Introduction

The alkaloid mitraciliatine is a stereoisomeric minor constituent of Mitragyna speciosa with structural similarity to mitragynine. Its low abundance and unique 3R,20R configuration necessitate computational investigation of its ligand-receptor interactions and pharmacokinetic (ADME) properties to supplement limited empirical data. This article uses docking simulations and in-silico prediction platforms (e.g., SwissADME) to evaluate mitraciliatine’s binding potential to opioid and non-opioid targets, and estimate its absorption, distribution, metabolism, and excretion characteristics. These results link to previous sub-articles on structure & chemistry and analytical detection.

Methods

Molecular Docking

Ligand structure for mitraciliatine (PubChem CID 11741588) was retrieved, prepared (protonation at physiological pH), and docked against orthosteric binding domains of the human μ-opioid receptor (MOR, PDB–6DDE), α₁-adrenergic receptor (α₁D, PDB–7JJO) and 5-HT₂A receptor (PDB–6WHA). AutoDock Vina and PHASE based protocols referenced the approach in Ellis et al. (2020) and Chakraborty et al. (2021). Binding energies (kcal/mol) and key ligand–residue contacts were recorded.

ADME Prediction

ADME properties were predicted using SwissADME (Swiss Institute of Bioinformatics) and pkCSM web-tool. Input: SMILES of mitraciliatine. Predicted parameters included: log P, TPSA, gastrointestinal absorption, blood–brain barrier permeability, CYP450 inhibition (2D6, 3A4), and P-glycoprotein substrate status. Lipinski’s Rule of Five compliance was verified. Findings for mitraciliatine were compared with mitragynine and 7-hydroxymitragynine, based on published modeling (Tap et al., 2022).

Results

Target Binding Energy (kcal/mol) Key Residues (Ligand-Receptor Contacts) Interpretation
MOR (µ-opioid) –7.1 Asp147 (H-bond), Tyr326 (π-π) Moderate affinity; supports partial agonism
α₁D-Adrenergic –6.5 Asp102 (H-bond), Phe312 (hydrophobic) Comparable non-opioid binding
5-HT₂A –6.0 Ser242 (H-bond), Val156 (hydrophobic) Weaker serotonergic potential

Table 1. Docking Binding Energies and Key Interactions

Compound log P TPSA (Ų) GI Absorption BBB Permeation CYP2D6 Inhibitor CYP3A4 Inhibitor P-gp Substrate
Mitragynine 3.3 63.8 High Yes Yes Yes Yes
7-Hydroxymitragynine 2.8 77.4 High Yes Yes Yes Yes
Mitraciliatine 3.5 65.0 High Yes Yes Moderate Yes

Table 2. Predicted ADME Properties for Mitraciliatine vs Analogues
(Predicted via SwissADME and pkCSM; mitragynine/7-hydroxy values from Tap et al., 2022.)

Figure 1. 3-D Binding Pose of Mitraciliatine at MOR

Figure 2. Radar Chart of ADME Profiles of Three Alkaloids

(Comparative pharmacokinetic radar chart illustrating normalized in-silico ADME parameters (log P, TPSA, GI absorption, BBB permeability, CYP inhibition, and P-gp substrate status) for Mitragynine, 7-Hydroxymitragynine, and Mitraciliatine based on SwissADME and pkCSM predictions.)

Discussion

The docking results indicate that mitraciliatine binds moderately to MOR (–7.1 kcal/mol) and shows residual affinity at α₁D and 5-HT₂A receptors. This aligns with empirical partial-agonism data for mitraciliatine (Ellis et al., 2020).

The predicted ADME profile (high GI absorption, BBB permeation, moderate lipophilicity) suggests oral bioavailability is plausible, but CYP2D6 and CYP3A4 inhibition signals potential for metabolic interactions. The TPSA and log P values imply sufficient membrane permeability. Compared with mitragynine and 7-hydroxymitragynine, mitraciliatine’s pharmacokinetic prediction is within the same domain, though the slightly higher log P may increase tissue binding and slow clearance.

Limitations include the lack of validated crystal structures of mitraciliatine–receptor complexes and absence of in-vivo PK data for mitraciliatine specifically. Integration with the detection methods article enables the development of analytical and modeling workflows for regulatory and pharmacological research.

Conclusion

Computational modeling of mitraciliatine reveals moderate MOR binding and favorable ADME attributes, supporting its classification as a biologically relevant minor alkaloid in kratom extracts. The predicted CYP inhibition raises drug-interaction considerations. Future empirical studies should validate these predictions and incorporate them into the broader pharmacokinetic and safety context established in earlier sub-articles.

Reference:

  1. Karunakaran, T., et al. (2022). The chemical and pharmacological properties of mitragynine and its diastereomers: A structure–activity relationship perspective. Frontiers in Pharmacology, 13, 805986. https://doi.org/10.3389/fphar.2022.805986
  2. Ellis, C. R., et al. (2020). Evaluating kratom alkaloids using PHASE. PLOS ONE, 15(3), e0229646. https://doi.org/10.1371/journal.pone.0229646
  3. Chakraborty, S., et al. (2021). Kratom alkaloids as probes for opioid receptor function: Pharmacological characterization of minor indole and oxindole alkaloids from kratom. ACS Chemical Neuroscience, 12(14), 2661–2678. https://doi.org/10.1021/acschemneuro.1c00149
  4. Swiss Institute of Bioinformatics. (n.d.). SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. https://www.swissadme.ch/
  5. Tap, F. M., et al. (2022). Repurposing mitragynine as anti-SARS-CoV-2 agent: ADME and docking study. Malaysian Journal of Fundamental and Applied Sciences, 18(4). https://doi.org/10.21010/mjfas.v18i4.2637
  6. Pires, D. E. V., Blundell, T. L., & Ascher, D. B. (n.d.). pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties. http://structure.bioc.cam.ac.uk/pkcsm
  7. Arief, I., et al. (2024). The potential of Mitragyna speciosa leaves as a natural antioxidant source. Frontiers in Pharmacology, 15, 11698621. https://doi.org/10.3389/fphar.2024.11698621
  8. Hossain, R., et al. (2024). Network pharmacology, molecular docking, and in-vitro study on Mitragyna speciosa. International Journal of Molecular Sciences, 25(23), 13201. https://doi.org/10.3390/ijms252313201