Content area
The aim of this study is to assess the sedative impact of palmatine chloride (PME) in thiopental sodium-induced sleeping chicks and its underlying molecular mechanism by using in vivo and in silico approaches. Chicks received PME per orally (p.o.) at doses of 1.25, 2.5, and 5 mg/kg per body weight (b.w.), while diazepam (DZP) (2 mg/kg) served as a positive control and vehicle as a negative control. For the purpose of evaluating the experimental compounds synergistic or antagonistic effects, a combination of PME and DZP was administered to the chicks. After thirty minutes, thiopental sodium (40 mg/kg, intraperitoneal (i.p.)) was administered to induce sleep, and latency to sleep onset and sleep duration were measured. In vivo results showed that PME reduced sleep latency and prolonged sleep duration in a dose-dependent manner, with the combination therapy producing a significant enhancement of these effects. In silico docking revealed PME binding to gamma-aminobutyric acid A (GABAA) receptor α1 and β2 subunits (–7.2 kcal/mol) with shared amino acids. Pharmacokinetic and toxicity analyses suggested favorable drug-like properties. These results indicate PME’s sedative potential, alone or with DZP, likely via GABAergic modulation, warranting further functional validation.
Introduction
Insomnia, characterized by persistent difficulty in initiating or maintaining sleep, is one of the most prevalent neurological disorders worldwide, impairing cognitive performance, emotional stability, and overall quality of life1,2. The neurobiology of sleep regulation is strongly governed by the GABA system, the principal inhibitory neurotransmitter pathway in the central nervous system. Within this network, the GABAA receptor plays a pivotal role, serving as the major molecular target of sedative-hypnotic drugs3,4.
Currently available therapies, including benzodiazepines, barbiturates, and related agents, are effective but remain clinically constrained by rapid tolerance, dependence, and severe adverse reactions such as psychomotor impairment, cardiovascular complications, and cognitive deficits5,6. These limitations underscore the necessity of identifying safer, mechanism-based alternatives.
Natural products continue to represent a powerful source of therapeutic innovation, offering structurally diverse molecules with central nervous system activity. Phytochemicals such as flavonoids, terpenes, and alkaloids have demonstrated the capacity to modulate GABAergic transmission, thereby exerting anxiolytic and sedative effects7,8. Among them, palmatine chloride (PME), a protoberberine isoquinoline alkaloid, has attracted growing attention. PME is a quaternary ammonium salt form of palmatine that exhibits superior solubility and stability, making it more suitable for pharmacological evaluation than the free base9,10. In addition to its wide pharmacological spectrum, including neuroprotective, anti-inflammatory, and antidepressant effects11,12. PME has been reported to influence neurotransmission by enhancing serotonergic and GABAergic pathways13. These findings suggest that PME may serve as a promising candidate for managing sleep disturbances.
Based on this evidence, we hypothesized that PME could exert sedative effects through modulation of the GABAA receptor, either alone or in combination with DZP, thereby potentiating GABAergic signaling. To test this, the present study investigated the effects of PME on thiopental sodium-induced sleeping chicks and assessed its potential synergism with DZP. Furthermore, to elucidate the molecular basis of its action, in silico docking was performed against GABAA receptor subunits, and its pharmacokinetic and toxicological profiles were evaluated to establish PME as a viable drug-like candidate.
Methods
In vivo study
Chemicals and reagents
The experimental compound PME (1.25, 2.5, and 5 mg/kg, p.o.) (CAS Number: 10605-02-4, Purity: 98% (HPLC)) was brought from Sigma-Aldrich (USA). Loba Chemie Pvt. Ltd. (India) supplied Tween-80 (0.5%). DZP (2 mg/kg, p.o.) and thiopental sodium (40 mg/kg, i.p.) were purchased from Square Pharmaceuticals Ltd. (Bangladesh).
Dose selection and Preparation
The test doses of PME (1.25, 2.5, and 5 mg/kg) were selected from the previous published literature review14. We prepared a mother solution at the concentration of 5 mg/kg by adding a sufficient amount of distilled water and Tween-80 (0.5%). At last, we dilute the mother solution at the concentrations of 2.5 and 1.25 mg/kg. On the other hand, DZP, supplied as an injectable solution (5 mg/ml), was diluted with distilled water to achieve a final concentration of 2 mg/kg for oral administration.
Experimental animals
Four-day-old (Grade-A) young broiler chickens (Gallus domesticus) of either sex, with a weight range of 42–48 g, were purchased from Aftab Hatchery Ltd., which is located in Bhagalpur, Bajitpur, Kishoreganj, Bangladesh. Chicks were chosen as a validated alternative neuropharmacological model because thiopental sodium reliably induces sleep in this species, and chicks are highly sensitive to GABAergic modulation, making them suitable for early-phase sedative screening15. Before our investigation, all of the chicks were kept in the pharmacology lab at Gopalganj Science and Technology University (GSTU) at a consistent temperature (26 ± 3 °C), with 65% relative humidity and under controlled lighting (12-hour dark/light cycle). During this time, they had unlimited access to standard feeds and water ad libitum. All animals were randomly assigned into six groups (n = 5 each). Randomization was performed using a random number generator, and the investigator recording sleep parameters was blinded to treatment groups. Before conducting the sedative test, chicks were fasted for about 12 h. The current experimental test was performed between 8:00 A.M. and 3:00 P.M. All experimental procedures were conducted in accordance with relevant institutional guidelines and regulations. All methods are reported in accordance with the ARRIVE guidelines (https://arriveguidelines.org). The experimental protocols used in this study were reviewed and approved by the Institutional Animal Ethics Committee (IAEC) of [Department of Pharmacy, GSTU], under approval number [#gstu-20PHR009-02].
Table 1. In chicks, several treatments and doses were examined.
Treatment Groups | Dose (mg/kg) | Route of Administration | Targeted Receptor |
|---|---|---|---|
NC (Vehicle): Distilled water containing 0.9% NaCl and 0.5% tween-80 | 10 ml/kg | p.o. | – |
DZP-2 | 2 | GABAA receptor | |
PME-1.25 | 1.25 | Under Investigation | |
PME-2.50 | 2.50 | ||
PME-5 | 5 | ||
PME-5 + DZP-2 | 5 + 2 |
NC: Negative control; DZP: Diazepam; PME: Palmatine Chloride; GABAA: Gama-aminobutyric acid type A receptor; p.o.: per orally
Thiopental sodium-induced sleeping test
The test was run following the procedure outlined by Dehar et al. (2012)16. PME: 1.25, 2.5, and 5 mg/kg; DZP: 2 mg/kg; and vehicle: 10 ml/kg were administered p.o. Then after 30 min of pretreatment, each chick received an i.p. injection of thiopental sodium at the dose of 40 mg/kg to induce sleep before being placed in the observation chamber. After administration of thiopental sodium, we observed two points: first, latency period (LP) and second, duration of sleeping time up to four hours (Table 1). The percentage of sleep incidence, increase in sleep time, and reduction in LP can be determined using the following equations.
% Incidence of sleep= .
% Reduction in LP = .
% Increase in the amount of time spent sleeping = .
Where, A = mean of LP in the NC group, B = mean of LP in the test or standard groups, X = mean of duration in the test or standard groups and Y = Mean of duration in the NC group.
Statistical analysis
Results are presented as mean ± standard deviation (SD) (n = 5). One-way ANOVA was used, followed by t-Student Tukey’s post-hoc test for pairwise comparisons (GraphPad Prism v9.5, San Diego, USA). A p-value < 0.05 was considered statistically significant.
In Silico study
Ligand Preparation
The 3D structures of DZP (Compound ID: 3016), and PME (Compound ID: 19009) were downloaded using the PubChem chemical database (https://pubchem.ncbi.nlm.nih.gov/, obtained on 4 March 2025) and stored in structure-data file (SDF) format. PME was used as palmatine chloride, but the chloride ion was removed for docking. Structures were energy-minimized using Chem3D 21.0.0. The 2D structure of chemical agents is shown in Fig. 1.
Fig. 1 [Images not available. See PDF.]
The two-dimensional chemical structures of diazepam and palmatine.
Protein selection and Preparation
The GABAA receptor was selected as the molecular target, focusing on α1 and β2 subunits (PDB ID: 6X3X), which are critically involved in sedative and hypnotic drug actions 3,17,18. The protein structure was prepared by removing heteroatoms, water molecules, and redundant residues using PyMol (v2.4.1). Further energy minimization was conducted using Swiss-PDB Viewer (v4.1.0) with the GROMOS96 43B1 force field 15,19.
Molecular Docking and visualization
Docking was performed with PyRx (v0.8) using AutoDock Vina. A grid box of 20.01 × 18.54 × 17.29 Å was set along the x-, y-, and z-axes, with 2000 calculation steps. Ligands were converted to PDBQT format, and binding affinities were expressed in kcal/mol. Docking validation was ensured by redocking DZP into the GABAA receptor binding pocket, which reproduced interactions consistent with previous reports. Visualization of hydrogen bonds and hydrophobic interactions was performed using Discovery Studio Visualizer (v21.1.0).
Prediction of pharmacokinetics and Drug-Likeness
Drug-likeness, absorption, distribution, metabolism, excretion (ADME), and toxicity profiles of PME and DZP were predicted using SwissADME (http://www.swissadme.ch/), pkCSM (https://biosig.lab.uq.edu.au/pkcsm/), and ProTox 3.0 (https://tox.charite.de/protox3/) online servers. Parameters included Lipinski’s rule of five, gastrointestinal absorption, blood–brain barrier penetration, CYP450 enzyme interactions, total clearance, and predicted LD50 values. As these analyses are computationally based, they provide preliminary predictions only, which require experimental validation.
Results
In vivo result
In vivo research, we observed thiopental sodium reliably produced 100% sedation in all tested animal groups. The NC group was characterized by delayed onset of sleep (3.49 ± 0.06 min) and acted as a baseline for comparison with other groups. However, the standard drug DZP-2 decreases LP (1.24 ± 0.14 min) compared to the NC group. On the other hand, PME produced dose-dependent effects on the thiopental sodium-induced animals and significantly (p < 0.05) reduced the LP when compared to the NC group. PME-1.25, PME-2.5, and PME-5 animal groups demonstrated LP at 3.28 ± 0.18, 2.24 ± 0.71, and 2.01 ± 0.44 min. Eventually, with PME-5 when co-treated with DZP-2 (PME-5 + DZP-2), we observed a synergistic effect and produced the lowest LP (1.02 ± 0.28 min) among all tested groups.
On the other hand, in the case of duration of sleep, DZP-2 increased duration of sleep (186.52 ± 7.18 min) compared to the NC (143.93 ± 7.75 min) group. Additionally, PME significantly (p < 0.05) and dose-dependently lengthens animals sleep duration. PME-1.25, PME-2.5, and PME-5 demonstrated durations of sleep at 156.30, 162.61, and 183.02 min, as mean values. When administering combination therapy (PME-5 + DZP-2), sleeping time significantly (p < 0.05) increased compared to the other treatment groups. However, details of in vivo findings are demonstrated in Table 2; Fig. 2.
Table 2. Latency period and duration of sleeping time observed in the palmatine chloride, diazepam, and combination treatment groups.
Treatments | Latency period (min) | Duration of sleep (min) | Sleep incidence (%) |
|---|---|---|---|
NC (Vehicle) | 3.49 ± 0.06 | 143.93 ± 7.75 | 100 |
DZP-2 | 1.24 ± 0.14*bcd | 186.52 ± 7.18*bcd | 100 |
PME-1.25 | 3.28 ± 0.18* | 156.30 ± 6.74* | 100 |
PME-2.5 | 2.24 ± 0.71*b | 162.61 ± 8.16*b | 100 |
PME-5 | 2.01 ± 0.44*bc | 183.02 ± 4.67*bc | 100 |
PME-5 + DZP-2 | 1.02 ± 0.28*abcd | 194.50 ± 4.25*abcd | 100 |
Values are mean ± SD (standard deviation) (n = 5); One-way ANOVA followed by t-Student Tukey’s as post-hoc test; p < 0.05 when compared to the *NC (vehicle), aDZP-2, bPME-1.25, cPME-2.5, dPME-5, ePME-5 + DZP-2; NC: Negative control (distilled water containing 0.9% NaCl and 0.5% tween 80); DZP: Diazepam; PME: Palmatine Chloride
Fig. 2 [Images not available. See PDF.]
Latency and duration of sleep in different treatment groups of animals. [Values are mean ± SD (standard deviation) (n = 5); one-way ANOVA followed by t-Student Tukey’s as post-hoc test; p < 0.05 when compared to the *NC (vehicle), aDZP-2, bPME-1.25, cPME-2.5, dPME-5, ePME-5 + DZP-2; NC: Negative control (distilled water containing 0.9% NaCl and 0.5% tween 80); DZP: Diazepam; PME: Palmatine Chloride]
In contrast, compared to the NC group, PME-1.25, PME-2.5, and PME-5 dose-dependently increased the percentage of sleeping time at 7.91, 11.48, and 21.35% (Table 3). However, when chicks received combination therapy (PME-5 + DZP-2), they exhibited the highest sleep duration among all groups at 26%. On the other hand, PME reduced the percentage of LP in a dose-dependent manner, which was 6.01, 35.81, and 42.40% for PME-1.25, PME-2.5, and PME-5, respectively. At last, the PME-5 + DZP-2 groups displayed the highest LP decrease at 70.77%.
Table 3. Percentage increase in duration of sleep and decrease in latency period in thiopental sodium-induced sleeping chicks of the palmatine chloride, diazepam, and combination treatment groups.
Treatments | % Reduction in latency Period | % Increase in the amount of time spent sleeping |
|---|---|---|
NC (Vehicle) | – | – |
DZP-2 | 64.46 | 22.83 |
PME-1.25 | 6.01 | 7.91 |
PME-2.5 | 35.81 | 11.48 |
PME-5 | 42.40 | 21.35 |
PME-5 + DZP-2 | 70.77 | 26 |
NC: Negative control (distilled water containing 0.9% NaCl and 0.5% tween 80); DZP: Diazepam; PME: Palmatine chloride
In Silico findings
Interactions of PME and DZP with GABAA (α1 and β2 subunits) receptor
For the GABAA receptor, DZP showed the highest binding affinity (− 8.3 kcal/mol) by the synthesis of a single hydrogen bond with LEU A: 285 amino acid residues. Additionally, DZP also formed several hydrophobic bonds, including PHE A: 289, PRO B: 233, MET B: 236, MET A: 286, and LEU B: 232 specific amino acid residues. Despite not forming hydrogen bonds, PME displayed reliable binding affinity (–7.2 kcal/mol) with the GABAA receptor. This interaction involved several hydrophobic bonds: PHE A: 289, PRO B: 233, MET B: 236, MET A: 286, LEU B: 232, LEU A: 285, MET A: 261, and VAL A: 290 amino acid residues. Diazepam and palmatine 3D and 2D non-bond interactions with the GABAA receptor (α1 and β2 types of subunits) were shown in Table 4; Fig. 3, together with information on the number of hydrogen bonds.
Table 4. Results of diazepam and palmatine molecular Docking against the GABAA (α1 and β2 types of subunits) receptor.
Ligands | Receptor (PDB ID) | BA (kcal/mol) | No of HB | Amino acid (AA) residues | |
|---|---|---|---|---|---|
HBs | Others | ||||
DZP | GABAA (α1 and β2 types of subunits) receptor (6X3X) | –8.3 | 1 | LEU A: 285 | PHE A: 289, MET A: 286, PRO B: 233, MET B: 236, LEU B: 232 |
PME | –7.2 | 0 | – | PHE A: 289, MET A: 286, LEU A: 285, MET A: 261, VAL A: 290, PRO B: 233, MET B: 236, LEU B: 232 | |
AA: Amino acid; BA: Binding affinity; DZP: Diazepam; GABAA: Gamma-aminobutyric acid type A receptor; HB: Hydrogen Bond; PDB ID: Protein Data Bank identification code; PME: Palmatine;
Fig. 3 [Images not available. See PDF.]
Diazepam and palmatine 3D and 2D non-bond connections with the GABAA receptor (α1 and β2 types of subunits).
Pharmacokinetics, drug-likeness properties and toxicological profiles
The pharmacokinetics study is crucial in drug development and discovery due to its evaluation of ADME and toxicity parameters. PME (C21H22NO4) and DZP (C16H13ClN2O) have molecular weights (MW) 352.40 g/mol and 284.74 g/mol. However, both compounds (PME and DZP) follow the five Lipinski rules without any violation: H-acceptor < 10, H-donor < 5, logP < 5, and MW < 500, as well as showing high gastrointestinal absorption (97.084 and 97.42%) and good solubility. Molar refractivity (MR) of the PME and DZP are 101.80 and 87.95, respectively, which was within limit (MR ≤ 140) and displayed bioavailability score of 0.55. Compared to DZP (0.294 log ml/min/kg), PME had a higher overall clearance (1.246 log ml/min/kg). In the metabolism part, PME and DZP do not have any effect on CYP2D6 and CYP3A4 substrates or inhibitors.
In this in silico toxicity investigation, PME demonstrates LD50 values of 200 mg/kg with toxicity class 3, whereas DZP exhibited LD50 values of 48 mg/kg with toxicity class 2. However, PME showed no toxic effect in terms of hepatotoxicity, nephrotoxicity, cardiotoxicity, clinical toxicity, and nutritional toxicity, while DZP showed adverse effects in the case of clinical toxicity, as shown in Table 5.
Table 5. Pharmacokinetics, drug likeness properties and toxicological profiles of palmatine and diazepam.
Properties | Factors | PME | DZP |
|---|---|---|---|
Physicochemical Properties | Formula | C21H22NO4 | C16H13ClN2O |
Molecular weight (g/mol) | 352.40 | 284.74 | |
Number of heavy atoms | 26 | 20 | |
Number of aromatic heavy atoms | 16 | 12 | |
Number of H-bond donors | 0 | 0 | |
Number of H-bond acceptors | 4 | 2 | |
Molar Refractivity | 101.80 | 87.95 | |
TPSA (Ų) | 40.80 | 32.67 | |
Lipophilicity | Log Po/w (XLOGP3) | 3.75 | 2.99 |
Drug-likeness | Lipinski | Yes; 0 violations | Yes; 0 violation |
Bioavailability Score | 0.55 | 0.55 | |
Water Solubility | Log S (ESOL) | −4.58 | −3.87 |
Class | Moderately soluble | Soluble | |
Absorption | Caco2 permeability (log Papp in 10 –6 cm/s) | 0.863 | 1.554 |
Intestinal absorption (human) Numeric (%) Absorbed | 97.084 | 97.42 | |
Skin Permeability | −2.554 | −2.205 | |
P-glycoprotein Substrate | Yes | Yes | |
P-glycoprotein I Inhibitor | Yes | No | |
P-glycoprotein II Inhibitor | Yes | No | |
Distribution | BBB permeability (log BB) | −0.112 | 0.331 |
VDss (human) (log L/kg) | 0.641 | 0.365 | |
CNS permeability (log PS) | −1.535 | −1.397 | |
Metabolism | CYP2D6 substrate | No | No |
CYP3A4 substrate | Yes | Yes | |
CYP1A2 inhibitor | Yes | Yes | |
CYP2C19 inhibitor | No | Yes | |
CYP2C9 inhibitor | No | Yes | |
CYP2D6 inhibitor | Yes | No | |
CYP3A4 inhibitor | No | No | |
Excretion | Total Clearance (log ml/min/kg) | 1.246 | 0.294 |
Renal OCT2 substrate | No | Yes | |
Toxicity | Toxicity class | 3 | 2 |
Predicted LD50 (mg/kg) | 200 | 48 | |
Hepatotoxicity | Inactive | Inactive | |
Nephrotoxicity | Inactive | Inactive | |
Cardiotoxicity | Inactive | Inactive | |
Clinical toxicity | Inactive | Active | |
Nutritional toxicity | Inactive | Inactive |
BBB: Blood-Brain Barrier; DZP: Diazepam; GI: Gastrointestinal; PME: Palmatine; TPSA: Topological Polar Surface Area; VDss: Volume of Distribution at Steady State
Discussion
The GABA transmitter is crucial to the advancement of contemporary sedation and anesthetic techniques20. Because GABA is the brain’s fundamental inhibitory neurotransmitter. It works by preventing nerve transmission, which lowers neuronal excitability. GABA function ameliorates epilepsy, Parkinson’s disease, insomnia, Alzheimer’s disease, and stable mood 21 22. Hypnotic medications, particularly benzodiazepines like DZP and barbiturates, are well-known for their sedative effects, which are mediated via amplifying GABAA receptor activity23. However, regular use of benzodiazepines produces patient tolerance (higher dosages are needed to achieve the same effect), leading to increasing mortality6. On the other hand, with long-term use of barbiturates, the body develops a dependency on barbiturates and produces withdrawal symptoms24. As a result, currently available hypnotic medication drugs have limitations in terms of safety. So, there is a need for the discovery of safer alternatives that can effectively address insomnia and other sleep disorders without severe adverse effects.
Thiopental sodium reversibly depresses excitable tissue in the CNS, which was used to provide the sedative effect and induce sleeping time in model25. Indeed, thiopental sodium, a barbiturate anesthetic, induces hypnosis by enhancing GABA inhibitory effects by modifying the GABAA receptor, leading to increased neuronal activity inhibition26. When thiopental sodium binds to the GABA receptor (part of the Cl– channel), chloride channels stay open longer, which causes neurons to become hyperpolarized and decrease neuronal activity17. Any sedative and anxiolytic medication, such as DZP, which lengthened the duration of sleep and reduced the beginning of thiopental sodium-induced sleep, may interact with GABAA receptor27.
The findings demonstrate that PME exhibits a dose-dependent sedative effect, which aligns with previous reports that palmatine enhances serotonergic and GABAergic signaling to modulate sleep and relaxation13. The marked reduction in sleep latency and prolongation of sleep duration observed at higher doses suggest that PME acts through potentiation of the GABAA receptor pathway, a mechanism shared by classical hypnotics such as benzodiazepines3,5. However, when a combination of multiple effects is higher than the sum of their individual impacts, this phenomenon is referred to as synergism or synergistic effects28. Notably, the synergistic interaction between PME and DZP highlights the possibility that PME may enhance benzodiazepine efficacy by facilitating chloride ion influx through GABAA receptor channels, thereby amplifying inhibitory neurotransmission. This synergism is pharmacologically important because it suggests that PME could be used to lower the effective dose of DZP, potentially reducing the adverse effects and dependence risks commonly associated with long-term benzodiazepine use6. Thus, PME emerges as an independent sedative agent and also as a potential adjunct therapy in insomnia management.
Computer-assisted drug discovery and design (CADD), it is now feasible to test more chemicals in a shorter time with lower cost. These technologies reduce reliance on animal models and help with the creation of new compounds and the investigation and analysis of the characteristics of natural items29. However, these have several limitations: molecular flexibility, validity and reliability, over-reliance on computational predictions and scoring functions, and algorithms30. Despite their drawbacks, computational approaches are anticipated to be essential to drug discovery and may accelerate the development of life-saving treatments.
Molecular docking is a computer simulation technology used to calculate the binding affinity of drug-receptor complexes in structural molecular biology and drug design31. Binding affinity is a crucial metric for evaluating the strength of a small molecule’s binding to its target32. Higher binding affinity indicates a strong drug-receptor interaction, which is crucial for successful drug development and effective cell signaling33. In this context, previous studies have shown that compounds capable of interacting with key residues in the GABAA receptor can enhance receptor activity and produce sleep-promoting effects. The current observations suggest that PME may occupy the classical benzodiazepine binding site and interact with amino acid residues such as LEU A: 285, PHE A: 289, MET A: 286, PRO B: 233, MET B: 236, and LEU B: 232, which are known to be critical for ligand binding and receptor modulation. These interactions indicate that PME could mimic the effects of standard GABAA modulators like DZP, supporting its potential as a sleep-enhancing agent. Nevertheless, while in silico predictions provide a valuable mechanistic hypothesis, further in vitro and in vivo studies are required to validate these interactions and elucidate the precise pharmacological mechanism of PME. The possible mechanism of sedative effects of PME is depicted in Fig. 4.
Fig. 4 [Images not available. See PDF.]
Proposed mechanism of the sedative effect of palmatine chloride (PME). [Following administration, PME binds to the GABAA receptor (α1 and β2 subunits) on postsynaptic neurons. This binding is predicted to facilitate chloride ion (Cl⁻) influx through the receptor channel, causing neuronal hyperpolarization. The resulting reduction in neuronal excitability contributes to increased relaxation, delayed sleep onset, and prolonged sleep duration, as observed in vivo. The schematic integrates our docking predictions with the behavioral outcomes, highlighting a possible GABAergic mechanism of action.]
Drug-likeness important guidelines for early drug discovery include drug-likeness, safety, Lipinski’s rule of five, desirable ADMET properties, and oral absorption34. Both compounds adhere to Lipinski’s rules with excellent bioavailability scores and outstanding solubility, indicating PME would be a promising drug candidate for oral bioavailability. Pharmacokinetics is the study of how the body interacts with administered drugs through ADME35. According to the ADME analysis, PME and DZP manifest high absorption at 97.084% and 97.42%, indicating that the GIT effectively absorbs these substances into the bloodstream. When comparing the LD50 of PME (200 mg/kg) and DZP (48 mg/kg) in a toxicity investigation, PME showed a greater lethal dose, suggesting it is less harmful at similar doses. Though, these LD50 values were derived from computational prediction (ProTox 3.0) and therefore are not linked to a specific route of administration. Our docking results suggest that PME interacts with key residues within the GABAA α1 and β2 subunits that overlap with the DZP-binding pocket, raising the possibility that PME modulates the same receptor site. This mechanistic hypothesis aligns with the observed in vivo sedative effects, where PME reduced sleep latency and prolonged sleep duration. Nevertheless, molecular docking is limited by rigid-receptor assumptions, reliance on scoring functions, and inability to capture dynamic conformational changes. Hence, while the computational evidence is supportive, experimental receptor-binding and electrophysiological studies are required to confirm PME’s precise mechanism of action.
Overall, our study presents several key findings regarding the sedative potential of PME. PME significantly reduced sleep latency and increased sleep duration in experimental animals, indicating strong sedative effects, and further exhibited a synergistic interaction when administered with DZP, although definitive confirmation through isobolographic analysis is needed. While the thiopental sodium-induced sleep test provided preliminary evidence, reliance on this single model limits the conclusiveness of our behavioral findings, and more comprehensive assays, such as the open field test and other locomotor activity paradigms, are warranted to robustly validate PME’s sedative activity. In silico analyses suggested that these effects may result from interactions with specific GABAA receptor amino acid residues, and computational pharmacokinetic and toxicity predictions indicated a favorable safety and efficacy profile compared to standard drugs. Nevertheless, these pharmacokinetic and toxicity assessments were purely predictive, generated using SwissADME, pkCSM, and ProTox 3.0 servers, which provide hypothesis-generating insights rather than confirmatory evidence, emphasizing that experimental validation of in vivo pharmacokinetics, metabolism, long-term toxicity, and overall safety is essential before clinical translation.
Conclusion
In vivo experiments confirmed that PME exerts significant sedative effects in thiopental sodium-induced sleeping chicks, as evidenced by a dose-dependent reduction in sleep latency and an increase in sleep duration. Importantly, the combination of PME with DZP produced a synergistic effect, suggesting that PME could enhance the efficacy of benzodiazepines while potentially reducing their required dose and associated adverse effects. Complementary in silico analyses provided mechanistic support for these findings by showing favorable interactions of PME with GABAA receptor subunits, consistent with modulation of the GABAergic pathway. Additionally, PME displayed satisfactory drug-likeness, pharmacokinetic, and safety profiles, reinforcing its potential as a drug-like candidate. Taken together, the results provide initial evidence that PME exerts sedative effects alone but may also enhance DZP efficacy, suggesting potential clinical utility as an adjunct therapy for insomnia or anxiety disorders. Such an approach could allow dose reduction of benzodiazepines, thereby mitigating risks of dependence and adverse effects.
Acknowledgements
AcknowledgmentsAuthors extend their appreciation to the University Higher Education Fund for funding this research work under the Research Support Program for Central Labs at King Khalid University through the project number CL/PRI/B/7.
Author contributions
Author ContributionsAll authors including Feroz Khan Nun, Mohammad Y. Alshahrani, Md. Sakib Al Hasan, Emon Mia, Na’il Saleh, Mashael A Almansoori, Mehedi Hasan Bappi, and Muhammad Torequl Islam made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis, and interpretation, or in all these areas, that is, revising or critically reviewing the article; giving final approval of the version to be published; agreeing on the journal to which the article has been submitted; and confirming to be accountable for all aspects of the work. All authors have read and agreed to the published version of the manuscript.
Funding
University Higher Education Fund number CL/PRI/B/7.
Data availability
The data that support the findings of this study are available from the corresponding author, Md. Sakib Al Hasan (email: [email protected]), upon reasonable request.
Declarations
Competing interests
The authors declare no competing interests.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
1. Himelfarb, M; Shatkin, JP. Pediatric insomnia. Child. Adolesc. Psychiatr Clin. N Am.; 2021; 30, pp. 117-129. [DOI: https://dx.doi.org/10.1016/j.chc.2020.08.004] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/33223056]
2. Everitt, H. et al. Antidepressants for insomnia in adults. Cochrane Database Syst. Rev.5 (Cd010753). https://doi.org/10.1002/14651858.CD010753.pub2 (2018).
3. Ghit, A; Assal, D; Al-Shami, AS; Hussein, DE. E. GABA(A) receptors: structure, function, pharmacology, and related disorders. J. Genet. Eng. Biotechnol.; 2021; 19, 123. [DOI: https://dx.doi.org/10.1186/s43141-021-00224-0] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34417930][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8380214]
4. Fogaça, MV; Duman, RS. Cortical GABAergic dysfunction in stress and depression: new insights for therapeutic interventions. Front. Cell. Neurosci.; 2019; 13, 87.1:CAS:528:DC%2BC1MXitlKmurjJ [DOI: https://dx.doi.org/10.3389/fncel.2019.00087] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/30914923][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6422907]
5. Peng, L; Morford, KL; Levander, XA. Benzodiazepines and related sedatives. Med. Clin. North. Am.; 2022; 106, pp. 113-129. [DOI: https://dx.doi.org/10.1016/j.mcna.2021.08.012] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34823725]
6. Edinoff, AN et al. Benzodiazepines: uses, dangers, and clinical considerations. Neurol. Int.; 2021; 13, pp. 594-607.1:CAS:528:DC%2BB3sXhtV2ltL3E [DOI: https://dx.doi.org/10.3390/neurolint13040059] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/34842811][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8629021]
7. Calixto, JB. The role of natural products in modern drug discovery. Acad. Bras. Cienc.; 2019; 91 Suppl 3, e20190105. [DOI: https://dx.doi.org/10.1590/0001-3765201920190105]
8. Akram, M; Daniyal, M; Munir, N; Mohiuddin, E; Sultana, S. Medicinal plants combating against insomnia: A green Anti-Insomnia approach. J. Nerv. Ment Dis.; 2019; 207, pp. 927-935. [DOI: https://dx.doi.org/10.1097/nmd.0000000000001052] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31663928]
9. Tarabasz, D; Kukula-Koch, W; Palmatine,. A review of Pharmacological properties and pharmacokinetics. Phytother Res.; 2020; 34, pp. 33-50. [DOI: https://dx.doi.org/10.1002/ptr.6504] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31496018]
10. Dhondale, M. R. et al. Co-Crystallization approach to enhance the stability of Moisture-Sensitive drugs. Pharmaceutics15https://doi.org/10.3390/pharmaceutics15010189 (2023).
11. Pei, H et al. Palmatine ameliorates LPS-induced HT-22 cells and mouse models of depression by regulating apoptosis and oxidative stress. J. Biochem. Mol. Toxicol.; 2023; 37, e23225.1:CAS:528:DC%2BB38XisFaqur7L [DOI: https://dx.doi.org/10.1002/jbt.23225] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/36169195]
12. Jahan Oni, M. I. et al. Botanical Sources, Pharmacokinetics, and Therapeutic Efficacy of Palmatine and Its Derivatives in the Management of Cancer: A Comprehensive Mechanistic Analysis. Journal of Food Biochemistry 8843855 (2024). (2024).
13. Du, J; Yu, Y; Qiu, L; Qiu, M; Zhu, Q. Palmatine plays a role in sedation and hypnosis by increasing 5-hydroxytryptamine. Cell. Mol. Biol. (Noisy-le-grand); 2019; 65, pp. 69-75. [DOI: https://dx.doi.org/10.14715/cmb/2019.65.4.11] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31078155]
14. Long, J; Palmatine, et al. A review of its pharmacology, toxicity and pharmacokinetics. Biochimie; 2019; 162, pp. 176-184.1:CAS:528:DC%2BC1MXpsVyju7g%3D [DOI: https://dx.doi.org/10.1016/j.biochi.2019.04.008] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31051209]
15. Islam, MT et al. Caffeine and sclareol take the edge off the sedative effects of linalool, possibly through the GABAA interaction pathway: molecular insights through in vivo and in Silico studies. Naunyn Schmiedebergs Arch. Pharmacol.; 2025; [DOI: https://dx.doi.org/10.1007/s00210-025-03915-4] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/40839005]
16. Dehar, N; Walia, R; Ratol, S. Potentiation of thiopentone sodium induced hypnosis by berberis aristata in rodents. Asian J. Pharm. Clin. Res.; 2012; 5, pp. 131-133.1:CAS:528:DC%2BC38XivVWhu7o%3D
17. Bhuia, MS et al. Trans-Ferulic acid reduces the sedative activity of diazepam in thiopental sodium-induced sleeping mice: A potential GABAergic transmission. Neurotoxicol Teratol; 2024; 106, 107403.1:CAS:528:DC%2BB2cXisFWns7rP [DOI: https://dx.doi.org/10.1016/j.ntt.2024.107403] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/39547315]
18. Al Hasan, MS et al. Assessment of sedative activity of chrysin: behavioral approach with pharmacokinetics, toxicological profile and molecular Docking. Sleep. Med.; 2025; 126, pp. 88-96. [DOI: https://dx.doi.org/10.1016/j.sleep.2024.12.007] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/39662277]
19. Bhuia, M. S. et al. Anxiolytic-like effects by trans-Ferulic acid possibly occur through GABAergic interaction pathways. Pharmaceuticals (Basel). 16https://doi.org/10.3390/ph16091271 (2023).
20. Philip, AB; Brohan, J; Goudra, B. The role of GABA receptors in anesthesia and sedation: an updated review. CNS Drugs; 2025; 39, pp. 39-54.1:CAS:528:DC%2BB2cXitlKgsL3K [DOI: https://dx.doi.org/10.1007/s40263-024-01128-6] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/39465449]
21. Yang, Z., Zou, Y. & Wang, L. Neurotransmitters in Prevention and Treatment of Alzheimer’s Disease. Int. J. Mol. Sci.24https://doi.org/10.3390/ijms24043841 (2023).
22. Zhu, W et al. GABA and its receptors’ mechanisms in the treatment of insomnia. Heliyon; 2024; 10, e40665.1:CAS:528:DC%2BB2cXis1KqsrrE [DOI: https://dx.doi.org/10.1016/j.heliyon.2024.e40665] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/39654705][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11626785]
23. Hassan, SH et al. Sclareol antagonizes the sedative effect of diazepam in thiopental sodium-induced sleeping animals: in vivo and in Silico studies. Biomed. Pharmacother; 2024; 176, 116939.1:CAS:528:DC%2BB2cXhtlCku7fE [DOI: https://dx.doi.org/10.1016/j.biopha.2024.116939] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/38870629]
24. Aston, R. In Chemical & Biological Aspects of Drug Dependence37–54 (CRC, 2019).
25. Ghosh, S; Kumar, A; Sachan, N; Aggarwal, I; Chandra, P. GABAergic and serotonergic system mediated psychoneuropharmacological activities of essential oil from the leaves of Aegle marmelos: an in vivo and in Silico approach. J. Essent. Oil Bearing Plants; 2020; 23, pp. 1265-1282.1:CAS:528:DC%2BB3MXhsVelur4%3D [DOI: https://dx.doi.org/10.1080/0972060X.2020.1867008]
26. Uddin, MJ et al. Antinociceptive and anxiolytic and sedative effects of methanol extract of anisomeles indica: an experimental assessment in mice and computer aided models. Front. Pharmacol.; 2018; 9, 246.1:CAS:528:DC%2BC1cXisVCgt77K [DOI: https://dx.doi.org/10.3389/fphar.2018.00246] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/29706888][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5907530]
27. Bappi, M. H. et al. Quercetin antagonizes the sedative effects of linalool, possibly through the GABAergic interaction pathway. Molecules28https://doi.org/10.3390/molecules28145616 (2023).
28. Stajkovic, AD; Luthans, F. Behavioral management and task performance in organizations: conceptual background, meta-analysis, and test of alternative models. Pers. Psychol.; 2003; 56, pp. 155-194. [DOI: https://dx.doi.org/10.1111/j.1744-6570.2003.tb00147.x]
29. Rahman, M. M. et al. Emerging promise of computational techniques in Anti-Cancer research: at a glance. Bioeng. (Basel). 9https://doi.org/10.3390/bioengineering9080335 (2022).
30. Ghosh, PR et al. Assessments of protodioscin’s antinociceptive and antidiarrheal properties: in vivo and in Silico investigations on macromolecule binding affinity and modulatory effects. Naunyn Schmiedebergs Arch. Pharmacol.; 2025; [DOI: https://dx.doi.org/10.1007/s00210-025-03860-2] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/40227309]
31. Dias, R; de Azevedo, WF. Jr. Molecular Docking algorithms. Curr. Drug Targets; 2008; 9, pp. 1040-1047.1:CAS:528:DC%2BD1MXhslelu7g%3D [DOI: https://dx.doi.org/10.2174/138945008786949432] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/19128213]
32. Siebenmorgen, T; Zacharias, M. Computational prediction of protein–protein binding affinities. Wiley Interdisciplinary Reviews: Comput. Mol. Sci.; 2020; 10, e1448.1:CAS:528:DC%2BB3cXhsVKntr7N
33. Kastritis, PL; Bonvin, AM. On the binding affinity of macromolecular interactions: daring to ask why proteins interact. J. R Soc. Interface; 2013; 10, 20120835.1:CAS:528:DC%2BC3sXivVGgtb8%3D [DOI: https://dx.doi.org/10.1098/rsif.2012.0835] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/23235262][PubMedCentral: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3565702]
34. Jia, CY; Li, JY; Hao, GF; Yang, G. F. A drug-likeness toolbox facilitates ADMET study in drug discovery. Drug Discov Today; 2020; 25, pp. 248-258.1:CAS:528:DC%2BC1MXitFelsrfL [DOI: https://dx.doi.org/10.1016/j.drudis.2019.10.014] [PubMed: https://www.ncbi.nlm.nih.gov/pubmed/31705979]
35. Sucharitha, P., Reddy, K. R., Satyanarayana, S. & Garg, T. In Computational Approaches for Novel Therapeutic and Diagnostic Designing To Mitigate SARS-CoV-2 Infection335–355 (Elsevier, 2022).
© The Author(s) 2025. This work is published under http://creativecommons.org/licenses/by-nc-nd/4.0/ (the "License"). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.