ANA 12; ANA-12; N-(2-(((Hexahydro-2-oxo-1H-azepin-3-yl)amino)carbonyl)phenyl)benzo(b)thiophene-2-carboxamide; N-[2-[[(Hexahydro-2-oxo-1H-azepin-3-yl)amino]carbonyl]phenyl]benzo[b]thiophene-2-carboxamide; ANA-12; 219766-25-3; N-(2-((2-oxoazepan-3-yl)carbamoyl)phenyl)benzo[b]thiophene-2-carboxamide; N-[2-[(2-oxoazepan-3-yl)carbamoyl]phenyl]-1-benzothiophene-2-carboxamide; ANA12
Size | Price | Stock | Qty |
---|---|---|---|
5mg |
|
||
10mg |
|
||
25mg |
|
||
50mg |
|
||
100mg |
|
||
250mg |
|
||
500mg |
|
||
Other Sizes |
|
Purity: ≥98%
ANA-12 (ANA12) is a novel, potent and selective TrkB inhibitor/antagonist with important biological activity. It has a Kd of 10 nM for the high affinity site of TrkB and 12 μM for the low affinity site.
Targets |
TrkB (Kd = 10 nM)
|
---|---|
ln Vitro |
ANA-12 has a direct and specific binding to TrkB, blocking its downstream processes without changing the functions of TrkA or TrkC. ANA-12, even at concentrations as low as 10 nM, inhibits brain-derived neurotrophic factor (BDNF)-induced neurite outgrowth in nnr5 PC12-TrkB cells.[1] ANA-12 blocks BDNF's ability to increase inward current in DRG neurons.[2]
Screening of N-T19 analogs reveals a potent TrkB antagonist. [1] KIRA-ELISA and neurite outgrowth assessments revealed that only N-T19 was able to maintain its effects in neurons and neuron-like systems. However, although N-T19 demonstrated a high efficacy in inhibiting TrkB activity, its relatively low potency prompted us to seek out analogs that keep the original high efficacy of N-T19 but with a better potency. For this purpose, a second round of in silico screening using the Bioinfo-DB database was undertaken to identify analogs of N-T19 sharing the same molecular scaffold (Figure 4A). Fourteen new molecules were identified as close analogs and were tested in a first set of functional screenings using KIRA-ELISA assays. Four compounds were found to have the highest activity but only 1 (ANA-12) demonstrated a submicromolar potency in recombinant cells. In-depth pharmacological characterizations of ANA-12 confirmed the high efficacy (full inhibition) and potency (submicromolar) of the molecule (Figure 4B). Like the parent compound NT-19, ANA-12 revealed a 2-site mode of action in neurons but surprisingly also in recombinant cells. Potencies on the low- and high-affinity sites were comparable between the 2 cell systems (50 μM and 50 nM, respectively). It is noteworthy that, of the 14 molecules tested, the structure of ANA-12 was the closest to that of the parent lead compound, both molecules differing only by an extra benzene moiety in ANA-12 (Figure 4A). ANA-12 binds directly and selectively to TrkB. [1] We then determined whether ANA-12 binds directly to TrkB by incubating the fluorescently labeled compound (see Methods) with chimeric TrkBECD-Fc or BSA or IgG-Fc as negative controls for nonspecific binding. As shown in Figure 5A, ANA-12 specifically bound to the extracellular domain of TrkB in a dose-dependent manner but not to BSA or IgG-Fc. Saturation binding studies with TrkBECD-Fc showed that ANA-12 binds TrkBECD-Fc with a Kd of 12 μM (Figure 5B), which corresponds to the functional low-affinity site previously observed in KIRA-ELISA. The detection of the high-affinity site was located in the nonlinear range of fluorescence detection and therefore appeared as a small indentation. Linearization and extrapolation analyses showed that ANA-12 also binds the high-affinity site with a Kd of approximately 10 nM (data not shown). A 2-site fitting model of ANA-12 binding to TrkB demonstrated that the high-affinity site accounts for 20% of the total binding (data not shown), a value similar to the 30% observed in KIRA-ELISA assays (Figure 4B). To further study the binding properties of ANA-12, BDNF was added to the ANA-12/TrkB complex (Figure 5B). This resulted in a 60% decrease in the maximal amount of ANA-12 bound to TrkBECD-Fc with no shift of the curve to the right (Kd, 16 μM), suggesting a noncompetitive mechanism. Together, these data suggest that both the high- and low-affinity binding sites coexist on the extracellular domain of TrkB and that BDNF and ANA-12 do not compete for the same sites on TrkB. Computational docking of the new compound to TrkB-d5 demonstrated that the putative binding mode was similar to that of N-T19 (Figure 5C): the lactame moiety of ANA-12 interacts with the main chain atoms of His299 and His300 of TrkB, while hydrogen bonds are formed between the acyclic amide moiety of the compound and the side chains of TrkB-specific Gln347 and Asp298. The shape of the ligand fits into the TrkB-d5 ADEB β-sheet, notably through hydrophobic contacts between the 7-membered ring and the accessible disulfide bridge Cys302-Cys347, and through aromatic-aromatic interactions with a bundle of histidine residues (His299, His300, His335). ANA-12 affects cell functions associated with TrkB but not TrkA nor TrkC. [1] Neurite outgrowth was used to verify the effects of ANA-12 on cellular processes (Figure 5, D and E). We observed that in the TrkB-expressing cells, ANA-12 prevented BDNF-induced neurite outgrowth at concentrations as low as 10 nM, confirming the high potency and efficacy observed in KIRA-ELISA assays (Figure 5D). At concentrations up to 10–100 μM, ANA-12 completely abolished the effects of BDNF since no single neurite or branching could be observed even after 3 days of incubation. To assess the selectivity of the compound toward TrkB, we used 2 other nnr5-PC12 cell lines expressing either TrkA or TrkC for which neurite outgrowth is dependent on NGF and NT-3, respectively (Figure 5E). In these cell lines, ANA-12 had no effect on neurite outgrowth. Three days incubation with concentrations as high as 100 μM of ANA-12 did not affect NGF- and NT-3–dependent neurite length and branching, confirming its specificity to TrkB-related signaling. |
ln Vivo |
ANA-12 (0.5 mg/kg, i.p.) reduces anxiety and depression-related behaviors in adult C57BL6/129SveV F1s mice without compromising neuron survival. It also lowers TrKB activity in the brain.[1] Analogous amideamide-12 (0.5 mg/kg, i.p.) reverses the depressive-like effects of lipopolysaccharide-induced behavior in male C57BL/6 mice.[3] The effect of medial nucleus tractus solitarius (mNTS) BDNF on decreasing food intake is blocked in male Sprague-Dawley rats by ANA-12 (3 μg/dose).[4] ANA-12 reverses ethanol intake and causes D3 receptor downregulation in male wild-type mice, but it has no effect on D3R-/-mice.[5] The reduced self-administration of cocaine in male CocSired rats is reversed by ANA-12 (0.5 mg/kg, i.p.).[6]
NMDA receptors in primary afferent terminals can contribute to hyperalgesia by increasing neurotransmitter release. In rats and mice, we found that the ability of intrathecal NMDA to induce neurokinin 1 receptor (NK1R) internalization (a measure of substance P release) required a previous injection of BDNF. Selective knock-down of NMDA receptors in primary afferents decreased NMDA-induced NK1R internalization, confirming the presynaptic location of these receptors. The effect of BDNF was mediated by tropomyosin-related kinase B (trkB) receptors and not p75 neurotrophin receptors (p75(NTR) ), because it was not produced by proBDNF and was inhibited by the trkB antagonist ANA-12 but not by the p75(NTR) inhibitor TAT-Pep5. These effects are probably mediated through the truncated form of the trkB receptor as there is little expression of full-length trkB in dorsal root ganglion (DRG) neurons. Src family kinase inhibitors blocked the effect of BDNF, suggesting that trkB receptors promote the activation of these NMDA receptors by Src family kinase phosphorylation. Western blots of cultured DRG neurons revealed that BDNF increased Tyr(1472) phosphorylation of the NR2B subunit of the NMDA receptor, known to have a potentiating effect. Patch-clamp recordings showed that BDNF, but not proBDNF, increased NMDA receptor currents in cultured DRG neurons. NMDA-induced NK1R internalization was also enabled in a neuropathic pain model or by activating dorsal horn microglia with lipopolysaccharide. These effects were decreased by a BDNF scavenger, a trkB receptor antagonist and a Src family kinase inhibitor, indicating that BDNF released by microglia potentiates NMDA receptors in primary afferents during neuropathic pain. [1] LPS caused a reduction of BDNF in the CA3 and dentate gyrus (DG) of the hippocampus and prefrontal cortex (PFC), whereas LPS increased BDNF in the nucleus accumbens (NAc). Dexamethason suppression tests showed hyperactivity of the hypothalamic-pituitary-adrenal axis in LPS-treated mice. Intraperitoneal (i.p.) administration of 7,8-DHF showed antidepressant effects on LPS-induced depression-like behavior, and i.p. pretreatment with ANA-12 blocked its antidepressant effects. Surprisingly, ANA-12 alone showed antidepressant-like effects on LPS-induced depression-like behavior. Furthermore, bilateral infusion of ANA-12 into the NAc showed antidepressant effects. Moreover, LPS caused a reduction of spine density in the CA3, DG, and PFC, whereas LPS increased spine density in the NAc. Interestingly, 7,8-DHF significantly attenuated LPS-induced reduction of p-TrkB and spine densities in the CA3, DG, and PFC, whereas ANA-12 significantly attenuated LPS-induced increases of p-TrkB and spine density in the NAc. Conclusions: The results suggest that LPS-induced inflammation may cause depression-like behavior by altering BDNF and spine density in the CA3, DG, PFC, and NAc, which may be involved in the antidepressant effects of 7,8-DHF and ANA-12, respectively. [2] To localize the neurons mediating the energy balance effects of hindbrain ventricle-delivered BDNF, ventricle subthreshold doses were delivered directly to medial nucleus tractus solitarius (mNTS). mNTS BDNF administration reduced food intake significantly, and this effect was blocked by preadministration of a highly selective TrkB receptor antagonist {[N2-2-2-Oxoazepan-3-yl amino]carbonyl phenyl benzo (b)thiophene-2-carboxamide (ANA-12)}, suggesting that TrkB receptor activation mediates hindbrain BDNF's effect on food intake. Because both BDNF and leptin interact with melanocortin signaling to reduce food intake, we also examined whether the intake inhibitory effects of hindbrain leptin involve hindbrain-specific BDNF/TrkB activation. BDNF protein content within the dorsal vagal complex of the hindbrain was increased significantly by hindbrain leptin delivery. To assess if BDNF/TrkB receptor signaling acts downstream of leptin signaling in the control of energy balance, leptin and ANA-12 were coadministered into the mNTS. Administration of the TrkB receptor antagonist attenuated the intake-suppressive effects of leptin, suggesting that mNTS TrkB receptor activation contributes to the mediation of the anorexigenic effects of hindbrain leptin. Collectively, these results indicate that TrkB-mediated signaling in the mNTS negatively regulates food intake and, in part, the intake inhibitory effects of leptin administered into the NTS. [3] Indeed, blockade of the BDNF pathway by the TrkB selective antagonist ANA-12 reversed chronic stable ethanol intake and strongly decreased the striatal expression of D3R. Finally, we evaluated buspirone, an approved drug for anxiety disorders endowed with D3R antagonist activity (confirmed by molecular modeling analysis), that resulted effective in inhibiting ethanol intake. Thus, DA signaling via D3R is essential for ethanol-related reward and consumption and may represent a novel therapeutic target for weaning. [4] Administration of a BDNF receptor antagonist (the TrkB receptor antagonist ANA-12) reversed the diminished cocaine self-administration in male cocaine-sired rats. In addition, the association of acetylated histone H3 with Bdnf promoters was increased in the sperm of sires that self-administered cocaine. Collectively, these findings indicate that voluntary paternal ingestion of cocaine results in epigenetic reprogramming of the germline, having profound effects on mPFC gene expression and resistance to cocaine reinforcement in male offspring. [5] |
Enzyme Assay |
Various concentrations of Trk BECD -Fc, 20 mg/ml BSA, or 1 mg/mL IgG-Fc (polyclonal anti-TrkB) are coated on Maxisorp ELISA 96-well plates and left overnight at 4°C in a carbonate buffer with a pH of 9.6. After two hours of room temperature solubility in 0.5% BSA in PBS, plates are thoroughly cleaned in PBS-Tween 0.05%. Following an hour of room temperature incubation in 0.5% PBS-BSA, BDNF is added to the solution and incubated for an additional hour. This process is repeated with Bodipy-ANA-12. The amount of bodipy-ANA-12 bound is measured by fluorescence at 520 ± 10 nm following thorough washes in PBS-Tween 0.05%. To determine the detectability range for extrapolation analysis, ELISA plates are coated with bodipy-ANA-12, and the fluorescence at 520 ± 10 nm is measured.
ANA-12 binding assays [1] Maxisorp ELISA 96-well plates were coated with various concentrations of TrkBECD-Fc (as indicated in the figures), 20 mg/ml BSA, or 1 mg/ml IgG-Fc (polyclonal anti-TrkB;) in a carbonate buffer (pH 9.6) overnight at 4°C. Plates were saturated with 0.5% BSA in PBS for 2 hours at room temperature and extensively washed in PBS-Tween 0.05%. Bodipy–ANA-12 was then incubated in 0.5% PBS-BSA for 1 hour at room temperature before the addition of BDNF in 0.5% PBS-BSA for another hour, as indicated in the figures. After extensive washes in PBS-Tween 0.05%, the amount of bodipy–ANA-12 bound was quantified by fluorescence at 520 ± 10 nm. Detectability range for extrapolation analysis was assessed by coating ELISA plates with bodipy–ANA-12 and reading fluorescence at 520 ± 10 nm. |
Cell Assay |
In nnr5 PC12-TrkB, -TrkA, and -TrkC cells, the effects of molecules on neurite outgrowth are evaluated following the addition of BDNF (1 nM), NGF (2 nM), and NT-3 (10 nM), respectively. A microscope counts the number of cells in each counting field—three wells per condition, two fields per well—that have neurites longer than two cells in diameter. For three days, the counting is done in the dark every 24 hours.
KIRA-ELISA [1] TrkB receptor autophosphorylation was quantified using a modified version of KIRA-ELISA, as described earlier. TetOn-rhTrkB cells were seeded on flat-bottom 96-well culture plates (4 × 104 cells per well) and were incubated overnight with 1000 ng/ml doxycycline to induce the expression of TrkB receptors. Cortical neurons were seeded on polyornithin-coated flat-bottom 96-well culture plates (12 × 104 cells per well) and cultured 7 to 8 days at 37°C in 5% CO2. Fluorescence levels in TetOn-rhTrkB cells were verified before each assay. Cells were carefully washed 4 times with DMEM before being treated with compounds for 20 minutes and stimulated for 20 minutes with BDNF (recombinant cells, 4 nM; neurons, 0.4 nM) in DMEM containing 0.5% BSA and 25 mM HEPES (control medium) at 37°C in 5% CO2. Assay was stopped by removing the medium on ice, and membranes were solubilized by adding a solubilization buffer (150 mM NaCl, 50 mM Hepes, 0.5% Triton X-100, 0.01% thimerosal, 2 mM sodium orthovanadate, supplemented with a cocktail of protease inhibitor) for 1 hour at room temperature. Lysates were transferred to an ELISA microtiter plate precoated with either anti-GFP (1:5000) for rhTrkB or anti-TrkB (1 μg/ml) for neuronal TrkB. Phosphorylation was revealed by incubation with biotinylated anti-phosphotyrosine (0.5 μg/ml) and HRP-conjugated streptavidin (1:4000). After addition of TMB and acidification with 1 N hydrochloric acid, absorbance was read at 450 nm. Total TrkB was also quantified using KIRA-ELISA using either a monoclonal anti-GFP (1:3000) for rhTrkB or a monoclonal anti-TrkB (1:1000) for neuronal TrkB to precipitate the receptor and either a polyclonal anti-GFP (1:5000) for rhTrkB or a polyclonal anti-TrkB (1 μg/ml) for neuronal TrkB to detect the receptor. In cell cultures, the total TrkB signal was found to be unchanged in all treatment conditions. In brain tissues, the amount of total TrkB was variable and was used to normalize the signal obtained for phospho-TrkB. |
Animal Protocol |
In a vehicle of 17% dimethyl sulfoxide (DMSO) in phosphate-buffered saline, ketamine (ketamine hydrochloride, 10 mg/kg), 7,8-dihydroxyflavone (7,8-DHF; 10 mg/kg), and ANA-12, N2-(2-{[(2-oxoazepan-3-yl) amino]carbonyl}phenyl)benzo[b]thiophene-2-carboxamide (0.5 mg/kg) are prepared on the day of injection. Ketamine (10 mg/kg), 7,8-DHF (10 mg/kg), and ANA-12 (0.5 mg/kg) are the doses that have been chosen. Mice receive intraperitoneal (i.p.) administration of all compounds.
Administration of ANA-12 and in vivo KIRA-ELISA analysis [1] F1 hybrids obtained by crossing C57BL/6 and 129SveV mice were used following the recommendation of the Banbury Conference on genetic background for studying mouse behavior. Adult C57BL6/129SveV F1s (3 months old) were randomly distributed into saline (1% DMSO dissolved in 0.9% NaCl solution) and ANA-12 (dissolved in saline solution) groups. A volume of 10 μl/g body weight was injected i.p. for saline and ANA-12 (0.5 mg/kg body weight) solutions. After 2 or 4 hours, mice were decapitated and brains were rapidly removed on ice. Striatum, cortex, and hippocampus were subsequently dissected when needed. Tissues were rapidly washed in ice-cold PBS, transferred into ice-cold KIRA-ELISA solubilization buffer, and left overnight at 4°C. Protein concentrations were determined, equal amounts of proteins were loaded, and KIRA-ELISA assays were performed as described above. Analysis of ANA-12 stability and bioavailability in mouse brains [1] Analysis of stability in mouse serum and quantification in mouse brain were performed by TechMedILL facilities (École Supérieure de Biotechnologie de Strasbourg, Illkirch, France). Stability was assessed by incubating ANA-12 in mouse serum for 15, 30, 45, and 60 minutes at 37°C. Mixtures were homogenized and proteins precipitated with acetonitrile before being analyzed by liquid chromatography–mass spectrometry (Agilent LC-MS ESI qTOF connected to a C18-1 × 10 × 1.9 column). Bioavailability in mouse brain was assessed by injecting mice i.p. with ANA-12 (0.5 mg/kg) as described above. After 30 minutes, 1, 2, 4, and 6 hours (3 mice/time point), brains were removed and mashed in NaCl 0.9% before treatment with acetonitrile. The mixture was cleared by ultracentrifugation, and the compound was extracted using a series of dehydrations and resuspensions in acetonitrile/water (1/1 v/v) before being subjected to LC-MS for detection and analysis. Reference samples were prepared by adding known amounts of compound to blank mixtures (brains from saline-treated mice). Concentrations of ANA-12 (ng/g brain) were derived from the reference samples by calculating the area under the detection peak. In vivo cell death analysis [1] The effects of subchronic treatments of ANA-12 on cell death in mouse brains were assessed using the TUNEL assay. Mice were randomly distributed into 4 groups and were i.p. injected with saline (containing 1% DMSO) or 0.5, 1.0, or 2.0 mg/kg of ANA-12 once a day for 1 week. Mice were sacrificed 24 hours after the last injection and perfused transcardially with PBS followed by 4% paraformaldehyde. Brains were postfixed overnight in 4% paraformaldehyde, and 50 μm–thick coronal sections were obtained using a vibratome. Free-floating sections were extensively washed in PBS and permeabilized in PBS containing 0.5% Triton X-100 for 90 minutes at room temperature. Sections were then rinsed and mounted on slides. Labeling of 3′ OH-DNA strand breaks was performed using the DeadEnd Fluorometric TUNEL system according to the manufacturer’s instruction. The reaction was terminated by rinsing the slides in SSC 2× and washing extensively with PBS. Sections were mounted with VECTASHIELD plus DAPI, and fluorescein was visualized at 520 nm using fluorescence microscopy. Positive controls were obtained by pretreating sections from saline-treated animals with DNAse (10 U/ml) for 90 minutes at 37°C in PBS containing 0.5% Triton X-100. Negative controls (absence of fluorescent spots) were obtained by processing the sections as described above without the terminal transferase. Behavioral testing of ANA-12 [1] All behavioral testing was started at the same time of the day (1:00 pm) in quiet and separated rooms under bright ambient light conditions (800–900 lux, except for the open field, 300–400 lux). Behavioral tasks were performed 4 hours after injection of 0.5 mg/kg of ANA-12 using the same procedure as described above. To eliminate odor cues, all testing apparatus was thoroughly cleaned after each animal using the disinfectant Roccal. Effects of ANA-12 on anxiety-related behaviors [1] Anxiety-related behaviors were tested using the open field test, elevated plus maze, and novelty-suppressed feeding paradigm. Drug Administration [2] ANA-12, N2-(2-{[(2-oxoazepan-3-yl) amino]carbonyl}phenyl)benzo[b]thiophene-2-carboxamide (0.5mg/kg, i.p.), was dissolved in 1% dimethylsulfoxide in physiological saline. The doses of 7,8-DHF and ANA-12 were also selected as previously reported (Ren et al., 2013 2014; Cazorla et al., 2011). Surgery and Bilateral Injection of ANA-12 into NAc [2] Mice were anesthetized with pentobarbital (5mg/kg), and placed in a stereotaxic frame. Microinjection needles were placed bilaterally into the NAc shell (+1.7 AP, ±0.75 ML, -3.6 DV) (Paxinos and Watson, 1998). Twenty-four hours after surgery, LPS (0.5mg/kg) or saline (10ml/kg) was injected i.p. Twenty-three hours after injection of LPS (or saline), ANA-12 (0.1 nmol/L, 0.1 μL/min for 5min) or vehicle was injected bilaterally. Behavioral evaluation was performed 4 and 6 hours after the final infusion (Figure 3B). Experiment 4: BDNF/TrkB receptor signaling within the mNTS. [3] All rats (n = 9) received two unilateral NTS intraparenchyma injections that were ∼20 min apart, and food intake was determined at 1, 3, 6, and 24 h after the second injection. Conditions were counterbalanced and were as follows: control condition (100 nl of DMSO followed by 100 nl of aCSF), ANA-12 condition (1 or 3 μg of ANA-12 followed by aCSF), BDNF condition (DMSO followed by 0.2 μg of BDNF), and the combination condition (either 1 or 3 μg of ANA-12 followed by 0.2 μg of BDNF). Body weight measurements were made immediately before and 24 h after the injection of the drugs. Experiment 6: hindbrain TrkB receptor signaling and leptin. [3] All rats (n = 9) received two unilateral NTS parenchyma injections that were ∼20 min apart, and food intake was measured at 1, 3, 6, and 24 h after the second injection. The four counterbalanced conditions were as follows: control condition (100 nl of DMSO followed by 100 nl of sodium bicarbonate), ANA-12 condition (3 μg of ANA-12 followed by sodium bicarbonate), leptin condition (DMSO followed by 0.2 μg of leptin), and the combination condition (3 μg of ANA-12 followed by 0.2 μg of leptin). Body weight measurements were made immediately before and 24 h after the injection of drugs. Drugs and Treatments [4] Ethanol, U99194A maleate, SB277011A hydrochloride, buspirone hydrochloride, 8-OH-DPAT and ANA-12 were dissolved in saline and intraperitoneally (i.p.) injected (in a volume of 10 ml/kg), except ANA-12 that was dissolved in 10% dimethyl sulfoxide. U99194A was used at 10 mg/kg (Harrison and Nobrega, 2009), SB277011A was used at 10 mg/kg (Song et al, 2012), buspirone was used in the range 0.1–10 mg/kg (Martin et al, 1992), 8-OH-DPAT was used at 1 mg/kg (Martin et al, 1992), and ANA-12 was used at 0.5 mg/kg (Cazorla et al, 2011). In the two-bottle choice paradigm, after 30 days of voluntary alcohol-drinking procedure, D3R−/− and WT were randomly allocated to the eight experimental groups (n=6/10 per group): WT/vehicle, WT/U99194A, WT/SB277011A, WT/buspirone, D3R−/−/vehicle, D3R−/−/U99194A, D3R−/−/SB277011A, and D3R−/−/buspirone. Animals were i.p. injected once a day, for 14 consecutive days. On day 14, animals were sacrificed 1 h after the last administration and brain tissues were taken. In another set of experiments, after 30 days of voluntary alcohol-drinking procedure, mice were randomly allocated to five experimental groups (n=5/7 per group): WT naïve, WT/vehicle, WT/ANA-12, D3R−/−/vehicle, and D3R−/−/ANA-12. Animals were i.p. injected once a day, for 4 consecutive days with the selective Trkb antagonist ANA-12 at 0.5 mg/kg (Cazorla et al, 2011; Vassoler et al, 2013). On day 4, animals were sacrificed 1 h after the last administration and brain tissues were taken. |
ADME/Pharmacokinetics |
Systemic administration of ANA-12 inhibits TrkB in the brain.[1]
The purpose of this study was to develop a small molecule that can inhibit TrkB in the adult mammalian brain after systemic administration. We first tested whether ANA-12 was stable and not degraded into breakdown products in mouse serum before reaching the brain. To this end, ANA-12 was incubated in mouse serum for 15, 30, 45, and 60 minutes at 37°C. Liquid chromatography–mass spectrometry (LC-MS) analysis of the mixture did not reveal the presence of breakdown products and no degradation of ANA-12 over time was observed (Figure 6A). We then determined whether ANA-12 crosses the blood-brain barrier and reaches the brain after systemic administration. For this purpose, ANA-12 (0.5 mg/kg) was injected i.p. into adult mice. Animals were sacrificed 0.5, 1, 2, 4, or 6 hours later and brains were processed for quantification of ANA-12 by LC-MS. Figure 6B shows that active concentrations of ANA-12 could be detected in the brain as early as 30 minutes (~400 nM) and up to 6 hours after the i.p. injection (~10 nM). We then determined whether ANA-12 inhibits TrkB in the adult brain and determined the magnitude of TrkB inhibition in the brain 2 and 4 hours after the injection of 0.5 mg/kg of ANA-12. This time course was chosen based on what we previously observed with cyclotraxin-B, for which a minimum of 3 hours was required for TrkB deactivation in the brain. KIRA-ELISA quantification of phospho-TrkB revealed that 0.5 mg/kg of ANA-12 partially inhibited the total endogenous TrkB activity in the whole brain (8% at 2 hours, 25% at 4 hours; Figure 6C). Although TrkB is widely distributed in the brain, it is possible that ANA-12 does not inhibit the receptor uniformly in different brain areas. This could lead to an apparent partial inhibition of TrkB in the whole brain with some structures being fully inhibited while others are not or are very little affected. We therefore determined the amplitude of inhibition between different brain areas. Again, 0.5 mg/kg of ANA-12 was injected into adult mice, and different brain structures (striatum, cortex, and hippocampus) were collected after 2 or 4 hours (Figure 6D). KIRA-ELISA analysis showed that TrkB inhibition was more effective in the striatum than in hippocampus and cortex 2 hours after injection. At 4 hours, inhibition was comparable among all the structures that were analyzed (25%–30%), yet TrkB in the striatum appeared to be slightly more inhibited than in the hippocampus and cortex. Together, these observations suggest that very low doses of ANA-12 are sufficient to partially inhibit TrkB activity homogenously throughout the brain after 4 hours. |
Toxicity/Toxicokinetics |
ANA-12 does not affect neuron survival. [1]
Since inhibition of the BDNF/TrkB signaling can induce neuronal death in the central nervous system, we verified whether ANA-12 can be chronically administrated without toxic effects for the brain. For that purpose, adult mice received a daily injection of different doses of ANA-12 (0.5, 1.0, and 2.0 mg/kg) or saline solution for a week. After the last injection, mice were sacrificed and brains were processed for apoptosis detection using fluorescent TUNEL staining (Figure 9). Little or no TUNEL-positive cells could be detected in the brains of mice injected with saline or 0.5 or 1.0 mg/kg of ANA-12. It is noteworthy that examination of the whole brain revealed that the apoptotic cells were present only in the dentate gyrus of the hippocampus. However, a small increase in the number of TUNEL-positive cells was observed in the dentate gyrus of mice that received 2.0 mg/kg of ANA-12. No staining was detected in any other areas of investigation (cortex, striatum, CA1-3 areas of the hippocampus, hypothalamus, thalamus, substantia nigra, ventral tegmental area, pallidum, and raphe nucleus). |
References | |
Additional Infomation |
ANA-12 is a secondary carboxamide that is anthranilic acid in which the carboxy group has undergone condensation with the primary amino group of alpha-amino-epsilon-caprolactam, while the aryl-amino group has undergone condensation with the carboxy group of 1-benzothiophene-2-carboxylic acid. It is a selective, non-competitive antagonist of tropomyosin receptor kinase B (TrkB, also known as tyrosine receptor kinase B). It has a role as a tropomyosin-related kinase B receptor antagonist, an antidepressant and an anxiolytic drug. It is a secondary carboxamide, a member of caprolactams and a member of 1-benzothiophenes. It is functionally related to a 2-aminohexano-6-lactam and an anthranilic acid.
Finally, other compounds sharing the 3-(acylamino)-ε-caprolactam scaffold of N-T19 and ANA-12 have been described as procognitive agents in mice, although the precise mechanism of action is not described. Given the central role of the BDNF/TrkB coupling in cognition and memory, it would be interesting to evaluate the effect of these compounds on TrkB receptors and to compare them with ANA-12 and N-T19. To the best of our knowledge, ANA-12 is the first nonpeptide antagonist of TrkB receptor that elicits strong and specific effects in vivo. This proteolytically stable small molecule constitutes a valuable pharmacological tool to enable us to better investigate the role of BDNF/TrkB signaling in pathophysiological situations and will serve as a lead compound for the design of potent orally bioavailable TrkB modulators. [1] In conclusion, our study shows that LPS-induced inflammation caused depression-like behavior, as well as alterations in BDNF protein and spine density within the hippocampus, PFC, and NAc. Furthermore, antidepressant effects were shown on LPS-induced depressive behavior by normalizing altered dendritic spines in the hippocampus and PFC with TrkB agonist 7,8-DHF and in the NAc with antagonist ANA-12. Therefore, abnormal BDNF-TrkB signaling in the hippocampus, PFC, and NAc may play a role in inflammation-induced depression. Finally, in MDD, TrkB agonists and TrkB antagonists could act as potential therapeutic drugs for patients with lower BDNF levels in the hippocampus and PFC, and those with higher levels of BDNF in the NAc. [2] Mesolimbic dopamine (DA) controls drug- and alcohol-seeking behavior, but the role of specific DA receptor subtypes is unclear. We tested the hypothesis that D3R gene deletion or the D3R pharmacological blockade inhibits ethanol preference in mice. D3R-deficient mice (D3R(-/-)) and their wild-type (WT) littermates, treated or not with the D3R antagonists SB277011A and U99194A, were tested in a long-term free choice ethanol-drinking (two-bottle choice) and in a binge-like ethanol-drinking paradigm (drinking in the dark, DID). The selectivity of the D3R antagonists was further assessed by molecular modeling. Ethanol intake was negligible in D3R(-/-) and robust in WT both in the two-bottle choice and DID paradigms. Treatment with D3R antagonists inhibited ethanol intake in WT but was ineffective in D3R(-/-) mice. Ethanol intake increased the expression of RACK1 and brain-derived neurotrophic factor (BDNF) in both WT and D3R(-/-); in WT there was also a robust overexpression of D3R. Thus, increased expression of D3R associated with activation of RACK1/BDNF seems to operate as a reinforcing mechanism in voluntary ethanol intake. Indeed, blockade of the BDNF pathway by the TrkB selective antagonist ANA-12 reversed chronic stable ethanol intake and strongly decreased the striatal expression of D3R. Finally, we evaluated buspirone, an approved drug for anxiety disorders endowed with D3R antagonist activity (confirmed by molecular modeling analysis), that resulted effective in inhibiting ethanol intake. Thus, DA signaling via D3R is essential for ethanol-related reward and consumption and may represent a novel therapeutic target for weaning. [4] We delineated a heritable phenotype resulting from the self-administration of cocaine in rats. We observed delayed acquisition and reduced maintenance of cocaine self-administration in male, but not female, offspring of sires that self-administered cocaine. Brain-derived neurotrophic factor (Bdnf) mRNA and BDNF protein were increased in the medial prefrontal cortex (mPFC), and there was an increased association of acetylated histone H3 with Bdnf promoters in only the male offspring of cocaine-experienced sires. Administration of a BDNF receptor antagonist (the TrkB receptor antagonist ANA-12) reversed the diminished cocaine self-administration in male cocaine-sired rats. In addition, the association of acetylated histone H3 with Bdnf promoters was increased in the sperm of sires that self-administered cocaine. Collectively, these findings indicate that voluntary paternal ingestion of cocaine results in epigenetic reprogramming of the germline, having profound effects on mPFC gene expression and resistance to cocaine reinforcement in male offspring. [5] |
Molecular Formula |
C22H21N3O3S
|
|
---|---|---|
Molecular Weight |
407.49
|
|
Exact Mass |
407.13
|
|
Elemental Analysis |
C, 64.85; H, 5.19; N, 10.31; O, 11.78; S, 7.87
|
|
CAS # |
219766-25-3
|
|
Related CAS # |
|
|
PubChem CID |
2799722
|
|
Appearance |
White to off-white solid powder
|
|
LogP |
4.786
|
|
Hydrogen Bond Donor Count |
3
|
|
Hydrogen Bond Acceptor Count |
4
|
|
Rotatable Bond Count |
4
|
|
Heavy Atom Count |
29
|
|
Complexity |
628
|
|
Defined Atom Stereocenter Count |
0
|
|
SMILES |
S1C2=C([H])C([H])=C([H])C([H])=C2C([H])=C1C(N([H])C1=C([H])C([H])=C([H])C([H])=C1C(N([H])C1([H])C(N([H])C([H])([H])C([H])([H])C([H])([H])C1([H])[H])=O)=O)=O
|
|
InChi Key |
TUSCYCAIGRVBMD-UHFFFAOYSA-N
|
|
InChi Code |
InChI=1S/C22H21N3O3S/c26-20(25-17-10-5-6-12-23-21(17)27)15-8-2-3-9-16(15)24-22(28)19-13-14-7-1-4-11-18(14)29-19/h1-4,7-9,11,13,17H,5-6,10,12H2,(H,23,27)(H,24,28)(H,25,26)
|
|
Chemical Name |
N-[2-[(2-oxoazepan-3-yl)carbamoyl]phenyl]-1-benzothiophene-2-carboxamide
|
|
Synonyms |
|
|
HS Tariff Code |
2934.99.9001
|
|
Storage |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month |
|
Shipping Condition |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
Solubility (In Vitro) |
|
|||
---|---|---|---|---|
Solubility (In Vivo) |
Solubility in Formulation 1: 1.43 mg/mL (3.51 mM) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (add these co-solvents sequentially from left to right, and one by one), suspension solution; with sonication.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 14.3 mg/mL clear DMSO stock solution to 400 μL PEG300 and mix evenly; then add 50 μL Tween-80 to the above solution and mix evenly; then add 450 μL normal saline to adjust the volume to 1 mL. Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution. Solubility in Formulation 2: ≥ 1.43 mg/mL (3.51 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution. For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 14.3 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly. View More
Solubility in Formulation 3: 1 mg/mL (2.45 mM) in 5% DMSO + 40% PEG300 + 5% Tween80 + 50% Saline (add these co-solvents sequentially from left to right, and one by one), suspension solution; with ultrasonication. Solubility in Formulation 4: ≥ 0.45 mg/mL (1.10 mM) (saturation unknown) in 5% DMSO + 95% (20% SBE-β-CD in Saline) (add these co-solvents sequentially from left to right, and one by one), clear solution. Preparation of 20% SBE-β-CD in Saline (4°C,1 week): Dissolve 2 g SBE-β-CD in 10 mL saline to obtain a clear solution. Solubility in Formulation 5: ≥ 0.45 mg/mL (1.10 mM) (saturation unknown) in 5% DMSO + 95% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution. Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution. Solubility in Formulation 6: 2% DMSO+30% PEG 300+2% Tween 80+ddH2O: 2mg/mL . |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 2.4540 mL | 12.2702 mL | 24.5405 mL | |
5 mM | 0.4908 mL | 2.4540 mL | 4.9081 mL | |
10 mM | 0.2454 mL | 1.2270 mL | 2.4540 mL |
*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.
Calculation results
Working concentration: mg/mL;
Method for preparing DMSO stock solution: mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.
Method for preparing in vivo formulation::Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.
(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
(2) Be sure to add the solvent(s) in order.
Effects of 7,8-DHF and ANA-12 on LPS-induced changes in phosphorylation of TrkB in the mouse brain. Int J Neuropsychopharmacol. 2015 Feb; 18(4): pyu077. td> |
Role of TrkB and mTORC1 in the antidepressant action of 7,8-DHF and ANA-12 on LPS-induced depression-like behavior. Int J Neuropsychopharmacol. 2014 Oct 31;18(4). td> |
Role of TrkB and mTORC1 in the antidepressant action of 7,8-DHF and ANA-12 on LPS-induced depression-like behavior. Int J Neuropsychopharmacol. 2014 Oct 31;18(4). td> |