Skip to main content
Neuroscience Bulletin logoLink to Neuroscience Bulletin
. 2024 Mar 14;40(6):743–758. doi: 10.1007/s12264-024-01188-0

Positron Emission Tomography Imaging of Synaptic Dysfunction in Parkinson’s Disease

Jiaqi Niu 1,2,3, Yan Zhong 1,2,3, Chentao Jin 1,2,3, Peili Cen 1,2,3, Jing Wang 1,2,3, Chunyi Cui 1,2,3, Le Xue 1,2,3, Xingyue Cui 1,2,3, Mei Tian 1,2,3,6,, Hong Zhang 1,2,3,4,5,
PMCID: PMC11178751  PMID: 38483697

Abstract

Parkinson’s disease (PD) is one of the most common neurodegenerative diseases with a complex pathogenesis. Aggregations formed by abnormal deposition of alpha-synuclein (αSyn) lead to synapse dysfunction of the dopamine and non-dopamine systems. The loss of dopaminergic neurons and concomitant alterations in non-dopaminergic function in PD constitute its primary pathological manifestation. Positron emission tomography (PET), as a representative molecular imaging technique, enables the non-invasive visualization, characterization, and quantification of biological processes at cellular and molecular levels. Imaging synaptic function with PET would provide insights into the mechanisms underlying PD and facilitate the optimization of clinical management. In this review, we focus on the synaptic dysfunction associated with the αSyn pathology of PD, summarize various related targets and radiopharmaceuticals, and discuss applications and perspectives of PET imaging of synaptic dysfunction in PD.

Keywords: Parkinson’s disease, Positron emission tomography, Synapse dysfunction, Alpha-synuclein, Dopamine system, Non-dopamine system

Introduction

Parkinson's disease (PD) is the second most common neurodegenerative disease. Its pathogenesis is complex and multifaceted. But what is clear is that the synapse serves as the fundamental functional unit of neurons and plays the crucial node within neural circuits, and its degree of dysfunction determines disease symptoms and severity. The misfolding and deposition of alpha-synuclein (αSyn) lead to irreversible synapse dysfunction of dopamine and non-dopamine systems in PD. The progressive degeneration of dopamine terminals in the substantia nigra (SN) is the main pathological feature. When a certain number of dopaminergic neurons are lost, the typical motor symptoms in PD occur. Furthermore, in response to the dramatic alterations in dopamine function, various non-dopaminergic systems change accordingly. It is increasingly acknowledged that the presence of many PD symptoms is also attributed to disturbances in non-dopaminergic regulatory systems. For example, abnormal activation of non-dopamine systems such as the serotonin or cholinergic system is involved in the pathogenesis of Parkinsonian motor behavior [1, 2]. Non-dopamine system dysfunction is also a main cause of non-motor symptoms in PD, such as depression, anxiety, and insomnia. These motor and non-motor symptoms resulting from synaptic dysfunction greatly impact the quality of life of PD patients and may even elevate the risk of disability in severe cases [3]. At present, the traditional diagnosis of PD mainly relies on medical history, clinical manifestations, and neurological examination, which has a lag and low accuracy. A large amount of data indicates that early diagnosis and intervention in PD can evidently improve patients’ quality of life and prolong their lifespan. Therefore, it is important to diagnose PD as early as possible for effective prevention and treatment.

With the development of precision medicine, molecular imaging has gained increasing attention, by which detecting early subtle changes in neurological diseases can be supporting evidence in guiding clinical management [46]. As a representative molecular imaging technology, positron emission tomography (PET) can measure the distribution of radiotracers in vivo, with outstanding capabilities to visualize, characterize, and quantify biological processes at molecular and cellular levels. Increasingly, PET molecular imaging has been successfully applied in clinical diagnosis, treatment, and research on PD. To date, a series of imaging probes have been developed for the visualization of synaptic function in PD.

In this review, we focus on the synaptic dysfunction related to the αSyn pathology of PD. Next, we summarize various targets and radiopharmaceuticals related to dopaminergic and non-dopaminergic synaptic dysfunction in PD (Table 1, Fig. 1), and discuss their applications. Finally, we present the current limitations of PET imaging of synaptic dysfunction in PD and analyze future directions of development, providing new perspectives and suggestions for further research and applications.

Table 1.

PET radiopharmaceuticals for synaptic dysfunction of dopamine and non-dopamine systems in Parkinson’s disease.

Targets Radiopharmaceuticals References
Dopamine system
Dopaminergic system Aromatic L-amino acid decarboxylase [18F]DOPA Ref.[18]
Vesicular monoamine transporter 2 [11C]TBZ, [11C]MTBZ, [11C]DTBZ Ref.[23, 92, 93]
[18F]FP-DTBZ Ref.[25]
Dopamine transporter [11C]beta-CFT, [11C]PE2I Ref.[28, 30]
[18F]FP-CIT, [18F]FE-PE2I Ref.[76, 82]
Dopamine receptor [11C]raclopride, [11C]NMSP, [11C]NPA, [11C]MNPA, [11C]PHNO Ref.[33, 34, 37, 92, 93]
[18F]Fallypride Ref.[35]
Non-dopamine system
Cholinergic system Vesicular acetylcholine transporter [18F]FMV, [18F]FAA, [18F]NEFA, [18F]FBT, [18F]FEOBV, [18F]FBVM Ref.[38, 9496]
Nicotinic acetylcholine receptor [11C]nicotine, [11C]MPA, [11C]ABT-418, [11C]MeQAA Ref.[39, 40]
2-[18F]FA-85380, 6-[18F]FA-85380, [18F]Flubatine, [18F]AZAN, [18F]XTRA, [18F]ASEM Ref.[42, 43, 46, 97, 98]
Acetylcholinesterase [11C]AMP, [11C]PMP, [11C]donepezil Ref.[44, 46]
Serotonergic system Serotonin transporter [11C]MCN-5652, [11C]DASB Ref.[54, 57]
5-Hydroxytryptamine receptor [11C]WAY-100635 Ref.[58]
[18F]FCWAY, [18F]MPPF, [18F]MeFWAY Ref.[59, 60, 99]
Norepinephrine system Norepinephrine transporter [11C]Methylreboxetine, [11C]NS8880 Ref.[63, 65]
(S, S)-[18F]FMeNER-D2 Ref.[64]
Cannabinoid system Cannabinoid receptor [11C]JHU75528, [11C]MePPEP Ref.[100, 101]
[18F]MK-9470, [18F]FMPEP-d2, [18F]FPATPP Ref.[67, 68, 101]

Fig. 1.

Fig. 1

Clinical symptoms in Parkinson’s disease and schematic of PET targets related to synaptic dysfunction. DAT, dopamine transporter; VMAT2, vesicular monoamine transporter 2; AADC, aromatic L-amino acid decarboxylase; VAChT, vesicular acetylcholine transporter; AChE, acetylcholinesterase; SERT, serotonin transporter; NET, norepinephrine transporter; CB1/CB2R, cannabinoid type 1 and type 2 receptors; D1/D2R, dopamine type 1 and type 2 receptors; nAChR, nicotinic acetylcholine receptor; 5-HT(1A)R, 5-hydroxytryptamine 1A receptor. (Parkinson’s patient section created with BioRender.com).

The Key Mechanism of Synaptic Dysfunction in PD: Alpha-synuclein (αSyn)

PD is essentially an alpha-synucleinopathy. αSyn is a natural presynaptic neuronal protein encoded by the SNCA gene and is widely expressed in the healthy central nervous system (CNS) [7]. In PD, αSyn undergoes misfolding and pathological deposition to form Lewy body aggregates. These aggregates subsequently spread throughout the brain in a prion-like manner [8]. The substance has different biological effects on the PD mechanism by localizing in different parts. In the mitochondrion, αSyn aggregates can cause mitochondrial dysfunction and induce the production of reactive oxygen species, which greatly increases the level of oxidative stress [9]. This is a common feature of neurodegenerative diseases. The existence of αSyn inclusions has also been confirmed in astrocytes and microglia, which leads to immune system disorders and neuroinflammation [10]. This pathological process causes glial cells to lose their neuroprotective properties and trigger synaptic dysfunction. Importantly, the potential synergistic effect between mitochondrial dysfunction and neuroinflammation greatly prompts the deterioration of synaptic function and eventual apoptosis of neurons as a whole [11] (Fig. 2). In addition, mutations in αSyn-related genes, such as SNCA and A30P, result in increased concentrations of αSyn and faster aggregation, which are also strongly associated with early-onset or hereditary PD [12, 13].

Fig. 2.

Fig. 2

Pathological mechanism of synaptic dysfunction in PD. The core pathology of PD is characterized by the misfolding and aggregation of alpha-synuclein, resulting in the formation of Lewy bodies. This aggregation induces neuroinflammation through the activation of microglia and astrocytes, as well as causing mitochondrial dysfunction and oxidative stress. These changes lead to synaptic dysfunction and neuronal degeneration or loss ultimately. ROS, reactive oxygen species.

αSyn enables early diagnosis by targeting the fundamental mechanisms of PD, making it the most valuable and direct biomarker for monitoring disease. However, the development of αSyn-targeted probes has been in its infancy. Although little progress has been made in this field, such as [18F]BQ2 and [11C]MODAG-001, these compounds have the limitation of poor specificity and cannot provide satisfactory image quality [14, 15]. Recently, Ye's team developed the specific probe [18F]F0502B for αSyn for the first time and successfully verified it in animals, filling in the gap in the field of αSyn molecular probes at last [16]. This radiopharmaceutical can achieve priority and tight binding with αSyn, and it has no affinity for Abeta and tau fibers, showing high specificity to be a promising radiopharmaceutical for αSyn. At present, the team is conducting clinical transformation and application research in Huashan Hospital. Anyway, the development of an αSyn-specific PET probe remains a challenging task.

Dopaminergic Synapse Dysfunction in PD

Dopaminergic impairment is the most evident alteration resulting from synaptic dysfunction in PD. The role of the dopamine system in synapses generally consists of four components, that is synthesis, transport, binding, and reuptake. Initially, L-tyrosine is hydroxylated to form L-levodopa, which is subsequently decarboxylated by aromatic L-amino acid decarboxylase (AADC) to produce dopamine. Next, dopamine is transported into vesicles by a membrane protein located on the vesicles of monoaminergic neurons, vesicular monoamine transporter protein 2 (VMAT2). After depolarizing neuronal excitation, dopamine in vesicles is released into the synaptic gap and acts by binding to dopamine receptors in the postsynaptic membrane. Parts of the dopamine in the synaptic gap can be reuptaken into the presynaptic neuron cytoplasm through active transport of the dopamine transporter (DAT) on the presynaptic membrane for degradation or reuse. Dopaminergic impairment is the core mechanism of PD, which has a major impact on the nigrostriatal circuit responsible for motor coordination and gives rise to main extrapyramidal symptoms. In the potential mechanism of 1-methyl-4-phenyl-1,2,3,6-tetra-hydropyridine, which is the common method for modeling PD, the lethal toxin, 1-methyl-4-phenylpyridinium, is mistakenly taken up by the DAT, inhibiting the mitochondrial respiratory chain. This mainly leads to the damage or loss of dopaminergic neurons and the nigrostriatal axons, inducing irreversible PD [17]. Many anatomical pathological and imaging studies have confirmed the loss of dopaminergic neurons and the reduction in dopaminergic activity in early PD patients. Therefore, it can be inferred that targets on dopaminergic synapses play a pivotal role in the pathogenesis of PD.

Dopamine Synthesis: Aromatic L-amino Acid Decarboxylase (AADC)

Dihydroxyphenylalanine (DOPA) is the precursor of dopamine, and its accumulation can reflect the activity of AADC and the integrity of presynaptic dopaminergic function. [18F]DOPA is a fluorinated analog of natural levodopa. After being injected and transported into cells, it is decarboxylated by AADC to form [18F]dopamine. Since the 1980s, [18F]DOPA has been successfully synthesized and used in clinical examinations [18]. It was initially used to evaluate the integrity of the dopaminergic system in PD. [18F]DOPA has a maximal uptake range and the highest standardized uptake value in the basal ganglia, but very low uptake in normal tissues, providing an outstanding lesion/background ratio and better capacity for disease monitoring [19]. However, early decarboxylase frequently exhibits a compensatory phenomenon that makes its fluctuations inconsequential or even normal, limiting the sensitivity of [18F]DOPA [20]. Moreover, its radiosynthesis process is complex, and finding a simpler and more effective manufacturing strategy has been a challenge [21]. There are no other radiopharmaceuticals targeting AADC, and this area is continuously evolving.

Dopamine Transport: Vesicular Monoamine Transporter Protein 2 (VMAT2)

The initial development of VMAT2 radiopharmaceuticals relied upon the metabolites of tetrabenazine (TBZ) or its analog (2-O-methyl)dihydrotetrabenazine (MTBZ). [11C]TBZ, the earliest radioactive tracer for PET imaging of VMAT2, has been developed and approved for use in many countries since the 1960s. In 1993, Kilbourn and colleagues effectively conducted VMAT2 imaging in the human brain using [11C]TBZ. Subsequently, the case of the highly specific binding of TBZ and MTBZ has aroused other exploration. Dihydrotetrabenzoxazine (DTBZ) is one of the metabolites of TBZ that blocks the storage of monoamine neurotransmitters in presynaptic vesicles by binding to VMAT2 [22]. After modification and common 11C labeling, it has been used in imaging studies of neurodegenerative diseases. [11C]DTBZ is highly sensitive to dopaminergic terminal loss and remains unaffected by compensation or drug regulation. This makes it the most widely used VMAT2 PET molecular probe to evaluate the integrity of presynaptic dopamine terminals [23]. However, this substance is essentially generated through the complete reduction and rapid formation of TBZ under the action of carbonyl reductase. Consequently, its imaging effect does not significantly differ from TBZ-related radiopharmaceuticals [24]. Afterward, a novel radioligand [18F]FP-DTBZ ([18F]AV-133) was developed and showed excellent kinetics and affinity in both animal and human brains, offering an improved approach for detecting VMAT2 [25, 26]. The diversity of PET probes for VMAT2 has helped to comprehensively understand dopaminergic presynaptic function.

Dopamine Reuptake: Dopamine Transporter (DAT)

Because of the high affinity and sensitivity of cocaine and its derivatives for the DAT, many DAT imaging ligands have been developed based on phenyltropane cocaine analogs. 2beta-carbomethoxy-3beta-(4-fluorophenyl)tropane (beta-CFT or FP-CIT) is an effective ligand developed earlier for quantifying DAT density and is purified from nor-beta-CFT. We usually label 11C at 0-methyl as a simpler and easier method with a shorter synthesis time. However, there is a serious limitation in that the kinetics of [11C]beta-CFT are so slow that it is only absorbed in the striatum 24 h after injection, which leads to the DAT only being adequately imaged the next day [27]. Later, [18F]FP-CIT became an excellent substitute taking only 2 h after injection to reach a plateau [28]. Thus, [18F]FP-CIT has become an excellent substitute because of its more convenient performance in clinical examination. In 1999, a new ligand, N-(3-iodoprop-2E-enyl)-2beta-carbomethoxy-3beta-(4'-methylphenyl)nortropane (PE2I), was developed in an autoradiography study of normal postmortem human brain, showing the main distribution of the DAT in the basal ganglia. It has high structural homology with CIT compounds. At the same time, it has a lower binding force to norepinephrine and the serotonin transporter (SERT), due to the combination of 4-methyl substituted by a benzene ring with 3-iodopropyl-2-enyl [29]. In a competitive in vitro study, it was confirmed that [11C]PE2I has high selectivity and affinity for the DAT, and serves as one of the most selective ligands. However, [11C]PE2I has significant variability in the quantitative measurement of the DAT, and the radioactive metabolites produced by 4-hydroxymethyl bind to the DAT in the striatum, interfering with quantitative monitoring [30]. In comparison, [18F]FE-PE2I, a novel probe, was evaluated to have a faster elution rate and stronger kinetic activity [31]. More importantly, it produces fewer radiometabolites with intermediate lipophilicity that interferes with quantification [31, 32]. So [18F]FE-PE2I may be a more suitable choice for the quantitative display of the DAT in vivo.

Binding to Dopamine: Dopamine Receptor

Dopamine receptors are mainly categorized into D1 receptors (D1, D5) and D2 receptors (D2, D3, and D4), and most research is still focused on D2 receptors. [11C] Raclopride is a common D2 antagonist imaging agent with high selectivity and sensitivity, and it shows strong reliability in many clinical trials. [11C] Raclopride PET was used for the first time to report the release of DA from the striatum in humans in 1998. Subsequently, Volkow et al. used modafinil to occupy the DAT and raise dopamine levels and successfully presented this fluctuation with [11C]raclopride, which proved the feasibility of [11C]raclopride as an indicator of changes in extracellular dopamine [33]. However low affinity is a disadvantage that cannot be ignored. In comparison, the radioligand [11C]NMSP has a higher affinity (0.1–0.3 nmol/L) for the D2 receptor. However, it also has the capability to bind to both 5-HT receptors simultaneously, which poses a major concern [34]. With further exploration, [18F]Fallypride was reported as a D2 radioligand with high affinity and specificity at the same time and is capable of detecting complex defects within the cortico-striato-thalamic circuit [35]. PET studies have reported that D2 agonists are more susceptible to endogenous competition than traditional D2 antagonists, resulting in more sensitive detection of extracellular DA changes [36, 37]. And its affinity is higher and more saturated in vivo. A series of agonists for the quantitative assessment of D2 receptors, such as [11C]NPA, [11C]MNPA, and [11C]PHNO, have been developed as excellent PET radiotracers. In rodent studies, [11C]PHNO showed strong binding signals in the striatum, with a specific/non-specific ratio >90% [37]. Since 2008, researchers have evaluated the performance of these agonist probes in humans, all of which display fast dynamics and an ideal imaging effect with low noise and variability.

Non-dopaminergic Synaptic Dysfunction in PD

It is well known that PD is highly heterogeneous with multiple phenotypes; this is the combined effect of diverse systems and neurotransmitter factors caused by a complex neurochemical imbalance in this disease. Although dopaminergic neuron depletion is the main feature of PD, it is undeniable that the non-dopaminergic system also has significant changes in its pathogenesis (Fig. 3). Non-dopaminergic synaptic dysfunction in PD involves many aspects, such as the cholinergic, serotonergic, norepinephrine, and cannabinoid systems.

Fig. 3.

Fig. 3

Applications of PET imaging of non-dopamine systems in PD. A Cholinergic PET images of [18F]FEOBV VAChT (upper row) and [18F]Flubatine α4β2 nAChR (lower row) in a single PD patient. In columns A and B, both radioligands show increased uptake in the anterior frontotemporal cortex. In columns C and D, [18F]FEOBV shows increased uptake in the striatum and thalamus, and [18F]Flubatine shows increased uptake in the thalamus but not significantly in the striatum. In columns E–G, [18F]FEOBV shows significantly enhanced radioactivity in the whole cerebellum, while [18F]Flubatine shows diffusely reduced uptake in the cerebellum. Adapted with permission from Ref.[94]. B Axial, sagittal, and coronal [11C]DASB PET imaging among healthy controls, early unilateral idiopathic Parkinson’s disease (IPD), and advanced bilateral IPD patients, evaluating the availability of 5-HT1ARs. Adapted with permission from Ref.[102]. C [11C]MeNER PET images of NET sites in healthy controls and patients with PD. Adapted with permission from Ref.[103].

Cholinergic System

For imaging synaptic function in the brain, the cholinergic system consists of the vesicular acetylcholine transporter (VAChT) in the presynaptic terminals, nicotinic acetylcholine receptors (nAChRs) in the postsynaptic terminals, and acetylcholinesterase (AChE) in the synaptic cleft. Neuroimaging studies have shown reduced cholinergic neurons in cortical and subcortical regions of PD patients. The principal non-motor symptom in PD is cognitive impairment, which is widely recognized as a highly incapacitating process involving the cholinergic neurotransmission system. In addition, the cholinergic system partly influences the development of motor symptoms in PD. It has been shown that mice with VAChT deletion are more vulnerable to neurotoxic damage that mimics the dopamine imbalance pathogenesis of PD. This evidence indicates a close relationship between cholinergic dysfunction and PD.

Vesicular Acetylcholine Transporter (VAChT)

In the early stage, a series of vesical analogs labeled with radionuclides, such as [18F]FMV, [18F]FAA, and [18F]NEFA, were developed as PET agents in the quantitative study of VAChT. However, they have little or no value for human application. [18F] Fluorobenzyltrozamicol ([18F]FBT) is an effective ligand for cholinergic terminal density. Its PET imaging can detect both acute and chronic changes in cholinergic activity. Related studies using the [18F]FBT analog (+)-meta-125I-iodobenzyltrozamicol also suggested the consistency of its fluctuations with cholinergic changes, which provides evidence for the clinical imaging application of neuropsychiatric disease in the future. (-)-5-[18F]fluoroethoxybenzovesamicol ([18F]FEOBV), a visamicol derivative, is a novel VAChT-specific radioligand that was recently developed. Currently, it is widely used in the quantitative study of cholinergic function in clinical practice to demonstrate substantial efficacy in the assessment of neurodegenerative dementias such as PD and AD.

Recently, a number of promising radiopharmaceuticals have been further developed and promoted for VAChT, such as [18F]FBVM. It can sensitively detect cholinergic deprivation even in brain regions with low VAChT density, such as the hippocampus and cerebral cortex. Compared with [18F]FEOBV, it exhibits up to 50 times higher affinity, an enhanced signal-to-noise ratio for brain uptake, and superior cholinergic PET images [38]. However, [18F]FBVM is currently limited to preclinical stages and is waiting to be applied in clinical practice.

Nicotinic Acetylcholine Receptor (nAChR)

The nAChR, a barrel-shaped hetero-oligomer, is most abundant in its heteromeric α4β2 and α7 subtypes in the CNS. It acts on many physiological and behavioral processes by modifying the effects of nicotine. [11C] Nicotine was developed as the first nAChR radioligand to be used in neurodegenerative disorders, but it is affected by various limitations in the brain like blood flow and the blood-brain barrier (BBB) [39]. Most fatally, it is dominated by non-specific binding, resulting in low-quality images. Two other nicotine analogs, [11C]MPA and [11C]ABT-418, have a similar disadvantage [40]. Later, 3-pyridyl ether derivatives such as 2-[18F]FA-85380 and 6-[18F]FA-85380 were developed. They are nicotine agonists with higher affinity and specificity for α4β2 nAChRs, and the toxicity to the organism is greatly curtailed [41]. But they also have a slow kinetics limitation.

In order to solve these problems, novel α4β2 subtype radioligands with fast kinetics have emerged, representatively [18F]Flubatine, [18F]AZAN, and [18F]XTRA. Due to the effective anesthetic and analgesic properties of nAChRs, this kind of radiopharmaceutical was developed on the basis of epibatidine and its derivatives isolated from Ecuadorian poison frogs. In the first human quantitative study, [18F]Flubatine showed high metabolic stability and a rapid kinetic response [42]. However, [18F]AZAN and [18F]XTRA are mostly metabolized after injection, resulting in a serious problem of instability [43]. For the α7 subtype, only a few agents have been developed, such as [11C]MeQAA and [18F]ASEM, leaving a lot of space to be explored.

Acetylcholinesterase (AChE)

AChE regulates and maintains the concentration of ACh, which keeps the cholinergic system in a state of dynamic equilibrium. Once the hydrolysis of AChE is blocked, ACh does not dissociate from receptors of the postsynaptic membrane, which leads to many neurocognitive disorders in PD. Here are the two most common AChE radioactive probes, [11C]AMP ([11C]MP4A) and [11C]PMP ([11C]MP4P). They both belong to substrate-type ligands for AChE PET imaging. In addition, [11C]AMP and [11C]PMP are small molecular lipophilic analogs of ACh that can easily cross the BBB [44]. It was found that the radioactivity uptake after injection was significantly correlated with the AChE density in different brain regions. Besides, the pharmacokinetics of these compounds are so fast that they can enter the brain and be completely hydrolyzed into radioactive metabolites by AChE only 1 min after injection. The permeability of the BBB to this radioactive metabolite and its elimination is so slow that it can be retained in the brain for a long time, providing a stable time window for observation [45]. At present, these two radiopharmaceuticals are extensively used in clinical and research settings. However, their inability to provide peripheral imaging remains a major limitation to be considered [45].

According to the quantitative results of [11C]AMP on cholinergic inhibition, we found that the inhibitory effect induced by the AChE inhibitor, donepezil, is closely related to the plasma IC50 in a dose-dependent manner. Subsequently, based on the strong affinity and specificity of this reversible inhibitor, a new radiopharmaceutical, [11C]donepezil, has been developed. It is classified as a ligand-type probe and enables visualization of AChE density in peripheral organs, giving access to more extracerebral information than substrate-based probes [46].

Serotonergic System

The serotonergic system, including SERT and 5-hydroxytryptamine (5-HT) receptors, engages in the regulation of cognition, emotion, and motor behavior. Alterations in serotonin signaling are a non-neglectable component in PD. Pathology reports have confirmed that the serotonergic system is damaged in PD at the early stage, which is characterized by decreased expression in several basal ganglia nuclei, such as the putamen, caudate, and ventral caudate-putamen [47, 48]. This may be related to the negative modulation of serotonergic activity by αSyn in PD [49]. Multiple pieces of evidence suggest that serotonergic terminals can contribute to the development of levodopa-induced dyskinesia by modulating the dopamine system and promoting the non-physiologic release of dopamine in the striatum [50, 51]. Besides, some non-motor symptoms of PD, such as cognition, depression, and anxiety, also have a tight association with serotonergic dysfunction [52, 53].

Serotonin Transporter (SERT)

[11C]MCN-5652 is the first successfully applied PET radiotracer for the SERT. It was found to have a good correlation with serotonin uptake sites (r = 0.86) in mice 1 h after injection. At present, [11C]MCN-5652 has been applied to several diseases involving the serotonin system by evaluating the level of the SERT. However, it was found that [11C]MCN-5652 is only suitable for quantitatively visualizing regions with high SERT concentrations. Nonetheless, the specific binding may be omitted in areas with relatively low density, such as the prefrontal cortex [54]. This limitation makes it difficult to obtain reliable quantitative SERT data.

Later, [11C]3-amino-4-(2-dimethylaminomethylphenylsulfanyl)-benzonitrile ([11C]DASB) was discovered as a better probe for visualizing serotonergic function in the brain. [11C]DASB presents clear advantages over [11C]MCN-5652, including a higher plasma-free fraction and faster kinetics. Its specificity and non-specific binding rate to SERT are higher, resulting in a stronger contrast imaging effect [55, 56]. The feasibility of monitoring SERT by [11C]DASB has been evaluated in many rat studies, and the results show extremely high sensitivity and saturation binding (ED50 = 56 nmol/kg) to SERT in the brain and have good reliability and reproducibility [57].

5-Hydroxytyrptamine (5-HT) Receptor

[11C]WAY-100635 is a 5-HT1A antagonist with high affinity and selectivity that displays a significant specific signal in the serotonergic regions of the human cerebral cortex and raphe nuclei. In a study related to PD tremor, it was used to detect a generalized decrease in postsynaptic 5-HT1A receptor binding potentials (BPND) [58]. Due to the limitations of the 11C marker, researchers developed [18F]FCWAY and [18F]MPPF based on the analogs of WAY-100635, which have been used in human research. However, the huge defluorination of [18F]FCWAY hinders the accurate quantification of 5-HT1A receptor binding in the brain [59]. In order to solve this problem, a new radiopharmaceutical, [18F]MeFWAY, was developed with excellent metabolic stability. In the first human study, it showed excellent measurement of 5-HT1A receptors [60]. Although in a comparative study, the kinetic indicators, the distribution volume ratio, and the area under the regional time-activity curve ratio in [18F]MeFWAY were not as good as [18F]FCWAY, it is still a promising radiopharmaceutical [61].

Norepinephrine System

The relationship between the norepinephrine system and PD is multifaceted. In PD, the density and expression of norepinephrine neurons are generally reduced at the locus coeruleus (LC). In the meantime, the presynaptic transporter-mediated function of rapid norepinephrine clearance is diminished as well. Depletion of norepinephrine terminals may also be one of the reasons for the progression of symptoms. A PET imaging study revealed that norepinephrine-related LC degeneration is consistent with postmortem pathology in early PD, pointing to the possibility that norepinephrine injury may result in nonmotor symptoms in PD [62]. Therefore, it is widely agreed that norepinephrine synaptic dysfunction may contribute to the development and progression of PD.

The norepinephrine transporter (NET) regulates neural transmission by reabsorbing up to 80–90% of norepinephrine in the extracellular space. Based on reboxetine analogs known as norepinephrine reuptake inhibitors, many NET ligands have been developed and entered the field of molecular imaging. [11C]Methylreboxetine ([11C]MRB or [11C]MeNER) is a mature probe on the NET occupancy that has a very high affinity for the NET, making its affinity with other targets negligible [63]. PET imaging with [11C]MRB can effectively draw the map of norepinephrine defects in vivo. However, it has certain limitations in application due to its low absorption rate, sensitivity, and reliability. (S, S)-[18F]FMeNER-D2, as a new type of radioactive ligand, has a more stable determination time and peak uptake than [11C]MRB. But its BPND is so low that it is only 0.4–0.6 in the anterior cingulate cortex [64]. There are also many other NET radioligands, like [11C]NS8880, all of which have greatly improved properties over previous developers [65]. However, they all need to be validated in subsequent clinical trials.

Cannabinoid System

The endogenous cannabinoid system (ECS) was discovered in humans and other mammals as early as 1992. At present, the two main known receptors are cannabinoid type 1 (CB1R) and cannabinoid type 2 (CB2R), which play important roles in regulating various psychological and physiological functions through their interaction with endocannabinoids. CB1R is mainly found on the presynaptic terminals of dopaminergic neurons, and its activation increases the release of dopamine. It has been found that the overexpression of CB1R in PD may contribute to neuroinflammation, oxidative stress, and cell death processes. The above sections are all vital contributors to synaptic dysfunction. CB2R exists in peripheral immune cells and can reduce inflammation in the brain, implying an intrinsic molecular link to PD. Through the above mechanisms, the activity of cannabinoid receptors may influence the symptoms of PD to a large extent.

Due to the lipid signal transduction features of the ECS, the successful preparation of cannabinoid receptor radioligand requires moderate lipophilicity [66]. The first generation of imaging agents based on rimonabant are unsuitable for clinical application due to their high lipophilicity or poor BBB penetration. By continuously improving the lipophilicity of imaging agents, a series of second-generation CB1R PET agents have been successfully developed. [18F]MK-9470 is one of the most representative imaging agents and is widely used. As a potent reverse agonist, it has high selectivity (Ki = 0.7 nmol/L) and moderate lipophilicity (logD7.4 = 4.7) and can accomplish a forceful but reversible binding to CB1Rs. It is known that CB1Rs are mainly distributed in the SN, globus pallidus, putamen, hippocampus, and cerebellum of the human brain. A study of [18F]MK-9470 imaging of the rhesus monkey brain shows that the binding site of this radioactive ligand is highly compatible with CB1Rs. However, in the process of non-invasive imaging in humans, the kinetics of [18F]MK-9470 are so slow that it takes 2 h to reach the plateau after injection [67]. There are also some other second-generation imaging agents, including [11C]JHU75528 ([11C]OMAR), [11C]MePPEP, and [18F]FMPEP-d2, that balance selectivity, affinity, and lipophilicity but have the limitations of elution speed and defluorination capacity. Recently, it has been reported that a novel CB1R radioligand [18F]FPATPP has high specificity, high stability, and low defluorination, but only one case has been studied in mice, and it still needs our continuous exploration [68].

Applications of Synaptic Dysfunction PET Imaging in PD

Diagnostic Performance

According to the latest diagnosis criteria for PD, clinical symptoms still play a leading role. The major pathological changes in PD are the degeneration and loss of dopaminergic neurons in the SN pars compacta. However, due to the substantial compensatory effect of PD in its initial stage, typical symptoms frequently manifest after the loss of ~50% of dopaminergic neurons, which greatly contributes to the diagnostic lag. Radionuclide imaging has proven to be a potent tool for PD diagnosis; it can recognize early changes from an internal molecular perspective and greatly improve the diagnosis rate.

The axons of dopaminergic neurons are the most vulnerable because they are the earliest target of αSyn accumulation in PD, thus, presynaptic dopaminergic molecular imaging is recommended here for diagnosis. The Movement Disorder Society also regards normal presynaptic dopaminergic PET images as the absolute exclusion criterion of PD [69]. Initially, the diagnosis of PD was made by [18F]DOPA, reflecting AADC activity, to assess dopaminergic synthesis. In a [18F]DOPA PET study, the caudate nucleus and anterior thalamus were found to be as accurate as 0.93 in the diagnosis of PD by analyzing the striatal subregions [70]. However, due to the compensatory increase of the synthesis and dopa decarboxylase activity of residual dopamine neurons, the level of dopamine does not decrease significantly in the early stage. Therefore, [18F]DOPA PET imaging only has good diagnostic value in patients with middle and advanced PD. In contrast, the diagnostic power of DAT imaging is particularly outstanding in early PD (Fig. 4). Recognized as the most sensitive molecular imaging marker of PD, it displays a significant reduction in uptake early and tends to further decrease due to compensatory mechanisms [71]. A study used [18F]FE-PE2I PET imaging to measure the availability of DAT. The loss of DAT was found to be significantly detectable in de novo PD patients, even before the appearance of clinical symptoms [72]. Another quantitative brain imaging study in PD patients suggested that the correlation between BPND and the standardized uptake value ratio in [18F]FE-PE2I PET images could already be satisfactory earlier on [73]. Recently, other dopaminergic presynaptic targets have appeared to have greater performance in PD. For example, VMAT2 is less susceptible to dopamine drugs and compensatory regulation than AADC and DAT, and it is almost always concentrated in dopaminergic axons [74]. These make VMAT2 a more objective target for presynaptic dopaminergic imaging. A PET study with [18F]FP-DTBZ to diagnose PD, and its blinded validation, showed a high clinical diagnostic compliance rate of 0.933, pointing out that targeted VMAT2 imaging is also of high application potential in effectively distinguishing PD from healthy individuals [75].

Fig. 4.

Fig. 4

PET images of the dopamine transporter in PD. A [18F]FE-PE2I cross-sectional images of SRTM BPND and SUVR parameters, involving 2 reference areas of the cerebellum and occipital cortex, and 3 acquisition time windows. (a) a 52-year-old female with a healthy control. (b) a 67-year-old male PD patient (H-Y = 2), [18F]FE-PE2I uptake in the striatum is significantly reduced. Adapted with permission from Ref.[73] B Comparison of [18F]FP-CIT PET images in the striatum between a healthy control, an early-stage PD patient, and a late-stage PD patient. (a) healthy control, [18F]FP-CIT PET/MR image; (b) healthy control; (c) early-stage PD, [18F]FP-CIT uptake in the striatum is significantly reduced; (d) late-stage PD, as the disease progresses, [18F]FP-CIT uptake is further reduced. SRTM, simplified reference tissue model; BPND, binding potential; SUVR, standardized uptake value ratio; H–Y, Hoehn–Yahr stage. Adapted with permission from Ref.[104].

Differential Diagnosis of Parkinsonian Syndromes

Due to the significant overlap of clinical manifestations and mechanisms in Parkinsonian disorders, the differentiation of PD is also a link that cannot be ignored in the diagnostic process. Many PET scanning studies of dopamine synapses have demonstrated their high clinical value for the differential diagnosis of PD (Fig. 5). In a dual-phase PET/CT imaging study, [18F]FP-CIT was noted to achieve 91.7% specificity and 80% accuracy in differentiating idiopathic PD from atypical Parkinson's syndrome [76]. The semi-quantitative calculation of depth information from DAT PET images showed a significant difference between PD, multiple system atrophy (MSA), and progressive supranuclear palsy (PSP) (P <0.001) [77]. A comparative study found that in a series of Parkinsonian syndromes, a single [11C]PE2I PET examination can effectively replace the dual examination of [123I]FP-CIT SPECT and [18F]FDG PET in identifying them [78].

Fig. 5.

Fig. 5

[18F]FP-CIT PET images for differential diagnosis of Non-PD, IPD, MSA-P, MSA-C, and PSP. A, B Axial and coronal slices of delayed phase [18F]FP-CIT PET. IPD patients show decreased metabolism of the bilateral dorsal posterior putamen and ventral putamen. For MSA-C and MSA-P, decreased metabolism is also found in the posterior putamen and ventral putamen. PSP patients were characterized by extensive reduction in DAT binding of the putamen and caudate nuclei. C In the early phase [18F]FP-CIT PET, IPD has normal or hypermetabolic striatal metabolism in the bilateral dorsolateral putamen. MSA shows hypometabolism in the cerebellar cortex or basal ganglia region, which can differentiate between MSA-C and MSA-P. PSP shows hypometabolism in the medial frontal cortices and midbrain. Non-PD, people without Parkinson’s disease; IPD, idiopathic Parkinson’s disease; MSA-C, multiple system atrophy-cerebellar type; MSA-P, multiple system atrophy-Parkinson type; PSP, progressive supranuclear palsy; DAT, dopamine transporter. Adapted with permission from Ref.[105].

In addition, screening for non-dopaminergic alterations can also provide additional information for the diagnosis of PD. A [11C]DASB PET study targeting SERT successfully explored the imaging differences between PD and MSA [79]. Compared to PD, MSA patients have more severe serotonergic deficits in the brainstem and some subcortical regions. Other studies have found that PD and PSP can be distinguished by quantifying AChE density by [11C]PMP PET scanning. This method revealed a decrease in cholinergic AChE activity in the cerebral cortex in PD patients, while the decline is more remarkable in the subcortical region, such as the thalamus, in PSP and MSA patients [80, 81].

Efficacy Detection and Prognosis Evaluation

In terms of monitoring treatment changes and predicting prognosis, synaptic PET imaging also has tremendous advantages. In a group of early and stable PD patients, Jessia et al. synthetically analyzed dopaminergic and serotonergic states in combination with [11C]DTBZ, [11C]methylphenidate, [11C]DASB, and [11C]raclopride. The results showed a significant correlation with the degree of risk of developing dyskinesia after 3 years of levodopa treatment [51]. In another longitudinal trial, [18F]FE-PE2I PET was performed in non-advanced PD patients following 2 years of treatment, and little change was found in DAT availability in the SN, suggesting slow progression or compensatory changes [82]. Based on the monitored treatment in real-time, we can also adjust and optimize therapy regimens promptly. A PET study targeting VMAT2 by [11C]DTBZ suggested that the synaptic density had a determination on the decay time of the levodopa response, so that PD patients with a lower synaptic density may necessitate alternative or more aggressive therapies [83]. These precise and prompt assessments by PET molecular imaging of synaptic function will greatly benefit individuals with PD.

Exploration of Mechanism and Heterogeneity

PD is a complex and long-progressive disease with high heterogeneity that is affected by clinical, pathological, genetic, and other factors. Different individuals or stages lead to differences in phenotype and prognosis. Pathologically, these manifestations depend on the damage to specific brain regions caused by the abnormal deposition of αSyn. However, although a specific probe for αSyn has been developed recently, it is still in the preclinical trial stage [16]. In order to better understand intrinsic pathology and stratify PD patients, imaging of different kinds of synaptic function provides excellent options, which allows us to visualize the pathological condition more comprehensively.

In the Dutch Parkinson cohort study protocol, cholinergic and dopaminergic PET imaging were involved as one of the criteria for classifying PD subtypes [84]. AChE-related cholinergic PET imaging is commonly used to assess symptoms associated with visceral parasympathetic nervous system disorders. Studies have shown that [11C]donepezil binding is significantly reduced in the small intestine and pancreas in PD patients with symptoms such as constipation and gastroparesis [85]. This method provides feasibility for studying the temporal and spatial heterogeneity in PD. Knudsen et al. combined [11C]donepezil, [11C]MeNER, and [18F]DOPA PET to evaluate the Braak I, II, and III stages of PD, respectively [86]. Another study also revealed two types of body priority and brain priority in PD through cholinergic PET imaging [87]. Molecular imaging based on different synapses brings great convenience to exploring the whole picture and heterogeneity of PD pathology.

As the premise for in-depth analysis of all the above applications, quantitative PET molecular imaging should not be neglected; it helps to establish the direct relationship between imaging parameters and biofunctional indicators. Manual piece-by-piece analysis is a typical and traditional strategy. However, this method is time-consuming and laborious and depends on the operator’s judgment. By contrast, semi-automated or fully-automated analysis methods serve as commendable alternatives. Many comparative studies have demonstrated that the automatic definition of regions of interest has lower test-retest variability and better reliability than manual methods [88]. Nevertheless, in these automated approaches, special attention should be paid to target area conditions and the preprocessing steps. An inappropriate situation or treatment will lead to a deviation in the results. Besides, in constructing classification or prediction models, fully automated feature extraction by deep learning can achieve performance comparable to or superior to traditional radionics [89, 90]. However, this advantage is always based on the large scale of datasets, which severely limits the practical application of fully automatic modeling. Nowadays, multi-center collaborative research has become a trend. However, while sharing imaging data, we also need to consider several problems of data privacy and security. All in all, weighing convenience and accuracy, low data costs, optimal performance, and expanding sample centers under compliance remain worth thinking about, especially in the era of big data.

In addition, there are some limitations of PET imaging in PD applications. Firstly, PET technology has an obvious inherent defect in its low spatial resolution, which is not conducive to recognizing some subtle anatomical structures. Secondly, due to the short half-life (T1/2), most radiopharmaceuticals, such as 11C (T1/2 = 20 min), 13N (T1/2 = 10 min), and 15O (T1/2 = 2 min), must rely on an internal cyclotron for immediate production, which greatly limits their availability. In contrast, 18F has a long imaging window (T1/2 = 110 min) and convenient synthesis conditions that can be readily supplied by commercial radiopharmacies. Nevertheless, some targets still lack radiotracers labeled with 18F or other isotopes possessing a long T1/2. Finally, synaptic dysfunction in PD is fundamentally attributed to the misfolding and deposition of αSyn, but the development of molecular probes related to αSyn is currently not adequate for clinical trials.

Conclusions and Future Perspectives

PD is the second-most common neurodegenerative disease after Alzheimer's disease, with complex pathological mechanisms that are closely associated with different targets on synapses. It has typical motor symptoms, accompanied by some non-motor symptoms. However, there is often a latent period in the clinical diagnosis based on symptoms, which makes it impossible for doctors to make a timely diagnosis. PD not only affects the quality of life of a large number of patients but also imposes a heavy burden on society in many aspects. Therefore, it is important to make an early diagnosis and risk assessment of PD so as to further develop treatment strategies and greatly improve the prognosis.

PET is an excellent molecular imaging technique that enables non-invasive, dynamic visualization of metabolic and functional changes in vivo [91]. Given the succession of pathological events associated with synaptic dysfunction in PD, we should focus on various crucial molecular imaging targets, comprising not only αSyn but also the dopamine and non-dopamine systems. PET imaging of these biomarkers will be helpful for the early diagnosis of PD and is also of great importance in its treatment and prognostic evaluation.

In summary, nuclear molecular imaging technology has great potential in PD. Our research direction should focus on developing αSyn-specific probes to delve into PD fundamentally. We must continue to enrich novel PD-related synaptic function imaging tracers, especially those with a long T1/2. Moreover, the use of multimodal imaging in PD applications should be increased to address the intrinsic weaknesses of various imaging techniques and to acquire diverse dimensional data from images. In the context of the big data era, we should actively summarize the experience of PET quantification, look for a balance between data cost and performance, and safeguard data privacy. In the future, further studies, including in vitro screening and in vivo verification, are needed in this field in order to accelerate clinical application and find more possibilities for PET applications in PD and even other kinds of neurodegenerative disorders, so that patients can receive more benefits quickly.

Acknowledgements

This review was supported by the National Key Research and Development Program of China (2022YFC2009900, 2021YFE0108300, 2022YFE0118000, and 2021YFA1101700), the National Natural Science Foundation of China (82030049), and Fundamental Research Funds for the Central Universities of China.

Conflict of interest

The authors declare no competing interests.

Contributor Information

Mei Tian, Email: tianmei@fudan.edu.cn.

Hong Zhang, Email: hzhang21@zju.edu.cn.

References

  • 1.Brumberg J, Küsters S, Al-Momani E, Marotta G, Cosgrove KP, van Dyck CH, et al. Cholinergic activity and levodopa-induced dyskinesia: A multitracer molecular imaging study. Ann Clin Transl Neurol. 2017;4:632–639. doi: 10.1002/acn3.438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Li LX, Li YL, Wu JT, Song JZ, Li XM. Glutamatergic neurons in the caudal zona incerta regulate parkinsonian motor symptoms in mice. Neurosci Bull. 2022;38:1–15. doi: 10.1007/s12264-021-00775-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bloem BR, Okun MS, Klein C. Parkinson’s disease. Lancet. 2021;397:2284–2303. doi: 10.1016/S0140-6736(21)00218-X. [DOI] [PubMed] [Google Scholar]
  • 4.Tian M, Zuo C, Civelek AC, Carrio I, Watanabe Y, Kang KW, et al. International nuclear medicine consensus on the clinical use of amyloid positron emission tomography in Alzheimer’s disease. Phenomics. 2022;3:375–389. doi: 10.1007/s43657-022-00068-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Tian M, Civelek AC, Carrio I, Watanabe Y, Kang KW, Murakami K, et al. International consensus on the use of tau PET imaging agent 18F-flortaucipir in Alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2022;49:895–904. doi: 10.1007/s00259-021-05673-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Tian M, Watanabe Y, Kang KW, Murakami K, Chiti A, Carrio I, et al. International consensus on the use of[18F]-FDG PET/CT in pediatric patients affected by epilepsy. Eur J Nucl Med Mol Imag. 2021;48:3827–3834. doi: 10.1007/s00259-021-05524-8. [DOI] [PubMed] [Google Scholar]
  • 7.Wassouf Z, Schulze-Hentrich JM. Alpha-synuclein at the nexus of genes and environment: the impact of environmental enrichment and stress on brain health and disease. J Neurochem. 2019;150:591–604. doi: 10.1111/jnc.14787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Uchihara T, Giasson BI. Propagation of alpha-synuclein pathology: Hypotheses, discoveries, and yet unresolved questions from experimental and human brain studies. Acta Neuropathol. 2016;131:49–73. doi: 10.1007/s00401-015-1485-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Haque ME, Akther M, Azam S, Kim IS, Lin Y, Lee YH, et al. Targeting α-synuclein aggregation and its role in mitochondrial dysfunction in Parkinson’s disease. Br J Pharmacol. 2022;179:23–45. doi: 10.1111/bph.15684. [DOI] [PubMed] [Google Scholar]
  • 10.Wang C, Yang T, Liang M, Xie J, Song N. Astrocyte dysfunction in Parkinson’s disease: from the perspectives of transmitted α-synuclein and genetic modulation. Transl Neurodegener. 2021;10:39. doi: 10.1186/s40035-021-00265-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rostami J, Mothes T, Kolahdouzan M, Eriksson O, Moslem M, Bergström J, et al. Crosstalk between astrocytes and microglia results in increased degradation of α-synuclein and amyloid-β aggregates. J Neuroinflammation. 2021;18:124. doi: 10.1186/s12974-021-02158-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–2047. doi: 10.1126/science.276.5321.2045. [DOI] [PubMed] [Google Scholar]
  • 13.Krüger R, Kuhn W, Müller T, Woitalla D, Graeber M, Kösel S, et al. Ala30Pro mutation in the gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet. 1998;18:106–108. doi: 10.1038/ng0298-106. [DOI] [PubMed] [Google Scholar]
  • 14.Kaide S, Watanabe H, Shimizu Y, Iikuni S, Nakamoto Y, Hasegawa M, et al. Identification and evaluation of bisquinoline scaffold as a new candidate for α-synuclein-PET imaging. ACS Chem Neurosci. 2020;11:4254–4261. doi: 10.1021/acschemneuro.0c00523. [DOI] [PubMed] [Google Scholar]
  • 15.Kuebler L, Buss S, Leonov A, Ryazanov S, Schmidt F, Maurer A, et al. 11C]MODAG-001-towards a PET tracer targeting α-synuclein aggregates. Eur J Nucl Med Mol Imaging. 2021;48:1759–1772. doi: 10.1007/s00259-020-05133-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Xiang J, Tao Y, Xia Y, Luo S, Zhao Q, Li B, et al. Development of an α-synuclein positron emission tomography tracer for imaging synucleinopathies. Cell. 2023;186:3350–3367.e19. doi: 10.1016/j.cell.2023.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.González-Rodríguez P, Zampese E, Stout KA, Guzman JN, Ilijic E, Yang B, et al. Disruption of mitochondrial complex I induces progressive Parkinsonism. Nature. 2021;599:650–656. doi: 10.1038/s41586-021-04059-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Chondrogiannis S, Marzola MC, Rubello D. 18F-DOPA PET/computed tomography imaging. PET Clin. 2014;9:307–321. doi: 10.1016/j.cpet.2014.03.007. [DOI] [PubMed] [Google Scholar]
  • 19.Chondrogiannis S, Marzola MC, Al-Nahhas A, Venkatanarayana TD, Mazza A, Opocher G, et al. Normal biodistribution pattern and physiologic variants of 18F-DOPA PET imaging. Nucl Med Commun. 2013;34:1141–1149. doi: 10.1097/MNM.0000000000000008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nandhagopal R, Kuramoto L, Schulzer M, Mak E, Cragg J, McKenzie J, et al. Longitudinal evolution of compensatory changes in striatal dopamine processing in Parkinson’s disease. Brain. 2011;134:3290–3298. doi: 10.1093/brain/awr233. [DOI] [PubMed] [Google Scholar]
  • 21.Neves ÂCB, Hrynchak I, Fonseca I, Alves VHP, Pereira MM, Falcão A, et al. Advances in the automated synthesis of 6-[18F]Fluoro-L-DOPA. EJNMMI Radiopharm Chem. 2021;6:11. doi: 10.1186/s41181-021-00126-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Henry JP, Scherman D. Radioligands of the vesicular monoamine transporter and their use as markers of monoamine storage vesicles. Biochem Pharmacol. 1989;38:2395–2404. doi: 10.1016/0006-2952(89)90082-8. [DOI] [PubMed] [Google Scholar]
  • 23.Cho SS, Christopher L, Koshimori Y, Li C, Lang AE, Houle S, et al. Decreased pallidal vesicular monoamine transporter type 2 availability in Parkinson’s disease: the contribution of the nigropallidal pathway. Neurobiol Dis. 2019;124:176–182. doi: 10.1016/j.nbd.2018.11.022. [DOI] [PubMed] [Google Scholar]
  • 24.Kilbourn MR, Koeppe RA. Classics in neuroimaging: radioligands for the vesicular monoamine transporter 2. ACS Chem Neurosci. 2019;10:25–29. doi: 10.1021/acschemneuro.8b00429. [DOI] [PubMed] [Google Scholar]
  • 25.Hsiao IT, Weng YH, Hsieh CJ, Lin WY, Wey SP, Kung MP, et al. Correlation of Parkinson disease severity and 18F-DTBZ positron emission tomography. JAMA Neurol. 2014;71:758–766. doi: 10.1001/jamaneurol.2014.290. [DOI] [PubMed] [Google Scholar]
  • 26.Jiang D, Kong Y, Ren S, Cai H, Zhang Z, Huang Z, et al. Decreased striatal vesicular monoamine transporter 2 (VMAT2) expression in a type 1 diabetic rat model: a longitudinal study using micro-PET/CT. Nucl Med Biol. 2020;82(83):89–95. doi: 10.1016/j.nucmedbio.2020.02.011. [DOI] [PubMed] [Google Scholar]
  • 27.Rinne JO, Laihinen A, Någren K, Ruottinen H, Ruotsalainen U, Rinne UK. PET examination of the monoamine transporter with[11C]beta-CIT and[11C]beta-CFT in early Parkinson’s disease. Synapse. 1995;21:97–103. doi: 10.1002/syn.890210202. [DOI] [PubMed] [Google Scholar]
  • 28.Lundkvist C, Halldin C, Swahn CG, Hall H, Karlsson P, Nakashima Y, et al. O-methyl-11C]beta-CIT-FP, a potential radioligand for quantitation of the dopamine transporter: preparation, autoradiography, metabolite studies, and positron emission tomography examinations. Nucl Med Biol. 1995;22:905–913. doi: 10.1016/0969-8051(95)00029-W. [DOI] [PubMed] [Google Scholar]
  • 29.Hall H, Halldin C, Guilloteau D, Chalon S, Emond P, Besnard J, et al. Visualization of the dopamine transporter in the human brain postmortem with the new selective ligand[125I]PE2I. NeuroImage. 1999;9:108–116. doi: 10.1006/nimg.1998.0366. [DOI] [PubMed] [Google Scholar]
  • 30.Shetty HU, Zoghbi SS, Liow JS, Ichise M, Hong J, Musachio JL, et al. Identification and regional distribution in rat brain of radiometabolites of the dopamine transporter PET radioligand[11C]PE2I. Eur J Nucl Med Mol Imaging. 2007;34:667–678. doi: 10.1007/s00259-006-0277-1. [DOI] [PubMed] [Google Scholar]
  • 31.Varrone A, Tóth M, Steiger C, Takano A, Guilloteau D, Ichise M, et al. Kinetic analysis and quantification of the dopamine transporter in the nonhuman primate brain with 11C-PE2I and 18F-FE-PE2I. J Nucl Med. 2011;52:132–139. doi: 10.2967/jnumed.110.077651. [DOI] [PubMed] [Google Scholar]
  • 32.Varrone A, Steiger C, Schou M, Takano A, Finnema SJ, Guilloteau D, et al. In vitro autoradiography and in vivo evaluation in cynomolgus monkey of[18F]FE-PE2I, a new dopamine transporter PET radioligand. Synapse. 2009;63:871–880. doi: 10.1002/syn.20670. [DOI] [PubMed] [Google Scholar]
  • 33.Volkow ND, Fowler JS, Logan J, Alexoff D, Zhu W, Telang F, et al. Effects of modafinil on dopamine and dopamine transporters in the male human brain: clinical implications. JAMA. 2009;301:1148–1154. doi: 10.1001/jama.2009.351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Olsson H, Halldin C, Swahn CG, Farde L. Quantification of[11C]FLB 457 binding to extrastriatal dopamine receptors in the human brain. J Cereb Blood Flow Metab. 1999;19:1164–1173. doi: 10.1097/00004647-199910000-00013. [DOI] [PubMed] [Google Scholar]
  • 35.Ceccarini J, Vrieze E, Koole M, Muylle T, Bormans G, Claes S, et al. Optimized in vivo detection of dopamine release using 18F-fallypride PET. J Nucl Med. 2012;53:1565–1572. doi: 10.2967/jnumed.111.099416. [DOI] [PubMed] [Google Scholar]
  • 36.Narendran R, Mason NS, Laymon CM, Lopresti BJ, Velasquez ND, May MA, et al. A comparative evaluation of the dopamine D(2/3) agonist radiotracer[11C](-)-N-propyl-norapomorphine and antagonist[11C]raclopride to measure amphetamine-induced dopamine release in the human striatum. J Pharmacol Exp Ther. 2010;333:533–539. doi: 10.1124/jpet.109.163501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Wilson AA, McCormick P, Kapur S, Willeit M, Garcia A, Hussey D, et al. Radiosynthesis and evaluation of[11C]-(+)-4-propyl-3, 4, 4a, 5, 6, 10b-hexahydro-2H-naphtho[1, 2-b][1, 4]oxazin-9-ol as a potential radiotracer for in vivo imaging of the dopamine D2 high-affinity state with positron emission tomography. J Med Chem. 2005;48:4153–4160. doi: 10.1021/jm050155n. [DOI] [PubMed] [Google Scholar]
  • 38.Vercouillie J, Buron F, Sérrière S, Rodrigues N, Gulhan Z, Chartier A, et al. Development and preclinical evaluation of[18F]FBVM as a new potent PET tracer for vesicular acetylcholine transporter. Eur J Med Chem. 2022;244:114794. doi: 10.1016/j.ejmech.2022.114794. [DOI] [PubMed] [Google Scholar]
  • 39.Nordberg A, Hartvig P, Lilja A, Viitanen M, Amberla K, Lundqvist H, et al. Decreased uptake and binding of 11C-nicotine in brain of Alzheimer patients as visualized by positron emission tomography. J Neural Transm Park Dis Dement Sect. 1990;2:215–224. doi: 10.1007/BF02257652. [DOI] [PubMed] [Google Scholar]
  • 40.Sihver W, Fasth KJ, Ogren M, Lundqvist H, Bergström M, Watanabe Y, et al. In vivo positron emission tomography studies on the novel nicotinic receptor agonist[11C]MPA compared with[11C]ABT-418 and (S) (-)[11C]nicotine in rhesus monkeys. Nucl Med Biol. 1999;26:633–640. doi: 10.1016/S0969-8051(99)00034-7. [DOI] [PubMed] [Google Scholar]
  • 41.Valette H, Bottlaender M, Dollé F, Guenther I, Fuseau C, Coulon C, et al. Imaging central nicotinic acetylcholine receptors in baboons with[18F]fluoro-A-85380. J Nucl Med. 1999;40:1374–1380. [PubMed] [Google Scholar]
  • 42.Sabri O, Becker GA, Meyer PM, Hesse S, Wilke S, Graef S, et al. First-in-human PET quantification study of cerebral α4β2* nicotinic acetylcholine receptors using the novel specific radioligand (-)-[(18)F]Flubatine. NeuroImage. 2015;118:199–208. doi: 10.1016/j.neuroimage.2015.05.065. [DOI] [PubMed] [Google Scholar]
  • 43.Wong DF, Kuwabara H, Kim J, Brasic JR, Chamroonrat W, Gao Y, et al. PET imaging of high-affinity α4β2 nicotinic acetylcholine receptors in humans with 18F-AZAN, a radioligand with optimal brain kinetics. J Nucl Med. 2013;54:1308–1314. doi: 10.2967/jnumed.112.108001. [DOI] [PubMed] [Google Scholar]
  • 44.Kilbourn MR, Snyder SE, Sherman PS, Kuhl DE. In vivo studies of acetylcholinesterase activity using a labeled substrate, N-[11C]methylpiperdin-4-yl propionate ([11C]PMP) Synapse. 1996;22:123–131. doi: 10.1002/(SICI)1098-2396(199602)22:2&#x0003c;123::AID-SYN5&#x0003e;3.0.CO;2-F. [DOI] [PubMed] [Google Scholar]
  • 45.Gjerløff T, Jakobsen S, Nahimi A, Munk OL, Bender D, Alstrup AKO, et al. In vivo imaging of human acetylcholinesterase density in peripheral organs using 11C-donepezil: dosimetry, biodistribution, and kinetic analyses. J Nucl Med. 2014;55:1818–1824. doi: 10.2967/jnumed.114.143859. [DOI] [PubMed] [Google Scholar]
  • 46.Hiraoka K, Okamura N, Funaki Y, Watanuki S, Tashiro M, Kato M, et al. Quantitative analysis of donepezil binding to acetylcholinesterase using positron emission tomography and[5-(11)C-methoxy]donepezil. NeuroImage. 2009;46:616–623. doi: 10.1016/j.neuroimage.2009.03.006. [DOI] [PubMed] [Google Scholar]
  • 47.de Natale ER, Wilson H, Politis M. Serotonergic imaging in Parkinson’s disease. Prog Brain Res. 2021;261:303–338. doi: 10.1016/bs.pbr.2020.11.001. [DOI] [PubMed] [Google Scholar]
  • 48.Kish SJ, Tong J, Hornykiewicz O, Rajput A, Chang LJ, Guttman M, et al. Preferential loss of serotonin markers in caudate versus putamen in Parkinson’s disease. Brain. 2008;131:120–131. doi: 10.1093/brain/awm239. [DOI] [PubMed] [Google Scholar]
  • 49.Wersinger C, Rusnak M, Sidhu A. Modulation of the trafficking of the human serotonin transporter by human alpha-synuclein. Eur J Neurosci. 2006;24:55–64. doi: 10.1111/j.1460-9568.2006.04900.x. [DOI] [PubMed] [Google Scholar]
  • 50.Pagano G, Niccolini F, Politis M. The serotonergic system in Parkinson’s patients with dyskinesia: evidence from imaging studies. J Neural Transm. 2018;125:1217–1223. doi: 10.1007/s00702-017-1823-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Fu JF, Matarazzo M, McKenzie J, Neilson N, Vafai N, Dinelle K, et al. Serotonergic system impacts levodopa response in early Parkinson’s and future risk of dyskinesia. Mov Disord. 2021;36:389–397. doi: 10.1002/mds.28340. [DOI] [PubMed] [Google Scholar]
  • 52.Joling M, van den Heuvel OA, Berendse HW, Booij J, Vriend C. Serotonin transporter binding and anxiety symptoms in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2018;89:89–94. doi: 10.1136/jnnp-2017-316193. [DOI] [PubMed] [Google Scholar]
  • 53.Prange S, Metereau E, Maillet A, Klinger H, Schmitt E, Lhommée E, et al. Limbic serotonergic plasticity contributes to the compensation of apathy in early Parkinson’s disease. Mov Disord. 2022;37:1211–1221. doi: 10.1002/mds.28971. [DOI] [PubMed] [Google Scholar]
  • 54.Parsey RV, Kegeles LS, Hwang DR, Simpson N, Abi-Dargham A, Mawlawi O, et al. In vivo quantification of brain serotonin transporters in humans using[11C]McN 5652. J Nucl Med. 2000;41:1465–1477. [PubMed] [Google Scholar]
  • 55.Szabo Z, McCann UD, Wilson AA, Scheffel U, Owonikoko T, Mathews WB, et al. Comparison of (+)-(11)C-McN5652 and (11)C-DASB as serotonin transporter radioligands under various experimental conditions. J Nucl Med. 2002;43:678–692. [PMC free article] [PubMed] [Google Scholar]
  • 56.Huang Y, Hwang DR, Narendran R, Sudo Y, Chatterjee R, Bae SA, et al. Comparative evaluation in nonhuman Primates of five PET radiotracers for imaging the serotonin transporters: [11C]McN 5652, [11C]ADAM, [11C]DASB, [11C]DAPA, and[11C]AFM. J Cereb Blood Flow Metab. 2002;22:1377–1398. doi: 10.1097/01.WCB.0000040948.67415.05. [DOI] [PubMed] [Google Scholar]
  • 57.Walker M, Ehrlichmann W, Stahlschmidt A, Pichler BJ, Fischer K. In vivo evaluation of 11C-DASB for quantitative SERT imaging in rats and mice. J Nucl Med. 2016;57:115–121. doi: 10.2967/jnumed.115.163683. [DOI] [PubMed] [Google Scholar]
  • 58.Doder M, Rabiner EA, Turjanski N, Lees AJ, Brooks DJ, Study 1W 1P Tremor in Parkinson’s disease and serotonergic dysfunction: An 11C-WAY 100635 PET study. Neurology. 2003;60:601–605. doi: 10.1212/01.WNL.0000031424.51127.2B. [DOI] [PubMed] [Google Scholar]
  • 59.Tipre DN, Zoghbi SS, Liow JS, Green MV, Seidel J, Ichise M, et al. PET imaging of brain 5-HT1A receptors in rat in vivo with 18F-FCWAY and improvement by successful inhibition of radioligand defluorination with miconazole. J Nucl Med. 2006;47:345–353. [PubMed] [Google Scholar]
  • 60.Hillmer AT, Wooten DW, Bajwa AK, Higgins AT, Lao PJ, Betthauser TJ, et al. First-in-human evaluation of 18F-mefway, a PET radioligand specific to serotonin-1A receptors. J Nucl Med. 2014;55:1973–1979. doi: 10.2967/jnumed.114.145151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Choi JY, Lyoo CH, Kim JS, Kim KM, Kang JH, Choi SH, et al. 18F-Mefway PET imaging of serotonin 1A receptors in humans: a comparison with 18F-FCWAY. PLoS One. 2015;10:e0121342. doi: 10.1371/journal.pone.0121342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sommerauer M, Fedorova TD, Hansen AK, Knudsen K, Otto M, Jeppesen J, et al. Evaluation of the noradrenergic system in Parkinson’s disease: an 11C-MeNER PET and neuromelanin MRI study. Brain. 2018;141:496–504. doi: 10.1093/brain/awx348. [DOI] [PubMed] [Google Scholar]
  • 63.Severance AJ, Milak MS, Kumar JSD, Prabhakaran J, Majo VJ, Simpson NR, et al. In vivo assessment of[11C]MRB as a prospective PET ligand for imaging the norepinephrine transporter. Eur J Nucl Med Mol Imaging. 2007;34:688–693. doi: 10.1007/s00259-006-0312-2. [DOI] [PubMed] [Google Scholar]
  • 64.Takano A, Halldin C, Farde L. SERT and NET occupancy by venlafaxine and milnacipran in nonhuman Primates: a PET study. Psychopharmacology. 2013;226:147–153. doi: 10.1007/s00213-012-2901-z. [DOI] [PubMed] [Google Scholar]
  • 65.Vase KH, Peters D, Nielsen EØ, Alstrup AKO, Bender D. 11C]NS8880, a promising PET radiotracer targeting the norepinephrine transporter. Nucl Med Biol. 2014;41:758–764. doi: 10.1016/j.nucmedbio.2014.06.004. [DOI] [PubMed] [Google Scholar]
  • 66.Hou L, Rong J, Haider A, Ogasawara D, Varlow C, Schafroth MA, et al. Positron emission tomography imaging of the endocannabinoid system: Opportunities and challenges in radiotracer development. J Med Chem. 2021;64:123–149. doi: 10.1021/acs.jmedchem.0c01459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Burns HD, Van Laere K, Sanabria-Bohórquez S, Hamill TG, Bormans G, Eng WS, et al. 18F]MK-9470, a positron emission tomography (PET) tracer for in vivo human PET brain imaging of the cannabinoid-1 receptor. Proc Natl Acad Sci U S A. 2007;104:9800–9805. doi: 10.1073/pnas.0703472104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Lahdenpohja S, Rajala NA, Helin JS, Haaparanta-Solin M, Solin O, López-Picón FR, et al. Ruthenium-mediated 18F-fluorination and preclinical evaluation of a new CB1 receptor imaging agent[18F]FPATPP. ACS Chem Neurosci. 2020;11:2009–2018. doi: 10.1021/acschemneuro.0c00313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Postuma RB, Berg D, Stern M, Poewe W, Olanow CW, Oertel W, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 2015;30:1591–1601. doi: 10.1002/mds.26424. [DOI] [PubMed] [Google Scholar]
  • 70.Stormezand GN, Chaves LT, Vállez García D, Doorduin J, De Jong BM, Leenders KL, et al. Intrastriatal gradient analyses of 18F-FDOPA PET scans for differentiation of Parkinsonian disorders. Neuroimage Clin. 2020;25:102161. doi: 10.1016/j.nicl.2019.102161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Palermo G, Ceravolo R. Molecular imaging of the dopamine transporter. Cells. 2019;8:872. doi: 10.3390/cells8080872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mo SJ, Axelsson J, Jonasson L, Larsson A, Ögren MJ, Ögren M, et al. Dopamine transporter imaging with[18f]fe-pe2i pet and[123i]fp-cit spect-a clinical comparison. EJNMMI Res. 2018;8:100. doi: 10.1186/s13550-018-0450-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Delva A, Van Weehaeghe D, van Aalst J, Ceccarini J, Koole M, Baete K, et al. Quantification and discriminative power of 18F-FE-PE2I PET in patients with Parkinson’s disease. Eur J Nucl Med Mol Imaging. 2020;47:1913–1926. doi: 10.1007/s00259-019-04587-y. [DOI] [PubMed] [Google Scholar]
  • 74.Lee CS, Samii A, Sossi V, Ruth TJ, Schulzer M, Holden JE, et al. In vivo positron emission tomographic evidence for compensatory changes in presynaptic dopaminergic nerve terminals in Parkinson’s disease. Ann Neurol. 2000;47:493–503. doi: 10.1002/1531-8249(200004)47:4&#x0003c;493::AID-ANA13&#x0003e;3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 75.Liu XL, Liu SY, Barret O, Tamagnan GD, Qiao HW, Song TB, et al. Diagnostic value of striatal 18F-FP-DTBZ PET in Parkinson’s disease. Front Aging Neurosci. 2022;14:931015. doi: 10.3389/fnagi.2022.931015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cheon M, Kim SM, Ha SW, Kang MJ, Yang HE, Yoo J. Diagnostic performance for differential diagnosis of atypical parkinsonian syndromes from Parkinson’s disease using quantitative indices of 18F-FP-CIT PET/CT. Diagnostics. 2022;12:1402. doi: 10.3390/diagnostics12061402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zhao Y, Wu P, Wu J, Brendel M, Lu J, Ge J, et al. Decoding the dopamine transporter imaging for the differential diagnosis of Parkinsonism using deep learning. Eur J Nucl Med Mol Imaging. 2022;49:2798–2811. doi: 10.1007/s00259-022-05804-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Appel L, Jonasson M, Danfors T, Nyholm D, Askmark H, Lubberink M, et al. Use of 11C-PE2I PET in differential diagnosis of parkinsonian disorders. J Nucl Med. 2015;56:234–242. doi: 10.2967/jnumed.114.148619. [DOI] [PubMed] [Google Scholar]
  • 79.Chou KL, Dayalu P, Koeppe RA, Gilman S, Spears CC, Albin RL, et al. Serotonin transporter imaging in multiple system atrophy and Parkinson’s disease. Mov Disord. 2022;37:2301–2307. doi: 10.1002/mds.29220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Shinotoh H, Namba H, Yamaguchi M, Fukushi K, Nagatsuka S, Iyo M, et al. Positron emission tomographic measurement of acetylcholinesterase activity reveals differential loss of ascending cholinergic systems in Parkinson’s disease and progressive supranuclear palsy. Ann Neurol. 1999;46:62–69. doi: 10.1002/1531-8249(199907)46:1&#x0003c;62::AID-ANA10&#x0003e;3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 81.Gilman S, Koeppe RA, Nan B, Wang CN, Wang X, Junck L, et al. Cerebral cortical and subcortical cholinergic deficits in parkinsonian syndromes. Neurology. 2010;74:1416–1423. doi: 10.1212/WNL.0b013e3181dc1a55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kerstens VS, Fazio P, Sundgren M, Brumberg J, Halldin C, Svenningsson P, et al. Longitudinal DAT changes measured with[18F]FE-PE2I PET in patients with Parkinson’s disease; a validation study. NeuroImage Clin. 2023;37:103347. doi: 10.1016/j.nicl.2023.103347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kumar A, Mann S, Sossi V, Ruth TJ, Stoessl AJ, Schulzer M, et al. 11C]DTBZ-PET correlates of levodopa responses in asymmetric Parkinson’s disease. Brain. 2003;126:2648–2655. doi: 10.1093/brain/awg270. [DOI] [PubMed] [Google Scholar]
  • 84.Boertien JM, van der Zee S, Chrysou A, Gerritsen MJJ, Jansonius NM, Spikman JM, et al. Study protocol of the DUtch PARkinson Cohort (DUPARC): a prospective, observational study of de novo Parkinson’s disease patients for the identification and validation of biomarkers for Parkinson’s disease subtypes, progression and pathophysiology. BMC Neurol. 2020;20:245. doi: 10.1186/s12883-020-01811-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Gjerløff T, Fedorova T, Knudsen K, Munk OL, Nahimi A, Jacobsen S, et al. Imaging acetylcholinesterase density in peripheral organs in Parkinson’s disease with 11C-donepezil PET. Brain. 2015;138:653–663. doi: 10.1093/brain/awu369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Knudsen K, Fedorova TD, Hansen AK, Sommerauer M, Otto M, Svendsen KB, et al. In-vivo staging of pathology in REM sleep behaviour disorder: a multimodality imaging case-control study. Lancet Neurol. 2018;17:618–628. doi: 10.1016/S1474-4422(18)30162-5. [DOI] [PubMed] [Google Scholar]
  • 87.Horsager J, Andersen KB, Knudsen K, Skjærbæk C, Fedorova TD, Okkels N, et al. Brain-first versus body-first Parkinson’s disease: a multimodal imaging case-control study. Brain. 2020;143:3077–3088. doi: 10.1093/brain/awaa238. [DOI] [PubMed] [Google Scholar]
  • 88.Son HJ, Oh JS, Oh M, Lee SJ, Oh SJ, Chung SJ, et al. Test-retest reproducibility of dopamine transporter density measured with[18F]FP-CIT PET in patients with essential tremor and Parkinson’s disease. Ann Nucl Med. 2021;35:299–306. doi: 10.1007/s12149-020-01561-9. [DOI] [PubMed] [Google Scholar]
  • 89.Choi BW, Kang S, Kim HW, Kwon OD, Vu HD, Youn SW. Faster region-based convolutional neural network in the classification of different Parkinsonism patterns of the Striatum on maximum intensity projection images of[18F]FP-CIT positron emission tomography. Diagnostics. 2021;11:1557. doi: 10.3390/diagnostics11091557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sun X, Ge J, Li L, Zhang Q, Lin W, Chen Y, et al. Use of deep learning-based radiomics to differentiate Parkinson’s disease patients from normal controls: a study based on[18F]FDG PET imaging. Eur Radiol. 2022;32:8008–8018. doi: 10.1007/s00330-022-08799-z. [DOI] [PubMed] [Google Scholar]
  • 91.Tian M, He X, Jin C, He X, Wu S, Zhou R, et al. Transpathology: Molecular imaging-based pathology. Eur J Nucl Med Mol Imaging. 2021;48:2338–2350. doi: 10.1007/s00259-021-05234-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.DaSilva JN, Kilbourn MR. In vivo binding of[11C]tetrabenazine to vesicular monoamine transporters in mouse brain. Life Sci. 1992;51:593–600. doi: 10.1016/0024-3205(92)90228-H. [DOI] [PubMed] [Google Scholar]
  • 93.Kilbourn MR, Sherman PS, Abbott LC. Mutant mouse strains as models for in vivo radiotracer evaluations: [11C]methoxytetrabenazine ([11C]MTBZ) in tottering mice. Nucl Med Biol. 1995;22:565–567. doi: 10.1016/0969-8051(95)00005-I. [DOI] [PubMed] [Google Scholar]
  • 94.Bohnen NI, Kanel P, Müller MLTM. Molecular imaging of the cholinergic system in Parkinson’s disease. Int Rev Neurobiol. 2018;141:211–250. doi: 10.1016/bs.irn.2018.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Rogers GA, Stone-Elander S, Ingvar M, Eriksson L, Parsons SM, Widén L. 18F-labelled vesamicol derivatives: syntheses and preliminary in vivo small animal positron emission tomography evaluation. Nucl Med Biol. 1994;21:219–230. doi: 10.1016/0969-8051(94)90012-4. [DOI] [PubMed] [Google Scholar]
  • 96.Gage HD, Voytko ML, Ehrenkaufer RL, Tobin JR, Efange SM, Mach RH. Reproducibility of repeated measures of cholinergic terminal density using. J Nucl Med. 2000;41:2069–2076. [PubMed] [Google Scholar]
  • 97.Coughlin JM, Slania S, Du Y, Rosenthal HB, Lesniak WG, Minn I, et al. 18F-XTRA PET for enhanced imaging of the extrathalamic α4β2 nicotinic acetylcholine receptor. J Nucl Med. 2018;59:1603–1608. doi: 10.2967/jnumed.117.205492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Horti AG, Gao Y, Kuwabara H, Wang Y, Abazyan S, Yasuda RP, et al. 18F-ASEM, a radiolabeled antagonist for imaging the α7-nicotinic acetylcholine receptor with PET. J Nucl Med. 2014;55:672–677. doi: 10.2967/jnumed.113.132068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Aznavour N, Zimmer L. 18F]MPPF as a tool for the in vivo imaging of 5-HT1A receptors in animal and human brain. Neuropharmacology. 2007;52:695–707. doi: 10.1016/j.neuropharm.2006.09.023. [DOI] [PubMed] [Google Scholar]
  • 100.Horti AG, Fan H, Kuwabara H, Hilton J, Ravert HT, Holt DP, et al. 11C-JHU75528: a radiotracer for PET imaging of CB1 cannabinoid receptors. J Nucl Med. 2006;47:1689–1696. [PubMed] [Google Scholar]
  • 101.Borgan F, Laurikainen H, Veronese M, Marques TR, Haaparanta-Solin M, Solin O, et al. In vivo availability of cannabinoid 1 receptor levels in patients with first-episode psychosis. JAMA Psychiatry. 2019;76:1074–1084. doi: 10.1001/jamapsychiatry.2019.1427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wilson H, Dervenoulas G, Pagano G, Koros C, Yousaf T, Picillo M, et al. Serotonergic pathology and disease burden in the premotor and motor phase of A53T α-synuclein Parkinsonism: a cross-sectional study. Lancet Neurol. 2019;18:748–759. doi: 10.1016/S1474-4422(19)30140-1. [DOI] [PubMed] [Google Scholar]
  • 103.Nahimi A, Sommerauer M, Kinnerup MB, Østergaard K, Winterdahl M, Jacobsen J, et al. Noradrenergic deficits in parkinson disease imaged with 11C-MeNER. J Nucl Med. 2018;59:659–664. doi: 10.2967/jnumed.117.190975. [DOI] [PubMed] [Google Scholar]
  • 104.Wang J, Zuo CT, Jiang YP, Guan YH, Chen ZP, Xiang JD, et al. 18F-FP-CIT PET imaging and SPM analysis of dopamine transporters in Parkinson’s disease in various Hoehn & Yahr stages. J Neurol. 2007;254:185–190. doi: 10.1007/s00415-006-0322-9. [DOI] [PubMed] [Google Scholar]
  • 105.Oh M, Lee N, Kim C, Son HJ, Sung C, Oh SJ, et al. Diagnostic accuracy of dual-phase 18F-FP-CIT PET imaging for detection and differential diagnosis of Parkinsonism. Sci Rep. 2021;11:14992. doi: 10.1038/s41598-021-94040-8. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neuroscience Bulletin are provided here courtesy of Springer

RESOURCES