Background
Dysregulation of adenosine homeostasis has been linked to aging-related neurodegenerative diseases [
1,
2]. We recently reported that a 7-day chronic intraperitoneal (i.p.) injection of the adenosine A1 receptor (A1R) agonist N
6-Cyclopentyladenosine (CPA) produced similar levels of α-synuclein (α-Syn) upregulation and neurodegeneration as in 5-week daily i.p. CPA injections [
3,
4]. Therefore, the 7-day CPA chronic injection represented a convenient α-synucleinopathy model to test novel compounds with neuroprotective potential and drugs that could increase the CPA-induced neurotoxicity. Some neuroprotective drugs have been found to bind to α-Syn and prevent further aggregation, including caffeine, nicotine, 1-aminoindan and metformin [
5]. Conversely, there are other drugs such as methamphetamine, cocaine, 2-aminoindan and the herbicides, paraquat and rotenone, which appear to be neurotoxic because they increase α-Syn misfolding and can be correlated with a higher incidence of PD [
6,
7,
8]. Nonetheless, even with drugs such as caffeine or nicotine, there are several problems including being unsafe at high doses and causing numerous side effects [
9]. However, it was argued that linking these compounds and other similar neuroprotective drugs in dimers or trimers would decrease the misfolding and aggregation of α-Syn without risking the safety of the patients [
10]. These compounds can be also referred to as bifunctional drugs [
11]. They are connected via a six-carbon alkyl linker to minimize solubility issues and retain the flexibility that will allow further binding to α-Syn. Their synthesis and purity have been previously described [
10].
The dimers were tested in vitro using techniques such as nanopore analysis and it was established that they bind to α-Syn and do not cause a compact conformation [
10]. Focusing on the caffeine-indan (C
8-6-I) dimer, the isothermal titration calorimetry results indicated that the dimer has 1 binding site with α-Syn and a binding constant of 5.3*10
4 M
-1, thus it appears to be neuroprotective [
10]. Furthermore, the C
8-6-I dimer was successful in rescuing one and two-copied α-Syn-Green Fluorescent Protein (GFP) yeast strain from the α-Syn-induced cell death under the control of a galactose promoter. This dimer was also effective in preventing large α-Syn foci in the one and two-copied α-Syn-GFP yeast [
10]. Present in vitro studies of the C
8-6-I dimer were conducted using molecular docking simulations [
12]. This technique allows the prediction of the binding conformation of the eight in vitro α-Syn structures published from the Chen group [
13,
14] with our dimer. This technique is helpful in further elucidating the effect of the dimer on the misfolding patterns of α-Syn. Moreover, in the present study, the C
8-6-I was tested in combination with the A1R agonist CPA. Following 7-day i.p. treatments with vehicle control or with CPA (3mg/kg) in absence or presence of C
8-6-I (3 or 5 mg/kg), the animals underwent a series of behavioural tests such as Y-maze, open field, and forced swim test. We hypothesized that systemic administration of the dimer would inhibit the CPA-induced upregulation and aggregation of α-Syn similar to the previously reported neuroprotective effects of the A1R antagonist DPCPX and 1-aminoindan [
3,
4]. We will use molecular docking to determine whether the dimer interacts with α-Syn to induce a neuroprotective “loop” conformation, which could explain reduced neurodegeneration and improved behavioural outcomes of animals.
To test the blood-brain barrier permeability and distribution of the dimer we also performed an in vivo study with CD-11 albino mice using PET imaging. Before PET imaging, the dimer was radiolabeled with fluoride-18 [
18F] radioisotope [
11]. Therefore, PET imaging allows the quantitation of drug concentration, distribution, and pharmacokinetics of radiotracers [
15], as well as providing information about physiology such as metabolism, receptor concentration, and transport across cell membranes [
16,
17], as a result, PET is a useful tool in drug development [
18,
19,
20]. Since the pharmacological activity of a drug depends on the concentration reaching the tissue of interest, establishing the concentration of free drug reaching organs/tissues of interest and its full biodistribution is an important starting point for early phase development [
17]. PET can be used to non-invasively infer molecule distribution in vivo and brain exposure [
17,
21]. Recent advances in radiochemistry now mean many drug molecules can be rapidly labelled with radionuclides such as
11C or
18F and used for biodistribution studies [
22,
23]. Radiotracers are commonly used in non-clinical studies to assess the drug absorption, distribution, metabolism and excretion (ADME) profile of a drug [
24]. To measure the biodistribution of C
8-6-I, we have now developed a rapid and robust synthesis of the
18F-labelled analogue (as seen in
Figure 1) for PET imaging and biodistribution studies. Here we also report the radiosynthesis, purification, and in vivo PET imaging and ex vivo biodistribution of
18F- C
8-6-I in healthy CD-1 mice.
Discussion
Many epidemiological and multipurpose drug studies have presented interesting observations, such as the consumption of coffee or tobacco significantly decreasing the risk of PD [
32,
33,
34,
35,
36,
37]. Recently it was shown that caffeine as well as nicotine are neuroprotective against wild-type and mutant parkin proteins [
9]. The substitution of glutamic acid to lysine (E28K) in the ubiquitin-like binding domain of the parkin protein is strongly associated with early onset PD but also with several cancers like onset melanoma [
38,
39]. Caffeine and nicotine were shown to bind directly to the 28th amino acid residue of both parkin and E28K mutant [
9]. These two drugs show neuroprotective effects by decreasing the misfolding of the two proteins. Another interesting finding was the unexpected effects of metformin, a drug used to treat type 2 diabetes, which has been reported to be neuroprotective against PD in long-term studies [
40,
41,
42]. Another study looked at the role of metformin in reducing mitochondrial hyperactivity [
43]. The dysfunction of the branched-chain amino acid (BCAA) metabolism pathway was linked to PD-like motor deficits and neurodegeneration. This neurodegeneration was linked directly to hyperactivity of the mitochondria; however, metformin decreased the mitochondria respiration levels. Mitochondria dysfunction has been previously linked with PD in numerous studies [
44,
45,
46]. Moreover, as previously mentioned, 1-aminoindan, a metabolite of rasagiline, is also shown to be neuroprotective [
47,
48,
49]. 1-aminoindan was shown to bind directly to α-Syn, thereby promoting a neuroprotective “loop” conformation which attenuates the α-Syn misfolding and aggregation in a yeast model overexpressing α-Syn or in an in vivo rodent model of α-Synucleinopathy [
3,
5,
6,
27].
Although drugs such as caffeine, nicotine or 1-aminoindan would be clinically relevant for reducing the risk of developing PD, these drugs may be highly toxic at high concentrations [
50,
51,
52]. In addition, there are several side effects of consuming high doses of caffeine or nicotine such as anxiety and nervousness, fast heart rate, heartburn, high blood pressure, cancer and others [
53,
54,
55]. Therefore, linking these compounds and other similar neuroprotective drugs into dimers or trimers may be a more effective strategy to reduce the misfolding and aggregation of α-Syn without risking the safety of the patients [
10,
11,
56]. These compounds were optimized with a six-carbon alkyl linker, to minimize solubility issues and retain the flexibility that will allow further binding to α-Syn and protect the protein from misfolding and aggregating [
10]. Thus, the present study investigated the role of C
8-6-I dimer as a promising neuroprotective agent in our established in vivo 7-day CPA injection α-synucleinopathy model, which displayed increased α-Syn expression, aggregation, and neurodegeneration [
3]. Determining the dimer concentration is difficult to attain confidently via conventional approaches, especially for tissue compartments with specialized epithelial or endothelial cell barriers such as the central nervous system (CNS) [
17]. For example, the blood-brain barrier (BBB) prevents compounds, typically hydrophilic, from entering the brain and is a major obstacle that must be overcome during drug development. A lack of adequate brain exposure to drugs often leads to the failure of CNS drug candidates [
22,
57]. Lipophilic compounds that can passively diffuse across membranes may also be substrates of efflux transporters (such as P-glycoproteins, organic anion transporters, and multidrug-resistance-associated proteins) at the BBB resulting in low brain penetration of pharmacologically active drug molecules [
22]. To then predict the BBB permeability of
18F-C
8-6-I, we calculated the CNS multiparameter optimization (MPO) desirability score using the compound’s physicochemical properties [
58]. For the CNS MPO score, six physicochemical parameters (calculated LogP, calculated LogD, molecular weight, topological polar surface area, hydrogen bond donor, and acidity constant) are normalized and converted to a function ranging from 0 to 1, and the sum of all the normalized physicochemical properties produce the CNS MPO score ranging from 0 to 6. Generally, therapeutic agents with preferable BBB permeability show CNS MPO scores of 4 or higher [
59,
60,
61,
62,
63,
64]. The CNS MPO score for
18F-C
8-6-I was calculated as 4.2. Based on these initial results from binding studies with isothermal titration calorimetry and in vitro yeast assay indicating binding to α-Syn [
10], in vitro metabolic studies [
11], kinetic measurement and pharmacokinetic predictions [
31], and CNS MPO desirability score, we synthesized
18F-C
8-6-I for in vivo evaluation in healthy mice. We found that
18F-C
8-6-I was distributed in the brain regions, including the hippocampus and midbrain, which supports the reduced α-Syn expression and aggregation and neurodegeneration observed in these regions. Although our PET-CT studies showed
18F-C
8-6-I biodistribution and slow elimination in different organs, including the brain, it is yet unclear whether this PET-tracer compound is binding directly to α-Syn or other targets in the brain to mediate the neuroprotection and mitigate behavioural deficits. However, our present molecular docking results and previous reports using nanopore analysis [
10] confirm that the C
8-6-I dimer compound likely binds to α-Syn to promote a neuroprotective “loop” conformation that reduces α-Syn misfolding and aggregation. Moreover, our molecular docking results also indicate that the C
8-6-I compound could bind to other targets, including adenosine receptors (see below), which further contributes to the observed neuroprotective effects of the dimer in the hippocampus and SN brain regions.
In previous studies, we demonstrated that chronic A1R stimulation with 5 mg/kg CPA for 5 weeks showed a significant increase in α-Syn in the midbrain region and produced motor and cognitive deficits in male Sprague-Dawley rats [
4]. Recently, we also reported a novel 7-day i.p. injection model of 3 mg/kg CPA indicating similar findings in increased α-Syn expression and aggregation in both the SN pars compacta and the hippocampus [
3]. This was demonstrated using confocal images as well as Western blotting techniques. We also showed that chronic CPA treatments increased neurodegeneration levels, as shown by the FJC staining. However, the co-administration of DPCPX or 1-aminoindan prevented the CPA-induced α-Syn aggregation and neurodegeneration [
3]. Interestingly, we showed comparable results with the C
8-6-I dimer. We tested two doses of the dimer, 3 and 5 mg/kg. Following the 7th day of injections, the Sprague-Dawley rats were tested for cognitive dysfunction using the Y-maze test, anxiety levels using the open field test, and kinetic/despair behaviour using the forced swim test. We showed that chronic administration of CPA induced hippocampal-dependent spatial memory deficit, which was attenuated by co-administration with both doses of the dimer. During the open field test, the CPA group spent significantly less time exploring the center as shown by the significantly lower percentage of time spent in the center square. The co-administration of the dimer compound improved the exploratory behaviour and reduced the anxiety caused by CPA. Moreover, the forced swim test indicated that the CPA-treated rats exhibited reduced vigour and success scores, indicating reduced motor behaviour possibly due to increased despair/helplessness behaviour. Both doses of C
8-6-I used (3 and 5 mg/kg) were effective in restoring cognitive function, anxiety levels and motor activity, and these results are consistent with the observed decrease in neurodegeneration in the hippocampus and SN pars compacta. We did not test the effects of higher doses due to the difficulty in synthesizing and obtaining a high yield of the dimer. However, the initial dose of 3-5 mg/kg C
8-6-I dimer was sufficient to produce neuroprotective benefits in our α-synucleinopathy model. It is also important to note that the C
8-6-I dimer is a racemic mixture, and it is not known whether one or both enantiomers are responsible for the activity.
In addition, previous studies with C
8-6-I and
18F-C
8-6-I indicated that these compounds were promising as CNS PET tracers [
11,
31]. We previously determined that C
8-6-I directly interacted with α-Syn in vitro and rescued yeast cells from α-Syn fibrillation mediated toxicity [
10]. In the present study, we reported that both the 3 and 5 mg/kg doses of the dimer prevented the CPA-induced α-Syn accumulation and aggregation in both the SN pars compacta and hippocampal CA1 region. This coincides with the significant reduction of FJC neurodegeneration levels in both the dopaminergic cells of the SN pars compacta and the pyramidal neurons of the CA1 region of the hippocampus. We further synthesized
19F-C
8-6-I and determined that
19F-C
8-6-I possessed a similar metabolic profile to C
8-6-I in mouse, rat, and human liver microsomes and the metabolites are 1-aminoindan hydroxylation, 1-aminoindan
N-dealkylation,
N3 and
N1-demethylation with the exception of alkyl hydroxylation only found in
19F-C
8-6-I [
11]. No defluorination was observed as this is important for the development of PET probes since fluoride ions can accumulate in the skull and bones, cause toxicity and confound the interpretation of PET images [
11]. To further understand the in vivo parameters of C
8-6-I and
19F-C
8-6-I, we measured and reported the unbound intrinsic clearance, CL
int, in mouse liver microsomes and extrapolated to predict in vivo clearance in mouse plasma, CL
p using the well-stirred model. We reported a predicted CL
p of 36.0 ml/min/kg for
19F-C
8-6-I compared to 34.3 ml/min/kg for C
8-6-I [
31]. These metabolic stability studies indicated that similar metabolites were observed in mouse, rat and human microsomes, suggesting that mice and rats are appropriate models for future animal studies of
18F-C
8-6-I. Emerging evidence supports the key role of the gut-brain connection in the pathogenesis of PD [
65,
66,
67]. Our pharmacokinetics studies demonstrated a relatively high level of intestinal localization of
18F-C
8-6-I dimer probes which further suggests the potential applicability of our probe in gut-brain studies.
We also tested whether C
8-6-I could act on other targets, including A1Rs and A2ARs, since this bifunctional compound contains the caffeine moiety which could retain its binding affinity for adenosine receptors. Whereas caffeine non-selectively binds to the respective orthosteric adenosine binding sites in A1Rs and A2ARs [
30], the C
8-6-I dimer appears to bind preferentially to the A1R orthosteric site. Using molecular docking, we found that C
8-6-I interacted with the A1R orthosteric binding site and additional putative allosteric binding sites were also identified. In contrast, C
8-6-I did not make similar contacts with the orthosteric binding site or allosteric binding site for A2AR. Important amino acid residues in the second extracellular loop (ECL2) of A1R have been identified to play a crucial role in A1R allosteric modulation. Previous studies using an all-atom Gaussian accelerated molecular dynamics (GaMD) simulation using an inactive A1R structure (PDP: 5UEN) supported a key role of residue E172 (in the ECL2 region) as an important binding determinant for the two positive allosteric modulators (PAMs), PD71723 and VCP171. Alanine substitution of E172 decreased the binding affinity of two allosteric modulators, PD81723 and VCP171 [
68,
69]. The residues E171 (also in ECL2) and N254 on TM6 are known to interact with orthosteric antagonists DU172 and PSB36 [
68] as well as endogenous agonist, adenosine (PDB:6D9H) [
28]. In the present study, we observed the binding of C
8-6-I to the PAM-binding residue E172 and the orthosteric agonist/antagonist binding residue F171. Since we found that C
8-6-I was effective in preventing neurodegeneration of hippocampal and substantia nigral neurons, which likely depended on preserving A1R expression, this suggests that C
8-6-I may act as an A1R PAM that promotes less A1R desensitization compared to A1R orthosteric agonists, adenosine, and CPA. However, the C
8-6-I binding to the orthosteric site F171 also suggests that C
8-6-I may reduce agonist binding and A1R activity. Although the influence of C
8-6-I allosteric binding on A1R binding and function has yet to be investigated, it is also possible that the allosteric mechanism may involve decreasing the rate of orthosteric agonist dissociation and enhancing agonist binding and function [
70]. This potential A1R allosteric ligand property of C
8-6-I may be therapeutically beneficial and warrants further studies due to potential therapeutic implications. If C
8-6-I is an allosteric modulator of A1R, it will be important to carry out further concentration-response analysis, as some A1R PAMs have been shown to act as inhibitors at high concentrations and cause a functional response in the absence of an agonist (e.g., T62) while some compounds (e.g., PD81723 derivative with bridged 3- and 4- positions) exhibited A1R antagonism at low concentrations, but PAM activity at higher concentrations [
68,
71].
In summary, we showed further evidence that a 7-day chronic CPA i.p. injection rat model generated increased α-synuclein expression and aggregation, which provides a convenient model to assay several neuroprotective drugs, including the bifunctional compound C8-6-I, without waiting several months to observe the presence of α-synucleinopathies normally seen in genetic mouse of models of PD (e.g., A53T mouse model). Our results indicate that the C8-6-I dimer compound promotes neuroprotection by decreasing the CPA-induced α-synucleinopathy and neurodegeneration in the hippocampus and SN pars compacta. This likely involves C8-6-I directly binding to α-Syn domains that promote the neuroprotective “loop” conformation that prevents α-Syn misfolding and aggregation. Moreover, directly linking caffeine to 1-aminoindan also appears to increase the caffeine moiety’s preferential binding to the A1R adenosine orthosteric binding site. This implies that the observed biodistribution of C8-6-I dimer inside the CNS makes it an ideal therapeutic for PD, as this bifunctional drug promotes neuroprotection by inhibiting α-Syn misfolding and aggregation and by suppressing chronic adenosine A1 receptor stimulation which is expected to prevent α-Syn over-expression. Finally, this study indicates that C8-6-I may be an appropriate PET-CT tracer that can be used for imaging diagnostics and treatment of PD.
Figure 1.
Radiosynthesis of the C8-6-I dimer and in vivo studies with the CD-1 mice. a. Formation reaction of 18F-C8-6-I from C8-6-I-OMs in 23 ± 5% rcy (decay corrected). b. All steps involved in the b.1 radiosynthesis of 18F-C8-6-I from [18F]fluorine purification, b.2 reaction with Kryptofix/K18F and b.3 further purification to b.4 PET-imaging and b.5 biodistribution of the 18F-C8-6-I in major organs. The data was then analyzed and graphed using GraphPad Prism 8 (San Diego, CA, USA). Created using BioRender.com.
Figure 1.
Radiosynthesis of the C8-6-I dimer and in vivo studies with the CD-1 mice. a. Formation reaction of 18F-C8-6-I from C8-6-I-OMs in 23 ± 5% rcy (decay corrected). b. All steps involved in the b.1 radiosynthesis of 18F-C8-6-I from [18F]fluorine purification, b.2 reaction with Kryptofix/K18F and b.3 further purification to b.4 PET-imaging and b.5 biodistribution of the 18F-C8-6-I in major organs. The data was then analyzed and graphed using GraphPad Prism 8 (San Diego, CA, USA). Created using BioRender.com.
Figure 2.
Behavioural tests conducted for male Sprague Dawley rats after the 7-day chronic injection of C8-6-I at 3 and 5 mg/kg. a. Y-maze test values of the 7-day chronic C8-6-I dimer (3 mg/kg and 5mg/kg) as percentage of time spend in each of the arms: S-arm (“start” arm), O-arm (“old” arm), and N-arm (“new’ arm). The percentages of the time spent in each arm were calculated from the 5-minute trial. Open field test values of the 7-day chronic C8-6-I dimer (3 mg/kg and 5mg/kg) as percentage of time spend in the center square. The animal is placed in the center square of the grid and free to explore the field for 10 minutes. b. The percentage of the time spend in the red center square. c. The total fecal boli count. Forced swim test results of the 7-day chronic C8-6-I dimer (3 mg/kg and 5mg/kg) treatment groups as measurements of swimming vigorously and successfully. The animals were placed on the forced swim tank and let free to swim for 10 minutes. Once the test was conducted the animals were scored for d. Vigor, the ability to purposely swim and use all limbs and e. Success, the ability to keep their head above water. f. The total time spent immobile was also measured to assess learned helplessness and despair. Each dimer treatment was repeated at least 10 times per treatment (n=10) and the average of each treatment is presented in bar graphs as means ± SEM. Significances were determined using One-way ANOVA, followed by Student-Newman-Keuls multiple comparison test with ns p > 0.05; * p < 0.05; ** p < 0.01; and *** p < 0.001.
Figure 2.
Behavioural tests conducted for male Sprague Dawley rats after the 7-day chronic injection of C8-6-I at 3 and 5 mg/kg. a. Y-maze test values of the 7-day chronic C8-6-I dimer (3 mg/kg and 5mg/kg) as percentage of time spend in each of the arms: S-arm (“start” arm), O-arm (“old” arm), and N-arm (“new’ arm). The percentages of the time spent in each arm were calculated from the 5-minute trial. Open field test values of the 7-day chronic C8-6-I dimer (3 mg/kg and 5mg/kg) as percentage of time spend in the center square. The animal is placed in the center square of the grid and free to explore the field for 10 minutes. b. The percentage of the time spend in the red center square. c. The total fecal boli count. Forced swim test results of the 7-day chronic C8-6-I dimer (3 mg/kg and 5mg/kg) treatment groups as measurements of swimming vigorously and successfully. The animals were placed on the forced swim tank and let free to swim for 10 minutes. Once the test was conducted the animals were scored for d. Vigor, the ability to purposely swim and use all limbs and e. Success, the ability to keep their head above water. f. The total time spent immobile was also measured to assess learned helplessness and despair. Each dimer treatment was repeated at least 10 times per treatment (n=10) and the average of each treatment is presented in bar graphs as means ± SEM. Significances were determined using One-way ANOVA, followed by Student-Newman-Keuls multiple comparison test with ns p > 0.05; * p < 0.05; ** p < 0.01; and *** p < 0.001.
Figure 3.
Molecular docking simulation of α-Syn structures a. C1, b. C2, c. C3, and d. C8 bound to C8-6-I. Bold black dashed lines and amino acid residues indicate hydrogen bonding, while the grey dashed lines and amino acid residues indicate hydrophobic interactions. a. The bifunctional dimer compound forms hydrogen bond with T81 located in the NAC region of α-Syn, and additional hydrophobic interactions are found with the α-Syn N-terminus and NAC region. b. The dimer compound interacts via hydrogen bonding to α-Syn N-terminus, and additional hydrophobic interactions occur with amino acid residues in N-terminus and NAC region. c. The dimer compound binds via hydrogen bonds to amino acid residues in α-Syn N-terminus and NAC region, and additional hydrophobic binding occurs with amino acids located in the distal N-terminus and NAC region. d. The dimer compound binds via hydrogen bonding with NAC amino acid residues (V66, T72) and through hydrophobic interactions with amino acid residues in N-terminus and NAC domain. C1, C2, C3 and C8 α-Syn structures bind to the dimer compound which is predicted to form a “loop” conformation of α-Syn.
Figure 3.
Molecular docking simulation of α-Syn structures a. C1, b. C2, c. C3, and d. C8 bound to C8-6-I. Bold black dashed lines and amino acid residues indicate hydrogen bonding, while the grey dashed lines and amino acid residues indicate hydrophobic interactions. a. The bifunctional dimer compound forms hydrogen bond with T81 located in the NAC region of α-Syn, and additional hydrophobic interactions are found with the α-Syn N-terminus and NAC region. b. The dimer compound interacts via hydrogen bonding to α-Syn N-terminus, and additional hydrophobic interactions occur with amino acid residues in N-terminus and NAC region. c. The dimer compound binds via hydrogen bonds to amino acid residues in α-Syn N-terminus and NAC region, and additional hydrophobic binding occurs with amino acids located in the distal N-terminus and NAC region. d. The dimer compound binds via hydrogen bonding with NAC amino acid residues (V66, T72) and through hydrophobic interactions with amino acid residues in N-terminus and NAC domain. C1, C2, C3 and C8 α-Syn structures bind to the dimer compound which is predicted to form a “loop” conformation of α-Syn.
Figure 4.
Molecular docking of C8-6-I with A1R and A2AR. a. Amino acid sequence alignment of human A1R and A2AR with distinct binding of C8-6-I to A1R and A2AR indicated (red asterisks, A1R binding; blue asterisks, A2AR binding). b. Molecular docking of C8-6-I with A1R showing binding to amino acid residues that are similarly found within the A1R orthosteric binding site for adenosine. c. Molecular docking showing C8-6-I binding to amino acid residues that do not resemble those associated with A2AR orthosteric binding site.
Figure 4.
Molecular docking of C8-6-I with A1R and A2AR. a. Amino acid sequence alignment of human A1R and A2AR with distinct binding of C8-6-I to A1R and A2AR indicated (red asterisks, A1R binding; blue asterisks, A2AR binding). b. Molecular docking of C8-6-I with A1R showing binding to amino acid residues that are similarly found within the A1R orthosteric binding site for adenosine. c. Molecular docking showing C8-6-I binding to amino acid residues that do not resemble those associated with A2AR orthosteric binding site.
Figure 5.
Summary of the surface area analysis of the dimer study’s pars compacta region of the substantia nigra of DAPI, TH, and α-Syn. a. Representative images of 40 μm pars compacta region of substantia nigra taken with 63X oil immersion objective of a confocal microscope (126 times magnification). Separate channels and montages of 7-day chronic intraperitoneal injections with 3 mg/kg of the following treatments: Control (DMSO/Saline), CPA, C8-6-I (3 mg/kg) + CPA, and C8-6-I (5 mg/kg) + CPA. Slices were probed for DAPI (Blue), TH (Green, Alexa Fluor 555), and α-Syn (Red, Alexa Fluor 647). b. Representative images from 40 μm nigral brain slices of rats after probing for DAPI, anti-α-Synuclein and Thioflavin S taken at 63 times magnification with a confocal microscope. Scale 50 μm. c. Bar charts showing the mean area intensities of α-Syn and Thioflavin S in the pars compacta region of the substantia nigra of the eight treatments, respectively. Similar areas of 100 by 100 μm ROI coordinates for lateral pars compacta of SN were quantified respectively for each slice and normalized by subtracting F0 (50 by 50 μm ROI coordinates) values of the background (non-cell body bottom area). Average intensity values and correlation coefficients in bars represent the average mean ± SEM from n=4 independent experiments. Significances were determined using One-way ANOVA, followed by Student-Newman-Keuls multiple comparison test with ns p > 0.05; * p < 0.05; ** p < 0.01; and *** p < 0.001.
Figure 5.
Summary of the surface area analysis of the dimer study’s pars compacta region of the substantia nigra of DAPI, TH, and α-Syn. a. Representative images of 40 μm pars compacta region of substantia nigra taken with 63X oil immersion objective of a confocal microscope (126 times magnification). Separate channels and montages of 7-day chronic intraperitoneal injections with 3 mg/kg of the following treatments: Control (DMSO/Saline), CPA, C8-6-I (3 mg/kg) + CPA, and C8-6-I (5 mg/kg) + CPA. Slices were probed for DAPI (Blue), TH (Green, Alexa Fluor 555), and α-Syn (Red, Alexa Fluor 647). b. Representative images from 40 μm nigral brain slices of rats after probing for DAPI, anti-α-Synuclein and Thioflavin S taken at 63 times magnification with a confocal microscope. Scale 50 μm. c. Bar charts showing the mean area intensities of α-Syn and Thioflavin S in the pars compacta region of the substantia nigra of the eight treatments, respectively. Similar areas of 100 by 100 μm ROI coordinates for lateral pars compacta of SN were quantified respectively for each slice and normalized by subtracting F0 (50 by 50 μm ROI coordinates) values of the background (non-cell body bottom area). Average intensity values and correlation coefficients in bars represent the average mean ± SEM from n=4 independent experiments. Significances were determined using One-way ANOVA, followed by Student-Newman-Keuls multiple comparison test with ns p > 0.05; * p < 0.05; ** p < 0.01; and *** p < 0.001.
Figure 6.
Summary of the surface area analysis of the CA1 region of the hippocampus of DAPI, α-Syn and Thio-S. a. Representative images from 40 μm hippocampal rat brain slices after probing for DAPI, anti-α-Synuclein and Thioflavin S taken at 63 times magnification with a confocal microscope for the following treatments: Control (DMSO/Saline), CPA, C8-6-I (3 mg/kg) + CPA, and C8-6-I (5 mg/kg) + CPA. Scale 50 μm. b. Bar charts showing the mean area intensities of α-Syn and Thioflavin S in the CA1 region of the hippocampus of the eight treatments. Fluorescence intensities from a 100 by 100 μm ROI from the CA1 pyramidal cell layer of the hippocampus were quantified using a similar method employed for pars compacta region. Average intensity values and correlation coefficients in bars represent the average mean ± SEM from n=4 independent experiments. Significances were determined using One-way ANOVA, followed by Student-Newman-Keuls multiple comparison test with ns p > 0.05; * p < 0.05; ** p < 0.01; and *** p < 0.001.
Figure 6.
Summary of the surface area analysis of the CA1 region of the hippocampus of DAPI, α-Syn and Thio-S. a. Representative images from 40 μm hippocampal rat brain slices after probing for DAPI, anti-α-Synuclein and Thioflavin S taken at 63 times magnification with a confocal microscope for the following treatments: Control (DMSO/Saline), CPA, C8-6-I (3 mg/kg) + CPA, and C8-6-I (5 mg/kg) + CPA. Scale 50 μm. b. Bar charts showing the mean area intensities of α-Syn and Thioflavin S in the CA1 region of the hippocampus of the eight treatments. Fluorescence intensities from a 100 by 100 μm ROI from the CA1 pyramidal cell layer of the hippocampus were quantified using a similar method employed for pars compacta region. Average intensity values and correlation coefficients in bars represent the average mean ± SEM from n=4 independent experiments. Significances were determined using One-way ANOVA, followed by Student-Newman-Keuls multiple comparison test with ns p > 0.05; * p < 0.05; ** p < 0.01; and *** p < 0.001.
Figure 7.
Fluoro-Jade C (FJC) staining in the SN pars compacta and in the hippocampus CA1 region of rats with 7-day chronic intraperitoneal injection of Control (DMSO/saline), CPA, C8-6-I (3 mg/kg) + CPA, and C8-6-I (5 mg/kg) + CPA. Representative images with 50 μm scale bar for the a. SN pars compacta and b. the CA1 region of the hippocampus. FJC fluorescence intensity in a 100 × 100 μm2 region was normalized to the control group (100%). Values are shown as mean ± SEM. The average FJC fluorescence values were obtained from n = 4 independent experiments. ns, non-significant; *P < 0.05; **P < 0.01; and ***P < 0.001 (one-way ANOVA followed by Student-Newman-Keuls post-hoc multiple comparison test).
Figure 7.
Fluoro-Jade C (FJC) staining in the SN pars compacta and in the hippocampus CA1 region of rats with 7-day chronic intraperitoneal injection of Control (DMSO/saline), CPA, C8-6-I (3 mg/kg) + CPA, and C8-6-I (5 mg/kg) + CPA. Representative images with 50 μm scale bar for the a. SN pars compacta and b. the CA1 region of the hippocampus. FJC fluorescence intensity in a 100 × 100 μm2 region was normalized to the control group (100%). Values are shown as mean ± SEM. The average FJC fluorescence values were obtained from n = 4 independent experiments. ns, non-significant; *P < 0.05; **P < 0.01; and ***P < 0.001 (one-way ANOVA followed by Student-Newman-Keuls post-hoc multiple comparison test).
Figure 8.
Ex vivo stability and biodistribution in CD-1 mice at five different time points (5, 10, 20, 40, and 60 min) and major organs. a. HPLC co-registration profiles of 18F-C8-6-I and 19F-C8-6-I based on a radio detector and ultraviolet detector, respectively. Analytical radio-HPLC chromatograms in mouse b. liver and c. pancreas extracts at 40 mins after injection of 18F-C8-6-I. d. The distribution of 18F-C8-6-I was calculated as percentage of the injected dose per gram of tissue (% ID/g) and results were displayed into two groups: Lower distribution—Blood, heart, bone, and brain (left panel) and higher distribution—Liver, duodenum, kidneys, spleen, lungs, and large intestine (right panel). The data were obtained from n = 3 independent animals. Values are presented as mean ± SEM.
Figure 8.
Ex vivo stability and biodistribution in CD-1 mice at five different time points (5, 10, 20, 40, and 60 min) and major organs. a. HPLC co-registration profiles of 18F-C8-6-I and 19F-C8-6-I based on a radio detector and ultraviolet detector, respectively. Analytical radio-HPLC chromatograms in mouse b. liver and c. pancreas extracts at 40 mins after injection of 18F-C8-6-I. d. The distribution of 18F-C8-6-I was calculated as percentage of the injected dose per gram of tissue (% ID/g) and results were displayed into two groups: Lower distribution—Blood, heart, bone, and brain (left panel) and higher distribution—Liver, duodenum, kidneys, spleen, lungs, and large intestine (right panel). The data were obtained from n = 3 independent animals. Values are presented as mean ± SEM.
Figure 9.
Representative PET/CT images in CD-1 mice at different time points throughout an hour as well as time activity curve in the brain. a. PET images at different time points (1 min, 5 min, 10 min, 20 min, 30 min, and 50 min), b. PET summation images for 60 min and c. 120 min dynamic imaging. d. Time-activity curve (TAC) of 18F-C8-6-I for whole brain and three region consisting of the cortex, midbrain, and hippocampus from the PET/CT imaging. Values are presented as standardized uptake value (SUV). The data were obtained from n = 3 independent animals.
Figure 9.
Representative PET/CT images in CD-1 mice at different time points throughout an hour as well as time activity curve in the brain. a. PET images at different time points (1 min, 5 min, 10 min, 20 min, 30 min, and 50 min), b. PET summation images for 60 min and c. 120 min dynamic imaging. d. Time-activity curve (TAC) of 18F-C8-6-I for whole brain and three region consisting of the cortex, midbrain, and hippocampus from the PET/CT imaging. Values are presented as standardized uptake value (SUV). The data were obtained from n = 3 independent animals.
Table 2.
Biodistribution of 18F-C8-6-I in healthy CD-1 mice. Data shown are percentage of injected dose per gram of tissue (%ID/g). The values are presented as mean ± SEM, n = 3.
Table 2.
Biodistribution of 18F-C8-6-I in healthy CD-1 mice. Data shown are percentage of injected dose per gram of tissue (%ID/g). The values are presented as mean ± SEM, n = 3.
Organ |
5 min |
10 min |
20 min |
40 min |
60 min |
Blood |
0.76 ± 0.040 |
0.78 ± 0.14 |
0.67 ± 0.19 |
0.48 ± 0.029 |
0.39 ± 0.11 |
Liver |
5.12 ± 1.65 |
6.91 ± 2.02 |
6.64 ± 1.44 |
4.76 ± 0.54 |
5.74 ± 1.12 |
Small intestine |
8.56 ± 2.02 |
11.18 ± 3.11 |
13.02 ± 1.24 |
9.35 ± 1.82 |
12.33 ± 0.81 |
Kidney |
16.26 ± 1.82 |
11.86 ± 1.74 |
11.02 ± 0.48 |
8.65± 1.21 |
4.27 ± 0.37 |
Spleen |
6.54 ± 0.88 |
5.49 ± 1.05 |
5.71 ± 0.69 |
5.80 ± 0.83 |
3.79 ± 0.33 |
Lung |
7.03 ± 1.24 |
5.51 ± 0.44 |
5.52 ± 0.72 |
3.00 ± 1.40 |
1.42 ±0.22 |
Heart |
2.34 ± 0.26 |
1.77 ± 0.17 |
1.14 ± 0.040 |
1.01 ± 0.16 |
0.49 ± 0.018 |
Large intestine |
2.98 ± 0.94 |
3.06 ± 0.73 |
2.83 ± 0.068 |
2.69 ± 0.26 |
2.50 ± 0.93 |
Bone |
1.00 ± 0.18 |
1.24 ± 0.44 |
1.18 ± 0.005 |
1.04 ± 0.27 |
1.20 ± 0.34 |
Brain |
0.17 ± 0.014 |
0.14 ± 0.027 |
0.10 ± 0.013 |
0.10 ± 0.011 |
0.05 ± 0.005 |
Table 1.
HPLC gradient elution profile for the chromatographic purification of 18F-C8-6-I radiotracer. Mobile Phase A—0.1% (v/v) formic acid in water and mobile phase B—0.1% (v/v) formic acid in acetonitrile.
Table 1.
HPLC gradient elution profile for the chromatographic purification of 18F-C8-6-I radiotracer. Mobile Phase A—0.1% (v/v) formic acid in water and mobile phase B—0.1% (v/v) formic acid in acetonitrile.
Time (min) |
Flow Rate (ml/min) |
Mobile Phase A (%) |
Mobile Phase B (%) |
0.00 |
5 |
90 |
10 |
3.00 |
5 |
90 |
10 |
7.00 |
5 |
60 |
40 |
10.00 |
5 |
40 |
60 |
15.00 |
5 |
10 |
90 |
17.00 |
5 |
10 |
90 |
18.00 |
5 |
90 |
10 |
21.00 |
5 |
90 |
10 |