Skip to main content

REVIEW article

Front. Immunol., 23 September 2022
Sec. Inflammation
This article is part of the Research Topic Crosstalk between Peripheral and Local Immune Response in the Pathophysiology of Stroke and Neurodegeneration Diseases View all 30 articles

Novel brain PET imaging agents: Strategies for imaging neuroinflammation in Alzheimer’s disease and mild cognitive impairment

  • The First People’s Hospital of Linping District, Linping, China

Alzheimer’s disease (AD) is a devastating neurodegenerative disease with a concealed onset and continuous deterioration. Mild cognitive impairment (MCI) is the prodromal stage of AD. Molecule-based imaging with positron emission tomography (PET) is critical in tracking pathophysiological changes among AD and MCI patients. PET with novel targets is a promising approach for diagnostic imaging, particularly in AD patients. Our present review overviews the current status and applications of in vivo molecular imaging toward neuroinflammation. Although radiotracers can remarkably diagnose AD and MCI patients, a variety of limitations prevent the recommendation of a single technique. Recent studies examining neuroinflammation PET imaging suggest an alternative approach to evaluate disease progression. This review concludes that PET imaging towards neuroinflammation is considered a promising approach to deciphering the enigma of the pathophysiological process of AD and MCI.

1 Introduction

Alzheimer’s disease (AD) is a neurodegenerative disease with a concealed onset and continuous progress. It is a severe risk factor that is threatening the health and life of elderly individuals (1, 2). AD is known to accumulate two different insoluble protein aggregates. During the prodromal phase of AD, also called mild cognitive impairment (MCI), patients have a receding performance of cognitive domains (3). The neuropathological biomarkers for AD and MCI are β-amyloid (Aβ) plaques and intracellular tau neurofibrillary tangles (NFTs) (4, 5). Aβ cascade hypothesis is one of the dominant hypotheses of AD pathogenesis (6). It refers to the Aβ patch as the initial factor causing excessive phosphorylation of tau protein, microglial activation, neurotransmitter disorders, oxidative stress, and neuro-pathological changes (7). However, AD pathological changes may have been maintained for decades before symptoms occur. When the exact diagnosis has been made with the above approaches, patients may present with irreversible nervous system damages. Therefore, developing an early, non-invasive, and accurate diagnostic method could be a research hotspot.

The central nervous system (CNS) degeneration and disease neuropathology predate AD and MCI (8). One dominant theory indicates that the excess generation or disordered clearance of Aβ is the dominant event that initiates AD pathogenesis. However, about 30% of healthy individuals with Aβ deposition are without clinical symptoms. Nevertheless, most clinical studies revealed that high Aβ levels are associated with the severity of cognitive impairment. The hyperphosphorylated tau-based NFT pathology is positively associated with the severity of cognitive symptoms (9). However, anti-amyloid interventions have been reported to depict limited effects in clinical trials. Because of poor understanding of the pathogenesis, the clinical diagnosis of AD mainly depends on detailed medical history, imaging, and neuropsychological scale assessment. Therefore, besides the traditional biomarkers of AD pathology, more targets require further investigation in exploring AD.

Positron emission tomography (PET) is a neuroimaging approach to evaluate the molecular processes in the brain, effective and accurate for diagnostic purposes, clinical strategy planning, and assessing disease progression (10, 11). PET radioligands can bind to responding targets, including receptors, transporters, or enzymes. Furthermore, tracer binding or uptake degrees can serve as a quantitative neuropathology approach. With the advance of PET technology, targeted molecular probes toward Aβ deposition, tau protein, and neuroinflammation have been extensively developed and utilized in the clinical management of AD. PET biomarkers have also been recommended to ameliorate diagnostic accuracy for AD and MCI, including cerebral metabolism or Aβ deposition on amyloid PET (12). However, the rate of cerebral metabolism cannot explicitly elucidate potential neuropathology. Although measurement of amyloid and neuroinflammation through PET tracers provides excellent insight into the underlying process of AD and MCI, the latter has not been implemented in clinical practice. The present review describes the associated trials of PET imaging targeting neuroinflammation within AD and MCI.

2 AD and neuroinflammation

Currently, neuroinflammation in AD has been well illustrated (13). In the initial phase of neuroinflammation, immune cells aim to ameliorate neuronal injury. However, abnormal inflammatory responses involving prolonged microglial activation presented adverse effects and exacerbated neurodegeneration (14). According to histopathological studies, activated microglia localize to Aβ plaques and NFTs, which attached significant attention to the role of neuroinflammation in the AD process (15). Microglia are dominant immune cells in the central nervous system (CNS), essential in maintaining hemostasis and secreting inflammatory factors (16, 17). Under the hemostatic condition, microglia present M1-like status, while under pathological conditions, they convert to an M2 state (18). M1 microglia are pro-inflammatory and produce reactive oxygen species (ROS) to remove the foreign substance and trigger neuroinflammation, while M2 microglia present an anti-inflammatory function to protect the neurons. Additionally, reactive astrocyte is also a critical member of neuroinflammation, which precipitates both Aβ and tau and is closely linked to microgliosis (19, 20). Astrocytes can be classified into A1 and A2 subtypes based on their phenotype and genetic expression profiles (21). A1 astrocyte secretes and produces various inflammatory factors and neurotoxins, whereas A2 astrocyte produces neurotrophic substances (22, 23).

Neuroinflammation is considered to have an essential role in AD. It has been demonstrated that the persistent accumulation of Aβ levels in AD patients leads to activated neuroinflammation and elevated ROS, which induces cell death through apoptosis (7). Moreover, neuroinflammation initiated by infection has been revealed to exacerbate the tau pathological process in transgenic rodent AD models. Because elderly individuals are more prone to infections, the elevated Aβ burden with activated neuroinflammatory insult may accelerate the progression of neurodegeneration in patients at risk for AD (24). According to the above discussion, it is urgent to make international efforts to develop novel radiotracers for imaging neuroinflammation in AD patients with PET. Therefore, the following sections will briefly review the different strategies within advanced PET radiotracers targeting the potential molecules in the neuroinflammation process.

3 Application of PET imaging agents for neuroinflammation

The AD-associated neuroinflammatory biomarkers are divided into (1) enzymes or signal molecules, including 18-kDa transporters (TSPO), monoamine oxidase B (MAO-B), imidazoline binding sites I2 (I2BS), epoxidation enzyme, and arachidonic acid (AA); (2) G protein-coupled receptors, including purine P2X7 and P2Y receptors, and type 2 cannabinoid receptors (CB2R). Therefore, the corresponding PET tracers are developed and will be summarized in the following section.

3.1 TSPO PET radiotracer

It is reported that TSPO presents a variety of cellular functions, including cholesterol transport, inflammatory responses, and hormone synthesis. However, its exact role in the brain immune reaction is not fully understood. Under the hemostatic condition, the expression of TSPO maintains a low level in microglia within CNS, while after neuroinflammation, abnormal activation of microglia is associated with a high level of TSPO. Furthermore, upregulation of TSPO and activation of microglia colocalize spatially after a neurotoxic intervention was established through immunohistochemistry staining, suggesting that TSPO can detect activated microglia and can be a novel approach to measure neuroinflammation. Therefore, combining imaging agents with TSPO can serve as microglia activation and neuroinflammation biomarkers. 11C-PK11195 presents a high affinity to TSPO, the first classical probe used for PET neuroinflammation imaging. Accumulative evidence of PET images revealed that the region of the cingulate, temporoparietal cortex, and amygdala in patients with AD presented a high level of 11C-PK11195 absorption compared to healthy individuals. However, the low sensitivity, low bioavailability, high rate of nonspecific binding, and short half-life defects limited the application of 11C-PK11195 in patients with AD (2527). Therefore, the second- and third-generation imaging agents, including the 11C-DPA713, 18F-DPA714, 18F-GE180, and 11C-ER176, exert higher affinity, and brain uptake rate can detect TSPO in neuroinflammation in low expression.

3.1.1 First generation of TSPO PET radiotracers: 11C-PK11195

The dominant first generation of TSPO PET radiotracers was the 11C-PK11195, which is accompanied by either the racemic mixture or the active R-enantiomer. It has been considered the most widely used radiotracer for PET imaging in brain tissue. However, due to the intrinsic properties of the compound and complexity of carbon-11 radiolabeling within a short period of 20 min, the development of this technique for exploring neuroinflammation was impeded. Due to the poor blood–brain barrier (BBB) penetration and low brain uptake, 11C-PK11195 presents a poor signal-to-noise ratio. Moreover, 11C-PK11195 has various shortcomings, including low bioavailability and nonspecific binding, which limit its ability to determine subtle changes in TSPO expression in brain tissues (25, 27, 28). Therefore, the above difficulties challenge its application in clinical management and require further development of novel TSPO PET radiotracers.

3.1.2 Second generation of TSPO PET radiotracers

A wide range of second-generation TSPO PET radiotracers have emerged, which present a higher affinity to TSPO and better characteristics. Several radiotracers have already been widely used in patients, especially in AD. According to a previous study, 11C-PBR28 presents a higher specific signal towards microglial activity than 11C-PK11195 (29). Additionally, 11C-DPA-713 showed better sensitivity in the healthy brain than 11C-PK11195 in measuring increased TSPO expression in the brain (30). Moreover, 11C-DPA-713 determined increased TSPO density in widespread brain regions in AD patients than 11C-PK11195 (31). The evaluations of TSPO radioligands, including 11C-DPA-713, 18F-DPA-714, and 11C-PK11195, have been compared in acute neuroinflammation rat models (32). The results indicated that 18F-DPA-714 had the highest ipsilateral-to-contralateral uptake ratio and had a better binding potential than 11C-DPA-713 and 11C-PK11195 (31). Moreover, the ligand 11C-DAA1106 has demonstrated a higher affinity than 11C-PK11195 to activate microglia in various neurological disorders (33, 34). The development of novel molecules with higher affinity, greater bioavailability, and the possibility of radiolabel with 18F facilitates the easier development of inflammation imaging. Currently, a couple of radioligands labeled with 18F have also been established. Among these, 18F-FEDAA1106 presents a higher affinity to TSPO than DAA1106. Accumulated evidence has revealed the higher brain uptake of 18F-FEDAA1106 than 11C-PK11195 or 11C-DAA1106 (35). Moreover, 18F-FEMPA could be a suitable PET radiotracer for TSPO (36). It has been described that 18F-FEPPA is associated with higher affinity towards TSPO, higher brain penetration, and better pharmacokinetics than the first generation of TSPO PET radiotracers (37).

3.1.3 Third generation of TSPO PET radiotracers

A wide range of third-generation TSPO tracers, including 18F-GE-180 (R, S)-18F-GE-387, 11C-ER176, 11C-CB184, 11C-CB190, 11C-N′-MPB, and 18F-LW223, have been well established. A couple of clinical trials have compared the binding properties, brain uptake, and performance of TSPO radiotracers. The measurement of microglial activation through 18F-GE-180 was more sensitive than that by 18F-PBR06. A wide range of studies have reported on the value of 18F-GE-180 in assessing microglial activity in different rodent models. Furthermore, rising levels of 18F-GE-180 uptake indicate elevated microglial activation in patients with AD, semantic dementia, and MCI. However, another study on mouse stroke models suggested that 11C-DPA-713 PET presented higher accuracy and sensitivity on microglial activation measurement than 18F-GE-180. Moreover, a more favorable brain entrance property of 11C-PBR28 has also been reported compared to 18F-GE-180. 11C-ER176 has been revealed with a higher binding affinity than 11C-PK11195, 11C-PBR28, and 11C-DPA-71. A clinical trial of 11C-ER176 PET for accessing microglia activation in MCI and AD patients is still ongoing (NCT03744312).

Based on the evidence, the dominant concern for TSPO ligands is the cellular location of the signal. The different binding sites on glial and vascular TSPO have been reported for several TSPO ligands. The high-glial-TSPO-selectivity and polymorphism-sensitive ligand 18F-FEBMP has been developed. It presented a higher contrast to neuroinflammation than 11C-PK11195 in the PS19 tauopathy mouse model. Further studies assessing the binding selectivity to TSPO polymorphism among different generations of TSPO radiotracers are highly required.

3.2 TSPO PET imaging of neuroinflammation in AD

The following section will summarize the clinical effects secured from the clinical trials of PET imaging toward neuroinflammation among AD and MCI patients. An in-depth understanding of the underlying mechanisms of AD neuroinflammation will lead to novel therapeutic approaches to monitor disease progression using TSPO PET imaging.

3.2.1 First generation of TSPO PET imaging in AD

The radiotracer has been examined in accumulative studies involving AD and MCI patients (Table 1). The first clinical trial focusing on 11C-PK11195 failed to detect TSPO binding related to microglial activation in patients with mild to moderate dementia (51). Another AD study also reported a similar result, which suggested that microglial activation presented in later stages of AD or 11C-PK11195 is insensitive in mild to moderate AD (40). A recent study reveals only a small cluster of significantly elevated 11C-PK11195 binding in occipital lobes in AD dementia patients without any difference between clinically stable prodromal AD patients and those who progressed to dementia (43). The rising 11C-PK11195 brain uptake among AD patients was observed in two studies: (1) higher uptake was found in the frontal and right mesotemporal regions using SPECT (52); (2) higher uptake was seen in the frontal, temporal, occipital, and striatum using PET (39). Additionally, elevated regional 11C-PK11195 binding is observed in the entorhinal, temporoparietal, and cingulate cortex in patients having mild and early AD. The results were consistent with another study showing increased 11C-PK11195 signals (38, 53)

TABLE 1
www.frontiersin.org

Table 1 Studies examining regional brain uptake via first-generation TSPO tracers in AD and MCI.

Moreover, two other longitudinal studies provided the course of neuroinflammation in AD via 11C-PK11195. The first study revealed an increase in radiotracer signals in AD patients (44). On the contrary, another longitudinal study illustrated the evolution of 11C-PK11195 in eight MCI patients, four of whom revealed negative amyloid imaging. In contrast, a longitudinal reduction of microglial activation was observed in this population (47). The small sample sizes, different methods, and first-generation TSPO radiotracer limits could elaborate on these contradictory results.

3.2.2 Second generation of TSPO PET imaging in AD

The shortcomings of the second generation of TSPO radiotracers were improved with the development of advanced-generation TSPO ligands with an enhanced signal-to-noise ratio and higher binding affinity compared to 11C-PK11195 (54). Several studies assessed neuroinflammation with the measurement of radiotracer retention in AD patients through these advanced ligands (Table 2). This section will summarize the second-generation TSPO radiotracers widely used in AD or MCI. Elevated region-specific TSPO binding signals in a wide range of cortical areas are more illustrated in patients with AD than in normal individuals. The temporal pattern of neuroinflammation over the course of AD has also been well-characterized through longitudinal investigations (55, 60, 62). The role of neuroinflammation is revealed by a large longitudinal study with the administration of 18F-DPA-714 (60, 61). Participants with MCI and higher initial TSPO binding indicate a slower rate of decline due to dementia than those with lower initial TSPO binding. Combined with the other 11C-PK11195 studies, the proposal of a dual peak hypothesis of neuroinflammation in AD has been well-established (47). The hypothesis illustrated that neuroinflammation in the early phase of MCI patients is protective and beneficial to Aβ removal, while activated neuroinflammation in the later phase is detrimental. Notably, the results from TSPO imaging studies could reflect different PET signals to explore the association between TSPO expression and clinical outcomes. Similar to 11C-PK11195, enhanced TSPO signals are related to impairments in cognition and memory, visuospatial and language ability, executive functioning, dementia severity, and brain atrophy. Further research and clinical trials are urgently required when illustrating regional uptake patterns of TSPO ligands in MCI. Some studies indicated that striking patterns of high cortical tracer retention, especially in the temporal lobe, have been observed compared to healthy controls (47, 63, 65).

TABLE 2
www.frontiersin.org

Table 2 Studies examining regional brain uptake via advanced TSPO tracers in AD and MCI.

Nevertheless, it is necessary to understand the limitation of second-generation TSPO radiotracers, particularly sensitivity to the single-nucleotide polymorphism (SNP) of the TSPO gene. Genetic variation in the SNP rs6917 leads to different binding patterns in TSPO. Therefore, it is critical to evaluate these genetic polymorphisms when examining data from these radiotracers and exclude low-affinity binders (54, 67). Consequently, the advantage of 11C-PK11195 can be emphasized without any influence of TSPO polymorphism.

3.3 Other radiotracers targeting neuroinflammation

It is imperative to develop other novel and effective imaging radiotracers to measure neuroinflammation with higher specificity and affinity since TSPO is not specifically located in microglia. Therefore, promising targets should present precise localization in neuroinflammation and particular ligands to enable imaging measurement (6871). Further work is required to explore novel targets for the activated microglia and their ability to phagocytose Aβ. Transcriptional profiling of human microglia in plaque-associated and parenchymal tissues could decipher changes in whole tissue RNA, leading to novel targets identified and prioritized for further investigation (72). In the following section, we summarized the other neuroinflammation-targeted ligands for imaging neuroinflammation.

3.3.1 CSF1R PET imaging

Colony-stimulating factor 1 receptor (CSF1R) is dominantly expressed in the microglia, macrophages/monocytes, and dendritic cells in the brain parenchyma. CSF1R presents an essential role in microglia growth, proliferation, and survival. Previous studies have determined that the growth factors of colony stimulating factor-1 (GCSF1) and interleukin-34 (IL-34) are endogenous ligands toward CSF1R (73). It has also been demonstrated that the upregulation of CSF1R is responsible for injury and AD-related neuropathology (74, 75). Due to the role of CSF1R, a novel CSF1R-targeting radiotracer 11C-CPPC was developed. In animal acute inflammation models, the encephalomyelitis model of multiple sclerosis, and APPsi with cerebral Ab pathology, increased microglial levels of CSF1R have been captured through this radiotracer (76). Moreover, a recent study compared novel CSF1R tracers 11C-GW2580 with 11C-CPPC in detecting both acute inflammation induced by LPS injection and chronic inflammation in APP knockin mice. The results revealed that 11C-GW2580 signal changes in CSF1R showed higher sensitivity than 11C-CPPC, associated with increased TSPO pattern in the brain (77).

3.3.2 COX1 and COX2 PET imaging

Cyclooxygenase (COX) plays an essential role in the generation of prostaglandin H2, the substrate for prostaglandins and thromboxanes. There are two isoforms of COX, COX-1 and COX-2, determined to present a critical role in neuroinflammation and links to various neurodegenerative diseases, especially AD. According to the results of immunochemical evidence, COX-1 and COX-2 are located in both microglia and neuron in the CNS (78). Various radiotracers for COX-1 and COX-2 have been well established, such as 18F-TMI, 18F-triacoxib, 11C-rofecoxib, 11C-KTP-Me, 11C-PS13, and 11C-MC1 (7986). A few studies demonstrated that 11C-KTP-Me harbors a greater BBB entrance and selective sensitivity towards COX-1 (80, 86). Moreover, clinical trials with 11C-KTP-Me revealed an elevated brain signal in AD patients compared to normal individuals. 11C-KTP-Me accumulation can be seen in activated microglia surrounding Aβ plaques within the frontal cortex and hippocampus. Similar in vivo studies can also be observed in APPswe (Tg2576) mice compared to wild-type mice (80, 81). Additionally, previous studies illustrated that both 11C-PS1 (COX-1 PET imaging) and 11C-MC1 (COX-2 PET imaging) radiotracers showed specific detection patterns after LPS-induced neuroinflammation in monkey brain and human inflammatory tissues (82, 83).

3.3.3 P2X7R and P2Y12R PET imaging

The upregulated levels of the purinergic P2X7 receptor (P2X7R) can activate neuroinflammation, especially in M1 microglia. P2X7R had various biological functions, including inflammasome activation, cytokine secretion, T lymphocyte differentiation, and cell death (87). Microglia monitors through P2Y12R-dependent junctions are associated with mitochondrial activity in neurons (88). Brain injuries altered somatic junctions that induced P2Y12R-dependent neuroprotective effects through calcium load and functional connectivity in neurons (89, 90). Based on the immunohistochemical staining results, levels of P2Y12R were decreased in the brains of AD patients (91). Accumulative evidence has revealed that various P2X7R-targeting radiotracers have been developed currently, such as 11C-GSK1482160, 11C-JNJ-47965567, 18F-JNJ-64413739, 11C-JNJ-54173717, 11C-SMW139, and 18F-PTTP (9298). A previous study overexpressed human P2X7R in a rat model by rAAV3flag-hP2X7R and demonstrated that 11C-SMW139 had higher affinity and specificity to the P2X7R (99). Moreover, brains of AD patients revealed higher 11C-SMW139 binding compared to healthy individuals through autoradiography, consistent with histological staining results (99). An ongoing clinical trial applied 11C-SMW139 as a PET imaging radiotracer toward neuroinflammation in Parkinson’s disease (PRI-PD: 2018-000405-23). The other probes are P2Y12R-based radiotracers, including 11C-AZD1283, 11C-P2Y12R-ant, and 11C-5, assessed among animal models (94, 100, 101). A previous study has demonstrated that the P2Y12R radiotracer, 11C-AZD1283, distinctly responds to tau and amyloid deposits. The levels of P2Y12R binding increase in APP23 and APPNL-F/NL-F mice (101). However, 11C-AZD1283 PET imaging showed no signal in the wild-type mouse brain.

Additionally, two other radiotracers, 11C-P2Y12R and 11C-5, exerted sufficient brain uptake, high affinity, and promising results in experimental autoimmune encephalomyelitis and stroke models, detecting anti-inflammatory microglia (48) (94, 100).

3.3.4 MAO-B PET imaging

In several clinical trials, 11C-deuterium-L-deprenyl (DED) MAO-B inhibitors have also been applied in PET imaging toward neuroinflammation. It has been proved that early astrocytosis can be measured via 11C-DED in sporadic and autosomal dominant AD patients and amyloidosis mouse models (102110). In addition, 18F-fluorodeprenyl-D2 showed favorable kinetic properties and ameliorated affinity in MAO-B imaging (111). However, the technical problems of irreversible inhibitors impede the accuracy of imaging. Therefore, several reversible-binding inhibitors have been developed and validated, including 11C-Cou, 11C-SL25.1188, and 11C-SMBT-1 (69, 112, 113). Among the advanced PET imaging based on reversible-binding inhibitors, a specific elevated regional retention of 11C-SMBT-1 within the cortical and hippocampal regions can be seen in patients with AD compared to healthy individuals (114).

3.3.5 I2BS PET imaging

I2BS is located on both monoamine oxidases A (MAO-A) and B (MAO-B), which is another novel target for PET imaging toward neuroinflammation (115118). 11C-FTIMD presented the specific binging to I₂BS in the monkey brain (119). A previous study has revealed that the activated astroglia determined using 11C-BU99008 PET in the early period of Parkinson’s disease is responsible for α-synuclein accumulation (116). A previous in vitro study demonstrated that 3H-BU99008 revealed high specificity in brain tissues from AD patients and colocalized with glial fibrillary acidic protein staining of astrocytes (120). Moreover, a clinical study reported that an increasing 3H-BU99008 binding signal could be detected in the brain of patients with AD compared to healthy individuals. Similar results were reported in the cortical region via 11C-BU99008, consistent with the high cerebral Aβ load evaluated by 18F-florbetaben in MCI and AD patients (121). A previous study demonstrated that increased 11C-BU99008 signaling could be detected in earlier stages with low Aβ loads. At the same time, reduced astrocytosis can be observed in the advanced stages with a more significant Aβ load and atrophy (122).

3.3.6 CB2R PET imaging

Cannabinoid type 2 receptor (CB2R) is a member of the endogenous cannabinoid system. CB2R has a low concentration in the brain during healthy conditions. However, Aβ deposition activates microglia leading to the high expression of CB2R, another widely considered dominant marker of AD neuroinflammation. Various PET radiotracers with higher affinity towards CB2R have been well established. Among the developed CB2R imaging agents, 11C-NE40 can specifically and reversibly bind to CB2R. Previous studies have depicted that the uptake of 11C-NE40 was reduced in the brain of AD patients due to loss of neuron-based CB2R expression, contrary to the expectations from preclinical studies. This inconsistency may be due to the low expression of CB2R and insufficient selectivity for CB2R and CB1R (123). The other CB2R agonists with high affinity are under exploration, including 11C-MA2, 18F-MA3, or 18F-RS126 (124).

3.3.7 11C-AA PET imaging

AA, a kind of n-6 polyunsaturated fatty acid and an essential component of the metabolic network of inflammation, is abundant in the brain parenchyma and participates in cell signal transduction. Microglia in the CNS release the inflammatory cytokines and bind to receptors on the surface of astrocytes, leading to the secretion of phospholipase and cytoplasmic phosphatase and mediated AA release. Therefore, AA detection can serve as an indirect marker of microglial activation (125). Based on the above results, 11C-AA and PET imaging can determine brain phospholipase activity. By injecting 11C-AA, the regional brain incorporation coefficients and metabolic loss of AA in the brain can be determined (125). Therefore, elevated signals of 11C-AA could evaluate the disordered metabolism of AA due to neuroinflammation.

3.3.8 Nicotinic acetylcholine receptors

It has been demonstrated that nicotinic acetylcholine receptors (nAChRs) were closely related to neuroinflammation. Moreover, the ligand 2-18F-A85380 (2-FA) towards nAChR has also presented similar patterns of uptake with 11C-PK11195 in activated microglia and astrocytes (126). Additionally, 18F-flubatine has been established with more favorable kinetic profile, which leads to a better understanding of nAChRs in neuroinflammation (126, 127). The homomeric nAChRs are colocalized in neuritic plaques in patients’ brain with AD, and Aβ1–42 has been reported to bind to the α7 nAChRs with high affinity (128). α7 nAChRs are also strongly expressed on astrocytes and microglia, the activation of which has been shown to suppress inflammatory processes (128). Recently, several new compound-based bio-tracers, such as 18F-ASEM and 18F-DBT-10, presented more promising results (129).

4 Methodological issues in radiotracers quantification by PET

A variety of quantification methods have been established due to the tracers. It appears that it is a great challenge for quantification by PET and that various factors should be taken into account. (1) Genetic polymorphism and affinity: The main limitation of the second-generation TSPO radiotracers is their sensitivity to a polymorphism of the TSPO gene. This polymorphism leads to differential affinity of these ligands to TSPO (67). (2) Arterial plasma input function: The kinetics model analyses are based on the input function, which requires the relative traumatic placement of an arterial catheter and the development of radioanalytical methods to accurately identify the plasma metabolite fractions (56). (3) Reference region definition: The reference region should be devoid of specific ligand binding to the target and only share the same free and non-specific binding with the region expressing the target and remain unaffected by the disease.

5 Conclusion

The interplay between amyloid, tau, and neuroinflammation is a brand new area of investigation that has only recently become possible through the development of an expanded repertoire of PET tracers. Therefore, we reviewed various strategies for PET imaging for neuroinflammation and summarized the dominant results from previous clinical trials on AD patients. There are a variety of approaches explaining the exploration of neuroinflammation. Although determining neuroinflammation by PET imaging in AD is widely used, there is a conflict on whether neuroinflammation is beneficial or detrimental to the evolution of the symptoms, neuronal injury, and cognitive deficits. This review also highlighted are a number of areas of uncertainty and various radiotracers’ limitations. The challenges of radiotracer establishment and accurate binding quantification, the complicated neuroinflammation in the brain tissue of AD patients, and its extensive roles in different stages have also been emphasized. PET imaging towards neuroinflammation is a potential approach to deciphering the pathophysiological process of AD patients. It clarifies the links between amyloid/tau pathologies and neuroinflammation, influencing different clinical symptoms and pathophysiological progression. More efforts are also required to improve the approaches to determine the binding signal from the PET imaging based on the current radiotracers and develop novel radiotracers. Moreover, deciding on a consensus to standardize the PET data analysis is critical. Given the proposal that the role of neuroinflammation in AD pathogenesis changes over the disease course, this is a prerequisite to conducting multi-tracer longitudinal studies based on multiple centers and elucidating the multifaced role of neuroinflammation in AD and MCI patients.

Author contributions

The author confirms being the sole contributor of this work and has approved it for publication.

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Wang W, Zhou Q, Jiang T, Li S, Ye J, Zheng J, et al. A novel small-molecule PROTAC selectively promotes tau clearance to improve cognitive functions in Alzheimer-like models. Theranostics (2021) 11:5279–95. doi: 10.7150/thno.55680

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Chen ML, Hong CG, Yue T, Li HM, Duan R, Hu WB, et al. Inhibition of miR-331-3p and miR-9-5p ameliorates alzheimer’s disease by enhancing autophagy. Theranostics (2021) 11:2395–409. doi: 10.7150/thno.47408

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Blackman J, Swirski M, Clynes J, Harding S, Leng Y, Coulthard E. Pharmacological and non-pharmacological interventions to enhance sleep in mild cognitive impairment and mild alzheimer’s disease: A systematic review. J Sleep Res (2021) 30:e13229. doi: 10.1111/jsr.13229

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Zhang L, Chopp M, Jiang Q, Zhang Z. Role of the glymphatic system in ageing and diabetes mellitus impaired cognitive function. Stroke Vasc Neurol (2019) 4:90–2. doi: 10.1136/svn-2018-000203

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Qu Y, Ma YH, Huang YY, Ou YN, Shen XN, Chen SD, et al. Blood biomarkers for the diagnosis of amnestic mild cognitive impairment and alzheimer’s disease: A systematic review and meta-analysis. Neurosci Biobehav Rev (2021) 128:479–86. doi: 10.1016/j.neubiorev.2021.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Tolar M, Abushakra S, Sabbagh M. The path forward in alzheimer’s disease therapeutics: Reevaluating the amyloid cascade hypothesis. Alzheimers Dement (2020) 16:1553–60. doi: 10.1016/j.jalz.2019.09.075

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat Rev Neurol (2021) 17:157–72. doi: 10.1038/s41582-020-00435-y

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Masuoka N, Yoshimine C, Hori M, Tanaka M, Asada T, Abe K, et al. Effects of Anserine/Carnosine supplementation on mild cognitive impairment with APOE4. Nutrients (2019) 11:7. doi: 10.3390/nu11071626

CrossRef Full Text | Google Scholar

9. Bloom GS. Amyloid-β and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol (2014) 71:505–8. doi: 10.1001/jamaneurol.2013.5847

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Maddula M, Sprigg N, Bath PM, Munshi S. Cerebral misery perfusion due to carotid occlusive disease. Stroke Vasc Neurol (2017) 2:88–93. doi: 10.1136/svn-2017-000067

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Spence JD, Song H, Cheng G. Appropriate management of asymptomatic carotid stenosis. Stroke Vasc Neurol (2016) 1:64–71. doi: 10.1136/svn-2016-000016

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Chandra A, Valkimadi PE, Pagano G, Cousins O, Dervenoulas G, Politis M. Applications of amyloid, tau, and neuroinflammation PET imaging to alzheimer’s disease and mild cognitive impairment. Hum Brain Mapp (2019) 40:5424–42. doi: 10.1002/hbm.24782

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Zhang M, Qian C, Zheng ZG, Qian F, Wang Y, Thu PM, et al. Jujuboside a promotes aβ clearance and ameliorates cognitive deficiency in alzheimer’s disease through activating Axl/HSP90/PPARγ pathway. Theranostics (2018) 8:4262–78. doi: 10.7150/thno.26164

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell (2010) 140:918–34. doi: 10.1016/j.cell.2010.02.016

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Heppner FL, Ransohoff RM, Becher B. Immune attack: The role of inflammation in Alzheimer disease. Nat Rev Neurosci (2015) 16:358–72. doi: 10.1038/nrn3880

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Liu H, Leak RK, Hu X. Neurotransmitter receptors on microglia. Stroke Vasc Neurol (2016) 1:52–8. doi: 10.1136/svn-2016-000012

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Zhang A, Xu Y, Xu H, Ren J, Meng T, Ni Y, et al. Lactate-induced M2 polarization of tumor-associated macrophages promotes the invasion of pituitary adenoma by secreting CCL17. Theranostics (2021) 11:3839–52. doi: 10.7150/thno.53749

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Liang C, Zou T, Zhang M, Fan W, Zhang T, Jiang Y, et al. MicroRNA-146a switches microglial phenotypes to resist the pathological processes and cognitive degradation of alzheimer’s disease. Theranostics (2021) 11:4103–21. doi: 10.7150/thno.53418

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Chun H, Im H, Kang YJ, Kim Y, Shin JH, Won W, et al. Severe reactive astrocytes precipitate pathological hallmarks of alzheimer’s disease. Via H(2)O(2)(-) Production Nat Neurosci (2020) 23:1555–66. doi: 10.1038/s41593-020-00735-y

CrossRef Full Text | Google Scholar

20. Habib N, McCabe C, Medina S, Varshavsky M, Kitsberg D, Dvir-Szternfeld R, et al. Disease-associated astrocytes in alzheimer’s disease and aging. Nat Neurosci (2020) 23:701–6. doi: 10.1038/s41593-020-0624-8

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Li T, Liu T, Chen X, Li L, Feng M, Zhang Y, et al. Microglia induce the transformation of A1/A2 reactive astrocytes via the CXCR7/PI3K/Akt pathway in chronic post-surgical pain. J Neuroinflamm (2020) 17:211. doi: 10.1186/s12974-020-01891-5

CrossRef Full Text | Google Scholar

22. 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: 10.1038/nature21029

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Escartin C, Galea E, Lakatos A, O'Callaghan JP, Petzold GC, Serrano-Pozo A, et al. Reactive astrocyte nomenclature, definitions, and future directions. Nat Neurosci (2021) 24:312–25. doi: 10.1038/s41593-020-00783-4

PubMed Abstract | CrossRef Full Text | Google Scholar

24. James ML, Belichenko NP, Shuhendler AJ, Hoehne A, Andrews LE, Condon C, et al. [(18)F]GE-180 PET detects reduced microglia activation after LM11A-31 therapy in a mouse model of alzheimer’s disease. Theranostics (2017) 7:1422–36. doi: 10.7150/thno.17666

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Banati RB, Newcombe J, Gunn RN, Cagnin A, Turkheimer F, Heppner F, et al. The peripheral benzodiazepine binding site in the brain in multiple sclerosis: Quantitative in vivo imaging of microglia as a measure of disease activity. Brain: J Neurol (2000) 123(Pt11):2321–37. doi: 10.1093/brain/123.11.2321

CrossRef Full Text | Google Scholar

26. Belloli S, Moresco RM, Matarrese M, Biella G, Sanvito F, Simonelli P, et al. Evaluation of three quinoline-carboxamide derivatives as potential radioligands for the in vivo pet imaging of neurodegeneration. Neurochem Int (2004) 44:433–40. doi: 10.1016/j.neuint.2003.08.006

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Lockhart A, Davis B, Matthews JC, Rahmoune H, Hong G, Gee A, et al. The peripheral benzodiazepine receptor ligand PK11195 binds with high affinity to the acute phase reactant alpha1-acid glycoprotein: Implications for the use of the ligand as a CNS inflammatory marker. Nucl Med Biol (2003) 30:199–206. doi: 10.1016/s0969-8051(02)00410-9

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Ching AS, Kuhnast B, Damont A, Roeda D, Tavitian B, Dollé F. Current paradigm of the 18-kDa translocator protein (TSPO) as a molecular target for PET imaging in neuroinflammation and neurodegenerative diseases. Insights Imaging (2012) 3:111–9. doi: 10.1007/s13244-011-0128-x

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Kreisl WC, Fujita M, Fujimura Y, Kimura N, Jenko KJ, Kannan P, et al. Comparison of [(11)C]-(R)-PK 11195 and [(11)C]PBR28, two radioligands for translocator protein (18 kDa) in human and monkey: Implications for positron emission tomographic imaging of this inflammation biomarker. Neuroimage (2010) 49:2924–32. doi: 10.1016/j.neuroimage.2009.11.056

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Endres CJ, Pomper MG, James M, Uzuner O, Hammoud DA, Watkins CC, et al. Initial evaluation of 11C-DPA-713, a novel TSPO PET ligand, in humans. J Nucl Med (2009) 50:1276–82. doi: 10.2967/jnumed.109.062265

PubMed Abstract | CrossRef Full Text | Google Scholar

31. 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 [(11)C]DPA713 versus [(11)C](R)PK11195. J Cereb Blood Flow Metab (2017) 37:877–89. doi: 10.1177/0271678x16646788

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Chauveau F, Van Camp N, Dollé F, Kuhnast B, Hinnen F, Damont A, et al. Comparative evaluation of the translocator protein radioligands 11C-DPA-713, 18F-DPA-714, and 11C-PK11195 in a rat model of acute neuroinflammation. J Nucl Med (2009) 50:468–76. doi: 10.2967/jnumed.108.058669

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Gulyás B, Makkai B, Kása P, Gulya K, Bakota L, Várszegi S, et al. A comparative autoradiography study in post mortem whole hemisphere human brain slices taken from Alzheimer patients and age-matched controls using two radiolabelled DAA1106 analogues with high affinity to the peripheral benzodiazepine receptor (PBR) system. Neurochem Int (2009) 54:28–36. doi: 10.1016/j.neuint.2008.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Venneti S, Wang G, Nguyen J, Wiley CA. The positron emission tomography ligand DAA1106 binds with high affinity to activated microglia in human neurological disorders. J Neuropathol Exp Neurol (2008) 67:1001–10. doi: 10.1097/NEN.0b013e318188b204

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Varrone A, Mattsson P, Forsberg A, Takano A, Nag S, Gulyás B, et al. In vivo imaging of the 18-kDa translocator protein (TSPO) with [18F]FEDAA1106 and PET does not show increased binding in alzheimer’s disease patients. Eur J Nucl Med Mol Imaging (2013) 40:921–31. doi: 10.1007/s00259-013-2359-1

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Varrone A, Oikonen V, Forsberg A, Joutsa J, Takano A, Solin O, et al. Positron emission tomography imaging of the 18-kDa translocator protein (TSPO) with [18F]FEMPA in alzheimer’s disease patients and control subjects. Eur J Nucl Med Mol Imaging (2015) 42:438–46. doi: 10.1007/s00259-014-2955-8

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Rusjan PM, Wilson AA, Bloomfield PM, Vitcu I, Meyer JH, Houle S, et al. Quantitation of translocator protein binding in human brain with the novel radioligand [18F]-FEPPA and positron emission tomography. J Cereb Blood Flow Metab (2011) 31:1807–16. doi: 10.1038/jcbfm.2011.55

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer FE, et al. et al. in-vivo measurement of activated microglia in dementia. Lancet (London England) (2001) 358:461–7. doi: 10.1016/s0140-6736(01)05625-2

CrossRef Full Text | Google Scholar

39. Edison P, Archer HA, Gerhard A, Hinz R, Pavese N, Turkheimer FE, et al. Microglia, amyloid, and cognition in alzheimer’s disease: An [11C](R)PK11195-PET and [11C]PIB-PET study. Neurobiol Dis (2008) 32:412–9. doi: 10.1016/j.nbd.2008.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Wiley CA, Lopresti BJ, Venneti S, Price J, Klunk WE, DeKosky ST, et al. Carbon 11-labeled Pittsburgh compound b and carbon 11-labeled (R)-PK11195 positron emission tomographic imaging in Alzheimer disease. Arch Neurol (2009) 66:60–7. doi: 10.1001/archneurol.2008.511

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Okello A, Koivunen J, Edison P, Archer HA, Turkheimer FE, Någren K, et al. Conversion of amyloid positive and negative MCI to AD over 3 years: an 11C-PIB PET study. Neurology (2009) 73:754–60. doi: 10.1212/WNL.0b013e3181b23564

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Yokokura M, Mori N, Yagi S, Yoshikawa E, Kikuchi M, Yoshihara Y, et al. In vivo changes in microglial activation and amyloid deposits in brain regions with hypometabolism in alzheimer’s disease. Eur J Nucl Med Mol Imaging (2011) 38:343–51. doi: 10.1007/s00259-010-1612-0

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Schuitemaker A, Kropholler MA, Boellaard R, van der Flier WM, Kloet RW, van der Doef TF, et al. Microglial activation in alzheimer’s disease: An (R)-[¹¹C]PK11195 positron emission tomography study. Neurobiol Aging (2013) 34:128–36. doi: 10.1016/j.neurobiolaging.2012.04.021

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Fan Z, Okello AA, Brooks DJ, Edison P. Longitudinal influence of microglial activation and amyloid on neuronal function in alzheimer’s disease. Brain: J Neurol (2015) 138:3685–98. doi: 10.1093/brain/awv288

CrossRef Full Text | Google Scholar

45. Fan Z, Aman Y, Ahmed I, Chetelat G, Landeau B, Ray Chaudhuri K, et al. Influence of microglial activation on neuronal function in alzheimer’s and parkinson’s disease dementia. Alzheimers Dement (2015) 11:608–621.e607. doi: 10.1016/j.jalz.2014.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Femminella GD, Ninan S, Atkinson R, Fan Z, Brooks DJ, Edison P, et al. Does microglial activation influence hippocampal volume and neuronal function in alzheimer’s disease and parkinson’s disease dementia? J Alzheimers Dis (2016) 51:1275–89. doi: 10.3233/jad-150827

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Fan Z, Brooks DJ, Okello A, Edison P. An early and late peak in microglial activation in alzheimer’s disease trajectory. Brain: J Neurol (2017) 140:792–803. doi: 10.1093/brain/aww349

CrossRef Full Text | Google Scholar

48. Parbo P, Ismail R, Hansen KV, Amidi A, Mårup FH, Gottrup H, et al. Brain inflammation accompanies amyloid in the majority of mild cognitive impairment cases due to alzheimer’s disease. Brain: J Neurol (2017) 140:2002–11. doi: 10.1093/brain/awx120

CrossRef Full Text | Google Scholar

49. Passamonti L, Rodríguez PV, Hong YT, Allinson KSJ, Bevan-Jones WR, Williamson D, et al. [(11)C]PK11195 binding in Alzheimer disease and progressive supranuclear palsy. Neurology (2018) 90:e1989–96. doi: 10.1212/wnl.0000000000005610

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Parbo P, Ismail R, Sommerauer M, Stokholm MG, Hansen AK, Hansen KV, et al. Does inflammation precede tau aggregation in early alzheimer’s disease? A PET study. Neurobiol Dis (2018) 117:211–6. doi: 10.1016/j.nbd.2018.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Groom GN, Junck L, Foster NL, Frey KA, Kuhl DE. PET of peripheral benzodiazepine binding sites in the microgliosis of alzheimer’s disease. J Nucl Med (1995) 36:2207–10. doi: 10.1093/brain/awy079

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Versijpt JJ, Dumont F, Van Laere KJ, Decoo D, Santens P, Audenaert K, et al. Assessment of neuroinflammation and microglial activation in alzheimer’s disease with radiolabelled PK11195 and single photon emission computed tomography. A pilot study. Eur Neurol (2003) 50:39–47. doi: 10.1159/000070857

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Okello A, Edison P, Archer HA, Turkheimer FE, Kennedy J, Bullock R, et al. Microglial activation and amyloid deposition in mild cognitive impairment: a PET study. Neurology (2009) 72:56–62. doi: 10.1212/01.wnl.0000338622.27876.0d

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Edison P, Brooks DJ. Role of neuroinflammation in the trajectory of alzheimer’s disease and in vivo quantification using PET. J Alzheimers Dis (2018) 64:S339–s351. doi: 10.3233/jad-179929

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Kreisl WC, Lyoo CH, McGwier M, Snow J, Jenko KJ, Kimura N, et al. In vivo radioligand binding to translocator protein correlates with severity of alzheimer’s disease. Brain: J Neurol (2013) 136:2228–38. doi: 10.1093/brain/awt145

CrossRef Full Text | Google Scholar

56. Lyoo CH, Ikawa M, Liow JS, Zoghbi SS, Morse CL, Pike VW, et al. Cerebellum can serve as a pseudo-reference region in Alzheimer disease to detect neuroinflammation measured with PET radioligand binding to translocator protein. J Nucl Med (2015) 56:701–6. doi: 10.2967/jnumed.114.146027

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Kreisl WC, Lyoo CH, Liow JS, Wei M, Snow J, Page E, et al. (11)C-PBR28 binding to translocator protein increases with progression of alzheimer’s disease. Neurobiol Aging (2016) 44:53–61. doi: 10.1016/j.neurobiolaging.2016.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Fan Z, Dani M, Femminella GD, Wood M, Calsolaro V, Veronese M, et al. Parametric mapping using spectral analysis for (11)C-PBR28 PET reveals neuroinflammation in mild cognitive impairment subjects. Eur J Nucl Med Mol Imaging (2018) 45:1432–41. doi: 10.1007/s00259-018-3984-5

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Dani M, Wood M, Mizoguchi R, Fan Z, Walker Z, Morgan R, et al. Microglial activation correlates in vivo with both tau and amyloid in alzheimer’s disease. Brain: J Neurol (2018) 141:2740–54. doi: 10.1093/brain/awy188

CrossRef Full Text | Google Scholar

60. Hamelin L, Lagarde J, Dorothée G, Leroy C, Labit M, Comley RA, et al. Early and protective microglial activation in alzheimer’s disease: a prospective study using 18F-DPA-714 PET imaging. Brain: J Neurol (2016) 139:1252–64. doi: 10.1093/brain/aww017

CrossRef Full Text | Google Scholar

61. Hamelin L, Lagarde J, Dorothée G, Potier MC, Corlier F, Kuhnast B, et al. Distinct dynamic profiles of microglial activation are associated with progression of alzheimer’s disease. Brain: J Neurol (2018) 141:1855–70. doi: 10.1093/brain/awy079

CrossRef Full Text | Google Scholar

62. Yasuno F, Ota M, Kosaka J, Ito H, Higuchi M, Doronbekov TK, et al. Increased binding of peripheral benzodiazepine receptor in alzheimer’s disease measured by positron emission tomography with [11C]DAA1106. Biol Psychiatry (2008) 64:835–41. doi: 10.1016/j.biopsych.2008.04.021

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Yasuno F, Kosaka J, Ota M, Higuchi M, Ito H, Fujimura Y, et al. Increased binding of peripheral benzodiazepine receptor in mild cognitive impairment-dementia converters measured by positron emission tomography with [¹¹C]DAA1106. Psychiatry Res (2012) 203:67–74. doi: 10.1016/j.pscychresns.2011.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Suridjan I, Pollock BG, Verhoeff NP, Voineskos AN, Chow T, Rusjan PM, et al. In-vivo imaging of grey and white matter neuroinflammation in alzheimer’s disease: A positron emission tomography study with a novel radioligand, [18F]-FEPPA. Mol Psychiatry (2015) 20:1579–87. doi: 10.1038/mp.2015.1

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Knezevic D, Verhoeff NPL, Hafizi S, Strafella AP, Graff-Guerrero A, Rajji T, et al. Imaging microglial activation and amyloid burden in amnestic mild cognitive impairment. J Cereb Blood Flow Metab (2018) 38:1885–95. doi: 10.1177/0271678x17741395

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Vettermann FJ, Harris S, Schmitt J, Unterrainer M, Lindner S, Rauchmann BS, et al. Impact of TSPO receptor polymorphism on [(18)F]GE-180 binding in healthy brain and pseudo-reference regions of neurooncological and neurodegenerative disorders. Life (Basel) (2021) 11(6):3592–5. doi: 10.3390/life11060484

CrossRef Full Text | Google Scholar

67. Yoder KK, Nho K, Risacher SL, Kim S, Shen L, Saykin AJ. Influence of TSPO genotype on 11C-PBR28 standardized uptake values. J Nucl Med (2013) 54:1320–2. doi: 10.2967/jnumed.112.118885

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Kreisl WC, Kim MJ, Coughlin JM, Henter ID, Owen DR, Innis RB. PET imaging of neuroinflammation in neurological disorders. Lancet Neurol (2020) 19:940–50. doi: 10.1016/s1474-4422(20)30346-x

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Narayanaswami V, Drake LR, Brooks AF, Meyer JH, Houle S, Kilbourn MR, et al. Classics in neuroimaging: Development of PET tracers for imaging monoamine oxidases. ACS Chem Neurosci (2019) 10:1867–71. doi: 10.1021/acschemneuro.9b00081

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Janssen B, Mach RH. Development of brain PET imaging agents: Strategies for imaging neuroinflammation in alzheimer’s disease. Prog Mol Biol Transl Sci (2019) 165:371–99. doi: 10.1016/bs.pmbts.2019.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Janssen B, Vugts DJ, Windhorst AD, Mach RH. PET imaging of microglial activation-beyond targeting TSPO. Molecules (2018) 23(3):251–61. doi: 10.3390/molecules23030607

CrossRef Full Text | Google Scholar

72. Srinivasan K, Friedman BA, Larson JL, Lauffer BE, Goldstein LD, Appling LL, et al. Untangling the brain’s neuroinflammatory and neurodegenerative transcriptional responses. Nat Commun (2016) 7:11295. doi: 10.1038/ncomms11295

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Lin H, Lee E, Hestir K, Leo C, Huang M, Bosch E, et al. Discovery of a cytokine and its receptor by functional screening of the extracellular proteome. Science (2008) 320:807–11. doi: 10.1126/science.1154370

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Hamilton JA. Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol (2008) 8:533–44. doi: 10.1038/nri2356

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Murphy MC, Curran GL, Glaser KJ, Rossman PJ, Huston J 3rd, Poduslo JF, et al. Magnetic resonance elastography of the brain in a mouse model of alzheimer’s disease: initial results. Magn Reson Imaging (2012) 30:535–9. doi: 10.1016/j.mri.2011.12.019

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Horti AG, Naik R, Foss CA, Minn I, Misheneva V, Du Y, et al. PET imaging of microglia by targeting macrophage colony-stimulating factor 1 receptor (CSF1R). Proc Natl Acad Sci USA (2019) 116:1686–91. doi: 10.1073/pnas.1812155116

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Zhou X, Ji B, Seki C, Nagai Y, Minamimoto T, Fujinaga M, et al. PET imaging of colony-stimulating factor 1 receptor: A head-to-head comparison of a novel radioligand, (11)C-GW2580, and (11)C-CPPC, in mouse models of acute and chronic neuroinflammation and a rhesus monkey. J Cereb Blood Flow Metab (2021) 41:2410–22. doi: 10.1177/0271678x211004146

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Choi SH, Aid S, Bosetti F. The distinct roles of cyclooxygenase-1 and -2 in neuroinflammation: Implications for translational research. Trends Pharmacol Sci (2009) 30:174–81. doi: 10.1016/j.tips.2009.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Kumar JSD, Zanderigo F, Prabhakaran J, Rubin-Falcone H, Parsey RV, Mann JJ. In vivo evaluation of [(11)C]TMI, a COX-2 selective PET tracer, in baboons. Bioorg Med Chem Lett (2018) 28:3592–5. doi: 10.1016/j.bmcl.2018.10.049

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Ohnishi A, Senda M, Yamane T, Sasaki M, Mikami T, Nishio T, et al. Human whole-body biodistribution and dosimetry of a new PET tracer, [(11)C]ketoprofen methyl ester, for imagings of neuroinflammation. Nucl Med Biol (2014) 41:594–9. doi: 10.1016/j.nucmedbio.2014.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Shukuri M, Mawatari A, Ohno M, Suzuki M, Doi H, Watanabe Y, et al. Detection of cyclooxygenase-1 in activated microglia during amyloid plaque progression: PET studies in alzheimer’s disease model mice. J Nucl Med (2016) 57:291–6. doi: 10.2967/jnumed.115.166116

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Shrestha S, Singh P, Cortes-Salva MY, Jenko KJ, Ikawa M, Kim MJ, 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 [(11)C]PS13 for quantitative imaging. ACS Chem Neurosci (2018) 9:2620–7. doi: 10.1021/acschemneuro.8b00103

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Shrestha S, Kim MJ, Eldridge M, Lehmann ML, Frankland M, Liow JS, 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: 10.1186/s12974-020-01804-6

CrossRef Full Text | Google Scholar

84. Litchfield M, Wuest M, Glubrecht D, Wuest F. Radiosynthesis and biological evaluation of [(18)F]Triacoxib: A new radiotracer for PET imaging of COX-2. Mol Pharm (2020) 17:251–61. doi: 10.1021/acs.molpharmaceut.9b00986

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Ji B, Kumata K, Onoe H, Kaneko H, Zhang MR, Seki C, et al. Assessment of radioligands for PET imaging of cyclooxygenase-2 in an ischemic neuronal injury model. Brain Res (2013) 1533:152–62. doi: 10.1016/j.brainres.2013.08.026

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Shukuri M, Takashima-Hirano M, Tokuda K, Takashima T, Matsumura K, Inoue O, et al. In vivo expression of cyclooxygenase-1 in activated microglia and macrophages during neuroinflammation visualized by PET with 11C-ketoprofen methyl ester. J Nucl Med (2011) 52:1094–101. doi: 10.2967/jnumed.110.084046

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Di Virgilio F, Dal Ben D, Sarti AC, Giuliani AL, Falzoni S. The P2X7 receptor in infection and inflammation. Immunity (2017) 47:15–31. doi: 10.1016/j.immuni.2017.06.020

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Cserép C, Pósfai B, Lénárt N, Fekete R, László ZI, Lele Z, et al. Microglia monitor and protect neuronal function through specialized somatic purinergic junctions. Science (2020) 367:528–37. doi: 10.1126/science.aax6752

PubMed Abstract | CrossRef Full Text | Google Scholar

89. 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: J Neurol (2017) 140:1900–13. doi: 10.1093/brain/awx113

CrossRef Full Text | Google Scholar

90. Badimon A, Strasburger HJ, Ayata P, Chen X, Nair A, Ikegami A, et al. Negative feedback control of neuronal activity by microglia. Nature (2020) 586:417–23. doi: 10.1038/s41586-020-2777-8

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Mildner A, Huang H, Radke J, Stenzel W, Priller J. P2Y(12) receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia (2017) 65:375–87. doi: 10.1002/glia.23097

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Han J, Liu H, Liu C, Jin H, Perlmutter JS, Egan TM, et al. Pharmacologic characterizations of a P2X7 receptor-specific radioligand, [11C]GSK1482160 for neuroinflammatory response. Nucl Med Commun (2017) 38:372–82. doi: 10.1097/mnm.0000000000000660

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Territo PR, Meyer JA, Peters JS, Riley AA, McCarthy BP, Gao M, et al. Characterization of (11)C-GSK1482160 for targeting the P2X7 receptor as a biomarker for neuroinflammation. J Nucl Med (2017) 58:458–65. doi: 10.2967/jnumed.116.181354

PubMed Abstract | CrossRef Full Text | Google Scholar

94. 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: 10.1186/s12974-017-1034-z

CrossRef Full Text | Google Scholar

95. Koole M, Schmidt ME, Hijzen A, Ravenstijn P, Vandermeulen C, Van Weehaeghe D, et al. (18)F-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: 10.2967/jnumed.118.216747

PubMed Abstract | CrossRef Full Text | Google Scholar

96. 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: 10.2967/jnumed.119.231985

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Beaino W, Janssen B, Kooijman E, Vos R, Schuit RC, O'Brien-Brown J, et al. PET imaging of P2X(7)R in the experimental autoimmune encephalomyelitis model of multiple sclerosis using [(11)C]SMW139. J Neuroinflamm (2020) 17:300. doi: 10.1186/s12974-020-01962-7

CrossRef Full Text | Google Scholar

98. Fu Z, Lin Q, Hu B, Zhang Y, Chen W, Zhu J, et al. P2X7 PET radioligand (18)F-PTTP for differentiation of lung tumor from inflammation. J Nucl Med (2019) 60:930–6. doi: 10.2967/jnumed.118.222547

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Janssen B, Vugts DJ, Wilkinson SM, Ory D, Chalon S, Hoozemans JJM, et al. Identification of the allosteric P2X(7) receptor antagonist [(11)C]SMW139 as a PET tracer of microglial activation. Sci Rep (2018) 8:6580. doi: 10.1038/s41598-018-24814-0

PubMed Abstract | CrossRef Full Text | Google Scholar

100. 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: 10.7150/thno.25572

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Maeda J, Minamihisamatsu T, Shimojo M, Zhou X, Ono M, Matsuba Y, et al. Distinct microglial response against alzheimer’s amyloid and tau pathologies characterized by P2Y12 receptor. Brain Commun (2021) 3:fcab011. doi: 10.1093/braincomms/fcab011

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Ni R, Röjdner J, Voytenko L, Dyrks T, Thiele A, Marutle A, et al. In vitro characterization of the regional binding distribution of amyloid PET tracer florbetaben and the glia tracers deprenyl and PK11195 in autopsy alzheimer’s brain tissue. J Alzheimers Dis (2021) 80:1723–37. doi: 10.3233/jad-201344

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Vilaplana E, Rodriguez-Vieitez E, Ferreira D, Montal V, Almkvist O, Wall A, et al. Cortical microstructural correlates of astrocytosis in autosomal-dominant Alzheimer disease. Neurology (2020) 94:e2026–36. doi: 10.1212/wnl.0000000000009405

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Lemoine L, Gillberg PG, Bogdanovic N, Nennesmo I, Saint-Aubert L, Viitanen M, et al. Amyloid, tau, and astrocyte pathology in autosomal-dominant alzheimer’s disease variants: AβPParc and PSEN1DE9. Mol Psychiatry (2021) 26:5609–19. doi: 10.1038/s41380-020-0817-2

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Marutle A, Gillberg PG, Bergfors A, Yu W, Ni R, Nennesmo I, et al. ³H-deprenyl and ³H-PIB autoradiography show different laminar distributions of astroglia and fibrillar β-amyloid in Alzheimer brain. J Neuroinflamm (2013) 10:90. doi: 10.1186/1742-2094-10-90

CrossRef Full Text | Google Scholar

106. Carter SF, Chiotis K, Nordberg A, Rodriguez-Vieitez E. Longitudinal association between astrocyte function and glucose metabolism in autosomal dominant alzheimer’s disease. Eur J Nucl Med Mol Imaging (2019) 46:348–56. doi: 10.1007/s00259-018-4217-7

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Rodriguez-Vieitez E, Carter SF, Chiotis K, Saint-Aubert L, Leuzy A, Schöll M, et al. Comparison of early-phase 11C-Deuterium-l-Deprenyl and 11C-Pittsburgh compound b PET for assessing brain perfusion in Alzheimer disease. J Nucl Med (2016) 57:1071–7. doi: 10.2967/jnumed.115.168732

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Schöll M, Carter SF, Westman E, Rodriguez-Vieitez E, Almkvist O, Thordardottir S, et al. Early astrocytosis in autosomal dominant alzheimer’s disease measured in vivo by multi-tracer positron emission tomography. Sci Rep (2015) 5:16404. doi: 10.1038/srep16404

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Rodriguez-Vieitez E, Ni R, Gulyás B, Tóth M, Häggkvist J, Halldin C, et al. Astrocytosis precedes amyloid plaque deposition in Alzheimer APPswe transgenic mouse brain: A correlative positron emission tomography and in vitro imaging study. Eur J Nucl Med Mol Imaging (2015) 42:1119–32. doi: 10.1007/s00259-015-3047-0

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Olsen M, Aguilar X, Sehlin D, Fang XT, Antoni G, Erlandsson A, et al. Astroglial responses to amyloid-beta progression in a mouse model of alzheimer’s disease. Mol Imaging Biol (2018) 20:605–14. doi: 10.1007/s11307-017-1153-z

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Nag S, Fazio P, Lehmann L, Kettschau G, Heinrich T, Thiele A, et al. In vivo and In vitro characterization of a novel MAO-b inhibitor radioligand, 18F-labeled deuterated fluorodeprenyl. J Nucl Med (2016) 57:315–20. doi: 10.2967/jnumed.115.161083

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Moriguchi S, Wilson AA, Miler L, Rusjan PM, Vasdev N, Kish SJ, et al. Monoamine oxidase b total distribution volume in the prefrontal cortex of major depressive disorder: An [11C]SL25.1188 positron emission tomography study. JAMA Psychiatry (2019) 76:634–41. doi: 10.1001/jamapsychiatry.2019.0044

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Drake LR, Brooks AF, Mufarreh AJ, Pham JM, Koeppe RA, Shao X, et al. Deuterium kinetic isotope effect studies of a potential in vivo metabolic trapping agent for monoamine oxidase b. ACS Chem Neurosci (2018) 9:3024–7. doi: 10.1021/acschemneuro.8b00219

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Harada R, Hayakawa Y, Ezura M, Lerdsirisuk P, Du Y, Ishikawa Y, et al. (18)F-SMBT-1: A selective and reversible PET tracer for monoamine oxidase-b imaging. J Nucl Med (2021) 62:253–8. doi: 10.2967/jnumed.120.244400

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Kumar A, Koistinen NA, Malarte ML, Nennesmo I, Ingelsson M, Ghetti B, et al. Astroglial tracer BU99008 detects multiple binding sites in alzheimer’s disease brain. Mol Psychiatry (2021) 26:5833–47. doi: 10.1038/s41380-021-01101-5

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Wilson H, Dervenoulas G, Pagano G, Tyacke RJ, Polychronis S, Myers J, et al. Imidazoline 2 binding sites reflecting astroglia pathology in parkinson’s disease: an in vivo11C-BU99008 PET study. Brain: J Neurol (2019) 142:3116–28. doi: 10.1093/brain/awz260

CrossRef Full Text | Google Scholar

117. Tyacke RJ, Myers JFM, Venkataraman A, Mick I, Turton S, Passchier J, et al. Evaluation of (11)C-BU99008, a PET ligand for the Imidazoline(2) binding site in human brain. J Nucl Med (2018) 59:1597–602. doi: 10.2967/jnumed.118.208009

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Tesson F, Limon-Boulez I, Urban P, Puype M, Vandekerckhove J, Coupry I, et al. Localization of I2-imidazoline binding sites on monoamine oxidases. J Biol Chem (1995) 270:9856–61. doi: 10.1074/jbc.270.17.9856

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Kawamura K, Maeda J, Hatori A, Okauchi T, Nagai Y, Higuchi M, et al. In vivo and in vitro imaging of I₂ imidazoline receptors in the monkey brain. Synapse (2011) 65:452–5. doi: 10.1002/syn.20897

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Parker CA, Nabulsi N, Holden D, Lin SF, Cass T, Labaree D, et al. Evaluation of 11C-BU99008, a PET ligand for the imidazoline2 binding sites in rhesus brain. J Nucl Med (2014) 55:838–44. doi: 10.2967/jnumed.113.131854

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Calsolaro V, Matthews PM, Donat CK, Livingston NR, Femminella GD, Guedes SS, et al. Astrocyte reactivity with late-onset cognitive impairment assessed in vivo using (11)C-BU99008 PET and its relationship with amyloid load. Mol Psychiatry (2021) 26:5848–55. doi: 10.1038/s41380-021-01193-z

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Livingston NR, Calsolaro V, Hinz R, Nowell J, Raza S, Gentleman S, et al. Relationship between astrocyte reactivity, using novel (11)C-BU99008 PET, and glucose metabolism, grey matter volume and amyloid load in cognitively impaired individuals. Mol Psychiatry (2022) 27:2019–29. doi: 10.1038/s41380-021-01429-y

PubMed Abstract | CrossRef Full Text | Google Scholar

123. 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: 10.1007/s00259-016-3457-7

PubMed Abstract | CrossRef Full Text | Google Scholar

124. 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: 10.1111/jnc.13716

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Esposito G, Giovacchini G, Liow JS, Bhattacharjee AK, Greenstein D, Schapiro M, et al. Imaging neuroinflammation in alzheimer’s disease with radiolabeled arachidonic acid and PET. J Nucl Med (2008) 49:1414–21. doi: 10.2967/jnumed.107.049619

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Albrecht DS, Granziera C, Hooker JM, Loggia ML. In vivo imaging of human neuroinflammation. ACS Chem Neurosci (2016) 7:470–83. doi: 10.1021/acschemneuro.6b00056

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Lagarde J, Sarazin M, Chauviré V, Stankoff B, Kas A, Lacomblez L, et al. Cholinergic changes in aging and Alzheimer disease: An [18F]-F-A-85380 exploratory PET study. Alzheimer Dis Assoc Disord (2017) 31:8–12. doi: 10.1097/wad.0000000000000163

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Wang HY, Lee DH, D'Andrea MR, Peterson PA, Shank RP, Reitz AB. Beta-Amyloid(1-42) binds to alpha7 nicotinic acetylcholine receptor with high affinity. implications for alzheimer’s disease pathology. J Biol Chem (2000) 275:5626–32. doi: 10.1074/jbc.275.8.5626

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Hillmer AT, Li S, Zheng MQ, Scheunemann M, Lin SF, Nabulsi N, et al. PET imaging of α(7) nicotinic acetylcholine receptors: A comparative study of [(18)F]ASEM and [(18)F]DBT-10 in nonhuman primates, and further evaluation of [(18)F]ASEM in humans. Eur J Nucl Med Mol Imaging (2017) 44:1042–50. doi: 10.1007/s00259-017-3621-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Alzheimer’s disease, positron emission tomography, neuroinflammation, mild cognitive impairment, review

Citation: Huang J (2022) Novel brain PET imaging agents: Strategies for imaging neuroinflammation in Alzheimer’s disease and mild cognitive impairment. Front. Immunol. 13:1010946. doi: 10.3389/fimmu.2022.1010946

Received: 03 August 2022; Accepted: 06 September 2022;
Published: 23 September 2022.

Edited by:

Anwen Shao, Zhejiang University, China

Reviewed by:

Jiang Wu, Soochow University, China
Qiang Hu, Nanjing Medical University, China

Copyright © 2022 Huang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jie Huang, 344925209@qq.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.