Nuclear receptors (NRs) are important biological macromolecular transcription factors that are implicated in multiple biological pathways and may interact with other xenobiotics that are endocrine disruptors present in the environment. Examples of important NRs include the androgen receptor (AR), estrogen receptors (ER), and the pregnane X receptor (PXR). In this study we have utilized the Ligand Activity by Surface Similarity Order (LASSO) method, a ligand-based virtual screening strategy to derive structural (surface/shape) molecular features used to generate predictive models of biomolecular activity for AR, ER, and PXR. For PXR, twenty-five models were built using between 8 to 128 agonists and tested using 3000, 8000, and 24,000 drug-like decoys including PXR inactive compounds . Preliminary studies with AR and ER using LASSO suggested the utility of this approach with 2-fold enrichment factors at 20%. We found that models with 64–128 PXR actives provided enrichment factors of 10-fold (10% actives in the top 1% of compounds screened). The LASSO models for AR and ER have been deployed and are freely available online, and they represent a ligand-based prediction method for putative NR activity of compounds in this database. 1. Introduction The nuclear receptor (NRs) family of transcription factors are important targets for therapeutic interventions for multiple diseases [1] and also may interact with other xenobiotics that are endocrine disruptors present in the environment [2]. It is therefore important to identify compounds that may specifically bind NRs and act as endocrine disruptors and develop synthetic compounds that can selectively (in a cell-type and/or tissue-selective manner) modulate NR pharmacology (reviewed in [3–9]). NRs including the androgen receptor (AR; NR3C4), estrogen receptors α and β, (ERα and ERβ; NR3A1 and NR3A2) and pregnane X receptor (PXR; NR1I2) are particularly important as both therapeutic targets and for xenobiotics to mediate off-target effects. The ERs are activated by 17β-estradiol while the AR is activated by testosterone and dihydrotestosterone and these receptors are transcriptional regulators of many genes [10] with important physiological functions [11–16]. The human PXR [17–19] similarly transcriptionally regulates genes involved in xenobiotic metabolism and excretion, as well as other cellular processes, including apoptosis [20–24]. Human PXR is a broad specificity NR, binding a wide variety of molecules [25] and the activation of this NR can cause drug-drug interactions [23]. Multiple QSAR and machine
References
[1]
A. G. Smith and G. E. O. Muscat, “Skeletal muscle and nuclear hormone receptors: implications for cardiovascular and metabolic disease,” International Journal of Biochemistry and Cell Biology, vol. 37, no. 10, pp. 2047–2063, 2005.
[2]
A. K. Hotchkiss, C. V. Rider, C. R. Blystone et al., “Fifteen years after “wingspread”—environmental endocrine disrupters and human and wildlife health: where we are today and where we need to go,” Toxicological Sciences, vol. 105, no. 2, pp. 235–259, 2008.
[3]
S. Tenbaum and A. Baniahmad, “Nuclear receptors: structure, function and involvement in disease,” International Journal of Biochemistry and Cell Biology, vol. 29, no. 12, pp. 1325–1341, 1997.
[4]
A. Zimber and C. Gespach, “Bile acids and derivatives, their nuclear receptors FXR, PXR and ligands: role in health and disease and their therapeutic potential,” Anti-Cancer Agents in Medicinal Chemistry, vol. 8, no. 5, pp. 540–563, 2008.
[5]
P. L. Feldman, M. H. Lambert, and B. R. Henke, “PPAR modulators and PPAR pan agonists for metabolic diseases: the next generation of drugs targeting peroxisome proliferator-activated receptors?” Current Topics in Medicinal Chemistry, vol. 8, no. 9, pp. 728–749, 2008.
[6]
X. Ma, J. R. Idle, and F. J. Gonzalez, “The pregnane X receptor: from bench to bedside,” Expert Opinion on Drug Metabolism and Toxicology, vol. 4, no. 7, pp. 895–908, 2008.
[7]
M. Bertolotti, C. Gabbi, C. Anzivino, L. Carulli, P. Loria, and N. Carulli, “Nuclear receptors as potential molecular targets in cholesterol accumulation conditions: insights from evidence on hepatic cholesterol degradation and gallstone disease in humans,” Current Medicinal Chemistry, vol. 15, no. 22, pp. 2271–2284, 2008.
[8]
E. E. Baulieu, M. Atger, and M. Best Belpomme, “Steroid hormone receptors,” Vitamins and Hormones, vol. 33, pp. 649–736, 1975.
[9]
D. V. Henley and K. S. Korach, “Endocrine-disrupting chemicals use distinct mechanisms of action to modulate endocrine system function,” Endocrinology, vol. 147, no. 6, pp. S25–S32, 2006.
[10]
M. D. Krasowski, E. J. Reschly, and S. Ekins, “Intrinsic disorder in nuclear hormone receptors,” Journal of Proteome Research, vol. 7, no. 10, pp. 4359–4372, 2008.
[11]
A. C. W. Pike, A. M. Brzozowski, R. E. Hubbard et al., “Structure of the ligand-binding domain of oestrogen receptor beta in the presence of a partial agonist and a full antagonist,” EMBO Journal, vol. 18, no. 17, pp. 4608–4618, 1999.
[12]
A. M. Brzozowski, A. C. W. Pike, Z. Dauter et al., “Molecular basis of agonism and antagonism in the oestrogen receptor,” Nature, vol. 389, no. 6652, pp. 753–758, 1997.
[13]
J. W. R. Schwabe, L. Chapman, J. T. Finch, D. Rhodes, and D. Neuhaus, “DNA recognition by the oestrogen receptor: from solution to the crystal,” Structure, vol. 1, no. 3, pp. 187–204, 1993.
[14]
J. S. Sack, K. F. Kish, C. Wang et al., “Crystallographic structures of the ligand-binding domains of the androgen receptor and its T877A mutant complexed with the natural agonist dihydrotestosterone,” Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 9, pp. 4904–4909, 2001.
[15]
C. E. Bohl, C. Chang, M. L. Mohler et al., “A ligand-based approach to identify quantitative structure-activity relationships for the androgen receptor,” Journal of Medicinal Chemistry, vol. 47, no. 15, pp. 3765–3776, 2004.
[16]
P. L. Shaffer, A. Jivan, D. E. Dollins, F. Claessens, and D. T. Gewirth, “Structural basis of androgen receptor binding to selective androgen response elements,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 14, pp. 4758–4763, 2004.
[17]
G. Bertilsson, J. Heidrich, K. Svensson et al., “Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 21, pp. 12208–12213, 1998.
[18]
B. Blumberg, W. Sabbagh Jr., H. Juguilon et al., “SXR, a novel steroid and xenobiotic-sensing nuclear receptor,” Genes and Development, vol. 12, no. 20, pp. 3195–3205, 1998.
[19]
S. A. Kliewer, J. T. Moore, L. Wade et al., “An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway,” Cell, vol. 92, no. 1, pp. 73–82, 1998.
[20]
S. Verma, M. M. Tabb, and B. Blumberg, “Activation of the steroid and xenobiotic receptor, SXR, induces apoptosis in breast cancer cells,” BMC Cancer, vol. 9, article 3, 2009.
[21]
D. Gupta, M. Venkatesh, H. Wang et al., “Expanding the roles for pregnane X receptor in cancer: proliferation and drug resistance in ovarian cancer,” Clinical Cancer Research, vol. 14, no. 17, pp. 5332–5340, 2008.
[22]
J. Zhou, M. Liu, Y. Zhai, and W. Xie, “The antiapoptotic role of pregnane X receptor in human colon cancer cells,” Molecular Endocrinology, vol. 22, no. 4, pp. 868–880, 2008.
[23]
A. Biswas, S. Mani, M. R. Redinbo, et al., “Elucidating the ‘Jekyll and Hyde’ nature of PXR: the case for discovering antagonists,” Pharmaceutical Research, vol. 26, no. 8, pp. 1807–1815, 2009.
[24]
B. L. Urquhart, R. G. Tirona, and R. B. Kim, “Nuclear receptors and the regulation of drug-metabolizing enzymes and drug transporters: implications for interindividual variability in response to drugs,” Journal of Clinical Pharmacology, vol. 47, no. 5, pp. 566–578, 2007.
[25]
S. Ekins, S. Kortagere, M. Iyer et al., “Challenges predicting ligand-receptor interactions of promiscuous proteins: the nuclear receptor PXR,” PLoS Computational Biology, vol. 5, no. 12, Article ID e1000594, 2009.
[26]
N. Ai, R. K. DeLisle, S. J. Yu, and W. J. Welsh, “Computational models for predicting the binding affinities of ligands for the wild-type androgen receptor and a mutated variant associated with human prostate cancer,” Chemical Research in Toxicology, vol. 16, no. 12, pp. 1652–1660, 2003.
[27]
M. N. Jacobs, “In silico tools to aid risk assessment of endocrine disrupting chemicals,” Toxicology, vol. 205, no. 1-2, pp. 43–53, 2004.
[28]
W. Tong, R. Perkins, L. Xing, W. J. Welsh, and D. M. Sheehan, “QSAR models for binding of estrogenic compounds to estrogen receptor α and β subtypes,” Endocrinology, vol. 138, no. 9, pp. 4022–4025, 1997.
[29]
S. Ekins, L. Mirny, and E. G. Schuetz, “A ligand-based approach to understanding selectivity of nuclear hormone receptors PXR, CAR, FXR, LXRα, and LXRβ,” Pharmaceutical Research, vol. 19, no. 12, pp. 1788–1800, 2002.
[30]
T. Z. Bao, G.-Z. Han, J.-Y. Shim, Y. Wen, and X.-R. Jiang, “Quantitative structure-activity relationship of various endogenous estrogen metabolites for human estrogen receptor α and β subtypes: insights into the structural determinants favoring a differential subtype binding,” Endocrinology, vol. 147, no. 9, pp. 4132–4150, 2006.
[31]
D. Plewczynski, M. Von Grotthuss, S. A. H. Spieser et al., “Target specific compound identification using a support vector machine,” Combinatorial Chemistry and High Throughput Screening, vol. 10, no. 3, pp. 189–196, 2007.
[32]
L. Zhao and R. D. Brinton, “Structure-based virtual screening for plant-based EBβ-selective ligands as potential preventative therapy against age-related neurodegenerative diseases,” Journal of Medicinal Chemistry, vol. 48, no. 10, pp. 3463–3466, 2005.
[33]
W. H. Bisson, A. V. Cheltsov, N. Bruey-Sedano et al., “Discovery of antiandrogen activity of nonsteroidal scaffolds of marketed drugs,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 29, pp. 11927–11932, 2007.
[34]
S. Laws, et al., DSSTox EPA Estrogen Receptor Ki Binding Study (Laws et al.) Database—(KIERBL): SDF file and documentation. 2009 http://www.epa.gov/ncct/dsstox/sdf_kierbl.html.
[35]
S. C. Laws, S. Yavanhxay, R. L. Cooper, and J. C. Eldridge, “Nature of the binding interaction for 50 structurally diverse chemicals with rat estrogen receptors,” Toxicological Sciences, vol. 94, no. 1, pp. 46–56, 2006.
[36]
J. R. Rabinowitz, S. B. Little, S. C. Laws, and M.-R. Goldsmith, “Molecular modeling for screening environmental chemicals for estrogenicity: use of the toxicant-target approach,” Chemical Research in Toxicology, vol. 22, no. 9, pp. 1594–1602, 2009.
[37]
H. Fang, W. Tong, W. S. Branham, et al., “Study of 202 natural, synthetic, and environmental chemicals for binding to the androgen receptor,” Chemical Research in Toxicology, vol. 16, no. 10, pp. 1338–1358, 2003.
[38]
D. Ding, L. Xu, H. Fang et al., “The EDKB: an established knowledge base for endocrine disrupting chemicals,” BMC Bioinformatics, vol. 11, no. 6, article 5, 2010.
[39]
R. E. Watkins, P. R. Davis-Searles, M. H. Lambert, and M. R. Redinbo, “Coactivator binding promotes the specific interaction between ligand and the pregnane X receptor,” Journal of Molecular Biology, vol. 331, no. 4, pp. 815–828, 2003.
[40]
R. E. Watkins, J. M. Maglich, L. B. Moore et al., “2.1 ? crystal structure of human PXR in complex with the St. John's wort compound hyperforin,” Biochemistry, vol. 42, no. 6, pp. 1430–1438, 2003.
[41]
Y. Xue, E. Chao, W. J. Zuercher, T. M. Willson, J. L. Collins, and M. R. Redinbo, “Crystal structure of the PXR-T1317 complex provides a scaffold to examine the potential for receptor antagonism,” Bioorganic and Medicinal Chemistry, vol. 15, no. 5, pp. 2156–2166, 2007.
[42]
J. E. Chrencik, J. Orans, L. B. Moore et al., “Structural disorder in the complex of human pregnane X receptor and the macrolide antibiotic rifampicin,” Molecular Endocrinology, vol. 19, no. 5, pp. 1125–1134, 2005.
[43]
D. G. Teotico, J. J. Bischof, L. Peng, S. A. Kliewer, and M. R. Redinbo, “Structural basis of human pregnane X receptor activation by the hops constituent colupulone,” Molecular Pharmacology, vol. 74, no. 6, pp. 1512–1520, 2008.
[44]
R. E. Watkins, G. B. Wisely, L. B. Moore et al., “The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity,” Science, vol. 292, no. 5525, pp. 2329–2333, 2001.
[45]
Y. Xue, L. B. Moore, J. Orans et al., “Crystal structure of the pregnane X receptor-estradiol complex provides insights into endobiotic recognition,” Molecular Endocrinology, vol. 21, no. 5, pp. 1028–1038, 2007.
[46]
K. Bachmann, H. Patel, Z. Batayneh et al., “PXR and the regulation of apoA1 and HDL-cholesterol in rodents,” Pharmacological Research, vol. 50, no. 3, pp. 237–246, 2004.
[47]
S. Ekins, C. Chang, S. Mani et al., “Human pregnane X receptor antagonists and agonists define molecular requirements for different binding sites,” Molecular Pharmacology, vol. 72, no. 3, pp. 592–603, 2007.
[48]
S. Ekins and J. A. Erickson, “A pharmacophore for human pregnane X receptor ligands,” Drug Metabolism and Disposition, vol. 30, no. 1, pp. 96–99, 2002.
[49]
D. Schuster and T. Langer, “The identification of ligand features essential for PXR activation by pharmacophore modeling,” Journal of Chemical Information and Modeling, vol. 45, no. 2, pp. 431–439, 2005.
[50]
K. Yasuda, A. Ranade, R. Venkataramanan et al., “A comprehensive in vitro and in silico analysis of antibiotics that activate pregnane X receptor and induce CYP3A4 in liver and intestine,” Drug Metabolism and Disposition, vol. 36, no. 8, pp. 1689–1697, 2008.
[51]
A. Khandelwal, M. D. Krasowski, E. J. Reschly, M. W. Sinz, P. W. Swaan, and S. Ekins, “Machine learning methods and docking for predicting human pregnane X receptor activation,” Chemical Research in Toxicology, vol. 21, no. 7, pp. 1457–1467, 2008.
[52]
S. Kortagere, D. Chekmarev, W. J. Welsh, and S. Ekins, “Hybrid scoring and classification approaches to predict human pregnane X receptor activators,” Pharmaceutical Research, vol. 26, no. 4, pp. 1001–1011, 2009.
[53]
S. Ekins, E. J. Reschly, L. R. Hagey, and M. D. Krasowski, “Evolution of pharmacologic specificity in the pregnane X receptor,” BMC Evolutionary Biology, vol. 8, no. 1, article 103, 2008.
[54]
S. Kortagere, D. Chekmarev, W. J. Welsh, and S. Ekins, “Hybrid scoring and classification approaches to predict human pregnane X receptor activators,” Pharmaceutical Research, vol. 26, no. 4, pp. 1001–1011, 2009.
[55]
S. Kortagere, M. D. Krasowski, E. J. Reschly, M. Venkatesh, S. Mani, and S. Ekins, “Evaluation of computational docking to identify pregnane X receptor agonists in the toxcast database,” Environmental Health Perspectives, vol. 118, no. 10, pp. 1412–1417, 2010.
[56]
A. Khandelwal, M. D. Krasowski, E. J. Reschly, M. W. Sinz, P. W. Swaan, and S. Ekins, “Machine learning methods and docking for predicting human pregnane X receptor activation,” Chemical Research in Toxicology, vol. 21, no. 7, pp. 1457–1467, 2008.
[57]
S. Kortagere and S. Ekins, “Troubleshooting computational methods in drug discovery,” Journal of Pharmacological and Toxicological Methods, vol. 61, no. 2, pp. 67–75, 2010.
[58]
N. Ai, M. D. Krasowski, W. J. Welsh, and S. Ekins, “Understanding nuclear receptors using computational methods,” Drug Discovery Today, vol. 14, no. 9-10, pp. 486–494, 2009.
[59]
D. Reid, B. S. Sadjad, Z. Zsoldos, and A. Simon, “LASSO—ligand activity by surface similarity order: a new tool for ligand based virtual screening,” Journal of Computer-Aided Molecular Design, vol. 22, no. 6-7, pp. 479–487, 2008.
[60]
N. Huang, B. K. Shoichet, and J. J. Irwin, “Benchmarking sets for molecular docking,” Journal of Medicinal Chemistry, vol. 49, no. 23, pp. 6789–6801, 2006.
[61]
C. Y. Ung, H. Li, C. W. Yap, and Y. Z. Chen, “In silico prediction of pregnane X receptor activators by machine learning approaches,” Molecular Pharmacology, vol. 71, no. 1, pp. 158–168, 2007.
[62]
D. Weininger, “SMILES, a chemical language and information system. 1. Introduction to methodology and encoding rules,” Journal of Chemical Information and Computer Sciences, vol. 28, pp. 31–36, 1988.
[63]
G. B. McGaughey, R. P. Sheridan, C. I. Bayly et al., “Comparison of topological, shape, and docking methods in virtual screening,” Journal of Chemical Information and Modeling, vol. 47, no. 4, pp. 1504–1519, 2007.
[64]
L. R. Murthy, M. P. Johnson, and D. R. Rowley, “Characterization of steroid receptors in human prostate using mibolerone,” Prostate, vol. 8, no. 3, pp. 241–253, 1986.
[65]
A. M. Voutchkova, T. G. Osimitz, and P. T. Anastas, “Toward a comprehensive molecular design framework for reduced hazard,” Chemical Reviews, vol. 110, no. 10, pp. 5845–5882, 2010.