Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

AI-enhanced virtual screening approach to hit identification for GluN1/GluN3A NMDA receptor

Abstract

N-methyl-D-aspartate receptors (NMDARs) are calcium-permeable ionotropic glutamate receptors broadly expressed throughout the central nervous system, where they play crucial roles in neuronal development and synaptic plasticity. Among the various subtypes, the GluN1/GluN3A receptor represents a unique glycine-gated NMDAR with notably low calcium permeability. Despite its distinctive properties, GluN1/GluN3A remains understudied, particularly with respect to pharmacological tools development. This scarcity poses challenges for deeper investigation into its physiological functions and therapeutic relevance. In this study, we employed a hybrid virtual screening (VS) pipeline that integrates ligand-based and structure-based approaches for the efficient and precise identification of small-molecule candidates targeting GluN1/GluN3A. A large compound library comprising 18 million molecules was screened using an AI-enhanced multi-stage method. The initial phase utilized shape similarity ranking via ROCS-BART, followed by refinement with a graph neural network (GNN)-based drug-target interaction model to enhance docking accuracy. Functional validation using calcium flux (FDSS/μCell) identified two compounds with IC50 values below 10 μM. Of these, one candidate exhibited potent inhibitory activity with an IC50 of 5.31 ± 1.65 μM, which was further confirmed through manual patch-clamp recordings. These findings highlight an AI-enhanced VS workflow that achieves both efficiency and precision, providing a promising framework for exploring elusive targets such as GluN1/GluN3A.

This is a preview of subscription content, access via your institution

Access options

Buy this article

USD 39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: AI-enhanced virtual screening workflow.
Fig. 2: Diversity analysis of ROCS scoring.
Fig. 3: Workflow for ROCS scoring prediction using a pretrained unsupervised model.
Fig. 4: GluN1/GluN3A structure and potential allosteric binding sites.
Fig. 5: The performance of the ROCS-BART model in screening.
Fig. 6: Inhibitory activity and predicted binding mode of LP-2 on GluN1/GluN3A receptor.

Similar content being viewed by others

References

  1. Hansen KB, Wollmuth LP, Bowie D, Furukawa H, Menniti FS, Sobolevsky AI, et al. Structure, function, and pharmacology of glutamate receptor ion channels. Pharmacol Rev. 2021;73:298–487.

    PubMed  PubMed Central  Google Scholar 

  2. Paoletti P, Bellone C, Zhou Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci. 2013;14:383–400.

    PubMed  Google Scholar 

  3. Hansen KB, Yi F, Perszyk RE, Furukawa H, Wollmuth LP, Gibb AJ, et al. Structure, function, and allosteric modulation of NMDA receptors. J Gen Physiol. 2018;150:1081–105.

    PubMed  PubMed Central  Google Scholar 

  4. Ahmed H, Haider A, Ametamey SM. N-Methyl-D-Aspartate (NMDA) receptor modulators: a patent review (2015-present). Expert Opin Ther Pat. 2020;30:743–67.

    PubMed  Google Scholar 

  5. Egunlusi AO, Joubert J. NMDA receptor antagonists: emerging insights into molecular mechanisms and clinical applications in neurological disorders. Pharmaceuticals. 2024;17:639

    PubMed  PubMed Central  Google Scholar 

  6. Chatterton JE, Awobuluyi M, Premkumar LS, Takahashi H, Talantova M, Shin Y, et al. Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature. 2002;415:793–8.

    PubMed  Google Scholar 

  7. Sasaki YF, Rothe T, Premkumar LS, Das S, Cui J, Talantova MV, et al. Characterization and comparison of the NR3A subunit of the NMDA receptor in recombinant systems and primary cortical neurons. J Neurophysiol. 2002;87:2052–63.

    PubMed  Google Scholar 

  8. Pérez-Otaño I, Larsen RS, Wesseling JF. Emerging roles of GluN3-containing NMDA receptors in the CNS. Nat Rev Neurosci. 2016;17:623–35.

    PubMed  Google Scholar 

  9. Marco S, Giralt A, Petrovic MM, Pouladi MA, Martínez-Turrillas R, Martínez-Hernández J, et al. Suppressing aberrant GluN3A expression rescues synaptic and behavioral impairments in Huntington’s disease models. Nat Med. 2013;19:1030–8.

    PubMed  PubMed Central  Google Scholar 

  10. Yuan T, Mameli M, O’Connor EC, Dey PN, Verpelli C, Sala C, et al. Expression of cocaine-evoked synaptic plasticity by GluN3A-containing NMDA receptors. Neuron. 2013;80:1025–38.

    PubMed  Google Scholar 

  11. Mueller HT, Meador-Woodruff JH. NR3A NMDA receptor subunit mRNA expression in schizophrenia, depression and bipolar disorder. Schizophr Res. 2004;71:361–70.

    PubMed  Google Scholar 

  12. Beesley S, Sullenberger T, Crotty K, Ailani R, D’Orio C, Evans K, et al. D-serine mitigates cell loss associated with temporal lobe epilepsy. Nat Commun. 2020;11:4966.

    PubMed  PubMed Central  Google Scholar 

  13. Pfisterer U, Petukhov V, Demharter S, Meichsner J, Thompson JJ, Batiuk MY, et al. Identification of epilepsy-associated neuronal subtypes and gene expression underlying epileptogenesis. Nat Commun. 2020;11:5038.

    PubMed  PubMed Central  Google Scholar 

  14. Grand T, Abi Gerges S, David M, Diana MA, Paoletti P. Unmasking GluN1/GluN3A excitatory glycine NMDA receptors. Nat Commun. 2018;9:4769.

    PubMed  PubMed Central  Google Scholar 

  15. Kvist T, Greenwood JR, Hansen KB, Traynelis SF, Bräuner-Osborne H. Structure-based discovery of antagonists for GluN3-containing N-methyl-D-aspartate receptors. Neuropharmacology. 2013;75:324–36.

    PubMed  Google Scholar 

  16. Zhu Z, Yi F, Epplin MP, Liu D, Summer SL, Mizu R, et al. Negative allosteric modulation of GluN1/GluN3 NMDA receptors. Neuropharmacology. 2020;176:108117.

    PubMed  PubMed Central  Google Scholar 

  17. Zeng Y, Zheng Y, Zhang T, Ye F, Zhan L, Kou Z, et al. Identification of a subtype-selective allosteric inhibitor of GluN1/GluN3 NMDA receptors. Front Pharmacol. 2022;13:888308.

    PubMed  PubMed Central  Google Scholar 

  18. Karakas E, Simorowski N, Furukawa H. Subunit arrangement and phenylethanolamine binding in GluN1/GluN2B NMDA receptors. Nature. 2011;475:249–53.

    PubMed  PubMed Central  Google Scholar 

  19. Bettini E, Sava A, Griffante C, Carignani C, Buson A, Capelli AM, et al. Identification and characterization of novel NMDA receptor antagonists selective for NR2A- over NR2B-containing receptors. J Pharmacol Exp Ther. 2010;335:636–44.

    PubMed  Google Scholar 

  20. Yi F, Mou TC, Dorsett KN, Volkmann RA, Menniti FS, Sprang SR, et al. Structural basis for negative allosteric modulation of GluN2A-containing NMDA receptors. Neuron. 2016;91:1316–29.

    PubMed  PubMed Central  Google Scholar 

  21. Hackos DH, Lupardus PJ, Grand T, Chen Y, Wang TM, Reynen P, et al. Positive allosteric modulators of GluN2A-containing NMDARs with distinct modes of action and impacts on circuit function. Neuron. 2016;89:983–99.

    PubMed  Google Scholar 

  22. Vázquez J, López M, Gibert E, Herrero E, Luque FJ. Merging ligand-based and structure-based methods in drug discovery: an overview of combined virtual screening approaches. Molecules. 2020;25:4723.

    PubMed  PubMed Central  Google Scholar 

  23. Hawkins PC, Skillman AG, Nicholls A. Comparison of shape-matching and docking as virtual screening tools. J Med Chem. 2007;50:74–82.

    PubMed  Google Scholar 

  24. Lewis M, Yinhan L, Goyal N, Ghazvininejad M, Mohamed A, Levy O, et al. BART: denoising sequence-to-sequence pre-training for natural language generation, translation, and comprehension. In: Proceedings of the 58th annual meeting of the association for computational linguistics. Association for Computational; 2020. pp. 7871–80. online.

  25. Moon S, Hwang S-Y, Lim J, Kim WY. PIGNet2: a versatile deep learning-based protein–ligand interaction prediction model for binding affinity scoring and virtual screening. Digital Discov. 2024;3:287–99.

    Google Scholar 

  26. Yao Y, Belcher J, Berger AJ, Mayer ML, Lau AY. Conformational analysis of NMDA receptor GluN1, GluN2, and GluN3 ligand-binding domains reveals subtype-specific characteristics. Structure. 2013;21:1788–99.

    PubMed  Google Scholar 

  27. Fiser A, Sali A. Modeller: generation and refinement of homology-based protein structure models. Methods Enzymol. 2003;374:461–91.

    PubMed  Google Scholar 

  28. Huey R, Morris GM, Olson AJ, Goodsell DS. A semiempirical free energy force field with charge-based desolvation. J Comput Chem. 2007;28:1145–52.

    PubMed  Google Scholar 

  29. Berendsen HJC, van der Spoel D, van Drunen R. GROMACS: a message-passing parallel molecular dynamics implementation. Comput Phys Commun. 1995;91:43–56.

    Google Scholar 

  30. Abraham MJ, Murtola T, Schulz R, Páll S, Smith JC, Hess B, et al. GROMACS: High performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX. 2015;1-2:19–25.

  31. Vanommeslaeghe K, Hatcher E, Acharya C, Kundu S, Zhong S, Shim J, et al. CHARMM general force field: a force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. J Comput Chem. 2010;31:671–90.

    PubMed  PubMed Central  Google Scholar 

  32. Bepari AK, Reza HM. Identification of a novel inhibitor of SARS-CoV-2 3CL-PRO through virtual screening and molecular dynamics simulation. PeerJ. 2021;9:e11261.

    PubMed  PubMed Central  Google Scholar 

  33. Colovos C, Yeates TO. Verification of protein structures: patterns of nonbonded atomic interactions. Protein Sci. 1993;2:1511–9.

    PubMed  PubMed Central  Google Scholar 

  34. Laskowski RA, MacArthur MW, Moss DS, Thornton JM. PROCHECK: a program to check the stereochemical quality of protein structures. J Appl Cryst. 1993;26:283–91.

    Google Scholar 

  35. Lovell SC, Davis IW, Arendall WB 3rd, de Bakker PI, Word JM, Prisant MG, et al. Structure validation by Calpha geometry: phi, psi and Cbeta deviation. Proteins. 2003;50:437–50.

    PubMed  Google Scholar 

  36. Volgraf M, Sellers BD, Jiang Y, Wu G, Ly CQ, Villemure E, et al. Discovery of GluN2A-selective NMDA receptor positive allosteric modulators (PAMs): tuning deactivation kinetics via structure-based design. J Med Chem. 2016;59:2760–79.

    PubMed  Google Scholar 

  37. Irwin R, Dimitriadis S, He J, Bjerrum EJ. Chemformer: a pre-trained transformer for computational chemistry. Mach Learn Sci Technol. 2022;3:015022.

    Google Scholar 

  38. Friesner RA, Banks JL, Murphy RB, Halgren TA, Klicic JJ, Mainz DT, et al. Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J Med Chem. 2004;47:1739–49.

    PubMed  Google Scholar 

  39. Shen C, Zhang X, Hsieh CY, Deng Y, Wang D, Xu L, et al. A generalized protein-ligand scoring framework with balanced scoring, docking, ranking and screening powers. Chem Sci. 2023;14:8129–46.

    PubMed  PubMed Central  Google Scholar 

  40. Bossi S, Pizzamiglio L, Paoletti P. Excitatory GluN1/GluN3A glycine receptors (eGlyRs) in brain signaling. Trends Neurosci. 2023;46:667–81.

    PubMed  Google Scholar 

  41. Villemure E, Volgraf M, Jiang Y, Wu G, Ly CQ, Yuen PW, et al. GluN2A-selective pyridopyrimidinone series of NMDAR positive allosteric modulators with an improved in vivo profile. ACS Med Chem Lett. 2016;8:84–9.

    PubMed  PubMed Central  Google Scholar 

  42. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596:583–9.

    PubMed  PubMed Central  Google Scholar 

  43. Abramson J, Adler J, Dunger J, Evans R, Green T, Pritzel A, et al. Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature. 2024;630:493–500.

    PubMed  PubMed Central  Google Scholar 

  44. Bondi A. van der Waals volumes and radii. J Phys Chem. 1964;68:441–51.

    Google Scholar 

Download references

Acknowledgements

This work was supported by the Strategic Priority Research Program of the Chinese Academy of Sciences (Grant ID: XDB0830403) and the National Science and Technology Innovation 2030 Major Program (Grant ID: 2021ZD0200900). The authors gratefully acknowledge Professor Mao-lin Wang from the First Affiliated Hospital of Shantou University Medical College for his invaluable technical support and assistance with the OpenEye software.

Author information

Authors and Affiliations

Authors

Contributions

YK and ZBG designed the study. YK, YSJ, SFH, BCZ, SWL, and CL designed the virtual screening strategy. BCZ built the homology model and analyzed the structure-activity relationship. SWL, SFH and YK constructed the ROCS-BART model and deployed PIGNet2. YSJ calculated the virtual screening scores. YSJ and BCZ analyzed the virtual screening results and selected the candidate compounds. ZBG and YZ designed wet-lab experiments. YZ, HCW and AYW conducted FDSS/μCell screening and whole-cell patch clamp recording. YSJ, SFH and YZ wrote the manuscript. All authors reviewed the manuscript before submission.

Corresponding authors

Correspondence to Zhao-bing Gao or Yue Kong.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ji, Ys., Zeng, Y., Hu, Sf. et al. AI-enhanced virtual screening approach to hit identification for GluN1/GluN3A NMDA receptor. Acta Pharmacol Sin 47, 41–52 (2026). https://doi.org/10.1038/s41401-025-01644-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Version of record:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41401-025-01644-1

Keywords

This article is cited by

Search

Quick links