PDF
Abstract
Background: Alzheimer's disease (AD) is a growing healthcare crisis with limited effective therapies. This study aims to identify new candidate drugs that can be repurposed using key transcriptional regulators (DERs) in AD as therapeutic targets.
Methods: Multi-cohort single-nucleus RNA sequencing (snRNA-seq) data from the prefrontal cortex were analysed to identify DERs. Molecular docking and dynamic simulations analysis evaluated interactions between DERs and 2200 Food and Drug Administration-approved drugs to assess binding stability, whilst pharmacokinetic parameters relevant to blood–brain barrier permeability were evaluated.
Results: We identified 20 key DERs associated with AD. Lasmiditan stood out as the most promising drug amongst other drug candidates (Vorapaxar, Bictegravir, Tonaftate, Fluspirilene, Lisuride, Olaparib) interacting with five DERs: ZEB2, APP, PAX6, ETV6, and ST18. Lasmiditan–ETV6 complex showed the best binding stability (RMSD: 2.98 Å, H-bonds: 68.38) and optimal passive diffusion (LogP3–4, TPSA 60–75 Å2).
Discussion: Lasmiditan is a potential AD therapeutic candidate that warrants further preclinical validation.
Keywords
Alzheimer's disease
/
drug repurposing
/
gene regulators
/
Lasmiditan
/
snRNA-Seq
Cite this article
Download citation ▾
Martin Nwadiugwu, Selim Reza, Boluwatife Afolabi, Demetrius M. Maraganore, Hui Shen, Hongwen Deng.
Integrative snRNA-seq, molecular docking and dynamics simulations identifies Lasmiditan as drug candidate for Alzheimer's disease.
Clinical and Translational Medicine, 2025, 15(8): e70443 DOI:10.1002/ctm2.70443
| [1] |
Grabher BJ. Effects of Alzheimer disease on patients and their family. J Nucl Med Technol. 2018; 46: 335-340.
|
| [2] |
Alzheimer's Association. 2022 Alzheimer's disease facts and figures. Alzheimers Dement. 2022; 18: 700-789.
|
| [3] |
Palmqvist S, Janelidze S, Stomrud E, et al. Performance of fully automated plasma assays as screening tests for Alzheimer disease–related β-amyloid status. JAMA Neurol. 2019; 76: 1060.
|
| [4] |
Centers for Medicare & Medicaid Services (CMS). Amyloid PET. 2024. Accessed May 4, 2025. https://www.cms.gov/medicare/coverage/evidence/amyloid-pet
|
| [5] |
Mintun MA, Lo AC, Duggan Evans C, et al. Donanemab in early Alzheimer's disease. N Engl J Med. 2021; 384: 1691-1704.
|
| [6] |
Brockmann R, Nixon J, Love BL, Yunusa I. Impacts of FDA approval and Medicare restriction on antiamyloid therapies for Alzheimer's disease: patient outcomes, healthcare costs, and drug development. Lancet. 2023; 20: 100467.
|
| [7] |
Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of early Alzheimer's disease: clinical practice in 2021. J Prev Alzheimers Dis. 2021; 8: 371-386.
|
| [8] |
Nwadiugwu M. Early-onset dementia: key issues using a relationship-centred care approach. Postgrad Med J. 2021; 97: 598-604.
|
| [9] |
Nwadiugwu M, Shen H, Deng H-W. Potential Molecular Mechanisms of Alzheimer's Disease from Genetic Studies. Biology (Basel). 2023; 12: 602.
|
| [10] |
Nwadiugwu M, Onwuekwe I, Ezeanolue E, Deng H. Beyond amyloid: a machine learning-driven approach reveals properties of potent GSK-3β inhibitors targeting neurofibrillary tangles. Int J Mol Sci. 2024; 25: 2646.
|
| [11] |
Albrecht B, C K, A S, G D, M P Pursuing breakthroughs in cancer-drug development. 2025. Accessed May 4, 2025. https://www.mckinsey.com/industries/life-sciences/our-insights/pursuing-breakthroughs-in-cancer-drug-development. https://www.mckinsey.com/industries/life-sciences/our-insights/pursuing-breakthroughs-in-cancer-drug-development
|
| [12] |
Cummings J. Lessons learned from Alzheimer disease: clinical trials with negative outcomes. Clin Transl Sci. 2018; 11: 147-152.
|
| [13] |
Cummings J, Zhou Y, Lee G, Zhong K, Fonseca J, Cheng F. Alzheimers Dement. 2024; 10: e12465.
|
| [14] |
Zhuang J-J, Liu Q, Wu D-L, Tie L. Current strategies and progress for targeting the ‘undruggable’ transcription factors. Acta Pharmacol Sin. 2022; 43: 2474-2481.
|
| [15] |
Xie X, Yu T, Li X, et al. Recent advances in targeting the “undruggable” proteins: from drug discovery to clinical trials. Signal Transduct Target Ther. 2023; 8: 335.
|
| [16] |
Ezebuo FC, Uzochukwu IC. Drug repurposing for schistosomiasis: molecular docking and dynamics investigations. J Biomol Struct Dyn. 2022; 40: 995-1009.
|
| [17] |
Sadybekov AV, Katritch V. Computational approaches streamlining drug discovery. Nature. 2023; 616: 673-685.
|
| [18] |
Saikia S, Bordoloi M. Molecular docking: challenges, advances and its use in drug discovery perspective. Curr Drug Targets. 2019; 20: 501-521.
|
| [19] |
Knox C, Wilson M, Klinger CM, et al. DrugBank 6.0: the DrugBank Knowledgebase for 2024. Nucleic Acids Res. 2024; 52: D1265-D1275.
|
| [20] |
Dolinsky TJ, Czodrowski P, Li H, et al. PDB2PQR: expanding and upgrading automated preparation of biomolecular structures for molecular simulations. Nucleic Acids Res. 2007; 35: W522-5.
|
| [21] |
Gordon JC, Myers JB, Folta T, Shoja V, Heath LS, Onufriev A. H++: a server for estimating pKas and adding missing hydrogens to macromolecules. Nucleic Acids Res. 2005; 33: W368-W371.
|
| [22] |
Hanwell MD, Curtis DE, Lonie DC, Vandermeersch T, Zurek E, Hutchison GR. Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. J Cheminform. 2012; 4: 17.
|
| [23] |
Wu Q, Peng Z, Zhang Y, Yang J. COACH-D: improved protein-ligand binding sites prediction with refined ligand-binding poses through molecular docking. Nucleic Acids Res. 2018; 46: W438-W442.
|
| [24] |
Morris GM, Huey R, Lindstrom W, et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J Comput Chem. 2009; 30: 2785-2791.
|
| [25] |
Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010; 31: 455-461.
|
| [26] |
Hollingsworth SA, Dror RO. Molecular dynamics simulation for all. Neuron. 2018; 99: 1129-1143.
|
| [27] |
Phillips JC, Braun R, Wang W, et al. Scalable molecular dynamics with NAMD. J Comput Chem. 2005; 26: 1781-1802.
|
| [28] |
Jo S, Kim T, Iyer VG, Im W. CHARMM-GUI: a web-based graphical user interface for CHARMM. J Comput Chem. 2008; 29: 1859-1865.
|
| [29] |
Shrake A, Rupley JA. Environment and exposure to solvent of protein atoms. Lysozyme and insulin. J Mol Biol. 1973; 79: 351-371.
|
| [30] |
McGibbon RT, Beauchamp KA, Harrigan MP, et al. MDTraj: a Modern Open Library for the analysis of molecular dynamics trajectories. Biophys J. 2015; 109: 1528-1532.
|
| [31] |
Gaulton A, Bellis LJ, Bento AP, et al. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 2012; 40: D1100-7.
|
| [32] |
RDKit. Open-Source Cheminformatics Software. Accessed Nov 24, 2022. https://rdkit.org
|
| [33] |
Clark DE. Rapid calculation of polar molecular surface area and its application to the prediction of transport phenomena. 1. Prediction of intestinal absorption. J Pharm Sci. 1999; 88: 807-814.
|
| [34] |
Anderson AG, Rogers BB, Loupe JM, et al. Single nucleus multiomics identifies ZEB1 and MAFB as candidate regulators of Alzheimer's disease-specific cis-regulatory elements. Cell Genomics. 2023; 3: 100263.
|
| [35] |
Morabito S, Miyoshi E, Michael N, et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer's disease. Nat Genet. 2021; 53: 1143-1155.
|
| [36] |
Mathys H, Davila-Velderrain J, Peng Z, et al. Single-cell transcriptomic analysis of Alzheimer's disease. Nature. 2019; 570: 332-337.
|
| [37] |
Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell. 2021; 184: 3573-3587.e29.
|
| [38] |
Aran D, Looney AP, Liu L, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019; 20: 163-172.
|
| [39] |
Adams D, Altucci L, Antonarakis SE, et al. BLUEPRINT to decode the epigenetic signature written in blood. Nature Biotechnology. 2012; 30: 224-226.
|
| [40] |
ENCODE Project Consortium, Moore JE, Purcaro MJ, et al. Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature. 2020; 583: 699-710.
|
| [41] |
Cao K, Gong Q, Hong Y, Wan L. A unified computational framework for single-cell data integration with optimal transport. Nat Commun. 2022; 13: 7419.
|
| [42] |
Alvarez MJ, Shen Y, Giorgi FM, et al. Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nat Genet. 2016; 48: 838-847.
|
| [43] |
Becht E, McInnes L, Healy J, et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol. 2019; 37: 104749.
|
| [44] |
Blondel VD, Guillaume J-L, Lambiotte R, Lefebvre E. Fast unfolding of communities in large networks—Abstract. J Stat Mech 2008; 2008: P10008.
|
| [45] |
Rousseeuw PJ. Silhouettes: a graphical aid to the interpretation and validation of cluster analysis. J Comput Appl Math. 1987; 20: 53-65.
|
| [46] |
Kim SC, Lee SJ, Lee WJ, et al. Stouffer's test in a large scale simultaneous hypothesis testing. PLoS One. 2013; 8: e63290.
|
| [47] |
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15: 550.
|
| [48] |
RStudio Team. RStudio: Integrated Development for R. (2022).
|
| [49] |
Keenan AB, Torre D, Lachmann A, et al. ChEA3: transcription factor enrichment analysis by orthogonal omics integration. Nucleic Acids Res. 2019; 47: W212-W224.
|
| [50] |
Ashraf SA, Elkhalifa AEO, Mehmood K, et al. Multi-targeted molecular docking, pharmacokinetics, and drug-likeness evaluation of okra-derived ligand abscisic acid targeting signaling proteins involved in the development of diabetes. Molecules. 2021; 26: 5957.
|
| [51] |
Lachmann A, Giorgi FM, Lopez G, Califano A. ARACNe-AP: gene network reverse engineering through adaptive partitioning inference of mutual information. Bioinformatics. 2016; 32: 2233-2235.
|
| [52] |
Zhou F, Chen B. Acute myeloid leukemia carrying ETV6 mutations: biologic and clinical features. Hematology. 2018; 23: 608-612.
|
| [53] |
Tang Y, Xu G, Hu B, Zhu Y. HIVEP3 as a potential prognostic factor promotes the development of acute myeloid leukemia. Growth Factors. 2023; 41: 43-56.
|
| [54] |
El Menshawy N, El-Ghonemy MS, Ebrahim MA, et al. Aberrant ecotropic viral integration site-1 (EVI-1) and myocyte enhancer factor 2 C gene (MEF2C) in adult acute myeloid leukemia are associated with adverse t (9:22) & 11q23 rearrangements. Ann Hematol. 2024; 103: 2355-2364.
|
| [55] |
Ninkovic J, Steiner-Mezzadri A, Jawerka M, et al. The BAF complex interacts with Pax6 in adult neural progenitors to establish a neurogenic cross-regulatory transcriptional network. Cell Stem Cell. 2013; 13: 403-418.
|
| [56] |
VandenBosch LS, Wohl SG, Wilken MS, et al. Developmental changes in the accessible chromatin, transcriptome and Ascl1-binding correlate with the loss in Müller Glial regenerative potential. Sci Rep. 2020; 10: 13615.
|
| [57] |
García-Ortegón M, Simm GNC, Tripp AJ, Hernández-Lobato JM, Bender A, Bacallado S. DOCKSTRING: easy molecular docking yields better benchmarks for ligand design. J Chem Inf Model. 2022; 62: 3486-3502.
|
| [58] |
Dankwa B, Broni E, Enninful KS, Kwofie SK, Wilson MD. Consensus docking and MM-PBSA computations identify putative furin protease inhibitors for developing potential therapeutics against COVID-19. Struct Chem. 2022; 33: 2221-2241.
|
| [59] |
Guedes IA, Costa LSC, dos Santos KB, et al. Drug design and repurposing with DockThor-VS web server focusing on SARS-CoV-2 therapeutic targets and their non-synonym variants. Sci Rep. 2021; 11: 5543.
|
| [60] |
Lapins M, Arvidsson S, Lampa S, et al. A confidence predictor for logD using conformal regression and a support-vector machine. J Cheminform. 2018; 10: 17.
|
| [61] |
Hughes JD, Blagg J, Price DA, et al. Physiochemical drug properties associated with in vivo toxicological outcomes. Bioorg Med Chem Lett. 2008; 18: 4872-4875.
|
| [62] |
Sozio P, Cerasa LS, Marinelli L, Di Stefano A. Transdermal donepezil on the treatment of Alzheimer's disease. Neuropsychiatr Dis Treat. 2012; 8: 361-368.
|
| [63] |
National Center for Biotechnology Information. PubChem Compound Summary for CID 5073, Risperidone. 2025. Accessed May 4, 2025. https://pubchem.ncbi.nlm.nih.gov/compound/Risperidone
|
| [64] |
Rana N, Solanki P, Mehra R, Manhas A. Identification of natural compound inhibitors for substrate-binding site of MTHFD2 enzyme: insights from structure-based drug design and biomolecular simulations. Chem Phys Impact. 2025; 10: 100809.
|
| [65] |
Claus JJ, de Koning I, van Harskamp F, et al. Lisuride treatment of Alzheimer's disease. A preliminary placebo-controlled clinical trial of safety and therapeutic efficacy. Clin Neuropharmacol. 1998; 21: 190-195.
|
| [66] |
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther. 2023; 8: 359.
|
| [67] |
Villar J, Cros A, De Juan A, et al. ETV3 and ETV6 enable monocyte differentiation into dendritic cells by repressing macrophage fate commitment. Nat Immunol. 2023; 24: 84-95.
|
| [68] |
Rao C, Semrau S, Fossati V. Decoding microglial functions in Alzheimer's disease: insights from human models. Trends Immunol. 2025; 46: 310-323.
|
| [69] |
Miao J, Ma H, Yang Y, et al. Microglia in Alzheimer's disease: pathogenesis, mechanisms, and therapeutic potentials. Front Aging Neurosci. 2023; 15: 1201982.
|
| [70] |
Garner LC, Amini A, FitzPatrick MEB, et al. Single-cell analysis of human MAIT cell transcriptional, functional and clonal diversity. Nat Immunol. 2023; 24: 1565-1578.
|
| [71] |
Wager TT, Hou X, Verhoest PR, Villalobos A. Moving beyond rules: the development of a central nervous system multiparameter optimization (CNS MPO) approach to enable alignment of druglike properties. ACS Chem Neurosci. 2010; 1: 435-449.
|
| [72] |
Berger AA, Winnick A, Popovsky D, et al. Lasmiditan for the treatment of migraines with or without aura in adults. Psychopharmacol Bull. 2020; 50: 163-188.
|
| [73] |
Clemow DB, Johnson KW, Hochstetler HM, Ossipov MH, Hake AM, Blumenfeld AM. Lasmiditan mechanism of action—review of a selective 5-HT1F agonist. J Headache Pain. 2020; 21: 71.
|
| [74] |
Mitsikostas DD, Sanchez del Rio M, Moskowitz MA, Waeber C. Both 5-HT1B and 5-HT1F receptors modulate c-fos expression within rat trigeminal nucleus caudalis. Eur J Pharmacol. 1999; 369: 271-277.
|
| [75] |
Xiao Y, Richter JA, Hurley JH. Release of glutamate and CGRP from trigeminal ganglion neurons: role of calcium channels and 5-HT1 receptor signaling. Mol Pain. 2008; 4: 12.
|
| [76] |
Beauchene JK, Levien TLL. Acute migraine treatment without vasoconstriction. A review. J Pharm Technol. 2021; 37: 244-253.
|
| [77] |
Hou M, Xing H, Li C, et al. Short-term efficacy and safety of lasmiditan, a novel 5-HT1F receptor agonist, for the acute treatment of migraine: a systematic review and meta-analysis. J Headache Pain. 2020; 21: 66.
|
| [78] |
Glatfelter GC, Pottie E, Partilla JS, Stove CP, Baumann MH. Comparative pharmacological effects of lisuride and lysergic acid diethylamide revisited. ACS Pharmacol Transl Sci. 2024; 7: 641-653.
|
| [79] |
Drugs and Lactation Database (LactMed®). In: Lisuride. National Institute of Child Health and Human Development; 2006. [Updated 2021 Jul 19]. Accessed May 4, 2025. https://www.ncbi.nlm.nih.gov/books/NBK500804/
|
| [80] |
Horvath J, Fross RD, Kleiner-Fisman G, et al. Severe multivalvular heart disease: a new complication of the ergot derivative dopamine agonists. Mov Disord. 2004; 19: 656-662.
|
| [81] |
Jain H, Bhat AR, Dalvi H, Godugu C, Singh SB, Srivastava S. Repurposing approved therapeutics for new indication: addressing unmet needs in psoriasis treatment. Curr Res Pharmacol Drug Discov. 2021; 2: 100041.
|
| [82] |
Pushpakom S, Iorio F, Eyers PA, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov. 2019; 18: 41-58.
|
RIGHTS & PERMISSIONS
2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.