Skip to main content

Main menu

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Journal of Nuclear Medicine
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart
Journal of Nuclear Medicine

Advanced Search

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • View or Listen to JNM Podcast
  • Visit JNM on Facebook
  • Join JNM on LinkedIn
  • Follow JNM on Twitter
  • Subscribe to our RSS feeds
Research ArticleClinical Investigation

Imaging Neuroinflammation in Neurodegenerative Disorders

Joseph C. Masdeu, Belen Pascual and Masahiro Fujita
Journal of Nuclear Medicine June 2022, 63 (Supplement 1) 45S-52S; DOI: https://doi.org/10.2967/jnumed.121.263200
Joseph C. Masdeu
1Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Belen Pascual
1Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Masahiro Fujita
1Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas; and
2PET Core, Houston Methodist Research Institute, Weill Cornell Medicine, Houston, Texas
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • PDF
Loading

Abstract

Neuroinflammation plays a major role in the etiopathology of neurodegenerative diseases, including Alzheimer and Parkinson diseases, frontotemporal lobar degeneration, and amyotrophic lateral sclerosis. In vivo monitoring of neuroinflammation using PET is critical to understand this process, and data are accumulating in this regard, thus a review is useful. From PubMed, we retrieved publications using any of the available PET tracers to image neuroinflammation in humans as well as selected articles dealing with experimental animal models or the chemistry of currently used or potential radiotracers. We reviewed 280 articles. The most common PET neuroinflammation target, translocator protein (TSPO), has limitations, lacking cellular specificity and the ability to separate neuroprotective from neurotoxic inflammation. However, TSPO PET is useful to define the amount and location of inflammation in the brain of people with neurodegenerative disorders. We describe the characteristics of TSPO and other potential PET neuroinflammation targets and PET tracers available or in development. Despite target and tracer limitations, in recent years there has been a sharp increase in the number of reports of neuroinflammation PET in humans. The most studied has been Alzheimer disease, in which neuroinflammation seems initially neuroprotective and neurotoxic later in the progression of the disease. We describe the findings in all the major neurodegenerative disorders. Neuroinflammation PET is an indispensable tool to understand the process of neurodegeneration, particularly in humans, as well as to validate target engagement in therapeutic clinical trials.

  • molecular imaging
  • neurology
  • PET
  • Alzheimer’s disease
  • neurodegeneration
  • neuroinflammation
  • positron emission tomography
  • TSPO

Neuroinflammation is being increasingly recognized as a key component of the etiopathology of neurodegenerative diseases, spanning from Alzheimer disease (AD) through Parkinson disease (PD), frontotemporal lobar degeneration (FTD) and amyotrophic lateral sclerosis (ALS) (1,2). However, while PET biomarkers for some of these disorders, such as β-amyloid and tau tracers for AD, are widely used in both clinical and research work, the use of neuroinflammation tracers remains restricted to a small number of research centers. This limited use can be explained by the lack of cellular specificity of the currently used neuroinflammation target, the translocator protein 18 kDa (TSPO); its ubiquity in the brain, which precludes uptake quantification by comparing a brain region harboring the target with a region devoid of it (3); and the low affinity of the original neuroinflammation PET tracers. However, tracer development, improved quantification (3,4), and the increasing awareness of the importance of neuroinflammation in neurodegenerative disorders have resulted in a sharp increase in the number of publications. For instance, on clinical AD, 4 times as many articles were published between 2021 (the date of this review) and 2013 than between 2013 and 1995 (the date of the first publication). We hope that this review will encourage additional work in this important field. After a description of the characteristics of available PET tracers and their application to the study of various neurodegenerative disorders, we discuss the characteristics and applicability of potentially new PET inflammation biomarkers.

TSPO PET: CURRENT TARGET TO IMAGE NEUROINFLAMMATION

What Is TSPO?

TSPO is an ion-channel-type receptor located on the outer membrane of mitochondria. Initially reported as transporting cholesterol, porphyrin, and Ca2+ (5), its function is still under investigation and likely involved in steroidogenesis, apoptosis, mitochondrial respiration, and processing reactive oxygen species (6). TSPO is not selectively expressed by microglia but also by astrocytes, vascular endothelial cells, some neurons, immune cells, and some tumor cells. Thus, frequent statements in papers that TSPO PET measured an increase in “activated” microglia oversimplify the matter (7). Furthermore, some (8,9), but not all (10,11), postmortem studies of AD brains have failed to find significant increases in TSPO, fueling skepticism on the use of TSPO PET to image neuroinflammation in neurodegenerative disorders. But even the negative studies, which contained small samples, showed in some patients much higher TSPO levels in areas typically affected in AD, such as the frontal lobe, than in the cerebellum, little involved in this disorder (8). In other diseases as well, TSPO is increased specifically in areas characteristically affected by the pathology, such as the motor cortex in ALS (12). Thus, despite its limitations, TSPO PET is useful to measure neuroinflammation in neurodegeneration.

PET Ligands to Image TSPO

Since the 1980s initial PET studies on neuroinflammation used 11C-PK11195. However, later, 11C-PK11195 was found to have low specific binding (13,14). Around 2000, a new series of TSPO ligands with different chemical structures was published (15), and subsequently an original chemical, 11C-DAA1106, and analogs such as 11C-PBR28 have been used in PET studies (16,17). PET ligands with different structure have also been developed including 11C-DPA-713 (18). 18F-GE180, useful for rodent studies, does not penetrate the intact blood–brain barrier in humans (19), with K1 (rate constant for transfer from arterial plasma to tissue, according to Innis et al. (20)) of only 0.01 mL/g/min (21). 18F-GE180 is thus questionable for human studies of neuroinflammation. Radioligands developed after PK11195 are called second-generation PET TSPO ligands. Animal PET studies showed that most of the second-generation ligands have much greater specific binding to TSPO than PK11195 (13).

A limitation of nearly all second-generation TSPO ligands is that binding is affected by the single nucleotide polymorphism (SNP) rs6971 (22). Depending on the SNP they carry, subjects can be high-, mixed-, or low-affinity binders. In low-affinity binders, the low signal precludes useful PET using most ligands. The effect of the SNP needs to be included in the analysis of PET data. Therefore, a potential improvement over the second-generation ligands is to minimize the influence of the SNP.

To select a PET ligand to study changes in TSPO, several factors need to be considered including the equilibrium ratio of specific-to-nondisplaceable binding, BPND; influences of radiometabolites in the quantification; and feasibility to measure binding in low-affinity binders. Greater BPND provides PET signals more sensitive to the changes in specific binding. BPND is typically measured in animals by comparing scans under baseline and near complete binding blockade. However, because both in vitro and PET studies show large species differences in the density of TSPO (23–25) it is necessary to measure BPND of each ligand in humans. The Lassen plot allows measurement of BPND with only partial blockade of the binding, which is unlikely to cause pharmacologic effects (26). Thus, this method has been used in humans for 4 11C-labeled TSPO ligands: 11C-R-PK11195, 11C-PBR28, 11C-DPA-713, and 11C-ER176 (14). Feasibility of measuring TSPO in low-affinity binders and possible influences of radiometabolites have also been investigated for these 4 ligands (14). On the basis of these most comprehensive investigations in humans so far, 11C-ER176 is the current choice to study TSPO. 11C-ER176 has an adequate BPND even in low-affinity binders and shows the least influence from radiometabolites.

In neurodegeneration, the permeability of the blood–brain barrier may be altered (27). In theory, because greater permeability increases the movement of chemical compounds both in (K1) and out (k2) of the brain, changes in permeability are not expected to influence equilibrium parameters such as BPND and total distribution volume (VT).

NEUROINFLAMMATION PET IN NEURODEGENERATIVE DISORDERS

Alzheimer Disease (AD)

The discovery of AD risk genes involved in inflammation signaling (28) has emphasized the critical role of neuroinflammation for the onset and progression of AD. In AD transgenic mouse models, innate immune microglia are initial responders to neuronal release of amyloid-β (Aβ), activating microglial pattern recognition toll-like receptors, and intracellular NLRP3 inflammasomes, thereby inducing tau hyperphosphorylation and aggregation (1). Subsequent release of truncated phosphorylated tau also enhances immune cell activation, promoting the release of inflammatory mediators and a self-propagating cascade of synaptic dysfunction, neuronal injury, and cell death (1). Current understanding of the role of inflammation in AD stems largely from work with rodents. However, interspecies differences with humans may be large, as shown, for instance, by profiling microglial RNA from frozen AD and control brains (29). In human brain, the proportion of morphologically activated microglia in cortical tissue is associated with β-amyloid, tau-related neuropathology, and the rate of cognitive decline (30). However, neuroinflammation may be neuroprotective or harmful in neurodegeneration (31). It is possible that the predominant role of inflammation may shift at various stages of AD, being neuroprotective at earlier stages and neurotoxic at advanced stages, but this is yet to be proven in human AD. TSPO PET provides a tool to characterize brain inflammation at the various stages of human AD (Fig. 1). A preclinical stage, when the person is still cognitively unimpaired, but brain β-amyloid is already high, may be most amenable to therapy but has been least studied by PET. As the disease worsens, the patient has mild cognitive impairment, already with some degree of memory or language impairment, but not enough to interfere seriously with activities of daily living as it is in the next stage, clinical AD. Clinical AD, still not severe enough to render the patient uncooperative for brain PET, is the stage at which most early TSPO PET studies were performed and will be described first below. Although single photon emission tomography has been used to image TSPO in AD, most groups have used PET. The 2 tracers most often used have been 11C-PK11195, in 28 publications, and 11C-PBR28, in 16 publications. The first article (32) using 11C-PK11195 in AD was published in 1995 and the first ones (33,34) using 11C-PBR28, in 2013. Several studies have used an arterial input function (Supplemental Table 1; supplemental materials are available at http://jnm.snmjournals.org), but in AD the cerebellum is spared until the disease is very advanced and may serve as a pseudoreference region, that is, a region with TSPO receptors but no change in patients (4).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Translocator protein 18 kDa (TSPO) PET in AD. Inflammation imaging at presymptomatic, MCI, and dementia stages of AD process. In each subject, multimodal imaging is shown, to illustrate spatial correlation of inflammation, in bottom row of every individual scan, with amyloid and tau imaging. CDR = clinical dementia rating scale; HAB = TSPO-high-affinity binder subject; MAB = TSPO-mixed-affinity binder subject; SUVR = standardized uptake value ratio; VT = volume of distribution.

TSPO PET at the Clinical Alzheimer Disease (AD) Stage

At the clinical AD stage, most studies have detected significant neuroinflammation. Brain regions most often affected have been the medial temporal region, including the entorhinal cortex; temporoparietal association cortex, including precuneus; and cingulate cortex (35–37). The localization of inflammation matched the clinical syndrome (37) and correlated negatively with metabolism measured with 18F-FDG PET (35–37). Furthermore, the degree of TSPO uptake correlated with worse cognition (33,38,39), better than the degree of amyloid deposition and, together with the degree of tau deposition, predicted the degree of cognitive worsening over 3 y (40). Inflammation correlated with white matter changes (41) and impaired large-scale functional connectivity on MRI (42).

TSPO PET at the Mild Cognitive Impairment (MCI) Stage

Abnormal TSPO density (43) has not been found as consistently in MCI as in AD, with some studies reporting no difference with controls (33). Most cross-sectional studies reported greater neuroinflammation in the AD than in the MCI stage (36,44), although greater inflammation at the MCI stage has also been reported (45). To this variability may contribute the behavior of neuroinflammation at the MCI stage. A biphasic inflammation peak has been postulated by following MCI patients longitudinally with repeated TSPO PET over 2 y. Some patients with greater initial inflammation had decreased inflammation in the second scan, although their β-amyloid levels continued to rise (31,46). This observation supported the notion that initial inflammation may be neuroprotective (31), a concept supported by slower decline in patients with more initial inflammation at the MCI stage (47) and the inverse correlation between inflammation and neurofilament light levels (48). As at the AD stage, in MCI a negative correlation has been found between regional inflammation and metabolism (49). The relation of TSPO density to a marker of neurodegeneration, cortical atrophy on MRI, is complex. At the AD and MCI stages, inflammation was associated with decreased cortical thickness in the neocortex but not in the hippocampus (50). However, at the early MCI stage, inflammation was associated with increased gray matter volume, including in the hippocampus (51).

TSPO PET at the Presymptomatic Stage

A few studies at the presymptomatic stage, defined by normal cognition but abnormal amyloid brain deposition, have reported variable findings. Although an association of inflammation with amyloid deposition, but not with tau, has been reported in presymptomatic individuals (52), a closer correlation of inflammation with amyloid has been reported at the stage when amyloid PET is still negative, rather than when it becomes positive, although then the absolute level of inflammation is higher (53). This intriguing finding suggests that inflammation may be a very early factor in the neurodegenerative cascade (44), just as shown in experimental animal models.

TSPO PET in Normal Aging

Initial PET studies, when amyloid imaging was not consistently performed, found increased TSPO binding in subcortical structures and, particularly, in the thalamus of cognitively unimpaired older people (54,55). This aging effect may interfere with the interpretation of studies in diseases that affect the basal ganglia, such as corticobasal degeneration or progressive supranuclear palsy unless controls are closely matched by age. Increased inflammation with aging has also been reported in cortical regions (44,52).

TSPO Versus β-Amyloid and Tau Brain Density

To ensure that the cognitive impairment is due to AD, most groups are currently reporting the β-amyloid and tau status of the subjects studied. Thus, the relation of neuroinflammation to the proteins characteristic of AD is being clarified (Fig. 1). The scant studies at the presymptomatic stage are discussed in the previous section. At the MCI and AD stages, the degree and location of neuroinflammation are correlated with both amyloid and tau deposition but reported r values oscillate around 0.35 for both associations (44), reflecting in part that amyloid and tau deposition are only partially colocalized (45). Inflammation may correlate more closely with amyloid at the MCI stage and with tau at the AD stage (45,46).

Dementia with Lewy-Bodies (DLB)

DLB is characterized clinically by progressive cognitive impairment with fluctuating cognition; visual hallucinations; REM sleep behavior disorder, which may precede cognitive decline; and one or more cardinal features of parkinsonism (56). Pathologically, the protein α-synuclein is present in intraneuronal Lewy bodies and neurites spread throughout the cortex, hippocampus, and amygdala (57). About 50% of DLB patients have associated AD pathology (58). This association is more frequent with advancing age and confers a worse prognosis (58). Several studies have reported increased neuroinflammation (59,60) and a negative correlation between inflammation and metabolism (35,61). A positive correlation with tau has also been reported (62). Individuals with glucocerebrosidase mutations are predisposed to DLB (63). They have been reported to have increased neuroinflammation (64).

Parkinson Disease (PD)

Clinically, PD presents with bradykinesia, rigidity, and resting tremor. Pathologically, while there are neuronal loss and Lewy bodies in the substantia nigra, these changes are not widespread in the cortex, hippocampus, and amygdala, as they are in DLB. PET TSPO findings have not been uniform. Increased inflammation in the midbrain or other regions of the brain has been found in some studies (61,65) but not in others (66,67). In a large study, it was reported that multiple-system atrophy, which presents with parkinsonian clinical findings, was associated with increased brain 11C-PBR28 uptake, which PD is not, thus allowing for a ready differentiation of either disease (66).

Frontotemporal Dementia (FTD)

This heterogeneous group of diseases, known also as frontotemporal lobar degeneration, encompasses disorders that begin with behavioral, language, or motor impairment and are associated with tau or TDP-43 deposition in the brain. Neuroinflammation plays a major role in FTD (2) and it has been shown to be increased in the likely location of pathology (68–70), which differs among FTD variants. Thus, inflammation is greatest in the frontal and temporal poles in behavioral variant, premotor cortex in nonfluent primary progressive aphasia, in superomedial convexity in corticobasal degeneration, and temporal lobe in semantic dementia (Fig. 2). Although in semantic dementia the damage begins and is greatest in the temporal tip, inflammation is greatest at the periphery of the affected region, suggesting a major role for inflammation in damage propagation (Fig. 2) (71). In progressive supranuclear palsy, greater neuroinflammation may predict faster worsening (72).

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

TSPO PET in FTD. Inflammation imaging in behavioral variant of FTD and in the nonfluent and semantic variants of primary progressive aphasia (PPA), also categorized as FTD. Amyloid PET was negative in all subjects. In semantic variant PPA, peak of inflammation is at periphery of most affected area, which has atrophy on MRI. HAB = TSPO-high-affinity binder subject; MAB = TSPO-mixed-affinity binder subject; SUVR = standardized uptake value ratio; VT = volume of distribution. (Lower 2 panels are adapted with permission of (71)).

Amyotrophic Lateral Sclerosis (ALS)

Inflammation has been found in the paracentral cortex, which is most affected in ALS (73). Although an abnormal signal was detected with the TSPO tracers 11C-PBR28 and 18F-DPA714 (12), in a small study no signal was detected in ALS using the purine receptor P2 × 7 tracer 11C-JNJ54173717 (74).

Other Neurodegenerative Disorders

Huntington’s Chorea

Genetic testing allows for the determination of the carrier state at the presymptomatic stage of the fully penetrant autosomal dominantly inherited disorder. Some (75) but not others (76) have found increased neuroinflammation in subjects at risk, whereas there is consensus on the presence of neuroinflammation in symptomatic patients (76,77).

Nieman–Pick Disease

In adult Nieman–Pick disease, neuroinflammation was increased in the white matter and correlated with decreased fractional anisotropy (78).

Chronic Traumatic Encephalopathy

Chronic traumatic encephalopathy may be considered to have a neurodegenerative component, as have many other diseases, such as multiple sclerosis. Although these disorders are not covered in this review, some of the studies of neuroinflammation in chronic traumatic encephalopathy are listed in Supplemental Table 1.

LIMITATIONS OF TSPO PET

As a marker of neuroinflammation, TSPO has 2 major limitations: it is not specific for activated microglia and does not differentiate between microglia that protect or harm neurons.

No Selectivity to Cell Type

Although activated microglia are often considered as the main neuroinflammatory cell, astrocytes play critical roles in neuroinflammation. Both microglia and astrocytes respond to Aβ plaques but the response is different, possibly because microglia respond to the chemotactic effects of Aβ whereas astrocytes respond to neuritic damage (79). Astrocytes are heavily involved in the clearance of Aβ (80). However, close interactions between these 2 types of glia may be linked to neurodegenerative pathology. Activated microglia induce neurotoxic reactive astrocytes (81). The interaction of the microglial receptor TREM2 with lipoparticles is integral for the transfer of cholesterol from astrocytes to microglia (82), and mutations in TREM2 increase AD risk (83). All these rich interactions are not identifiable by TSPO PET, which provides a snapshot of the regional density in the brain of both microglia and astrocytes.

No Differentiation Between Beneficial and Detrimental Effects of Inflammation

Neuroinflammation has both protective and damaging effects. Although activation of microglia at early stages facilitates phagocytosis of Aβ plaques and maintains neuronal survival, chronic inflammation becomes skewed toward a proinflammatory pattern, which might be neurotoxic (84). Ongoing inflammation may also facilitate phosphorylation and truncation of tau, causing further damage to neurons (85). TSPO PET studies in both humans (reviewed in the section “TSPO PET at the Mild Cognitive Impairment [MCI] Stage”) and mice suggest that TSPO presents 2 peaks in the AD process: an earlier peak, associated with neuroprotection, and a later peak as the disease worsens, associated with neurotoxicity (31,86,87). The 2 facets of inflammation indicate that simple suppression of inflammation may not help AD. Therefore, novel therapies are being developed to shift microglia from neurotoxic to neuroprotective (88).

NEW INFLAMMATION IMAGING TARGETS TO OVERCOME THE LIMITATIONS OF TSPO PET

To overcome these limitations, new and improved imaging markers are being investigated.

Cell-Type Selective Imaging

Colony-stimulating factor 1 receptor (CSF1R) is predominantly expressed by microglia and macrophages and plays a key role in differentiation and survival of immune cells (89). Inhibitors of CSF1R decrease microglia and prevent loss of neurons and memory (90). A novel ligand, 11C-CPPC, successfully detected an increase in CSF1R in rodent and nonhuman primate after bacterial lipopolysaccharide was administered (91), but it had off-target and low specific binding (92). Another ligand, 11C-GW2580, detected increased microglia in a rodent model of neuroinflammation with greater sensitivity than 11C-CPPC (93).

Monoamine oxidase-B (MAO-B) is a potential PET marker of astrocytes. MAO-B is highly expressed by astrocytes and serotonin (5-HT)-releasing neurons but not by microglia (94). MAO-B is upregulated in reactive astrocytes (95) and correlates with glial fibrillary acidic protein (96). To image MAO-B, L-11C-deprenyl has been used since the 1980s (97). The irreversible nature of tracer uptake makes the PET measurement more sensitive to blood flow than to the enzyme activity, especially in areas with high MAO-B activity (98). To cope with this limitation, a deuterium-substituted PET tracer, L-11C-deprenyl-D2, was developed (97). Still, the specific-to-nondisplaceable ratio of L-11C-deprenyl is only 1.5 in humans (99). Semiquantitative analyses in healthy humans indicated that a new ligand to image MAO-B, 18F-SMBT-1 (100), has a BPND of approximately 6, which is about 4 times greater than that of L-11C-deprenyl (101).

Differentiating Beneficial and Detrimental Effects of Neuroinflammation

Because TSPO PET imaging does not differentiate between beneficial and detrimental effects of neuroinflammation, better PET markers are needed to monitor novel immunomodulatory therapies. For this purpose, potential markers are two adenosine diphosphate (ADP) / adenosine triphosphate (ATP) receptors with markedly different functions, P2X7 and P2Y12. Both P2X7 and P2Y12 are highly expressed by microglia. P2X7 has proinflammatory functions. Increased ATP and ADP in the extracellular space activates P2X7 and leads to proinflammatory cytokines (102). On the contrary, in basic studies, P2Y12 is used as a marker of homeostatic microglia and may protect neurons by regulating somatic microglia-neuron junctions (103). Postmortem studies on AD showed increased P2X7 (104,105) and decreased P2Y12 (106), consistent with greater detrimental than neuroprotective effects of neuroinflammation in moderate-advanced stages of AD. To image P2X7, 18F-JNJ-64413739 (107) and 11C-JNJ54173717 (108) have been successfully used in humans. PET ligands to image P2Y12 are still under development.

OTHER PET MARKERS TO STUDY NEUROINFLAMMATION

In the following, we list a few additional markers of neuroinflammation, for which PET scans have detected specific binding.

Type 2 Cannabinoid Receptor (CB2)

CB2 is involved in immunomodulatory functions and expressed by microglia, astrocytes, and neurons (109). Activation of CB2 shows protective effects against Aβ (110). Several PET ligands are being developed to image CB2 (111). Among these, the most tested is 11C-NE40, which has been used to compare healthy controls and AD patients and showed a decrease in AD (112). Because type 1 cannabinoid receptor (CB1) is highly and widely expressed in brain, high selectivity against CB1 is required to interpret CB2 PET results.

Mitochondrial Function and Reactive Oxygen Species

Because mitochondrial dysfunction has been reported in neurodegenerative disorders such as AD and PD and linked to neuroinflammation (113), there have been attempts to image mitochondrial activity. The most explored imaging target is mitochondrial complex 1 (MC1) imaged with 18F-BCPP-EF (114). MC1 is the first enzyme complex in the electron transfer chain, having a critical role in oxidative phosphorylation in mitochondria. AD patients had reduced 18F-BCPP-EF binding in the medial temporal cortex, which negatively correlated with tau (113). Recent attempts to image reactive oxygen species detected an increase induced by the local injection of bacterial lipopolysaccharide (115).

Phosphodiesterase Type 4B (PDE4B)

Cyclic nucleotide phosphodiesterases are enzymes that hydrolyze the second messengers, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate. Among 11 families of PDE, phosphodiesterase type 4, expressed by neurons and immune cells, is one of the main enzymes hydrolyzing cAMP. Among the 4 subtypes, A, B, C, and D, subtype B (PDE4B) is involved in microglial functions in neuroinflammation (116). Proinflammatory cytokines such as IL-1β and TNF-α increase PDE4B. A PET ligand, 18F-PF-06445974, has been developed to selectively image PDE4B (117).

LINKAGE BETWEEN PERIPHERAL AND CENTRAL INFLAMMATION

In addition to inflammation within the brain, inflammation in peripheral tissue and organs and interactions between peripheral and central inflammation are increasingly being recognized as important in pathogenesis. A pathogen of periodontitis was found in the brain of AD patients (118), and Aβ protects against microbial infection (119). Gut bacteria are involved in the production of α-synuclein, which accumulates in PD and DLB brain. Newly developed total body scanners allow for the gathering of full dynamic data in both brain and peripheral tissues and organs and open new opportunities to explore interactions between peripheral and central inflammation. In the brain, the choroid plexus is the major component of the blood–cerebrospinal fluid barrier, which may be a major player to connect peripheral and central inflammation (120). The function of the choroid plexus in neuroinflammation and neurodegeneration can be studied by brain imaging.

DISCLOSURE

Joseph Masdeu is a consultant and received research funding from Eli Lilly, parent co. of Avid Radiopharmaceuticals, manufacturer of 18F-flortaucipir. No other potential conflict of interest relevant to this article was reported.

  • © 2022 by the Society of Nuclear Medicine and Molecular Imaging.

REFERENCES

  1. 1.↵
    1. Ising C,
    2. Venegas C,
    3. Zhang S,
    4. et al
    . NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669–673.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Bright F,
    2. Werry EL,
    3. Dobson-Stone C,
    4. et al
    . Neuroinflammation in frontotemporal dementia. Nat Rev Neurol. 2019;15:540–555.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Guo Q,
    2. Owen DR,
    3. Rabiner EA,
    4. Turkheimer FE,
    5. Gunn RN
    . A graphical method to compare the in vivo binding potential of PET radioligands in the absence of a reference region: application to [¹¹C]PBR28 and [¹18F]PBR111 for TSPO imaging. J Cereb Blood Flow Metab. 2014;34:1162–1168.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Zanotti-Fregonara P,
    2. Kreisl WC,
    3. Innis RB,
    4. Lyoo CH
    . Automatic extraction of a reference region for the noninvasive quantification of translocator protein in brain using 11C-PBR28. J Nucl Med. 2019;60:978–984.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    1. Papadopoulos V,
    2. Baraldi M,
    3. Guilarte TR,
    4. et al
    . Translocator protein (18kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci. 2006;27:402–409.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Notter T,
    2. Coughlin JM,
    3. Gschwind T,
    4. et al
    . Translational evaluation of translocator protein as a marker of neuroinflammation in schizophrenia. Mol Psychiatry. 2018;23:323–334.
    OpenUrlCrossRef
  7. 7.↵
    1. Notter T,
    2. Schalbetter SM,
    3. Clifton NE,
    4. et al
    . Neuronal activity increases translocator protein (TSPO) levels. Mol Psychiatry. 2021;26:2025–2037.
    OpenUrl
  8. 8.↵
    1. Gui Y,
    2. Marks JD,
    3. Das S,
    4. Hyman BT,
    5. Serrano-Pozo A
    . Characterization of the 18 kDa translocator protein (TSPO) expression in post-mortem normal and Alzheimer’s disease brains. Brain Pathol. 2020;30:151–164.
    OpenUrl
  9. 9.↵
    1. Xu J,
    2. Sun J,
    3. Perrin RJ,
    4. et al
    . Translocator protein in late stage Alzheimer’s disease and Dementia with Lewy bodies brains. Ann Clin Transl Neurol. 2019;6:1423–1434.
    OpenUrl
  10. 10.↵
    1. Metaxas A,
    2. Thygesen C,
    3. Briting SRR,
    4. Landau AM,
    5. Darvesh S,
    6. Finsen B
    . Increased inflammation and unchanged density of synaptic vesicle glycoprotein 2A (SV2A) in the postmortem frontal cortex of Alzheimer’s Disease patients. Front Cell Neurosci. 2019;13:538.
    OpenUrl
  11. 11.↵
    1. Ni R,
    2. Röjdner J,
    3. Voytenko L,
    4. et al
    . In vitro characterization of the regional binding distribution of amyloid PET tracer florbetaben and the glia tracers deprenyl and PK11195 in autopsy Alzheimer’s brain tissue. J Alzheimers Dis. 2021;80:1723–1737.
    OpenUrl
  12. 12.↵
    1. Van Weehaeghe D,
    2. Babu S,
    3. De Vocht J,
    4. et al
    . Moving toward multicenter therapeutic trials in amyotrophic lateral sclerosis: feasibility of data pooling using different translocator protein PET radioligands. J Nucl Med. 2020;61:1621–1627.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Kreisl WC,
    2. Fujita M,
    3. Fujimura Y,
    4. et al
    . Comparison of [11C]-(R)-PK 11195 and [11C]PBR28, two radioligands for translocator protein (18 kDa) in human and monkey: Implications for positron emission tomographic imaging of this inflammation biomarker. Neuroimage. 2010;49:2924–2932.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Fujita M,
    2. Kobayashi M,
    3. Ikawa M,
    4. 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;7:84.
    OpenUrl
  15. 15.↵
    1. Okuyama S,
    2. Chaki S,
    3. Yoshikawa R,
    4. et al
    . Neuropharmacological profile of peripheral benzodiazepine receptor agonists, DAA1097 and DAA1106. Life Sci. 1999;64:1455–1464.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Zhang MR,
    2. Kida T,
    3. Noguchi J,
    4. et al
    . [11C]DAA1106: radiosynthesis and in vivo binding to peripheral benzodiazepine receptors in mouse brain. Nucl Med Biol. 2003;30:513–519.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Briard E,
    2. Zoghbi SS,
    3. Imaizumi M,
    4. et al
    . Synthesis and evaluation in monkey of two sensitive 11C-labeled aryloxyanilide ligands for imaging brain peripheral benzodiazepine receptors in vivo. J Med Chem. 2008;51:17–30.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. James ML,
    2. Fulton RR,
    3. Henderson DJ,
    4. et al
    . Synthesis and in vivo evaluation of a novel peripheral benzodiazepine receptor PET radioligand. Bioorg Med Chem. 2005;13:6188–6194.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Zanotti-Fregonara P,
    2. Pascual B,
    3. Rizzo G,
    4. et al
    . Head-to-head comparison of 11C-PBR28 and 18F-GE180 for quantification of the translocator protein in the human brain. J Nucl Med. 2018;59:1260–1266.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    1. Innis RB,
    2. Cunningham VJ,
    3. Delforge J,
    4. et al
    . Consensus nomenclature for in vivo imaging of reversibly binding radioligands. J Cereb Blood Flow Metab. 2007;27:1533–1539.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Feeney C,
    2. Scott G,
    3. Raffel J,
    4. et al
    . Kinetic analysis of the translocator protein positron emission tomography ligand [18F]GE-180 in the human brain. Eur J Nucl Med Mol Imaging. 2016;43:2201–2210.
    OpenUrl
  22. 22.↵
    1. Owen DR,
    2. Yeo AJ,
    3. Gunn RN,
    4. 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;32:1–5.
    OpenUrlCrossRefPubMed
  23. 23.↵
    1. Cymerman U,
    2. Pazos A,
    3. Palacios JM
    . Evidence for species differences in ‘peripheral’ benzodiazepine receptors: an autoradiographic study. Neurosci Lett. 1986;66:153–158.
    OpenUrlCrossRefPubMed
  24. 24.
    1. Imaizumi M,
    2. Briard E,
    3. Zoghbi SS,
    4. et al
    . Brain and whole-body imaging in nonhuman primates of [11C]PBR28, a promising PET radioligand for peripheral benzodiazepine receptors. Neuroimage. 2008;39:1289–1298.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Fujita M,
    2. Imaizumi M,
    3. Zoghbi SS,
    4. et al
    . Kinetic analysis in healthy humans of a novel positron emission tomography radioligand to image the peripheral benzodiazepine receptor, a potential biomarker for inflammation. Neuroimage. 2008;40:43–52.
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Cunningham VJ,
    2. Rabiner EA,
    3. Slifstein M,
    4. Laruelle M,
    5. Gunn RN
    . Measuring drug occupancy in the absence of a reference region: the Lassen plot re-visited. J Cereb Blood Flow Metab. 2010;30:46–50.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. Montagne A,
    2. Nation DA,
    3. Sagare AP,
    4. et al
    . APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature. 2020;581:71–76.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Schwartzentruber J,
    2. Cooper S,
    3. Liu JZ,
    4. et al
    . Genome-wide meta-analysis, fine-mapping and integrative prioritization implicate new Alzheimer’s disease risk genes. Nat Genet. 2021;53:392–402.
    OpenUrl
  29. 29.↵
    1. Srinivasan K,
    2. Friedman BA,
    3. Etxeberria A,
    4. et al
    . Alzheimer’s patient microglia exhibit enhanced aging and unique transcriptional activation. Cell Rep. 2020;31:107843.
    OpenUrlCrossRef
  30. 30.↵
    1. Felsky D,
    2. Roostaei T,
    3. Nho K,
    4. et al
    . Neuropathological correlates and genetic architecture of microglial activation in elderly human brain. Nat Commun. 2019;10:409.
    OpenUrlCrossRef
  31. 31.↵
    1. Fan Z,
    2. Brooks DJ,
    3. Okello A,
    4. Edison P
    . An early and late peak in microglial activation in Alzheimer’s disease trajectory. Brain. 2017;140:792–803.
    OpenUrl
  32. 32.↵
    1. Groom GN,
    2. Junck L,
    3. Foster NL,
    4. Frey KA,
    5. Kuhl DE
    . PET of peripheral benzodiazepine binding sites in the microgliosis of Alzheimer’s disease. J Nucl Med. 1995;36:2207–2210.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    1. Kreisl WC,
    2. Lyoo CH,
    3. McGwier M,
    4. et al
    ; Biomarkers Consortium PET Radioligand Project Team. In vivo radioligand binding to translocator protein correlates with severity of Alzheimer’s disease. Brain. 2013;136:2228–2238.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Kim S,
    2. Nho K,
    3. Risacher SL,
    4. et al
    . PARP1 gene variation and microglial activity on [11C]PBR28 PET in older adults at risk for Alzheimer’s disease. Multimodal Brain Image Anal. 2013;8159:150–158.
    OpenUrl
  35. 35.↵
    1. Fan Z,
    2. Aman Y,
    3. Ahmed I,
    4. et al
    . Influence of microglial activation on neuronal function in Alzheimer’s and Parkinson’s disease dementia. Alzheimers Dement. 2015;11:608–21.e7.
    OpenUrlPubMed
  36. 36.↵
    1. Kreisl WC,
    2. Lyoo CH,
    3. Liow JS,
    4. et al
    . 11C-PBR28 binding to translocator protein increases with progression of Alzheimer’s disease. Neurobiol Aging. 2016;44:53–61.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Tondo G,
    2. Iaccarino L,
    3. Caminiti SP,
    4. et al
    . The combined effects of microglia activation and brain glucose hypometabolism in early-onset Alzheimer’s disease. Alzheimers Res Ther. 2020;12:50.
    OpenUrl
  38. 38.↵
    1. Lindgren N,
    2. Tuisku J,
    3. Vuoksimaa E,
    4. et al
    . Association of neuroinflammation with episodic memory: a [11C]PBR28 PET study in cognitively discordant twin pairs. Brain Commun. 2020;2:fcaa024.
    OpenUrl
  39. 39.↵
    1. Passamonti L,
    2. Rodríguez PV,
    3. Hong YT,
    4. et al
    . [11C]PK11195 binding in Alzheimer disease and progressive supranuclear palsy. Neurology. 2018;90:e1989–e1996.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Malpetti M,
    2. Kievit RA,
    3. Passamonti L,
    4. et al
    . Microglial activation and tau burden predict cognitive decline in Alzheimer’s disease. Brain. 2020;143:1588–1602.
    OpenUrlPubMed
  41. 41.↵
    1. Low A,
    2. Mak E,
    3. Malpetti M,
    4. et al
    . In vivo neuroinflammation and cerebral small vessel disease in mild cognitive impairment and Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2020;92:45–52.
    OpenUrl
  42. 42.↵
    1. Passamonti L,
    2. Tsvetanov KA,
    3. Jones PS,
    4. et al
    . Neuroinflammation and functional connectivity in Alzheimer’s disease: interactive influences on cognitive performance. J Neurosci. 2019;39:7218–7226.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    1. Fan Z,
    2. Dani M,
    3. Femminella GD,
    4. et al
    . Parametric mapping using spectral analysis for 11C-PBR28 PET reveals neuroinflammation in mild cognitive impairment subjects. Eur J Nucl Med Mol Imaging. 2018;45:1432–1441.
    OpenUrl
  44. 44.↵
    1. Pascoal TA,
    2. Benedet AL,
    3. Ashton NJ,
    4. et al
    . Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27:1592–1599.
    OpenUrlCrossRef
  45. 45.↵
    1. Dani M,
    2. Wood M,
    3. Mizoguchi R,
    4. et al
    . Microglial activation correlates in vivo with both tau and amyloid in Alzheimer’s disease. Brain. 2018;141:2740–2754.
    OpenUrlCrossRefPubMed
  46. 46.↵
    1. Ismail R,
    2. Parbo P,
    3. Madsen LS,
    4. et al
    . The relationships between neuroinflammation, beta-amyloid and tau deposition in Alzheimer’s disease: a longitudinal PET study. J Neuroinflammation. 2020;17:151.
    OpenUrl
  47. 47.↵
    1. Hamelin L,
    2. Lagarde J,
    3. Dorothée G,
    4. et al
    . Distinct dynamic profiles of microglial activation are associated with progression of Alzheimer’s disease. Brain. 2018;141:1855–1870.
    OpenUrlCrossRef
  48. 48.↵
    1. Parbo P,
    2. Madsen LS,
    3. Ismail R,
    4. et al
    . Low plasma neurofilament light levels associated with raised cortical microglial activation suggest inflammation acts to protect prodromal Alzheimer’s disease. Alzheimers Res Ther. 2020;12:3.
    OpenUrl
  49. 49.↵
    1. Tondo G,
    2. Boccalini C,
    3. Caminiti SP,
    4. et al
    . Brain metabolism and microglia activation in mild cognitive impairment: A combined [18F]FDG and [11C]-(R)-PK11195 PET study. J Alzheimers Dis. 2021;80:433–445.
    OpenUrl
  50. 50.↵
    1. Nicastro N,
    2. Malpetti M,
    3. Mak E,
    4. et al
    . Gray matter changes related to microglial activation in Alzheimer’s disease. Neurobiol Aging. 2020;94:236–242.
    OpenUrlCrossRef
  51. 51.↵
    1. Femminella GD,
    2. Dani M,
    3. Wood M,
    4. et al
    . Microglial activation in early Alzheimer trajectory is associated with higher gray matter volume. Neurology. 2019;92:e1331–e1343.
    OpenUrl
  52. 52.↵
    1. Zou J,
    2. Tao S,
    3. Johnson A,
    4. et al
    . Microglial activation, but not tau pathology, is independently associated with amyloid positivity and memory impairment. Neurobiol Aging. 2020;85:11–21.
    OpenUrl
  53. 53.↵
    1. Toppala S,
    2. Ekblad LL,
    3. Tuisku J,
    4. et al
    . Association of early beta-amyloid accumulation and neuroinflammation measured with [11C]PBR28 in elderly individuals without dementia. Neurology. 2021;96:e1608–e1619.
    OpenUrl
  54. 54.↵
    1. Kumar A,
    2. Muzik O,
    3. Shandal V,
    4. Chugani D,
    5. Chakraborty P,
    6. Chugani HT
    . Evaluation of age-related changes in translocator protein (TSPO) in human brain using 11C-[R]-PK11195 PET. J Neuroinflammation. 2012;9:232.
    OpenUrlCrossRefPubMed
  55. 55.↵
    1. Schuitemaker A,
    2. van der Doef TF,
    3. Boellaard R,
    4. et al
    . Microglial activation in healthy aging. Neurobiol Aging. 2012;33:1067–1072.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. McKeith IG,
    2. Boeve BF,
    3. Dickson DW,
    4. et al
    . Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB Consortium. Neurology. 2017;89:88–100.
    OpenUrlCrossRefPubMed
  57. 57.↵
    1. Dickson DW
    . Dementia with Lewy bodies: neuropathology. J Geriatr Psychiatry Neurol. 2002;15:210–216.
    OpenUrlPubMed
  58. 58.↵
    1. Irwin DJ,
    2. Grossman M,
    3. Weintraub D,
    4. et al
    . Neuropathological and genetic correlates of survival and dementia onset in synucleinopathies: a retrospective analysis. Lancet Neurol. 2017;16:55–65.
    OpenUrlCrossRefPubMed
  59. 59.↵
    1. Iannaccone S,
    2. Cerami C,
    3. Alessio M,
    4. et al
    . In vivo microglia activation in very early dementia with Lewy bodies, comparison with Parkinson’s disease. Parkinsonism Relat Disord. 2013;19:47–52.
    OpenUrlCrossRefPubMed
  60. 60.↵
    1. Surendranathan A,
    2. Su L,
    3. Mak E,
    4. et al
    . Early microglial activation and peripheral inflammation in dementia with Lewy bodies. Brain. 2018;141:3415–3427.
    OpenUrlPubMed
  61. 61.↵
    1. Edison P,
    2. Ahmed I,
    3. Fan Z,
    4. et al
    . Microglia, amyloid, and glucose metabolism in Parkinson’s disease with and without dementia. Neuropsychopharmacology. 2013;38:938–949.
    OpenUrlCrossRefPubMed
  62. 62.↵
    1. Mak E,
    2. Nicastro N,
    3. Malpetti M,
    4. et al
    . Imaging tau burden in dementia with Lewy bodies using [18F]-AV1451 positron emission tomography. Neurobiol Aging. 2021;101:172–180.
    OpenUrl
  63. 63.↵
    1. Goker-Alpan O,
    2. Masdeu JC,
    3. Kohn PD,
    4. et al
    . The neurobiology of glucocerebrosidase-associated parkinsonism: a positron emission tomography study of dopamine synthesis and regional cerebral blood flow. Brain. 2012;135:2440–2448.
    OpenUrlCrossRefPubMed
  64. 64.↵
    1. Mullin S,
    2. Stokholm MG,
    3. Hughes D,
    4. et al
    . Brain microglial activation increased in glucocerebrosidase (GBA) mutation carriers without Parkinson’s disease. Mov Disord. 2021;36:774–779.
    OpenUrl
  65. 65.↵
    1. Lavisse S,
    2. Goutal S,
    3. Wimberley C,
    4. et al
    . Increased microglial activation in patients with Parkinson disease using [18F]-DPA714 TSPO PET imaging. Parkinsonism Relat Disord. 2021;82:29–36.
    OpenUrl
  66. 66.↵
    1. Jucaite A,
    2. Cselenyi Z,
    3. Kreisl WC,
    4. et al
    . Glia imaging differentiates multiple system atrophy from Parkinson’s disease: a positron emission tomography study with [11C]PBR28 and machine learning analysis. Mov Disord. 2022;37:119–129.
    OpenUrl
  67. 67.↵
    1. Varnäs K,
    2. Cselényi Z,
    3. Jucaite A,
    4. et al
    . PET imaging of [11C]PBR28 in Parkinson’s disease patients does not indicate increased binding to TSPO despite reduced dopamine transporter binding. Eur J Nucl Med Mol Imaging. 2019;46:367–375.
    OpenUrl
  68. 68.↵
    1. Bevan-Jones WR,
    2. Cope TE,
    3. Jones PS,
    4. et al
    . Neuroinflammation and protein aggregation co-localize across the frontotemporal dementia spectrum. Brain. 2020;143:1010–1026.
    OpenUrlCrossRefPubMed
  69. 69.
    1. Malpetti M,
    2. Rittman T,
    3. Jones PS,
    4. et al
    . In vivo PET imaging of neuroinflammation in familial frontotemporal dementia. J Neurol Neurosurg Psychiatry. 2021;92:319–322.
    OpenUrlAbstract/FREE Full Text
  70. 70.↵
    1. Zhang J
    . Mapping neuroinflammation in frontotemporal dementia with molecular PET imaging. J Neuroinflammation. 2015;12:108.
    OpenUrl
  71. 71.↵
    1. Pascual B,
    2. Funk Q,
    3. Zanotti-Fregonara P,
    4. et al
    . Neuroinflammation is highest in areas of disease progression in semantic dementia. Brain. 2021;144:1565–1575.
    OpenUrl
  72. 72.↵
    1. Malpetti M,
    2. Passamonti L,
    3. Jones PS,
    4. et al
    . Neuroinflammation predicts disease progression in progressive supranuclear palsy. J Neurol Neurosurg Psychiatry. 2021;92:769–775.
    OpenUrlAbstract/FREE Full Text
  73. 73.↵
    1. Alshikho MJ,
    2. Zürcher NR,
    3. Loggia ML,
    4. et al
    . Integrated magnetic resonance imaging and [11C]-PBR28 positron emission tomographic imaging in amyotrophic lateral sclerosis. Ann Neurol. 2018;83:1186–1197.
    OpenUrlCrossRefPubMed
  74. 74.↵
    1. Van Weehaeghe D,
    2. Van Schoor E,
    3. De Vocht J,
    4. et al
    . TSPO versus P2X7 as a target for neuroinflammation: An in vitro and in vivo study. J Nucl Med. 2020;61:604–607.
    OpenUrlAbstract/FREE Full Text
  75. 75.↵
    1. Politis M,
    2. Lahiri N,
    3. Niccolini F,
    4. et al
    . Increased central microglial activation associated with peripheral cytokine levels in premanifest Huntington’s disease gene carriers. Neurobiol Dis. 2015;83:115–121.
    OpenUrlCrossRefPubMed
  76. 76.↵
    1. Rocha NP,
    2. Charron O,
    3. Latham LB,
    4. et al
    . Microglia activation in basal ganglia is a late event in Huntington disease pathophysiology. Neurol Neuroimmunol Neuroinflamm. 2021;8:e984.
    OpenUrlAbstract/FREE Full Text
  77. 77.↵
    1. Lois C,
    2. González I,
    3. Izquierdo-García D,
    4. et al
    . Neuroinflammation in Huntington’s disease: new insights with 11C-PBR28 PET/MRI. ACS Chem Neurosci. 2018;9:2563–2571.
    OpenUrlCrossRefPubMed
  78. 78.↵
    1. Walterfang M,
    2. Di Biase MA,
    3. Cropley VL,
    4. et al
    . Imaging of neuroinflammation in adult Niemann-Pick type C disease: a cross-sectional study. Neurology. 2020;94:e1716–e1725.
    OpenUrl
  79. 79.↵
    1. Serrano-Pozo A,
    2. Muzikansky A,
    3. Gómez-Isla T,
    4. et al
    . Differential relationships of reactive astrocytes and microglia to fibrillar amyloid deposits in Alzheimer disease. J Neuropathol Exp Neurol. 2013;72:462–471.
    OpenUrlCrossRefPubMed
  80. 80.↵
    1. Iliff JJ,
    2. Wang M,
    3. Liao Y,
    4. et al
    . A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med. 2012;4:147ra111.
    OpenUrlAbstract/FREE Full Text
  81. 81.↵
    1. Liddelow SA,
    2. Guttenplan KA,
    3. Clarke LE,
    4. et al
    . Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–487.
    OpenUrlCrossRefPubMed
  82. 82.↵
    1. Bohlen CJ,
    2. Bennett FC,
    3. Tucker AF,
    4. Collins HY,
    5. Mulinyawe SB,
    6. Barres BA
    . Diverse requirements for microglial survival, specification, and function revealed by defined-medium cultures. Neuron. 2017;94:759–773.e8.
    OpenUrlCrossRefPubMed
  83. 83.↵
    1. Jonsson T,
    2. Stefansson H,
    3. Steinberg S,
    4. et al
    . Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med. 2013;368:107–116.
    OpenUrlCrossRefPubMed
  84. 84.↵
    1. Del-Aguila JL,
    2. Li Z,
    3. Dube U,
    4. et al
    . A single-nuclei RNA sequencing study of Mendelian and sporadic AD in the human brain. Alzheimers Res Ther. 2019;11:71.
    OpenUrlCrossRef
  85. 85.↵
    1. Lee DC,
    2. Rizer J,
    3. Selenica ML,
    4. et al
    . LPS- induced inflammation exacerbates phospho-tau pathology in rTg4510 mice. J Neuroinflammation. 2010;7:56.
    OpenUrlCrossRefPubMed
  86. 86.↵
    1. Hamelin L,
    2. Lagarde J,
    3. Dorothée G,
    4. et al
    ; Clinical IMABio3 team. Early and protective microglial activation in Alzheimer’s disease: a prospective study using 18F-DPA-714 PET imaging. Brain. 2016;139:1252–1264.
    OpenUrlCrossRefPubMed
  87. 87.↵
    1. Focke C,
    2. Blume T,
    3. Zott B,
    4. et al
    . Early and longitudinal microglial activation but not amyloid accumulation predicts cognitive outcome in PS2APP mice. J Nucl Med. 2019;60:548–554.
    OpenUrlAbstract/FREE Full Text
  88. 88.↵
    1. Cummings J,
    2. Lee G,
    3. Ritter A,
    4. Sabbagh M,
    5. Zhong K
    . Alzheimer’s disease drug development pipeline: 2020. Alzheimers Dement (N Y). 2020;6:e12050.
    OpenUrl
  89. 89.↵
    1. Elmore MR,
    2. Najafi AR,
    3. Koike MA,
    4. et al
    . Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron. 2014;82:380–397.
    OpenUrlCrossRefPubMed
  90. 90.↵
    1. Han J,
    2. Chitu V,
    3. Stanley ER,
    4. Wszolek ZK,
    5. Karrenbauer VD,
    6. Harris RA
    . Inhibition of colony stimulating factor-1 receptor (CSF-1R) as a potential therapeutic strategy for neurodegenerative diseases: opportunities and challenges. Cell Mol Life Sci. 2022;79:219.
    OpenUrl
  91. 91.↵
    1. Horti AG,
    2. Naik R,
    3. Foss CA,
    4. et al
    . PET imaging of microglia by targeting macrophage colony-stimulating factor 1 receptor (CSF1R). Proc Natl Acad Sci USA. 2019;116:1686–1691.
    OpenUrlAbstract/FREE Full Text
  92. 92.↵
    1. Knight AC,
    2. Varlow C,
    3. Zi T,
    4. et al
    . In vitro evaluation of [3H]CPPC as a tool radioligand for CSF-1R. ACS Chem Neurosci. 2021;12:998–1006.
    OpenUrl
  93. 93.↵
    1. Zhou X,
    2. Ji B,
    3. Seki C,
    4. et al
    . PET imaging of colony-stimulating factor 1 receptor: a head-to-head comparison of a novel radioligand, 11C-GW2580, and 11C-CPPC, in mouse models of acute and chronic neuroinflammation and a rhesus monkey. J Cereb Blood Flow Metab. 2021;41:2410–2422.
    OpenUrl
  94. 94.↵
    1. Saura J,
    2. Kettler R,
    3. Da Prada M,
    4. Richards JG
    . Quantitative enzyme radioautography with 3H-Ro 41-1049 and 3H-Ro 19-6327 in vitro: localization and abundance of MAO-A and MAO-B in rat CNS, peripheral organs, and human brain. J Neurosci. 1992;12:1977–1999.
    OpenUrlAbstract/FREE Full Text
  95. 95.↵
    1. Ekblom J,
    2. Jossan SS,
    3. Bergström M,
    4. Oreland L,
    5. Walum E,
    6. Aquilonius SM
    . Monoamine oxidase-B in astrocytes. Glia. 1993;8:122–132.
    OpenUrlCrossRefPubMed
  96. 96.↵
    1. Gulyás B,
    2. Pavlova E,
    3. Kása P,
    4. et al
    . Activated MAO-B in the brain of Alzheimer patients, demonstrated by [11C]-L-deprenyl using whole hemisphere autoradiography. Neurochem Int. 2011;58:60–68.
    OpenUrlCrossRefPubMed
  97. 97.↵
    1. Fowler JS,
    2. Wolf AP,
    3. MacGregor RR,
    4. et al
    . Mechanistic positron emission tomography studies: demonstration of a deuterium isotope effect in the monoamine oxidase-catalyzed binding of [11C]L-deprenyl in living baboon brain. J Neurochem. 1988;51:1524–1534.
    OpenUrlCrossRefPubMed
  98. 98.↵
    1. Lammertsma AA,
    2. Bench CJ,
    3. Price GW,
    4. et al
    . Measurement of cerebral monoamine oxidase B activity using L-[11C]deprenyl and dynamic positron emission tomography. J Cereb Blood Flow Metab. 1991;11:545–556.
    OpenUrlCrossRefPubMed
  99. 99.↵
    1. Freedman NM,
    2. Mishani E,
    3. Krausz Y,
    4. et al
    . In vivo measurement of brain monoamine oxidase B occupancy by rasagiline, using 11C-l-deprenyl and PET. J Nucl Med. 2005;46:1618–1624.
    OpenUrlAbstract/FREE Full Text
  100. 100.↵
    1. Harada R,
    2. Hayakawa Y,
    3. Ezura M,
    4. et al
    . 18F-SMBT-1: a selective and reversible PET tracer for monoamine oxidase-B imaging. J Nucl Med. 2021;62:253–258.
    OpenUrlAbstract/FREE Full Text
  101. 101.↵
    1. Villemagne VL,
    2. Harada R,
    3. Dore V,
    4. et al
    . First-in-human evaluation of 18F-SMBT-1, a novel 18F-labeled MAO-B PET tracer for imaging reactive astrogliosis. J Nucl Med. January 27, 2022 [Epub ahead of print].
  102. 102.↵
    1. Bhattacharya A,
    2. Biber K
    . The microglial ATP-gated ion channel P2X7 as a CNS drug target. Glia. 2016;64:1772–1787.
    OpenUrlCrossRefPubMed
  103. 103.↵
    1. Cserép C,
    2. Pósfai B,
    3. Lénárt N,
    4. et al
    . Microglia monitor and protect neuronal function through specialized somatic purinergic junctions. Science. 2020;367:528–537.
    OpenUrlAbstract/FREE Full Text
  104. 104.↵
    1. Martin E,
    2. Amar M,
    3. Dalle C,
    4. et al
    . New role of P2X7 receptor in an Alzheimer’s disease mouse model. Mol Psychiatry. 2019;24:108–125.
    OpenUrl
  105. 105.↵
    1. McLarnon JG,
    2. Ryu JK,
    3. Walker DG,
    4. Choi HB
    . Upregulated expression of purinergic P2X(7) receptor in Alzheimer disease and amyloid-beta peptide-treated microglia and in peptide-injected rat hippocampus. J Neuropathol Exp Neurol. 2006;65:1090–1097.
    OpenUrl
  106. 106.↵
    1. Mildner A,
    2. Huang H,
    3. Radke J,
    4. Stenzel W,
    5. Priller J
    . P2Y12 receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia. 2017;65:375–387.
    OpenUrlCrossRefPubMed
  107. 107.↵
    1. Koole M,
    2. Schmidt ME,
    3. Hijzen A,
    4. et al
    . 18F-JNJ-64413739, a novel PET ligand for the P2X7 ion channel: radiation dosimetry, kinetic modeling, test-retest variability, and occupancy of the P2X7 antagonist JNJ-54175446. J Nucl Med. 2019;60:683–690.
    OpenUrlAbstract/FREE Full Text
  108. 108.↵
    1. Van Weehaeghe D,
    2. Koole M,
    3. Schmidt ME,
    4. et al
    . [11C]JNJ54173717, a novel P2X7 receptor radioligand as marker for neuroinflammation: human biodistribution, dosimetry, brain kinetic modelling and quantification of brain P2X7 receptors in patients with Parkinson’s disease and healthy volunteers. Eur J Nucl Med Mol Imaging. 2019;46:2051–2064.
    OpenUrl
  109. 109.↵
    1. Bisogno T,
    2. Oddi S,
    3. Piccoli A,
    4. Fazio D,
    5. Maccarrone M
    . Type-2 cannabinoid receptors in neurodegeneration. Pharmacol Res. 2016;111:721–730.
    OpenUrl
  110. 110.↵
    1. Janefjord E,
    2. Mååg JL,
    3. Harvey BS,
    4. Smid SD
    . Cannabinoid effects on β amyloid fibril and aggregate formation, neuronal and microglial-activated neurotoxicity in vitro. Cell Mol Neurobiol. 2014;34:31–42.
    OpenUrl
  111. 111.↵
    1. Ni R,
    2. Mu L,
    3. Ametamey S
    . Positron emission tomography of type 2 cannabinoid receptors for detecting inflammation in the central nervous system. Acta Pharmacol Sin. 2019;40:351–357.
    OpenUrl
  112. 112.↵
    1. Ahmad R,
    2. Postnov A,
    3. Bormans G,
    4. Versijpt J,
    5. Vandenbulcke M,
    6. Van Laere K
    . Decreased in vivo availability of the cannabinoid type 2 receptor in Alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2016;43:2219–2227.
    OpenUrl
  113. 113.↵
    1. Terada T,
    2. Therriault J,
    3. Kang MSP,
    4. et al
    . Mitochondrial complex I abnormalities is associated with tau and clinical symptoms in mild Alzheimer’s disease. Mol Neurodegener. 2021;16:28.
    OpenUrl
  114. 114.↵
    1. Fukumoto D,
    2. Nishiyama S,
    3. Harada N,
    4. Yamamoto S,
    5. Tsukada H
    . Detection of ischemic neuronal damage with [¹18F]BMS-747158-02, a mitochondrial complex-1 positron emission tomography ligand: small animal PET study in rat brain. Synapse. 2012;66:909–917.
    OpenUrlCrossRefPubMed
  115. 115.↵
    1. Hou C,
    2. Hsieh CJ,
    3. Li S,
    4. et al
    . Development of a positron emission tomography radiotracer for imaging elevated levels of superoxide in neuroinflammation. ACS Chem Neurosci. 2018;9:578–586.
    OpenUrl
  116. 116.↵
    1. Pearse DD,
    2. Hughes ZA
    . PDE4B as a microglia target to reduce neuroinflammation. Glia. 2016;64:1698–1709.
    OpenUrl
  117. 117.↵
    1. Zhang L,
    2. Chen L,
    3. Beck EM,
    4. et al
    . The discovery of a novel phosphodiesterase (PDE) 4B-preferring radioligand for positron emission tomography (PET) imaging. J Med Chem. 2017;60:8538–8551.
    OpenUrl
  118. 118.↵
    1. Dominy SS,
    2. Lynch C,
    3. Ermini F,
    4. et al
    . Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv. 2019;5:eaau3333.
    OpenUrlFREE Full Text
  119. 119.↵
    1. Kumar DK,
    2. Choi SH,
    3. Washicosky KJ,
    4. et al
    . Amyloid-β peptide protects against microbial infection in mouse and worm models of Alzheimer’s disease. Sci Transl Med. 2016;8:340ra72.
    OpenUrlAbstract/FREE Full Text
  120. 120.↵
    1. Carloni S,
    2. Bertocchi A,
    3. Mancinelli S,
    4. et al
    . Identification of a choroid plexus vascular barrier closing during intestinal inflammation. Science. 2021;374:439–448.
    OpenUrlPubMed
  • Received for publication January 15, 2022.
  • Revision received May 3, 2022.
PreviousNext
Back to top

In this issue

Journal of Nuclear Medicine: 63 (Supplement 1)
Journal of Nuclear Medicine
Vol. 63, Issue Supplement 1
June 1, 2022
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Complete Issue (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Journal of Nuclear Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Imaging Neuroinflammation in Neurodegenerative Disorders
(Your Name) has sent you a message from Journal of Nuclear Medicine
(Your Name) thought you would like to see the Journal of Nuclear Medicine web site.
Citation Tools
Imaging Neuroinflammation in Neurodegenerative Disorders
Joseph C. Masdeu, Belen Pascual, Masahiro Fujita
Journal of Nuclear Medicine Jun 2022, 63 (Supplement 1) 45S-52S; DOI: 10.2967/jnumed.121.263200

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Imaging Neuroinflammation in Neurodegenerative Disorders
Joseph C. Masdeu, Belen Pascual, Masahiro Fujita
Journal of Nuclear Medicine Jun 2022, 63 (Supplement 1) 45S-52S; DOI: 10.2967/jnumed.121.263200
Twitter logo Facebook logo LinkedIn logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Bookmark this article

Jump to section

  • Article
    • Abstract
    • TSPO PET: CURRENT TARGET TO IMAGE NEUROINFLAMMATION
    • NEUROINFLAMMATION PET IN NEURODEGENERATIVE DISORDERS
    • LIMITATIONS OF TSPO PET
    • NEW INFLAMMATION IMAGING TARGETS TO OVERCOME THE LIMITATIONS OF TSPO PET
    • OTHER PET MARKERS TO STUDY NEUROINFLAMMATION
    • LINKAGE BETWEEN PERIPHERAL AND CENTRAL INFLAMMATION
    • DISCLOSURE
    • REFERENCES
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • PDF

Related Articles

  • PubMed
  • Google Scholar

Cited By...

  • Progressive lifespan modifications in the corpus callosum following a single juvenile concussion in male mice monitored by diffusion MRI
  • Molecular Imaging of Neurodegeneration: The Way to New Horizons
  • Google Scholar

More in this TOC Section

  • First-in-Human Study of 18F-Labeled PET Tracer for Glutamate AMPA Receptor [18F]K-40: A Derivative of [11C]K-2
  • Detection of HER2-Low Lesions Using HER2-Targeted PET Imaging in Patients with Metastatic Breast Cancer: A Paired HER2 PET and Tumor Biopsy Analysis
  • [11C]Carfentanil PET Whole-Body Imaging of μ-Opioid Receptors: A First in-Human Study
Show more Clinical Investigation

Similar Articles

Keywords

  • Molecular imaging
  • Neurology
  • PET
  • Alzheimer’s disease
  • neurodegeneration
  • neuroinflammation
  • Positron Emission Tomography
  • TSPO
SNMMI

© 2025 SNMMI

Powered by HighWire