2023.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
The apolipoprotein ε4 (APOE ε4) allele is most commonly associated with increased risk for late-onset Alzheimer's disease (AD). However, recent longitudinal studies suggest that these risks are overestimated; most ε4 carriers will not develop dementia in their lifetime. In this article, we review new evidence regarding the impact of APOE ε4 on cognition among healthy older adults. We discuss emerging work from animal models suggesting that ε4 impacts brain structure and function in multiple ways that may lead to age-related cognitive impairment, independent from AD pathology. We discuss the importance of taking an individualized approach in future studies by incorporating biomarkers and neuroimaging methods that may better disentangle the phenotypic influences of APOE ε4 on the aging brain from prodromal AD pathology.
Collapse
Affiliation(s)
- Justin M Palmer
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| | - Matthew Huentelman
- Translational Genomics Research Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Lee Ryan
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|
25
|
Ekblad LL, Tuisku J, Koivumäki M, Helin S, Rinne JO, Snellman A. Insulin resistance and body mass index are associated with TSPO PET in cognitively unimpaired elderly. J Cereb Blood Flow Metab 2023; 43:1588-1600. [PMID: 37113066 PMCID: PMC10414007 DOI: 10.1177/0271678x231172519] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/27/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023]
Abstract
Metabolic risk factors are associated with peripheral low-grade inflammation and an increased risk for dementia. We evaluated if metabolic risk factors i.e. insulin resistance, body mass index (BMI), serum cholesterol values, or high sensitivity C-reactive protein associate with central inflammation or beta-amyloid (Aβ) accumulation in the brain, and if these associations are modulated by APOE4 gene dose. Altogether 60 cognitively unimpaired individuals (mean age 67.7 years (SD 4.7); 63% women; 21 APOE3/3, 20 APOE3/4 and 19 APOE4/4) underwent positron emission tomography with [11C]PK11195 targeting TSPO (18 kDa translocator protein) and [11C]PIB targeting fibrillar Aβ. [11C]PK11195 distribution value ratios and [11C]PIB standardized uptake values were calculated in a cortical composite region of interest typical for Aβ accumulation in Alzheimer's disease. Associations between metabolic risk factors, [11C]PK11195, and [11C]PIB uptake were evaluated with linear models adjusted for age and sex. Higher logarithmic HOMA-IR (standardized beta 0.40, p = 0.002) and BMI (standardized beta 0.27, p = 0.048) were associated with higher TSPO availability. Voxel-wise analyses indicated that this association was mainly seen in the parietal cortex. Higher logarithmic HOMA-IR was associated with higher [11C]PIB (standardized beta 0.44, p = 0.02), but only in APOE4/4 homozygotes. BMI and HOMA-IR seem to influence TSPO availability in the brain.
Collapse
Affiliation(s)
- Laura L Ekblad
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Mikko Koivumäki
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- InFLAMES Reseach Flagship Center, University of Turku, Turku, Finland
| | - Anniina Snellman
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
26
|
Tournier B, Bouteldja F, Amossé Q, Nicolaides A, Duarte Azevedo M, Tenenbaum L, Garibotto V, Ceyzériat K, Millet P. 18 kDa Translocator Protein TSPO Is a Mediator of Astrocyte Reactivity. ACS OMEGA 2023; 8:31225-31236. [PMID: 37663488 PMCID: PMC10468775 DOI: 10.1021/acsomega.3c03368] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/01/2023] [Indexed: 09/05/2023]
Abstract
An increase in astrocyte reactivity has been described in Alzheimer's disease and seems to be related to the presence of a pro-inflammatory environment. Reactive astrocytes show an increase in the density of the 18 kDa translocator protein (TSPO), but TSPO involvement in astrocyte functions remains poorly understood. The goal of this study was to better characterize the mechanisms leading to the increase in TSPO under inflammatory conditions and the associated consequences. For this purpose, the C6 astrocytic cell line was used in the presence of lipopolysaccharide (LPS) or TSPO overexpression mediated by the transfection of a plasmid encoding TSPO. The results show that nonlethal doses of LPS induced TSPO expression at mRNA and protein levels through a STAT3-dependent mechanism and increased the number of mitochondria per cell. LPS stimulated reactive oxygen species (ROS) production and decreased glucose consumption (quantified by the [18F]FDG uptake), and these effects were diminished by FEPPA, a TSPO antagonist. The transfection-mediated overexpression of TSPO induced ROS production, and this effect was blocked by FEPPA. In addition, a synergistic effect of overexpression of TSPO and LPS on ROS production was observed. These data show that the increase of TSPO in astrocytic cells is involved in the regulation of glucose metabolism and in the pro-inflammatory response. These data suggest that the overexpression of TSPO by astrocytes in Alzheimer's disease would have rather deleterious effects by promoting the pro-inflammatory response.
Collapse
Affiliation(s)
- Benjamin
B. Tournier
- Department
of Psychiatry, University Hospitals of Geneva, Geneva 1206, Switzerland
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Farha Bouteldja
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Quentin Amossé
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Alekos Nicolaides
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| | - Marcelo Duarte Azevedo
- Laboratory
of Cellular and Molecular Neurotherapies, Center for Neuroscience
Research, Clinical Neuroscience Department, Lausanne University Hospital, Lausanne 1011, Switzerland
| | - Liliane Tenenbaum
- Laboratory
of Cellular and Molecular Neurotherapies, Center for Neuroscience
Research, Clinical Neuroscience Department, Lausanne University Hospital, Lausanne 1011, Switzerland
| | - Valentina Garibotto
- Division
of Nuclear Medicine, Diagnostic Department, University Hospitals of Geneva, Geneva 1206, Switzerland
- CIBM
Center for BioMedical Imaging; NIMTLab, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Kelly Ceyzériat
- Department
of Psychiatry, University Hospitals of Geneva, Geneva 1206, Switzerland
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
- Division
of Nuclear Medicine, Diagnostic Department, University Hospitals of Geneva, Geneva 1206, Switzerland
- CIBM
Center for BioMedical Imaging; NIMTLab, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Philippe Millet
- Department
of Psychiatry, University Hospitals of Geneva, Geneva 1206, Switzerland
- Department
of Psychiatry, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
27
|
Garland EF, Dennett O, Lau LC, Chatelet DS, Bottlaender M, Nicoll JAR, Boche D. The mitochondrial protein TSPO in Alzheimer's disease: relation to the severity of AD pathology and the neuroinflammatory environment. J Neuroinflammation 2023; 20:186. [PMID: 37580767 PMCID: PMC10424356 DOI: 10.1186/s12974-023-02869-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023] Open
Abstract
The 18kD translocator protein (TSPO) is used as a positron emission tomography (PET) target to quantify neuroinflammation in patients. In Alzheimer's disease (AD), the cerebellum is the pseudo-reference region for comparison with the cerebral cortex due to the absence of AD pathology and lower levels of TSPO. However, using the cerebellum as a pseudo-reference region is debated, with other brain regions suggested as more suitable. This paper aimed to establish the neuroinflammatory differences between the temporal cortex and cerebellar cortex, including TSPO expression. Using 60 human post-mortem samples encompassing the spectrum of Braak stages (I-VI), immunostaining for pan-Aβ, hyperphosphorylated (p)Tau, TSPO and microglial proteins Iba1, HLA-DR and MSR-A was performed in the temporal cortex and cerebellum. In the cerebellum, Aβ but not pTau, increased over the course of the disease, in contrast to the temporal cortex, where both proteins were significantly increased. TSPO increased in the temporal cortex, more than twofold in the later stages of AD compared to the early stages, but not in the cerebellum. Conversely, Iba1 increased in the cerebellum, but not in the temporal cortex. TSPO was associated with pTau in the temporal cortex, suggesting that TSPO positive microglia may be reacting to pTau itself and/or neurodegeneration at later stages of AD. Furthermore, the neuroinflammatory microenvironment was examined, using MesoScale Discovery assays, and IL15 only was significantly increased in the temporal cortex. Together this data suggests that the cerebellum maintains a more homeostatic environment compared to the temporal cortex, with a consistent TSPO expression, supporting its use as a pseudo-reference region for quantification in TSPO PET scans.
Collapse
Affiliation(s)
- Emma F Garland
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Oliver Dennett
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Laurie C Lau
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - David S Chatelet
- Biomedical Imaging Unit, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Michel Bottlaender
- CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frederic Joliot, Paris-Sacaly University, 91400, Orsay, France
- UNIACT Neurospin, CEA, Gif-Sur-Yvette, 91191, France
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Trust, Southampton, SO16 6YD, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK.
| |
Collapse
|
28
|
Nieuwland JM, Nutma E, Philippens IHCHM, Böszörményi KP, Remarque EJ, Bakker J, Meijer L, Woerdman N, Fagrouch ZC, Verstrepen BE, Langermans JAM, Verschoor EJ, Windhorst AD, Bontrop RE, de Vries HE, Stammes MA, Middeldorp J. Longitudinal positron emission tomography and postmortem analysis reveals widespread neuroinflammation in SARS-CoV-2 infected rhesus macaques. J Neuroinflammation 2023; 20:179. [PMID: 37516868 PMCID: PMC10387202 DOI: 10.1186/s12974-023-02857-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/19/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) patients initially develop respiratory symptoms, but they may also suffer from neurological symptoms. People with long-lasting effects after acute infections with severe respiratory syndrome coronavirus 2 (SARS-CoV-2), i.e., post-COVID syndrome or long COVID, may experience a variety of neurological manifestations. Although we do not fully understand how SARS-CoV-2 affects the brain, neuroinflammation likely plays a role. METHODS To investigate neuroinflammatory processes longitudinally after SARS-CoV-2 infection, four experimentally SARS-CoV-2 infected rhesus macaques were monitored for 7 weeks with 18-kDa translocator protein (TSPO) positron emission tomography (PET) using [18F]DPA714, together with computed tomography (CT). The baseline scan was compared to weekly PET-CTs obtained post-infection (pi). Brain tissue was collected following euthanasia (50 days pi) to correlate the PET signal with TSPO expression, and glial and endothelial cell markers. Expression of these markers was compared to brain tissue from uninfected animals of comparable age, allowing the examination of the contribution of these cells to the neuroinflammatory response following SARS-CoV-2 infection. RESULTS TSPO PET revealed an increased tracer uptake throughout the brain of all infected animals already from the first scan obtained post-infection (day 2), which increased to approximately twofold until day 30 pi. Postmortem immunohistochemical analysis of the hippocampus and pons showed TSPO expression in cells expressing ionized calcium-binding adaptor molecule 1 (IBA1), glial fibrillary acidic protein (GFAP), and collagen IV. In the hippocampus of SARS-CoV-2 infected animals the TSPO+ area and number of TSPO+ cells were significantly increased compared to control animals. This increase was not cell type specific, since both the number of IBA1+TSPO+ and GFAP+TSPO+ cells was increased, as well as the TSPO+ area within collagen IV+ blood vessels. CONCLUSIONS This study manifests [18F]DPA714 as a powerful radiotracer to visualize SARS-CoV-2 induced neuroinflammation. The increased uptake of [18F]DPA714 over time implies an active neuroinflammatory response following SARS-CoV-2 infection. This inflammatory signal coincides with an increased number of TSPO expressing cells, including glial and endothelial cells, suggesting neuroinflammation and vascular dysregulation. These results demonstrate the long-term neuroinflammatory response following a mild SARS-CoV-2 infection, which potentially precedes long-lasting neurological symptoms.
Collapse
Affiliation(s)
- Juliana M Nieuwland
- Department of Neurobiology and Aging, Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
| | - Erik Nutma
- Department of Neurobiology and Aging, Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
| | - Ingrid H C H M Philippens
- Department of Neurobiology and Aging, Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands
| | - Kinga P Böszörményi
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Edmond J Remarque
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Jaco Bakker
- Department of Radiology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Lisette Meijer
- Department of Radiology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Noor Woerdman
- Department of Radiology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Zahra C Fagrouch
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Babs E Verstrepen
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Jan A M Langermans
- Department of Animal Sciences, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
- Department Population Health Sciences, Unit Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Ernst J Verschoor
- Department of Virology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam (TCA), Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ronald E Bontrop
- Department of Comparative Genetics and Refinement, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
- Department of Biology, Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Marieke A Stammes
- Department of Radiology, Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Jinte Middeldorp
- Department of Neurobiology and Aging, Biomedical Primate Research Centre (BPRC), Lange Kleiweg 161, 2288GJ, Rijswijk, The Netherlands.
| |
Collapse
|
29
|
Butler T, Wang XH, Chiang GC, Li Y, Zhou L, Xi K, Wickramasuriya N, Tanzi E, Spector E, Ozsahin I, Mao X, Razlighi QR, Fung EK, Dyke JP, Maloney T, Gupta A, Raj A, Shungu DC, Mozley PD, Rusinek H, Glodzik L. Choroid Plexus Calcification Correlates with Cortical Microglial Activation in Humans: A Multimodal PET, CT, MRI Study. AJNR Am J Neuroradiol 2023; 44:776-782. [PMID: 37321857 PMCID: PMC10337614 DOI: 10.3174/ajnr.a7903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/04/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND PURPOSE The choroid plexus (CP) within the brain ventricles is well-known to produce cerebrospinal fluid (CSF). Recently, the CP has been recognized as critical in modulating inflammation. MRI-measured CP enlargement has been reported in neuroinflammatory disorders like MS as well as with aging and neurodegeneration. The basis of MRI-measured CP enlargement is unknown. On the basis of tissue studies demonstrating CP calcification as a common pathology associated with aging and disease, we hypothesized that previously unmeasured CP calcification contributes to MRI-measured CP volume and may be more specifically associated with neuroinflammation. MATERIALS AND METHODS We analyzed 60 subjects (43 healthy controls and 17 subjects with Parkinson's disease) who underwent PET/CT using 11C-PK11195, a radiotracer sensitive to the translocator protein expressed by activated microglia. Cortical inflammation was quantified as nondisplaceable binding potential. Choroid plexus calcium was measured via manual tracing on low-dose CT acquired with PET and automatically using a new CT/MRI method. Linear regression assessed the contribution of choroid plexus calcium, age, diagnosis, sex, overall volume of the choroid plexus, and ventricle volume to cortical inflammation. RESULTS Fully automated choroid plexus calcium quantification was accurate (intraclass correlation coefficient with manual tracing = .98). Subject age and choroid plexus calcium were the only significant predictors of neuroinflammation. CONCLUSIONS Choroid plexus calcification can be accurately and automatically quantified using low-dose CT and MRI. Choroid plexus calcification-but not choroid plexus volume-predicted cortical inflammation. Previously unmeasured choroid plexus calcium may explain recent reports of choroid plexus enlargement in human inflammatory and other diseases. Choroid plexus calcification may be a specific and relatively easily acquired biomarker for neuroinflammation and choroid plexus pathology in humans.
Collapse
Affiliation(s)
- T Butler
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - X H Wang
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - G C Chiang
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - Y Li
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - L Zhou
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - K Xi
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - N Wickramasuriya
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - E Tanzi
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - E Spector
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - I Ozsahin
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - X Mao
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - Q R Razlighi
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - E K Fung
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - J P Dyke
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - T Maloney
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - A Gupta
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| | - A Raj
- Department of Radiology (A.R.), University of California, San Francisco, San Francisco, California
| | - D C Shungu
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - P D Mozley
- Department of Radiology (X.M., E.K.F., J.P.D., D.C.S., P.D.M.), Weill Cornell Medicine, New York, New York
| | - H Rusinek
- Department of Radiology (H.R.), New York University, New York, New York
| | - L Glodzik
- From the Brain Health Imaging Institute (T.B., X.H.W., G.C.C., Y.L., L.Z., K.X., N.W., E.T., E.S., I.O., X.M., Q.R.R., T.M., A.G., L.G.)
| |
Collapse
|
30
|
Syed AU, Liang C, Patel KK, Mondal R, Kamalia VM, Moran TR, Ahmed ST, Mukherjee J. Comparison of Monoamine Oxidase-A, Aβ Plaques, Tau, and Translocator Protein Levels in Postmortem Human Alzheimer's Disease Brain. Int J Mol Sci 2023; 24:10808. [PMID: 37445985 PMCID: PMC10341404 DOI: 10.3390/ijms241310808] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Increased monoamine oxidase-A (MAO-A) activity in Alzheimer's disease (AD) may be detrimental to the point of neurodegeneration. To assess MAO-A activity in AD, we compared four biomarkers, Aβ plaques, tau, translocator protein (TSPO), and MAO-A in postmortem AD. Radiotracers were [18F]FAZIN3 for MAO-A, [18F]flotaza and [125I]IBETA for Aβ plaques, [124/125I]IPPI for tau, and [18F]FEPPA for TSPO imaging. Brain sections of the anterior cingulate (AC; gray matter GM) and corpus callosum (CC; white matter WM) from cognitively normal control (CN, n = 6) and AD (n = 6) subjects were imaged using autoradiography and immunostaining. Using competition with clorgyline and (R)-deprenyl, the binding of [18F]FAZIN3 was confirmed to be selective to MAO-A levels in the AD brain sections. Increases in MAO-A, Aβ plaque, tau, and TSPO activity were found in the AD brains compared to the control brains. The [18F]FAZIN3 ratio in AD GM versus CN GM was 2.80, suggesting a 180% increase in MAO-A activity. Using GM-to-WM ratios of AD versus CN, a >50% increase in MAO-A activity was observed (AD/CN = 1.58). Linear positive correlations of [18F]FAZIN3 with [18F]flotaza, [125I]IBETA, and [125I]IPPI were measured and suggested an increase in MAO-A activity with increases in Aβ plaques and tau activity. Our results support the finding that MAO-A activity is elevated in the anterior cingulate cortex in AD and thus may provide a new biomarker for AD in this brain region.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
31
|
Vicente-Rodríguez M, Mancuso R, Peris-Yague A, Simmons C, Gómez-Nicola D, Perry VH, Turkheimer F, Lovestone S, Parker CA, Cash D. Pharmacological modulation of TSPO in microglia/macrophages and neurons in a chronic neurodegenerative model of prion disease. J Neuroinflammation 2023; 20:92. [PMID: 37032328 PMCID: PMC10084680 DOI: 10.1186/s12974-023-02769-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 03/20/2023] [Indexed: 04/11/2023] Open
Abstract
Neuroinflammation is an important component of many neurodegenerative diseases, whether as a primary cause or a secondary outcome. For that reason, either as diagnostic tools or to monitor progression and/or pharmacological interventions, there is a need for robust biomarkers of neuroinflammation in the brain. Mitochondrial TSPO (18 kDa Translocator protein) is one of few available biomarkers of neuroinflammation for which there are clinically available PET imaging agents. In this study, we further characterised neuroinflammation in a mouse model of prion-induced chronic neurodegeneration (ME7) including a pharmacological intervention via a CSF1R inhibitor. This was achieved by autoradiographic binding of the second-generation TSPO tracer, [3H]PBR28, along with a more comprehensive examination of the cellular contributors to the TSPO signal changes by immunohistochemistry. We observed regional increases of TSPO in the ME7 mouse brains, particularly in the hippocampus, cortex and thalamus. This increased TSPO signal was detected in the cells of microglia/macrophage lineage as well as in astrocytes, endothelial cells and neurons. Importantly, we show that the selective CSF1R inhibitor, JNJ-40346527 (JNJ527), attenuated the disease-dependent increase in TSPO signal, particularly in the dentate gyrus of the hippocampus, where JNJ527 attenuated the number of Iba1+ microglia and neurons, but not GFAP+ astrocytes or endothelial cells. These findings suggest that [3H]PBR28 quantitative autoradiography in combination with immunohistochemistry are important translational tools for detecting and quantifying neuroinflammation, and its treatments, in neurodegenerative disease. Furthermore, we demonstrate that although TSPO overexpression in the ME7 brains was driven by various cell types, the therapeutic effect of the CSF1R inhibitor was primarily to modulate TSPO expression in microglia and neurons, which identifies an important route of biological action of this particular CSF1R inhibitor and provides an example of a cell-specific effect of this type of therapeutic agent on the neuroinflammatory process.
Collapse
Affiliation(s)
- Marta Vicente-Rodríguez
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK.
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain.
| | - Renzo Mancuso
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Alba Peris-Yague
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Camilla Simmons
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
| | - Diego Gómez-Nicola
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - V Hugh Perry
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Federico Turkheimer
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
| | - Simon Lovestone
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- Janssen Medical Ltd, High Wycombe, UK
| | - Christine A Parker
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
- GlaxoSmithKline, Stevenage, London, UK
| | - Diana Cash
- Department of Neuroimaging, BRAIN Centre (Biomarker Research and Imaging for Neuroscience), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- The Wellcome Trust Consortium for the Neuroimmunology of Mood Disorders and Alzheimer's Disease (NIMA), London, UK
| |
Collapse
|
32
|
Ferrari-Souza JP, Lussier FZ, Leffa DT, Therriault J, Tissot C, Bellaver B, Ferreira PC, Malpetti M, Wang YT, Povala G, Benedet AL, Ashton NJ, Chamoun M, Servaes S, Bezgin G, Kang MS, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, O’Brien JT, Rowe JB, Cohen AD, Lopez OL, Tudorascu DL, Karikari TK, Klunk WE, Villemagne VL, Soucy JP, Gauthier S, Souza DO, Zetterberg H, Blennow K, Zimmer ER, Rosa-Neto P, Pascoal TA. APOEε4 associates with microglial activation independently of Aβ plaques and tau tangles. SCIENCE ADVANCES 2023; 9:eade1474. [PMID: 37018391 PMCID: PMC10075966 DOI: 10.1126/sciadv.ade1474] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/02/2023] [Indexed: 06/01/2023]
Abstract
Animal studies suggest that the apolipoprotein E ε4 (APOEε4) allele is a culprit of early microglial activation in Alzheimer's disease (AD). Here, we tested the association between APOEε4 status and microglial activation in living individuals across the aging and AD spectrum. We studied 118 individuals with positron emission tomography for amyloid-β (Aβ; [18F]AZD4694), tau ([18F]MK6240), and microglial activation ([11C]PBR28). We found that APOEε4 carriers presented increased microglial activation relative to noncarriers in early Braak stage regions within the medial temporal cortex accounting for Aβ and tau deposition. Furthermore, microglial activation mediated the Aβ-independent effects of APOEε4 on tau accumulation, which was further associated with neurodegeneration and clinical impairment. The physiological distribution of APOE mRNA expression predicted the patterns of APOEε4-related microglial activation in our population, suggesting that APOE gene expression may regulate the local vulnerability to neuroinflammation. Our results support that the APOEε4 genotype exerts Aβ-independent effects on AD pathogenesis by activating microglia in brain regions associated with early tau deposition.
Collapse
Affiliation(s)
- João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Firoza Z. Lussier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Douglas T. Leffa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- ADHD Outpatient Program and Development Psychiatry Program, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Cécile Tissot
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Maura Malpetti
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Guilherme Povala
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andréa L. Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Artificial Intelligence and Computational Neurosciences lab, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nina Margherita Poltronetti
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - John T. O’Brien
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B. Rowe
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Ann D. Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas K. Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Diogo O. Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Tharick A. Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Chaudhran PA, Sharma A. Progress in the Development of Imidazopyridine-Based Fluorescent Probes for Diverse Applications. Crit Rev Anal Chem 2022; 54:2148-2165. [PMID: 36562726 DOI: 10.1080/10408347.2022.2158720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Different classes of Imidazopyridine i.e., Imidazo[1,2-a]pyridine, Imidazo[1,5-a] pyridine, Imidazo[4,5-b]pyridine, have shown versatile applications in various fields. In this review, we have concisely presented the usefulness of the fluorescent property of imidazopyridine in different fields such as imaging tools, optoelectronics, metal ion detection, etc. Fluorescence mechanisms such as excited state intramolecular proton transfer, photoinduced electron transfer, fluorescence resonance energy transfer, intramolecular charge transfer, etc. are incorporated in the designed fluorophore to make it for fluorescent applications. It has been widely employed for metal ion detection, where selective metal ion detection is possible with triazole-attached imidazopyridine, β-carboline imidazopyridine hybrid, quinoline conjugated imidazopyridine, and many more. Also, other popular applications involve organic light emitting diodes and cell imaging. This review shed a light on recent development in this area especially focusing on the optical properties of the molecules with their usage which would be helpful in designing application-based new imidazopyridine derivatives.
Collapse
Affiliation(s)
- Preeti AshokKumar Chaudhran
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, India
| | - Abha Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, India
| |
Collapse
|
34
|
Kosyreva AM, Sentyabreva AV, Tsvetkov IS, Makarova OV. Alzheimer’s Disease and Inflammaging. Brain Sci 2022; 12:brainsci12091237. [PMID: 36138973 PMCID: PMC9496782 DOI: 10.3390/brainsci12091237] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 09/10/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer’s disease is one of the most common age-related neurodegenerative disorders. The main theory of Alzheimer’s disease progress is the amyloid-β cascade hypothesis. However, the initial mechanisms of insoluble forms of amyloid-β formation and hyperphosphorylated tau protein in neurons remain unclear. One of the factors, which might play a key role in senile plaques and tau fibrils generation due to Alzheimer’s disease, is inflammaging, i.e., systemic chronic low-grade age-related inflammation. The activation of the proinflammatory cell phenotype is observed during aging, which might be one of the pivotal mechanisms for the development of chronic inflammatory diseases, e.g., atherosclerosis, metabolic syndrome, type 2 diabetes mellitus, and Alzheimer’s disease. This review discusses the role of the inflammatory processes in developing neurodegeneration, activated during physiological aging and due to various diseases such as atherosclerosis, obesity, type 2 diabetes mellitus, and depressive disorders.
Collapse
|
35
|
Butler T, Glodzik L, Wang XH, Xi K, Li Y, Pan H, Zhou L, Chiang GCY, Morim S, Wickramasuriya N, Tanzi E, Maloney T, Harvey P, Mao X, Razlighi QR, Rusinek H, Shungu DC, de Leon M, Atwood CS, Mozley PD. Positron Emission Tomography reveals age-associated hypothalamic microglial activation in women. Sci Rep 2022; 12:13351. [PMID: 35922659 PMCID: PMC9349172 DOI: 10.1038/s41598-022-17315-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
In rodents, hypothalamic inflammation plays a critical role in aging and age-related diseases. Hypothalamic inflammation has not previously been assessed in vivo in humans. We used Positron Emission Tomography (PET) with a radiotracer sensitive to the translocator protein (TSPO) expressed by activated microglia, to assess correlations between age and regional brain TSPO in a group of healthy subjects (n = 43, 19 female, aged 23-78), focusing on hypothalamus. We found robust age-correlated TSPO expression in thalamus but not hypothalamus in the combined group of women and men. This pattern differs from what has been described in rodents. Prominent age-correlated TSPO expression in thalamus in humans, but in hypothalamus in rodents, could reflect evolutionary changes in size and function of thalamus versus hypothalamus, and may be relevant to the appropriateness of using rodents to model human aging. When examining TSPO PET results in women and men separately, we found that only women showed age-correlated hypothalamic TSPO expression. We suggest this novel result is relevant to understanding a stark sex difference in human aging: that only women undergo loss of fertility-menopause-at mid-life. Our finding of age-correlated hypothalamic inflammation in women could have implications for understanding and perhaps altering reproductive aging in women.
Collapse
Affiliation(s)
- Tracy Butler
- Department of Radiology, Weill Cornell Medicine, New York, USA.
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA.
| | - Lidia Glodzik
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Xiuyuan Hugh Wang
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Ke Xi
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Yi Li
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Hong Pan
- Department of Psychiatry, Brigham and Women's Hospital, Boston, USA
| | - Liangdong Zhou
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | | | - Simon Morim
- Department of Radiology, Weill Cornell Medicine, New York, USA
| | - Nimmi Wickramasuriya
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Emily Tanzi
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Thomas Maloney
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Patrick Harvey
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Xiangling Mao
- Department of Radiology, Weill Cornell Medicine, New York, USA
| | - Qolamreza Ray Razlighi
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Henry Rusinek
- Department of Radiology, New York University, New York, USA
| | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medicine, New York, USA
| | - Mony de Leon
- Department of Radiology, Weill Cornell Medicine, New York, USA
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 405 E 61st St, New York, NY, 10065, USA
| | - Craig S Atwood
- Department of Gerontology, University of Wisconsin, Madison, Madison, USA
| | - P David Mozley
- Department of Radiology, Weill Cornell Medicine, New York, USA
| |
Collapse
|
36
|
Gu X, Lai D, Liu S, Chen K, Zhang P, Chen B, Huang G, Cheng X, Lu C. Hub Genes, Diagnostic Model, and Predicted Drugs Related to Iron Metabolism in Alzheimer's Disease. Front Aging Neurosci 2022; 14:949083. [PMID: 35875800 PMCID: PMC9300955 DOI: 10.3389/fnagi.2022.949083] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disease, remains unclear in terms of its underlying causative genes and effective therapeutic approaches. Meanwhile, abnormalities in iron metabolism have been demonstrated in patients and mouse models with AD. Therefore, this study sought to find hub genes based on iron metabolism that can influence the diagnosis and treatment of AD. First, gene expression profiles were downloaded from the GEO database, including non-demented (ND) controls and AD samples. Fourteen iron metabolism-related gene sets were downloaded from the MSigDB database, yielding 520 iron metabolism-related genes. The final nine hub genes associated with iron metabolism and AD were obtained by differential analysis and WGCNA in brain tissue samples from GSE132903. GO analysis revealed that these genes were mainly involved in two major biological processes, autophagy and iron metabolism. Through stepwise regression and logistic regression analyses, we selected four of these genes to construct a diagnostic model of AD. The model was validated in blood samples from GSE63061 and GSE85426, and the AUC values showed that the model had a relatively good diagnostic performance. In addition, the immune cell infiltration of the samples and the correlation of different immune factors with these hub genes were further explored. The results suggested that these genes may also play an important role in immunity to AD. Finally, eight drugs targeting these nine hub genes were retrieved from the DrugBank database, some of which were shown to be useful for the treatment of AD or other concomitant conditions, such as insomnia and agitation. In conclusion, this model is expected to guide the diagnosis of patients with AD by detecting the expression of several genes in the blood. These hub genes may also assist in understanding the development and drug treatment of AD.
Collapse
Affiliation(s)
- Xuefeng Gu
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Xuefeng Gu
| | - Donglin Lai
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Shuang Liu
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Kaijie Chen
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Peng Zhang
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Bing Chen
- Department of Neurosurgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Gang Huang
| | - Xiaoqin Cheng
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- Xiaoqin Cheng
| | - Changlian Lu
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- *Correspondence: Changlian Lu
| |
Collapse
|
37
|
Masdeu JC, Pascual B, Fujita M. Imaging Neuroinflammation in Neurodegenerative Disorders. J Nucl Med 2022; 63:45S-52S. [PMID: 35649654 DOI: 10.2967/jnumed.121.263200] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation plays a major role in the etiopathology of neurodegenerative diseases, including Alzheimer and Parkinson diseases, frontotemporal lobar degeneration, and amyotrophic lateral sclerosis. In vivo monitoring of neuroinflammation using PET is critical to understand this process, and data are accumulating in this regard, thus a review is useful. From PubMed, we retrieved publications using any of the available PET tracers to image neuroinflammation in humans as well as selected articles dealing with experimental animal models or the chemistry of currently used or potential radiotracers. We reviewed 280 articles. The most common PET neuroinflammation target, translocator protein (TSPO), has limitations, lacking cellular specificity and the ability to separate neuroprotective from neurotoxic inflammation. However, TSPO PET is useful to define the amount and location of inflammation in the brain of people with neurodegenerative disorders. We describe the characteristics of TSPO and other potential PET neuroinflammation targets and PET tracers available or in development. Despite target and tracer limitations, in recent years there has been a sharp increase in the number of reports of neuroinflammation PET in humans. The most studied has been Alzheimer disease, in which neuroinflammation seems initially neuroprotective and neurotoxic later in the progression of the disease. We describe the findings in all the major neurodegenerative disorders. Neuroinflammation PET is an indispensable tool to understand the process of neurodegeneration, particularly in humans, as well as to validate target engagement in therapeutic clinical trials.
Collapse
Affiliation(s)
- Joseph C Masdeu
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and
| | - Belen Pascual
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and
| | - Masahiro Fujita
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and.,PET Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas
| |
Collapse
|
38
|
Murray J, Meloni G, Cortes EP, KimSilva A, Jacobs M, Ramkissoon A, Crary JF, Morgello S. Frontal lobe microglia, neurodegenerative protein accumulation, and cognitive function in people with HIV. Acta Neuropathol Commun 2022; 10:69. [PMID: 35526056 PMCID: PMC9080134 DOI: 10.1186/s40478-022-01375-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Microglia are implicated in Alzheimer's Disease (AD) pathogenesis. In a middle-aged cohort enriched for neuroinflammation, we asked whether microgliosis was related to neocortical amyloid beta (A[Formula: see text]) deposition and neuronal phosphorylated tau (p-tau), and whether microgliosis predicted cognition. Frontal lobe tissue from 191 individuals autopsied with detectable (HIV-D) and undetectable (HIV-U) HIV infection, and 63 age-matched controls were examined. Immunohistochemistry (IHC) was used to evaluate A[Formula: see text] plaques and neuronal p-tau, and quantitate microgliosis with markers Iba1, CD163, and CD68 in large regions of cortex. Glia in the A[Formula: see text] plaque microenvironment were quantitated by immunofluorescence (IF). The relationship of microgliosis to cognition was evaluated. No relationship between A[Formula: see text] or p-tau accumulation and overall severity of microgliosis was discerned. Individuals with uncontrolled HIV had the greatest microgliosis, but fewer A[Formula: see text] plaques; they also had higher prevalence of APOE [Formula: see text]4 alleles, but died earlier than other groups. HIV group status was the only variable predicting microgliosis over large frontal regions. In contrast, in the A[Formula: see text] plaque microenvironment, APOE [Formula: see text]4 status and sex were dominant predictors of glial infiltrates, with smaller contributions of HIV status. Cognition correlated with large-scale microgliosis in HIV-D, but not HIV-U, individuals. In this autopsy cohort, over large regions of cortex, HIV status predicts microgliosis, whereas in the A[Formula: see text] plaque microenvironment, traditional risk factors of AD (APOE [Formula: see text]4 and sex) are stronger determinants. While microgliosis does not predict neurodegenerative protein deposition, it does predict cognition in HIV-D. Increased neuroinflammation does not initiate amyloid deposition in a younger group with enhanced genetic risk. However, once A[Formula: see text] deposits are established, APOE [Formula: see text]4 predicts increased plaque-associated inflammation.
Collapse
Affiliation(s)
- Jacinta Murray
- Department of Neurology, The Icahn School of Medicine at Mount Sinai, Box 1137, Mount Sinai Medical Center, New York City, NY, 10029, USA
| | - Gregory Meloni
- Department of Neurology, The Icahn School of Medicine at Mount Sinai, Box 1137, Mount Sinai Medical Center, New York City, NY, 10029, USA
| | - Etty P Cortes
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Ariadna KimSilva
- Department of Neurology, The Icahn School of Medicine at Mount Sinai, Box 1137, Mount Sinai Medical Center, New York City, NY, 10029, USA
| | - Michelle Jacobs
- Department of Neurology, The Icahn School of Medicine at Mount Sinai, Box 1137, Mount Sinai Medical Center, New York City, NY, 10029, USA
| | - Alyssa Ramkissoon
- Department of Neurology, The Icahn School of Medicine at Mount Sinai, Box 1137, Mount Sinai Medical Center, New York City, NY, 10029, USA
| | - John F Crary
- Department of Neuroscience, The Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Artificial Intelligence and Human Health, Ronald M. Loeb Center for Alzheimer's Disease, The Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Susan Morgello
- Department of Neurology, The Icahn School of Medicine at Mount Sinai, Box 1137, Mount Sinai Medical Center, New York City, NY, 10029, USA.
- Department of Neuroscience, The Friedman Brain Institute, The Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
39
|
Nicotinic Acetylcholine Receptors and Microglia as Therapeutic and Imaging Targets in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092780. [PMID: 35566132 PMCID: PMC9102429 DOI: 10.3390/molecules27092780] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022]
Abstract
Amyloid-β (Aβ) accumulation and tauopathy are considered the pathological hallmarks of Alzheimer’s disease (AD), but attenuation in choline signaling, including decreased nicotinic acetylcholine receptors (nAChRs), is evident in the early phase of AD. Currently, there are no drugs that can suppress the progression of AD due to a limited understanding of AD pathophysiology. For this, diagnostic methods that can assess disease progression non-invasively before the onset of AD symptoms are essential, and it would be valuable to incorporate the concept of neurotheranostics, which simultaneously enables diagnosis and treatment. The neuroprotective pathways activated by nAChRs are attractive targets as these receptors may regulate microglial-mediated neuroinflammation. Microglia exhibit both pro- and anti-inflammatory functions that could be modulated to mitigate AD pathogenesis. Currently, single-cell analysis is identifying microglial subpopulations that may have specific functions in different stages of AD pathologies. Thus, the ability to image nAChRs and microglia in AD according to the stage of the disease in the living brain may lead to the development of new diagnostic and therapeutic methods. In this review, we summarize and discuss the recent findings on the nAChRs and microglia, as well as their methods for live imaging in the context of diagnosis, prophylaxis, and therapy for AD.
Collapse
|
40
|
Inflammation in dementia with Lewy bodies. Neurobiol Dis 2022; 168:105698. [DOI: 10.1016/j.nbd.2022.105698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/03/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
|
41
|
Garland EF, Hartnell IJ, Boche D. Microglia and Astrocyte Function and Communication: What Do We Know in Humans? Front Neurosci 2022; 16:824888. [PMID: 35250459 PMCID: PMC8888691 DOI: 10.3389/fnins.2022.824888] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia and astrocytes play essential roles in the central nervous system contributing to many functions including homeostasis, immune response, blood-brain barrier maintenance and synaptic support. Evidence has emerged from experimental models of glial communication that microglia and astrocytes influence and coordinate each other and their effects on the brain environment. However, due to the difference in glial cells between humans and rodents, it is essential to confirm the relevance of these findings in human brains. Here, we aim to review the current knowledge on microglia-astrocyte crosstalk in humans, exploring novel methodological techniques used in health and disease conditions. This will include an in-depth look at cell culture and iPSCs, post-mortem studies, imaging and fluid biomarkers, genetics and transcriptomic data. In this review, we will discuss the advantages and limitations of these methods, highlighting the understanding these methods have brought the field on these cells communicative abilities, and the knowledge gaps that remain.
Collapse
Affiliation(s)
| | | | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
42
|
Harada R, Furumoto S, Kudo Y, Yanai K, Villemagne VL, Okamura N. Imaging of Reactive Astrogliosis by Positron Emission Tomography. Front Neurosci 2022; 16:807435. [PMID: 35210989 PMCID: PMC8862631 DOI: 10.3389/fnins.2022.807435] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Many neurodegenerative diseases are neuropathologically characterized by neuronal loss, gliosis, and the deposition of misfolded proteins such as β-amyloid (Aβ) plaques and tau tangles in Alzheimer’s disease (AD). In postmortem AD brains, reactive astrocytes and activated microglia are observed surrounding Aβ plaques and tau tangles. These activated glial cells secrete pro-inflammatory cytokines and reactive oxygen species, which may contribute to neurodegeneration. Therefore, in vivo imaging of glial response by positron emission tomography (PET) combined with Aβ and tau PET would provide new insights to better understand the disease process, as well as aid in the differential diagnosis, and monitoring glial response disease-specific therapeutics. There are two promising targets proposed for imaging reactive astrogliosis: monoamine oxidase-B (MAO-B) and imidazoline2 binding site (I2BS), which are predominantly expressed in the mitochondrial membranes of astrocytes and are upregulated in various neurodegenerative conditions. PET tracers targeting these two MAO-B and I2BS have been evaluated in humans. [18F]THK-5351, which was originally designed to target tau aggregates in AD, showed high affinity for MAO-B and clearly visualized reactive astrocytes in progressive supranuclear palsy (PSP). However, the lack of selectivity of [18F]THK-5351 binding to both MAO-B and tau, severely limits its clinical utility as a biomarker. Recently, [18F]SMBT-1 was developed as a selective and reversible MAO-B PET tracer via compound optimization of [18F]THK-5351. In this review, we summarize the strategy underlying molecular imaging of reactive astrogliosis and clinical studies using MAO-B and I2BS PET tracers.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
- *Correspondence: Ryuichi Harada,
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Department of New Therapeutics Innovation for Alzheimer’s and Dementia, Institute of Development and Aging, Tohoku University, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Victor L. Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Nobuyuki Okamura,
| |
Collapse
|
43
|
Muñoz-Castro C, Noori A, Magdamo CG, Li Z, Marks JD, Frosch MP, Das S, Hyman BT, Serrano-Pozo A. Cyclic multiplex fluorescent immunohistochemistry and machine learning reveal distinct states of astrocytes and microglia in normal aging and Alzheimer's disease. J Neuroinflammation 2022; 19:30. [PMID: 35109872 PMCID: PMC8808995 DOI: 10.1186/s12974-022-02383-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Astrocytes and microglia react to Aβ plaques, neurofibrillary tangles, and neurodegeneration in the Alzheimer's disease (AD) brain. Single-nuclei and single-cell RNA-seq have revealed multiple states or subpopulations of these glial cells but lack spatial information. We have developed a methodology of cyclic multiplex fluorescent immunohistochemistry on human postmortem brains and image analysis that enables a comprehensive morphological quantitative characterization of astrocytes and microglia in the context of their spatial relationships with plaques and tangles. METHODS Single FFPE sections from the temporal association cortex of control and AD subjects were subjected to 8 cycles of multiplex fluorescent immunohistochemistry, including 7 astroglial, 6 microglial, 1 neuronal, Aβ, and phospho-tau markers. Our analysis pipeline consisted of: (1) image alignment across cycles; (2) background subtraction; (3) manual annotation of 5172 ALDH1L1+ astrocytic and 6226 IBA1+ microglial profiles; (4) local thresholding and segmentation of profiles; (5) machine learning on marker intensity data; and (6) deep learning on image features. RESULTS Spectral clustering identified three phenotypes of astrocytes and microglia, which we termed "homeostatic," "intermediate," and "reactive." Reactive and, to a lesser extent, intermediate astrocytes and microglia were closely associated with AD pathology (≤ 50 µm). Compared to homeostatic, reactive astrocytes contained substantially higher GFAP and YKL-40, modestly elevated vimentin and TSPO as well as EAAT1, and reduced GS. Intermediate astrocytes had markedly increased EAAT2, moderately increased GS, and intermediate GFAP and YKL-40 levels. Relative to homeostatic, reactive microglia showed increased expression of all markers (CD68, ferritin, MHC2, TMEM119, TSPO), whereas intermediate microglia exhibited increased ferritin and TMEM119 as well as intermediate CD68 levels. Machine learning models applied on either high-plex signal intensity data (gradient boosting machines) or directly on image features (convolutional neural networks) accurately discriminated control vs. AD diagnoses at the single-cell level. CONCLUSIONS Cyclic multiplex fluorescent immunohistochemistry combined with machine learning models holds promise to advance our understanding of the complexity and heterogeneity of glial responses as well as inform transcriptomics studies. Three distinct phenotypes emerged with our combination of markers, thus expanding the classic binary "homeostatic vs. reactive" classification to a third state, which could represent "transitional" or "resilient" glia.
Collapse
Affiliation(s)
- Clara Muñoz-Castro
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Sevilla, Spain
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Ayush Noori
- Harvard College, Boston, MA, 02138, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
| | - Colin G Magdamo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
| | - Zhaozhi Li
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
| | - Jordan D Marks
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- MassGeneral Institute for Neurodegenerative Disease, 114 16th Street, Charlestown, MA, 02129, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
44
|
Gouilly D, Saint-Aubert L, Ribeiro MJ, Salabert AS, Tauber C, Péran P, Arlicot N, Pariente J, Payoux P. Neuroinflammation PET imaging of the translocator protein (TSPO) in Alzheimer's disease: an update. Eur J Neurosci 2022; 55:1322-1343. [PMID: 35083791 DOI: 10.1111/ejn.15613] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/28/2022]
Abstract
Neuroinflammation is a significant contributor to Alzheimer's disease (AD). Until now, PET imaging of the translocator protein (TSPO) has been widely used to depict the neuroimmune endophenotype of AD. The aim of this review was to provide an update to the results from 2018 and to advance the characterization of the biological basis of TSPO imaging in AD by re-examining TSPO function and expression and the methodological aspects of interest. Although the biological basis of the TSPO PET signal is obviously related to microglia and astrocytes in AD, the observed process remains uncertain and might not be directly related to neuroinflammation. Further studies are required to re-examine the cellular significance underlying a variation in the PET signal in AD and how it can be impacted by a disease-modifying treatment.
Collapse
Affiliation(s)
- Dominique Gouilly
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Laure Saint-Aubert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Maria-Joao Ribeiro
- Department of Nuclear Medicine, CHU, Tours, France.,UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Anne-Sophie Salabert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| | | | - Patrice Péran
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, France.,Inserm CIC 1415, CHRU, Tours, France
| | - Jérémie Pariente
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU, Toulouse, France.,Center of Clinical Investigations (CIC1436), CHU, Toulouse, France
| | - Pierre Payoux
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, France.,Department of Nuclear Medicine, CHU, Toulouse, France
| |
Collapse
|
45
|
The 18 kDa translocator protein is associated with microglia in the hippocampus of non-demented elderly subjects. AGING BRAIN 2022; 2:100045. [PMID: 36908874 PMCID: PMC9997180 DOI: 10.1016/j.nbas.2022.100045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Increase in the brain expression of the 18 kDa translocator protein (TSPO) is considered as a marker of neuroinflammation in the context of brain diseases, such as Alzheimer's disease (AD). However, in non-demented subjects with Alzheimer's neuropathology, TSPO accumulation in hippocampus subdivisions has not been fully characterized. To determine if TSPO is associated with the presence of amyloid β plaques and/or phosphorylated Tau accumulation, we analyzed hippocampal sections using immunohistochemistry of 14 non-demented subjects with positive staining for Aβ and/or phosphorylated Tau. TSPO expression was heterogenous with higher accumulation in the CA2/3 and subiculum subfields of the hippocampus. Its distribution closely resembled that of the microglial IBA1 marker and of the Aβ42 amyloid form. In addition, positive correlations were observed between TSPO and IBA1 densities in CA4, CA2/3 and the subiculum but not with either the astrocyte GFAP marker or the AD-type Aβ and Tau markers. This study sustains the hypothesis that TSPO is mainly associated with microglia and in Aβ42-rich subdivisions in the hippocampus of non-demented elderly individuals.
Collapse
|
46
|
Harada R. [Is TSPO PET a biomarker of activated microglia?]. Nihon Yakurigaku Zasshi 2022; 157:385. [PMID: 36047160 DOI: 10.1254/fpj.22053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
|
47
|
Delage C, Vignal N, Guerin C, Taib T, Barboteau C, Mamma C, Khacef K, Margaill I, Sarda-Mantel L, Rizzo-Padoin N, Hontonnou F, Marchand-Leroux C, Lerouet D, Hosten B, Besson V. From positron emission tomography to cell analysis of the 18-kDa Translocator Protein in mild traumatic brain injury. Sci Rep 2021; 11:24009. [PMID: 34907268 PMCID: PMC8671393 DOI: 10.1038/s41598-021-03416-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 11/17/2021] [Indexed: 11/24/2022] Open
Abstract
Traumatic brain injury (TBI) leads to a deleterious neuroinflammation, originating from microglial activation. Monitoring microglial activation is an indispensable step to develop therapeutic strategies for TBI. In this study, we evaluated the use of the 18-kDa translocator protein (TSPO) in positron emission tomography (PET) and cellular analysis to monitor microglial activation in a mild TBI mouse model. TBI was induced on male Swiss mice. PET imaging analysis with [18F]FEPPA, a TSPO radiotracer, was performed at 1, 3 and 7 days post-TBI and flow cytometry analysis on brain at 1 and 3 days post-TBI. PET analysis showed no difference in TSPO expression between non-operated, sham-operated and TBI mice. Flow cytometry analysis demonstrated an increase in TSPO expression in ipsilateral brain 3 days post-TBI, especially in microglia, macrophages, lymphocytes and neutrophils. Moreover, microglia represent only 58.3% of TSPO+ cells in the brain. Our results raise the question of the use of TSPO radiotracer to monitor microglial activation after TBI. More broadly, flow cytometry results point the lack of specificity of TSPO for microglia and imply that microglia contribute to the overall increase in TSPO in the brain after TBI, but is not its only contributor.
Collapse
Affiliation(s)
- Clément Delage
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France.
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France.
| | - Nicolas Vignal
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
| | - Coralie Guerin
- Université de Paris, Innovative Therapies in Haemostasis, Inserm, 75006, Paris, France
- Institut Curie, Cytometry Core, 75005, Paris, France
- Université de Paris, Inserm UMS 3612 CNRS - US25 Inserm -Faculté de Pharmacie de Paris, Paris, France
| | - Toufik Taib
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Clément Barboteau
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Célia Mamma
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Kahina Khacef
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Isabelle Margaill
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1140, Paris, France
| | - Laure Sarda-Mantel
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
| | - Nathalie Rizzo-Padoin
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- CHU de Martinique, Service Pharmacie, Hôpital Pierre Zobda-Quitman, Fort-de-France, France
| | - Fortune Hontonnou
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- Université de Paris, Inserm UMR-S 942, Hôpital Lariboisière, Paris, France
| | - Catherine Marchand-Leroux
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Dominique Lerouet
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Benoit Hosten
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service Pharmacie, Hôpital Saint-Louis, Paris, France
| | - Valérie Besson
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| |
Collapse
|
48
|
Aramideh JA, Vidal-Itriago A, Morsch M, Graeber MB. Cytokine Signalling at the Microglial Penta-Partite Synapse. Int J Mol Sci 2021; 22:ijms222413186. [PMID: 34947983 PMCID: PMC8708012 DOI: 10.3390/ijms222413186] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022] Open
Abstract
Microglial cell processes form part of a subset of synaptic contacts that have been dubbed microglial tetra-partite or quad-partite synapses. Since tetrapartite may also refer to the presence of extracellular matrix components, we propose the more precise term microglial penta-partite synapse for synapses that show a microglial cell process in close physical proximity to neuronal and astrocytic synaptic constituents. Microglial cells are now recognised as key players in central nervous system (CNS) synaptic changes. When synaptic plasticity involving microglial penta-partite synapses occurs, microglia may utilise their cytokine arsenal to facilitate the generation of new synapses, eliminate those that are not needed anymore, or modify the molecular and structural properties of the remaining synaptic contacts. In addition, microglia–synapse contacts may develop de novo under pathological conditions. Microglial penta-partite synapses have received comparatively little attention as unique sites in the CNS where microglial cells, cytokines and other factors they release have a direct influence on the connections between neurons and their function. It concerns our understanding of the penta-partite synapse where the confusion created by the term “neuroinflammation” is most counterproductive. The mere presence of activated microglia or the release of their cytokines may occur independent of inflammation, and penta-partite synapses are not usually active in a neuroimmunological sense. Clarification of these details is the main purpose of this review, specifically highlighting the relationship between microglia, synapses, and the cytokines that can be released by microglial cells in health and disease.
Collapse
Affiliation(s)
- Jason Abbas Aramideh
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Andres Vidal-Itriago
- Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (A.V.-I.); (M.M.)
| | - Marco Morsch
- Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (A.V.-I.); (M.M.)
| | - Manuel B. Graeber
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence:
| |
Collapse
|
49
|
Nutma E, Ceyzériat K, Amor S, Tsartsalis S, Millet P, Owen DR, Papadopoulos V, Tournier BB. Cellular sources of TSPO expression in healthy and diseased brain. Eur J Nucl Med Mol Imaging 2021; 49:146-163. [PMID: 33433698 PMCID: PMC8712293 DOI: 10.1007/s00259-020-05166-2] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022]
Abstract
The 18 kDa translocator protein (TSPO) is a highly conserved protein located in the outer mitochondrial membrane. TSPO binding, as measured with positron emission tomography (PET), is considered an in vivo marker of neuroinflammation. Indeed, TSPO expression is altered in neurodegenerative, neuroinflammatory, and neuropsychiatric diseases. In PET studies, the TSPO signal is often viewed as a marker of microglial cell activity. However, there is little evidence in support of a microglia-specific TSPO expression. This review describes the cellular sources and functions of TSPO in animal models of disease and human studies, in health, and in central nervous system diseases. A discussion of methods of analysis and of quantification of TSPO is also presented. Overall, it appears that the alterations of TSPO binding, their cellular underpinnings, and the functional significance of such alterations depend on many factors, notably the pathology or the animal model under study, the disease stage, and the involved brain regions. Thus, further studies are needed to fully determine how changes in TSPO binding occur at the cellular level with the ultimate goal of revealing potential therapeutic pathways.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Division of Nuclear medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
- Division of Radiation Oncology, Department of Oncology, University Hospitals of Geneva, Geneva, Switzerland
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - David R Owen
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
50
|
Guilarte TR, Rodichkin AN, McGlothan JL, Acanda De La Rocha AM, Azzam DJ. Imaging neuroinflammation with TSPO: A new perspective on the cellular sources and subcellular localization. Pharmacol Ther 2021; 234:108048. [PMID: 34848203 DOI: 10.1016/j.pharmthera.2021.108048] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/04/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Translocator Protein 18 kDa (TSPO), previously named Peripheral Benzodiazepine Receptor, is a well-validated and widely used biomarker of neuroinflammation to assess diverse central nervous system (CNS) pathologies in preclinical and clinical studies. Many studies have shown that in animal models of human neurological and neurodegenerative disease and in the human condition, TSPO levels increase in the brain neuropil, and this increase is driven by infiltration of peripheral inflammatory cells and activation of glial cells. Therefore, a clear understanding of the dynamics of the cellular sources of the TSPO response is critically important in the interpretation of Positron Emission Tomography (PET) studies and for understanding the pathophysiology of CNS diseases. Within the normal brain compartment, there are tissues and cells such as the choroid plexus, ependymal cells of the lining of the ventricles, and vascular endothelial cells that also express TSPO at even higher levels than in glial cells. However, there is a paucity of knowledge if these cell types respond and increase TSPO in the diseased brain. These cells do provide a background signal that needs to be accounted for in TSPO-PET imaging studies. More recently, there are reports that TSPO may be expressed in neurons of the adult brain and TSPO expression may be increased by neuronal activity. Therefore, it is essential to study this topic with a great deal of detail, methodological rigor, and rule out alternative interpretations and imaging artifacts. High levels of TSPO are present in the outer mitochondrial membrane. Recent studies have provided evidence of its localization in other cellular compartments including the plasma membrane and perinuclear regions which may define functions that are different from that in mitochondria. A greater understanding of the TSPO subcellular localization in glial cells and infiltrating peripheral immune cells and associated function(s) may provide an additional layer of information to the understanding of TSPO neurobiology. This review is an effort to outline recent advances in understanding the cellular sources and subcellular localization of TSPO in brain cells and to examine remaining questions that require rigorous investigation.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America.
| | - Alexander N Rodichkin
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| | - Jennifer L McGlothan
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| | - Arlet Maria Acanda De La Rocha
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| | - Diana J Azzam
- Brain, Behavior, & the Environment Program, Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, United States of America
| |
Collapse
|