1
|
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
|
2
|
Barriola S, Delgado-García LM, Cartas-Cejudo P, Iñigo-Marco I, Fernández-Irigoyen J, Santamaría E, López-Mascaraque L. Orosomucoid-1 Arises as a Shared Altered Protein in Two Models of Multiple Sclerosis. Neuroscience 2023; 535:203-217. [PMID: 37949310 DOI: 10.1016/j.neuroscience.2023.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023]
Abstract
Multiple sclerosis (MS) is a complex autoimmune and neurodegenerative disorder that affects the central nervous system (CNS). It is characterized by a heterogeneous disease course involving demyelination and inflammation. In this study, we utilized two distinct animal models, cuprizone (CPZ)-induced demyelination and experimental autoimmune encephalomyelitis (EAE), to replicate various aspects of the disease. We aimed to investigate the differential CNS responses by examining the proteomic profiles of EAE mice during the peak disease (15 days post-induction) and cuprizone-fed mice during the acute phase (38 days). Specifically, we focused on two different regions of the CNS: the dorsal cortex (Cx) and the entire spinal cord (SC). Our findings revealed varied glial, synaptic, dendritic, mitochondrial, and inflammatory responses within these regions for each model. Notably, we identified a single protein, Orosomucoid-1 (Orm1), also known as Alpha-1-acid glycoprotein 1 (AGP1), that consistently exhibited alterations in both models and regions. This study provides insights into the similarities and differences in the responses of these regions in two distinct demyelinating models.
Collapse
Affiliation(s)
- Sonsoles Barriola
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Consejo Superior de Investigaciones Científicas-CSIC, Madrid 28002, Spain; Ph.D. Program in Neuroscience, Autónoma de Madrid University-Cajal Institute, Madrid 28029, Spain
| | - Lina María Delgado-García
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Consejo Superior de Investigaciones Científicas-CSIC, Madrid 28002, Spain; Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo UNIFESP, São Paulo 04039032, Brazil
| | - Paz Cartas-Cejudo
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IDISNA, Pamplona 31008, Spain
| | - Ignacio Iñigo-Marco
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IDISNA, Pamplona 31008, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IDISNA, Pamplona 31008, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), IDISNA, Pamplona 31008, Spain
| | - Laura López-Mascaraque
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Consejo Superior de Investigaciones Científicas-CSIC, Madrid 28002, Spain.
| |
Collapse
|
3
|
Jiang H, Zhou C, Qiu L, Gropler RJ, Brier MR, Wu GF, Cross AH, Perlmutter JS, Benzinger TLS, Tu Z. Quantitative Analysis of S1PR1 Expression in the Postmortem Multiple Sclerosis Central Nervous System. ACS Chem Neurosci 2023; 14:4039-4050. [PMID: 37882753 PMCID: PMC11037862 DOI: 10.1021/acschemneuro.3c00581] [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] [Indexed: 10/27/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease that is characterized by demyelination and inflammation in the central nervous system (CNS). Previous studies demonstrated that sphingosine-1-phosphate receptor (S1PR) modulators effectively inhibit S1PR1 in immune cell trafficking and reduce entry of pathogenic cells into the CNS. Studies have also implicated a nonimmune, inflammatory role of S1PR1 within the CNS in MS. In this study, we explored the expression of S1PR1 in the development and progression of demyelinating pathology of MS by quantitative assessment of S1PR1 expression using our S1PR1-specific radioligand, [3H]CS1P1, in the postmortem human CNS tissues including cortex, cerebellum, and spinal cord of MS cases and age- and sex-matched healthy cases. Immunohistochemistry with whole slide scanning for S1PR1 and various myelin proteins was also performed. Autoradiographic analysis using [3H]CS1P1 showed that the expression of S1PR1 was statistically significantly elevated in lesions compared to nonlesion regions in the MS cases, as well as normal healthy controls. The uptake of [3H]CS1P1 in the gray matter and nonlesion white matter did not significantly differ between healthy and MS CNS tissues. Saturation autoradiography analysis showed an increased binding affinity (Kd) of [3H]CS1P1 to S1PR1 in both gray matter and white matter of MS brains compared to healthy brains. Our blocking study using NIBR-0213, a S1PR1 antagonist, indicated [3H]CS1P1 is highly specific to S1PR1. Our findings demonstrated the activation of S1PR1 and an increased uptake of [3H]CS1P1 in the lesions of MS CNS. In summary, our quantitative autoradiography analysis using [3H]CS1P1 on human postmortem tissues shows the feasibility of novel imaging strategies for MS by targeting S1PR1.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Charles Zhou
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Lin Qiu
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Robert J Gropler
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Matthew R Brier
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Gregory F Wu
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Anne H Cross
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri 63110, United States
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri 63110, United States
| |
Collapse
|
4
|
Yan X, Siméon FG, Liow JS, Morse CL, Montero Santamaria JA, Jenkins M, Manly LS, Van Buskirk M, Zoghbi SS, Pike VW, Innis RB, Zanotti-Fregonara P. In vivo evaluation of a novel 18F-labeled PET radioligand for translocator protein 18 kDa (TSPO) in monkey brain. Eur J Nucl Med Mol Imaging 2023; 50:2962-2970. [PMID: 37249618 PMCID: PMC10382351 DOI: 10.1007/s00259-023-06270-9] [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: 10/14/2022] [Accepted: 05/18/2023] [Indexed: 05/31/2023]
Abstract
PURPOSE [18F]SF51 was previously found to have high binding affinity and selectivity for 18 kDa translocator protein (TSPO) in mouse brain. This study sought to assess the ability of [18F]SF51 to quantify TSPO in rhesus monkey brain. METHODS Positron emission tomography (PET) imaging was performed in monkey brain (n = 3) at baseline and after pre-blockade with the TSPO ligands PK11195 and PBR28. TSPO binding was calculated as total distribution volume corrected for free parent fraction in plasma (VT/fP) using a two-tissue compartment model. Receptor occupancy and nondisplaceable uptake were determined via Lassen plot. Binding potential (BPND) was calculated as the ratio of specific binding to nondisplaceable uptake. Time stability of VT was used as an indirect probe to detect radiometabolite accumulation in the brain. In vivo and ex vivo experiments were performed in mice to determine the distribution of the radioligand. RESULTS After [18F]SF51 injection, the concentration of brain radioactivity peaked at 2.0 standardized uptake value (SUV) at ~ 10 min and declined to 30% of the peak at 180 min. VT/fP at baseline was generally high (203 ± 15 mL· cm-3) and decreased by ~ 90% after blockade with PK11195. BPND of the whole brain was 7.6 ± 4.3. VT values reached levels similar to terminal 180-min values by 100 min and remained relatively stable thereafter with excellent identifiability (standard errors < 5%), suggesting that no significant radiometabolites accumulated in the brain. Ex vivo experiments in mouse brain showed that 96% of radioactivity was parent. No significant uptake was observed in the skull, suggesting a lack of defluorination in vivo. CONCLUSION The results demonstrate that [18F]SF51 is an excellent radioligand that can quantify TSPO with a good ratio of specific to nondisplaceable uptake and has minimal radiometabolite accumulation in brain. Collectively, the results suggest that [18F]SF51 warrants further evaluation in humans.
Collapse
Affiliation(s)
- Xuefeng Yan
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA.
| | - Fabrice G Siméon
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Jose A Montero Santamaria
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Madeline Jenkins
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Lester S Manly
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Maia Van Buskirk
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, NIH, 10 Center Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
5
|
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: 1.0] [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
|
6
|
Lopresti BJ, Royse SK, Mathis CA, Tollefson SA, Narendran R. Beyond monoamines: I. Novel targets and radiotracers for Positron emission tomography imaging in psychiatric disorders. J Neurochem 2023; 164:364-400. [PMID: 35536762 DOI: 10.1111/jnc.15615] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
With the emergence of positron emission tomography (PET) in the late 1970s, psychiatry had access to a tool capable of non-invasive assessment of human brain function. Early applications in psychiatry focused on identifying characteristic brain blood flow and metabolic derangements using radiotracers such as [15 O]H2 O and [18 F]FDG. Despite the success of these techniques, it became apparent that more specific probes were needed to understand the neurochemical bases of psychiatric disorders. The first neurochemical PET imaging probes targeted sites of action of neuroleptic (dopamine D2 receptors) and psychoactive (serotonin receptors) drugs. Based on the centrality of monoamine dysfunction in psychiatric disorders and the measured success of monoamine-enhancing drugs in treating them, the next 30 years witnessed the development of an armamentarium of PET radiopharmaceuticals and imaging methodologies for studying monoamines. Continued development of monoamine-enhancing drugs over this time however was less successful, realizing only modest gains in efficacy and tolerability. As patent protection for many widely prescribed and profitable psychiatric drugs lapsed, drug development pipelines shifted away from monoamines in search of novel targets with the promises of improved efficacy, or abandoned altogether. Over this period, PET radiopharmaceutical development activities closely paralleled drug development priorities resulting in the development of new PET imaging agents for non-monoamine targets. Part one of this review will briefly survey novel PET imaging targets with relevance to the field of psychiatry, which include the metabotropic glutamate receptor type 5 (mGluR5), purinergic P2 X7 receptor, type 1 cannabinoid receptor (CB1 ), phosphodiesterase 10A (PDE10A), and describe radiotracers developed for these and other targets that have matured to human subject investigations. Current limitations of the targets and techniques will also be discussed.
Collapse
Affiliation(s)
- Brian J Lopresti
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah K Royse
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chester A Mathis
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Savannah A Tollefson
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rajesh Narendran
- Departments of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Departments of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Kim J, Kim YK. Molecular Imaging of Neuroinflammation in Alzheimer's Disease and Mild Cognitive Impairment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:301-326. [PMID: 36949316 DOI: 10.1007/978-981-19-7376-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent neurocognitive disorder. Due to the ineffectiveness of treatments targeting the amyloid cascade, molecular biomarkers for neuroinflammation are attracting attention with increasing knowledge about the role of neuroinflammation in the pathogenesis of AD. This chapter will explore the results of studies using molecular imaging for diagnosing AD and mild cognitive impairment (MCI). Because it is critical to interpreting the data to understand which substances are targeted in molecular imaging, this chapter will discuss the two most significant targets, microglia and astrocytes, as well as the best-known radioligands for each. Then, neuroimaging results with PET neuroinflammation imaging will be reviewed for AD and MCI. Although a growing body of evidence has suggested that these molecular imaging biomarkers for neuroinflammation may have a role in the diagnosis of AD and MCI, the findings are inconsistent or cross-sectional, which indicates that it is difficult to apply the contents in practice due to the need for additional study. In particular, because the results of multiple interventions targeting neuroinflammation were inconclusive, molecular imaging markers for neuroinflammation can be used in combination with conventional markers to select appropriate patients for early intervention for neuroinflammation rather than as a single marker.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Psychiatry, Korea University College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Ansan, Republic of Korea.
| |
Collapse
|
8
|
Huang J. Novel brain PET imaging agents: Strategies for imaging neuroinflammation in Alzheimer’s disease and mild cognitive impairment. Front Immunol 2022; 13:1010946. [PMID: 36211392 PMCID: PMC9537554 DOI: 10.3389/fimmu.2022.1010946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disease with a concealed onset and continuous deterioration. Mild cognitive impairment (MCI) is the prodromal stage of AD. Molecule-based imaging with positron emission tomography (PET) is critical in tracking pathophysiological changes among AD and MCI patients. PET with novel targets is a promising approach for diagnostic imaging, particularly in AD patients. Our present review overviews the current status and applications of in vivo molecular imaging toward neuroinflammation. Although radiotracers can remarkably diagnose AD and MCI patients, a variety of limitations prevent the recommendation of a single technique. Recent studies examining neuroinflammation PET imaging suggest an alternative approach to evaluate disease progression. This review concludes that PET imaging towards neuroinflammation is considered a promising approach to deciphering the enigma of the pathophysiological process of AD and MCI.
Collapse
|
9
|
Schubert J, Tonietto M, Turkheimer F, Zanotti-Fregonara P, Veronese M. Supervised clustering for TSPO PET imaging. Eur J Nucl Med Mol Imaging 2021; 49:257-268. [PMID: 33779770 PMCID: PMC8712290 DOI: 10.1007/s00259-021-05309-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE This technical note seeks to act as a practical guide for implementing a supervised clustering algorithm (SVCA) reference region approach and to explain the main strengths and limitations of the technique in the context of 18-kilodalton translocator protein (TSPO) positron emission tomography (PET) studies in experimental medicine. BACKGROUND TSPO PET is the most widely used imaging technique for studying neuroinflammation in vivo in humans. Quantifying neuroinflammation with PET can be a challenging and invasive procedure, especially in frail patients, because it often requires blood sampling from an arterial catheter. A widely used alternative to arterial sampling is SVCA, which identifies the voxels with minimal specific binding in the PET images, thus extracting a pseudo-reference region for non-invasive quantification. Unlike other reference region approaches, SVCA does not require specification of an anatomical reference region a priori, which alleviates the limitation of TSPO contamination in anatomically-defined reference regions in individuals with underlying inflammatory processes. Furthermore, SVCA can be applied to any TSPO PET tracer across different neurological and neuropsychiatric conditions, providing noninvasivequantification of TSPO expression. METHODS We provide an overview of the development of SVCA as well as step-by-step instructions for implementing SVCA with suggestions for specific settings. We review the literature on SVCAapplications using first- and second- generation TSPO PET tracers and discuss potential clinically relevant limitations and applications. CONCLUSIONS The correct implementation of SVCA can provide robust and reproducible estimates of brain TSPO expression. This review encourages the standardisation of SVCA methodology in TSPO PET analysis, ultimately aiming to improve replicability and comparability across study sites.
Collapse
Affiliation(s)
- Julia Schubert
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Matteo Tonietto
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Paolo Zanotti-Fregonara
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
10
|
Direct Comparison of [ 18F]F-DPA with [ 18F]DPA-714 and [ 11C]PBR28 for Neuroinflammation Imaging in the same Alzheimer's Disease Model Mice and Healthy Controls. Mol Imaging Biol 2021; 24:157-166. [PMID: 34542805 PMCID: PMC8760190 DOI: 10.1007/s11307-021-01646-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/22/2021] [Accepted: 08/23/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE In this study we compared the recently developed TSPO tracer [18F]F-DPA, with [18F]DPA-714 and [11C]PBR28 by performing in vivo PET imaging on the same Alzheimer's disease mouse model APP/PS1-21 (TG) and wild-type (WT) mice with all three radiotracers. PROCEDURES To compare the radiotracer uptake, percentage of injected dose/mL (%ID/mL), standardized uptake value ratios to cerebellum (SUVRCB), and voxel-wise analyses were performed. RESULTS The peak uptake of [18F]F-DPA was higher than 4.3% ID/mL, while [18F]DPA-714 reached just over 3% ID/mL, and [11C]PBR28 was over 4% ID/mL in only one brain region in the WT mice. The peak/60-min uptake ratios of [18F]F-DPA were significantly higher (p < 0.001) than those of [18F]DPA-714 and [11C]PBR28. The differences in [18F]F-DPA SUVRCB between WT and TG mice were highly significant (p < 0.001) in the three studied time periods after injection. [18F]DPA-714 uptake was significantly higher in TG mice starting in the 20-40-min timeframe and increased thereafter, whereas [11C]PBR28 uptake became significant at 10-20 min (p < 0.05). The voxel-wise analysis confirmed the differences between the radiotracers. CONCLUSIONS [18F]F-DPA displays higher brain uptake, higher TG-to-WT SUVRCB ratios, and faster clearance than [18F]DPA-714 and [11C]PBR28, and could prove useful for detecting low levels of inflammation and allow for shorter dynamic PET scans.
Collapse
|
11
|
Veronese M, Tuosto M, Marques TR, Howes O, Pascual B, Yu M, Masdeu JC, Turkheimer F, Bertoldo A, Zanotti-Fregonara P. Parametric Mapping for TSPO PET Imaging with Spectral Analysis Impulsive Response Function. Mol Imaging Biol 2021; 23:560-571. [PMID: 33475944 PMCID: PMC8277653 DOI: 10.1007/s11307-020-01575-9] [Citation(s) in RCA: 4] [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: 07/06/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this study was to investigate the use of spectral analysis (SA) for voxel-wise analysis of TSPO PET imaging studies. TSPO PET quantification is methodologically complicated by the heterogeneity of TSPO expression and its cell-dependent modulation during neuroinflammatory response. Compartmental models to account for this complexity exist, but they are unreliable at the high noise typical of voxel data. On the contrary, SA is noise-robust for parametric mapping and provides useful information about tracer kinetics with a free compartmental structure. PROCEDURES SA impulse response function (IRF) calculated at 90 min after tracer injection was used as main parameter of interest in 3 independent PET imaging studies to investigate its sensitivity to (1) a TSPO genetic polymorphism (rs6971) known to affect tracer binding in a cross-sectional analysis of healthy controls scanned with [11C]PBR28 PET; (2) TSPO density with [11C]PBR28 in a competitive blocking study with a TSPO blocker, XBD173; and (3) the higher affinity of a second radiotracer for TSPO, by using data from a head-to-head comparison between [11C]PBR28 and [11C]ER176 scans. RESULTS SA-IRF produced parametric maps of visually good quality. These were sensitive to TSPO genotype (mean relative difference between high- and mixed-affinity binders = 25 %) and TSPO availability (mean signal displacement after 90 mg oral administration of XBD173 = 39 %). Regional averages of voxel-wise IRF estimates were strongly associated with regional total distribution volume (VT) estimated with a 2-tissue compartmental model with vascular compartment (Pearson's r = 0.86 ± 0.11) but less strongly with standard 2TCM-VT (Pearson's r = 0.76 ± 0.32). Finally, SA-IRF estimates for [11C]ER176 were significantly higher than [11C]PBR28 ones, consistent with the higher amount of specific binding of the former tracer. CONCLUSIONS SA-IRF can be used for voxel-wise quantification of TSPO PET data because it generates high-quality parametric maps, it is sensitive to TSPO availability and genotype, and it accounts for the complexity of TSPO tracer kinetics with no additional assumptions.
Collapse
Affiliation(s)
- Mattia Veronese
- Department of Neuroimaging, IoPPN, King's College London, London, UK.
| | - Marcello Tuosto
- Department of Information Engineering, Padova University, Padova, Italy
| | - Tiago Reis Marques
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
| | - Oliver Howes
- Department of Psychosis Studies, IoPPN, King's College London, London, UK
- MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Belen Pascual
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Meixiang Yu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | - Joseph C Masdeu
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| | | | - Alessandra Bertoldo
- Department of Information Engineering, Padova University, Padova, Italy
- Padova Neuroscience Centre, Padova University, Padova, Italy
| | - Paolo Zanotti-Fregonara
- Nantz National Alzheimer Center and Houston Methodist Research Neurological Institute, and Weill Cornell Medicine, 6670 Bertner Ave, Houston, TX, 77030, USA
| |
Collapse
|
12
|
Łoś K, Waszkiewicz N. Biological Markers in Anxiety Disorders. J Clin Med 2021; 10:1744. [PMID: 33920547 PMCID: PMC8073190 DOI: 10.3390/jcm10081744] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Anxiety disorders are one of the most commonly reported disorders in psychiatry, causing a high medical and socio-economic burden. Recently, there has been a soaring interest in the biological basis of anxiety disorders, which is reflected in an increasing number of articles related to the topic. Due to the ambiguity of the diagnosis and a large number of underdiagnosed patients, researchers are looking for laboratory tests that could facilitate the diagnosis of anxiety disorders in clinical practice and would allow for the earliest possible implementation of appropriate treatment. Such potential biomarkers may also be useable in monitoring the efficacy of pharmacological therapy for anxiety disorders. Therefore this article reviews the literature of potential biomarkers such as components of saliva, peripheral blood, cerebrospinal fluid (CSF), and neuroimaging studies. There are promising publications in the literature that can be useful. The most valuable and promising markers of saliva are cortisol, lysozyme, and α-amylase (sAA). In the blood, in turn, we can distinguish serotonin, brain-derived serum neurotrophic factor (BDNF), cortisol, and microRNA. Structural changes in the amygdala and hippocampus are promising neuroimaging markers, while in CSF, potential markers include oxytocin and 5-Hydroxyindoleacetic acid (5-HIAA). Unfortunately, research in the field of biomarkers is hampered by insufficient knowledge about the etiopathogenesis of anxiety disorders, the significant heterogeneity of anxiety disorders, frequent comorbidities, and low specificity of biomarkers. The development of appropriate biomarker panels and their assessment using new approaches may have the prospective to overcome the above-mentioned obstacles.
Collapse
Affiliation(s)
- Kacper Łoś
- Department of Psychiatry, Medical University of Bialystok, Plac Brodowicza 1, 16-070 Choroszcz, Poland;
| | | |
Collapse
|
13
|
Adhikari A, Singh P, Mahar KS, Adhikari M, Adhikari B, Zhang MR, Tiwari AK. Mapping of Translocator Protein (18 kDa) in Peripheral Sterile Inflammatory Disease and Cancer through PET Imaging. Mol Pharm 2021; 18:1507-1529. [PMID: 33645995 DOI: 10.1021/acs.molpharmaceut.1c00002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Positron emission tomography (PET) imaging of the translocator 18 kDa protein (TSPO) with radioligands has become an effective means of research in peripheral inflammatory conditions that occur in many diseases and cancers. The peripheral sterile inflammatory diseases (PSIDs) are associated with a diverse group of disorders that comprises numerous enduring insults including the cardiovascular, respiratory, gastrointestinal, or musculoskeletal system. TSPO has recently been introduced as a potential biomarker for peripheral sterile inflammatory diseases (PSIDs). The major critical issue related to PSIDs is its timely characterization and localization of inflammatory foci for proper therapy of patients. As an alternative to metabolic imaging, protein imaging expressed on immune cells after activation is of great importance. The five transmembrane domain translocator protein-18 kDa (TSPO) is upregulated on the mitochondrial cell surface of macrophages during inflammation, serving as a potential ligand for PET tracers. Additionally, the overexpressed TSPO protein has been positively correlated with various tumor malignancies. In view of the association of escalated TSPO expression in both disease conditions, it is an immensely important biomarker for PET imaging in oncology and PSIDs. In this review, we summarize the most outstanding advances on TSPO-targeted PSIDs and cancer in the development of TSPO ligands as a potential diagnostic tool, specifically discussing the last five years.
Collapse
Affiliation(s)
- Anupriya Adhikari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, (A Central University), Lucknow, Uttar Pradesh 226025, India
| | - Priya Singh
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, A Central University, Lucknow, Uttar Pradesh 226025, India
| | - Kamalesh S Mahar
- Birbal Sahni Institute of Palaeosciences, Lucknow, Uttar Pradesh 226007, India
| | - Manish Adhikari
- The George Washington University, Washington, D.C. 20052, United States
| | - Bhawana Adhikari
- Plasma Bio-science Research Center, Kwangwoon University, Seoul 01897, South Korea
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Anjani Kumar Tiwari
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, (A Central University), Lucknow, Uttar Pradesh 226025, India
| |
Collapse
|
14
|
Pinpointing Brain TREM2 Levels in Two Mouse Models of Alzheimer's Disease. Mol Imaging Biol 2021; 23:665-675. [PMID: 33620643 PMCID: PMC8410720 DOI: 10.1007/s11307-021-01591-3] [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: 09/02/2020] [Revised: 01/11/2021] [Accepted: 02/11/2021] [Indexed: 11/01/2022]
Abstract
PURPOSE The triggering receptor expressed on myeloid cells 2 (TREM2) is expressed by brain microglia. Microglial activation, as observed in Alzheimer's disease (AD) as well as in transgenic mice expressing human amyloid-beta, appears to increase soluble TREM2 (sTREM2) levels in CSF and brain. In this study, we used two different transgenic mouse models of AD pathology and investigated the potential of TREM2 to serve as an in vivo biomarker for microglial activation in AD. PROCEDURES We designed and generated a bispecific antibody based on the TREM2-specific monoclonal antibody mAb1729, fused to a single-chain variable fragment of the transferrin receptor binding antibody 8D3. The 8D3-moiety enabled transcytosis of the whole bispecific antibody across the blood-brain barrier. The bispecific antibody was radiolabeled with I-125 (ex vivo) or I-124 (PET) and administered to transgenic AD and wild-type (WT) control mice. Radioligand retention in the brain of transgenic animals was compared to WT mice by isolation of brain tissue at 24 h or 72 h, or with in vivo PET at 24 h, 48 h, and 72 h. Intrabrain distribution of radiolabeled mAb1729-scFv8D3CL was further studied by autoradiography, while ELISA was used to determine TREM2 brain concentrations. RESULTS Transgenic animals displayed higher total exposure, calculated as the AUC based on SUV determined at 24h, 48h, and 72h post injection, of PET radioligand [124I]mAb1729-scFv8D3CL than WT mice. However, differences were not evident in single time point PET images or SUVs. Ex vivo autoradiography confirmed higher radioligand concentrations in cortex and thalamus in transgenic mice compared to WT, and TREM2 levels in brain homogenates were considerably higher in transgenic mice compared to WT. CONCLUSION Antibody-based radioligands, engineered to enter the brain, may serve as PET radioligands to follow changes of TREM2 in vivo, but antibody formats with faster systemic clearance to increase the specific signal in relation to that from blood in combination with antibodies showing higher affinity for TREM2 must be developed to further progress this technique for in vivo use.
Collapse
|
15
|
Schubert JJ, Veronese M, Fryer TD, Manavaki R, Kitzbichler MG, Nettis MA, Mondelli V, Pariante CM, Bullmore ET, Turkheimer FE. A Modest Increase in 11C-PK11195-Positron Emission Tomography TSPO Binding in Depression Is Not Associated With Serum C-Reactive Protein or Body Mass Index. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 6:716-724. [PMID: 33515765 PMCID: PMC8264953 DOI: 10.1016/j.bpsc.2020.12.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/27/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023]
Abstract
Background Immune mechanisms have been implicated in the pathogenesis of depression. Translocator protein (TSPO)–targeted positron emission tomography (PET) has been used to assess neuroinflammation in major depressive disorder. We aimed to 1) test the hypothesis of significant case-control differences in TSPO binding in the anterior cingulate cortex, prefrontal cortex, and insula regions; and 2) explore the relationship between cerebral TSPO binding and peripheral blood C-reactive protein (CRP) concentration. Methods A total of 51 depressed subjects with Hamilton Depression Rating Scale score >13 (median 17; interquartile range, 16–22) and 25 healthy control subjects underwent dynamic brain 11C-PK11195 PET and peripheral blood immune marker characterization. Depressed subjects were divided into high CRP (>3 mg/L; n = 20) and low CRP (<3 mg/L; n = 31). Results Across the three regions, TSPO binding was significantly increased in depressed versus control subjects (η2p = .09; F1,71 = 6.97, p = .01), which was not influenced by body mass index. The case-control difference was greatest in the anterior cingulate cortex (d = 0.49; t74 = 2.00, p = .03) and not significant in the prefrontal cortex or insula (d = 0.27 and d = 0.36, respectively). Following CRP stratification, significantly higher TSPO binding was observed in low-CRP depression compared with controls (d = 0.53; t54 = 1.96, p = .03). These effect sizes are comparable to prior major depressive disorder case-control TSPO PET data. No significant correlations were observed between TSPO and CRP measures. Conclusions Consistent with previous findings, there is a modest increase in TSPO binding in depressed patients compared with healthy control subjects. The lack of a significant correlation between brain TSPO binding and blood CRP concentration or body mass index poses questions about the interactions between central and peripheral immune responses in the pathogenesis of depression.
Collapse
Affiliation(s)
- Julia J Schubert
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - Tim D Fryer
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom; Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom
| | - Roido Manavaki
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Manfred G Kitzbichler
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Maria A Nettis
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; National Institute for Health and Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; National Institute for Health and Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; National Institute for Health and Research Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, United Kingdom
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom; Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| |
Collapse
|
16
|
Turkheimer FE, Althubaity N, Schubert J, Nettis MA, Cousins O, Dima D, Mondelli V, Bullmore ET, Pariante C, Veronese M. Increased serum peripheral C-reactive protein is associated with reduced brain barriers permeability of TSPO radioligands in healthy volunteers and depressed patients: implications for inflammation and depression. Brain Behav Immun 2021; 91:487-497. [PMID: 33160089 DOI: 10.1016/j.bbi.2020.10.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 01/08/2023] Open
Abstract
The relationship between peripheral and central immunity and how these ultimately may cause depressed behaviour has been the focus of a number of imaging studies conducted with Positron Emission Tomography (PET). These studies aimed at testing the immune-mediated model of depression that proposes a direct effect of peripheral cytokines and immune cells on the brain to elicit a neuroinflammatory response via a leaky blood-brain barrier and ultimately depressive behaviour. However, studies conducted so far using PET radioligands targeting the neuroinflammatory marker 18 kDa translocator protein (TSPO) in patient cohorts with depression have demonstrated mild inflammatory brain status but no correlation between central and peripheral immunity. To gain a better insight into the relationship between heightened peripheral immunity and neuroinflammation, we estimated blood-to-brain and blood-to-CSF perfusion rates for two TSPO radiotracers collected in two separate studies, one large cross-sectional study of neuroinflammation in normal and depressed cohorts (N = 51 patients and N = 25 controls) and a second study where peripheral inflammation in N = 7 healthy controls was induced via subcutaneous injection of interferon (IFN)-α. In both studies we observed a consistent negative association between peripheral inflammation, measured with c-reactive protein P (CRP), and radiotracer perfusion into and from the brain parenchyma and CSF. Importantly, there was no association of this effect with the marker of BBB leakage S100β, that was unchanged. These results suggest a different model of peripheral-to-central immunity interaction whereas peripheral inflammation may cause a reduction in BBB permeability. This effect, on the long term, is likely to disrupt brain homeostasis and induce depressive behavioural symptoms.
Collapse
Affiliation(s)
- Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Noha Althubaity
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Julia Schubert
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Maria A Nettis
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Oliver Cousins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Danai Dima
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychology, School of Arts and Social Sciences, City, University of London, London, UK
| | - Valeria Mondelli
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, UK; Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Carmine Pariante
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
17
|
Wright P, Veronese M, Mazibuko N, Turkheimer FE, Rabiner EA, Ballard CG, Williams SCR, Hari Narayanan AK, Osrah B, Williams R, Marques TR, Howes OD, Roncaroli F, O'Sullivan MJ. Patterns of Mitochondrial TSPO Binding in Cerebral Small Vessel Disease: An in vivo PET Study With Neuropathological Comparison. Front Neurol 2020; 11:541377. [PMID: 33178101 PMCID: PMC7596201 DOI: 10.3389/fneur.2020.541377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Small vessel disease (SVD) is associated with cognitive impairment in older age and be implicated in vascular dementia. Post-mortem studies show proliferation of activated microglia in the affected white matter. However, the role of inflammation in SVD pathogenesis is incompletely understood and better biomarkers are needed. We hypothesized that expression of the 18 kDa translocator protein (TSPO), a marker of microglial activation, would be higher in SVD. Positron emission tomography (PET) was performed with the second-generation TSPO ligand [11C]PBR28 in 11 participants with SVD. TSPO binding was evaluated by a two-tissue compartment model, with and without a vascular binding component, in white matter hyperintensities (WMH) and normal-appearing white matter (NAWM). In post-mortem tissue, in a separate cohort of individuals with SVD, immunohistochemistry was performed for TSPO and a pan-microglial marker Iba1. Kinetic modeling showed reduced tracer volume and blood volume fraction in WMH compared with NAWM, but a significant increase in vascular binding. Vascular [11C]PBR28 binding was also increased compared with normal-appearing white matter of healthy participants free of SVD. Immunohistochemistry showed a diffuse increase in microglial staining (with Iba1) in sampled tissue in SVD compared with control samples, but with only a subset of microglia staining positively for TSPO. Intense TSPO staining was observed in the vicinity of damaged small blood vessels, which included perivascular macrophages. The results suggest an altered phenotype of activated microglia, with reduced TSPO expression, in the areas of greatest white matter ischemia in SVD, with implications for the interpretation of TSPO PET studies in older individuals or those with vascular risk factors.
Collapse
Affiliation(s)
- Paul Wright
- Department of Neuroimaging, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Ndabezinhle Mazibuko
- Department of Neuroimaging, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico E. Turkheimer
- Department of Neuroimaging, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Eugenii A. Rabiner
- Department of Neuroimaging, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
- Invicro, London, United Kingdom
| | - Clive G. Ballard
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Steven C. R. Williams
- Department of Neuroimaging, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Avinash Kumar Hari Narayanan
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Manchester Centre for Clinical Neuroscience, Salford Royal Foundation Trust, Salford, United Kingdom
| | - Bahiya Osrah
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Manchester Centre for Clinical Neuroscience, Salford Royal Foundation Trust, Salford, United Kingdom
| | - Ricky Williams
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Manchester Centre for Clinical Neuroscience, Salford Royal Foundation Trust, Salford, United Kingdom
| | - Tiago R. Marques
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Oliver D. Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Manchester Centre for Clinical Neuroscience, Salford Royal Foundation Trust, Salford, United Kingdom
| | - Michael J. O'Sullivan
- Department of Neuroimaging, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
- University of Queensland Centre for Clinical Research, Brisbane, QLD, Australia
- Department of Neurology, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| |
Collapse
|
18
|
Nettis MA, Veronese M, Nikkheslat N, Mariani N, Lombardo G, Sforzini L, Enache D, Harrison NA, Turkheimer FE, Mondelli V, Pariante CM. PET imaging shows no changes in TSPO brain density after IFN-α immune challenge in healthy human volunteers. Transl Psychiatry 2020; 10:89. [PMID: 32152285 PMCID: PMC7063038 DOI: 10.1038/s41398-020-0768-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
Depression is associated with peripheral inflammation, but its link with brain microglial activity remains unclear. In seven healthy males, we used repeated translocator protein-Positron Emission Tomography (TSPO-PET) dynamic scans with [11C]PBR28 to image brain microglial activation before and 24 h after the immune challenge interferon (IFN)-α. We also investigated the association between changes in peripheral inflammation, changes in microglial activity, and changes in mood. IFN-α administration decreased [11C]PBR28 PET tissue volume of distribution (Vt) across the brain (-20 ± 4%; t6 = 4.1, p = 0.01), but after correction for radioligand free-plasma fraction there were no longer any changes (+23 ± 31%; t = 0.1, p = 0.91). IFN-α increased serum IL-6 (1826 ± 513%, t6 = -7.5, p < 0.001), IL-7 (39 ± 12%, t6 = -3.6, p = 0.01), IL-10 (328 ± 48%, t6 = -12.8, p < 0.001), and IFN-γ (272 ± 64%, t6 = -7.0, p < 0.001) at 4-6 h, and increased serum TNF-α (49 ± 7.6%, t6 = -7.5, p < 0.001), IL-8 (39 ± 12%, t6 = -3.5, p = 0.013), and C-reactive protein (1320 ± 459%, t6 = -7.2, p < 0.001) at 24 h. IFN-α induced temporary mood changes and sickness symptoms after 4-6 h, measured as an increase in POMS-2 total mood score, confusion and fatigue, and a decrease in vigor and friendliness (all p ≤ 0.04). No association was found between changes in peripheral inflammation and changes in PET or mood measures. Our work suggests that brain TSPO-PET signal is highly dependent of inflammation-induced changes in ligand binding to plasma proteins. This limits its usefulness as a sensitive marker of neuroinflammation and consequently, data interpretation. Thus, our results can be interpreted as showing either that [11C]PBR28 is not sensitive enough under these conditions, or that there is simply no microglial activation in this model.
Collapse
Affiliation(s)
- M A Nettis
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK.
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| | - M Veronese
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
- Institute of Psychiatry, Psychology and Neuroscience, King's College London Department of Neuroimaging, London, UK
| | - N Nikkheslat
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
| | - N Mariani
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
| | - G Lombardo
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
| | - L Sforzini
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
- Universita' degli Studi di Milano, Psychiatry Unit, Department of Biomedical and Clinical Sciences, Luigi Sacco Hospital, Milan, Italy
| | - D Enache
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Stockholm, Sweden
| | - N A Harrison
- Cardiff University Brain Research Imaging Centre (CUBRIC), Cardiff University, Cardiff, UK
| | - F E Turkheimer
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
- Institute of Psychiatry, Psychology and Neuroscience, King's College London Department of Neuroimaging, London, UK
| | - V Mondelli
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - C M Pariante
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Department of Psychological Medicine, London, UK
- National Institute for Health and Research Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
19
|
Chandra A, Valkimadi PE, Pagano G, Cousins O, Dervenoulas G, Politis M. Applications of amyloid, tau, and neuroinflammation PET imaging to Alzheimer's disease and mild cognitive impairment. Hum Brain Mapp 2019; 40:5424-5442. [PMID: 31520513 PMCID: PMC6864887 DOI: 10.1002/hbm.24782] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/29/2019] [Accepted: 08/18/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating and progressive neurodegenerative disease for which there is no cure. Mild cognitive impairment (MCI) is considered a prodromal stage of the disease. Molecular imaging with positron emission tomography (PET) allows for the in vivo visualisation and tracking of pathophysiological changes in AD and MCI. PET is a very promising methodology for differential diagnosis and novel targets of PET imaging might also serve as biomarkers for disease-modifying therapeutic interventions. This review provides an overview of the current status and applications of in vivo molecular imaging of AD pathology, specifically amyloid, tau, and microglial activation. PET imaging studies were included and evaluated as potential biomarkers and for monitoring disease progression. Although the majority of radiotracers showed the ability to discriminate AD and MCI patients from healthy controls, they had various limitations that prevent the recommendation of a single technique or tracer as an optimal biomarker. Newer research examining amyloid, tau, and microglial PET imaging in combination suggest an alternative approach in studying the disease process.
Collapse
Affiliation(s)
- Avinash Chandra
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Polytimi-Eleni Valkimadi
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Gennaro Pagano
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Oliver Cousins
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - George Dervenoulas
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| | - Marios Politis
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London (KCL), London, UK
| |
Collapse
|
20
|
Song YS. Perspectives in TSPO PET Imaging for Neurologic Diseases. Nucl Med Mol Imaging 2019; 53:382-385. [PMID: 31867073 DOI: 10.1007/s13139-019-00620-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 01/30/2023] Open
Abstract
The translocator protein (18 kDa) (TSPO) is a mitochondrial transmembrane protein, which has brought attention as a neuroinflammatory biomarker. Positron emission tomography (PET) imaging studies have been done for several decades, since neuroinflammation has been implicated as an important pathophysiology of several common neurologic disorders. However, despite numerous previous studies with positive findings, its clinical significance is not yet clear. Various attempts to overcome the limitations are ongoing, in order to bring acceptance for use in clinics.
Collapse
Affiliation(s)
- Yoo Sung Song
- Seoul National University Bundang Hospital, Seongnam, South Korea
| |
Collapse
|
21
|
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: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
22
|
Dahoun T, Calcia MA, Veronese M, Bloomfield P, Reis Marques T, Turkheimer F, Howes OD. The association of psychosocial risk factors for mental health with a brain marker altered by inflammation: A translocator protein (TSPO) PET imaging study. Brain Behav Immun 2019; 80:742-750. [PMID: 31112791 DOI: 10.1016/j.bbi.2019.05.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 12/11/2022] Open
Abstract
Psychiatric disorders associated with psychosocial risk factors, including depression and psychosis, have been shown to demonstrate increased microglia activity. Whilst preclinical studies indicate that psychosocial stress leads to increased levels of microglia in the frontal cortex, no study has yet been performed in humans. This study aimed at investigating whether psychosocial risk factors for depression and/or psychosis would be associated with alterations in a brain marker expressed by microglia, the translocator specific protein (TSPO) in humans. We used [11C]-PBR28 Positron Emission Tomography on healthy subjects exposed to childhood and adulthood psychosocial risk factors (high-risk group, N = 12) and age- and sex-matched healthy controls not exposed to childhood and adulthood psychosocial risk factors (low-risk group, N = 12). The [11C]-PBR28 volume of distribution (VT) and Distribution Volume Ratio (DVR) were measured in the total gray matter, and frontal, parietal, temporal, occipital lobes. Levels of childhood trauma, anxiety and depression were measured using respectively the Childhood Trauma Questionnaire, State-anxiety questionnaire and Beck Depression Inventory. Compared to the low-risk group, the high-risk group did not exhibit significant differences in the mean [11C]-PBR28 VT (F(1,20) = 1.619, p = 0.218) or DVR (F(1,22) = 0.952, p = 0.340) on any region. There were no significant correlations between the [11C]-PBR28 VT and DVRs in total gray matter and frontal lobe and measures of childhood trauma, anxiety and depression. Psychosocial risk factors for depression and/or psychosis are unlikely to be associated with alterations in [11C]-PBR28 binding, indicating that alterations in TSPO expression reported in these disorders is unlikely to be caused by psychosocial risk factors alone.
Collapse
Affiliation(s)
- Tarik Dahoun
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX37 JX, UK
| | - Marilia A Calcia
- Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Peter Bloomfield
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK
| | - Tiago Reis Marques
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK
| | - Oliver D Howes
- Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London W12 0NN, UK; Institute of Psychiatry, Neurology and Neuroscience (IoPPN), King's College London, London SE5 8AF, UK.
| |
Collapse
|
23
|
Dani M, Wood M, Mizoguchi R, Fan Z, Walker Z, Morgan R, Hinz R, Biju M, Kuruvilla T, Brooks DJ, Edison P. Microglial activation correlates in vivo with both tau and amyloid in Alzheimer's disease. Brain 2019; 141:2740-2754. [PMID: 30052812 DOI: 10.1093/brain/awy188] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 05/29/2018] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease is characterized by the histopathological presence of amyloid-β plaques and tau-containing neurofibrillary tangles. Microglial activation is also a recognized pathological component. The relationship between microglial activation and protein aggregation is still debated. We investigated the relationship between amyloid plaques, tau tangles and activated microglia using PET imaging. Fifty-one subjects (19 healthy controls, 16 mild cognitive impairment and 16 Alzheimer's disease subjects) participated in the study. All subjects had neuropsychometric testing, MRI, amyloid (18F-flutemetamol), and microglial (11C-PBR28) PET. All subjects with mild cognitive impairment and Alzheimer's disease and eight of the controls had tau (18F-AV1451) PET. 11C-PBR28 PET was analysed using Logan graphical analysis with an arterial plasma input function, while 18F-flutemetamol and 18F-AV1451 PET were analysed as target:cerebellar ratios to create parametric standardized uptake value ratio maps. Biological parametric mapping in the Statistical Parametric Mapping platform was used to examine correlations between uptake of tracers at a voxel-level. There were significant widespread clusters of positive correlation between levels of microglial activation and tau aggregation in both the mild cognitive impairment (amyloid-positive and amyloid-negative) and Alzheimer's disease subjects. The correlations were stronger in Alzheimer's disease than in mild cognitive impairment, suggesting that these pathologies increase together as disease progresses. Levels of microglial activation and amyloid deposition were also correlated, although in a different spatial distribution; correlations were stronger in mild cognitive impairment than Alzheimer's subjects, in line with a plateauing of amyloid load with disease progression. Clusters of positive correlations between microglial activation and protein aggregation often targeted similar areas of association cortex, indicating that all three processes are present in specific vulnerable brain areas. For the first time using PET imaging, we show that microglial activation can correlate with both tau aggregation and amyloid deposition. This confirms the complex relationship between these processes. These results suggest that preventative treatment for Alzheimer's disease should target all three processes.
Collapse
Affiliation(s)
- Melanie Dani
- Neurology Imaging Unit, Department of Medicine, Imperial College London, Hammersmith Hospital, UK
| | - Melanie Wood
- Neurology Imaging Unit, Department of Medicine, Imperial College London, Hammersmith Hospital, UK
| | - Ruth Mizoguchi
- Neurology Imaging Unit, Department of Medicine, Imperial College London, Hammersmith Hospital, UK
| | - Zhen Fan
- Neurology Imaging Unit, Department of Medicine, Imperial College London, Hammersmith Hospital, UK
| | - Zuzana Walker
- Division of Psychiatry, University College London, UK.,Essex Partnership University NHS Foundation Trust, UK
| | | | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, UK
| | - Maya Biju
- Gether NHS Foundation Trust, Gloucester, UK
| | | | - David J Brooks
- Neurology Imaging Unit, Department of Medicine, Imperial College London, Hammersmith Hospital, UK.,Department of Nuclear Medicine, Aarhus University, Denmark.,Institute of Neuroscience, University of Newcastle upon Tyne, UK
| | - Paul Edison
- Neurology Imaging Unit, Department of Medicine, Imperial College London, Hammersmith Hospital, UK
| |
Collapse
|
24
|
Ghadery C, Best LA, Pavese N, Tai YF, Strafella AP. PET Evaluation of Microglial Activation in Non-neurodegenerative Brain Diseases. Curr Neurol Neurosci Rep 2019; 19:38. [PMID: 31139952 PMCID: PMC6538572 DOI: 10.1007/s11910-019-0951-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF THE REVIEW Microglial cell activation is an important component of neuroinflammation, and it is generally well accepted that chronic microglial activation is indicative of accumulating tissue damage in neurodegenerative conditions, particularly in the earlier stages of disease. Until recently, there has been less focus on the role of neuroinflammation in other forms of neurological and neuropsychiatric conditions. Through this review, we hope to demonstrate the important role TSPO PET imaging has played in illuminating the pivotal role of neuroinflammation and microglial activation underpinning these conditions. RECENT FINDINGS TSPO is an 18 kDa protein found on the outer membrane of mitochondria and can act as a marker of microglial activation using nuclear imaging. Through the development of radiopharmaceuticals targeting TSPO, researchers have been able to better characterise the spatial-temporal evolution of chronic neurological conditions, ranging from the focal autoimmune reactions seen in multiple sclerosis to the Wallerian degeneration at remote parts of the brain months following acute cerebral infarction. Development of novel techniques to investigate neuroinflammation within the central nervous system, for the purposes of diagnosis and therapeutics, has flourished over the past few decades. TSPO has proven itself a robust and sensitive biomarker of microglial activation and neuroimaging affords a minimally invasive technique to characterise neuroinflammatory processes in vivo.
Collapse
Affiliation(s)
- Christine Ghadery
- The Edmond J. Safra Program in Parkinson's Disease & Movement Disorder Unit, Toronto Western Hospital & Krembil Research Institute, University Health Network; Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Laura A Best
- Clinical Ageing Research Unit, Newcastle University, Campus for Ageing and Vitality, Westgate Road, Newcastle Upon Tyne, UK.
| | - Nicola Pavese
- Clinical Ageing Research Unit, Newcastle University, Campus for Ageing and Vitality, Westgate Road, Newcastle Upon Tyne, UK
- PET centre, University of Aarhus Denmark, Aarhus, Denmark
| | - Yen Foung Tai
- Imperial College London South Kensington Campus, London, UK
| | - Antonio P Strafella
- The Edmond J. Safra Program in Parkinson's Disease & Movement Disorder Unit, Toronto Western Hospital & Krembil Research Institute, University Health Network; Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
(R)-[ 18F]NEBIFQUINIDE: A promising new PET tracer for TSPO imaging. Eur J Med Chem 2019; 176:410-418. [PMID: 31125895 DOI: 10.1016/j.ejmech.2019.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/19/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) imaging of the 18 kDa translocator protein (TSPO), has a high diagnostic potential in neurodegenerative disorders and cancer. However, TSPO is considered a challenge for molecular imaging due to the poor availability of suitable radiotracers with adequate pharmacokinetic properties. Here, we describe the development of a radiofluorinated pyridinyl isoquinoline analogue of the established TSPO PET tracer (R)-[11C]PK11195 with improved binding properties in all known human TSPO phenotypes. We conducted a complete preclinical evaluation using in vitro, in vivo and ex vivo methods to assess the performance of this novel radiotracer and observed high specific binding of the radiotracer to TSPO, as well as high metabolic stability. Therefore, we propose this radiolabeled compound for further evaluation in animal models as well as in clinical trials.
Collapse
|
26
|
Rizzo G, Veronese M, Tonietto M, Bodini B, Stankoff B, Wimberley C, Lavisse S, Bottlaender M, Bloomfield PS, Howes O, Zanotti-Fregonara P, Turkheimer FE, Bertoldo A. Generalization of endothelial modelling of TSPO PET imaging: Considerations on tracer affinities. J Cereb Blood Flow Metab 2019; 39:874-885. [PMID: 29135382 PMCID: PMC6501510 DOI: 10.1177/0271678x17742004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The 18 kDa translocator protein (TSPO) is a marker of microglia activation and the main target of positron emission tomography (PET) ligands for neuroinflammation. Previous works showed that accounting for TSPO endothelial binding improves PET quantification for [11C]PBR28, [18F]DPA714 and [11C]-R-PK11195. It is still unclear, however, whether the vascular signal is tracer-dependent. This work aims to explore the relationship between the TSPO vascular and tissue components for PET tracers with varying affinity, also assessing the impact of affinity towards the differentiability amongst kinetics and the ensuing ligand amenability to cluster analysis for the extraction of a reference region. First, we applied the compartmental model accounting for vascular binding to [11C]-R-PK11195 data from six healthy subjects. Then, we compared the [11C]-R-PK11195 vascular binding estimates with previously published values for [18F]DPA714 and [11C]PBR28. Finally, we determined the suitability for reference region extraction by calculating the angle between grey and white matter kinetics. Our results showed that endothelial binding is common to all TSPO tracers and proportional to their affinity. By consequence, grey and white matter kinetics were most similar for the radioligand with the highest affinity (i.e. [11C]PBR28), hence poorly suited for the extraction of a reference region using supervised clustering.
Collapse
Affiliation(s)
- Gaia Rizzo
- 1 Department of Information Engineering, Padova University, Padova, Italy
| | - Mattia Veronese
- 2 Department of Neuroimaging, King's College London, London, UK
| | - Matteo Tonietto
- 3 UPMC, Institut du Cerveau et de la Moelle épinière, Hôpital de la Pitié Salpêtrière, Sorbonne Universités, Paris, France
| | - Benedetta Bodini
- 3 UPMC, Institut du Cerveau et de la Moelle épinière, Hôpital de la Pitié Salpêtrière, Sorbonne Universités, Paris, France.,4 Assistance Publique des Hopitaux de Paris, APHP, Hôpital Saint Antoine, Paris, France
| | - Bruno Stankoff
- 3 UPMC, Institut du Cerveau et de la Moelle épinière, Hôpital de la Pitié Salpêtrière, Sorbonne Universités, Paris, France.,4 Assistance Publique des Hopitaux de Paris, APHP, Hôpital Saint Antoine, Paris, France.,5 IMIV, Inserm, CEA, Paris-Sud Univ, Université Paris Saclay, Orsay, France
| | - Catriona Wimberley
- 5 IMIV, Inserm, CEA, Paris-Sud Univ, Université Paris Saclay, Orsay, France
| | - Sonia Lavisse
- 6 Département de Recherche Fondamentale (DRF), Institut d'Imagerie Biomédicale (I2BM), Fontenay-aux-Roses, France.,7 Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Michel Bottlaender
- 5 IMIV, Inserm, CEA, Paris-Sud Univ, Université Paris Saclay, Orsay, France.,8 Neurospin, CEA, Gif-sur-Yvette, France
| | | | - Oliver Howes
- 9 Institute of Clinical Sciences, Imperial College London, London, UK.,10 Department of Psychosis Studies, King's College London, London, UK
| | - Paolo Zanotti-Fregonara
- 11 Houston Methodist Hospital, PET Core Facility, Research Institute, Stanley H. Appel Department of Neurology, Houston, Texas, USA
| | | | - Alessandra Bertoldo
- 1 Department of Information Engineering, Padova University, Padova, Italy.,12 Padua Neuroscience Center, University of Padova, Padova, Italy
| |
Collapse
|
27
|
Stadulytė A, Alcaide-Corral CJ, Walton T, Lucatelli C, Tavares AAS. Analysis of PK11195 concentrations in rodent whole blood and tissue samples by rapid and reproducible chromatographic method to support target-occupancy PET studies. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1118-1119:33-39. [PMID: 31005772 PMCID: PMC6522057 DOI: 10.1016/j.jchromb.2019.04.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022]
Abstract
In Positron Emission Tomography (PET) research, it is important to assess not only pharmacokinetics of a radiotracer in vivo, but also of the drugs used in blocking/displacement PET studies. Typically, pharmacokinetic/pharmacodynamic (PK/PD) analyses of drugs used in rodent PET studies are based on population average pharmacokinetic profiles of the drugs due to limited blood volume withdrawal while simultaneously maintaining physiological homeostasis. This likely results in bias of PET data quantification, including unknown bias of target occupancy (TO) measurements. This study aimed to develop a High Performance Liquid Chromatography (HPLC) method for PK/PD quantification of drugs used in preclinical rodent PET research, specifically the translocator 18 kDa protein (TSPO) selective drug, PK11195, that used sub-millilitre blood volumes. The lowest detection limit for the proposed HPLC method ranged between 7.5 and 10 ng/mL depending on the method used to calculate the limit of detection, and the measured average relative standard deviation for intermediate precision was equal to 17.2%. Most importantly, we were able to demonstrate a significant difference between calculated PK11195 concentrations at 0.5, 1, 2, 3, 5, 15 and 30 min post-administration and individually measured whole blood levels (significance level range from p < 0.05 to p < 0.001; one-way ANOVA, Dunnet's post hoc test, p < 0.05). The HPLC method developed here uses sub-millilitre sample volumes to reproducibly assess PK/PD of PK11195 in rodent blood. This study highlights the importance of individually measured PK/PD drug concentrations when quantifying the TO from blocking/displacement rodent PET experiments.
Collapse
Affiliation(s)
- Agnė Stadulytė
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, UK; Edinburgh Preclinical Imaging (EPI), University of Edinburgh, UK.
| | - Carlos José Alcaide-Corral
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, UK; Edinburgh Preclinical Imaging (EPI), University of Edinburgh, UK
| | - Tashfeen Walton
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, UK; Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, UK
| | - Christophe Lucatelli
- Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, UK
| | - Adriana Alexandre S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, UK; Edinburgh Preclinical Imaging (EPI), University of Edinburgh, UK
| |
Collapse
|
28
|
Sestini S, Alongi P, Berti V, Calcagni ML, Cecchin D, Chiaravalloti A, Chincarini A, Cistaro A, Guerra UP, Pappatà S, Tiraboschi P, Nobili F. The role of molecular imaging in the frame of the revised dementia with Lewy body criteria. Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00321-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Synthesis and in vitro evaluation of new translocator protein ligands designed for positron emission tomography. Future Med Chem 2019; 11:539-550. [PMID: 30888874 DOI: 10.4155/fmc-2018-0444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
AIM Dysregulated levels of the translocator protein TSPO 18 KDa have been reported in several disorders, particularly neurodegenerative diseases. This makes TSPO an interesting target for the development of diagnostic biomarkers. Even though several radioligands have already been developed for in vivo TSPO imaging, the ideal TSPO radiotracer has still not been found. RESULTS Here, we report the chemical synthesis of a set of new TSPO ligands designed for future application in positron emission tomography, together with the determination of their biological activity and applied 11C-labeling strategy. CONCLUSION The lead compound of our series, (R)-[11C]Me@NEBIQUINIDE, showed very promising results and is therefore proposed to be further evaluated under in vivo settings.
Collapse
|
30
|
VanElzakker MB, Brumfield SA, Lara Mejia PS. Neuroinflammation and Cytokines in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): A Critical Review of Research Methods. Front Neurol 2019; 9:1033. [PMID: 30687207 PMCID: PMC6335565 DOI: 10.3389/fneur.2018.01033] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 11/16/2018] [Indexed: 01/18/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is the label given to a syndrome that can include long-term flu-like symptoms, profound fatigue, trouble concentrating, and autonomic problems, all of which worsen after exertion. It is unclear how many individuals with this diagnosis are suffering from the same condition or have the same underlying pathophysiology, and the discovery of biomarkers would be clarifying. The name "myalgic encephalomyelitis" essentially means "muscle pain related to central nervous system inflammation" and many efforts to find diagnostic biomarkers have focused on one or more aspects of neuroinflammation, from periphery to brain. As the field uncovers the relationship between the symptoms of this condition and neuroinflammation, attention must be paid to the biological mechanisms of neuroinflammation and issues with its potential measurement. The current review focuses on three methods used to study putative neuroinflammation in ME/CFS: (1) positron emission tomography (PET) neuroimaging using translocator protein (TSPO) binding radioligand (2) magnetic resonance spectroscopy (MRS) neuroimaging and (3) assays of cytokines circulating in blood and cerebrospinal fluid. PET scanning using TSPO-binding radioligand is a promising option for studies of neuroinflammation. However, methodological difficulties that exist both in this particular technique and across the ME/CFS neuroimaging literature must be addressed for any results to be interpretable. We argue that the vast majority of ME/CFS neuroimaging has failed to use optimal techniques for studying brainstem, despite its probable centrality to any neuroinflammatory causes or autonomic effects. MRS is discussed as a less informative but more widely available, less invasive, and less expensive option for imaging neuroinflammation, and existing studies using MRS neuroimaging are reviewed. Studies seeking to find a peripheral circulating cytokine "profile" for ME/CFS are reviewed, with attention paid to the biological and methodological reasons for lack of replication among these studies. We argue that both the biological mechanisms of cytokines and the innumerable sources of potential variance in their measurement make it unlikely that a consistent and replicable diagnostic cytokine profile will ever be discovered.
Collapse
Affiliation(s)
- Michael B. VanElzakker
- Division of Neurotherapeutics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | | |
Collapse
|
31
|
Felger JC. Imaging the Role of Inflammation in Mood and Anxiety-related Disorders. Curr Neuropharmacol 2018; 16:533-558. [PMID: 29173175 PMCID: PMC5997866 DOI: 10.2174/1570159x15666171123201142] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 10/23/2017] [Accepted: 11/21/2017] [Indexed: 02/08/2023] Open
Abstract
Background Studies investigating the impact of a variety of inflammatory stimuli on the brain and behavior have reported evidence that inflammation and release of inflammatory cytokines affect circuitry relevant to both reward and threat sensitivity to contribute to behavioral change. Of relevance to mood and anxiety-related disorders, biomarkers of inflammation such as inflammatory cytokines and acute-phase proteins are reliably elevated in a significant proportion of patients with major depressive disorder (MDD), bipolar disorder, anxiety disorders and post-traumatic stress disorder (PTSD). Methods This review summarized clinical and translational work demonstrating the impact of peripheral inflammation on brain regions and neurotransmitter systems relevant to both reward and threat sensitivity, with a focus on neuroimaging studies involving administration of inflammatory stimuli. Recent translation of these findings to further understand the role of inflammation in mood and anxiety-related disorders is also discussed. Results Inflammation was consistently found to affect basal ganglia and cortical reward and motor circuits to drive reduced motivation and motor activity, as well as anxiety-related brain regions including amygdala, insula and anterior cingulate cortex, which may result from cytokine effects on monoamines and glutamate. Similar relationships between inflammation and altered neurocircuitry have been observed in MDD patients with increased peripheral inflammatory markers, and such work is on the horizon for anxiety disorders and PTSD. Conclusion Neuroimaging effects of inflammation on reward and threat circuitry may be used as biomarkers of inflammation for future development of novel therapeutic strategies to better treat mood and anxiety-related disorders in patients with high inflammation.
Collapse
Affiliation(s)
- Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States.,Winship Cancer Institute, Emory University, Atlanta, GA, United States
| |
Collapse
|
32
|
TSPO in diverse CNS pathologies and psychiatric disease: A critical review and a way forward. Pharmacol Ther 2018; 194:44-58. [PMID: 30189290 DOI: 10.1016/j.pharmthera.2018.09.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The use of Translocator Protein 18 kDa (TSPO) as a clinical neuroimaging biomarker of brain injury and neuroinflammation has increased exponentially in the last decade. There has been a furious pace in the development of new radiotracers for TSPO positron emission tomography (PET) imaging and its use has now been extensively described in many neurological and mental disorders. This fast pace of research and the ever-increasing number of new laboratories entering the field often times lack an appreciation of the historical perspective of the field and introduce dogmatic, but unproven facts, related to the underlying neurobiology of the TSPO response to brain injury and neuroinflammation. Paradoxically, while in neurodegenerative disorders and in all types of CNS pathologies brain TSPO levels increase, a new observation in psychiatric disorders such as schizophrenia is decreased brain levels of TSPO measured by PET. The neurobiological bases for this new finding is currently not known, but rigorous experimental design using multiple experimental approaches and careful interpretation of results is critically important to provide the methodological and/or biological underpinnings to this new observation. This review provides a perspective of the early history of validating TSPO as a biomarker of brain injury and neuroinflammation and a critical analysis of controversial topics in the literature related to the cellular sources of the TSPO response. The latter is important in order to provide the correct interpretation of PET studies in neurodegenerative and psychiatric disorders. Furthermore, this review proposes some yet to be explored explanations to new findings in psychiatric disorders and new approaches to quantitatively assess the glial sources of the TSPO response in order to move the field forward.
Collapse
|
33
|
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
|
34
|
Edison P, Donat CK, Sastre M. In vivo Imaging of Glial Activation in Alzheimer's Disease. Front Neurol 2018; 9:625. [PMID: 30131755 PMCID: PMC6090997 DOI: 10.3389/fneur.2018.00625] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by memory loss and decline of cognitive function, associated with progressive neurodegeneration. While neuropathological processes like amyloid plaques and tau neurofibrillary tangles have been linked to neuronal death in AD, the precise role of glial activation on disease progression is still debated. It was suggested that neuroinflammation could occur well ahead of amyloid deposition and may be responsible for clearing amyloid, having a neuroprotective effect; however, later in the disease, glial activation could become deleterious, contributing to neuronal toxicity. Recent genetic and preclinical studies suggest that the different activation states of microglia and astrocytes are complex, not as polarized as previously thought, and that the heterogeneity in their phenotype can switch during disease progression. In the last few years, novel imaging techniques e.g., new radiotracers for assessing glia activation using positron emission tomography and advanced magnetic resonance imaging technologies have emerged, allowing the correlation of neuro-inflammatory markers with cognitive decline, brain function and brain pathology in vivo. Here we review all new imaging technology in AD patients and animal models that has the potential to serve for early diagnosis of the disease, to monitor disease progression and to test the efficacy and the most effective time window for potential anti-inflammatory treatments.
Collapse
Affiliation(s)
- Paul Edison
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Cornelius K Donat
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Magdalena Sastre
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
35
|
Veronese M, Reis Marques T, Bloomfield PS, Rizzo G, Singh N, Jones D, Agushi E, Mosses D, Bertoldo A, Howes O, Roncaroli F, Turkheimer FE. Kinetic modelling of [ 11C]PBR28 for 18 kDa translocator protein PET data: A validation study of vascular modelling in the brain using XBD173 and tissue analysis. J Cereb Blood Flow Metab 2018; 38:1227-1242. [PMID: 28580888 PMCID: PMC6434448 DOI: 10.1177/0271678x17712388] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The 18 kDa translocator protein (TSPO) is a marker of microglia activation in the central nervous system and represents the main target of radiotracers for the in vivo quantification of neuroinflammation with positron emission tomography (PET). TSPO PET is methodologically challenging given the heterogeneous distribution of TSPO in blood and brain. Our previous studies with the TSPO tracers [11C]PBR28 and [11C]PK11195 demonstrated that a model accounting for TSPO binding to the endothelium improves the quantification of PET data. Here, we performed a validation of the kinetic model with the additional endothelial compartment through a displacement study. Seven subjects with schizophrenia, all high-affinity binders, underwent two [11C]PBR28 PET scans before and after oral administration of 90 mg of the TSPO ligand XBD173. The addition of the endothelial component provided a signal compartmentalization much more consistent with the underlying biology, as only in this model, the blocking study produced the expected reduction in the tracer concentration of the specific tissue compartment, whereas the non-displaceable compartment remained unchanged. In addition, we also studied TSPO expression in vessels using 3D reconstructions of histological data of frontal lobe and cerebellum, demonstrating that TSPO positive vessels account for 30% of the vascular volume in cortical and white matter.
Collapse
Affiliation(s)
- Mattia Veronese
- Department of Neuroimaging, IoPPN,
King’s College London, London, UK
| | - Tiago Reis Marques
- Department of Psychosis Studies, IoPPN,
King’s College London, London, UK
- Institute of Clinical Sciences, Imperial
College London, London, UK
| | | | - Gaia Rizzo
- Department of Information Engineering,
Padova University, Padova, Italy
| | - Nisha Singh
- Department of Neuroimaging, IoPPN,
King’s College London, London, UK
| | - Deborah Jones
- Department of Cellular Pathology,
Salford Royal Foundation Trust, Salford, UK
| | - Erjon Agushi
- Division of Neuroscience and
Experimental Psychology, University of Manchester, UK
| | - Dominic Mosses
- Division of Neuroscience and
Experimental Psychology, University of Manchester, UK
| | - Alessandra Bertoldo
- Department of Information Engineering,
Padova University, Padova, Italy
- Padua Neuroscience Center, University of
Padova, Padova, Italy
| | - Oliver Howes
- Department of Psychosis Studies, IoPPN,
King’s College London, London, UK
- Institute of Clinical Sciences, Imperial
College London, London, UK
| | - Federico Roncaroli
- Division of Neuroscience and
Experimental Psychology, University of Manchester, UK
| | - Federico E Turkheimer
- Department of Neuroimaging, IoPPN,
King’s College London, London, UK
- Federico E Turkheimer, Centre for
Neuroimaging Sciences, IoPPN, King’s College London, P089, De Crespigny Park,
Denmark Hill, London SE5 8AF, UK.
| |
Collapse
|
36
|
Selvaraj S, Bloomfield PS, Cao B, Veronese M, Turkheimer F, Howes OD. Brain TSPO imaging and gray matter volume in schizophrenia patients and in people at ultra high risk of psychosis: An [ 11C]PBR28 study. Schizophr Res 2018; 195:206-214. [PMID: 28893493 PMCID: PMC6027955 DOI: 10.1016/j.schres.2017.08.063] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/31/2017] [Accepted: 08/31/2017] [Indexed: 12/12/2022]
Abstract
Patients with schizophrenia show whole brain and cortical gray matter (GM) volume reductions which are progressive early in their illness. Microglia, the resident immune cells in the CNS, phagocytose neurons and synapses. Some post mortem and in vivo studies in schizophrenia show evidence for elevated microglial activation compared to matched controls. However, it is currently unclear how these results relate to changes in cortical structure. METHODS Fourteen patients with schizophrenia and 14 ultra high risk for psychosis (UHR) subjects alongside two groups of age and genotype matched healthy controls received [11C]PBR28 PET scans to index TSPO expression, a marker of microglial activation and a 3T MRI scan. We investigated the relationship between the volume changes of cortical regions and microglial activation in cortical GM (as indexed by [11C]PBR28 distribution volume ratio (DVR). RESULTS The total cortical GM volume was significantly lower in SCZ than the controls [mean (SD)/cm3: SCZ=448.83 (39.2) and controls=499.6 (59.2) (p=0.02) but not in UHR (mean (SD)=503.06 (57.9) and controls=524.46 (45.3) p=0.3). Regression model fitted the total cortical GM DVR values with the cortical regional volumes in SCZ (r=0.81; p<0.001) and in UHR (r=0.63; p=0.02). We found a significant negative correlation between the TSPO signal and total cortical GM volume in SCZ with the highest absolute correlation coefficient in the right superior-parietal cortex (r=-0.72; p=0.006). CONCLUSIONS These findings suggest that microglial activity is related to the altered cortical volume seen in schizophrenia. Longitudinal investigations are required to determine whether microglial activation leads to cortical gray matter loss.
Collapse
Affiliation(s)
- Sudhakar Selvaraj
- Department of Psychiatry and Behavioural Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; Psychiatric Imaging Group, MRC Clinical Sciences Centre, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.
| | - Peter S Bloomfield
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Bo Cao
- Department of Psychiatry and Behavioural Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, IoPPN, King's College London, Box PO89, De Crespigny Park, London SE5 8AF, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, IoPPN, King's College London, Box PO89, De Crespigny Park, London SE5 8AF, UK
| | - Oliver D Howes
- Psychiatric Imaging Group, MRC Clinical Sciences Centre, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| |
Collapse
|
37
|
Guedes CES, Dias BRS, Petersen ALDOA, Cruz KP, Almeida NDJ, Andrade DR, Menezes JPBD, Borges VDM, Veras PST. In vitro evaluation of the anti-leishmanial activity and toxicity of PK11195. Mem Inst Oswaldo Cruz 2018; 113:e170345. [PMID: 29412342 PMCID: PMC5851033 DOI: 10.1590/0074-02760170345] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/30/2017] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Leishmaniasis, one of the most neglected diseases, is a serious public health problem in many countries, including Brazil. Currently available treatments require long-term use and have serious side effects, necessitating the development of new therapeutic interventions. Because translocator protein (TSPO) levels are reduced in Leishmania amazonensis-infected cells and because this protein participates in apoptosis and immunomodulation, TSPO represents a potential target for Leishmania chemotherapy. The present study evaluated PK11195, a ligand of this protein, as an anti-leishmanial agent. OBJECTIVE To evaluate the leishmanicidal activity of PK11195 against L. amazonensis in infected CBA mouse macrophages in vitro. METHODS The viability of axenic L. amazonensis, Leishmania major, and Leishmania braziliensis promastigotes was assessed after 48 h treatment with PK11195 (0.2-400 µM). Additionally, intracellular parasite viability was evaluated to determine IC50 values and the number of viable parasites in infected macrophages treated with PK11195 (50-100 µM). Infected macrophages were then treated with PK11195 (25-100 µM) to determine the percentage of L. amazonensis-infected cells and the number of parasites per infected cell. Electron microscopy was used to investigate morphological changes caused by PK11195. The production of free oxygen radicals, nitric oxide, and pro-inflammatory cytokines was also evaluated in infected macrophages treated with PK11195 and primed or not primed with IFN-γ. FINDINGS Median IC50 values for PK11195 were 14.2 µM for L. amazonensis, 8.2 µM for L. major, and 3.5 µM for L. braziliensis. The selective index value for L. amazonensis was 13.7, indicating the safety of PK11195 for future testing in mammals. Time- and dose-dependent reductions in the percentage of infected macrophages, the number of parasites per infected macrophage, and the number of viable intracellular parasites were observed. Electron microscopy revealed some morphological alterations suggestive of autophagy. Interestingly, MCP-1 and superoxide levels were reduced in L. amazonensis-infected macrophages treated with PK11195. MAIN CONCLUSIONS PK11195 causes the killing of amastigotes in vitro by mechanisms independent of inflammatory mediators and causes morphological alterations within Leishmania parasites, suggestive of autophagy, at doses that are non-toxic to macrophages. Thus, this molecule has demonstrated potential as an anti-leishmanial agent.
Collapse
Affiliation(s)
- Carlos Eduardo Sampaio Guedes
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Gonçalo Moniz, Laboratório de Patologia e Biointervenção, Salvador, BA, Brasil
| | - Beatriz Rocha Simões Dias
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Gonçalo Moniz, Laboratório de Patologia e Biointervenção, Salvador, BA, Brasil
| | | | - Kercia Pinheiro Cruz
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Gonçalo Moniz, Laboratório de Patologia e Biointervenção, Salvador, BA, Brasil
| | - Niara de Jesus Almeida
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Gonçalo Moniz, Laboratório de Patologia e Biointervenção, Salvador, BA, Brasil
| | - Daniela Rodrigues Andrade
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Gonçalo Moniz, Laboratório Integrado de Microbiologia e Imunoregulação, Salvador, BA, Brasil
| | | | - Valéria de Matos Borges
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Gonçalo Moniz, Laboratório Integrado de Microbiologia e Imunoregulação, Salvador, BA, Brasil
| | - Patricia Sampaio Tavares Veras
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Gonçalo Moniz, Laboratório de Patologia e Biointervenção, Salvador, BA, Brasil
| |
Collapse
|
38
|
Cumming P, Burgher B, Patkar O, Breakspear M, Vasdev N, Thomas P, Liu GJ, Banati R. Sifting through the surfeit of neuroinflammation tracers. J Cereb Blood Flow Metab 2018; 38:204-224. [PMID: 29256293 PMCID: PMC5951023 DOI: 10.1177/0271678x17748786] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/26/2017] [Accepted: 11/09/2017] [Indexed: 01/09/2023]
Abstract
The first phase of molecular brain imaging of microglial activation in neuroinflammatory conditions began some 20 years ago with the introduction of [11C]-( R)-PK11195, the prototype isoquinoline ligand for translocator protein (18 kDa) (TSPO). Investigations by positron emission tomography (PET) revealed microgliosis in numerous brain diseases, despite the rather low specific binding signal imparted by [11C]-( R)-PK11195. There has since been enormous expansion of the repertoire of TSPO tracers, many with higher specific binding, albeit complicated by allelic dependence of the affinity. However, the specificity of TSPO PET for revealing microglial activation not been fully established, and it has been difficult to judge the relative merits of the competing tracers and analysis methods with respect to their sensitivity for detecting microglial activation. We therefore present a systematic comparison of 13 TSPO PET and single photon computed tomography (SPECT) tracers belonging to five structural classes, each of which has been investigated by compartmental analysis in healthy human brain relative to a metabolite-corrected arterial input. We emphasize the need to establish the non-displaceable binding component for each ligand and conclude with five recommendations for a standard approach to define the cellular distribution of TSPO signals, and to characterize the properties of candidate TSPO tracers.
Collapse
Affiliation(s)
- Paul Cumming
- School of Psychology and Counselling and IHBI, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- QIMR Berghofer Institute, Brisbane, Australia
| | - Bjorn Burgher
- QIMR Berghofer Institute, Brisbane, Australia
- Metro North Mental Health Service, Brisbane, Australia
| | - Omkar Patkar
- School of Psychology and Counselling and IHBI, Faculty of Health, Queensland University of Technology, Brisbane, Australia
- QIMR Berghofer Institute, Brisbane, Australia
| | - Michael Breakspear
- QIMR Berghofer Institute, Brisbane, Australia
- Metro North Mental Health Service, Brisbane, Australia
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Paul Thomas
- Herston Imaging Research Facility, Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation, Lucas Heights, Australia
- National Imaging Facility, Brain and Mind Centre and Faculty of Health Sciences, University of Sydney, Camperdown, Australia
| | - Richard Banati
- Australian Nuclear Science and Technology Organisation, Lucas Heights, Australia
- National Imaging Facility, Brain and Mind Centre and Faculty of Health Sciences, University of Sydney, Camperdown, Australia
| |
Collapse
|
39
|
Albrecht DS, Normandin MD, Shcherbinin S, Wooten DW, Schwarz AJ, Zürcher NR, Barth VN, Guehl NJ, Akeju O, Atassi N, Veronese M, Turkheimer F, Hooker JM, Loggia ML. Pseudoreference Regions for Glial Imaging with 11C-PBR28: Investigation in 2 Clinical Cohorts. J Nucl Med 2018; 59:107-114. [PMID: 28818984 PMCID: PMC5750517 DOI: 10.2967/jnumed.116.178335] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 07/17/2017] [Indexed: 12/12/2022] Open
Abstract
The translocator protein (TSPO) is a commonly used imaging target to investigate neuroinflammation. Although TSPO imaging demonstrates great promise, its signal exhibits substantial interindividual variability, which needs to be accounted for to uncover group effects that are truly reflective of neuroimmune activation. Recent evidence suggests that relative metrics computed using pseudoreference approaches can minimize within-group variability and increase sensitivity to detect physiologically meaningful group differences. Here, we evaluated various ratio approaches for TSPO imaging and compared them with standard kinetic modeling techniques, analyzing 2 different disease cohorts. Patients with chronic low back pain (cLBP) or amyotrophic lateral sclerosis (ALS) and matching healthy controls received 11C-PBR28 PET scans. The occipital cortex, cerebellum and whole brain were first evaluated as candidate pseudoreference regions by testing for the absence of group differences in SUV and distribution volume (VT) estimated with an arterial input function. The SUV from target regions (cLBP study, thalamus; ALS study, precentral gyrus) was normalized with the SUV from candidate pseudoreference regions (i.e., occipital cortex, cerebellum, and whole brain) to obtain SUVRoccip, SUVRcereb, and SUVRWB The sensitivity to detect group differences in target regions was compared using various SUVR approaches, as well as distribution volume ratio (DVR) estimated with (blDVR) or without arterial input function (refDVR), and VT Additional voxelwise SUVR group analyses were performed. We observed no significant group differences in pseudoreference VT or SUV, excepting whole-brain VT, which was higher in cLBP patients than controls. Target VT elevations in patients (P = 0.028 and 0.051 in cLBP and ALS, respectively) were similarly detected by SUVRoccip and SUVRWB, and by refDVR and blDVR (less reliably by SUVRcereb). In voxelwise analyses, SUVRoccip, but not SUVRcereb, identified regional group differences initially observed with SUVRWB, and in additional areas suspected to be affected in the pathology examined. All ratio metrics were highly cross-correlated, but generally were not associated with VT. Although important caveats need to be considered when using relative metrics, ratio analyses appear to be similarly sensitive to detect pathology-related group differences in 11C-PBR28 signal as classic kinetic modeling techniques. The occipital cortex may be a suitable pseudoreference region, at least for the populations evaluated, pending further validation in larger cohorts.
Collapse
Affiliation(s)
- Daniel S Albrecht
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
- Gordon Center for Medical Imaging, NMMI, Radiology Department, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | - Marc D Normandin
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | | | - Dustin W Wooten
- Gordon Center for Medical Imaging, NMMI, Radiology Department, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | | | - Nicole R Zürcher
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | | | - Nicolas J Guehl
- Gordon Center for Medical Imaging, NMMI, Radiology Department, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts
| | - Oluwaseun Akeju
- Department of Anesthesiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nazem Atassi
- Neurological Clinical Research Institute, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Marco L Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| |
Collapse
|
40
|
Vomacka L, Albert NL, Lindner S, Unterrainer M, Mahler C, Brendel M, Ermoschkin L, Gosewisch A, Brunegraf A, Buckley C, Kümpfel T, Rupprecht R, Ziegler S, Kerschensteiner M, Bartenstein P, Böning G. TSPO imaging using the novel PET ligand [ 18F]GE-180: quantification approaches in patients with multiple sclerosis. EJNMMI Res 2017; 7:89. [PMID: 29150726 PMCID: PMC5693838 DOI: 10.1186/s13550-017-0340-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/20/2017] [Indexed: 01/12/2023] Open
Abstract
Background PET ligands targeting the translocator protein (TSPO) represent promising tools to visualise neuroinflammation. Here, we analysed parameters obtained in dynamic and static PET images using the novel TSPO ligand [18F]GE-180 in patients with relapsing remitting multiple sclerosis (RRMS) and an approach for semi-quantitative assessment of this disease in clinical routine. Seventeen dynamic [18F]GE-180 PET scans of RRMS patients were evaluated (90 min). A pseudo-reference region (PRR) was defined after identification of the least disease-affected brain area by voxel-based comparison with six healthy controls (HC) and upon exclusion of voxels suspected of being affected in static 60–90 min p.i. images. Standardised uptake value ratios (SUVR) obtained from static images normalised to PRR were correlated to the distribution volume ratios (DVR) derived from dynamic data with Logan reference tissue model. Results Group comparison with HC revealed white matter and thalamus as most affected regions. Fewest differences were found in grey matter, and normalisation to frontal cortex (FC) yielded the greatest reduction in variability of healthy grey and white matter. Hence, FC corrected for affected voxels was chosen as PRR, leading to time-activity curves of FC which were congruent to HC data (SUV60–90 0.37, U test P = 0.42). SUVR showed a very strong correlation with DVR (Pearson ρ > 0.9). Focal MS lesions exhibited a high SUVR (range, 1.3–3.2). Conclusions This comparison with parameters from dynamic data suggests that SUVR normalised to corrected frontal cortex as PRR is suitable for the quantification of [18F]GE-180 uptake in lesions and different brain regions of RRMS patients. This efficient diagnostic protocol based on static [18F]GE-180 PET scans acquired 60–90 min p.i. allows the semi-quantitative assessment of neuroinflammation in RRMS patients in clinical routine.
Collapse
Affiliation(s)
- Lena Vomacka
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Nathalie Lisa Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Christoph Mahler
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Larissa Ermoschkin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Astrid Gosewisch
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Anika Brunegraf
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | | | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Guido Böning
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
41
|
Di Biase MA, Zalesky A, O'keefe G, Laskaris L, Baune BT, Weickert CS, Olver J, McGorry PD, Amminger GP, Nelson B, Scott AM, Hickie I, Banati R, Turkheimer F, Yaqub M, Everall IP, Pantelis C, Cropley V. PET imaging of putative microglial activation in individuals at ultra-high risk for psychosis, recently diagnosed and chronically ill with schizophrenia. Transl Psychiatry 2017; 7:e1225. [PMID: 28850113 PMCID: PMC5611755 DOI: 10.1038/tp.2017.193] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 06/23/2017] [Indexed: 01/22/2023] Open
Abstract
We examined putative microglial activation as a function of illness course in schizophrenia. Microglial activity was quantified using [11C](R)-(1-[2-chrorophynyl]-N-methyl-N-[1-methylpropyl]-3 isoquinoline carboxamide (11C-(R)-PK11195) positron emission tomography (PET) in: (i) 10 individuals at ultra-high risk (UHR) of psychosis; (ii) 18 patients recently diagnosed with schizophrenia; (iii) 15 patients chronically ill with schizophrenia; and, (iv) 27 age-matched healthy controls. Regional-binding potential (BPND) was calculated using the simplified reference-tissue model with four alternative reference inputs. The UHR, recent-onset and chronic patient groups were compared to age-matched healthy control groups to examine between-group BPND differences in 6 regions: dorsal frontal, orbital frontal, anterior cingulate, medial temporal, thalamus and insula. Correlation analysis tested for BPND associations with gray matter volume, peripheral cytokines and clinical variables. The null hypothesis of equality in BPND between patients (UHR, recent-onset and chronic) and respective healthy control groups (younger and older) was not rejected for any group comparison or region. Across all subjects, BPND was positively correlated to age in the thalamus (r=0.43, P=0.008, false discovery rate). No correlations with regional gray matter, peripheral cytokine levels or clinical symptoms were detected. We therefore found no evidence of microglial activation in groups of individuals at high risk, recently diagnosed or chronically ill with schizophrenia. While the possibility of 11C-(R)-PK11195-binding differences in certain patient subgroups remains, the patient cohorts in our study, who also displayed normal peripheral cytokine profiles, do not substantiate the assumption of microglial activation in schizophrenia as a regular and defining feature, as measured by 11C-(R)-PK11195 BPND.
Collapse
Affiliation(s)
- M A Di Biase
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC Australia
| | - A Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC Australia
- Melbourne School of Engineering, The University of Melbourne, Parkville, VIC Australia
| | - G O'keefe
- Department of Molecular Imaging and Therapy, The University of Melbourne, Heidelberg, VIC Australia
- Department of Medicine, The University of Melbourne, and La Trobe University, Austin Hospital, Heidelberg, VIC, Australia
| | - L Laskaris
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC Australia
| | - B T Baune
- Discipline of Psychiatry, The University of Adelaide, Adelaide, SA, Australia
| | - C S Weickert
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Neuroscience Research Australia, Randwick, NSW, Australia
- Schizophrenia Research Institute, Randwick, NSW, Australia
- School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - J Olver
- Department of Psychiatry, The University of Melbourne, Parkville, VIC Australia
- Department of Molecular Imaging and Therapy, The University of Melbourne, Heidelberg, VIC Australia
- Department of Medicine, The University of Melbourne, and La Trobe University, Austin Hospital, Heidelberg, VIC, Australia
| | - P D McGorry
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - G P Amminger
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
| | - B Nelson
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia
| | - A M Scott
- Department of Molecular Imaging and Therapy, The University of Melbourne, Heidelberg, VIC Australia
- Department of Medicine, The University of Melbourne, and La Trobe University, Austin Hospital, Heidelberg, VIC, Australia
| | - I Hickie
- Brain & Mind Centre, The University of Sydney, Camperdown, NSW, Australia
| | - R Banati
- Medical Radiation Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - F Turkheimer
- Department of Neuroimaging, King’s College London, London, UK
| | - M Yaqub
- VU University Medical Center, Amsterdam, The Netherlands
| | - I P Everall
- Department of Psychiatry, The University of Melbourne, Parkville, VIC Australia
- North Western Mental Health, Melbourne Health, Parkville, VIC, Australia
- Florey Institute for Neurosciences and Mental Health, Parkville, VIC, Australia
- Centre for Neural Engineering, Department of Electrical and Electronic Engineering, The University of Melbourne, Carlton South, VIC, Australia
- Cooperative Research Centre for Mental Health, Carlton, VIC, Australia
| | - C Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC Australia
- North Western Mental Health, Melbourne Health, Parkville, VIC, Australia
- Florey Institute for Neurosciences and Mental Health, Parkville, VIC, Australia
- Centre for Neural Engineering, Department of Electrical and Electronic Engineering, The University of Melbourne, Carlton South, VIC, Australia
- Cooperative Research Centre for Mental Health, Carlton, VIC, Australia
| | - V Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
- Department of Psychiatry, The University of Melbourne, Parkville, VIC Australia
| |
Collapse
|
42
|
Mondelli V, Vernon AC, Turkheimer F, Dazzan P, Pariante CM. Brain microglia in psychiatric disorders. Lancet Psychiatry 2017; 4:563-572. [PMID: 28454915 DOI: 10.1016/s2215-0366(17)30101-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 12/30/2022]
Abstract
The role of immune activation in psychiatric disorders has attracted considerable attention over the past two decades, contributing to the rise of a new era for psychiatry. Microglia, the macrophages of the brain, are progressively becoming the main focus of the research in this field. In this Review, we assess the literature on microglia activation across different psychiatric disorders, including post-mortem and in-vivo studies in humans and experimental studies in animals. Although microglia activation has been noted in all types of psychiatric disorder, no association was seen with specific diagnostic categories. Furthermore, the findings from these studies highlight that not all psychiatric patients have microglial activation. Therefore, the cause of the neuroinflammation in these cohorts and its implications are unclear. We discuss psychosocial stress as one of the main factors determining microglial activation in patients with psychiatric disorders, and explore the relevance of these findings for future treatment strategies.
Collapse
Affiliation(s)
- Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Paola Dazzan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
43
|
Donat CK, Scott G, Gentleman SM, Sastre M. Microglial Activation in Traumatic Brain Injury. Front Aging Neurosci 2017; 9:208. [PMID: 28701948 PMCID: PMC5487478 DOI: 10.3389/fnagi.2017.00208] [Citation(s) in RCA: 282] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/12/2017] [Indexed: 12/15/2022] Open
Abstract
Microglia have a variety of functions in the brain, including synaptic pruning, CNS repair and mediating the immune response against peripheral infection. Microglia rapidly become activated in response to CNS damage. Depending on the nature of the stimulus, microglia can take a number of activation states, which correspond to altered microglia morphology, gene expression and function. It has been reported that early microglia activation following traumatic brain injury (TBI) may contribute to the restoration of homeostasis in the brain. On the other hand, if they remain chronically activated, such cells display a classically activated phenotype, releasing pro-inflammatory molecules, resulting in further tissue damage and contributing potentially to neurodegeneration. However, new evidence suggests that this classification is over-simplistic and the balance of activation states can vary at different points. In this article, we review the role of microglia in TBI, analyzing their distribution, morphology and functional phenotype over time in animal models and in humans. Animal studies have allowed genetic and pharmacological manipulations of microglia activation, in order to define their role. In addition, we describe investigations on the in vivo imaging of microglia using translocator protein (TSPO) PET and autoradiography, showing that microglial activation can occur in regions far remote from sites of focal injuries, in humans and animal models of TBI. Finally, we outline some novel potential therapeutic approaches that prime microglia/macrophages toward the beneficial restorative microglial phenotype after TBI.
Collapse
Affiliation(s)
| | | | | | - Magdalena Sastre
- Division of Brain Sciences, Department of Medicine, Imperial College LondonLondon, United Kingdom
| |
Collapse
|
44
|
Heurling K, Leuzy A, Jonasson M, Frick A, Zimmer ER, Nordberg A, Lubberink M. Quantitative positron emission tomography in brain research. Brain Res 2017; 1670:220-234. [PMID: 28652218 DOI: 10.1016/j.brainres.2017.06.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
The application of positron emission tomography (PET) in brain research has increased substantially during the past 20years, and is still growing. PET provides a unique insight into physiological and pathological processes in vivo. In this article we introduce the fundamentals of PET, and the methods available for acquiring quantitative estimates of the parameters of interest. A short introduction to different areas of application is also given, including basic research of brain function and in neurology, psychiatry, drug receptor occupancy studies, and its application in diagnostics of neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease. Our aim is to inform the unfamiliar reader of the underlying basics and potential applications of PET, hoping to inspire the reader into considering how the technique could be of benefit for his or her own research.
Collapse
Affiliation(s)
- Kerstin Heurling
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Antoine Leuzy
- Department Neurobiology, Care Sciences and Society, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden
| | - My Jonasson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Andreas Frick
- Department of Psychology, Uppsala University, Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Eduardo R Zimmer
- Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Agneta Nordberg
- Department Neurobiology, Care Sciences and Society, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden; Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mark Lubberink
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
45
|
Lagarde J, Sarazin M, Bottlaender M. In vivo PET imaging of neuroinflammation in Alzheimer's disease. J Neural Transm (Vienna) 2017; 125:847-867. [PMID: 28516240 DOI: 10.1007/s00702-017-1731-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests that neuroinflammation contributes to the pathophysiology of many neurodegenerative diseases, especially Alzheimer's disease (AD). Molecular imaging by PET may be a useful tool to assess neuroinflammation in vivo, thus helping to decipher the complex role of inflammatory processes in the pathophysiology of neurodegenerative diseases and providing a potential means of monitoring the effect of new therapeutic approaches. For this objective, the main target of PET studies is the 18 kDa translocator protein (TSPO), as it is overexpressed by activated microglia. In the present review, we describe the most widely used PET tracers targeting the TSPO, the methodological issues in tracer quantification and summarize the results obtained by TSPO PET imaging in AD, as well as in neurodegenerative disorders associated with AD, in psychiatric disorders and ageing. We also briefly describe alternative PET targets and imaging modalities to study neuroinflammation. Lastly, we question the meaning of PET imaging data in the context of a highly complex and multifaceted role of neuroinflammation in neurodegenerative diseases. This overview leads to the conclusion that PET imaging of neuroinflammation is a promising way of deciphering the enigma of the pathophysiology of AD and of monitoring the effect of new therapies.
Collapse
Affiliation(s)
- Julien Lagarde
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Marie Sarazin
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Michel Bottlaender
- UNIACT, NeuroSpin, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91191, Gif-sur-Yvette, France. .,Laboratoire Imagerie Moléculaire in Vivo, UMR 1023, Service Hospitalier Frédéric Joliot, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91400, Orsay, France.
| |
Collapse
|
46
|
Reduction of PK11195 uptake observed in multiple sclerosis lesions after natalizumab initiation. Mult Scler Relat Disord 2017. [PMID: 28641769 DOI: 10.1016/j.msard.2017.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The objective of this study is to longitudinally analyze the uptake of [11C]PK11195-PET in multiple sclerosis patients after 3 and 6 months of natalizumab treatment. METHODS Eighteen MS patients, starting treatment with monocloncal anti-VLA-4, were enrolled in a longitudinal PK-PET study. PK uptake was quantified by volume of distribution (VT) calculation using image-derived input function at baseline, 3 and 6 months. Pharmacokinetic quantification was done using a segmented MRI, and selected areas included white matter, gadolinium enhancing lesions, non-enhancing lesions, cortical grey matter and thalamus. VTs of lesions were calculated in reference to each patient's white matter (VT ratio=VTr), to consider physiologic variability. RESULTS Test-retest variability was stable for healthy control (HC). Quantification of PK uptake was completed in 18 patients, and baseline uptake was compared to 6-month uptake. After the start of natalizumab VTr significantly decreased in 13 individual enhancing lesions present within 5 patients (p=0.001). Moreover, VTr of the sum of non-enhancing lesions showed a moderate decrease (p=0.03). No longitudinal changes were detected in normal appearing white matter, the thalamus and cortical grey matter. CONCLUSION A reduction in PK11195 uptake was observed in both enhancing and chronic lesions after the start of natalizumab. PK11195 PET can be used as tool to assess the longitudinal change in MS lesions.
Collapse
|
47
|
Schain M, Kreisl WC. Neuroinflammation in Neurodegenerative Disorders—a Review. Curr Neurol Neurosci Rep 2017; 17:25. [DOI: 10.1007/s11910-017-0733-2] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
Kalk NJ, Guo Q, Owen D, Cherian R, Erritzoe D, Gilmour A, Ribeiro AS, McGonigle J, Waldman A, Matthews P, Cavanagh J, McInnes I, Dar K, Gunn R, Rabiner EA, Lingford-Hughes AR. Decreased hippocampal translocator protein (18 kDa) expression in alcohol dependence: a [ 11C]PBR28 PET study. Transl Psychiatry 2017; 7:e996. [PMID: 28072413 PMCID: PMC5545729 DOI: 10.1038/tp.2016.264] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 11/02/2016] [Accepted: 11/13/2016] [Indexed: 01/05/2023] Open
Abstract
Repeated withdrawal from alcohol is clinically associated with progressive cognitive impairment. Microglial activation occurring during pre-clinical models of alcohol withdrawal is associated with learning deficits. We investigated whether there was microglial activation in recently detoxified alcohol-dependent patients (ADP), using [11C]PBR28 positron emission tomography (PET), selective for the 18kDa translocator protein (TSPO) highly expressed in activated microglia and astrocytes. We investigated the relationship between microglial activation and cognitive performance. Twenty healthy control (HC) subjects (45±13; M:F 14:6) and nine ADP (45±6, M:F 9:0) were evaluated. Dynamic PET data were acquired for 90 min following an injection of 331±15 MBq [11C]PBR28. Regional volumes of distribution (VT) for regions of interest (ROIs) identified a priori were estimated using a two-tissue compartmental model with metabolite-corrected arterial plasma input function. ADP had an ~20% lower [11C]PBR28 VT, in the hippocampus (F(1,24) 5.694; P=0.025), but no difference in VT in other ROIs. Hippocampal [11C]PBR28 VT was positively correlated with verbal memory performance in a combined group of HC and ADP (r=0.720, P<0.001), an effect seen in HC alone (r=0.738; P=0.001) but not in ADP. We did not find evidence for increased microglial activation in ADP, as seen pre-clinically. Instead, our findings suggest lower glial density or an altered activation state with lower TSPO expression. The correlation between verbal memory and [11C]PBR28 VT, raises the possibility that abnormalities of glial function may contribute to cognitive impairment in ADP.
Collapse
Affiliation(s)
- N J Kalk
- National Addictions Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK,National Addictions Centre, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 4 Windsor Walk, London SE5 8BB, UK. E-mail:
| | - Q Guo
- Neuroimaging Department, Kings College London, London, UK,Centre for Neuropsychopharmacology, Imperial College London, London, UK
| | - D Owen
- Division of Brain Sciences, Imperial College London, London, UK
| | - R Cherian
- West London Mental Health NHS Trust, London, UK
| | - D Erritzoe
- Centre for Neuropsychopharmacology, Imperial College London, London, UK
| | - A Gilmour
- Centre for Infection, Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - A S Ribeiro
- Centre for Neuropsychopharmacology, Imperial College London, London, UK
| | - J McGonigle
- Centre for Neuropsychopharmacology, Imperial College London, London, UK
| | - A Waldman
- Division of Brain Sciences, Imperial College London, London, UK
| | - P Matthews
- Division of Brain Sciences, Imperial College London, London, UK
| | - J Cavanagh
- Institute of Health and Well-being, University of Glasgow, Glasgow, UK
| | - I McInnes
- Centre for Infection, Inflammation and Immunity, University of Glasgow, Glasgow, UK
| | - K Dar
- Central and North West London NHS Trust, London, UK
| | - R Gunn
- Imanova Limited, London, UK
| | | | | |
Collapse
|
49
|
Scott G, Mahmud M, Owen DR, Johnson MR. Microglial positron emission tomography (PET) imaging in epilepsy: Applications, opportunities and pitfalls. Seizure 2017; 44:42-47. [DOI: 10.1016/j.seizure.2016.10.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022] Open
|
50
|
Felger JC, Treadway MT. Inflammation Effects on Motivation and Motor Activity: Role of Dopamine. Neuropsychopharmacology 2017; 42:216-241. [PMID: 27480574 PMCID: PMC5143486 DOI: 10.1038/npp.2016.143] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/13/2016] [Accepted: 07/27/2016] [Indexed: 01/18/2023]
Abstract
Motivational and motor deficits are common in patients with depression and other psychiatric disorders, and are related to symptoms of anhedonia and motor retardation. These deficits in motivation and motor function are associated with alterations in corticostriatal neurocircuitry, which may reflect abnormalities in mesolimbic and mesostriatal dopamine (DA). One pathophysiologic pathway that may drive changes in DAergic corticostriatal circuitry is inflammation. Biomarkers of inflammation such as inflammatory cytokines and acute-phase proteins are reliably elevated in a significant proportion of psychiatric patients. A variety of inflammatory stimuli have been found to preferentially target basal ganglia function to lead to impaired motivation and motor activity. Findings have included inflammation-associated reductions in ventral striatal neural responses to reward anticipation, decreased DA and DA metabolites in cerebrospinal fluid, and decreased availability, and release of striatal DA, all of which correlated with symptoms of reduced motivation and/or motor retardation. Importantly, inflammation-associated symptoms are often difficult to treat, and evidence suggests that inflammation may decrease DA synthesis and availability, thus circumventing the efficacy of standard pharmacotherapies. This review will highlight the impact of administration of inflammatory stimuli on the brain in relation to motivation and motor function. Recent data demonstrating similar relationships between increased inflammation and altered DAergic corticostriatal circuitry and behavior in patients with major depressive disorder will also be presented. Finally, we will discuss the mechanisms by which inflammation affects DA neurotransmission and relevance to novel therapeutic strategies to treat reduced motivation and motor symptoms in patients with high inflammation.
Collapse
Affiliation(s)
- Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Michael T Treadway
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Psychology, Emory University, Atlanta, GA, USA
| |
Collapse
|