Abstract
There is increasing evidence showing the heterogeneity of microglia activation in neuroinflammatory and neurodegenerative diseases. It has been hypothesized that pro‐inflammatory microglia are detrimental and contribute to disease progression, while anti‐inflammatory microglia play a role in damage repair and remission. The development of therapeutics targeting the deleterious glial activity and modulating it into a regenerative phenotype relies heavily upon a clearer understanding of the microglia dynamics during disease progression and the ability to monitor therapeutic outcome in vivo. To that end, molecular imaging techniques are required to assess microglia dynamics and study their role in disease progression as well as to evaluate the outcome of therapeutic interventions. Positron emission tomography (PET) is such a molecular imaging technique, and provides unique capabilities for non‐invasive quantification of neuroinflammation and has the potential to discriminate between microglia phenotypes and define their role in the disease process. However, several obstacles limit the possibility for selective in vivo imaging of microglia phenotypes mainly related to the poor characterization of specific targets that distinguish the two ends of the microglia activation spectrum and lack of suitable tracers. PET tracers targeting translocator protein 18 kDa (TSPO) have been extensively explored, but despite the success in evaluating neuroinflammation they failed to discriminate between microglia activation statuses. In this review, we highlight the current knowledge on the microglia phenotypes in the major neuroinflammatory and neurodegenerative diseases. We also discuss the current and emerging PET imaging targets, the tracers and their potential in discriminating between the pro‐ and anti‐inflammatory microglia activation states.
Keywords: microglia phenotypes, neuroinflammation, PET imaging
The development of therapeutics targeting the deleterious glial activity and modulating it into a regenerative phenotype heavily relies on a better understanding of the microglia dynamics during disease progression and the ability to monitor therapeutic outcome in vivo. In this review, we will highlight the current knowledge on the microglia phenotypes in the major neuroinflammatory and neurodegenerative diseases. We will also discuss the current and emerging PET imaging targets, the tracers, and their potential in discriminating between the pro‐ and anti‐inflammatory microglia activation states.

INTRODUCTION
Microglia are the resident immune cells of the central nervous system (CNS). They originate from the yolk sac where progenitors, during development, migrate and populate the brain [1, 2]. Following this migration, the microglia population is maintained during the life‐span by self‐renewal through apoptosis and proliferative processes [3]. During development, microglia play a key function in shaping the neuronal network by supporting neural development and synapsis pruning [4, 5], while in the adult CNS they carry surveillance functions to maintain homeostasis. Taking advantage of their ramified plastic morphology and their high mobility, they continuously sense their environment for pathological insults and harmful stimuli [6]. Microglia are the first line of defense in the brain. In response to injury and disease pathogens they undergo cellular activation characterized by morphological change (from ramified into amoeboid shape), gliosis and functional changes, including alteration of cell surface receptors expression and production of cytokines and chemokines. Depending on the nature of the stimuli, activated microglia have been largely classified in two phenotypes, namely pro‐inflammatory and anti‐inflammatory, sitting at opposite ends of the activation spectrum. Pro‐inflammatory microglia are generally described as producers of inflammatory mediators, including tumor necrosis factor (TNF)‐α, interleukin (IL)‐1β, IL‐6, macrophage inflammatory protein (MIP)‐1 α, nitric oxide synthase (NOS) and reactive oxygen species (ROS), via which they exert their detrimental effects [7]. Conversely, anti‐inflammatory microglia are involved in dampening the inflammatory response and promoting the repair mechanisms. They are characterized by secretion of anti‐inflammatory cytokines such as IL‐4, IL‐10, IL‐13 and transforming growth factor (TGF)‐β and up‐regulation of markers such as arginase‐1 (Arg‐1), FIZZ‐1, CD206 and YM‐1 [7]. However, recent single‐cell transcriptomics and proteomics studies disputed this pro‐/anti‐inflammatory dichotomy and identified a multitude of activated microglia phenotypes due to a variety of gene signatures associated with diseases and depending on their environmental context [8, 9, 10].
Despite this tremendous progress in identifying the different microglia phenotypes in vitro and in vivo, a clear understanding of these processes in the human brain in vivo is still lacking. A non‐invasive imaging technique such as positron emission tomography (PET) is well suited for quantitative investigation of neuroinflammation and possesses the potential to discriminate between the different microglia phenotypes. Although it is now appreciated that the simple pro‐ and anti‐inflammatory phenotype may not capture the whole diversity of microglia activation [11], this classification serves as a crucial guide for PET tracer development. In this context, we understand the pro‐inflammatory response to be associated with disease progression and neurotoxicity, while the anti‐inflammatory response is linked to dampening of harmful effects and recovery. Most of the development in PET imaging of neuroinflammation has focused upon targeting translocator protein 18 kDa (TSPO). Despite the large panel of tracers developed, TSPO appeared to be a less than ideal target due to, among others, its expression on different brain cells and the lack of ability to differentiate between pro‐ and anti‐inflammatory microglia. These drawbacks encouraged the investigation of other targets that might be more selective for microglia and can differentiate between the activated phenotypes.
In the current review, we first summarize the different microglia activation phenotypes in the main chronic neuroinflammatory and neurodegenerative diseases, focusing on multiple sclerosis (MS), Alzheimer’s disease (AD) and Parkinson’s disease (PD). Then, we discuss the current and emerging PET imaging targets and their tracers and highlight, when characterized, their potential in discriminating between microglia activation phenotypes.
Microglia activation phenotypes in neuroinflammatory and neurodegenerative diseases
Neuroinflammation is an inflammatory process within the CNS and occurs as response to various pathological insults, such as infection, protein aggregation, trauma and ischemia. It is mainly driven by microglia and astrocytes and in certain cases by infiltrated immune cells from the blood, especially when the blood–brain barrier (BBB) is inflamed and compromised. The secretion of pro‐inflammatory cytokines and chemokines, nitric oxide (NO) and ROS usually mark this process. In an acute event, the mounted neuroinflammatory response will resolve when the triggering insults are eliminated, and microglia will transition into an anti‐inflammatory and regenerative phenotype. However, in neurodegenerative diseases, neuroinflammation tends to become a chronic process that fails to resolve itself due to continuous stimulation. This chronic activation leads to a variety of microglia phenotypes that potentially contributes to neuronal damage and death and becomes a major driver of disease progression.
In this section we concisely highlight the activation phenotypes of microglia in neuroinflammatory and neurodegenerative diseases and how they contribute to the disease pathology, focusing upon MS, AD and PD (Table 1).
TABLE 1.
Activated microglia phenotypes and the expressed cytokines and markers in neuroinflammatory and neurodegenerative diseases
| Disease | Microglia phenotype | Expression of markers and cytokines | Reference |
|---|---|---|---|
| Multiple sclerosis | |||
| Initial and early white matter active lesions | Pro‐inflammatory | CD68, MHC‐I, MHC‐II, CD86 | [14] |
| ROS, p22phox | |||
| Late active white matter lesions | Intermediate phenotype | MIP‐1β (CCL4), OPN | [15, 16, 17] |
| HLA‐DR, CD11c, AXL, CD45, CD68, CD206, CD163 | |||
| Slowly expanding white matter lesions (active rim) | Pro‐inflammatory | CD40, CD68, p22phox, iNOS, ferritin | [14, 18] |
| IL‐1, IL‐6 | |||
| Cortical lesions | MS 1 | ↓ P2Y12R, ↑HLA‐II, ↑CD68 | [21] |
| MS 2 | ↓ P2Y12R, hyper‐ramified morphology | [21] | |
| EAE animal model (early phase) | Pro‐inflammatory | CD86, CD40, MHC‐II | [22] |
| TNF‐α, IFN‐γ, IL‐12, IL‐6, iNOS, NO | |||
| EAE animal model (recovery phase) | Anti‐inflammatory | IL‐4, IL‐10 and IL‐13 | [22, 23] |
| Alzheimer’s disease | |||
| AD (late stage) | Pro‐inflammatory | IL‐1β, TNF‐α, p40, MHC‐II, iNOS | [9, 31] |
| AD1 (associated with Aβ) | ↓ P2Y12R | [10] | |
| ↑ TREM2, ↑ ApoE4, ↑ITGAX | |||
| AD2 (associated with pTau) | ↑ GRID2, P2Y12R, ↑CX3CR1 | [10] | |
| AD transgenic mice | Anti‐inflammatory (early Aβ pathology) | YM1 | [30] |
| Dynamic shift towards pro‐inflammatory (later stage) | |||
| DAM (phagocytic) | ↓ P2y12, ↓ CX3CR1, ↓ TMEM119 | [32] | |
| ↑ ApoE4, ↑ Tyrobp, ↑TREM2 | |||
| Dark microglia | CD11b, TREM2 | [39] | |
| Parkinson’s disease | |||
| PD | Pro‐inflammatory | COX‐2, iNOS | [43, 44] |
| IL‐1β, IL‐6, TNF‐α | |||
| PD animal models | Pro‐inflammatory | IL‐1β, IL‐6, TNF‐α, NOS2 | [55] |
| ↓ IGF‐1, ↓ MRC1 |
Abbreviations: ↑, Over‐expression; ↓, down‐regulation; AD, Alzheimer’s disease; ApoE4, apolipoprotein E‐4; CX3CR1, CX3C chemokine receptor‐1; EAE, experimental autoimmune encephalomyelitis; GRID2, glutamate receptor, ionotropic, delta 2; HLA‐DR, human leukocyte antigen D‐related; IFN, interferon; IGF, insulin‐like growth factor; IL, interleukin; iNOS, inducible nitric oxide synthase; ITGAX, integrin subunit alpha X; MHC, major histocompatibility complex; MIP, macrophage inflammatory protein; MRC, mannose receptor C; NO, nitric oxide; OPN, osteopontin; P2Y12R, purinergic 2Y receptor type 12; ROS, reactive oxygen species; TNF, tumor necrosis factor; TREM, triggering receptor expressed on myeloid cells; Tyrobp, transmembrane immune signaling adaptor.
Multiple sclerosis (MS)
MS is a chronic inflammatory disease of the CNS. The main pathological hallmark of MS is the presence of focal lesions in the white matter associated with extensive glia activation, demyelination and neurodegeneration [12, 13]. Activated microglia are at the core of the neuroinflammatory response in MS pathology. Recent discoveries of specific markers and molecular signatures of microglia [transmembrane protein 119 (TMEM119) and purinergic 2Y receptor type 12 (P2Y12R)] indicated their predominance over monocyte‐derived macrophages in MS lesions [14, 15]. Active demyelination and axonal injury are associated with the focal accumulation of activated microglia; however, the dynamics of microglia activation and their phenotypes in association with lesion types and disease progression is still not well understood. In white matter initial and early active lesion stages, microglia predominantly showed a pro‐inflammatory phenotype with expression of phagocytic and antigen presentation markers [CD68, major histocompatibility complex (MHC) classes I and II and CD86] and production of ROS and enzymes that produce them (p22phox). In later stages of active lesions and inactive microglia‐containing cores of active lesions, microglia adopted an intermediate phenotype and co‐expressed anti‐inflammatory markers (CD206 and CD163) together with pro‐inflammatory markers [14, 16, 17]. Single‐cell mass cytometry analysis on active lesions of progressive MS revealed enriched clusters of cells with higher expression of phagocytosis‐related markers [human leucocyte antigen D‐related (HLA‐DR), CD11c, AXL, CD45, CD68] and inflammatory molecules such as inflammatory cytokines MIP‐1β (CCL4) and osteopontin (OPN) [15]. In slowly expanding lesions, microglia present at the active rim adopted mainly a pro‐inflammatory phenotype with expression of CD40, CD68, p22phox, inducible nitric oxide synthase (iNOS) markers and ferritin, while expression of anti‐inflammatory markers was very low [14, 18]. This phenotype was supported by data from RNA microarrays on micro‐dissected tissues showing strong expression of complement factors and interleukins (IL‐1 and IL‐6), which play a role in pro‐inflammatory processes [18]. It is evident that pro‐inflammatory microglia contribute to the demyelination and axonal injury in the early active lesions and the active rim of chronic lesions [19]. The increased expression of antigen‐presenting markers, inflammatory cytokines and ROS support their strong role in the development of MS pathology. Active remyelination in MS lesions is most prominent in active white matter lesions, which harbor a high content of microglia with intermediate phenotype. In contrast, no or minimal remyelination is seen in slowly expanding lesions [20] where mainly pro‐inflammatory microglia are present. Cortical lesions are also present in MS; a recent study identified the presence of two distinct MS‐specific microglia in these lesions named MS1, defined by a low expression of P2Y12R, high HLA‐II and CD68 and MS2 characterized by a hyper‐ramified morphology with low P2Y12R expression [21]. Interestingly, only MS2 microglia were associated with neuronal loss in the cortical lesions [21].
Experimental autoimmune encephalomyelitis (EAE) is the most used model to mimic the pathology observed in MS. Pro‐inflammatory microglia are predominantly present in the early phase of EAE and express high levels of CD86, CD40 and MHC‐II on their surface. The prominent expression of pro‐inflammatory cytokines such as TNF‐α, interferon (IFN)‐γ, IL‐12, IL‐6, iNOS, NO and proteases highlight their important role in the early phase of EAE development [22]. The level of anti‐inflammatory microglia increases with EAE progression and peaks at the maximum of the clinical symptoms and in the recovery phase at the expense of pro‐inflammatory microglia. This dynamic shift towards anti‐inflammatory microglia dominance and the release of a variety of cytokines such as IL‐4, IL‐10 and IL‐13 play an important role in the resolution of inflammation and repair mechanisms [22, 23]. This pro‐ and anti‐inflammatory polarization balance highly correlates with the demyelination/remyelination process in acute demyelination models, probably via modulation of the inflammatory niche and oligodendrocyte progenitor cell response [24, 25].
Microglia activation in MS shows a very complex spatial and temporal pattern, and the exact role and contribution of microglia phenotypes to the evolution of MS lesions at different stages of the disease is still not totally clear.
Alzheimer’s disease (AD)
AD is the most common type of neurodegenerative diseases of the CNS and is associated with progressive cognitive decline and memory loss. Pathologically, AD is characterized by extracellular deposition of amyloid beta (Aβ) plaques and intraneuronal accumulation of phosphorylated tau tangles [26]. Neuroinflammation is a prominent feature of AD. Microglia have been identified around Aβ plaques and shown to undergo dramatic morphological and electrophysiological changes in comparison to plaque‐distant microglia, indicating an activated state [27, 28]. In AD, microglia are able to bind soluble Aβ and Aβ fibrils via cell‐surface and Toll‐like receptors, and microglia phagocytic activity of Aβ has been identified in vitro and in vivo, exhibiting their role in Aβ clearing [29]. Microglia surrounding the plaques express YM‐1 and are thus suggested to adopt an anti‐inflammatory activation phenotype [30]. An age‐related dynamic shift of microglia phenotype is reported in the PS1M146LxAPP751SL double mutated mice. Anti‐inflammatory microglia with phagocytic characteristics at the beginning of Aβ pathology could switch to a pro‐inflammatory phenotype at the advanced stage of disease in this model, which is induced in part by the age‐dependent accumulation of soluble Aβ oligomers [30]. Activated microglia in a later AD stage showed an increased expression of pro‐inflammatory markers such as IL‐1β, TNF‐α, p40, MHC‐II and iNOS, but not the anti‐inflammatory factors IL‐4 or IL‐10 [9, 31].
Recent transcriptomics studies identified a microglia phenotype associated with neurodegenerative diseases, termed disease‐associated microglia (DAM) [32]. DAM stained positively for intracellular Aβ particles and are spatially localized in the proximity of Aβ plaques in the 5XFAD mice, a transgenic animal model of AD, and in human post‐mortem brain [32]. DAM demonstrated significant changes in gene expression, including down‐regulation of homeostatic genes such as P2y12, CX3C chemokine receptor‐1 (Cx3cr1) and TMEM119, and up‐regulation of known AD risk factors such as apolipoprotein E‐4 (ApoE4), transmembrane immune signaling adaptor (Tyrobp) and TREM2 [32, 33]. Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglia‐specific cell surface receptor, and loss‐of‐function mutations in TREM2 are associated with increased risk of developing AD [34]. TREM2 binds Aβ and works as a sensor molecule for microglia activation [35]. TREM2‐deficient microglia showed reduced uptake of Aβ‐lipoprotein complexes in vitro and minimal evidence of Aβ internalization in vivo [36, 37]. The stimulation of TREM2 initiates signal transduction pathways that promote microglial chemotaxis, phagocytosis, survival and proliferation, suggesting an anti‐inflammatory and protective role of DAM in AD [34, 38]. A new phenotype of microglia, referred to as ‘dark microglia’, was identified in the APP/PS1 AD mouse and demonstrated a highly activated state with strong expression of CD11b and TREM2 [39]. Microglia activation in AD is complex, and results in a large variety of phenotypes [9] due to the presence of different stimuli and variety of activation pathways. Microglia activation and phenotypes evolve as a function of disease progression. A loss of homeostatic signature and transition of microglia into DAM population is a function of disease progression and occurs in two phases with a TREM2‐independent initial step and TREM2‐dependent second step [9, 32]. Recently, in a snRNAseq study on human AD tissues, two distinct AD‐associated microglia profiles were identified that associated either with Aβ (termed ‘AD1’) or with hyper‐phosphorylated tau (termed ‘AD2’) [10]. AD1 microglia correlated with phagocytic/activated markers including ITGAX, LPL, GPNMB, MYO1E and SPP1, while AD2 microglia were enriched with homeostasis genes such as CXCR1, P2Y12R and neuron‐related genes such as GRID2, ADGRB3 and DPP10 [10]. Interestingly, AD1 microglia showed a similar gene expression profile as the DAM identified in the amyloid mice model [32] with increased expression of TREM2 and APOE and down‐regulation of P2Y12R [10].
Current evidence suggests that at the early stages of AD, microglia have a protective role marked by phagocytosis and clearance of Aβ and secretion of anti‐inflammatory cytokines. However, as AD progresses, chronic stimulation and activation cause phenotypical changes in microglia towards a pro‐inflammatory phenotype. The pro‐inflammatory environment can alter the phagocytic activity of microglia, which possibly leads to increased accumulation of Aβ [38], and can stimulate neurotoxic astrocytes via IL‐1α and TNF [40]. In addition, complement‐mediated (C3R) synapse phagocytosis and loss induced by pro‐inflammatory microglia contribute to neurodegeneration and disease progression [38].
Parkinson’s disease (PD)
Progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the presence of intraneuronal Lewy body inclusions and neurites containing protein aggregates are the main pathological hallmarks of PD [41]. In PD patients, dopaminergic neuronal death results in progressive motor symptoms, including tremor at rest, bradykinesia, rigidity and postural instability. During the last decade it has become evident that neuroinflammation is associated with PD and activated microglia are present in the SNpc of PD patients [42]. In post‐mortem PD tissues, activated microglia express high levels of inflammatory cytokines such as IL‐1β, IL‐6 and TNF‐α, as well as iNOS and cyclooxygenase‐2 (COX‐2), suggesting a pro‐inflammatory activated phenotype [43, 44]. Analysis of cerebrospinal fluid (CSF) and peripheral blood plasma from PD patients also showed elevated levels of IL‐1β, IL‐6 and TNF‐α [44, 45, 46]. These inflammatory cytokines were also increased in the 6‐hydroxydopamine (6‐OHDA) and 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP) models of PD, while anti‐inflammatory cytokines and markers such as TGF‐β and CD206 were decreased [47, 48]. In non‐human primates, long‐term glial activation and high levels of TNF‐α and IFN‐γ were detected several years following MPTP treatment [49]. Importantly, the amounts of IFN‐γ and TNF‐α in the SNpc correlated with the degree of neurodegeneration and motor impairment [49]. The exposure of dopaminergic neurons to inflammatory cytokines has been shown to promote neuronal death. Over‐expression of TNF‐α or IL‐1β, pro‐inflammatory microglia cytokines, in the SNpc using an adenovirus vector was sufficient to induce nigral neuronal degeneration [50, 51]. However, neutralization of soluble TNF by intranigral administration of engineered dominant negative TNF compound XENP345 reduced the retrograde nigral degeneration in the 6‐OHDA model by 50% [52]. This emphasizes the cardinal role of pro‐inflammatory microglia and cytokines in dopaminergic neuronal degeneration. Another feature of PD pathology is the presence of α‐synuclein (α‐syn) aggregates that constitute the main component of the Lewy bodies and neurites. Extracellular α‐syn oligomers function as damage‐associated molecular patterns. Signaling via Toll‐like receptor (TLR)‐2 and ‐4, α‐syn activates microglia into a pro‐inflammatory phenotype and induces the secretion of inflammatory cytokines and markers such as TNF, IL‐1β, IL‐6, COX‐2, NOS and ROS [53, 54]. Administration of preformed fibrils in mouse brains induced the expression of pro‐inflammatory markers by microglia including NOS2, IL‐6, TNF and IL‐1β without affecting or decreasing anti‐inflammatory markers such as IFN regulatory factor‐4 (IRF‐4), insulin‐like growth factor‐1 (IGF‐1) and mannose receptor C‐1 (MRC‐1) [55]. Additionally, activated microglia play an active role in the process of α‐syn transmission to neurons via exosomes that is further amplified by the presence of pro‐inflammatory cytokines [56].
The role of pro‐inflammatory microglia as initiator or secondary effector in PD is still not clear. Chronically activated microglia and the secretion of high levels of pro‐inflammatory mediators that damage the neurons and induce the release of α‐syn further activate microglia resulting in a feed‐forward effect, promoting inflammation and neurodegeneration.
Translocator protein 18 kDa (TSPO) – gold standard for PET imaging of activated microglia
PET imaging research of microglial activation and neuroinflammation has been traditionally focused upon TSPO. TSPO is predominantly expressed on the outer mitochondrial membrane in multiple cell types in periphery and CNS and is linked to several critical cellular functions, including inflammatory pathways. TSPO PET, particularly using (R)‐[11C]PK11195, is often referred to as the gold standard for PET imaging of neuroinflammation. Limitations of (R)‐[11C]PK11195, including the low signal‐to‐noise ratio, have led to an extensive search for a more optimal TSPO radioligand, both taking into account an increase in affinity to reduce non‐specific binding (second‐generation TSPO ligands) and the difference in ligand binding affinity induced by single‐nucleotide polymorphism rs6971 (third‐generation TSPO ligands), reviewed in detail recently [57, 58]. Next to limitations of the PET ligands, targeting TSPO for neuroinflammation has challenges and limitations in itself. First, knowledge on levels and cellular localization of TSPO expression in healthy and diseased human brain largely stems from post‐mortem studies using TSPO radioligands, but results are highly inconsistent [59, 60]. Cellular sources of TSPO expression in healthy and diseased brain have recently been reviewed by Nutma et al. [61]. In short, immunohistochemical studies for TSPO in healthy human brain revealed that TSPO is expressed in glial cells (microglia and astrocytes), oligodendrocytes and vascular endothelial cells [62, 63]. In post‐mortem human AD brain samples, TSPO expression was demonstrated in microglia, macrophages and astrocytes but also in cells surrounding Aβ plaques [62] and only marginally significant differences in TSPO protein expression were found in temporal and frontal cortex between AD patients and healthy controls [63]. Furthermore, the TSPO burden did not correlate with neuropathology or with the glial response (microglia and astrocytes). In active and chronic active MS lesions, a strong correlation was shown between radioligand binding [(R)‐[3H]PK11195 and [3H]PBR28], but astrocytic TSPO accounted for up to 65% of TSPO‐positive cells in these lesions [64]. In PD rodent models, α‐syn aggregates have been linked to microglial activation and correlated with expression of TSPO, but in PD patients TSPO PET has thus far yielded inconclusive results (recently reviewed by [65]).
Moreover, TSPO expression was also shown not to be phenotype‐specific in microglia in active and chronic active MS lesions [64]. To directly correlate tracer binding to protein expression and distinguish cell types responsible for the radioligand uptake, Tournier et al. coupled fluorescence‐activated cell sorting (FACS) to readout in radioligand‐treated tissues (RTT) [66]. Using FACS‐RTT, cellular origin of TSPO expression was shown to be highly dependent upon neuroinflammation type [66], and thus should be investigated for every pathology in order to attribute correct conclusions regarding TSPO PET imaging in human disease.
In line with these recent findings regarding TSPO protein expression, preclinical and clinical research results using TSPO PET in neurological diseases show highly variable outcomes. Nevertheless, some interesting recent imaging studies are outlined here. In a transgenic rat model of AD, using second‐generation ligand [18F]FEPPA, researchers were unable to detect activated microglia that were MHC‐II‐positive. Similarly, in a rat model of stroke, TSPO PET signal was only found in the insult region, but not in remotely present MHC‐II‐positive microglia [67].
In healthy humans, a significant correlation between volume of distribution (VT) and age was found using second‐generation ligand [11C]PBR28 [59]. Interestingly, another study showed that the binding potential (BPND) values of second‐generation ligand [11C]DPA713 but not those of (R)‐[11C]PK11195 increased with age, although [11C]DPA713 BPND values also did not reach significance [68]. Similarly, increased BPND values for [11C]DPA713 were found in AD patients in all ROIs, but for (R)‐[11C]PK11195 BPND values were only elevated in precuneus [68], although patient cohorts for each tracer were different, so no direct comparison can be made.
(R)‐[11C]PK11195 was used in a cohort of MS patients to follow‐up on disease progress over 4 years [69]. Tracer uptake in normal‐appearing white matter (NAWM) and thalamus of MS patients was significantly higher compared with healthy controls at the baseline scan, but no difference in cortical gray matter was observed. Interestingly, this is the first clinical study, to our knowledge, showing that higher TSPO binding predicts greater clinical disability, independent of relapsing status [69].
Taken together, being the ‘gold standard’ in PET imaging of neuroinflammation in the clinic, TSPO PET has had some success in revealing neuroinflammatory processes in human aging and disease. However, due to TSPO expression being non‐specific for either pro‐ or anti‐inflammatory status and even non‐specific for a single cell type, there is still a need for the evaluation of other, more specific biological targets for PET imaging of neuroinflammation in human disease.
Emerging targets and PET tracers for imaging of activated microglia
To overcome the limitations of TSPO PET, other neuroinflammation and microglia imaging targets are being explored (recently reviewed in Narayanaswami et al. [70]). In the next part of this review, we describe the emerging targets for PET imaging of microglia and the most recent preclinical and clinical results. Focus lies in the most promising targets for which significant advances were made with regard to target characterization and for which PET tracer development includes initial biological evaluation (Table 2).
TABLE 2.
Activated microglia targets and their association with microglia phenotypes and potential tracers for PET imaging
| Molecular target | Cellular localization | Pro‐/anti‐inflammatory expression | Tracers | In vivo (pre‐)clinical research stage | References |
|---|---|---|---|---|---|
| P2X7R | Microglia (mainly), to a lower extend on oligodendrocytes and astrocytes | Potentially specific to pro‐inflammatory phenotype | [11C]GSK1482160 | First‐in‐human results in healthy volunteers warrant further clinical evaluation | [80, 81, 82] |
| [11C]SMW139 | First‐in‐human results in healthy volunteers and MS patients warrant further clinical evaluation | [75, 83] | |||
| [11C]JNJ‐54173717 | No differences in uptake between healthy volunteers and PD or ALS patients | [84, 85, 86, 143] | |||
| [18F]JNJ‐64413739 | High inter‐individual signal variability in non‐human primates and human | [87, 88, 89] | |||
| P2Y12R | Microglia | Potentially specific to anti‐inflammatory phenotype | [11C]5 | No in vivo data | [25, 94] |
| CB2R | Microglia, astrocytes, neurons, endothelial cells | No data on microglia phenotype expression | [11C]NE40 | Lower uptake in AD patients than healthy volunteers | [111, 112] |
| [11C]RS‐028 | Unsuitable for clinic due to rapid washout of mouse spleen | [107] | |||
| [18F]11 | No difference between ALS mouse model and wild‐type | [110] | |||
| [18F]29 | Rapidly metabolized in mice | [109] | |||
| COX‐1 | Microglia, neurons | No data on microglia phenotype expression | [11C]KTP‐Me | Unsuitable for further clinical evaluation due to rapid washout observed in healthy volunteers and MCI/AD patients | [125] |
| [11C]PS13 | Specific uptake in non‐human primates, no increase after LPS treatment, good test‐retest reliability in healthy volunteers | [126, 128, 129 | |||
| [18F]PS13 | Specific uptake in non‐human primates | 130 | |||
| COX‐2 | Microglia, neurons | No data on microglia phenotype expression | [11C]MC1 | Increased uptake after LPS treatment in non‐human primates, higher uptake in inflamed joints (and interestingly brain) of RA patients | [126, 127] |
| [11C]TMI | Metabolically stable in non‐human primates | [131] | |||
| [11C]MPbP | Metabolically stable in mice | [133] | |||
| [18F]1 | Increased uptake in LPS‐treated rats | [132] | |||
| CSF‐1R | Microglia | No data on microglia phenotype expression | [11C]AZ683 | Low brain uptake in rats and non‐human primates | [114] |
| [11C]CPPC | Increased uptake in LPS‐treated, AD and EAE mice and LPS‐treated non‐human primates, in vitro data in human AD tissue warrants clinical evaluation | [115] | |||
| [11C]BLZ945 | Efflux transporter substrate, unsuitable for clinical application | [122] | |||
| [11C]5 | Does not cross BBB in rats | [123] |
Abbreviations: AD, Alzheimer’s disease; ALS, amyotrophic lateral sclerosis; BBB, blood–brain barrier; CB, cannabinoid receptor; COX, cyclooxygenase; CSF‐1R, colony stimulating factor 1 receptor; EAE, experimental autoimmune encephalomyelitis; LPS, lipopolysaccharide; MS, multiple sclerosis; P2X7R, purinergic 2X receptor type 7; P2Y12R, purinergic 2Y receptor type 12; PD, Parkinson’s disease; PET, positron emission tomography; RA, rheumatoid arthritis.
Purinergic 2X receptor type 7 (P2X7R)
P2X7R is an ATP‐gated ion channel expressed on cells of the myeloid lineage, lymphoid immune cells and mast cells. In CNS, the highest P2X7R density is found in microglia, but receptor expression is also found in astrocytes and oligodendrocytes, albeit to a lesser extent [71, 72]. The involvement of P2X7R in the immune response is best described by its role in the mechanism of IL‐1β secretion [71]. Following P2X7R activation by ATP, the efflux of large amounts of K+ drive the assembly of NLRP3 inflammasome and subsequent caspase‐1 activation [71, 73, 74]. High ATP concentrations (mM), generally an effect of cell damage or injury, are required to trigger receptor activation [73, 74]. P2X7R is therefore regarded silent in normal physiological conditions; however, it is highly up‐regulated during microglial activation. The increased microglial expression of P2X7R in neuroinflammatory disease is associated with a pro‐inflammatory activation phenotype. In vitro in polarized adult human microglia, P2X7R is highly over‐expressed in the pro‐inflammatory but not the anti‐inflammatory phenotype [25]. P2X7R was also over‐expressed in inflammatory microglia in white matter of active and chronic active MS lesions [25]. Similarly, P2X7R expression was increased in activated microglia in the EAE rat model during the disease phase [25, 75]. In AD mouse models, neuroinflammation induced by Aβ peptides increased the expression of P2X7R on microglia in senile plaques at advanced and late pathological stages [76], and P2X7R expression levels in plaque associated microglia progressively rose with age in line with the development of AD pathogenesis [77]. Altogether, P2X7R could serve as potential target for PET imaging of pro‐inflammatory microglia.
The recent development of several PET tracers targeting P2X7R by different research groups (for overviews on purinergic receptor imaging see Janssen et al. and Zarrinmayeh and Territo [78, 79]) highlights P2X7R as a promising target. In vitro autoradiography studies with [11C]GSK1482160 in EAE rats displayed increased tracer binding in lumbar spinal cord at 12–14 days post‐immunization (peak of the disease) compared with spinal cord of healthy rats [80]. However, these results could not be replicated in vivo due to too‐low affinity of [11C]GSK1482160 for rat P2X7R. When inducing neuroinflammation in mice using lipopolysaccharide (LPS), [11C]GSK1482160 uptake was higher in brains of LPS‐treated mice (72 h post‐treatment) compared with saline‐treated animals [81]. In a first‐in‐human study in healthy volunteers, brain uptake of [11C]GSK1482160 was demonstrated with a SUV of 1.0 at 20 min p.i. [82], deeming [11C]GSK1482160 suitable for further clinical evaluation.
Adamantyl benzamide [11C]SMW139 was used in a preclinical PET imaging study in EAE rats, displaying significantly elevated tracer uptake in spinal cord at the maximum of clinical symptoms in severe–relapsing EAE rats compared with recovery phase and control animals, but not in rats with mild–acute disease progression [75], probably due to milder inflammatory response. A first‐in‐human study showed good pharmacokinetics and quantifiable brain uptake of [11C]SMW139 in a small set of healthy volunteers and MS patients [83]. For every brain region investigated, VT and BPND values were higher for MS patients compared with healthy subjects, but only VT differed significantly.
To date, two Janssen R&D (Raritan, New Jersey, USA) compounds have been evaluated as a P2X7R PET tracer, both preclinically and clinically. In an acute rat model of PD (6‐OHDA injection), [11C]JNJ‐54173717 showed a time‐dependent increase in tracer binding in vitro, co‐localizing with Iba1 expression, but failed to show differences in tracer binding in a chronic A53T viral vector model of PD [84]. [11C]JNJ‐54173717 showed desirable and quantifiable brain uptake in human, but no significant differences were detected between brain uptake in healthy volunteers and PD [85] or ALS patients [86].
In rats unilaterally treated with LPS, [18F]JNJ‐64413739 uptake was significantly higher in the ipsilateral hemisphere compared with the contralateral hemisphere, in ex vivo autoradiography and in in vivo PET imaging, but not in in vitro autoradiography, suggesting differences in binding mechanism between in vitro and in vivo experiments [87]. A 90‐min dynamic PET acquisition in healthy humans showed limited bias, good test–retest variability and reliability [88], but no appropriate reference region could be determined both in human and non‐human primates [88, 89]. In addition, both [11C]JNJ‐54173717 and [18F]JNJ‐64413739 displayed high inter‐individual signal variability, possibly due to a binding effect of a P2X7R polymorphism [85, 88]; therefore, this needs to be further explored (Figure 1).
FIGURE 1.

Positron emission tomography (PET) tracers for purinergic 2X receptor type 7 (P2X7R)
Purinergic 2Y receptor type 12 (P2Y12R)
P2Y12R is a G‐protein coupled receptor involved in chemotaxis. Following the increase in ATP and ADP concentrations post‐brain injury, ADP acts as chemotactic signal that recruits microglia to sites of injury via activation of P2Y12R [90]. Receptor expression in the CNS is mainly restricted to microglia and P2Y12R is absent in peripheral macrophages, making it an ideal microglial marker [91]. P2Y12R is used as a homeostatic marker; however, in vitro studies also demonstrated significant up‐regulation of P2Y12R expression in anti‐inflammatory polarized microglia and down‐regulation upon pro‐inflammatory stimuli [25, 92, 93]. In rats, intracranial injection of recombinant IL‐4 increased P2Y12R expression in microglia in the injected hemisphere compared to the contralateral side, and the expression co‐localized with the anti‐inflammatory marker CD206 [94]. In a model of parasitic brain infection known to induce a T helper type 2 (Th‐2) response that promotes anti‐inflammatory microglia polarization, P2Y12R expression was highly increased on CD163‐positive microglia [92]. Conversely, P2Y12R expression is down‐regulated in pro‐inflammatory conditions. In MS, P2Y12R expression is absent in the active and chronic active lesions [14, 91] and in AD a loss of P2Y12R expression is observed in microglia surrounding Aβ plaques and Tau tangles [95]. Thus, P2Y12R is a promising target for PET imaging of anti‐inflammatory microglia. One prospective PET tracer ([11C]5) has been reported, displaying decreased tracer binding in the affected brain regions in in vitro autoradiography experiments in post‐mortem human and mouse brain tissue following stroke [94] and in brain tissue of MS patients and EAE rats [25]. In EAE rats, tracer binding increased in the recovery phase following the peak of clinical symptoms [25]. These results warrant the investigation of P2Y12R as a PET target for neuroprotective neuroinflammation, but unfortunately further preclinical evaluation of [11C]5 was discontinued due to its inability to cross the BBB (Figure 2).
FIGURE 2.

Prospective positron emission tomography (PET) tracer for purinergic 2Y receptor type 12 (P2Y12R)
Cannabinoid receptor type 2 (CB2)
CB2 is mainly expressed in immune cells in the periphery, but in CNS under healthy conditions low expression is found in microglia and neurons [96, 97]. Expression of CB2 on microglia significantly increases under neuroinflammatory conditions and in pathological conditions such as MS, AD and PD [98, 99]. Limited data are available on the association of CB2 expression with microglia activation phenotypes. However, CB2 in microglia has been extensively investigated as a therapeutic target to help limit the inflammatory process. Activation of CB2 receptors by cannabinoids modulates microglial immune function by increasing their beneficial properties such as phagocytosis, as well as reducing their production of pro‐inflammatory mediators such as TNF‐α and free radicals (recently reviewed by Duffey et al. [100]).
As CB1 is constitutively expressed in CNS, PET tracers need to be highly selective for CB2 over CB1. The latest preclinical and clinical results in CB2 tracer development are reviewed here; for a complete overview the reader is referred to reviews by Ametamey and co‐workers [101, 102].
As a proof of principle for PET tracers dedicated to imaging of inflammation in CNS, LPS‐induced neuroinflammation in rodents is a much‐used tool. A number of CB2 targeting PET tracers have been evaluated in this model [103, 104, 105, 106, 107, 108] with mixed results, the majority showing no significant increase in tracer uptake compared with control animals or non‐specific binding that is too high to pick up on small differences in radioactivity read‐out. A‐836339 analogue [18F]29 showed an increased uptake in brain of LPS‐treated mice versus control mice, but its high metabolic rate (7% intact at 30 min p.i.) limits the applicability of this tracer [109].
[11C]RS‐028 showed significantly higher tracer binding in post‐mortem spinal cord tissue of ALS patients versus healthy subjects, but was deemed unsuitable for in vivo purposes due to rapid washout in spleen (CB2‐rich organ) of mice [107].
Diastereomeric mixture [18F]11 was first tested in vitro, with similar levels of specific binding in control mouse brain (31%) compared with SOD1 mouse brain (ALS model), but the pattern of binding differed between the two [110]. In vivo PET imaging also showed no significant differences in time‐activity curves between animals, unless using a Logan plot with the left heart ventricle as a reference region, showing slightly higher values for symptomatic versus asymptomatic mice only [110].
[11C]NE40 is to date the only CB2 targeting tracer investigated in a clinical setting, showing rapid brain uptake and washout in healthy volunteers [111]. Brain uptake of [11C]NE40 was higher in healthy volunteers than in AD patients [112], which can probably be attributed to neuronal loss in late‐stage AD (Figure 3).
FIGURE 3.

Positron emission tomography (PET) tracers for cannabinoid receptor type 2 (CB2) receptor
Colony stimulating factor 1 receptor (CSF‐1R)
Colony stimulating factor 1 receptor (CSF‐1R) is a cell surface, class III receptor tyrosine kinase mainly expressed by microglia in the brain, but expression has also been found on cells of the neuronal lineage [113]. Endogenous ligands of CSF‐1R are cytokines CSF‐1 and IL‐34. CSF‐1R is directly involved in controlling development, survival and maintenance of microglia [114, 115]. In transgenic AD mice, high CSF‐1R expression is found in close proximity to Aβ aggregates [116, 117]. More recently, plaque‐associated microglia were found to be more resistant to microglia depletion by CSF‐1R inhibition than non‐plaque‐associated microglia in a mouse model displaying both Aβ and tau pathology [118]. In MS lesions, CSF‐1R expression was lower than in NAWM and in EAE mice, CSF‐1R inhibitors reduced disease severity [113]. Activation of microglia through CSF‐1R was recently shown to induce demyelination [119]. In line with this, increased levels of CSF‐1 in EAE mice correlated with microglial activation and neuronal loss [120]. In human brain tissue of MS patients, CSF‐1R was only modestly increased in relapse–remitting MS but significantly elevated in progressive MS compared to NAWM from non‐MS patients [121]. In addition, increased CSF‐1 could be detected in cerebrospinal fluid of progressive MS patients [121].
Four PET tracers targeting CSF‐1R have been evaluated in vivo to date (Figure 4). [11C]BLZ945 was evaluated in healthy mice but showed a substantial level of non‐specific binding in brain, both in vitro and in vivo [122]. In addition, low brain uptake due to strong efflux transporter substrate behavior was demonstrated, rendering [11C]BLZ945 unsuitable for clinical application. [11C]5 showed high specific binding in vitro, but selectivity for CSF‐1R was limited [123]. In healthy rats, [11C]5 did not cross the BBB, even when efflux transporters were inhibited, and thus [11C]5 is unfit for use in neuroimaging. [11C]AZ683 showed low brain uptake and retention in both rats and non‐human primates [114], but [11C]CPPC showed increased tracer uptake in mouse and baboon brains after LPS‐induced neuroinflammation [115]. Transgenic AD and EAE mice also showed an increase in PET signal, which correlated with disease severity in the EAE mice. [11C]CPPC binding was highly increased (75–99%) in post‐mortem brain of AD patients compared with healthy control brain in in vitro autoradiography studies [115]. The increasingly evident role of CSF‐1R in microglial activation and the first promising results in PET tracer development warrant further investigation of CSF‐1R as a PET target for neuroinflammation imaging.
FIGURE 4.

Positron emission tomography (PET) tracers for colony stimulating factor 1 receptor (CSF)‐1R
Cyclooxygenases
Cyclooxygenase (COX) enzymes are involved in activation of inflammatory pathways leading to prostaglandin production and chemokine, cytokine and ROS release [124]. Two known isoforms, COX‐1 and COX‐2, are both expressed in brain, although cell type localization and regulatory function differ. While COX‐1 is constitutively expressed in CNS, COX‐2 expression increases in inflammatory conditions. To accurately assess the role of both COX‐1 and COX‐2 in neuroinflammation by PET imaging, tracers targeting each isoform need to be highly selective over the other. In an attempt to develop a CNS‐applicable radiotracer for intracellular target COX‐1, a pro‐drug of COX‐1 inhibitor ketoprofen (ketoprofen methyl ester) was labeled with carbon‐11 ([11C]KTP‐Me) and evaluated in healthy volunteers and MCI/AD patients [125]. Initial brain uptake was followed by a very rapid washout in all groups, probably due to hydrolysis of the ester moiety, and [11C]KTP‐Me is therefore rendered unsuitable as PET tracer. Recently, two isoform selective tracers, [11C]PS13 (COX‐1) and [11C]MC1 (COX‐2), were studied in a head‐to‐head comparison in which [11C]PS13 showed high specific uptake in non‐human primates, while uptake of [11C]MC1 was mainly non‐specific at baseline conditions [126, 127, 128]. However, scans acquired post‐LPS treatment revealed significant up‐regulation of COX‐2 but not COX‐1 [126, 127]. Nevertheless, [11C]PS13 was evaluated in healthy humans, showing good test–retest reliability, and thus it would be interesting to investigate binding differences between healthy subjects and, for example, AD patients [129]. To provide a radiotracer with a longer half‐life, a [18F]PS13 was developed, showing similar promising results in non‐human primates, however, optimization of the radiolabeling procedure is required to obtain [18F]PS13 with sufficient molar activity [130]. [11C]MC1 was evaluated in two healthy volunteers and two patients with rheumatoid arthritis as a proof of principle, and notably higher binding in inflamed joints was observed in patients versus controls [127]. Unexpectedly, [11C]MC1 uptake in brain of rheumatoid arthritis patients could be blocked by celecoxib, indicating a potential central response to peripheral inflammation [127]. As celecoxib is a well‐known COX‐2 inhibitor, but subject to rapid metabolism, its scaffold has been explored to improve metabolic stability, as well as selectivity over COX‐1. Isoxazole derivative [11C]TMI showed strongly enhanced metabolic stability in non‐human primates, with a significant amount of radiometabolites (22% at 90 min p.i.) only observed in a blocking study using meloxicam [131]. Fluorinated derivative [18F]1 showed high brain uptake and excellent metabolic stability in mice [132], but could only be obtained in low yields and low molar activity, hampering further evaluation of this tracer. Exploring a different scaffold, [11C]MPbP showed increased uptake in brains of LPS‐treated rats compared with non‐treated rats; however, selectivity of this tracer over COX‐1 is limited [133] (Figure 5).
FIGURE 5.

Positron emission tomography (PET) tracers for cyclooxygenase (COX)‐1 and COX‐2
Summarizing, COX‐2 seems to be a legitimate target for PET imaging of neuroinflammation, whereas for COX‐1 this remains unclear.
Therapeutic perspectives of modulating microglia phenotypes
The increased understanding of microglia phenotypes and their contribution to disease development and progression offers novel approaches for therapeutic intervention. As anti‐inflammatory microglia promote regeneration and repair mechanisms, strategies aiming at modulating microglia into the anti‐inflammatory phenotype has gained momentum in recent years. In animals, brain delivery of anti‐inflammatory microglia inducers such as IL‐4, IL‐10 and IL‐13 have been shown to be effective in reducing the clinical symptoms and the demyelination in the EAE model [134]. Similarly, intracranial delivery of IL‐4 reduced the Aβ plaque load and improved cognition in the APP23 AD mice [134]. Although the results with these agents seem promising, the translation into human remains a challenge, as most of the cytokines do not cross the BBB. Extensive effort has been made to target other immunomodulators, such as the peroxisome proliferator‐activated receptor‐γ (PPARγ). PPARγ is a transcription factor that has long been known to modulate microglia phenotype. PPARγ agonists are able to induce the anti‐inflammatory microglia phenotype and have been shown to facilitate the clearance of Aβ in AD models and to exhibit a protective function against striatal dopamine depletion and restore cognitive function in the MPTP model of PD [135]. However, in a Phase III double‐blind randomized placebo‐controlled study in AD patients, PPARγ agonist rosiglitazone showed no efficacy nor improvement in cognition [136]. This was probably due to the poor BBB permeability of the compound, which raises uncertainties as to whether target occupancy was sufficient. Recently, a Phase IIa study with a new PPARγ/δ agonist T3D‐959 evaluated brain pharmacology of the drug by monitoring brain metabolism with [18F]FDG‐PET [137]. They were able to show that T3D‐959 alters glucose metabolism in the brain in a dose‐dependent manner. The study also provided an indication of improving cognition and higher executive function in mild‐to‐moderate AD supporting further clinical investigation [137]. Minocycline is a brain‐penetrating antibiotic that was shown to have an anti‐inflammatory effect in vitro and in animal models via reduction of inflammatory mediators such as IL‐1β, IL‐6, COX‐2 and iNOS [138]. A randomized controlled trial in MS patients showed that the risk of conversion from a clinically isolated syndrome to MS was significantly lower with minocycline than with placebo over 6 months [139]. Conversely, in a clinical study with AD patients, minocycline at a dose of 200 and 400 mg failed to delay the progress of cognitive or functional impairment in people with mild AD during a 2‐year period [Minocycline in Alzheimer’s Disease Efficacy (MADE) trial] [140]. Recently, more attention has been directed to the immunomodulatory characteristics of the sphingosine‐1‐phosphate receptor modulator fingolimod (FTY720), an immunosuppressor that is clinically approved for MS. In AD and stroke animal models, fingolimod was able to shift microglia into an anti‐inflammatory phenotype, which encourages further investigation [141, 142].
Although the modulation of microglia is a promising approach, successful clinical translation has been challenging. Our understanding of the dynamics of microglia activation and resulting phenotypes in the different neuroinflammatory and neurodegenerative diseases is still incomplete. Translation from animal models is also not straightforward as doses used to effectively modulate neuroinflammation in animals may not be tolerated in humans, and lower doses would not provide the wanted neuroprotective effect. Furthermore, the timing for modulation of pro‐ and anti‐inflammatory microglia is critical and needs to be tailored to the different diseases. Altogether, this stresses the need for reliable in vivo measurement methods that could enable the assessment of microglia phenotypes and their dynamics, define the therapeutic window and monitor the therapeutic efficacy to guide the clinical trial design.
DISCUSSION AND CONCLUSION
PET imaging is particularly well suited to quantitatively evaluate and discriminate the different microglia phenotypes, assuming that a selective target is identified and validated and proper radiotracers become available. Significant efforts have been made to go beyond TSPO PET and identify imaging biomarkers specific for microglia activation phenotypes. This proved not to be trivial, and only a couple of targets have been associated with pro‐ or anti‐inflammatory microglia phenotypes. This can be attributed to several limitations; (i) the strategies used to identify differential phenotypical markers mainly rely on in vitro polarized microglia that may not capture the complexity of the stimulus in a pathological environment; (ii) the lack of reliable antibody tools, which limits the validation of the identified biomarkers; and (iii) the limited availability and accessibility of human post‐mortem tissues that are crucial for exploration and validation of the biomarkers in different pathologies.
Among the emerging targets, P2X7R and P2Y12R stand out as more specific for pro‐ and anti‐inflammatory microglia, respectively. Several P2X7R tracers have been developed and evaluated in neuroinflammatory and neurodegenerative diseases but results were not consistent in showing clear differences between healthy control and disease state. In the case of P2Y12R, the lack of a BBB penetrable tracer makes it difficult to assess its value. The remaining targets are less well characterized with respect to association with microglia phenotypes. Additionally, in the case of CB2, its neuronal expression may hamper imaging interpretation in neurodegenerative diseases. It is unlikely that a single imaging marker would be able to discern the inflammatory environment, and a combination of two or three markers would be needed. One could imagine combining P2X7R with P2Y12R and evaluating their relative dynamics as an indication of microglia activation status at different time‐points during disease progression or treatment. This could be applied, for example, in the evaluation of disease‐modifying therapies in longitudinal studies. A decrease of P2X7R tracer uptake that coincides with an increase of P2Y12R uptake could be indicative of a shift in microglia towards an anti‐inflammatory phenotype. In addition to the targets described here, several other markers are being explored and more will emerge from mining microglia transcriptomic and snRNA sequencing studies. This stresses the importance to define basic characteristics for a PET target, which ideally should be (i) specifically expressed on microglia and minimal expression on other brain cells; (ii) localized on the cell membrane; (iii) differentially up‐regulated on activated microglia in pro‐ or anti‐inflammatory conditions; (iv) low homeostatic levels of expression; and (v) low level of polymorphism.
Additionally, radiotracer development for PET neuroimaging is a challenging task. An important criterion for a candidate PET tracer is its ability to cross the BBB. In the case of P2Y12R, this has been a bottleneck for the development of a candidate PET tracer. While the developed tracer displayed favorable in vitro binding properties, it failed to show BBB permeability and brain uptake. To address this problem, new computational approaches can potentially be used to predict the BBB permeability of compound libraries and their affinity to efflux transporters. This will help in identifying the most suitable candidate compounds for translation into PET tracers and increase their chances of success in in vivo evaluation. Other downfalls that limit clinical translation of tracers are related to their non‐specific uptake in the brain and the presence of radioactive metabolites that cross the BBB. This can present serious complications for accurate quantification of tracer uptake and the development of kinetic modeling approaches. It is also crucial for PET radiotracers to not be affected by the target polymorphism which was a hurdle for the TSPO PET tracer development.
It is now apparent that the recent advances in transcriptomic and snRNA sequencing have shone light upon the complexity of the microglia phenotypes in different pathologies and have taken it beyond the limits of the pro‐ and anti‐inflammatory dichotomy. Conversely, they also provided a powerful tool to identify specific targets that can be used to image these subsets. Thus, there is a call to action to identify and validate biomarkers that better capture these complex and dynamic microglia activation phenotypes in order to accurately investigate their role in disease progression and monitor therapeutic response. It is also crucial that imaging biomarkers are extensively validated and shown to provide predictive value for clinical or therapeutic outcome before investing time and resources in developing radiotracers.
CONFLICTS OF INTEREST
A.D.W. is editor‐in‐chief of Nuclear Medicine and Biology; the other authors declare no conflicts of interest.
AUTHOR CONTRIBUTIONS
W.B. designed the manuscript and together with B.J. equally contributed to the writing. D.J.V., H.E.deV. and A.D.W. critically reviewed the manuscript.
Beaino W, Janssen B, Vugts DJ, de Vries HE, Windhorst AD. Towards PET imaging of the dynamic phenotypes of microglia. Clin Exp Immunol. 2021;206:282–300. 10.1111/cei.13649
OTHER ARTICLES PUBLISHED IN THIS REVIEW SERIES
Imaging immune responses in neuroinflammatory diseases. Clinical and Experimental Immunology 2021, 206: 248–250.
Magnetic resonance imaging in neuromyelitis optica spectrum disorder. Clinical and Experimental Immunology 2021, 206: 251–265.
Clinical and neuroimaging findings in MOGAD‐MRI and OCT. Clinical and Experimental Immunology 2021, 206: 266–281.
Imaging immunological processes from blood to brain in amyotrophic lateral sclerosis. Clinical and Experimental Immunology 2021, 206: 301–313.
Neuroinflammation and immunoregulation in glioblastoma and brain metastases: Recent developments in imaging approaches. Clinical and Experimental Immunology 2021, 206: 314–324.
‘A picture is worth a thousand words’: The use of microscopy for imaging neuroinflammation. Clinical and Experimental Immunology 2021, 206: 325–345.
Wissam Beaino and Bieneke Janssen contributed equally to the writing of the manuscript.
REFERENCES
- 1. Alliot F, Godin I, Pessac B. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Dev Brain Res. 1999;117:145–52. [DOI] [PubMed] [Google Scholar]
- 2. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330:841–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Askew K, Li K, Olmos‐Alonso A, Garcia‐Moreno F, Liang Y, Richardson P, et al. Coupled proliferation and apoptosis maintain the rapid turnover of microglia in the adult brain. Cell Rep. 2017;18:391–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Schafer D, Lehrman E, Kautzman A, Koyama R, Mardinly A, Yamasaki R, et al. Microglia sculpt postnatal neural circuits in an activity and complement‐dependent manner. Neuron. 2012;74:691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, et al. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333:1456–8. [DOI] [PubMed] [Google Scholar]
- 6. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo . Science. 2005;308:1314–8. 10.1126/science.1110647. [DOI] [PubMed] [Google Scholar]
- 7. Colonna M, Butovsky O. Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol. 2017;35:441–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Miedema A, Wijering MHC, Eggen BJL, Kooistra SM. High‐resolution transcriptomic and proteomic profiling of heterogeneity of brain‐derived microglia in multiple sclerosis. Front Mol Neurosci. 2020;13:583811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Mathys H, Adaikkan C, Gao F, Young JZ, Manet E, Hemberg M, et al. Temporal tracking of microglia activation in neurodegeneration at single‐cell resolution. Cell Rep. 2017;21:366–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Gerrits E, Brouwer N, Kooistra SM, Woodbury ME, Vermeiren Y, Lambourne M, et al. Distinct amyloid‐β and tau‐associated microglia profiles in Alzheimer’s disease. Acta Neuropathol. 2021;141:681–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ransohoff RM. A polarizing question: do M1 and M2 microglia exist? Nat Neurosci. 2016;19:987–91. [DOI] [PubMed] [Google Scholar]
- 12. Popescu BFG, Lucchinetti CF. Pathology of demyelinating diseases. Annu Rev Pathol. 2012;7:185–217. [DOI] [PubMed] [Google Scholar]
- 13. Calabrese M, Filippi M, Gallo P. Cortical lesions in multiple sclerosis. Nat Rev Neurol. 2010;6:438–44. [DOI] [PubMed] [Google Scholar]
- 14. Zrzavy T, Hametner S, Wimmer I, Butovsky O, Weiner HL, Lassmann H. Loss of ‘homeostatic’ microglia and patterns of their activation in active multiple sclerosis. Brain. 2017;140:1900–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Böttcher C, van der Poel M, Fernández‐Zapata C, Schlickeiser S, Leman JKH, Hsiao C‐C, et al. Single‐cell mass cytometry reveals complex myeloid cell composition in active lesions of progressive multiple sclerosis. Acta Neuropathol Commun. 2020;8:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Peferoen LAN, Vogel DYS, Ummenthum K, Breur M, Heijnen PDAM, Gerritsen WH, et al. Activation status of human microglia is dependent on lesion formation stage and remyelination in multiple sclerosis. J Neuropathol Exp Neurol. 2015;74:48–63. [DOI] [PubMed] [Google Scholar]
- 17. Vogel DYS, Vereyken EJF, Glim JE, Heijnen PDAM, Moeton M, van der Valk P, et al. Macrophages in inflammatory multiple sclerosis lesions have an intermediate activation status. J Neuroinflamm. 2013;10:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Jäckle K, Zeis T, Schaeren‐Wiemers N, Junker A, van der Meer F, Kramann N, et al. Molecular signature of slowly expanding lfackesions in progressive multiple sclerosis. Brain. 2020;143:2073–88. [DOI] [PubMed] [Google Scholar]
- 19. Henderson APD, Barnett MH, Parratt JDE, Prineas JW. Multiple sclerosis: distribution of inflammatory cells in newly forming lesions. Ann Neurol. 2009;66:739–53. [DOI] [PubMed] [Google Scholar]
- 20. Bramow S, Frischer JM, Lassmann H, Koch‐Henriksen N, Lucchinetti CF, Sørensen PS, et al. Demyelination versus remyelination in progressive multiple sclerosis. Brain. 2010;133:2983–98. [DOI] [PubMed] [Google Scholar]
- 21. van Olst L, Rodriguez‐Mogeda C, Picon C, Kiljan S, James RE, Kamermans A, et al. Meningeal inflammation in multiple sclerosis induces phenotypic changes in cortical microglia that differentially associate with neurodegeneration. Acta Neuropathol. 2021;141:881–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Jiang Z, Jiang JX, Zhang G‐X. Macrophages: a double‐edged sword in experimental autoimmune encephalomyelitis. Immunol Lett. 2014;160:17–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Lassmann H. Pathology of inflammatory diseases of the nervous system: Human disease versus animal models. Glia. 2020;68:830–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Chu T, Zhang YP, Tian Z, Ye C, Zhu M, Shields LBE, et al. Dynamic response of microglia/macrophage polarization following demyelination in mice. J Neuroinflamm. 2019;16:188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Beaino W, Janssen B, Kooij G, van der Pol SMA, van Het Hof B, van Horssen J, et al. Purinergic receptors P2Y12R and P2X7R: potential targets for PET imaging of microglia phenotypes in multiple sclerosis. J Neuroinflamm. 2017;14:259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362:329–44. [DOI] [PubMed] [Google Scholar]
- 27. Plescher M, Seifert G, Hansen JN, Bedner P, Steinhäuser C, Halle A. Plaque‐dependent morphological and electrophysiological heterogeneity of microglia in an Alzheimer’s disease mouse model. Glia. 2018;66:1464–80. [DOI] [PubMed] [Google Scholar]
- 28. Mandrekar‐Colucci S, Landreth GE. Microglia and inflammation in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2010;9:156–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Heneka MT, Carson MJ, Khoury JE, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Jimenez S, Baglietto‐Vargas D, Caballero C, Moreno‐Gonzalez I, Torres M, Sanchez‐Varo R, et al. Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer’s disease: age‐dependent switch in the microglial phenotype from alternative to classic. J Neurosci. 2008;28:11650–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Martin E, Boucher C, Fontaine B, Delarasse C. Distinct inflammatory phenotypes of microglia and monocyte‐derived macrophages in Alzheimer’s disease models: effects of aging and amyloid pathology. Aging Cell. 2017;16:27–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Keren‐Shaul H, Spinrad A, Weiner A, Matcovitch‐Natan O, Dvir‐Szternfeld R, Ulland TK, et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell. 2017;169:1276–1290.e17. [DOI] [PubMed] [Google Scholar]
- 33. Sobue A, Komine O, Hara Y, Endo F, Mizoguchi H, Watanabe S, et al. Microglial gene signature reveals loss of homeostatic microglia associated with neurodegeneration of Alzheimer’s disease. Acta Neuropathol Commun. 2021;9:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Gratuze M, Leyns CEG, Holtzman DM. New insights into the role of TREM2 in Alzheimer’s disease. Mol Neurodegen. 2018;13:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Zhao Y, Wu X, Li X, Jiang L‐L, Gui X, Liu Y, et al. TREM2 Is a Receptor for β‐amyloid that mediates microglial function. Neuron. 2018;97:1023–1031.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M. TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid‐beta by microglia. Neuron. 2016;91:328–40. [DOI] [PubMed] [Google Scholar]
- 37. Yuan P, Condello C, Keene C, Wang Y, Bird T, Paul S, et al. TREM2 haplodeficiency in mice and humans impairs the microglia barrier function leading to decreased amyloid compaction and severe axonal dystrophy. Neuron. 2016;90:724–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer’s disease. J Cell Biol. 2018;217:459–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Bisht K, Sharma KP, Lecours C, Gabriela Sánchez M, El Hajj H, Milior G, et al. Dark microglia: a new phenotype predominantly associated with pathological states. Glia. 2016;64:826–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Dickson DW. Parkinson’s disease and parkinsonism: neuropathology. Cold Spring Harb Perspect Med. 2012;2:a009258. 10.1101/cshperspect.a009258 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Rocha NP, de Miranda AS, Teixeira AL. Insights into neuroinflammation in Parkinson’s disease: from biomarkers to anti‐inflammatory based therapies. Biomed Res Int. 2015;2015:628192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Knott C, Stern G, Wilkin GP. Inflammatory regulators in Parkinson’s disease: iNOS, lipocortin‐1, and cyclooxygenases‐1 and ‐2. Mol Cell Neurosci. 2000;16:724–39. [DOI] [PubMed] [Google Scholar]
- 44. Nagatsu T, Mogi M, Ichinose H, Togari A. Cytokines in Parkinson’s disease. J Neural Transm. 2000;578:143–51. [PubMed] [Google Scholar]
- 45. Mount MP, Lira A, Grimes D, Smith PD, Faucher S, Slack R, et al. Involvement of interferon‐gamma in microglial‐mediated loss of dopaminergic neurons. J Neurosci. 2007;27:3328–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Scalzo P, Kümmer A, Cardoso F, Teixeira AL. Serum levels of interleukin‐6 are elevated in patients with Parkinson’s disease and correlate with physical performance. Neurosci Lett. 2010;468:56–8. [DOI] [PubMed] [Google Scholar]
- 47. Pisanu A, Lecca D, Mulas G, Wardas J, Simbula G, Spiga S, et al. Dynamic changes in pro‐ and anti‐inflammatory cytokines in microglia after PPAR‐γ agonist neuroprotective treatment in the MPTPp mouse model of progressive Parkinson’s disease. Neurobiol Dis. 2014;71:280–91. [DOI] [PubMed] [Google Scholar]
- 48. Na SJ, DiLella AG, Lis EV, Jones K, Levine DM, Stone DJ, et al. Molecular profiling of a 6‐hydroxydopamine model of Parkinson's disease. Neurochem Res. 2010;35:761–72. [DOI] [PubMed] [Google Scholar]
- 49. Barcia C, Ros CM, Annese V, Gómez A, Ros‐Bernal F, Aguado‐Llera D, et al. IFN‐γ signaling, with the synergistic contribution of TNF‐α, mediates cell specific microglial and astroglial activation in experimental models of Parkinson’s disease. Cell Death Dis. 2011;2:e142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Ferrari CC, Pott Godoy MC, Tarelli R, Chertoff M, Depino AM, Pitossi FJ. Progressive neurodegeneration and motor disabilities induced by chronic expression of IL‐1beta in the substantia nigra. Neurobiol Dis. 2006;24:183–93. [DOI] [PubMed] [Google Scholar]
- 51. Lella Ezcurra AL, deChertoff M, Ferrari C, Graciarena M, Pitossi F. Chronic expression of low levels of tumor necrosis factor‐alpha in the substantia nigra elicits progressive neurodegeneration, delayed motor symptoms and microglia/macrophage activation. Neurobiol Dis. 2010;37:630–40. [DOI] [PubMed] [Google Scholar]
- 52. McCoy MK, Martinez TN, Ruhn KA, Szymkowski DE, Smith CG, Botterman BR, et al. Blocking soluble tumor necrosis factor signaling with dominant‐negative tumor necrosis factor inhibitor attenuates loss of dopaminergic neurons in models of Parkinson’s disease. J Neurosci. 2006;26:9365–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Kim C, Ho D‐H, Suk J‐E, You S, Michael S, Kang J, et al. Neuron‐released oligomeric α‐synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat Commun. 2013;4:1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Hughes CD, Choi ML, Ryten M, Hopkins L, Drews A, Botía JA, et al. Picomolar concentrations of oligomeric alpha‐synuclein sensitizes TLR4 to play an initiating role in Parkinson’s disease pathogenesis. Acta Neuropathol. 2019;137:103–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Sarkar S, Dammer EB, Malovic E, Olsen AL, Raza SA, Gao T, et al. Molecular signatures of neuroinflammation induced by αsynuclein aggregates in microglial cells. Front Immunol. 2020;11:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Guo M, Wang J, Zhao Y, Feng Y, Han S, Dong Q, et al. Microglial exosomes facilitate α‐synuclein transmission in Parkinson’s disease. Brain. 2020;143:1476–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Werry EL, Bright FM, Piguet O, Ittner LM, Halliday GM, Hodges JR, et al. Recent developments in TSPO PET imaging as a biomarker of neuroinflammation in neurodegenerative disorders. Int J Mol Sci 2019;20:3161. 10.3390/ijms20133161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Zhang L, Hu K, Shao T, Hou LU, Zhang S, Ye W, et al. Recent developments on PET radiotracers for TSPO and their applications in neuroimaging. Acta Pharm Sin B. 2021;11:373–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Paul S, Gallagher E, Liow J‐S, Mabins S, Henry K, Zoghbi SS, et al. Building a database for brain 18 kDa translocator protein imaged using 11CPBR28 in healthy subjects. J Cereb Blood Flow Metab. 2019;39:1138–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tong J, Williams B, Rusjan PM, Mizrahi R, Lacapère J‐J, McCluskey T, et al. Concentration, distribution, and influence of aging on the 18 kDa translocator protein in human brain: Implications for brain imaging studies. J Cereb Blood Flow Metab. 2020;40:1061–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Nutma E, Ceyzériat K, Amor S, Tsartsalis S, Millet P, Owen DR, et al. Cellular sources of TSPO expression in healthy and diseased brain. Eur J Nucl Med Mol Imaging. 2021. 10.1007/s00259-020-05166-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Cosenza‐Nashat M, Zhao M‐L, Suh H‐S, Morgan J, Natividad R, Morgello S, 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;35:306–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Gui Y, Marks JD, Das S, Hyman BT, Serrano‐Pozo A. Characterization of the 18 kDa translocator protein (TSPO) expression in post‐mortem normal and Alzheimer’s disease brains. Brain Pathol. 2020;30:151–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Nutma E, Stephenson JA, Gorter RP, de Bruin J, Boucherie DM, Donat CK, et al. A quantitative neuropathological assessment of translocator protein expression in multiple sclerosis. Brain. 2019;142:3440–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Belloli S, Morari M, Murtaj V, Valtorta S, Moresco RM, Gilardi MC. Translation imaging in Parkinson’s disease: focus on neuroinflammation. Front Aging Neurosci. 2020;12:152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Tournier BB, Tsartsalis S, Ceyzériat K, Medina Z, Fraser BH, Grégoire M‐C, et al. Fluorescence‐activated cell sorting to reveal the cell origin of radioligand binding. J Cereb Blood Flow Metab. 2020;40:1242–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Al‐Khishman NU, Qi QI, Roseborough AD, Levit A, Allman BL, Anazodo UC, et al. TSPO PET detects acute neuroinflammation but not diffuse chronically activated MHCII microglia in the rat. Eur J Nuclear Med Mol Imaging Res. 2020;10:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Yokokura M, Terada T, Bunai T, Nakaizumi K, Takebayashi K, Iwata Y, et al. Depiction of microglial activation in aging and dementia: positron emission tomography with 11CDPA713 versus 11C(R)PK11195. J Cereb Blood Flow Metab. 2017;37:877–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Sucksdorff M, Matilainen M, Tuisku J, Polvinen E, Vuorimaa A, Rokka J, et al. Brain TSPO‐PET predicts later disease progression independent of relapses in multiple sclerosis. Brain. 2020;143:3318–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Narayanaswami V, Dahl K, Bernard‐Gauthier V, Josephson L, Cumming P, Vasdev N. Emerging PET radiotracers and targets for imaging of neuroinflammation in neurodegenerative diseases: outlook beyond TSPO. Mol Imaging. 2018;17:1536012118792317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Illes P, Müller CE, Jacobson KA, Grutter T, Nicke A, Fountain SJ, et al. Update of P2X receptor properties and their pharmacology: IUPHAR Review 30. Br J Pharmacol. 2021;178:489–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Kaczmarek‐Hajek K, Zhang J, Kopp R, Grosche A, Rissiek B, Saul A, et al. Re‐evaluation of neuronal P2X7 expression using novel mouse models and a P2X7‐specific nanobody. eLife. 2018. 10.7554/eLife.36217 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Bartlett R, Stokes L, Sluyter R. The P2X7 receptor channel: recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev. 2014;66:638–75. [DOI] [PubMed] [Google Scholar]
- 74. Bhattacharya A, Biber K. The microglial ATP‐gated ion channel P2X7 as a CNS drug target. Glia. 2016;64:1772–87. [DOI] [PubMed] [Google Scholar]
- 75. Beaino W, Janssen B, Kooijman E, Vos R, Schuit RC, O’Brien‐Brown J, et al. PET imaging of P2X7R in the experimental autoimmune encephalomyelitis model of multiple sclerosis using 11CSMW139. J Neuroinflamm. 2020;17:300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Martínez‐Frailes C, Di Lauro C, Bianchi C, de Diego‐García L, Sebastián‐Serrano Á, Boscá L, et al. Amyloid peptide induced neuroinflammation increases the P2X7 receptor expression in microglial cells, impacting on its functionality. Front Cell Neurosci. 2019;13:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Lee HG, Won SM, Gwag BJ, Lee YB. Microglial P2X₇ receptor expression is accompanied by neuronal damage in the cerebral cortex of the APPswe/PS1dE9 mouse model of Alzheimer’s disease. Exp Mol Med. 2011;43:7–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Janssen B, Vugts DJ, Windhorst AD. PET imaging of purinergic receptors. In: Dierckx RA, Otte A, de Vries EF, van Waarde A, Lammertsma AA, eds. PET and SPECT of neurobiological systems. Cham: Springer International Publishing; 2021:879–89. [Google Scholar]
- 79. Zarrinmayeh H, Territo PR. Purinergic receptors of the central nervous system: biology, PET ligands, and their applications. Mol Imaging. 2020;19:1536012120927609. 10.1177/1536012120927609 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Han J, Liu H, Liu C, Jin H, Perlmutter JS, Egan TM, et al. Pharmacologic characterizations of a P2X7 receptor‐specific radioligand, 11CGSK1482160 for neuroinflammatory response. Nucl Med Commun. 2017;38:372–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Territo PR, Meyer JA, Peters JS, Riley AA, McCarthy BP, Gao M, et al. Characterization of 11C‐GSK1482160 for targeting the P2X7 receptor as a biomarker for neuroinflammation. J Nucl Med. 2017;58:458–65. [DOI] [PubMed] [Google Scholar]
- 82. Green M, Hutchins G, Fletcher J, Territo W, Polson H, Trussell H, et al. Distribution of the P2X7‐receptor‐targeted [11C]GSK1482160 radiopharmaceutical in normal human subjects. J Nucl Med. 2018;59:1009. [Google Scholar]
- 83. Hagens MHJ, Golla SSV, Janssen B, Vugts DJ, Beaino W, Windhorst AD, et al. The P2X7 receptor tracer 11CSMW139 as an in vivo marker of neuroinflammation in multiple sclerosis: a first‐in‐man study. Eur J Nucl Med Mol Imaging. 2020;47:379–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Crabbé M, Van der Perren A, Bollaerts I, Kounelis S, Baekelandt V, Bormans G, et al. Increased P2X7 receptor binding is associated with neuroinflammation in acute but not chronic rodent models for Parkinson’s disease. Front Neurosci. 2019;13:799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Van Weehaeghe D, Koole M, Schmidt ME, Deman S, Jacobs AH, Souche E, et al. 11CJNJ54173717, 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;46:2051–64. [DOI] [PubMed] [Google Scholar]
- 86. Van Weehaeghe D, Van Schoor E, De Vocht J, Koole M, Attili B, Celen S, et al. TSPO Versus P2X7 as a target for neuroinflammation: an in vitro and in vivo study. J Nucl Med. 2020;61:604–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Berdyyeva T, Xia C, Taylor N, He Y, Chen G, Huang C, et al. PET Imaging of the P2X7 ion channel with a novel tracer 18FJNJ‐64413739 in a rat model of neuroinflammation. Mol Imaging Biol. 2019;21:871–8. [DOI] [PubMed] [Google Scholar]
- 88. Koole M, Schmidt ME, Hijzen A, Ravenstijn P, Vandermeulen C, van Weehaeghe D, 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;60:683–90. [DOI] [PubMed] [Google Scholar]
- 89. Kolb HC, Barret O, Bhattacharya A, Chen G, Constantinescu C, Huang C, et al. Preclinical evaluation and nonhuman primate receptor occupancy study of 18F‐JNJ‐64413739, a PET radioligand for P2X7 receptors. J Nucl Med. 2019;60:1154–9. [DOI] [PubMed] [Google Scholar]
- 90. Jacobson KA, Delicado EG, Gachet C, Kennedy C, Kügelgen I, Li B, et al. Update of P2Y receptor pharmacology: IUPHAR Review 27. Br J Pharmacol. 2020;177:2413–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Mildner A, Huang H, Radke J, Stenzel W, Priller J. P2Y12 receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia. 2017;65:375–87. [DOI] [PubMed] [Google Scholar]
- 92. Moore CS, Ase AR, Kinsara A, Rao VTS, Michell‐Robinson M, Leong SY, et al. P2Y12 expression and function in alternatively activated human microglia. Neurol Neuroimmunol Neuroinflamm. 2015;2:e80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Walker DG, Tang TM, Mendsaikhan A, Tooyama I, Serrano GE, Sue LI, et al. Patterns of expression of purinergic receptor P2RY12, a putative marker for non‐activated microglia, in aged and Alzheimer’s disease brains. Int J Mol Sci. 2020;21:678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Villa A, Klein B, Janssen B, Pedragosa J, Pepe G, Zinnhardt B, et al. Identification of new molecular targets for PET imaging of the microglial anti‐inflammatory activation state. Theranostics. 2018;8:5400–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Gómez Morillas A, Besson VC, Lerouet D. Microglia and neuroinflammation: what place for P2RY12? Int J Mol Sci. 2021;22:1636. 10.3390/ijms22041636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Navarro G, Morales P, Rodríguez‐Cueto C, Fernández‐Ruiz J, Jagerovic N, Franco R. Targeting cannabinoid CB2 receptors in the central nervous system. Medicinal chemistry approaches with focus on neurodegenerative disorders. Front Neurosci. 2016;10:406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Savonenko AV, Melnikova T, Wang Y, Ravert H, Gao Y, Koppel J, et al. Cannabinoid CB2 receptors in a mouse model of Aβ amyloidosis: immunohistochemical analysis and suitability as a PET biomarker of neuroinflammation. PLOS ONE. 2015;10:e0129618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Yiangou Y, Facer P, Durrenberger P, Chessell IP, Naylor A, Bountra C, et al. COX‐2, CB2 and P2X7‐immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006;6:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Kelly R, Joers V, Tansey MG, McKernan DP, Dowd E. Microglial phenotypes and their relationship to the cannabinoid system: therapeutic implications for Parkinson’s disease. Molecules. 2020;25:453. 10.3390/molecules25030453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Duffy SS, Hayes JP, Fiore NT, Moalem‐Taylor G. The cannabinoid system and microglia in health and disease. Neuropharmacology. 2021;190:108555. [DOI] [PubMed] [Google Scholar]
- 101. Spinelli F, Mu L, Ametamey SM. Radioligands for positron emission tomography imaging of cannabinoid type 2 receptor. J Label Comp Radiopharm. 2018;61:299–308. [DOI] [PubMed] [Google Scholar]
- 102. Ni R, Mu L, Ametamey S. Positron emission tomography of type 2 cannabinoid receptors for detecting inflammation in the central nervous system. Acta Pharmacol Sin. a 2019;40:351–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Horti AG, Gao Y, Ravert HT, Finley P, Valentine H, Wong DF, et al. Synthesis and biodistribution of 11CA‐836339, a new potential radioligand for PET imaging of cannabinoid type 2 receptors (CB2). Bioorg Med Chem. 2010;18:5202–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Slavik R, Grether U, Müller Herde A, Gobbi L, Fingerle J, Ullmer C, et al. Discovery of a high affinity and selective pyridine analog as a potential positron emission tomography imaging agent for cannabinoid type 2 receptor. J Med Chem. 2015;58:4266–77. [DOI] [PubMed] [Google Scholar]
- 105. Slavik R, Herde AM, Bieri D, Weber M, Schibli R, Krämer SD, et al. Synthesis, radiolabeling and evaluation of novel 4‐oxo‐quinoline derivatives as PET tracers for imaging cannabinoid type 2 receptor. Eur J Med Chem. 2015;92:554–64. [DOI] [PubMed] [Google Scholar]
- 106. Slavik R, Müller Herde A, Haider A, Krämer SD, Weber M, Schibli R, et al. Discovery of a fluorinated 4‐oxo‐quinoline derivative as a potential positron emission tomography radiotracer for imaging cannabinoid receptor type 2. J Neurochem. 2016;138:874–86. [DOI] [PubMed] [Google Scholar]
- 107. Haider A, Spinelli F, Herde AM, Mu B, Keller C, Margelisch M, et al. Evaluation of 4‐oxo‐quinoline‐based CB2 PET radioligands in R6/2 chorea huntington mouse model and human ALS spinal cord tissue. Eur J Med Chem. 2018;145:746–59. [DOI] [PubMed] [Google Scholar]
- 108. Pottier G, Gómez‐Vallejo V, Padro D, Boisgard R, Dollé F, Llop J, et al. PET imaging of cannabinoid type 2 receptors with 11CA‐836339 did not evidence changes following neuroinflammation in rats. J Cereb Blood Flow Metab. 2017;37:1163–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Moldovan R‐P, Teodoro R, Gao Y, Deuther‐Conrad W, Kranz M, Wang Y, et al. Development of a high‐affinity PET radioligand for imaging cannabinoid subtype 2 receptor. J Med Chem. 2016;59:7840–55. [DOI] [PubMed] [Google Scholar]
- 110. Pascali G, Panetta D, De Simone M, Burchielli S, Lucchesi V, Sanguinetti E, et al. Preliminary investigation of a novel 18F radiopharmaceutical for imaging CB2 receptors in a SOD mouse model. Aust J Chem. 2021;74:443. 10.1071/CH20247 [DOI] [Google Scholar]
- 111. Ahmad R, Koole M, Evens N, Serdons K, Verbruggen A, Bormans G, et al. Whole‐body biodistribution and radiation dosimetry of the cannabinoid type 2 receptor ligand 11C‐NE40 in healthy subjects. Mol Imaging Biol. 2013;15:384–90. [DOI] [PubMed] [Google Scholar]
- 112. Ahmad R, Postnov A, Bormans G, Versijpt J, Vandenbulcke M, van Laere K. Decreased in vivo availability of the cannabinoid type 2 receptor in Alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2016;43:2219–27. [DOI] [PubMed] [Google Scholar]
- 113. Chitu V, Gokhan Ş, Nandi S, Mehler MF, Stanley ER. Emerging roles for CSF‐1 receptor and its ligands in the nervous system. Trends Neurosci. 2016;39:378–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Tanzey SS, Shao X, Stauff J, Arteaga J, Sherman P, Scott PJH, et al. Synthesis and initial in vivo evaluation of 11CAZ683 – a novel PET radiotracer for colony stimulating factor 1 receptor (CSF1R). Pharmaceuticals. 2018;11:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Horti AG, Naik R, Foss CA, Minn IL, Misheneva V, Du Y, et al. PET imaging of microglia by targeting macrophage colony‐stimulating factor 1 receptor (CSF1R). Proc Natl Acad Sci. 2019;116:1686–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Boissonneault V, Filali M, Lessard M, Relton J, Wong G, Rivest S. Powerful beneficial effects of macrophage colony‐stimulating factor on beta‐amyloid deposition and cognitive impairment in Alzheimer’s disease. Brain. 2009;132:1078–92. [DOI] [PubMed] [Google Scholar]
- 117. Murphy GM, Zhao F, Yang L, Cordell B. Expression of macrophage colony‐stimulating factor receptor is increased in the AβPPV717F transgenic mouse model of Alzheimer’s disease. Am J Pathol. 2000;157:895–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Lodder C, Scheyltjens I, Stancu IC, Botella Lucena P, Gutiérrez de Ravé M, Vanherle S, et al. CSF1R inhibition rescues tau pathology and neurodegeneration in an A/T/N model with combined AD pathologies, while preserving plaque associated microglia. Acta Neuropathol Commun. 2021;9:108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Marzan DE, Brügger‐Verdon V, West BL, Liddelow S, Samanta J, Salzer JL. Activated microglia drive demyelination via CSF1R signaling. Glia. 2021;69:1583–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Gushchina S, Pryce G, Yip PK, Wu D, Pallier P, Giovannoni G, et al. Increased expression of colony‐stimulating factor‐1 in mouse spinal cord with experimental autoimmune encephalomyelitis correlates with microglial activation and neuronal loss. Glia. 2018;66:2108–25. [DOI] [PubMed] [Google Scholar]
- 121. Hagan N, Kane JL, Grover D, Woodworth L, Madore C, Saleh J, et al. CSF1R signaling is a regulator of pathogenesis in progressive MS. Cell Death Dis. 2020;11:904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. van der Wildt B, Miao Z, Reyes ST, Park JH, Klockow JL, Zhao N, et al. BLZ945 derivatives for PET imaging of colony stimulating factor‐1 receptors in the brain. Nucl Med Biol. 2021;100–101:44–51. [DOI] [PubMed] [Google Scholar]
- 123. van der Wildt B, Nezam M, Kooijman EJM, Reyes ST, Shen B, Windhorst AD, et al. Evaluation of carbon‐11 labeled 5‐(1‐methyl‐1H‐pyrazol‐4‐yl)‐N‐(2‐methyl‐5‐(3‐(trifluoromethyl)benzamido)phenyl)nicotinamide as PET tracer for imaging of CSF‐1R expression in the brain. Bioorg Med Chem. 2021;42:116245. [DOI] [PubMed] [Google Scholar]
- 124. Aïd S, Bosetti F. Targeting cyclooxygenases‐1 and ‐2 in neuroinflammation: therapeutic implications. Biochimie. 2011;93:46–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Ohnishi A, Senda M, Yamane T, Mikami T, Nishida H, Nishio T, 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;43:438–44. [DOI] [PubMed] [Google Scholar]
- 126. Kim M‐J, Shrestha SS, Cortes M, Singh P, Morse C, Liow J‐S, et al. Evaluation of two potent and selective PET radioligands to image COX‐1 and COX‐2 in rhesus monkeys. J Nucl Med. 2018;59:1907–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Shrestha S, Kim M‐J, Eldridge M, Lehmann ML, Frankland M, Liow J‐S, et al. PET measurement of cyclooxygenase‐2 using a novel radioligand: upregulation in primate neuroinflammation and first‐in‐human study. J Neuroinflamm. 2020;17:140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Shrestha S, Singh P, Cortes‐Salva MY, Jenko KJ, Ikawa M, Kim M‐J, et al. 3‐Substituted 1,5‐diaryl‐1 H‐1,2,4‐triazoles as prospective PET radioligands for imaging brain COX‐1 in monkey. Part 2: Selection and evaluation of 11CPS13 for quantitative imaging. ACS Chem Neurosci. 2018;9:2620–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Kim M‐J, Lee J‐H, Juarez Anaya F, Hong J, Miller W, Telu S, et al. First‐in‐human evaluation of 11CPS13, a novel PET radioligand, to quantify cyclooxygenase‐1 in the brain. Eur J Nucl Med Mol Imaging. 2020;47:3143–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Taddei C, Morse CL, Kim M‐J, Liow J‐S, Montero Santamaria J, Zhang A, et al. Synthesis of 18FPS13 and evaluation as a PET radioligand for cyclooxygenase‐1 in monkey. ACS Chem Neurosci. 2021;12:517–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Kumar JSD, Zanderigo F, Prabhakaran J, Rubin‐Falcone H, Parsey RV, Mann JJ. In vivo evaluation of 11CTMI, a COX‐2 selective PET tracer, in baboons. Bioorg Med Chem Lett. 2018;28:3592–5. [DOI] [PubMed] [Google Scholar]
- 132. Lebedev A, Jiao J, Lee J, Yang F, Allison N, Herschman H, et al. Radiochemistry on electrodes: synthesis of an 18F‐labelled and in vivo stable COX‐2 inhibitor. PLOS ONE. 2017;12:e0176606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Kiseleva MM, Vaulina DD, Sivak KV, Alexandrov AG, Kuzmich NN, Viktorov NB, et al. Radiosynthesis of a novel 11 C‐labeled derivative of 4′‐O‐methylhonokiol and its preliminary evaluation in an LPS rat model of neuroinflammation. ChemistrySelect. 2020;5:2685–9. [Google Scholar]
- 134. Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflamm. 2014;11:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Strosznajder AK, Wójtowicz S, Jeżyna MJ, Sun GY, Strosznajder JB. Recent insights on the role of PPAR‐β/δ in neuroinflammation and neurodegeneration, and its potential target for therapy. NeuroMol Med. 2021;23:86–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Gold M, Alderton C, Zvartau‐Hind M, Egginton S, Saunders AM, Irizarry M, et al. Rosiglitazone monotherapy in mild‐to‐moderate Alzheimer's disease: results from a randomized, double‐blind, placebo‐controlled phase III study. Dement Geriatr Cogn Disord. 2010;30:131–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Chamberlain S, Gabriel H, Strittmatter W, Didsbury J. An exploratory Phase IIa study of the PPAR delta/gamma agonist T3D–959 assessing metabolic and cognitive function in subjects with mild to moderate Alzheimer’s disease. J Alzheimers Dis. 2020;73:1085–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Garrido‐Mesa N, Zarzuelo A, Gálvez J. Minocycline: far beyond an antibiotic. Br J Pharmacol. 2013;169:337–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Metz LM, Li DKB, Traboulsee AL, Duquette P, Eliasziw M, Cerchiaro G, et al. Trial of minocycline in a clinically isolated syndrome of multiple sclerosis. N Engl J Med. 2017;376:2122–33. [DOI] [PubMed] [Google Scholar]
- 140. Howard R, Zubko O, Bradley R, Harper E, Pank L, O’Brien J, et al. Minocycline at 2 different dosages vs placebo for patients with mild Alzheimer disease: a randomized clinical trial. JAMA Neurol. 2020;77:164–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Hu Z‐W, Zhou L‐Q, Yang S, Chen M, Yu H‐H, Tao R, et al. FTY720 Modulates microglia toward anti‐inflammatory phenotype by suppressing autophagy via STAT1 pathway. Cell Mol Neurobiol. 2021;41:353–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Qin C, Fan W‐H, Liu Q, Shang KE, Murugan M, Wu L‐J, et al. Fingolimod protects against ischemic white matter damage by modulating microglia toward M2 polarization via STAT3 pathway. Stroke. 2017;48:3336–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Ory D, Celen S, Gijsbers R, Van Den Haute C, Postnov A, Koole M, et al. Preclinical evaluation of a P2X7 receptor‐selective radiotracer: PET studies in a rat model with local overexpression of the human P2X7 receptor and in nonhuman primates. J Nucl Med. 2016;57:1436–41. [DOI] [PubMed] [Google Scholar]
