Screening a small molecule library to identify inhibitors of NF-κB inducing kinase and pro-labor genes in human placenta

Bingbing Wang, Nataliya Parobchak, Adriana Martin, Max Rosen, Lumeng Jenny Yu, Mary Nguyen, Kseniya Gololobova, Todd Rosen, Bingbing Wang, Nataliya Parobchak, Adriana Martin, Max Rosen, Lumeng Jenny Yu, Mary Nguyen, Kseniya Gololobova, Todd Rosen

Abstract

The non-canonical NF-κB signaling (RelB/p52) pathway drives pro-labor genes in the human placenta, including corticotropin-releasing hormone (CRH) and cyclooxygenase-2 (COX-2), making this a potential therapeutic target to delay onset of labor. Here we sought to identify small molecule compounds from a pre-existing chemical library of orally active drugs that can inhibit this NF-κB signaling, and in turn, human placental CRH and COX-2 production. We used a cell-based assay coupled with a dual-luciferase reporter system to perform an in vitro screening of a small molecule library of 1,120 compounds for inhibition of the non-canonical NF-κB pathway. Cell toxicity studies and drug efflux transport MRP1 assays were used to further characterize the lead compounds. We have found that 14 drugs have selective inhibitory activity against lymphotoxin beta complex-induced activation of RelB/p52 in HEK293T cells, several of which also inhibited expression of CRH and COX-2 in human term trophoblast. We identified sulfapyridine and propranolol with activity against CRH and COX-2 that deserve further study. These drugs could serve as the basis for development of orally active drugs to affect length of gestation, first in an animal model, and then in clinical trials to prevent preterm birth during human pregnancy.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Schematic presentation of strategy of drug screening. (A) Initial screening. HEK293T cells were transiently transfected with a mixture of firefly luciferase (FL) reporter and renilla luciferase (RL) reporter vectors. 48 hours later, the transfectants were exposed to individual compound (drug) for 2 hours, and in turn, lymph toxin- a1b2 (LT-α1β2) for 4 hours prior to dual-luciferase assay. Each chemical compound was run in triplicates and the averages of FL and RL in triplicates were obtained, respectively. With normalization to RL, the individual compound with ≥90% of FL inhibitory effect compared with the vehicle control was considered as a hit for inhibition of the non-canonical NF-κB signaling activity. (B) Secondary screening. HEK293T cells were transiently transfected with a mixture of vectors as described in (A). 48 hours later, the transfectants were exposed to individual hits from the initial screening, and in turn, TPA for 4 hours prior to dual-luciferase assay. Each chemical compound was run in triplicates and the averages of FL and RL in triplicates were obtained, respectively. With normalization to RL, the individual compound with ≤10% of FL inhibitory effect compared with the vehicle control was considered as the specific inhibitor for the non-canonical NF-κB signaling activity. (C) Screening of kinase inhibitors. HEK293T cells were transiently transfected with a mammalian expression vector for ectopic expression of an individual kinase (K), which was followed by a mixture of vectors as described in (A). Then the transfectants were treated with LT-α1β2 (for NIK and IKKα) or TPA (for IKKβ). Each experiment was run in triplicates. With normalization to RL activity, the individual compound with de-repressed FL activity compared with the empty vector control (p < 0.05) was considered as a candidate for inhibition of this kinase. (D) Representative gel pattern and analysis were obtained from 3 independent term placentas by Western blotting with abbreviations described in Table 1. The cropped gels from different Western blot analysis were divided by white space with full-length blots shown in Supplementary Fig. 1. Western blot results were quantified with use of ImageJ. The number below each lane representing relative protein levels of CRH or COX-2 treated with the individual drug relative to DMSO with normalization to β-actin was derived from the following formula: (peak area of CRH or COX-2 of each drug)/(peak area of β-actin of the same lane) of/(peak area of CRH or COX-2 of DMSO)/(peak area of β-actin of the same lane) × 100%. The relative abundance of CRH or COX-2 in the lane of DMSO was considered 100%.
Figure 2
Figure 2
Assays of efflux transporter MRP1 and cell viability. (A) This ATPase assay was performed in the basic way as detailed in Materials and Methods. The bars indicate the average of OD (595 nm) against each individual hit with error bars representing the standard deviation from experiments performed in 3 independent term placentas. *p < 0.01. (B) Primary term cytotrophoblast were treated with the individual compound and cell viability was measured with use of the CellTiter-Blue cell viability assay kit (Promega). The bars indicate the average ration of OD 560/590 (nm) against each individual hit with error bars representing the standard deviation from experiments performed in 3 independent term placentas. *p < 0.01.
Figure 3
Figure 3
Co-localization of NIK and sulfapyridine or propranolol in HEK293 cell. (A) HEK293 cells were treated with Alexa Fluor 647 cadaverine (AlexaFluor-647) or Alexa Fluor 647 cadaverine-labeled sulfapyridine (AlexaFluor-647-Sul) for 24 hr, followed by IF staining. (B) HEK293 cells were treated with AlexaFluor-647 or Alexa Fluor 647 cadaverine-labeled propranolol (AlexaFluor-647-Pro) for 24 hr, followed by IF staining. Each experiment was repeated three times. Arrows indicate co-localization.
Figure 4
Figure 4
Dose-dependent effects of sulfapyridine or propranolol on COX-2 and CRH in primary cytotrophoblast. Human primary cytotrophoblast were treated with sulfapyridine (A) or propranolol (B) at concentrations as indicated for 24 hrs. The total cell lysates were subjected to Western blot analysis (N = 3 individual experiments). The full-length blots are shown in Supplementary Fig. 3. Western blot results were quantified with use of ImageJ (right panels). The relative protein levels of CRH or COX-2 treated with each drug at different concentrations relative to non-treatment (0) with normalization to β-actin were derived from the following formula: (peak area of CRH or COX-2 of each concentration)/(peak area of β-actin of the same lane) of/(peak area of CRH or COX-2 of non-treatment)/(peak area of β-actin of the same lane) × 100%. The relative abundance of CRH or COX-2 in the lane of non-treatment was considered 100%. *p < 0.01, **p < 0.05.

References

    1. Matthews TJ, MacDorman MF. Infant mortality statistics from the 2010 period linked birth/infant death data set. National vital statistics reports: from the Centers for Disease Control and Prevention, National Center for Health Statistics, National Vital Statistics System. 2013;62:1–26.
    1. Behrman, R. E., Butler, A. S. & Institute of Medicine (U.S.). Committee on Understanding Premature Birth and Assuring Healthy Outcomes. Preterm birth: causes, consequences, and prevention. (National Academies Press, 2007).
    1. Petraglia, F., Imperatore, A. & Challis, J. R. Neuroendocrine mechanisms in pregnancy and parturition. Endocr Rev31, 783–816, doi:er.2009-0019 (2010).
    1. McLean M, et al. A placental clock controlling the length of human pregnancy. Nat Med. 1995;1:460–463. doi: 10.1038/nm0595-460.
    1. Wadhwa, P. D., Porto, M., Garite, T. J., Chicz-DeMet, A. & Sandman, C. A. Maternal corticotropin-releasing hormone levels in the early third trimester predict length of gestation in human pregnancy. Am J Obstet Gynecol179, 1079–1085, S0002937898702194 (1998).
    1. Hobel, C. J., Dunkel-Schetter, C., Roesch, S. C., Castro, L. C. & Arora, C. P. Maternal plasma corticotropin-releasing hormone associated with stress at 20 weeks’ gestation in pregnancies ending in preterm delivery. Am J Obstet Gynecol180, S257–263, doi:S0002-9378(99)70712-X (1999).
    1. Warren, W. B., Patrick, S. L. & Goland, R. S. Elevated maternal plasma corticotropin-releasing hormone levels in pregnancies complicated by preterm labor. Am J Obstet Gynecol166, 1198–1204; discussion 1204–1197 (1992).
    1. Simhan, H. N. & Caritis, S. N. Prevention of preterm delivery. N Engl J Med357, 477–487, 357/5/477 (2007).
    1. Challis JR, et al. Prostaglandins and mechanisms of preterm birth. Reproduction. 2002;124:1–17. doi: 10.1530/rep.0.1240001.
    1. Wang, B. et al. Glucocorticoid Receptor Signaling Contributes to Constitutive Activation of the Noncanonical NF-kappaB Pathway in Term Human Placenta. Mol Endocrinol27, 203–211, me.2012–1309 (2013).
    1. Wang, B., Parobchak, N., Rosen, M., Roche, N. & Rosen, T. Negative Effects of Progesterone Receptor Isoform-A on Human Placental Activity of the Noncanonical NF-kappaB Signaling. J Clin Endocrinol Metab99, E320–328, doi:jc.2013-2721 (2014).
    1. Wang, B., Parobchak, N. & Rosen, T. RelB/NF-kappaB2 Regulates Corticotropin-Releasing Hormone in the Human Placenta. Mol Endocrinol26, 1356–1369, doi:me.2012-1035 (2012).
    1. Di Stefano, V., Wang, B., Parobchak, N., Roche, N. & Rosen, T. RelB/p52-mediated NF-kappaB signaling alters histone acetylation to increase the abundance of corticotropin-releasing hormone in human placenta. Sci Signal8, ra85, 10.1126/scisignal.aaa9806 (2015).
    1. Sun, S. C. Non-canonical NF-kappaB signaling pathway. Cell Res21, 71–85, doi:cr2010177 (2011).
    1. Dejardin, E. The alternative NF-kappaB pathway from biochemistry to biology: pitfalls and promises for future drug development. Biochem Pharmacol72, 1161–1179, doi:S0006-2952(06)00500-4 (2006).
    1. Bhakar AL, et al. Constitutive nuclear factor-kappa B activity is required for central neuron survival. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2002;22:8466–8475.
    1. Lai, T. S. et al. Identification of chemical inhibitors to human tissue transglutaminase by screening existing drug libraries. Chem Biol15, 969–978, doi:S1074-5521(08)00285-8 (2008).
    1. Mordmuller B, Krappmann D, Esen M, Wegener E, Scheidereit C. Lymphotoxin and lipopolysaccharide induce NF-kappaB-p52 generation by a co-translational mechanism. EMBO Rep. 2003;4:82–87. doi: 10.1038/sj.embor.embor710.
    1. Kuai J, et al. Endogenous association of TRAF2, TRAF3, cIAP1, and Smac with lymphotoxin beta receptor reveals a novel mechanism of apoptosis. J Biol Chem. 2003;278:14363–14369. doi: 10.1074/jbc.M208672200.
    1. Kang, M. I. et al. A selective small-molecule nuclear factor-kappaB inhibitor from a high-throughput cell-based assay for “activator protein-1 hits”. Mol Cancer Ther8, 571–581, doi:1535-7163.MCT-08-0811 (2009).
    1. Kobori M, et al. Wedelolactone suppresses LPS-induced caspase-11 expression by directly inhibiting the IKK complex. Cell Death Differ. 2004;11:123–130. doi: 10.1038/sj.cdd.4401325.
    1. Ruegg UT, Burgess GM. Staurosporine, K-252 and UCN-01: potent but nonspecific inhibitors of protein kinases. Trends Pharmacol Sci. 1989;10:218–220. doi: 10.1016/0165-6147(89)90263-0.
    1. Nagashige, M. et al. Basal membrane localization of MRP1 in human placental trophoblast. Placenta24, 951–958, doi:S014340040300170X (2003).
    1. Keppler D, Leier I, Jedlitschky G. Transport of glutathione conjugates and glucuronides by the multidrug resistance proteins MRP1 and MRP2. Biol Chem. 1997;378:787–791.
    1. Cole SP. Targeting multidrug resistance protein 1 (MRP1, ABCC1): past, present, and future. Annu Rev Pharmacol Toxicol. 2014;54:95–117. doi: 10.1146/annurev-pharmtox-011613-135959.
    1. Mortier, J. et al. NF-kappaB inducing kinase (NIK) inhibitors: identification of new scaffolds using virtual screening. Bioorg Med Chem Lett20, 4515–4520, doi:S0960-894X(10)00801-2 (2010).
    1. Yu LJ, Wang B, Parobchak N, Roche N, Rosen T. STAT3 cooperates with the non-canonical NF-kappaB signaling to regulate pro-labor genes in the human placenta. Placenta. 2015;36:581–586. doi: 10.1016/j.placenta.2015.02.013.
    1. Mogadam M, Dobbins WO, Korelitz BI, Ahmed SW. Pregnancy in Inflammatory Bowel-Disease - Effect of Sulfasalazine and Corticosteroids on Fetal-Outcome. Gastroenterology. 1981;80:72–76.
    1. Wahl C, Liptay S, Adler G, Schmid RM. Sulfasalazine: a potent and specific inhibitor of nuclear factor kappa B. The Journal of clinical investigation. 1998;101:1163–1174. doi: 10.1172/JCI992.
    1. Norgard B, Pedersen L, Christensen LA, Sorensen HT. Therapeutic drug use in women with Crohn’s disease and birth outcomes: a Danish nationwide cohort study. Am J Gastroenterol. 2007;102:1406–1413. doi: 10.1111/j.1572-0241.2007.01216.x.
    1. Pruyn SC, Phelan JP, Buchanan GC. Long-term propranolol therapy in pregnancy: maternal and fetal outcome. American journal of obstetrics and gynecology. 1979;135:485–489. doi: 10.1016/0002-9378(79)90436-8.
    1. Gilboa SM, et al. Use of antihistamine medications during early pregnancy and isolated major malformations. Birth defects research. Part A, Clinical and molecular teratology. 2009;85:137–150. doi: 10.1002/bdra.20513.
    1. Canadian Preterm Labor Investigators, G Treatment of preterm labor with the beta-adrenergic agonist ritodrine. N Engl J Med. 1992;327:308–312. doi: 10.1056/NEJM199207303270503.
    1. Haas DM, Caldwell DM, Kirkpatrick P, McIntosh JJ, Welton NJ. Tocolytic therapy for preterm delivery: systematic review and network meta-analysis. BMJ. 2012;345:e6226. doi: 10.1136/bmj.e6226.
    1. Foster PM, Harris MW. Changes in androgen-mediated reproductive development in male rat offspring following exposure to a single oral dose of flutamide at different gestational ages. Toxicological sciences: an official journal of the Society of Toxicology. 2005;85:1024–1032. doi: 10.1093/toxsci/kfi159.
    1. Feinshtein V, et al. Nitrofurantoin transport by placental choriocarcinoma JAr cells: involvement of BCRP, OATP2B1 and other MDR transporters. Archives of gynecology and obstetrics. 2010;281:1037–1044. doi: 10.1007/s00404-009-1286-7.
    1. Memon, N. et al. Regional expression of the BCRP/ABCG2 transporter in term human placentas. Reprod Toxicol43, 72–77, S0890-6238(13)00378-X (2014).
    1. Annunziata, C. M. et al. Frequent engagement of the classical and alternative NF-kappaB pathways by diverse genetic abnormalities in multiple myeloma. Cancer Cell12, 115–130, doi:S1535-6108(07)00203-6 (2007).
    1. Keats JJ, et al. Promiscuous mutations activate the noncanonical NF-kappaB pathway in multiple myeloma. Cancer cell. 2007;12:131–144. doi: 10.1016/j.ccr.2007.07.003.
    1. Chang PY, et al. Propranolol Reduces Cancer Risk: A Population-Based Cohort Study. Medicine (Baltimore) 2015;94:e1097. doi: 10.1097/MD.0000000000001097.
    1. Assimes TL, Elstein E, Langleben A, Suissa S. Long-term use of antihypertensive drugs and risk of cancer. Pharmacoepidemiol Drug Saf. 2008;17:1039–1049. doi: 10.1002/pds.1656.
    1. Monami M, et al. Further data on beta-blockers and cancer risk: observational study and meta-analysis of randomized clinical trials. Curr Med Res Opin. 2013;29:369–378. doi: 10.1185/03007995.2013.772505.
    1. Tang Z, et al. Isolation of hofbauer cells from human term placentas with high yield and purity. Am J Reprod Immunol. 2011;66:336–348. doi: 10.1111/j.1600-0897.2011.01006.x.
    1. Berman HM, et al. The Protein Data Bank. Nucleic acids research. 2000;28:235–242. doi: 10.1093/nar/28.1.235.
    1. Halgren TAB. form, scope, parameterizaiton and performance of MMFF94. J. Comput Chem. 1996;17:490–519. doi: 10.1002/(SICI)1096-987X(199604)17:5/6<490::AID-JCC1>;2-P.
    1. Halgren TA. Merck molecular force field. II. MMFF94 van der Waals and electrostatic parameters for intermolecular interactions J. Comput Chem. 1996;17:520–552.

Source: PubMed

3
Tilaa