From single drug targets to synergistic network pharmacology in ischemic stroke

Ana I Casas, Ahmed A Hassan, Simon J Larsen, Vanessa Gomez-Rangel, Mahmoud Elbatreek, Pamela W M Kleikers, Emre Guney, Javier Egea, Manuela G López, Jan Baumbach, Harald H H W Schmidt, Ana I Casas, Ahmed A Hassan, Simon J Larsen, Vanessa Gomez-Rangel, Mahmoud Elbatreek, Pamela W M Kleikers, Emre Guney, Javier Egea, Manuela G López, Jan Baumbach, Harald H H W Schmidt

Abstract

Drug discovery faces an efficacy crisis to which ineffective mainly single-target and symptom-based rather than mechanistic approaches have contributed. We here explore a mechanism-based disease definition for network pharmacology. Beginning with a primary causal target, we extend this to a second using guilt-by-association analysis. We then validate our prediction and explore synergy using both cellular in vitro and mouse in vivo models. As a disease model we chose ischemic stroke, one of the highest unmet medical need indications in medicine, and reactive oxygen species forming NADPH oxidase type 4 (Nox4) as a primary causal therapeutic target. For network analysis, we use classical protein-protein interactions but also metabolite-dependent interactions. Based on this protein-metabolite network, we conduct a gene ontology-based semantic similarity ranking to find suitable synergistic cotargets for network pharmacology. We identify the nitric oxide synthase (Nos1 to 3) gene family as the closest target to Nox4 Indeed, when combining a NOS and a NOX inhibitor at subthreshold concentrations, we observe pharmacological synergy as evidenced by reduced cell death, reduced infarct size, stabilized blood-brain barrier, reduced reoxygenation-induced leakage, and preserved neuromotor function, all in a supraadditive manner. Thus, protein-metabolite network analysis, for example guilt by association, can predict and pair synergistic mechanistic disease targets for systems medicine-driven network pharmacology. Such approaches may in the future reduce the risk of failure in single-target and symptom-based drug discovery and therapy.

Keywords: NOX4; network analysis; network pharmacology; stroke.

Conflict of interest statement

Conflict of interest statement: H.H.H.W.S. is a cofounder of a biotech company, Vasopharm, engaged in the development of small-molecule NOS inhibitors, currently in stage III clinical development. However, H.H.H.W.S. has no operative role in the company and holds less than 1% of shares.

Copyright © 2019 the Author(s). Published by PNAS.

Figures

Fig. 1.
Fig. 1.
Computational workflow for target prioritization via network pharmacology. The computational target prioritization pipeline consists of three interdependent modules. The blue module extracts the metabolites interacting with the protein NOX4 from the Human Metabolome Database, performs curation of the metabolites, extracts the proteins interacting with them, and filters them based on the availability of drugs from the Therapeutic Target Database (TTD). The gray module uses the Integrated Interaction Database (IID) to extract protein–protein interactions of the proteins yielded by the blue module and constructs a network out of them. The green module calculates gene ontology-based semantic similarity scores of the output of the blue module compared with NOX4 using molecular function (MF) annotations, ranks the proteins based on their similarity scores, and excludes proteins with less than one molecular function. The output of the green module is used to annotate the network with the top four proteins.
Fig. 2.
Fig. 2.
Integrated NOX4-extended multilayer network of biomolecular interactions used for candidate extraction and the involved protein semantic similarity ranking. (A) The full network constructed using the primary protein, NOX4 (orange node), connected to its direct metabolic interactors (red nodes), which have been linked to the proteins (blue nodes) interacting with them. We also show all protein–protein and metabolite–protein interactions (gray edges). (B) The semantic similarity ranking based on molecular functions (MF SemSim) of proteins with the top four similar proteins is highlighted. (C) The simplified network with only the primary protein, and the top four similar proteins and metabolites shown individually, while the rest of the proteins are grouped as modules and their interactions are merged.
Fig. 3.
Fig. 3.
In vitro cotarget validation and drug identification of NOX4 and NOS inhibitors as a combinatory treatment. (A) Organotypic hippocampal cultures prepared from hippocampal slices were cultured for 4 d and subsequently subjected to 15 min of OGD period followed by 24-h treatment. Samples for gene expression detection were collected at 0, 2, 4, 8, 12, and 24 h post-OGD. (B) NOX4 expression was up-regulated at 4 and 8 h in comparison with the beginning of the ischemia period (*P < 0.05, ***P < 0.001; n = 3). (C) Inducible NOS (NOS2; square) was up-regulated only in the first 2 h post-OGD, while neuronal NOS (NOS1; circle) was up-regulated in the final 12 to 24 h after the OGD period. Similarly, endothelial NOS (NOS3; triangle) was also significantly up-regulated at 8, 12, and 24 h post-OGD (*P < 0.05, **P < 0.01; n = 4). Gene expression was normalized using β-actin as housekeeping gene. (D) Cell death was significantly reduced in OHCs treated with GKT136901 (0.01 µM) and L-NAME (0.3 µM) in combination (**P < 0.01; n = 8; green slashed bar) in comparison with control slices (#P < 0.05 with respect to basal; n = 8; gray bar). Individual treatments show no effect. (E) ROS formation was also significantly decreased in OHCs treated with the combination of GKT136901 (0.01 µM) and L-NAME (0.3 µM) in comparison with nontreated slices. Again, individual treatments show no effect on cell death. #P < 0.05 compared with basal conditions (gray bar; n = 5); **P < 0.01 with respect to nontreated slices (gray bar; n = 5). (F) Combinatory treatment of GKT136901 and L-NAME increases cell viability in human brain microvascular endothelial cells subjected to hypoxia/reoxygenation (Re-Ox). ##P < 0.01 with respect to basal conditions (n = 4; gray bar); *P < 0.05 with respect to nontreated cells (n = 4; green slashed bar). (G) Cell permeability was assessed by measuring Evans blue fluorescence post-OGD. Evans blue diffusion was significantly reduced in cells treated with GKT136901 (0.01 μM) and L-NAME (0.3 μM) in combination (#P < 0.05; n = 4; gray bar) in comparison with nontreated cells (*P < 0.05; n = 4; green slashed bar). Error bars are mean ± SD.
Fig. 4.
Fig. 4.
In vivo validation of network pharmacology for clinical translation. (A) Twenty-four hours after tMCAO infarct size was reduced in mice treated with GKT136901 (10 mg/kg) and L-NAME (3 mg/kg) in combination 1 h (**P < 0.01; n = 6) and 3 h poststroke (*P < 0.05; n = 5), while individual treatment showed no effect in reduction of infarct size. Infarct volume was also significantly reduced in aged animals treated with the combination (GKT+L-NAME) 1 h poststroke (**P < 0.01; n = 5). Similarly, combinatory treatment decreased infarct volume after permanent occlusion of the MCA in adult mice (*P < 0.05; n = 5). (B) With respect to the neurological outcome of the combinatory treatment in surviving mice, neurological outcome (Bederson score) was improved in the adult mice treated 1 h poststroke (*P < 0.05; n = 9), 3 h poststroke (*P < 0.05; n = 5), and the aged model (*P < 0.05; n = 4). (C) Likewise, the elevated body swing test indicated a significant increase for the right swing number/total swing number ratio in adult mice treated 1 h PO (*P < 0.05; n = 9) but not in the other groups. (D) Significantly improved motor outcome was detected after four-limb hanging test in all groups: 1 h PO (*P < 0.05; n = 9), 3 h PO (*P < 0.05; n = 5), and aged animals (*P < 0.05; n = 4). (E) Blood–brain barrier integrity assessed by Evans blue extravasation was preserved in treated animals compared with nontreated mice at day 1 after 1 h of tMCAO (*P < 0.05; n = 4). (F) Treated mice showed decreased ROS formation compared with their respective nontreated animals (*P < 0.05; n = 4). (G) N-Tyr–positive cells were significantly reduced with the combinatory therapy compared with nontreated mice. (*P < 0.05; n = 4). Error bars are mean ± SD.

References

    1. Yildirim MA, Goh K-I, Cusick ME, Barabási A-L, Vidal M. Drug-target network. Nat Biotechnol. 2007;25:1119–1126.
    1. Aguirre-Plans J, et al. Proximal pathway enrichment analysis for targeting comorbid diseases via network endopharmacology. Pharmaceuticals (Basel) 2018;11:E61.
    1. Hopkins AL. Network pharmacology. Nat Biotechnol. 2007;25:1110–1111.
    1. Scannell JW, Blanckley A, Boldon H, Warrington B. Diagnosing the decline in pharmaceutical R&D efficiency. Nat Rev Drug Discov. 2012;11:191–200.
    1. Menche J, et al. Disease networks. Uncovering disease-disease relationships through the incomplete interactome. Science. 2015;347:1257601.
    1. Langhauser F, et al. A diseasome cluster-based drug repurposing of soluble guanylate cyclase activators from smooth muscle relaxation to direct neuroprotection. NPJ Syst Biol Appl. 2018;4:8.
    1. Barabási A-L, Oltvai ZN. Network biology: Understanding the cell’s functional organization. Nat Rev Genet. 2004;5:101–113.
    1. Batra R, et al. On the performance of de novo pathway enrichment. NPJ Syst Biol Appl. 2017;3:6.
    1. Alcaraz N, et al. De novo pathway-based biomarker identification. Nucleic Acids Res. 2017;45:e151.
    1. Koduru P, Chaganti R. Congenital chromosome breakage clusters within Giemsa-light bands and identifies sites of chromatin instability. Cytogenet Cell Genet. 1988;49:269–274.
    1. Li X, et al. Prediction of synergistic anti-cancer drug combinations based on drug target network and drug induced gene expression profiles. Artif Intell Med. 2017;83:35–43.
    1. Meladze VG. [Temporal characteristics of the autoregulation process of local cerebral blood flow in hypo- and hypertension] Patol Fiziol Eksp Ter. 1985:29–32. Russian.
    1. Vitali F, Mulas F, Marini P, Bellazzi R. Network-based target ranking for polypharmacological therapies. J Biomed Inform. 2013;46:876–881.
    1. Chua HE, Bhowmick SS, Tucker-Kellogg L. Synergistic target combination prediction from curated signaling networks: Machine learning meets systems biology and pharmacology. Methods. 2017;129:60–80.
    1. Vilar S, Hripcsak G. The role of drug profiles as similarity metrics: Applications to repurposing, adverse effects detection and drug-drug interactions. Brief Bioinform. 2017;18:670–681.
    1. Guney E. Reproducible drug repurposing: When similarity does not suffice. Pac Symp Biocomput. 2017;22:132–143.
    1. Casas AI, et al. NOX4-dependent neuronal autotoxicity and BBB breakdown explain the superior sensitivity of the brain to ischemic damage. Proc Natl Acad Sci USA. 2017;114:12315–12320.
    1. Kleinschnitz C, et al. Post-stroke inhibition of induced NADPH oxidase type 4 prevents oxidative stress and neurodegeneration. PLoS Biol. 2010;8:e1000479.
    1. Li F, Xu W, Zhao S. Regulatory roles of metabolites in cell signaling networks. J Genet Genomics. 2013;40:367–374.
    1. Nisimoto Y, Diebold BA, Cosentino-Gomes D, Lambeth JD. Nox4: A hydrogen peroxide-generating oxygen sensor. Biochemistry. 2014;53:5111–5120, and erratum (2014) 53:5472.
    1. Piovesan D, Giollo M, Ferrari C, Tosatto SCE. Protein function prediction using guilty by association from interaction networks. Amino Acids. 2015;47:2583–2592.
    1. Pesquita C, et al. Metrics for GO based protein semantic similarity: A systematic evaluation. BMC Bioinformatics. 2008;9(Suppl 5):S4.
    1. Wang JZ, Du Z, Payattakool R, Yu PS, Chen C-F. A new method to measure the semantic similarity of GO terms. Bioinformatics. 2007;23:1274–1281.
    1. Kleikers PWM, et al. A combined pre-clinical meta-analysis and randomized confirmatory trial approach to improve data validity for therapeutic target validation. Sci Rep. 2015;5:13428.
    1. O’Collins VE, et al. 1,026 experimental treatments in acute stroke. Ann Neurol. 2006;59:467–477.
    1. Peña ID, Borlongan C, Shen G, Davis W. Strategies to extend thrombolytic time window for ischemic stroke treatment: An unmet clinical need. J Stroke. 2017;19:50–60.
    1. Chen R-L, Balami JS, Esiri MM, Chen L-K, Buchan AM. Ischemic stroke in the elderly: An overview of evidence. Nat Rev Neurol. 2010;6:256–265.
    1. Lehner C, et al. Oxidative stress and blood-brain barrier dysfunction under particular consideration of matrix metalloproteinases. Antioxid Redox Signal. 2011;15:1305–1323.
    1. Enciu A-M, Gherghiceanu M, Popescu BO. Triggers and effectors of oxidative stress at blood-brain barrier level: Relevance for brain ageing and neurodegeneration. Oxid Med Cell Longev. 2013;2013:297512.
    1. Zhao X-M, et al. Prediction of drug combinations by integrating molecular and pharmacological data. PLoS Comput Biol. 2011;7:e1002323.
    1. Huang L, et al. DrugComboRanker: Drug combination discovery based on target network analysis. Bioinformatics. 2014;30:i228–i236.
    1. Vidal M, Cusick ME, Barabási A-L. Interactome networks and human disease. Cell. 2011;144:986–998.
    1. Futschik ME, Chaurasia G, Herzel H. Comparison of human protein-protein interaction maps. Bioinformatics. 2007;23:605–611.
    1. Lehne B, Schlitt T. Protein-protein interaction databases: Keeping up with growing interactomes. Hum Genomics. 2009;3:291–297.
    1. Rolland T, et al. A proteome-scale map of the human interactome network. Cell. 2014;159:1212–1226.
    1. Mehta V, Trinkle-Mulcahy L. Recent advances in large-scale protein interactome mapping. F1000 Res. 2016;5:782.
    1. Amaral LAN. A truer measure of our ignorance. Proc Natl Acad Sci USA. 2008;105:6795–6796.
    1. Altenhöfer S, Radermacher KA, Kleikers PWM, Wingler K, Schmidt HHHW. Evolution of NADPH oxidase inhibitors: Selectivity and mechanisms for target engagement. Antioxid Redox Signal. 2015;23:406–427.
    1. Miettinen TP, Björklund M. NQO2 is a reactive oxygen species generating off-target for acetaminophen. Mol Pharm. 2014;11:4395–4404.
    1. Radi R. Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc Natl Acad Sci USA. 2018;115:5839–5848.
    1. Geiszt M. NADPH oxidases: New kids on the block. Cardiovasc Res. 2006;71:289–299.
    1. Espey MG, et al. A chemical perspective on the interplay between NO, reactive oxygen species, and reactive nitrogen oxide species. Ann N Y Acad Sci. 2002;962:195–206.
    1. Ghezzi P, Jaquet V, Marcucci F, Schmidt HHHW. The oxidative stress theory of disease: Levels of evidence and epistemological aspects. Br J Pharmacol. 2017;174:1784–1796.
    1. Kleinschnitz C, et al. NOS knockout or inhibition but not disrupting PSD-95-NOS interaction protect against ischemic brain damage. J Cereb Blood Flow Metab. 2016;36:1508–1512.
    1. Endres M, Laufs U, Liao JK, Moskowitz MA. Targeting eNOS for stroke protection. Trends Neurosci. 2004;27:283–289.
    1. Niwa M, et al. Time course of expression of three nitric oxide synthase isoforms after transient middle cerebral artery occlusion in rats. Neurol Med Chir (Tokyo) 2001;41:63–73.
    1. Dao VT-V, et al. Pharmacology and clinical drug candidates in redox medicine. Antioxid Redox Signal. 2015;23:1113–1129.
    1. Sun Q-A, Hess DT, Wang B, Miyagi M, Stamler JS. Off-target thiol alkylation by the NADPH oxidase inhibitor 3-benzyl-7-(2-benzoxazolyl)thio-1,2,3-triazolo[4,5-d]pyrimidine (VAS2870) Free Radic Biol Med. 2012;52:1897–1902.
    1. Ott C, et al. Effects of the nitric oxide synthase inhibitor ronopterin (VAS203) on renal function in healthy volunteers. Br J Clin Pharmacol. January 21, 2019 doi: 10.1111/bcp.13870.
    1. Mistry RK, et al. Transcriptional regulation of cystathionine-γ-lyase in endothelial cells by NADPH oxidase 4-dependent signaling. J Biol Chem. 2016;291:1774–1788.
    1. Chan SJ, et al. Cystathionine β-synthase inhibition is a potential therapeutic approach to treatment of ischemic injury. ASN Neuro. 2015;7:1759091415578711.
    1. Noronha-Dutra AA, Epperlein MM, Woolf N. Reaction of nitric oxide with hydrogen peroxide to produce potentially cytotoxic singlet oxygen as a model for nitric oxide-mediated killing. FEBS Lett. 1993;321:59–62.
    1. Hopkins AL. Network pharmacology: The next paradigm in drug discovery. Nat Chem Biol. 2008;4:682–690.
    1. Tang J, Aittokallio T. Network pharmacology strategies toward multi-target anticancer therapies: From computational models to experimental design principles. Curr Pharm Des. 2014;20:23–36.

Source: PubMed

3
Iratkozz fel