Elsevier

The Lancet Psychiatry

Volume 7, Issue 12, December 2020, Pages 1064-1074
The Lancet Psychiatry

Review
Neuroinflammation in psychiatric disorders: PET imaging and promising new targets

https://doi.org/10.1016/S2215-0366(20)30255-8Get rights and content

Summary

Neuroinflammation is a multifaceted physiological and pathophysiological response of the brain to injury and disease. Given imaging findings of 18 kDa translocator protein (TSPO) and the development of radioligands for other inflammatory targets, PET imaging of neuroinflammation is at a particularly promising stage. This Review critically evaluates PET imaging results of inflammation in psychiatric disorders, including major depressive disorder, schizophrenia and psychosis disorders, substance use, and obsessive-compulsive disorder. We also consider promising new targets that can be measured in the brain, such as monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, colony stimulating factor 1 receptor, and the purinergic P2X7 receptor. Thus far, the most compelling TSPO imaging results have arguably been found in major depressive disorder, for which consistent increases have been observed, and in schizophrenia and psychosis, for which patients show reduced TSPO levels. This pattern highlights the importance of validating brain biomarkers of neuroinflammation for each condition separately before moving on to patient stratification and treatment monitoring trials.

Introduction

PET imaging of neuroinflammation was applied first in neurological disorders and later in psychiatric disorders. In this context, neurological disorders, in which inflammation is an established pathological contributor, are ideal positive controls for assessing whether a new biomarker probe works in the human brain. For example, testing 18 kDa translocator protein (TSPO) as a biomarker of neuroinflammation in neurological disorders yielded promising results for Alzheimer's disease, which justified the extension of this probe to psychiatric disorders. Nevertheless, such an extension is associated with many hurdles.

Psychiatric disorders (eg, major depressive disorder and schizophrenia) are more heterogeneous than most neurological illnesses; that is, biological measurements of pathologies sampled from patients with specific diagnoses never completely separate from those of healthy controls and, because symptoms overlap across diagnoses, phenotypes are often shared. Moreover, large variability exists in the course of illness, in response to treatments, and, quite importantly, in the heterogeneous group of associated genetic polymorphisms. These factors probably explain why, to date, no biomarker has been found to fully separate individual patients with specific psychiatric diagnoses from healthy controls. In order to detect a significant difference in the mean of a phenotype measure—in this case, neuroinflammation—in moderately sized samples, the phenotype should be substantially different and reasonably common in the entire population with the disease. One advantage for such markers of more moderate effect sizes is that the prevalence of associated pathology is sufficient for these markers to stratify patients—in this case, as having high or low neuroinflammation—for subsequent studies or treatments. For instance, elevated TSPO binding has been observed in six of seven studies of unmedicated patients with major depressive disorder. By implication, anti-inflammatory treatments in patients with this disorder should preferentially improve symptoms in those patients with PET measures of inflammation at baseline. As will be described in this Review, a study found that elevated TSPO in unmedicated patients with major depressive disorder predicted response to celecoxib, a non-steroidal anti-inflammatory drug and selective inhibitor of cyclooxygenase (COX)-2.1

Regardless of the advantages or disadvantages of TSPO as a biomarker, additional biomarkers of neuroinflammation are needed to reflect the wide range of proinflammatory and anti-inflammatory responses that occur in the brain. This Review will critically evaluate PET imaging results of inflammation in psychiatric disorders, including major depressive disorder, schizophrenia and psychosis, and obsessive-compulsive disorder, as well as in studies of substance use. These illnesses were chosen because they are common major diseases in which quantitative studies have been completed with reasonably high-quality radioligands. The Review will also assess several promising novel PET radioligands of neuroinflammation, including those for COX-1, COX-2, monoamine oxidase B (MAO-B), colony stimulating factor 1 receptor (CSF1R), and the purinergic P2X7 receptor (P2X7R; table 1).

Section snippets

PET imaging of a biomarker

The most common clinical use of PET is for oncology, in which many tumours and their metastases have a high glycolytic rate, identified by uptake and trapping of a radiolabelled analogue of glucose, such as [18F]fluorodeoxyglucose (FDG). Because the underlying process (ie, energy metabolism) has numerous determinants, increased uptake might not have a single interpretation. For example, increased FDG uptake can reflect brain tumours and surrounding inflammation, making it difficult to establish

TSPO: the most commonly imaged biomarker of neuroinflammation

As is the case with many glial cell markers, the interpretation of TSPO is complex and discussed in more detail in a companion article in The Lancet Neurology.2 In rodents, TSPO binding is strongly related to the magnitude of expression of TSPO in activated microglia in studies of lipopolysaccharide administration, toxin, and stroke.9, 10 However, TSPO is expressed in both activated microglia and astroglia in human post-mortem studies of neuropsychiatric illnesses such as Alzheimer's disease,

Major depressive disorder

Studies of peripheral inflammatory biomarkers, such as C-reactive protein (CRP), tumour necrosis factor α (TNFα), and interleukin-6 (IL-6), have typically reported either increases or no change in individuals with major depressive disorder. Meta-analyses of cerebrospinal fluid measures have shown increased concentrations of IL-6 and TNFα in major depressive disorder.28, 29 Given the evidence of peripheral inflammation in major depressive disorder, the question remained whether central

MAO-B

MAO-B, an enzyme comprising 520 amino acids, is mainly located on the outer mitochondrial membranes within astrocytes and serotonin-releasing neurons. During astrogliosis, increased MAO-B expression is associated with similarly greater expression of the astrogliosis marker glial fibrillary acidic protein (GFAP), as seen in neuropsychiatric illnesses such as Alzheimer's disease, amyotrophic lateral sclerosis, multisystem atrophy, and progressive supranuclear palsy.60, 61, 62 MAO-B is an

Conclusion

PET studies of neuroinflammation in neurological disorders have the advantage that distinct disease subtypes can be identified by neuropathology and sometimes by genetic causality (eg, Alzheimer's disease, sporadic frontotemporal dementia, progranulin-caused frontotemporal dementia). By contrast, psychiatric disorders are more heterogeneous, have no consistent neuropathology, and yield wider variability of most biomarkers, including PET measurements of neuroinflammation.

Despite these

Search strategy and selection criteria

We searched PubMed for papers published between Jan 1, 2005, and Feb 29, 2020, with combinations of the following search terms: “PET”, “TSPO”, “translocator protein”, “peripheral benzodiazepine receptor”, “inflammation PET”, “microglia PET”, “depression”, “schizophrenia”, “psychosis”, “substance use”, “obsessive-compulsive disorder”, “MAO-B”, “cyclooxygenase”, “COX”, “CSF1R”, and “P2X7R”. We applied no language restrictions. We generated the final reference list on the basis of relevance to the

References (108)

  • P Plavén-Sigray et al.

    Positron emission tomography studies of the glial cell marker translocator protein in patients with psychosis: a meta-analysis using individual participant data

    Biol Psychiatry

    (2018)
  • J Saura et al.

    Increased monoamine oxidase B activity in plaque-associated astrocytes of Alzheimer brains revealed by quantitative enzyme radioautography

    Neuroscience

    (1994)
  • J Saura et al.

    Molecular neuroanatomy of human monoamine oxidases A and B revealed by quantitative enzyme radioautography and in situ hybridization histochemistry

    Neuroscience

    (1996)
  • T Herraiz et al.

    Human monoamine oxidase is inhibited by tobacco smoke: beta-carboline alkaloids act as potent and reversible inhibitors

    Biochem Biophys Res Commun

    (2005)
  • SH Choi et al.

    The distinct roles of cyclooxygenase-1 and -2 in neuroinflammation: implications for translational research

    Trends Pharmacol Sci

    (2009)
  • A Ohnishi et al.

    Exploratory human PET study of the effectiveness of (11)C-ketoprofen methyl ester, a potential biomarker of neuroinflammatory processes in Alzheimer's disease

    Nucl Med Biol

    (2016)
  • B Ji et al.

    Assessment of radioligands for PET imaging of cyclooxygenase-2 in an ischemic neuronal injury model

    Brain Res

    (2013)
  • JSD Kumar et al.

    In vivo evaluation of [11C]TMI, a COX-2 selective PET tracer, in baboons

    Bioorg Med Chem Lett

    (2018)
  • H Akiyama et al.

    Expression of the receptor for macrophage colony stimulating factor by brain microglia and its upregulation in brains of patients with Alzheimer's disease and amyotrophic lateral sclerosis

    Brain Res

    (1994)
  • N Rodriguez-Alvarez et al.

    Effects of P2X7 receptor antagonists on hypoxia-induced neonatal seizures in mice

    Neuropharmacology

    (2017)
  • LK Parvathenani et al.

    P2X7 mediates superoxide production in primary microglia and is up-regulated in a transgenic mouse model of Alzheimer's disease

    J Biol Chem

    (2003)
  • S Moriguchi et al.

    Monoamine oxidase B total distribution volume in the prefrontal cortex of major depressive disorder: an [11C]SL25.1188 positron emission tomography study

    JAMA Psychiatry

    (2019)
  • JK Mallajosyula et al.

    MAO-B elevation in mouse brain astrocytes results in Parkinson's pathology

    PLoS One

    (2008)
  • MJ Kim et al.

    First-in-human evaluation of [11C]PS13, a novel PET radioligand, to quantify cyclooxygenase-1 in the brain

    Eur J Nucl Med Mol Imaging

    (2020)
  • S Shrestha et al.

    PET measurement of cyclooxygenase-2 using a novel radioligand: upregulation in primate neuroinflammation and first-in-human study

    J Neuroinflammation

    (2020)
  • AG Horti et al.

    PET imaging of microglia by targeting macrophage colony-stimulating factor 1 receptor (CSF1R)

    Proc Natl Acad Sci USA

    (2019)
  • MHJ Hagens et al.

    The P2X7 receptor tracer [11C]SMW139 as an in vivo marker of neuroinflammation in multiple sclerosis: a first-in man study

    Eur J Nucl Med Mol Imaging

    (2020)
  • RB Banati et al.

    PK (‘peripheral benzodiazepine’)—binding sites in the CNS indicate early and discrete brain lesions: microautoradiographic detection of [3H]PK11195 binding to activated microglia

    J Neurocytol

    (1997)
  • A Martín et al.

    Evaluation of the PBR/TSPO radioligand [(18)F]DPA-714 in a rat model of focal cerebral ischemia

    J Cereb Blood Flow Metab

    (2010)
  • E Nutma et al.

    A quantitative neuropathological assessment of translocator protein expression in multiple sclerosis

    Brain

    (2019)
  • AM Barron et al.

    Ligand for translocator protein reverses pathology in a mouse model of Alzheimer's disease

    J Neurosci

    (2013)
  • E Simon-O'Brien et al.

    Etifoxine improves sensorimotor deficits and reduces glial activation, neuronal degeneration, and neuroinflammation in a rat model of traumatic brain injury

    J Neuroinflammation

    (2016)
  • HD Li et al.

    A translocator protein 18 kDa agonist protects against cerebral ischemia/reperfusion injury

    J Neuroinflammation

    (2017)
  • M Cosenza-Nashat et al.

    Expression of the translocator protein of 18 kDa by microglia, macrophages and astrocytes based on immunohistochemical localization in abnormal human brain

    Neuropathol Appl Neurobiol

    (2009)
  • S Venneti et al.

    The positron emission tomography ligand DAA1106 binds with high affinity to activated microglia in human neurological disorders

    J Neuropathol Exp Neurol

    (2008)
  • C Betlazar et al.

    Cellular sources and regional variations in the expression of the neuroinflammatory marker translocator protein (TSPO) in the normal brain

    Int J Mol Sci

    (2018)
  • R Rupprecht et al.

    Translocator protein (18 kDa) (TSPO) as a therapeutic target for neurological and psychiatric disorders

    Nat Rev Drug Discov

    (2010)
  • VM Milenkovic et al.

    CRISPR-Cas9 mediated TSPO gene knockout alters respiration and cellular metabolism in human primary microglia cells

    Int J Mol Sci

    (2019)
  • DR Owen et al.

    Pro-inflammatory activation of primary microglia and macrophages increases 18 kDa translocator protein expression in rodents but not humans

    J Cereb Blood Flow Metab

    (2017)
  • F Chauveau et al.

    Nuclear imaging of neuroinflammation: a comprehensive review of [11C]PK11195 challengers

    Eur J Nucl Med Mol Imaging

    (2008)
  • M Kobayashi et al.

    11C-DPA-713 has much greater specific binding to translocator protein 18 kDa (TSPO) in human brain than 11C-(R)-PK11195

    J Cereb Blood Flow Metab

    (2018)
  • DR Owen et al.

    An 18-kDa translocator protein (TSPO) polymorphism explains differences in binding affinity of the PET radioligand PBR28

    J Cereb Blood Flow Metab

    (2012)
  • DR Owen et al.

    Two binding sites for [3H]PBR28 in human brain: implications for TSPO PET imaging of neuroinflammation

    J Cereb Blood Flow Metab

    (2010)
  • M Fujita et al.

    Comparison of four 11C-labeled PET ligands to quantify translocator protein 18 kDa (TSPO) in human brain: (R)-PK11195, PBR28, DPA-713, and ER176-based on recent publications that measured specific-to-non-displaceable ratios

    EJNMMI Res

    (2017)
  • MA Nettis et al.

    PET imaging shows no changes in TSPO brain density after IFN-α immune challenge in healthy human volunteers

    Transl Psychiatry

    (2020)
  • EM Richards et al.

    PET radioligand binding to translocator protein (TSPO) is increased in unmedicated depressed subjects

    EJNMMI Res

    (2018)
  • E Setiawan et al.

    Role of translocator protein density, a marker of neuroinflammation, in the brain during major depressive episodes

    JAMA Psychiatry

    (2015)
  • TR Marques et al.

    Neuroinflammation in schizophrenia: meta-analysis of in vivo microglial imaging studies

    Psychol Med

    (2019)
  • S Attwells et al.

    Replicating predictive serum correlates of greater translocator protein distribution volume in brain

    Neuropsychopharmacology

    (2020)
  • JM Coughlin et al.

    In vivo markers of inflammatory response in recent-onset schizophrenia: a combined study using [(11)C]DPA-713 PET and analysis of CSF and plasma

    Transl Psychiatry

    (2016)
  • Cited by (140)

    • Reviewing the neurobiology of electroconvulsive therapy on a micro- meso- and macro-level

      2023, Progress in Neuro-Psychopharmacology and Biological Psychiatry
    View all citing articles on Scopus
    View full text