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
|
Kaurani L, Islam MR, Heilbronner U, Krüger DM, Zhou J, Methi A, Strauss J, Pradhan R, Schröder S, Burkhardt S, Schuetz AL, Pena T, Erlebach L, Bühler A, Budde M, Senner F, Kohshour MO, Schulte EC, Schmauß M, Reininghaus EZ, Juckel G, Kronenberg-Versteeg D, Delalle I, Odoardi F, Flügel A, Schulze TG, Falkai P, Sananbenesi F, Fischer A. Regulation of Zbp1 by miR-99b-5p in microglia controls the development of schizophrenia-like symptoms in mice. EMBO J 2024; 43:1420-1444. [PMID: 38528182 PMCID: PMC11021462 DOI: 10.1038/s44318-024-00067-8] [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: 05/03/2023] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 03/27/2024] Open
Abstract
Current approaches to the treatment of schizophrenia have mainly focused on the protein-coding part of the genome; in this context, the roles of microRNAs have received less attention. In the present study, we analyze the microRNAome in the blood and postmortem brains of schizophrenia patients, showing that the expression of miR-99b-5p is downregulated in both the prefrontal cortex and blood of patients. Lowering the amount of miR-99b-5p in mice leads to both schizophrenia-like phenotypes and inflammatory processes that are linked to synaptic pruning in microglia. The microglial miR-99b-5p-supressed inflammatory response requires Z-DNA binding protein 1 (Zbp1), which we identify as a novel miR-99b-5p target. Antisense oligonucleotides against Zbp1 ameliorate the pathological effects of miR-99b-5p inhibition. Our findings indicate that a novel miR-99b-5p-Zbp1 pathway in microglia might contribute to the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany.
| | - Md Rezaul Islam
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
| | - Dennis M Krüger
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Jiayin Zhou
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Aditi Methi
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Judith Strauss
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Ranjit Pradhan
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Sophie Schröder
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Susanne Burkhardt
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Anna-Lena Schuetz
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Tonatiuh Pena
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany
| | - Lena Erlebach
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany; Germany and German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Anika Bühler
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany; Germany and German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Monika Budde
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
| | - Fanny Senner
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
| | - Mojtaba Oraki Kohshour
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
| | - Eva C Schulte
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University Hospital Bonn, Medical Faculty, University of Bonn, Bonn, Germany
| | - Max Schmauß
- Clinic for Psychiatry, Psychotherapy and Psychosomatics, Augsburg University, Medical Faculty, Bezirkskrankenhaus Augsburg, Augsburg, 86156, Germany
| | - Eva Z Reininghaus
- Department of Psychiatry and Psychotherapeutic Medicine, Research Unit for Bipolar Affective Disorder, Medical University of Graz, Graz, 8036, Austria
| | - Georg Juckel
- Department of Psychiatry, Ruhr University Bochum, LWL University Hospital, Bochum, 44791, Germany
| | - Deborah Kronenberg-Versteeg
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany; Germany and German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Ivana Delalle
- Department of Pathology, Lifespan Academic Medical Center, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany.
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.
| | - Farahnaz Sananbenesi
- Research Group for Genome Dynamics in Brain Diseases, 37077, Göttingen, Germany.
| | - Andre Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37077, Göttingen, Germany.
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, 37077, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
3
|
Hartmann SM, Heider J, Wüst R, Fallgatter AJ, Volkmer H. Microglia-neuron interactions in schizophrenia. Front Cell Neurosci 2024; 18:1345349. [PMID: 38510107 PMCID: PMC10950997 DOI: 10.3389/fncel.2024.1345349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Multiple lines of evidence implicate increased neuroinflammation mediated by glial cells to play a key role in neurodevelopmental disorders such as schizophrenia. Microglia, which are the primary innate immune cells of the brain, are crucial for the refinement of the synaptic circuitry during early brain development by synaptic pruning and the regulation of synaptic plasticity during adulthood. Schizophrenia risk factors as genetics or environmental influences may further be linked to increased activation of microglia, an increase of pro-inflammatory cytokine levels and activation of the inflammasome resulting in an overall elevated neuroinflammatory state in patients. Synaptic loss, one of the central pathological hallmarks of schizophrenia, is believed to be due to excess removal of synapses by activated microglia, primarily affecting glutamatergic neurons. Therefore, it is crucial to investigate microglia-neuron interactions, which has been done by multiple studies focusing on post-mortem brain tissues, brain imaging, animal models and patient iPSC-derived 2D culture systems. In this review, we summarize the major findings in patients and in vivo and in vitro models in the context of neuron-microglia interactions in schizophrenia and secondly discuss the potential of anti-inflammatory treatments for the alleviation of positive, negative, and cognitive symptoms.
Collapse
Affiliation(s)
- Sophia-Marie Hartmann
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Johanna Heider
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Richard Wüst
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Andreas J. Fallgatter
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Hansjürgen Volkmer
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
4
|
Laricchiuta D, Papi M, Decandia D, Panuccio A, Cutuli D, Peciccia M, Mazzeschi C, Petrosini L. The role of glial cells in mental illness: a systematic review on astroglia and microglia as potential players in schizophrenia and its cognitive and emotional aspects. Front Cell Neurosci 2024; 18:1358450. [PMID: 38419655 PMCID: PMC10899480 DOI: 10.3389/fncel.2024.1358450] [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: 12/19/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Schizophrenia is a complex and severe mental disorder that affects approximately 1% of the global population. It is characterized by a wide range of symptoms, including delusions, hallucinations, disorganized speech and behavior, and cognitive impairment. Recent research has suggested that the immune system dysregulation may play a significant role in the pathogenesis of schizophrenia, and glial cells, such as astroglia and microglia known to be involved in neuroinflammation and immune regulation, have emerged as potential players in this process. The aim of this systematic review is to summarize the glial hallmarks of schizophrenia, choosing as cellular candidate the astroglia and microglia, and focusing also on disease-associated psychological (cognitive and emotional) changes. We conducted a systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We searched PubMed, Scopus, and Web of Science for articles that investigated the differences in astroglia and microglia in patients with schizophrenia, published in the last 5 years. The present systematic review indicates that changes in the density, morphology, and functioning of astroglia and microglia may be involved in the development of schizophrenia. The glial alterations may contribute to the pathogenesis of schizophrenia by dysregulating neurotransmission and immune responses, worsening cognitive capabilities. The complex interplay of astroglial and microglial activation, genetic/epigenetic variations, and cognitive assessments underscores the intricate relationship between biological mechanisms, symptomatology, and cognitive functioning in schizophrenia.
Collapse
Affiliation(s)
- Daniela Laricchiuta
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Martina Papi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Davide Decandia
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Anna Panuccio
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Debora Cutuli
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Maurizio Peciccia
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Claudia Mazzeschi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Laura Petrosini
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
5
|
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
|
6
|
Uzuegbunam BC, Rummel C, Librizzi D, Culmsee C, Hooshyar Yousefi B. Radiotracers for Imaging of Inflammatory Biomarkers TSPO and COX-2 in the Brain and in the Periphery. Int J Mol Sci 2023; 24:17419. [PMID: 38139248 PMCID: PMC10743508 DOI: 10.3390/ijms242417419] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammation involves the activation of innate immune cells and is believed to play an important role in the development and progression of both infectious and non-infectious diseases such as neurodegeneration, autoimmune diseases, pulmonary and cancer. Inflammation in the brain is marked by the upregulation of translocator protein (TSPO) in microglia. High TSPO levels are also found, for example, in macrophages in cases of rheumatoid arthritis and in malignant tumor cells compared to their relatively low physiological expression. The same applies for cyclooxgenase-2 (COX-2), which is constitutively expressed in the kidney, brain, thymus and gastrointestinal tract, but induced in microglia, macrophages and synoviocytes during inflammation. This puts TSPO and COX-2 in the spotlight as important targets for the diagnosis of inflammation. Imaging modalities, such as positron emission tomography and single-photon emission tomography, can be used to localize inflammatory processes and to track their progression over time. They could also enable the monitoring of the efficacy of therapy and predict its outcome. This review focuses on the current development of PET and SPECT tracers, not only for the detection of neuroinflammation, but also for emerging diagnostic measures in infectious and other non-infectious diseases such as rheumatic arthritis, cancer, cardiac inflammation and in lung diseases.
Collapse
Affiliation(s)
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Gießen, Germany;
- Center for Mind Brain and Behavior, Universities Giessen and Marburg, 35043 Marburg, Germany;
| | - Damiano Librizzi
- Department of Nuclear Medicine, Philipps University of Marburg, 35043 Marburg, Germany;
| | - Carsten Culmsee
- Center for Mind Brain and Behavior, Universities Giessen and Marburg, 35043 Marburg, Germany;
- Institute of Pharmacology and Clinical Pharmacy, Philipps University of Marburg, 35037 Marburg, Germany
| | | |
Collapse
|
7
|
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: 26] [Impact Index Per Article: 13.0] [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
|
8
|
Mawson ER, Morris BJ. A consideration of the increased risk of schizophrenia due to prenatal maternal stress, and the possible role of microglia. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110773. [PMID: 37116354 DOI: 10.1016/j.pnpbp.2023.110773] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Schizophrenia is caused by interaction of a combination of genetic and environmental factors. Of the latter, prenatal exposure to maternal stress is reportedly associated with elevated disease risk. The main orchestrators of inflammatory processes within the brain are microglia, and aberrant microglial activation/function has been proposed to contribute to the aetiology of schizophrenia. Here, we evaluate the epidemiological and preclinical evidence connecting prenatal stress to schizophrenia risk, and consider the possible mediating role of microglia in the prenatal stress-schizophrenia relationship. Epidemiological findings are rather consistent in supporting the association, albeit they are mitigated by effects of sex and gestational timing, while the evidence for microglial activation is more variable. Rodent models of prenatal stress generally report lasting effects on offspring neurobiology. However, many uncertainties remain as to the mechanisms underlying the influence of maternal stress on the developing foetal brain. Future studies should aim to characterise the exact processes mediating this aspect of schizophrenia risk, as well as focussing on how prenatal stress may interact with other risk factors.
Collapse
Affiliation(s)
- Eleanor R Mawson
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Brian J Morris
- School of Psychology and Neuroscience, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
9
|
Raval NR, Wetherill RR, Wiers CE, Dubroff JG, Hillmer AT. Positron Emission Tomography of Neuroimmune Responses in Humans: Insights and Intricacies. Semin Nucl Med 2023; 53:213-229. [PMID: 36270830 PMCID: PMC11261531 DOI: 10.1053/j.semnuclmed.2022.08.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/06/2022]
Abstract
The brain's immune system plays a critical role in responding to immune challenges and maintaining homeostasis. However, dysregulated neuroimmune function contributes to neurodegenerative disease and neuropsychiatric conditions. In vivo positron emission tomography (PET) imaging of the neuroimmune system has facilitated a greater understanding of its physiology and the pathology of some neuropsychiatric conditions. This review presents an in-depth look at PET findings from human neuroimmune function studies, highlighting their importance in current neuropsychiatric research. Although the majority of human PET studies feature radiotracers targeting the translocator protein 18 kDa (TSPO), this review also considers studies with other neuroimmune targets, including monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, nitric oxide synthase, and the purinergic P2X7 receptor. Promising new targets, such as colony-stimulating factor 1, Sphingosine-1-phosphate receptor 1, and the purinergic P2Y12 receptor, are also discussed. The significance of validating neuroimmune targets and understanding their function and expression is emphasized in this review to better identify and interpret PET results.
Collapse
Affiliation(s)
- Nakul R Raval
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT
| | - Reagan R Wetherill
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT; Department of Psychiatry, Yale University, New Haven, CT.
| |
Collapse
|
10
|
Royse SK, Lopresti BJ, Mathis CA, Tollefson S, Narendran R. Beyond monoamines: II. Novel applications for PET imaging in psychiatric disorders. J Neurochem 2023; 164:401-443. [PMID: 35716057 DOI: 10.1111/jnc.15657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/27/2022]
Abstract
Early applications of positron emission tomography (PET) in psychiatry sought to identify derangements of cerebral blood flow and metabolism. The need for more specific neurochemical imaging probes was soon evident, and these probes initially targeted the sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. For nearly 30 years, the centrality of monoamine dysfunction in psychiatric disorders drove the development of an armamentarium of monoaminergic PET radiopharmaceuticals and imaging methodologies. However, continued investments in monoamine-enhancing drug development realized only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely parallelled drug development priorities, resulting in the development of new PET imaging agents for non-monoamine targets. In part two of this review, we survey clinical research studies using the novel targets and radiotracers described in part one across major psychiatric application areas such as substance use disorders, anxiety disorders, eating disorders, personality disorders, mood disorders, and schizophrenia. Important limitations of the studies described are discussed, as well as key methodologic issues, challenges to the field, and the status of clinical trials seeking to exploit these targets for novel therapeutics.
Collapse
Affiliation(s)
- Sarah K Royse
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah Tollefson
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
11
|
Skorobogatov K, Autier V, Foiselle M, Richard JR, Boukouaci W, Wu CL, Raynal S, Carbonne C, Laukens K, Meysman P, Coppens V, le Corvoisier P, Barau C, De Picker L, Morrens M, Tamouza R, Leboyer M. Kynurenine pathway abnormalities are state-specific but not diagnosis-specific in schizophrenia and bipolar disorder. Brain Behav Immun Health 2023; 27:100584. [PMID: 36685639 PMCID: PMC9852293 DOI: 10.1016/j.bbih.2022.100584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Schizophrenia (SCZ) and bipolar disorder (BD) are associated with immunological dysfunctions that have been hypothesized to lead to clinical symptomatology in particular through kynurenine pathway abnormalities. The aim of this study was thus to investigate the impact of serum kynurenine metabolite levels on diagnosis, clinical state, symptom severity and clinical course in a large French transdiagnostic cohort of SCZ and BD patients. Four patient groups (total n = 507) were included in a cross-sectional observational study: 1) hospitalized acute bipolar patients (n = 205); 2) stable bipolar outpatients (n = 116); 3) hospitalized acute schizophrenia patients (n = 111) and 4) stable schizophrenia outpatients (n = 75), in addition to healthy controls (HC) (n = 185). The quantitative determination of serum kynurenine metabolites was performed using liquid chromatography-tandem mass spectrometry. Kynurenine levels were lower in all patients combined compared to HC while ANCOVA analyses did not reveal inter-diagnostic difference between SCZ and BD. Interestingly, hospitalized patients of both diagnostic groups combined displayed significantly lower kynurenine levels than stabilized outpatients. Psychotic symptoms were associated with lower quinaldic acid (F = 9.18, p=<.001), which is KAT-driven, whereas a longer duration of illness contributed to abnormalities in tryptophan (F = 5.41, p = .023), kynurenine (F = 16.93, p=<.001), xanthurenic acid (F = 9.34, p = .002), quinolinic acid (F = 9.18, p = .003) and picolinic acid (F = 4.15, p = .043), metabolized through the KMO-branch. These data confirm illness state rather than diagnosis to drive KP alterations in SCZ and BD. Lower levels of KP metabolites can thus be viewed as a transdiagnostic feature of SCZ and BD, independently associated with acute symptomatology and a longer duration of illness. Quinaldic acid has seldomly been investigated by previous studies and appears an important state marker in SCZ and BD. As serum samples are used in this study, it is not possible to extrapolate these findings to the brain.
Collapse
Affiliation(s)
- Katrien Skorobogatov
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | | | - Marianne Foiselle
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Jean-Romain Richard
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Wahid Boukouaci
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Ching-Lien Wu
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | | | | | - Kris Laukens
- Biomedical Informatics Research Center Antwerp (BIOMINA), University of Antwerp/Antwerp University Hospital, Antwerp, Belgium
- Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
| | - Pieter Meysman
- Biomedical Informatics Research Center Antwerp (BIOMINA), University of Antwerp/Antwerp University Hospital, Antwerp, Belgium
- Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
| | - Violette Coppens
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Philippe le Corvoisier
- Inserm, Centre d'Investigation Clinique 1430, AP-HP, Hôpital Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Caroline Barau
- Plateforme de Ressources Biologiques, HU Henri Mondor, F94010, France
| | - Livia De Picker
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Manuel Morrens
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Ryad Tamouza
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Marion Leboyer
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| |
Collapse
|
12
|
Rodrigues-Neves AC, Ambrósio AF, Gomes CA. Microglia sequelae: brain signature of innate immunity in schizophrenia. Transl Psychiatry 2022; 12:493. [PMID: 36443303 PMCID: PMC9705537 DOI: 10.1038/s41398-022-02197-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Schizophrenia is a psychiatric disorder with significant impact on individuals and society. The current pharmacologic treatment, which principally alleviates psychosis, is focused on neurotransmitters modulation, relying on drugs with severe side effects and ineffectiveness in a significant percentage of cases. Therefore, and due to difficulties inherent to diagnosis and treatment, it is vital to reassess alternative cellular and molecular drug targets. Distinct risk factors - genetic, developmental, epigenetic, and environmental - have been associated with disease onset and progression, giving rise to the proposal of different pathophysiological mechanisms and putative pharmacological targets. Immunity is involved and, particularly microglia - innate immune cells of the central nervous system, critically involved in brain development - have captured attention as cellular players. Microglia undergo marked morphologic and functional alterations in the human disease, as well as in animal models of schizophrenia, as reported in several original papers. We cluster the main findings of clinical studies by groups of patients: (1) at ultra-high risk of psychosis, (2) with a first episode of psychosis or recent-onset schizophrenia, and (3) with chronic schizophrenia; in translational studies, we highlight the time window of appearance of particular microglia alterations in the most well studied animal model in the field (maternal immune activation). The organization of clinical and translational findings based on schizophrenia-associated microglia changes in different phases of the disease course may help defining a temporal pattern of microglia changes and may drive the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- A. Catarina Rodrigues-Neves
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - António. F. Ambrósio
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Catarina A. Gomes
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| |
Collapse
|
13
|
18F-Radiolabeled Translocator Protein (TSPO) PET Tracers: Recent Development of TSPO Radioligands and Their Application to PET Study. Pharmaceutics 2022; 14:pharmaceutics14112545. [PMID: 36432736 PMCID: PMC9697781 DOI: 10.3390/pharmaceutics14112545] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a transmembrane protein in the mitochondrial membrane, which has been identified as a peripheral benzodiazepine receptor. TSPO is generally present at high concentrations in steroid-producing cells and plays an important role in steroid synthesis, apoptosis, and cell proliferation. In the central nervous system, TSPO expression is relatively modest under normal physiological circumstances. However, some pathological disorders can lead to changes in TSPO expression. Overexpression of TSPO is associated with several diseases, such as neurodegenerative diseases, neuroinflammation, brain injury, and cancers. TSPO has therefore become an effective biomarker of related diseases. Positron emission tomography (PET), a non-invasive molecular imaging technique used for the clinical diagnosis of numerous diseases, can detect diseases related to TSPO expression. Several radiolabeled TSPO ligands have been developed for PET. In this review, we describe recent advances in the development of TSPO ligands, and 18F-radiolabeled TSPO in particular, as PET tracers. This review covers pharmacokinetic studies, preclinical and clinical trials of 18F-labeled TSPO PET ligands, and the synthesis of TSPO ligands.
Collapse
|
14
|
van der Geest KSM, Sandovici M, Nienhuis PH, Slart RHJA, Heeringa P, Brouwer E, Jiemy WF. Novel PET Imaging of Inflammatory Targets and Cells for the Diagnosis and Monitoring of Giant Cell Arteritis and Polymyalgia Rheumatica. Front Med (Lausanne) 2022; 9:902155. [PMID: 35733858 PMCID: PMC9207253 DOI: 10.3389/fmed.2022.902155] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/13/2022] [Indexed: 12/26/2022] Open
Abstract
Giant cell arteritis (GCA) and polymyalgia rheumatica (PMR) are two interrelated inflammatory diseases affecting patients above 50 years of age. Patients with GCA suffer from granulomatous inflammation of medium- to large-sized arteries. This inflammation can lead to severe ischemic complications (e.g., irreversible vision loss and stroke) and aneurysm-related complications (such as aortic dissection). On the other hand, patients suffering from PMR present with proximal stiffness and pain due to inflammation of the shoulder and pelvic girdles. PMR is observed in 40-60% of patients with GCA, while up to 21% of patients suffering from PMR are also affected by GCA. Due to the risk of ischemic complications, GCA has to be promptly treated upon clinical suspicion. The treatment of both GCA and PMR still heavily relies on glucocorticoids (GCs), although novel targeted therapies are emerging. Imaging has a central position in the diagnosis of GCA and PMR. While [18F]fluorodeoxyglucose (FDG)-positron emission tomography (PET) has proven to be a valuable tool for diagnosis of GCA and PMR, it possesses major drawbacks such as unspecific uptake in cells with high glucose metabolism, high background activity in several non-target organs and a decrease of diagnostic accuracy already after a short course of GC treatment. In recent years, our understanding of the immunopathogenesis of GCA and, to some extent, PMR has advanced. In this review, we summarize the current knowledge on the cellular heterogeneity in the immunopathology of GCA/PMR and discuss how recent advances in specific tissue infiltrating leukocyte and stromal cell profiles may be exploited as a source of novel targets for imaging. Finally, we discuss prospective novel PET radiotracers that may be useful for the diagnosis and treatment monitoring in GCA and PMR.
Collapse
Affiliation(s)
- Kornelis S. M. van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Pieter H. Nienhuis
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - William F. Jiemy
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Toyohara J, Sakata M, Wagatsuma K, Tago T, Ishibashi K, Ishii K, Elsinga P, Ishiwata K. Test-retest reproducibility of cerebral adenosine A 2A receptor quantification using [ 11C]preladenant. Ann Nucl Med 2022; 36:15-23. [PMID: 34564828 DOI: 10.1007/s12149-021-01678-5] [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: 07/11/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To evaluate the reproducibility of cerebral adenosine A2A receptor (A2AR) quantification using [11C]preladenant ([11C]PLN) and PET in a test-retest study. METHODS Eight healthy male volunteers were enrolled. Dynamic 90 min PET scans were performed twice at the same time of the day to avoid the effect of diurnal variation. Subjects refrained from caffeine from 12 h prior to scanning, and serum caffeine was measured before radioligand injection. Arterial blood was sampled repeatedly during scanning and the fraction of the parent compound in plasma was determined. Total distribution volume (VT) was estimated using 1- and 2-tissue compartment models (1-TCM and 2-TCM, respectively) and Logan graphical analysis (Logan plot) (t* = 30 min). Plasma-free fraction (fP) of [11C]PLN was measured and used for correction of VT values. Distribution volume ratio (DVR) was calculated from VT of target and reference regions and obtained by noninvasive Logan graphical reference tissue model (LGAR) (t* = 30 min). Absolute test-retest variability (aTRV), and intra-class correlation coefficient (ICC) of VT and DVR were calculated as indexes of repeatability. Correlation between DVR and serum concentration of caffeine (a nonselective A2AR blocker) was analyzed by Pearson's correlation analysis. RESULTS Regional time-activity curves were well described by 2-TCM models. Estimation of VT by 2-TCM produced some erroneous values; therefore, the more robust Logan plot was selected as the appropriate model. Global mean aTRV was 20% for VT and 14% for VT/fP (ICC, 0.72 for VT and 0.87 for VT/fP). Global mean aTRV of DVR was 13% for Logan plot and 10% for LGAR (ICC, 0.70 for Logan plot and 0.81 for LGAR). DVR estimates using LGAR and Logan plot were in good agreement (r2 = 0.96). Coefficients of variation for VT, VT/fP, DVR (Logan plot), and DVR (LGAR) were 47%, 47%, 27%, and 18%, respectively. Despite low serum caffeine levels, significant concentration-dependent effects on [11C]PLN binding to target regions were observed (p < 0.01). CONCLUSIONS In this study, moderate test-retest reproducibility and large inter-subject differences were observed with [11C]PLN PET, possibly attributable to competition by baseline amount of caffeine. Analysis of plasma caffeine concentration is recommended during [11C]PLN PET studies. TRIAL REGISTRATION UMIN000030040.
Collapse
Affiliation(s)
- Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
| | - Muneyuki Sakata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kei Wagatsuma
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- School of Allied Health Science, Kitasato University, 1-15-1 Kitasato, Sagamihara, Kanagawa, 252-0373, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kenji Ishibashi
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Philip Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Institute of Cyclotron and Drug Discovery Research, Southern Tohoku Research Institute for Neuroscience, 7-115 Yatsuyamada, Koriyama, Fukushima, 963-8563, Japan
- Department of Biofunctional Imaging, Fukushima Medical University, 1 Hikariga-oka, Fukushima, 960-1295, Japan
| |
Collapse
|
16
|
De Picker LJ, Haarman BCM. Applicability, potential and limitations of TSPO PET imaging as a clinical immunopsychiatry biomarker. Eur J Nucl Med Mol Imaging 2021; 49:164-173. [PMID: 33735406 DOI: 10.1007/s00259-021-05308-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/08/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE TSPO PET imaging may hold promise as a single-step diagnostic work-up for clinical immunopsychiatry. This review paper on the clinical applicability of TSPO PET for primary psychiatric disorders discusses if and why TSPO PET imaging might become the first clinical immunopsychiatry biomarker and the investment prerequisites and scientific advancements needed to accommodate this transition from bench to bedside. METHODS We conducted a systematic search of the literature to identify clinical studies of TSPO PET imaging in patients with primary psychiatric disorders. We included both original case-control studies as well as longitudinal cohort studies of patients with a primary psychiatric diagnosis. RESULTS Thirty-one original studies met our inclusion criteria. In the field of immunopsychiatry, TSPO PET has until now mostly been studied in schizophrenia and related psychotic disorders, and to a lesser extent in mood disorders and neurodevelopmental disorders. Quantitative TSPO PET appears most promising as a predictive biomarker for the transdiagnostic identification of subgroups or disease stages that could benefit from immunological treatments, or as a prognostic biomarker forecasting patients' illness course. Current scanning protocols are still too unreliable, impractical and invasive for clinical use in symptomatic psychiatric patients. CONCLUSION TSPO PET imaging in its present form does not yet offer a sufficiently attractive cost-benefit ratio to become a clinical immunopsychiatry biomarker. Its translation to psychiatric clinical practice will depend on the prioritising of longitudinal research and the establishment of a uniform protocol rendering clinically meaningful TSPO uptake quantification at the shortest possible scan duration without arterial cannulation.
Collapse
Affiliation(s)
- Livia J De Picker
- University Psychiatric Hospital Campus Duffel, Stationsstraat 22C, 2570, Duffel, Belgium.
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Wilrijkstraat 1, 2650, Edegem, Belgium.
| | - Benno C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700RA, Groningen, The Netherlands
- Rob Giel Research Center (RGOc), University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700RA, Groningen, The Netherlands
| |
Collapse
|
17
|
Sepehrizadeh T, Jong I, DeVeer M, Malhotra A. PET/MRI in paediatric disease. Eur J Radiol 2021; 144:109987. [PMID: 34649143 DOI: 10.1016/j.ejrad.2021.109987] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Nuclear medicine and molecular imaging have a small but growing role in the management of paediatric and neonatal diseases. During the past decade, combined PET/MRI has emerged as a clinically important hybrid imaging modality in paediatric medicine due to diagnostic advantages and reduced radiation exposure compared to alternative techniques. The applications for nuclear medicine, radiopharmaceuticals and combined PET/MRI in paediatric diagnosis is broadly similar to adults, however there are some key differences. There are a variety of clinical applications for PET/MRI imaging in children including, but not limited to, oncology, neurology, cardiovascular, infection and chronic inflammatory diseases, and in renal-urological disorders. In this article, we review the applications of PET/MRI in paediatric and neonatal imaging, its current role, advantages and disadvantages over other hybrid imaging techniques such as PET/CT, and its future applications. Overall, PET/MRI is a powerful imaging technology in diagnostic medicine and paediatric diseases. Higher soft tissue contrasts and lower radiation dose of the MRI makes it the superior technology compared to other conventional techniques such as PET/CT or scintigraphy. However, this relatively new hybrid imaging has also some limitations. MRI based attenuation correction remains a challenge and although methodologies have improved significantly in the last decades, most remain under development.
Collapse
Affiliation(s)
| | - Ian Jong
- Department of diagnostic imaging, Monash Health, Melbourne, Australia
| | - Michael DeVeer
- Monash Biomedical Imaging, Monash University, Melbourne, Australia
| | - Atul Malhotra
- Monash Newborn, Monash Children's Hospital, Melbourne, Australia; Department of Paediatrics, Monash University, Melbourne, Australia
| |
Collapse
|
18
|
Skorobogatov K, De Picker L, Verkerk R, Coppens V, Leboyer M, Müller N, Morrens M. Brain Versus Blood: A Systematic Review on the Concordance Between Peripheral and Central Kynurenine Pathway Measures in Psychiatric Disorders. Front Immunol 2021; 12:716980. [PMID: 34630391 PMCID: PMC8495160 DOI: 10.3389/fimmu.2021.716980] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Disturbances in the kynurenine pathway have been implicated in the pathophysiology of psychotic and mood disorders, as well as several other psychiatric illnesses. It remains uncertain however to what extent metabolite levels detectable in plasma or serum reflect brain kynurenine metabolism and other disease-specific pathophysiological changes. The primary objective of this systematic review was to investigate the concordance between peripheral and central (CSF or brain tissue) kynurenine metabolites. As secondary aims we describe their correlation with illness course, treatment response, and neuroanatomical abnormalities in psychiatric diseases. Methods We performed a systematic literature search until February 2021 in PubMed. We included 27 original research articles describing a correlation between peripheral and central kynurenine metabolite measures in preclinical studies and human samples from patients suffering from neuropsychiatric disorders and other conditions. We also included 32 articles reporting associations between peripheral KP markers and symptom severity, CNS pathology or treatment response in schizophrenia, bipolar disorder or major depressive disorder. Results For kynurenine and 3-hydroxykynurenine, moderate to strong concordance was found between peripheral and central concentrations not only in psychiatric disorders, but also in other (patho)physiological conditions. Despite discordant findings for other metabolites (mainly tryptophan and kynurenic acid), blood metabolite levels were associated with clinical symptoms and treatment response in psychiatric patients, as well as with observed neuroanatomical abnormalities and glial activity. Conclusion Only kynurenine and 3-hydroxykynurenine demonstrated a consistent and reliable concordance between peripheral and central measures. Evidence from psychiatric studies on kynurenine pathway concordance is scarce, and more research is needed to determine the validity of peripheral kynurenine metabolite assessment as proxy markers for CNS processes. Peripheral kynurenine and 3-hydroxykynurenine may nonetheless represent valuable predictive and prognostic biomarker candidates for psychiatric disorders.
Collapse
Affiliation(s)
- Katrien Skorobogatov
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| | - Livia De Picker
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| | - Robert Verkerk
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Violette Coppens
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| | - Marion Leboyer
- INSERM U955, Equipe Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental - Hôpital Albert Chenevier - Pôle Psychiatrie, Créteil, France.,AP-HP, Hôpitaux Universitaires Henri Mondor, DHU Pepsy, Pôle de Psychiatrie et d'Addictologie, Créteil, France.,Université Paris Est Créteil, Faculté de Médecine, Creteil, France
| | - Norbert Müller
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, München, Germany
| | - Manuel Morrens
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| |
Collapse
|
19
|
Marques TR, Veronese M, Owen DR, Rabiner EA, Searle GE, Howes OD. Specific and non-specific binding of a tracer for the translocator-specific protein in schizophrenia: an [11C]-PBR28 blocking study. Eur J Nucl Med Mol Imaging 2021; 48:3530-3539. [PMID: 33825022 PMCID: PMC8440284 DOI: 10.1007/s00259-021-05327-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/21/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The mitochondrial 18-kDa translocator protein (TSPO) is expressed by activated microglia and positron emission tomography enables the measurement of TSPO levels in the brain. Findings in schizophrenia have shown to vary depending on the outcome measure used and this discrepancy in TSPO results could be explained by lower non-displaceable binding (VND) in schizophrenia, which could obscure increases in specific binding. In this study, we have used the TSPO ligand XBD173 to block the TSPO radioligand [11C]-PBR28 and used an occupancy plot to quantify VND in patients with schizophrenia. METHODS A total of 7 patients with a diagnosis of schizophrenia were recruited for this study. Each patient received two separate PET scans with [11C]PBR28, one at baseline and one after the administration of the TSPO ligand XBD173. All patients were high-affinity binders (HABs) for the TSPO gene. We used an occupancy plot to quantify the non-displaceable component (VND) using 2TCM kinetic estimates with and without vascular correction. Finally we computed the VND at a single subject level using the SIME method. RESULTS All patients showed a global and generalized reduction in [11C]PBR28 uptake after the administration of XBD173. Constraining the VND to be equal for all patients, the population VND was estimated to be 1.99 mL/cm3 (95% CI 1.90 to 2.08). When we used vascular correction, the fractional TSPO occupancy remained similar. CONCLUSIONS In schizophrenia patients, a substantial component of the [11C]PBR28 signal represents specific binding to TSPO. Furthermore, the VND in patients with schizophrenia is similar to that previously reported in healthy controls. These results suggest that changes in non-specific binding between schizophrenia patients and healthy controls do not account for discrepant PET findings in this disorder.
Collapse
Affiliation(s)
- Tiago Reis Marques
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith Hospital, Imperial College London, London, UK.
- Psychiatric Imaging Group, Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK.
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, King's College London, London, UK
| | - David R Owen
- Division of Brain Sciences, Department of Medicine, Imperial College, London, UK
| | - Eugenii A Rabiner
- Centre for Neuroimaging Sciences, Institute of Psychiatry, King's College London, London, UK
- Invicro, London, UK
| | | | - Oliver D Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith Hospital, Imperial College London, London, UK
- Psychiatric Imaging Group, Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| |
Collapse
|
20
|
Calsbeek JJ, González EA, Boosalis CA, Zolkowska D, Bruun DA, Rowland DJ, Saito NH, Harvey DJ, Chaudhari AJ, Rogawski MA, Garbow JR, Lein PJ. Strain differences in the extent of brain injury in mice after tetramethylenedisulfotetramine-induced status epilepticus. Neurotoxicology 2021; 87:43-50. [PMID: 34478772 PMCID: PMC8595842 DOI: 10.1016/j.neuro.2021.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/28/2021] [Accepted: 08/29/2021] [Indexed: 11/30/2022]
Abstract
Acute intoxication with tetramethylenedisulfotetramine (TETS) can trigger status epilepticus (SE) in humans. Survivors often exhibit long-term neurological effects, including electrographic abnormalities and cognitive deficits, but the pathogenic mechanisms linking the acute toxic effects of TETS to chronic outcomes are not known. Here, we use advanced in vivo imaging techniques to longitudinally monitor the neuropathological consequences of TETS-induced SE in two different mouse strains. Adult male NIH Swiss and C57BL/6J mice were injected with riluzole (10 mg/kg, i.p.), followed 10 min later by an acute dose of TETS (0.2 mg/kg in NIH Swiss; 0.3 mg/kg, i.p. in C57BL/6J) or an equal volume of vehicle (10% DMSO in 0.9% sterile saline). Different TETS doses were administered to trigger comparable seizure behavior between strains. Seizure behavior began within minutes of TETS exposure and rapidly progressed to SE that was terminated after 40 min by administration of midazolam (1.8 mg/kg, i.m.). The brains of vehicle and TETS-exposed mice were imaged using in vivo magnetic resonance (MR) and translocator protein (TSPO) positron emission tomography (PET) at 1, 3, 7, and 14 days post-exposure to monitor brain injury and neuroinflammation, respectively. When the brain scans of TETS mice were compared to those of vehicle controls, subtle and transient neuropathology was observed in both mouse strains, but more extensive and persistent TETS-induced neuropathology was observed in C57BL/6J mice. In addition, one NIH Swiss TETS mouse that did not respond to the midazolam therapy, but remained in SE for more than 2 h, displayed robust neuropathology as determined by in vivo imaging and confirmed by FluoroJade C staining and IBA-1 immunohistochemistry as readouts of neurodegeneration and neuroinflammation, respectively. These findings demonstrate that the extent of injury observed in the mouse brain after TETS-induced SE varied according to strain, dose of TETS and/or the duration of SE. These observations suggest that TETS-intoxicated humans who do not respond to antiseizure medication are at increased risk for brain injury.
Collapse
Affiliation(s)
- Jonas J Calsbeek
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, USA.
| | - Eduardo A González
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, USA.
| | - Casey A Boosalis
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, USA.
| | - Dorota Zolkowska
- Department of Neurology, University of California, Davis, School of Medicine, Davis, CA, 95616, USA.
| | - Donald A Bruun
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, USA.
| | - Douglas J Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA, 95616, USA.
| | - Naomi H Saito
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, CA, 95616, USA.
| | - Danielle J Harvey
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, CA, 95616, USA.
| | - Abhijit J Chaudhari
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA, 95616, USA.
| | - Michael A Rogawski
- Department of Neurology, University of California, Davis, School of Medicine, Davis, CA, 95616, USA.
| | - Joel R Garbow
- Biomedical Magnetic Resonance Laboratory, Mallinckrodt Institute of Radiology, Washington University in St. Louis, School of Medicine, St. Louis, MO, 63110, USA.
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, USA.
| |
Collapse
|
21
|
Abstract
The use of PET imaging agents in oncology, cardiovascular disease, and neurodegenerative disease shows the power of this technique in evaluating the molecular and biological characteristics of numerous diseases. These agents provide crucial information for designing therapeutic strategies for individual patients. Novel PET tracers are in continual development and many have potential use in clinical and research settings. This article discusses the potential applications of tracers in diagnostics, the biological characteristics of diseases, the ability to provide prognostic indicators, and using this information to guide treatment strategies including monitoring treatment efficacy in real time to improve outcomes and survival.
Collapse
|
22
|
The Potential Use of Peripheral Blood Mononuclear Cells as Biomarkers for Treatment Response and Outcome Prediction in Psychiatry: A Systematic Review. Mol Diagn Ther 2021; 25:283-299. [PMID: 33978935 DOI: 10.1007/s40291-021-00516-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Psychiatric disorders have a major impact on the global burden of disease while therapeutic interventions remain insufficient to adequately treat a large number of patients. Regrettably, the efficacy of several psychopharmacological treatment regimens becomes apparent only after 4-6 weeks, and at this point, a significant number of patients present as non-responsive. As such, many patients go weeks/months without appropriate treatment or symptom management. Adequate biomarkers for treatment success and outcome prediction are thus urgently needed. OBJECTIVE With this systematic review, we provide an overview of the use of peripheral blood mononuclear cells (PBMCs) and their signaling pathways in evaluating and/or predicting the effectiveness of different treatment regimens in the course of psychiatric illnesses. We highlight PBMC characteristics that (i) reflect treatment presence, (ii) allow differentiation of responders from non-responders, and (iii) prove predictive at baseline with regard to treatment outcome for a broad range of psychiatric intervention strategies. REVIEW METHODS A PubMed database search was performed to extract papers investigating the relation between any type of PBMC characteristic and treatment presence and/or outcome in patients suffering from severe mental illness. Criteria for eligibility were: written in English; psychiatric diagnosis based on DSM-III-R or newer; PBMC isolation via gradient centrifugation; comparison between treated and untreated patients via PBMC features; sample size ≥ n = 5 per experimental group. Papers not researching in vivo treatment effects between patients and healthy controls, non-clinical trials, and non-hypothesis-/data-driven (e.g., -omics designs) approaches were excluded. DATA SYNTHESIS Twenty-nine original articles were included and qualitatively summarized. Antidepressant and antipsychotic treatments were mostly reflected by intracellular inflammatory markers while intervention with mood stabilizers was evidenced through cell maturation pathways. Lastly, cell viability parameters mirrored predominantly non-pharmacological therapeutic strategies. As for response prediction, PBMC (subtype) counts and telomerase activity seemed most promising for antidepressant treatment outcome determination; full length brain-derived neurotrophic factor (BDNF)/truncated BDNF were shown to be most apt to prognosticate antipsychotic treatment. CONCLUSIONS We conclude that, although inherent limitations to and heterogeneity in study designs in combination with the scarce number of original studies hamper unambiguous identification, several PBMC characteristics-mostly related to inflammatory pathways and cell viability-indeed show promise towards establishment as clinically relevant treatment biomarkers.
Collapse
|
23
|
Morrens M, Coppens V, Walther S. Do Immune Dysregulations and Oxidative Damage Drive Mood and Psychotic Disorders? Neuropsychobiology 2021; 79:251-254. [PMID: 30991415 DOI: 10.1159/000496622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium, .,University Department of Psychiatry, Campus Duffel, Duffel, Belgium,
| | - Violette Coppens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium.,University Department of Psychiatry, Campus Duffel, Duffel, Belgium
| | - Sebastian Walther
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Imaging Biomarkers for Monitoring the Inflammatory Redox Landscape in the Brain. Antioxidants (Basel) 2021; 10:antiox10040528. [PMID: 33800685 PMCID: PMC8065574 DOI: 10.3390/antiox10040528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 12/27/2022] Open
Abstract
Inflammation is one key process in driving cellular redox homeostasis toward oxidative stress, which perpetuates inflammation. In the brain, this interplay results in a vicious cycle of cell death, the loss of neurons, and leakage of the blood–brain barrier. Hence, the neuroinflammatory response fuels the development of acute and chronic inflammatory diseases. Interrogation of the interplay between inflammation, oxidative stress, and cell death in neurological tissue in vivo is very challenging. The complexity of the underlying biological process and the fragility of the brain limit our understanding of the cause and the adequate diagnostics of neuroinflammatory diseases. In recent years, advancements in the development of molecular imaging agents addressed this limitation and enabled imaging of biomarkers of neuroinflammation in the brain. Notable redox biomarkers for imaging with positron emission tomography (PET) tracers are the 18 kDa translocator protein (TSPO) and monoamine oxygenase B (MAO–B). These findings and achievements offer the opportunity for novel diagnostic applications and therapeutic strategies. This review summarizes experimental as well as established pharmaceutical and biotechnological tools for imaging the inflammatory redox landscape in the brain, and provides a glimpse into future applications.
Collapse
|
25
|
Plavén-Sigray P, Matheson GJ, Coughlin JM, Hafizi S, Laurikainen H, Ottoy J, De Picker L, Rusjan P, Hietala J, Howes OD, Mizrahi R, Morrens M, Pomper MG, Cervenka S. Meta-analysis of the Glial Marker TSPO in Psychosis Revisited: Reconciling Inconclusive Findings of Patient-Control Differences. Biol Psychiatry 2021; 89:e5-e8. [PMID: 32682565 PMCID: PMC7899168 DOI: 10.1016/j.biopsych.2020.05.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Pontus Plavén-Sigray
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden,Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Granville J. Matheson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Jennifer M. Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Sina Hafizi
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Heikki Laurikainen
- Department of Psychiatry, University of Turku and Neuropsychiatric Imaging Group, Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Julie Ottoy
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Pablo Rusjan
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Jarmo Hietala
- Department of Psychiatry, University of Turku and Neuropsychiatric Imaging Group, Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Oliver D. Howes
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London,MRC London Institute of Medical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom,Hammersmith Hospital; and Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Romina Mizrahi
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Martin G. Pomper
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, Maryland,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.
| |
Collapse
|
26
|
Iliopoulou SM, Tsartsalis S, Kaiser S, Millet P, Tournier BB. Dopamine and Neuroinflammation in Schizophrenia - Interpreting the Findings from Translocator Protein (18kDa) PET Imaging. Neuropsychiatr Dis Treat 2021; 17:3345-3357. [PMID: 34819729 PMCID: PMC8608287 DOI: 10.2147/ndt.s334027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/09/2021] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia is a complex disease whose pathophysiology is not yet fully understood. In addition to the long prevailing dopaminergic hypothesis, the evidence suggests that neuroinflammation plays a role in the pathophysiology of the disease. Recent studies using positron emission tomography (PET) that target a 18kDa translocator protein (TSPO) in activated microglial cells in an attempt to measure neuroinflammation in patients have shown a decrease or a lack of an increase in TSPO binding. Many biological and methodological considerations have been formulated to explain these findings. Although dopamine has been described as an immunomodulatory molecule, its potential role in neuroinflammation has not been explored in the aforementioned studies. In this review, we discuss the interactions between dopamine and neuroinflammation in psychotic states. Dopamine may inhibit neuroinflammation in activated microglia. Proinflammatory molecules released from microglia may decrease dopaminergic transmission. This could potentially explain why the levels of neuroinflammation in the brain of patients with schizophrenia seem to be unchanged or decreased compared to those in healthy subjects. However, most data are indirect and are derived from animal studies or from studies performed outside the field of schizophrenia. Further studies are needed to combine TSPO and dopamine imaging to study the association between microglial activation and dopamine system function.
Collapse
Affiliation(s)
- Sotiria Maria Iliopoulou
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland
| | | | - Stefan Kaiser
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, 1204, Switzerland
| | - Philippe Millet
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, 1204, Switzerland
| | - Benjamin B Tournier
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, 1204, Switzerland
| |
Collapse
|
27
|
Personality traits in psychosis and psychosis risk linked to TSPO expression: a neuroimmune marker. PERSONALITY NEUROSCIENCE 2020; 3:e14. [PMID: 33354652 PMCID: PMC7737185 DOI: 10.1017/pen.2020.14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 08/19/2020] [Accepted: 08/25/2020] [Indexed: 12/23/2022]
Abstract
Personality has been correlated with differences in cytokine expression, an indicator of peripheral inflammation; however, the associations between personality and central markers of inflammation have never been investigated in vivo in humans. Microglia are the resident macrophages of the central nervous system, and the first responders to tissue damage and brain insult. Microglial activation is associated with elevated expression of translocator protein 18kDa (TSPO), which can be imaged with positron emission tomography (PET) to quantify immune activation in the human brain. This study aimed to investigate the association between personality and TSPO expression across the psychosis spectrum. A total of 61 high-resolution [18F]FEPPA PET scans were conducted in 28 individuals at clinical high risk (CHR) for psychosis, 19 First-Episode Psychosis (FEP), and 14 healthy volunteers (HVs), and analyzed using a two-tissue compartment model and plasma input function to obtain a total volume of distribution (VT) as an index of brain TSPO expression (controlling for the rs6971 TSPO polymorphism). Personality was assessed using the Revised NEO Personality Inventory (NEO-PI-R). We found TSPO expression to be specifically associated with neuroticism. A positive association between TSPO expression and neuroticism was found in HVs, in contrast to a nonsignificant, negative association in CHR and significant negative association in FEP. The TSPO-associated neuroticism trait indicates an unexplored connection between neuroimmune activation and personality that varies across the psychosis spectrum.
Collapse
|
28
|
Meyer JH, Cervenka S, Kim MJ, Kreisl WC, Henter ID, Innis RB. Neuroinflammation in psychiatric disorders: PET imaging and promising new targets. Lancet Psychiatry 2020; 7:1064-1074. [PMID: 33098761 PMCID: PMC7893630 DOI: 10.1016/s2215-0366(20)30255-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/14/2023]
Abstract
Neuroinflammation is a multifaceted physiological and pathophysiological response of the brain to injury and disease. Given imaging findings of 18 kDa translocator protein (TSPO) and the development of radioligands for other inflammatory targets, PET imaging of neuroinflammation is at a particularly promising stage. This Review critically evaluates PET imaging results of inflammation in psychiatric disorders, including major depressive disorder, schizophrenia and psychosis disorders, substance use, and obsessive-compulsive disorder. We also consider promising new targets that can be measured in the brain, such as monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, colony stimulating factor 1 receptor, and the purinergic P2X7 receptor. Thus far, the most compelling TSPO imaging results have arguably been found in major depressive disorder, for which consistent increases have been observed, and in schizophrenia and psychosis, for which patients show reduced TSPO levels. This pattern highlights the importance of validating brain biomarkers of neuroinflammation for each condition separately before moving on to patient stratification and treatment monitoring trials.
Collapse
Affiliation(s)
- Jeffrey H Meyer
- Campbell Family Mental Health Research Institute, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm, Sweden
| | - Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|
29
|
Dinesh AA, Islam J, Khan J, Turkheimer F, Vernon AC. Effects of Antipsychotic Drugs: Cross Talk Between the Nervous and Innate Immune System. CNS Drugs 2020; 34:1229-1251. [PMID: 32975758 DOI: 10.1007/s40263-020-00765-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2020] [Indexed: 12/11/2022]
Abstract
Converging lines of evidence suggest that activation of microglia (innate immune cells in the central nervous system [CNS]) is present in a subset of patients with schizophrenia. The extent to which antipsychotic drug treatment contributes to or combats this effect remains unclear. To address this question, we reviewed the literature for evidence that antipsychotic exposure influences brain microglia as indexed by in vivo neuroimaging and post-mortem studies in patients with schizophrenia and experimental animal models. We found no clear evidence from clinical studies for an effect of antipsychotics on either translocator protein (TSPO) radioligand binding (an in vivo neuroimaging measure of putative gliosis) or markers of brain microglia in post-mortem studies. In experimental animals, where drug and illness effects may be differentiated, we also found no clear evidence for consistent effects of antipsychotic drugs on TSPO radioligand binding. By contrast, we found evidence that chronic antipsychotic exposure may influence central microglia density and morphology. However, these effects were dependent on the dose and duration of drug exposure and whether an immune stimulus was present or not. In the latter case, antipsychotics were generally reported to suppress expression of inflammatory cytokines and inducible inflammatory enzymes such as cyclooxygenase and microglia activation. No clear conclusions could be drawn with regard to any effect of antipsychotics on brain microglia from current clinical data. There is evidence to suggest that antipsychotic drugs influence brain microglia in experimental animals, including possible anti-inflammatory actions. However, we lack detailed information on how these drugs influence brain microglia function at the molecular level. The clinical relevance of the animal data with regard to beneficial treatment effects and detrimental side effects of antipsychotic drugs also remains unknown, and further studies are warranted.
Collapse
Affiliation(s)
- Ayushi Anna Dinesh
- School of Medicine, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Juned Islam
- School of Medicine, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Javad Khan
- School of Medicine, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Centre for Neuroimaging Sciences, De Crespigny Park, London, SE5 8AF, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, United Kingdom
| | - Anthony C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, United Kingdom.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RT, United Kingdom.
| |
Collapse
|
30
|
Blood-based kynurenine pathway alterations in schizophrenia spectrum disorders: A meta-analysis. Schizophr Res 2020; 223:43-52. [PMID: 32981827 DOI: 10.1016/j.schres.2020.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 08/06/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The kynurenine pathway (KP) has been proposed as indirect link between systemic immune responses and clinical symptom development in schizophrenia spectrum disorders (SSD). Empirical evidence for such immune-related KP shifts in SSD has however resulted in divergent findings. METHODS We conducted a systematic literature search in PubMed. Thirty papers (total number of patients n = 1506; controls: n = 1432) reported on peripheral concentrations of KP metabolites in SSD patients versus controls. Six KP metabolites were included in a meta-analysis, with secondary analysis of covariate and subgroup effects of patients' symptomatic state, age and duration of illness. RESULTS Tryptophan (SMD: -0.30; p = .003) and Xanthurenic Acid (SMD: -0.80; p < .001) were significantly decreased in SSD compared to controls, while Quinolinic Acid (SMD: -0.40; p = .08) and Kynurenic Acid (SMD: -0.39; p = .04) were only significantly decreased in patients with acute or highly symptomatic illness. Finally, in relatively older patient cohorts Kynurenine (SMD: -0.31; p = .02) and Kynurenic Acid (SMD: -0.40; p = .002) were found to be decreased. CONCLUSION A partial downregulation of the KP is observed in SSD patients, in particular during acute symptomatic states and in older age, effects that were independent from each other. In contrast, younger and stable or remitted patients display limited to no KP metabolite abnormalities. The current meta-analysis illustrates the dynamic nature of KP abnormalities. It should be noted that all included studies investigated peripheral KP metabolites, which do not necessarily reflect central KP metabolite abnormalities in schizophrenic patients.
Collapse
|
31
|
Berdyyeva T, Xia C, Taylor N, He Y, Chen G, Huang C, Zhang W, Kolb H, Letavic M, Bhattacharya A, Szardenings AK. PET Imaging of the P2X7 Ion Channel with a Novel Tracer [ 18F]JNJ-64413739 in a Rat Model of Neuroinflammation. Mol Imaging Biol 2020; 21:871-878. [PMID: 30632003 DOI: 10.1007/s11307-018-01313-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE The P2X7 receptor, an adenosine triphosphate (ATP)-gated purinoreceptor, has emerged as one of the key players in neuroinflammatory processes. Therefore, developing a positron emission tomography (PET) tracer for imaging of P2X7 receptors in vivo presents a promising approach to diagnose, monitor, and study neuroinflammation in a variety of brain disorders. To fulfill the goal of developing a P2X7 PET ligand as a biomarker of neuroinflammation, [18F]JNJ-64413739 has been recently disclosed. PROCEDURES We evaluated [18F]JNJ-64413739 in a rat model of neuroinflammation induced by an intracerebral injection of lipopolysaccharide (LPS). In vivo brain uptake was determined by PET imaging. Upregulation of neuroinflammatory biomarkers was determined by quantitative polymerase chain reaction (qPCR). Distribution of the tracer in the brain was determined by ex vivo autoradiography (ARG). The specificity of [18F]JNJ-64413739 was confirmed by performing blocking experiments with the P2X7 antagonist JNJ-54175446. RESULTS Brain regions of rats injected with LPS had a significantly increased uptake (34 % ± 3 % s.e.m., p = 0.036, t test, standardized uptake value measured over the entire scanning period) of [18F]JNJ-64413739 relative to the corresponding brain regions of control animals injected with phosphate-buffered saline (PBS). The uptake in the contralateral regions and cerebellum was not significantly different between the groups of animals. The increase in uptake of [18F]JNJ-64413739 at the LPS-injected site observed by PET imaging was concordant with ex vivo ARG, upregulation of neuroinflammatory biomarkers, and elevated P2X7 expression levels. CONCLUSIONS While further work is needed to study [18F]JNJ-64413739 in other types of neuroinflammation, the current results favorably characterize [18F]JNJ-64413739 as a potential PET tracer of central neuroinflammation.
Collapse
Affiliation(s)
- Tamara Berdyyeva
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA.
| | - Chunfang Xia
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Natalie Taylor
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Yingbo He
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Gang Chen
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Chaofeng Huang
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Wei Zhang
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Hartmuth Kolb
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Michael Letavic
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Anindya Bhattacharya
- Janssen Research & Development LLC, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | | |
Collapse
|
32
|
Cavaliere C, Tramontano L, Fiorenza D, Alfano V, Aiello M, Salvatore M. Gliosis and Neurodegenerative Diseases: The Role of PET and MR Imaging. Front Cell Neurosci 2020; 14:75. [PMID: 32327973 PMCID: PMC7161920 DOI: 10.3389/fncel.2020.00075] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/13/2020] [Indexed: 12/16/2022] Open
Abstract
Glial activation characterizes most neurodegenerative and psychiatric diseases, often anticipating clinical manifestations and macroscopical brain alterations. Although imaging techniques have improved diagnostic accuracy in many neurological conditions, often supporting diagnosis, prognosis prediction and treatment outcome, very few molecular imaging probes, specifically focused on microglial and astrocytic activation, have been translated to a clinical setting. In this context, hybrid positron emission tomography (PET)/magnetic resonance (MR) scanners represent the most advanced tool for molecular imaging, combining the functional specificity of PET radiotracers (e.g., targeting metabolism, hypoxia, and inflammation) to both high-resolution and multiparametric information derived by MR in a single imaging acquisition session. This simultaneity of findings achievable by PET/MR, if useful for reciprocal technical adjustments regarding temporal and spatial cross-modal alignment/synchronization, opens still debated issues about its clinical value in neurological patients, possibly incompliant and highly variable from a clinical point of view. While several preclinical and clinical studies have investigated the sensitivity of PET tracers to track microglial (mainly TSPO ligands) and astrocytic (mainly MAOB ligands) activation, less studies have focused on MR specificity to this topic (e.g., through the assessment of diffusion properties and T2 relaxometry), and only few exploiting the integration of simultaneous hybrid acquisition. This review aims at summarizing and critically review the current state about PET and MR imaging for glial targets, as well as the potential added value of hybrid scanners for characterizing microglial and astrocytic activation.
Collapse
|
33
|
Arakawa R, Takano A, Stenkrona P, Stepanov V, Nag S, Jahan M, Grybäck P, Bolin M, Chen L, Zhang L, He P, Villalobos A, McCarthy TJ, Halldin C, Varrone A. PET imaging of beta-secretase 1 in the human brain: radiation dosimetry, quantification, and test-retest examination of [ 18F]PF-06684511. Eur J Nucl Med Mol Imaging 2020; 47:2429-2439. [PMID: 32140803 PMCID: PMC7396399 DOI: 10.1007/s00259-020-04739-5] [Citation(s) in RCA: 4] [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: 11/28/2019] [Accepted: 02/20/2020] [Indexed: 11/29/2022]
Abstract
Purpose Beta-secretase 1 (BACE1) enzyme is implicated in the pathophysiology of Alzheimer’s disease. [18F]PF-06684511 is a positron emission tomography (PET) radioligand for imaging BACE1. Despite favorable brain kinetic properties, the effective dose (ED) of [18F]PF-06684511 estimated in non-human primates was relatively high. This study was therefore designed to evaluate the whole-body distribution, dosimetry, quantification, and test-retest reliability of imaging brain BACE1 with [18F]PF-06684511 in healthy volunteers. Methods Five subjects were studied for the dosimetry study. Whole-body PET was performed for 366 min with 4 PET-CT sessions. Estimates of the absorbed radiation dose were calculated using the male adult model. Eight subjects participated in the test-retest study. Brain PET measurements were conducted for 123 min with an interval of 5 to 19 days between test and retest conditions. The total distribution volume (VT) was estimated with one-tissue (1T), two-tissue (2T), compartment model (CM), and graphical analysis. Test-retest variability (TRV) and intraclass correlation coefficient (ICC) of VT were calculated as reliability measures. Results In the dosimetry study, the highest uptake was found in the liver (25.2 ± 2.3 %ID at 0.5 h) and the largest dose was observed in the pancreas (92.9 ± 52.2 μSv/MBq). The calculated ED was 24.7 ± 0.8 μSv/MBq. In the test-retest study, 2TCM described the time-activity curves well. VT (2TCM) was the highest in the anterior cingulate cortex (6.28 ± 1.09 and 6.85 ± 0.81) and the lowest in the cerebellum (4.23 ± 0.88 and 4.20 ± 0.75). Mean TRV and ICC of VT (2TCM) were 16.5% (12.4–20.5%) and 0.496 (0.291–0.644). Conclusion The ED of [18F]PF-06684511 was similar to other 18F radioligands, allowing repeated PET measurements. 2TCM was the most appropriate quantification method. TRV of VT was similar to other radioligands without a reference region, albeit with lower ICC. These data indicated that [18F]PF-06684511 is a suitable radioligand to measure BACE1 level in the human brain. Trial registration EudraCT 2016-001110-19 (registered 2016-08-08)
Collapse
Affiliation(s)
- Ryosuke Arakawa
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.
| | - Akihiro Takano
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Per Stenkrona
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Vladimir Stepanov
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Sangram Nag
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Mahabuba Jahan
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Per Grybäck
- Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Bolin
- Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Laigao Chen
- Worldwide Research & Development, Pfizer Inc., Cambridge, MA, USA
| | - Lei Zhang
- Worldwide Research & Development, Pfizer Inc., Cambridge, MA, USA
| | - Ping He
- Worldwide Research & Development, Pfizer Inc., Cambridge, MA, USA
| | | | | | - Christer Halldin
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Andrea Varrone
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| |
Collapse
|
34
|
De Picker L, Fransen E, Coppens V, Timmers M, de Boer P, Oberacher H, Fuchs D, Verkerk R, Sabbe B, Morrens M. Immune and Neuroendocrine Trait and State Markers in Psychotic Illness: Decreased Kynurenines Marking Psychotic Exacerbations. Front Immunol 2020; 10:2971. [PMID: 32010121 PMCID: PMC6978914 DOI: 10.3389/fimmu.2019.02971] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 12/03/2019] [Indexed: 01/18/2023] Open
Abstract
Objective: Different patterns of immune system upregulation are present in the acute vs. post-treatment states of psychotic illness. We explored the existence of state and trait markers in the peripheral immune system and two immune-associated neuroendocrine pathways (IDO and GTP-CH1 pathway) in a longitudinal sample of psychosis patients. We also evaluated the association of these markers with neuropsychiatric symptomatology. Method: Plasma concentrations of peripheral blood markers were measured in a transdiagnostic group of 49 inpatients with acute psychosis and 52 matched healthy control subjects. Samples were obtained in patients within 48 h after hospital admission for an acute psychotic episode (before initiation of antipsychotics), after 1-2 weeks and again after 8 weeks of treatment. Kynurenine, kynurenic acid (KA), 3-hydroxykynurenine (3-HK), quinolinic acid (QA), phenylalanine, tyrosine, nitrite, and neopterin were measured using HPLC and LC-MS/MS analysis. Concentrations of CRP, CCL2 (MCP1) and cytokines were determined with multiplex immunoassay. PANSS interviews and cognitive tests were performed at baseline and follow-up. Mixed model analyses were used to identify trait and state markers. Results: Patients had significantly higher plasma concentrations of CRP, CCL2, IL1RA, and lower concentrations of KA and KA/Kyn at all time points (F7.5-17.5, all p < 0.001). Increased concentrations of IL6, IL8, IL1RA, TNFα, and CCL2 and decreased QA and 3-HK (F8.7-21.0, all p < 0.005) were found in the acute psychotic state and normalized after treatment. Low nitrite concentrations at admission rose sharply after initiation of antipsychotic medication (F42.4, p < 0.001). PANSS positive scale scores during the acute episode correlated with pro-inflammatory immune markers (r ≥ |0.5|), while negative scale scores correlated inversely with IDO pathway markers (r ≥ |0.4|). Normalization of KA and 3-HK levels between admission and follow-up corresponded to a larger improvement of negative symptoms (r = 0.5, p < 0.030) A reverse association was found between relative improvement of SDST scores and decreasing KA levels (r = 0.5, p < 0.010). Conclusion: The acute psychotic state is marked by state-specific increases of immune markers and decreases in peripheral IDO pathway markers. Increased CRP, CCL2, and IL1RA, and decreased KA and KA/Kyn are trait markers of psychotic illness.
Collapse
Affiliation(s)
- Livia De Picker
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,University Department of Psychiatry, Campus Duffel, Antwerp, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, Antwerp, Belgium
| | - Violette Coppens
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,University Department of Psychiatry, Campus Duffel, Antwerp, Belgium
| | - Maarten Timmers
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium.,Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Peter de Boer
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Herbert Oberacher
- Core Facility Metabolomics, Institute of Legal Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Fuchs
- Biocenter, Division of Biological Chemistry, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Verkerk
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Bernard Sabbe
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,University Department of Psychiatry, Campus Duffel, Antwerp, Belgium
| | - Manuel Morrens
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,University Department of Psychiatry, Campus Duffel, Antwerp, Belgium
| |
Collapse
|
35
|
Microglial activation in schizophrenia: Is translocator 18 kDa protein (TSPO) the right marker? Schizophr Res 2020; 215:167-172. [PMID: 31699629 DOI: 10.1016/j.schres.2019.10.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022]
Abstract
Positron emission tomography (PET) with translocator 18 kDa protein (TSPO) radioligands has frequently been used to investigate microglial activation in schizophrenia in vivo. However, the specificity of this marker is increasingly debated. Here we show that TSPO expression is 1) not increased in postmortem brain tissue of schizophrenia patients; 2) not correlated with expression of microglial activation markers; 3) not restricted to microglia; and 4) not upregulated in ex vivo activated human primary microglia. Our data are in line with recent reports showing that TSPO expression is not increased in schizophrenia and that it is not a specific marker for activated microglia. This study emphasizes the need for further development of tracers to study the role of microglial activation in schizophrenia and other diseases.
Collapse
|
36
|
De Picker L, Morrens M. Perspective: Solving the Heterogeneity Conundrum of TSPO PET Imaging in Psychosis. Front Psychiatry 2020; 11:362. [PMID: 32425835 PMCID: PMC7206714 DOI: 10.3389/fpsyt.2020.00362] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Positron emission tomography using ligands targeting translocator protein 18 kDa (TSPO PET) is an innovative method to visualize and quantify glial inflammatory responses in the central nervous system in vivo. Compared to some other neuropsychiatric disorders, findings of TSPO PET in schizophrenia and related psychotic disorders have been considerably more heterogeneous. Two conflicting meta-analyses have been published on the topic within the last year: one asserting evidence for decreased TSPO uptake, while the other observed increased TSPO uptake in a selection of studies. In this paper, we review and discuss five hypotheses which may explain the observed variability of TSPO PET findings in psychotic illness, namely that (1) an inflammatory phenotype is only present in a subgroup of psychosis patients; (2) heterogeneity is caused by interference of antipsychotic medication; (3) interference of other clinical confounders in the study populations (such as age, sex, BMI, smoking, and substance use); or (4) methodological variability between studies (such as choice of tracer and kinetic model, genotyping, study power, and diurnal effects); and (5) the glial responses underlying changes in TSPO expression are themselves heterogeneous and dynamic. Finally, we propose four key recommendations for future research proposals to mitigate these different causes of heterogeneity.
Collapse
Affiliation(s)
- Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,SINAPS, University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium.,SINAPS, University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| |
Collapse
|
37
|
Beaurain M, Salabert AS, Ribeiro MJ, Arlicot N, Damier P, Le Jeune F, Demonet JF, Payoux P. Innovative Molecular Imaging for Clinical Research, Therapeutic Stratification, and Nosography in Neuroscience. Front Med (Lausanne) 2019; 6:268. [PMID: 31828073 PMCID: PMC6890558 DOI: 10.3389/fmed.2019.00268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/01/2019] [Indexed: 01/06/2023] Open
Abstract
Over the past few decades, several radiotracers have been developed for neuroimaging applications, especially in PET. Because of their low steric hindrance, PET radionuclides can be used to label molecules that are small enough to cross the blood brain barrier, without modifying their biological properties. As the use of 11C is limited by its short physical half-life (20 min), there has been an increasing focus on developing tracers labeled with 18F for clinical use. The first such tracers allowed cerebral blood flow and glucose metabolism to be measured, and the development of molecular imaging has since enabled to focus more closely on specific targets such as receptors, neurotransmitter transporters, and other proteins. Hence, PET and SPECT biomarkers have become indispensable for innovative clinical research. Currently, the treatment options for a number of pathologies, notably neurodegenerative diseases, remain only supportive and symptomatic. Treatments that slow down or reverse disease progression are therefore the subject of numerous studies, in which molecular imaging is proving to be a powerful tool. PET and SPECT biomarkers already make it possible to diagnose several neurological diseases in vivo and at preclinical stages, yielding topographic, and quantitative data about the target. As a result, they can be used for assessing patients' eligibility for new treatments, or for treatment follow-up. The aim of the present review was to map major innovative radiotracers used in neuroscience, and explain their contribution to clinical research. We categorized them according to their target: dopaminergic, cholinergic or serotoninergic systems, β-amyloid plaques, tau protein, neuroinflammation, glutamate or GABA receptors, or α-synuclein. Most neurological disorders, and indeed mental disorders, involve the dysfunction of one or more of these targets. Combinations of molecular imaging biomarkers can afford us a better understanding of the mechanisms underlying disease development over time, and contribute to early detection/screening, diagnosis, therapy delivery/monitoring, and treatment follow-up in both research and clinical settings.
Collapse
Affiliation(s)
- Marie Beaurain
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Anne-Sophie Salabert
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Maria Joao Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Philippe Damier
- Inserm U913, Neurology Department, University Hospital, Nantes, France
| | | | - Jean-François Demonet
- Leenards Memory Centre, Department of Clinical Neuroscience, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pierre Payoux
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| |
Collapse
|
38
|
Marques TR, Ashok AH, Pillinger T, Veronese M, Turkheimer FE, Dazzan P, Sommer IE, Howes OD. Neuroinflammation in schizophrenia: meta-analysis of in vivo microglial imaging studies. Psychol Med 2019; 49:2186-2196. [PMID: 30355368 PMCID: PMC6366560 DOI: 10.1017/s0033291718003057] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Converging lines of evidence implicate an important role for the immune system in schizophrenia. Microglia are the resident immune cells of the central nervous system and have many functions including neuroinflammation, axonal guidance and neurotrophic support. We aimed to provide a quantitative review of in vivo PET imaging studies of microglia activation in patients with schizophrenia compared with healthy controls. METHODS Demographic, clinical and imaging measures were extracted from each study and meta-analysis was conducted using a random-effects model (Hedge's g). The difference in 18-kDa translocator protein (TSPO) binding between patients with schizophrenia and healthy controls, as quantified by either binding potential (BP) or volume of distribution (VT), was used as the main outcome. Sub-analysis and sensitivity analysis were carried out to investigate the effects of genotype, ligand and illness stage. RESULTS In total, 12 studies comprising 190 patients with schizophrenia and 200 healthy controls met inclusion criteria. There was a significant elevation in tracer binding in schizophrenia patients relative to controls when BP was used as an outcome measure, (Hedge's g = 0.31; p = 0.03) but no significant differences when VT was used (Hedge's g = -0.22; p = 0.29). CONCLUSIONS In conclusion, there is evidence for moderate elevations in TSPO tracer binding in grey matter relative to other brain tissue in schizophrenia when using BP as an outcome measure, but no difference when VT is the outcome measure. We discuss the relevance of these findings as well as the methodological issues that may underlie the contrasting difference between these outcomes.
Collapse
Affiliation(s)
- Tiago Reis Marques
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Du Cane Road, London W12 0NN, UK
- Psychiatric Imaging Group, London Institute of Medical Sciences (LMS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Abhishekh H Ashok
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Du Cane Road, London W12 0NN, UK
- Psychiatric Imaging Group, London Institute of Medical Sciences (LMS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - Toby Pillinger
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - Federico E. Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - Paola Dazzan
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - Iris E.C. Sommer
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Oliver D Howes
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Du Cane Road, London W12 0NN, UK
- Psychiatric Imaging Group, London Institute of Medical Sciences (LMS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| |
Collapse
|
39
|
Plavén-Sigray P, Cervenka S. Meta-analytic studies of the glial cell marker TSPO in psychosis - a question of apples and pears? Psychol Med 2019; 49:1624-1628. [PMID: 30739609 PMCID: PMC6601355 DOI: 10.1017/s003329171800421x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/20/2022]
Affiliation(s)
- P. Plavén-Sigray
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - S. Cervenka
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
40
|
More dampened monocytic Toll-like receptor 4 response to lipopolysaccharide and its association with cognitive function in Chinese Han first-episode patients with schizophrenia. Schizophr Res 2019; 206:300-306. [PMID: 30429077 DOI: 10.1016/j.schres.2018.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Accumulating evidence suggests alterations of the innate immune system are related to schizophrenia, although the precise mechanism remains to be elucidated. In this study, we aimed to detect the monocytic toll-like receptor 4 (TLR4) expression under basal and lipopolysaccharide (LPS)-stimulated conditions in first-episode (FE) Han Chinese patients with schizophrenia, as well as its association with cognitive function. METHODS Whole blood samples were taken in 42 FE schizophrenia patients and 36 healthy controls. Expressions of TLR4 on monocytes under basal and LPS-stimulated conditions were measured with flow cytometry. Psychopathological symptoms of schizophrenia were assessed by the Positive and Negative Syndrome Scale (PANSS) and the MATRICS Consensus Cognitive Battery (MCCB) was administered to all of the participants. RESULTS We found no differences in percentage and mean fluorescence intensity (MFI) of TLR4 expression on monocytes between patients and controls at basal status. However, LPS challenge resulted in a lower cell-surface level of TLR4 on monocytes in FE schizophrenia patients as compared to healthy controls (TLR4+%: F = 4.092, p = 0.047; TLR4 + MFI: F = 4.820, p = 0.031). In addition, correlation analysis together with multivariate linear regression analysis identified basal percentage of TLR4 in monocytes as the beneficial factor for visual learning and working memory in FE patients with schizophrenia. CONCLUSIONS Our findings suggested that TLR4 may be involved in the pathophysiology of schizophrenia, corroborating the role of innate immunity-related functional deficits in increased risk of schizophrenia.
Collapse
|
41
|
De Picker L, Ottoy J, Verhaeghe J, Deleye S, Wyffels L, Fransen E, Kosten L, Sabbe B, Coppens V, Timmers M, de Boer P, Van Nueten L, Op De Beeck K, Oberacher H, Vanhoenacker F, Ceyssens S, Stroobants S, Staelens S, Morrens M. State-associated changes in longitudinal [ 18F]-PBR111 TSPO PET imaging of psychosis patients: Evidence for the accelerated ageing hypothesis? Brain Behav Immun 2019; 77:46-54. [PMID: 30503836 DOI: 10.1016/j.bbi.2018.11.318] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/23/2018] [Accepted: 11/29/2018] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To determine whether state-associated changes in microglial activity, measured with translocator-protein positron emission tomography (TSPO PET), can be identified in psychosis patients through longitudinal evaluation of their regional tracer uptake over the clinical course from acute psychosis to post-treatment follow-up, and comparison to healthy controls. We also evaluated the relation between tracer uptake, clinical symptoms and peripheral immunological markers. METHOD Second-generation radioligand [18F]-PBR111 TSPO PET-CT was used for longitudinal dynamic imaging in 14 male psychosis patients and 17 male age-matched healthy control subjects. Patients were first scanned during an acute psychotic episode followed by a second scan after treatment. Prior genotyping of subjects for the rs6917 polymorphism distinguished high- and mixed-affinity binders. The main outcome was regional volume of distribution (VT), representing TSPO binding. Plasma concentrations of CRP, cytokines and kynurenines were measured at each timepoint. RESULTS We found a significant three-way interaction between time of scan, age and cohort (cortical grey matter F6.50, p.020). Age-dependent differences in VT existed between cohorts during the psychotic state, but not at follow-up. Patients' relative change in VT over time correlated with age (cortical grey matter Pearson's r.574). PANSS positive subscale scores correlated with regional VT during psychosis (cortical grey matter r.767). Plasma CRP and quinolinic acid were independently associated with lower VT. CONCLUSIONS We identified a differential age-dependent pattern of TSPO binding from psychosis to follow-up in our cohort of male psychosis patients. We recommend future TSPO PET studies in psychosis patients to differentiate between clinical states and consider potential age-related effects.
Collapse
Affiliation(s)
- Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium.
| | - Julie Ottoy
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Steven Deleye
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, Belgium
| | - Lauren Kosten
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Bernard Sabbe
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium
| | - Violette Coppens
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium
| | - Maarten Timmers
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium; Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Peter de Boer
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Luc Van Nueten
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Ken Op De Beeck
- Medical Genetics Research Group, University of Antwerp, Antwerp, Belgium
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Innsbruck, Austria
| | - Filip Vanhoenacker
- Department of Radiology, Sint-Maarten General Hospital, Mechelen, Belgium; Faculty of Medicine and Health Sciences, Universities of Antwerp and Ghent, Belgium
| | - Sarah Ceyssens
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sigrid Stroobants
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium
| |
Collapse
|
42
|
Abstract
Contrary to the notion that neurology but not psychiatry is the domain of disorders evincing structural brain alterations, it is now clear that there are subtle but consistent neuropathological changes in schizophrenia. These range from increases in ventricular size to dystrophic changes in dendritic spines. A decrease in dendritic spine density in the prefrontal cortex (PFC) is among the most replicated of postmortem structural findings in schizophrenia. Examination of the mechanisms that account for the loss of dendritic spines has in large part focused on genes and molecules that regulate neuronal structure. But the simple question of what is the effector of spine loss, ie, where do the lost spines go, is unanswered. Recent data on glial cells suggest that microglia (MG), and perhaps astrocytes, play an important physiological role in synaptic remodeling of neurons during development. Synapses are added to the dendrites of pyramidal cells during the maturation of these neurons; excess synapses are subsequently phagocytosed by MG. In the PFC, this occurs during adolescence, when certain symptoms of schizophrenia emerge. This brief review discusses recent advances in our understanding of MG function and how these non-neuronal cells lead to structural changes in neurons in schizophrenia.
Collapse
Affiliation(s)
| | - Ariel Y Deutch
- Neuroscience Program, Vanderbilt University, Nashville, TN
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| |
Collapse
|