SUMMARY
The human nuclear receptor (NR) family of transcription factors contains 48 proteins that bind lipophilic molecules. Approved NR therapies have had immense success treating various diseases, but lack of selectivity has hindered efforts to therapeutically target the majority of NRs due to unpredictable off-target effects. The synthetic ligand T0901317 was originally discovered as a potent agonist of liver X receptors (LXRα/β) but subsequently found to target additional NRs, with activation of pregnane X receptor (PXR) being as potent as that of LXRs. We previously showed that directed rigidity reduces PXR binding by T0901317 derivatives through unfavorable protein remodeling. Here, we use a similar approach to achieve selectivity for PXR over other T0901317-targeted NRs. One molecule, SJPYT-318, accomplishes selectivity by favorably utilizing PXR’s flexible binding pocket and surprisingly binding in a new mode distinct from the parental T0901317. Our work provides a structure-guided framework to achieve NR selectivity from promiscuous compounds.
Keywords: Nuclear receptor, Promiscuity, Drug Design, Pregnane X Receptor, Liver X Receptor
Graphical Abstract

eTOC Blurb
Huber et al. use structure-guided design to develop a selective PXR agonist from a classically known promiscuous nuclear receptor ligand. The newly derived analog binds PXR in a different pose compared to the parental molecule, highlighting the use of PXR’s flexible binding pocket to recognize ligands.
INTRODUCTION
The nuclear receptor (NR) superfamily is a group of ligand-activated transcription factors with diverse roles in reproduction, development, physiology, and disease.1 There are 48 NRs encoded in the human genome that regulate transcription of various target genes,2 and alternative splicing further expands NR functionality.3 NRs share a common modular structure consisting of a variable N-terminal domain, a DNA-binding domain (DBD), and a ligand-binding domain (LBD). An activation function 2 (AF-2) helix (alpha helix 12, α12) on the LBD C-terminus regulates ligand-dependent cofactor interactions and receptor activity.4 The LBD is responsible for ligand recognition and resulting cofactor interactions through ligand-modulated α12 conformations.5 Agonists induce receptor association with coactivators, such as steroid receptor coactivator 1 (SRC-1), and inverse agonists stabilize receptor complexes with nuclear receptor corepressors like NCOR1 and NCOR2.6 Thus, the LBD dictates transcriptional transactivation activity of NRs while the DBD determines target gene specificity. The variety of DNA-binding sequences and the propensity of different NRs to either homodimerize (e.g., androgen receptor) or heterodimerize with retinoid X receptors results in a diverse set of transcriptional targets2 including proteases (e.g., KLK3),7 regulators of inflammation (e.g., ANXA1),8 drug metabolism enzymes (e.g., CYP3A4),9-12 and transcription factors (e.g., FOS).13
Most of the >1,600 transcription factors are typically difficult to develop drugs toward due to their disordered structures, but NRs are uniquely druggable because of their dedicated ligand-binding pockets.14,15 These defined pockets combined with extensively characterized natural and synthetic ligands and broad biological roles have made NRs one of the most highly therapeutically targeted protein classes. However, drugs are only approved for approximately one-third of the 48 NRs.16,17 A recurring challenge in drug development campaigns is multi-NR modulation by a single molecule. NRs are activated by an array of lipophilic molecules such as steroid hormones, vitamins, fatty acids, and xenobiotics,18 and there is considerable overlap of NR modulation by individual ligands. Both activation and inhibition of specific receptors can be advantageous in context-dependent manners, so ligand specificity is key to achieving a desired biological impact without adverse nonspecific effects.
The synthetic compound T0901317 is a representative promiscuous NR ligand that exhibits a range of agonistic and inverse agonistic activities on various NRs. First identified as a highly potent and efficacious agonist of the liver X receptors (LXRα and LXRβ),19 T0901317 was later found to modulate activities of multiple NRs, with activation of pregnane X receptor (PXR) being as robust as that of the LXRs.20,21 PXR agonists have been proposed as potential treatments for inflammatory bowel disease (IBD),22 but lack of specificity of PXR agonists has prevented more meaningful studies and additional development. For example, LXR activation causes liver lipogenesis as well as additional systemic effects due to the more widespread expression of LXR compared to PXR, whose expression is generally limited to liver and intestine.23,24 The therapeutic mechanism of the IBD drug rifaximin has at least partly been attributed to PXR activation, but the extent of PXR’s role is unknown because rifaximin is also a potent antibiotic that alters gut microbiota makeup.25,26 Therefore, enhanced specificity of PXR agonists may be desirable for more effective IBD treatments. On the other hand, small molecule-mediated PXR activation might lead to drug-drug interactions and decreased drug efficacy, indicating that, like with other receptors, both activation and inhibition of PXR can be desirable in different contexts.
We previously used T0901317 as a case study chemical to reduce PXR binding by targeted expansion of the ligand binding pocket with N-position extensions of T0901317.27 PXR has a large pocket volume (~1,200 Å3) that can expand to >1,500 Å3 to incorporate large ligands,28 although use of the expansion mechanism is unfavorable to binding affinity and subsequent activation.27 Comparatively, LXRs have a small binding pocket volume of ~700-850 Å3,29,30 and we reasoned that N-position extensions of T0901317 would lose LXR activation disproportionately to PXR activation. Therefore, in this study, we expanded our T0901317 analog series and identified two molecules, SJPYT-305 and SJPYT-318, that are highly selective PXR agonists. Interestingly, while SJPYT-318 has a larger extension than SJPYT-319 (previously published as T0-BP),27 SJPYT-318 more potently activated PXR than SJPYT-319. To investigate this unexpected observation, we solved the crystal structure of PXR LBD bound to SJPYT-318 and found that the N-position substituent was rotated ~180° compared to SJPYT-319 and T0901317, forming a network of new interactions that included stabilizing hydrophobic contacts with α12. These results highlight PXR’s ability to mold its ligand binding pocket to diverse ligand shapes and demonstrate that PXR’s flexibility can be utilized to develop PXR-selective ligands from a promiscuous initial scaffold. In contrast, ligand flexibility should be avoided when taking the approach of increasing ligand bulkiness to reduce PXR binding affinity.
RESULTS
Identification of PXR-selective T0901317 derivatives
Using a limited set of compounds, we previously showed that N-position extensions of T0901317 reduce PXR binding potency and activation potential in a stepwise manner.27 Because T0901317 was originally reported as a potent LXR agonist19 and is known to be a promiscuous NR ligand,20 we expanded our T0901317 analog series (Figure 1) and tested compound activities on PXR, LXRα, and LXRβ. We used a time-resolved fluorescence resonance energy transfer (TR-FRET) assay to measure recruitment of a 23-amino acid coactivator peptide (SRC-1) to purified NR LBDs (Figure 2A-C). To accurately reflect both potency (half maximal effective concentration, EC50) and efficacy (maximal effect), we calculated area under the curve (AUC) for each compound (Table 1). We then analyzed the values along a linear gradient to detect PXR-selective molecules (Figure 2D). The large substitution in SJPYT-320 resulted in a complete loss of activity for all receptors, and we identified two analogs, SJPYT-305 and SJPYT-318, that exhibited selectivity for PXR LBD over LXR LBDs for recruiting SRC-1. Interestingly, the selectivity occurred in different ways for the two compounds. SJPYT-305 had more efficient SRC-1 recruitment than T0901317 for PXR LBD but became less efficient than T0901317 for LXR LBDs, thus resulting in PXR selectivity. SJPYT-318, however, was less efficient than T0901317 for all proteins but had a more pronounced activity loss for LXR LBDs than for PXR LBD.
Figure 1. Chemical structures of the studied compounds.
Figure 2. Identification of PXR-selective T0901317 derivatives.
(A) Schematic of TR-FRET assay used to measure recruitment of a 23-amino acid coactivator peptide (SRC-1) to purified NR LBDs. (B-C) The indicated compounds were assayed for SRC-1 recruitment to the LBDs of PXR (left panels), LXRα (middle panels), or LXRβ (right panels). Results are expressed as the mean ± standard deviation from four replicates. (D) The area under the curve (AUC) was calculated for each of the curves in (B-C), where larger AUC indicates more SRC-1 recruitment. See also Figure S3.
Table 1. SRC-1 recruitment to NR LBDs.
TR-FRET assays were performed with purified GST-tagged NR LBDs and a fluorescently labeled 23-amino acid SRC-1 peptide. 1AUC, area under curve. 2SD, standard deviation. 3FC, fold change compared to T0901317.
| Compound | PXR | LXRα | LXRβ | |||
|---|---|---|---|---|---|---|
| AUC1 ± SD2 | FC3 | AUC ± SD | FC | AUC ± SD | FC | |
| T0901317 | 124.0 ± 4.6 | 1 | 122.4 ± 2.6 | 1 | 201.5 ± 3.3 | 1 |
| SJPYT-299 | 101.8 ± 15.7 | 0.82 | 67.1 ± 2.9 | 0.55 | 133.1 ± 1.4 | 0.66 |
| SJPYT-300 | 102.2 ± 5.8 | 0.82 | 80.2 ± 1.9 | 0.66 | 136.4 ± 3.4 | 0.68 |
| SJPYT-301 | 128.8 ± 12.9 | 1.04 | 95.1 ± 1.8 | 0.78 | 154.1 ± 2.3 | 0.76 |
| SJPYT-302 | 101.9 ± 8.1 | 0.82 | 108.1 ± 4.4 | 0.88 | 148.9 ± 5.4 | 0.74 |
| SJPYT-303 | 134.1 ± 3.1 | 1.08 | 63.6 ± 1.9 | 0.52 | 119.6 ± 2.4 | 0.59 |
| SJPYT-304 | 112.6 ± 8.2 | 0.91 | 59.5 ± 1.8 | 0.49 | 124.9 ± 3.6 | 0.62 |
| SJPYT-305 | 161.4 ± 6.4 | 1.30 | 33.4 ± 0.5 | 0.27 | 76.1 ± 2.3 | 0.38 |
| SJPYT-315 | 57.4 ± 2.7 | 0.46 | 62.3 ± 0.4 | 0.51 | 117.1 ± 2.4 | 0.58 |
| SJPYT-316 | 38.7 ± 2.2 | 0.31 | 60.8 ± 0.7 | 0.50 | 143.8 ± 3.6 | 0.71 |
| SJPYT-317 | 39.5 ± 1.5 | 0.32 | 27.2 ± 1.4 | 0.22 | 71.3 ± 2.8 | 0.35 |
| SJPYT-318 | 47.1 ± 2.4 | 0.38 | 5.0 ± 0.3 | 0.04 | 21.8 ± 1.1 | 0.11 |
| SJPYT-319 | 23.6 ± 0.2 | 0.19 | 7.3 ± 0.2 | 0.06 | 21.5 ± 0.9 | 0.11 |
| SJPYT-320 | 5.0 ± 1.2 | 0.04 | 2.5 ± 0.4 | 0.02 | 6.5 ± 1.1 | 0.03 |
PXR selectivity of molecules is accentuated in a cellular environment
Positive results in the SRC-1 recruitment assay indicated that most analogs may be agonists of PXR and LXRα/β to varying degrees. To test if SJPYT-305 and SJPYT-318 activate PXR and maintain their PXR selectivity in a cellular context, we performed activity assays in 293T cells using NR LBDs fused to the GAL4 DNA binding domain and a reporter under the control of a minimal promoter containing GAL4 binding sites (Figure 3A). Rifampicin and GW3965 were used as PXR-specific and LXR-specific agonists, respectively (Figure S1). To find a concentration range suitable for cellular experiments, we treated 293T cells with compounds and found that concentrations above 1 μM consistently reduced cell viability (Figure S2); therefore, 1 μM was chosen as the upper cutoff for T0901317 analog treatments in the cellular reporter assays.
Figure 3. PXR selectivity of molecules is accentuated in a cellular environment.
(A) Schematic of NR reporter activity assays. An NR LBD is fused to the yeast GAL4 DNA binding domain, which binds to a reporter plasmid. Expression of the reporter (firefly luciferase for PXR or beta-lactamase for LXRs) is increased in the presence of an agonist. (B-C) The indicated compounds were assayed for activity of PXR (left panels), LXRα (middle panels), or LXRβ (right panels). Results are expressed as the mean ± standard deviation from three replicates. (D) AUC was calculated for each of the curves in (B-C), where larger AUC indicates more NR activity. See also Figures S1-S3.
Reporter activity was measured after 24 h compound treatment, and activities were analyzed in the same manner as the SRC-1 recruitment assays (Figure 3B-D, Table 2, and Figure S3). The compound efficiencies for reporter activation were largely equal to those for SRC-1 recruitment, with a slight preference for cellular reporter activity over SRC-1 recruitment for PXR and LXRα and a marginal preference toward SRC-1 recruitment for LXRβ (Table 1, Table 2, and Figure S3). As expected based on our previous observations,27 activation efficiency was roughly proportional to ligand size. Remarkably, however, despite the increased size of SJPYT-318 compared to SJPYT-319, SJPYT-318 had substantially enhanced PXR activation over SJPYT-319 (Figure 3C), and this observation was consistent with SRC-1 recruitment (Figure 2C). Furthermore, SJPYT-318 was strongly selective for PXR in cellular reporter activation, showing only marginal activation of both LXRα and LXRβ (Figure 3D). Because T0901317 is also a farnesoid X receptor (FXR) agonist and RAR-related orphan receptor gamma (RORγ) inverse agonist, we tested SJPYT-305 and SJPYT-318 and found that neither compound modulated these receptors (Figure 4A-B). The compounds also did not modulate additional receptors that have large ligand binding pockets, such as peroxisome proliferator-activated receptors delta and gamma (PPARδ and PPARγ) (Figure 4C-D). Thus, these molecules, particularly SJPYT-318, are highly selective PXR agonists.
Table 2. Cellular NR activation by T0901317 analogs.
NR LBDs are fused to the yeast GAL4 DNA binding domain, which binds to a reporter plasmid. Expression of the reporter (firefly luciferase for PXR or beta-lactamase for LXRs) is increased in the presence of an agonist.
| Compound | PXR | LXRα | LXRβ | |||
|---|---|---|---|---|---|---|
| AUC±SD | FC | AUC ± SD | FC | AUC ± SD | FC | |
| T0901317 | 175.9 ± 6.3 | 1 | 186.8 ± 11.4 | 1 | 192.6 ± 28 | 1 |
| SJPYT-299 | 111.4 ± 24.3 | 0.63 | 73.5 ± 3.5 | 0.39 | 78.4 ± 9.2 | 0.41 |
| SJPYT-300 | 150.9 ± 4.8 | 0.86 | 131.4 ± 8.2 | 0.70 | 134.7 ± 10.9 | 0.70 |
| SJPYT-301 | 164.8 ± 24.4 | 0.94 | 124.9 ± 10.1 | 0.67 | 137.4 ± 26.6 | 0.71 |
| SJPYT-302 | 107.3 ± 17.3 | 0.61 | 120.8 ± 7.6 | 0.65 | 130.7 ± 19 | 0.68 |
| SJPYT-303 | 154 ± 29.8 | 0.88 | 103.9 ± 2.9 | 0.56 | 107.9 ± 16.9 | 0.56 |
| SJPYT-304 | 145.2 ± 25.7 | 0.83 | 103.5 ± 4.1 | 0.55 | 86.1 ± 18.7 | 0.45 |
| SJPYT-305 | 119.9 ± 19.9 | 0.68 | 35.1 ± 4.1 | 0.19 | 70.4 ± 23.4 | 0.37 |
| SJPYT-315 | 101.6 ± 19.8 | 0.58 | 98.0 ± 3.4 | 0.52 | 104.2 ± 18.9 | 0.54 |
| SJPYT-316 | 67.8 ± 16.2 | 0.39 | 88.8 ± 5.9 | 0.48 | 108.7 ± 13 | 0.56 |
| SJPYT-317 | 70.6 ± 6.8 | 0.40 | 43.1 ± 4.7 | 0.23 | 56.5 ± 10.1 | 0.29 |
| SJPYT-318 | 102.5 ± 20.9 | 0.58 | 11.9 ± 4.1 | 0.06 | 36.4 ± 9.8 | 0.19 |
| SJPYT-319 | 34.9 ± 1.3 | 0.20 | 12.1 ± 1.3 | 0.06 | 38.7 ± 14 | 0.20 |
| SJPYT-320 | 9.1 ± 4.9 | 0.05 | 4.1 ± 3.3 | 0.02 | 21.1 ± 7.4 | 0.11 |
Figure 4. SJPYT-305 and SJPYT-318 do not modulate additional NRs.
NR reporter activity assays were performed for (A) FXR, (B) RORγ, (C) PPARδ, or (D) PPARγ. Cells were treated with DMSO or 1 μM of the indicated compounds for 24 h. T0901317 is a known FXR agonist and RORγ inverse agonist and served as the assay control for (A) and (B). T0901317 is not reported to modulate PPARδ or PPARγ, so GW0742 or rosiglitazone served as the respective control for (C) and (D). Results are plotted as fold change (FC) relative to the average DMSO control signal and are expressed as the mean ± standard deviation from at least three replicates.
SJPYT-305 and SJPYT-318 selectively activate endogenous PXR
Next, we aimed to investigate the impact of T0901317 analogs on PXR- and LXR-dependent transcription in a more biologically relevant context by examining endogenous CYP3A4 and ABCA1 RNA induction by endogenous PXR and LXRα/β, respectively. Because PXR activation in cell lines typically results in a low-magnitude transcription response, we first examined a panel of cell lines for the greatest CYP3A4 induction upon PXR activation. Guided by the Dependency Map portal (depmap.org),31 we selected eight cell lines with high PXR RNA expression, including SNU-C4 cells, which we have previously published for PXR studies.32,33 We confirmed PXR RNA levels and assessed basal and rifampicin-induced CYP3A4 RNA levels by RT-qPCR (Figure S4). While SNU-C4 indeed had rifampicin-inducible CYP3A4 RNA expression, SNU719 inducibility was significantly greater (Figure S4C). Furthermore, the LXR target gene ABCA1 was highly inducible by LXR agonists, and rifampicin and GW3965 maintained their receptor specificity in SNU719 (Figure S5). Therefore, we chose SNU719 as the cell model for further study.
Importantly, SJPYT-305 and SJPYT-318 both retained high PXR selectivity in the SNU719 model (Figure 5). CYP3A4 induction by SJPYT-305 was similar to that by T0901317, but SJPYT-305 also slightly induced ABCA1 expression (2.3-fold for SJPYT-305 vs. 11.1-fold for T0901317). SJPYT-318, on the other hand, robustly induced CYP3A4 RNA without increasing ABCA1 RNA. None of the compounds notably altered expression of the control gene GAPDH. These results validate the selectivity of SJPYT-318 and SJPYT-305 for PXR and highlight the ability of PXR to incorporate bulky ligands that other NRs cannot.
Figure 5. SJPYT-305 and SJPYT-318 selectively induce endogenous PXR.
SNU719 cells were treated with DMSO or the indicated concentrations of (A) T0901317, (B) SJPYT-305, or (C) SJPYT-318 for 24 h. RNA was isolated and subjected to RT-qPCR to measure expression of CYP3A4, ABCA1, or GAPDH. 18S RNA was used for normalization of all samples. FC, fold change relative to DMSO controls. Results are expressed as the mean ± standard deviation from three replicates. See also Figures S4-S5.
Crystal structure of PXR LBD bound to SJPYT-318 demonstrates cooperativity between PXR flexibility and ligand conformational freedom
We achieved PXR selectivity of T0901317 derivatives by adding inflexible, bulky groups at the N-position. We previously showed that for PXR, analogs with bulky N-position substituents had decreased PXR activity due to unfavorable ligand binding pocket rearrangements. In this prior study, we found that SJPYT-319 (containing a biphenyl extension) substantially altered the position of alpha helix 2 (α2) of PXR LBD.27 Oddly, SJPYT-318 has a larger group than SJPYT-319 at the N-position (phenoxyphenyl for SJPYT-318 vs. biphenyl for SJPYT-319), and yet, SJPYT-318 more potently activated PXR than SJPYT-319 (Figure 3C and Table 2). This unexpected finding combined with the selectivity of SJPYT-318 prompted us to investigate the binding mode of SJPYT-318 to PXR LBD, and we solved the co-crystal structure to 2.2 Å (Table 3).
Table 3. Data collection and model refinement statistics for the SJPYT-318-bound PXR LBD structure (PDB code 8SZV).
Values from the highest resolution shell are shown in parentheses. 1Generated with MolProbity.
| Data Collection | |
|---|---|
| Resolution range (Å) | 45.74-2.20 (2.27-2.20) |
| Space group | P 43 21 2 |
| Unit cell dimensions | |
| a, b, c (Å) | 91.47, 91.47, 85.07 |
| α, β, γ (°) | 90.0, 90.0, 90.0 |
| Wavelength (Å) | 1.00003 |
| Unique reflections | 18851 |
| Redundancy | 14.6 (15.3) |
| Completeness (%) | 99.9 (99.1) |
| I/σI | 13.8 (2.4) |
| RMerge | 0.100 (1.339) |
| CC1/2 | 0.998 (0.829) |
| Model Refinement | |
| Rwork/Rfree | 0.211/0.223 |
| Number of atoms | 2220 |
| Protein | 2137 |
| Ligand | 40 |
| Water | 43 |
| RMSD | |
| Bond length (Å) | 0.005 |
| Bond angles (°) | 0.769 |
| Ramachandran plot (%) | |
| Preferred | 98.52 |
| Outliers | 0.00 |
| Clashscore1 | 0.94 |
| MolProbity score1 | 0.78 |
| Average B-factor (Å2) | 56.29 |
| Protein | 55.76 |
| Ligand | 83.31 |
| Water | 57.57 |
As with all reported PXR LBD structures obtained in the absence of SRC-1 peptide, the asymmetric unit contained a single PXR LBD protomer (Figure 6A). The orientation of SJPYT-318 was clear, although the benzenesulfonamide moiety appeared to have at least two orientations – one toward the F288-W299-Y306 aromatic cage and one toward α2. The aromatic cage-oriented position seemed to be low occupancy, so we modeled only the α2 orientation (Figure 6B). The hexafluoro hydroxypropyl moiety was positioned similarly to T0901317 and previous derivatives, with the hydroxyl group forming a hydrogen bond with H407 (Figure 6C). Surprisingly, however, the benzenesulfonamide and N-position substituent of SJPYT-318 were rotated ~180° compared to the parental T0901317 and related SJPYT-319 (Figure 6D). Rather than being oriented over α2 like the biphenyl moiety of SJPYT-319, the phenoxyphenyl of SJPYT-318 was rotated toward α3 and α5, with the terminal benzene ring near α12 (also known as the AF-2 helix). This position required a rotamer flip of F251 and subsequent movement of L428 and F429 to accommodate the altered rotamer (Figure 6E). Interestingly, the position of the terminal benzene ring and resulting α12 interactions somewhat resembled that of our previously reported agonist SJB7, with M425, L428, and F429 forming a hydrophobic surface that interacts with the ligand (Figure 6F).32 The α3 residue F251 further stabilizes the complex by interacting with both the SJPYT-318 terminal benzene ring and the α12hydrophobic surface, pinning α12 to the ligand binding pocket (Figure 6F). Thus, SJPYT-318 is a more efficient PXR agonist than SJPYT-319 because it is reoriented to avoid the α2 clash, and other regions of the flexible ligand binding pocket move to not only accommodate the new ligand position, but to favorably incorporate the ligand in its new binding mode. The new ligand binding mode is made possible by 1) the flexibility of the PXR ligand binding pocket and 2) the flexibility imparted to the ligand by the addition of an oxygen between the two benzene rings.
Figure 6. SJPYT-318 adopts new binding mode compared to parental T0901317.
PXR LBD was co-crystal I ized with SJPYT-318 to a resolution of 2.2 Å. (A) One chain of PXR LBD was present in the asymmetric unit. The protein is shown as light blue cartoon, and SJPYT-318 is represented as deep blue spheres. (B) The 2Fo─Fc map for SJPYT-318 is contoured in mesh at 1.0 rmsd and carved around SJPYT-318 at 2.0 Å. (C) Residues of the ligand binding pocket within 5 Å of SJPYT-318 are shown as sticks. (D) Comparison of SJPYT-318 binding mode with binding modes of SJPYT-319 (PDB ID 8FPE) and T0901317 (PDB ID 2O9I, chain A). (E) SJPYT-318-bound PXR LBD (light blue protein with deep blue ligand) is overlaid with T0901317-bound PXR LBD (gray). α12 residues (L428 and F429) and the adjacent F251 move to incorporate the phenoxyphenyl group of SJPYT-318. (F) SJPYT-318-bound PXR LBD (light blue protein with deep blue ligand) is overlaid with SJB7-bound PXR LBD (PDB ID 5X0R, light teal). The α12 residues M425, L428, and F429 and the adjacent F251 are similarly arranged to incorporate the two ligands.
DISCUSSION
Both activation and inhibition of NRs can be beneficial or detrimental in context-specific manners. For example, oral contraceptives contain estrogen receptor agonists while estrogen receptor antagonists are staples of breast cancer treatments.34,35 Similarly, PXR antagonists have potential value for enhancing drug bioavailability and treating conditions such as drug- or hemorrhagic shock-induced liver injury,32,36,37 while agonists may be used for treatment of IBD.22 These dichotomies combined with multi-NR modulation of individual promiscuous chemicals reinforce the need for specificity in NR-targeting molecules. Achieving selectivity could open new avenues to treat pathologies in which NRs are known to play key roles.
Among the 48 human nuclear receptors, PXR is unique in its large ligand binding pocket volume (1200-1600 Å3)28 that allows binding of a vast array of molecules. Because of its propensity to be activated by diverse molecules and its role in regulating drug metabolism genes, PXR is often defined as a xenobiotic receptor. However, PXR also plays roles in homeostatic processes such as glucose metabolism, diabetes, intestinal barrier function, and general liver function.24 PXR has been shown to mitigate intestinal inflammation through interaction with gut microbial metabolites and reduction of toll-like receptor 4 (TLR4) signaling.38 Exogenous PXR agonists have also been shown to protect against IBD through crosstalk with nuclear factor kappa B (NF-κB) signaling.39 Furthermore, PXR activation by the IBD drug rifaximin has been proposed as rifaximin’s therapeutic mechanism, but the extent of PXR’s role is unknown because rifaximin is also a potent antibiotic that alters gut microbiome composition.25,26 PXR may therefore be a viable target for IBD treatment, but studies are currently limited by nonspecific agonists.
Though T0901317 has previously been used as a platform to generate modulators of NRs such as LXRs and retinoic acid-related orphan receptors,40,42 and attempts at deriving PXR antagonists have even been made,43 efforts aimed at enhancing PXR selectivity are lacking, and there is little information available specifically on the N-position. While the N-trifluoroethyl is directed at a flexible region of PXR LBD, it is in a relatively constrained area of the LXRβ LBD,44 and it was reasonable to predict that N-position extensions would disproportionately diminish LXR activity over PXR. However, we were surprised to find that the phenoxyphenyl substitution in SJPYT-318 not only imparted PXR selectivity, but also completely flipped the binding mode. This was made possible both by the rotatable bonds in the molecule and the permissive nature of the PXR ligand binding pocket. The rotation brought the phenoxyphenyl group in close proximity to α12, similar to our previously reported agonist SJB7, resulting in productive stabilizing interactions. Interestingly, as we have previously studied SJB7 analogs as PXR antagonists, similar approaches might be used to generate antagonists from SJPYT-318. Thus, SJPYT-318 represents a PXR-selective agonist that can serve as a starting point for further development of both agonists and antagonists.
STAR METHODS
RESOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Taosheng Chen (taosheng.chen@stjude.org).
Materials availability
All unique/stable reagents generated in this study are available from the lead contact with a completed materials transfer agreement. There are restrictions to the availability of new compounds due to the lack of an external centralized repository for their distribution and our need to produce and maintain the stocks. We are glad to share new compounds with reasonable compensation by the requestor for its production, processing, and shipping.
Data and code availability
The structure of SJPYT-318-bound PXR LBD was deposited in the Protein Data Bank (PDB) under accession code 8SZV.
This paper does not report original code.
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
293T/17 cells (referred to as 293T) were obtained from the American Type Culture Collection (ATCC, cat. # CRL-11268) and maintained in Dulbecco's Modified Eagle Medium (DMEM, ATCC, cat. # 30-2002) with 10% fetal bovine serum (FBS, HyClone, cat. # SH30396.03). LS1034 cells were obtained from ATCC (cat. # CRL-2158) and maintained in Roswell Park Memorial Institute 1640 (RPMI) medium (ATCC, cat. # 30-2001) with 10% FBS. SW948 (cat. # CCL-237), SW1417 (cat. # CCL-238), and SW1463 (cat. # CCL-234) cells were obtained from ATCC and maintained in Leibovitz's L-15 Medium (ATCC, cat. # 30-2008) with 10% FBS. SNU-C4 (cat. # 0000C4), SNU81 (cat. # 00081), SNU719 (cat. # 00719), and SNU620 (cat. # 00620) cells were obtained from the Korean Cell Line Bank (KCLB) and maintained in RPMI with 10% FBS. GeneBLAzer LXRα-UAS-bla HEK 293T Cells (cat. # K1692) and GeneBLAzer LXRβ-UAS-bla HEK 293T Cells (cat. # K1699) were obtained from Thermo Fisher Scientific and maintained in DMEM with 10% FBS, 100 μg/mL Hygromycin B (Thermo Fisher Scientific, cat. # 10687010), and 100 μg/mL Zeocin (Thermo Fisher Scientific, cat. # R25001). GeneBLAzer PPARδ-UAS-bla HEK 293T Cells (Thermo Fisher Scientific, cat. # K1690) were maintained in DMEM with 10% FBS, 80 μg/mL Hygromycin B, and 100 μg/mL Zeocin. GeneBLAzer PPARγ-UAS-bla HEK 293H Cells (Thermo Fisher Scientific, cat. # K1701) were maintained in DMEM with 10% FBS, 100 μg/mL Hygromycin B, and 500 μg/mL geneticin (Thermo Fisher Scientific, cat. # 10131027). The GeneBLAzer cells were cultured on plates coated with 0.25% Matrigel Growth Factor Reduced Basement Membrane Matrix (Corning, cat. # 356230), as per the manufacturer’s recommendations. All cells were routinely verified to be mycoplasma free by using the MycoProbe Mycoplasma Detection Kit (R&D Systems), and, apart from the SW lines, were incubated in a humidified atmosphere at 37°C with 5% CO2. SW948, SW1417, and SW1463 cells were incubated in a humidified atmosphere at 37°C without CO2, according to ATCC recommendations. Cell counts were obtained with a Countess II Automated Cell Counter using trypan blue staining. The “Assay Medium” referenced in the following subsections was DMEM, high glucose, HEPES, no phenol red (Thermo Fisher Scientific, cat. # 21063029) supplemented with 2% charcoal/dextran treated FBS (HyClone, cat. # SH30068.03), 1 mM sodium pyruvate (Thermo Fisher Scientific, cat. # 11360070), and 0.1 mM non-essential amino acids (Thermo Fisher Scientific, cat. # 11140050). For protein expression, TurboCells Competent E. coli BL21(DE3) (Genlantis, cat. # C302020) were grown in terrific broth at 37°C.
METHOD DETAILS
General chemistry methods and synthesis
Dimethyl sulfoxide (DMSO) was purchased from Fisher Scientific (cat. # BP231-100). T0901317 was purchased from Cayman Chemical (cat. # 71810). Rosiglitazone (cat. # HY-17386) and GW0742 (cat. # HY-13928) were purchased from MedChemExpress. Organic reagents were purchased from commercial suppliers unless otherwise noted and were used without further purification. All solvents were analytical or reagent grade and the solvents were dried using the Glass Contour Solvent Systems by SG Water USA. All reactions with water- and/or air-sensitive starting materials were carried out in pre-dried glassware under argon atmosphere with standard procedure. Flash column chromatography was performed by using Biotage Isolera Flash Systems and Biotage SNAP Ultra or Biotage® SNAP Ultra C18 columns (Biotage, Charlotte, NC). All reactions as well as compound purity were monitored by UPLC-MS by using a Waters Acquity UPLC MS system with a C18 column in a 2-min gradient (H2O + 0.1% formic acid → acetonitrile + 0.1% formic acid) and detectors of PDA (215–400 nm), ELSD, and Acquity SQD ESI-positive MS (Waters Corporation, Milford, MA). High-resolution mass spectra were determined by using a Waters Acquity UPLC system with a C18 column (H2O + 0.1% formic acid → acetonitrile + 0.1% formic acid gradient over 2.5 min) and Xevo G2Q-TOF ESI-positive MS in resolution mode. Compounds were internally normalized to leucine-enkephalin lock solution, with a calculated error of <5 ppm. All final compounds used for SAR studies have purity at 95% or greater (purity was calculated based on the average of the ELSD detector purity and UV detector purity). All NMR spectra were recorded on a Bruker 500 MHz spectrometer (Bruker Corporation) in the solvents indicated and spectra were processed using MestReNova (14.1.0) (Mestrelab Research). The chemical shift values are expressed in parts per million (ppm) relative to tetramethylsilane as the internal standard. Coupling constants (J) are reported in hertz (Hz). 1H NMR, 13C NMR, and HRMS spectra and HPLC chromatograms are shown in Data S1.
Syntheses of SJPYT-302, 315-317, and 319 have been described previously.27 Syntheses of SJPYT-300, 301, 303, 304, 318, and 320 were prepared by a similar procedure. Sulfonamidation of 2-(4-Aminophenyl)-1,1,1,3,3,3-hexafluoropropan-2-ol (1a) with benzenesulfonyl chloride in the presence of 2,6-Lutidine gave N-[4-(1,1,1,3,3,3-Hexafluoro-2-hydroxypropan-2-yl)phenyl]benzenesulfonamide (1b). The resulting sulfonamide (1b) was then alkylated with respective alkyl-halides in the presence of potassium carbonate and heating to produce various N-alkyl analogs (SJPYT-300 to SJPYT-304 and SJPYT-315 to SJPYT-320). For SJPYT-299 and SJPYT-305, the aniline 1a was alkylated under reductive amination conditions with corresponding aldehydes in the presence of sodium cyanoborohydride to generate the secondary aniline intermediates (2a and 2b) which were then converted to SJPYT-299 and SJPYT-305 via sulfonamidation with benzenesulfonyl chloride.41
Synthesis of SJPYT-299 to 305 and SJPYT-315 to 320

Reagents and conditions: (a) Benzenesulfonyl chloride, 2,6-lutidine, acetone, reflux; (b) RBr or RI, K2CO3, CH3CN, 70°C; (c) 3,3,3-trifluoropropanal or 4-hydroxybenzaldehyde, NaBH3CN, MeOH, room temperature; (d) PhSO2Cl, pyridine, DMAP (cat.), toluene, reflux.
N-ethyl-N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)benzenesulfonamide (SJPYT-300)
Benzenesulfonyl chloride (2.59 ml, 20.26 mmol) and 2,6-dimethylpyridine (4.13 g, 38.6 mmol) were added to a solution of 2-(4-aminophenyl)-1,1,1,3,3,3-hexafluoropropan-2-ol (1a, 5g, 19.29 mmol) in acetone (30 mL) at room temperature. The mixture was stirred at 60°C overnight. After mass spectrometry (MS) demonstrated the complete consumption of the starting material 1a, the reaction mixture was concentrated, and water was added (100 mL). The aqueous layer was extracted with EtOAc (100 mL × 2). The combined organic layer was washed with brine. The organic layer was then dried over MgSO4, filtered, and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0% to 100% acetonitrile in water) to give N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)benzenesulfonamide (1b) as an off-white solid (6.78 g, 88% yield). 1H NMR (500 MHz, DMSO-d6) δ 11.21 (s, 1H), 9.10 (s, 1H), 8.45 – 8.24 (m, 2H), 8.18 – 8.12 (m, 1H), 8.11 – 8.00 (m, 4H), 7.81 – 7.67 (m, 2H).
K2CO3 (138 mg, 1.002 mmol) and iodoethane (44.3 μL, 0.551 mmol) were added to a solution of N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)benzenesulfonamide (1b, 200 mg, 0.501 mmol) in 5 mL CH3CN at room temperature. The suspension was stirred at 70°C overnight. Water (50 mL was added, and the reaction mixture was extracted with EtOAc (50 mL × 2). The EtOAc layer was washed with water, dried with anhydrous Na2SO4, and concentrated. The residue was purified by silica gel chromatography (0% to 100% EA in hexane) to give product SJPYT-300 as a white solid, 161.3 mg, 75% yield, 96.86% purity. 1H NMR (500 MHz, DMSO-d6) δ 8.81 (s, 1H), 7.71 (tt, J = 5.9, 2.6 Hz, 1H), 7.67 (d, J = 8.4 Hz, 2H), 7.62 – 7.55 (m, 4H), 7.28 – 7.22 (m, 2H), 3.62 (q, J = 7.1 Hz, 2H), 0.97 (t, J = 7.1 Hz, 3H). 13C NMR (126 MHz, DMSO-d6) δ 140.64, 138.23, 133.74, 130.29, 129.86, 128.70, 128.09, 127.57, δ 123.29 (q, J = 288.7 Hz), 77.14 (p, J = 29.4 Hz), 45.31, 14.34. ESI-TOF HRMS: m/z 428.0760 (C17H15F6NO3S + H+ requires 428.0750).
N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-N-propylbenzenesulfonamide (SJPYT-301)
The title compound (SJPYT-301) was synthesized using a procedure similar to that described for SJPYT-300, with the utilization of 1b and 1-iodopropane to give a white solid, 173.1 mg, 70% yield, 97.92% purity. 1H NMR (500 MHz, DMSO-d6) δ 8.80 (s, 1H), 7.70 (ddt, J = 8.6, 6.9, 1.7 Hz, 1H), 7.66 (d, J = 8.4 Hz, 2H), 7.62 – 7.52 (m, 4H), 7.32 – 7.23 (m, 2H), 3.54 (t, J = 7.0 Hz, 2H), 1.31 (h, J = 7.2 Hz, 2H), 0.82 (t, J = 7.3 Hz, 3H). 13C NMR (126 MHz, DMSO-d6) δ 140.82, 138.13, 133.72, 130.30, 129.82, 128.68, 128.07, 127.58, 126.73, 124.44, 122.14, 119.84, 77.61, 77.38, 77.14, 76.91, 76.68, 51.68, 21.47, 11.21. ESI-TOF HRMS: m/z 442.0918 (C18H17F6NO3S + H+ requires 442.0906).
N-benzyl-N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)benzenesulfonamide (SJPYT-303)
The title compound (SJPYT-303) was synthesized using a procedure similar to that described for SJPYT-300, with the utilization of 1b and benzyl bromide to give a white solid, 179.9 mg, 73% yield, 98.34% purity. 1H NMR (500 MHz, DMSO-d6) δ 8.72 (s, 1H), 7.79 – 7.72 (m, 1H), 7.68 – 7.59 (m, 4H), 7.56 (d, J = 8.5 Hz, 2H), 7.32 – 7.24 (m, 6H), 7.19 (ddd, J = 8.7, 4.9, 3.9 Hz, 1H), 4.85 (s, 2H). 13C NMR (126 MHz, DMSO-d6) δ 140.81, 138.16, 136.49, 133.93, 130.06, 129.95, 128.86, 128.46, 128.34, 127.98, 127.87, 127.68, 126.66, 119.78, 77.54, 77.31, 77.07, 76.84, 76.61, 53.50. ESI-TOF HRMS: m/z 490.0905 (C22H17F6NO3S + H+requires 490.0906).
N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-N-(4-methoxybenzyl)benzenesulfonamide (SJPYT-304)
The title compound (SJPYT-304) was synthesized using a procedure similar to that described for SJPYT-300 with the utilization of 1b and 1-(bromomethyl)-4-methoxybenzene to give a white solid, 445.2 mg, 73% yield, 98.38% purity. 1H NMR (500 MHz, DMSO-d6) δ 8.73 (s, 1H), 7.73 (tt, J = 6.7, 1.9 Hz, 1H), 7.67 – 7.59 (m, 4H), 7.56 (d, J = 8.5 Hz, 2H), 7.27 – 7.22 (m, 2H), 7.18 – 7.13 (m, 2H), 6.83 – 6.77 (m, 2H), 4.76 (s, 2H), 3.67 (s, 3H). 13C NMR (126 MHz, DMSO-d6) δ 159.06, 140.73, 138.23, 133.87, 130.03, 129.93, 129.77, 128.55, 128.15, 127.83, 127.65, 126.67, 124.38, 122.08, 119.79, 114.25, 77.54, 77.31, 77.08, 76.85, 76.62, 55.44, 52.97. ESI-TOF HRMS: m/z 518.0881 (C23H19F6NO4S - H+ requires 518.0861).
N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-N-(4-phenoxybenzyl)benzenesulfonamide (SJPYT-318)
The title compound (SJPYT-318) was synthesized using a procedure similar to that described for SJPYT-300 with the utilization of 1b and 1-(bromomethyl)-4-phenoxybenzene to give a white solid, 205.9 mg, 71% yield, 95.38% purity. 1H NMR (500 MHz, DMSO-d6) δ 8.75 (s, 1H), 7.78 – 7.69 (m, 1H), 7.71 – 7.59 (m, 4H), 7.57 (d, J = 8.5 Hz, 2H), 7.39 – 7.31 (m, 2H), 7.30 – 7.23 (m, 4H), 7.16 – 7.06 (m, 1H), 6.94 – 6.85 (m, 4H), 4.82 (s, 2H). 13C NMR (126 MHz, DMSO-d6) δ 155.89, 155.31, 139.70, 137.08, 132.85, 130.36, 129.39, 129.13, 129.02, 128.88, 127.54, 126.79, 126.60, 125.60, 123.30, 122.89, 121.00, 118.72, 117.90, 76.23, 76.00, 75.77, 75.54, 51.96. ESI-TOF HRMS: m/z 580.1010 (C28H21F6NO4S - H+ requires 580.1017).
N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-N-(4-(1,2,2-triphenylvinyl)benzyl)benzenesulfonamide (SJPYT-320)
The title compound (SJPYT-320) was synthesized using a procedure similar to that described for SJPYT-300 with the utilization of 1b and (2-(4-(bromomethyl)phenyl)ethene-1,1,2-triyl)tribenzene to give a white solid, 304.8 mg, 82% yield, 99.39% purity.1H NMR (500 MHz, DMSO-d6) δ 8.77 (s, 1H), 7.75 – 7.69 (m, 1H), 7.67 – 7.54 (m, 6H), 7.18 – 7.14 (m, 2H), 7.12 – 6.98 (m, 11H), 6.95 – 6.91 (m, 2H), 6.90 – 6.77 (m, 6H), 4.75 (s, 2H). 13C NMR (126 MHz, DMSO-d6) δ 142.44, 142.37, 142.35, 141.77, 140.06, 139.66, 139.47, 137.24, 133.54, 132.81, 130.01, 129.95, 129.04, 128.86, 127.59, 127.20, 127.15, 127.05, 126.86, 126.71, 126.59, 125.94, 125.84, 125.64, 123.34, 121.05, 118.75, 76.49, 76.25, 76.02, 75.79, 75.55, 52.43. ESI-TOF HRMS: m/z 742.1843 (C42H31F6NO3S - H+ requires 742.1851).
N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-N-(3,3,3-trifluoropropyl)benzenesulfonamide (SJPYT-299)
NaCNBH3 (485 mg, 7.72 mmol) and 3,3,3-trifluoropropanal (173 mg, 1.543 mmol) were added to a solution of 2-(4-aminophenyl)- 1,1,1,3,3,3-hexafluoropropan-2-ol (1a, 400 mg, 1.543 mmol) in MeOH (5 mL) at room temperature. The suspension was stirred at room temperature overnight. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL × 2). The EtOAc layer was washed with water (50 mL × 2), dried with anhydrous Na2SO4, and concentrated. The residue (crude 2a) was mixed with pyridine (5 mL), toluene (10 mL), DMAP (cat.), and benzenesulfonyl chloride (296 μL, 2.32 mmol). The reaction mixture was under reflux overnight. The reaction mixture was poured into water (50 mL) and extracted with EtOAc (50 mL × 2). The EtOAc layer was washed with water (50 mL × 2), dried with anhydrous Na2SO4, and concentrated. The residue was purified by silica gel chromatography (0% to 100% EA in hexane) to give product SJPYT-299 as a white solid, 212.4 mg, 28% yield (two steps), 98.14% purity. 1H NMR (500 MHz, DMSO-d6) δ 8.83 (d, J = 0.9 Hz, 1H), 7.76 – 7.70 (m, 1H), 7.67 (d, J = 8.4 Hz, 2H), 7.62 – 7.53 (m, 4H), 7.30 – 7.24 (m, 2H), 3.87 (t, J = 6.7 Hz, 2H), 2.43 (qt, J = 11.1, 6.7 Hz, 2H). 13C NMR (126 MHz, DMSO-d6) δ 139.29, 136.31, 133.00, 129.71, 128.97, 128.85, 127.71, 127.15, 126.77, 126.62, 125.63, 124.56, 123.34, 122.36, 121.05, 118.76, 76.53, 76.30, 76.07, 75.83, 75.60, 43.15, 43.11, 43.09, 43.06, 31.88, 31.67, 31.45, 31.23. ESI-TOF HRMS: m/z 496.0630 (C18H14F9NO3S+ H+ requires 496.0623).
N-(4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl)-N-(4-hydroxybenzyl)benzenesulfonamide (SJPYT-305)
The title compound (SJPYT-305) was synthesized using a procedure similar to that described for SJPYT-299 with the utilization of 1a and 4-hydroxybenzaldehyde to give a white solid, 103.2 mg, 13% yield (two steps), 99.26% purity. 1H NMR (500 MHz, DMSO-d6) δ 9.34 (s, 1H), 8.72 (s, 1H), 7.73 (td, J = 6.7, 2.1 Hz, 1H), 7.66 – 7.59 (m, 4H), 7.55 (d, J = 8.5 Hz, 2H), 7.25 – 7.18 (m, 2H), 7.06 – 6.96 (m, 2H), 6.63 – 6.55 (m, 2H), 4.70 (s, 2H).1H NMR (500 MHz, DMSO-d6) δ 9.34 (s, 1H), 8.72 (s, 1H), 7.73 (td, J = 6.7, 2.1 Hz, 1H), 7.66 – 7.59 (m, 4H), 7.55 (d, J = 8.5 Hz, 2H), 7.25 – 7.18 (m, 2H), 7.06 – 6.96 (m, 2H), 6.63 – 6.55 (m, 2H), 4.70 (s, 2H). ESI-TOF HRMS: m/z 550.0728 (C22Hi7F6NO4S + HCOO− requires 550.5765).
Plasmids
The pG5luc vector containing five GAL4 binding sites upstream of a minimal TATA box and the firefly luciferase gene was obtained from Promega as a component of the CheckMate Mammalian Two-Hybrid System (cat. # E2440). The pBIND vector containing the yeast GAL4 DNA-binding domain upstream of a multiple cloning region was also a component of the CheckMate Mammalian Two-Hybrid System (Promega, cat. # E2440). The PXR expression vector pcDNA3-FLAG-PXR has been described previously.45 To generate pBIND-PXR LBD, which contains the GAL4 DNA-binding domain fused to PXR LBD (residues 130-434), the PXR LBD was amplified from pcDNA3-FLAG-PXR using primers 5’-TCTAGAGCGGCCGCAGGTACCAGTGAACGGACAGGGACTC-3’ and 5’-GGCCTTAGTTATTCAGGTACCTCAGCTACCTGTGATGCC-3’. The amplicon was then inserted into KpnI-digested pBIND using NEBuilder HiFi DNA Assembly Master Mix (New England Biolabs, cat. # E2621S). To generate pBIND-FXR LBD and pBIND-RORγ LBD, KpnI-digested pBIND was ligated to gBlocks (Integrated DNA Technologies) containing either FXR LBD (residues 214-472) or RORγ LBD (residues 251-518) using NEBuilder HiFi DNA Assembly Master Mix. The pETDuet-1 vector (Sigma-Aldrich, cat. # 71146) containing His-tagged PXR LBD (residues 130-434) and untagged mouse SRC-1 (residues 623-710) has been described previously.27,32
Time-resolved fluorescence resonance energy transfer (TR-FRET) PXR LBD coactivator recruitment assay
The TR-FRET PXR LBD coactivator recruitment assay measures PXR LBD interaction with a fluorescently labeled SRC-1 peptide (FAM-SRC1-B) and was performed as previously described with minor modifications.46 The fluorescently labeled peptide FAM-SRC1-B with the sequence of FAM-(PEG)5-CPSSHSSLTERHKILHRLLQEGSPS-NH2 was synthesized by the Macromolecular Synthesis Section of the Hartwell Center for Bioinformatics and Biotechnology at St. Jude Children's Research Hospital. The assay buffer composition was 50 mM Tris (pH 7.5), 20 mM MgCl2, 0.1 mg/mL bovine serum albumin (BSA), and 0.05 mM dithiothreitol (DTT). FAM-SRC1-B peptide solution (10 μL/well, 150 nM in assay buffer) was dispensed into ProxiPlate-384 Plus F black 384-shallow well microplates (PerkinElmer, cat. # 6008260). An Echo 650 Acoustic Liquid Handler then dispensed 15 nL/well of compound stocks or DMSO. Lastly, 5 μL/well of 15 nM LanthaScreen Tb-anti-GST antibody (Thermo Fisher Scientific, cat. # PV3550) and 15 nM GST-PXR LBD protein (Thermo Fisher Scientific, cat. # PV4841) in assay buffer was added. The final assay volume per well was 15 μL, and the final concentrations for the assay components were: 100 nM FAM-SRC1-B peptide, 5 nM Tb-anti-GST, 5 nM GST-PXR LBD, and 0.2% DMSO (0.1% from the compound addition and 0.1% from the peptide solution). 0.2% DMSO and 1 μM T0901317 were included in each plate as negative (0% signal) and positive (100% signal) controls, respectively. The plates were shaken at 900 rpm (80 × g) on an IKA MTS 2/4 digital microtiter shaker for 1 min then centrifuged at 200 × g for 30 s in an Eppendorf 5810 centrifuge equipped with an A-4-62 swing-bucket rotor. The plates were protected from light exposure and incubated for 90 min at room temperature. The TR-FRET signal from each well was collected with a PHERAstar FS microplate reader (BMG LABTECH) using a 340 nm excitation filter, 520 nm and 490 nm emission filters, a 100 μs delay, and a 200 μs integration time. The measured RFU were normalized for each well using equation 5,
| Equation 5 |
and equation 6,
| Equation 6 |
TR-FRET LXRα LBD and LXRβ LBD coactivator recruitment assays
The assays were performed similarly to the PXR LBD coactivator recruitment assay, with modifications. GST-LXRα LBD (cat. # PV4657) and GST-LXRβ LBD (cat. # PV4660) were purchased from Thermo Fisher Scientific. The assay buffer composition was 50 mM Tris (pH 7.5), 0.002% Pluronic F-127, 0.01% BSA, and 5 mM DTT. The final FAM-SRC1-B peptide concentrations were 50 nM for LXRα assay and 125 nM for the LXRβ assay.
Cytotoxicity assay
293T cells (5 × 103/well in 25 μL Assay Medium) were plated and treated as in the reporter assays. After 24 h compound treatment, the CellTiter-Glo Luminescent Cell Viability Assay (Promega, cat. # G7572) and an EnVision microplate reader were used to assess cell viability. Cells treated with 0.1% DMSO served as positive controls (100% cell viability), and wells without cells (medium only) served as negative controls (0% cell viability). The percent cell viability for each well was calculated using equation 4,
| Equation 4 |
PXR transactivation assay
293T cells (1 × 106/well in 2 mL/well DMEM with 10% FBS) were reverse transfected with pG5luc and pBIND-PXR LBD (1 μg/well each) using FuGENE HD Transfection Reagent (Promega, cat. # E2311). The following day, cells were trypsinized and resuspended in Assay Medium, and 5 × 103 cells/well in 25 μL were added to white CulturPlate-384 plates (PerkinElmer, cat. # 6007680). An Echo 650 Acoustic Liquid Handler (Beckman Coulter) was used to dispense 25 nL/well of DMSO or stock compounds, resulting in 0.1% DMSO and the indicated concentrations of compounds. Cells treated with 0.1% DMSO served as negative controls (0% activity), cells treated with 1 μM T0901317 served as positive controls (100% activity), and wells without cells (medium only) were used for background correction. After 24 h, the steadylite plus Reporter Gene Assay System (PerkinElmer, cat. # 6066751) and an EnVision microplate reader (PerkinElmer) were used to measure luminescence. Data normalization occurred in two stages. First, the average measured relative light units (RLU) of the medium-only wells were subtracted from the RLU for all wells to give background-corrected values for each reading. Second, the percent activity was calculated for each well using equation 1,
| Equation 1 |
LXRα and LXRβ GeneBLAzer assay
GeneBLAzer cells (1 × 104/well in 25 μL Assay Medium) were plated in black 384-well clear-bottom plates (Corning, cat. # 3764). An Echo 650 Acoustic Liquid Handler was used to dispense 25 nL/well of DMSO or stock compounds, resulting in 0.1% DMSO and the indicated concentrations of compounds. Cells treated with 0.1% DMSO served as negative controls (0% activity), cells treated with 1 μM T0901317 served as positive controls (100% activity), and wells without cells (medium only) were used for background correction. After 24 h, the LiveBLAzer FRET-B/G Loading Kit with CCF4-AM (Thermo Fisher Scientific, cat. # K1096) and a CLARIOstar Plus microplate reader (BMG LABTECH) were used to detect reporter activity. Readings were taken with excitation/emission wavelengths (nm) of 409/460 and 409/530. Data normalization occurred in three stages. First, the average relative fluorescence units (RFU) of the medium-only wells at 460 nm and 530 nm were subtracted from the RFU of all wells to give background-corrected values for each reading. Second, the “signal” for each well was calculated using equation 2,
| Equation 2 |
Third, the percent activity was calculated for each well using equation 3,
| Equation 3 |
FXR, RORγ, PPARδ, and PPARγ transactivation assays
Cellular FXR and RORγ assays were conducted by the same protocol as used for the PXR transactivation assay. However, rather than calculating percent activity, the background-corrected RLU values were normalized to DMSO control wells to yield fold change (FC). PPARδ and PPARγ GeneBLAzer assays were performed by the same protocol as used for LXRα and LXRβ. However, rather than calculating percent activity, the well signal values from equation 2 were normalized to DMSO control wells to yield FC.
Nuclear receptor target gene quantitative real-time polymerase chain reaction (RT-qPCR)
SNU-C4 cells (2 × 105/well in 1 mL/well RPMI with 10% FBS) were plated in tissue culture-treated 12-well plates (Corning, cat. # 3512). Cells were grown for six days with fresh medium added every two days. Cells were washed with PBS, and 800 μL Assay Medium containing 0.5% DMSO and the indicated concentrations of compounds was added. After 24 h, total RNA was isolated from cells with Maxwell 16 LEV SimplyRNA Tissue Kits (Promega, cat. # AS1280), and cDNA was generated from 1 μg of RNA with the Superscript VILO cDNA Synthesis Kit (Thermo Fisher Scientific, cat. # 11754050). RT-qPCR was conducted with 2 μL of cDNA using TaqMan Fast Advanced Master Mix (Thermo Fisher Scientific, cat. # 4444557) in an Applied Biosystems 7500 Fast Real-Time PCR System. TaqMan gene expression assays specific for CYP3A4 (Hs00604506_m1), ABCA1 (Hs01059118_m1), GAPDH (Hs03929097_g1), and RNA18S (Hs03928990_g1) were purchased from Thermo Fisher Scientific. Fold induction values were calculated according to the 2−ΔΔCt method, where ΔCt represents the differences in cycle threshold numbers between the target gene and reference gene and ΔΔCt represents the relative change in these differences between the control and treatment groups.47 RNA18S was used as the reference gene for relative quantification of all other genes.
Protein purification, crystallization, and structure determination
PXR LBD was expressed and purified as previously described,27,32 with minor modifications. Codon-optimized sequences for His-tagged PXR LBD (residues 130-434) and untagged mouse SRC-1 (residues 623-710) were cloned into pETDuet-1 (Novagen), which allows co-expression of two genes from separate inducible T7 promoters. The plasmid was transformed into TurboCells Competent E. coli BL21(DE3) (Genlantis, cat. # C302020), grown in terrific broth at 37°C to an OD600 of 3-4, and induced overnight at 16°C with 500 μM isopropyl β-D-thiogalactoside (IPTG). Cells were pelleted by centrifugation at 4,000 × g and resuspended in lysis buffer [20 mM Tris (pH 7.5), 250 mM NaCl, 5% (v/v) glycerol, 10 mM imidazole] supplemented with EDTA-free SIGMAFAST protease inhibitor cocktail Tablets (MilliporeSigma, cat. # S8830-20TAB) and 1 mg/mL lysozyme (Gold Biotechnology, cat. # 12650-88-3). The suspension was sonicated and then centrifuged at 20,000 × g for one hour, and the supernatant was applied to a 5 mL HisTrap FF column (Cytiva). The column was washed with 50 mL lysis buffer, 25 mL lysis buffer with 50 mM imidazole, and 25 mL lysis buffer with 100 mM imidazole, and bound proteins were eluted with lysis buffer containing 500 mM imidazole. Elution fractions were collected and analyzed by SDS-PAGE for protein amount and purity. Selected fractions were pooled, and a 2:1 molar ratio of SRC-1 peptide was added to stabilize PXR LBD (N-CPSSHSSLTERHKILHRLLQEGSPS-C, prepared by the Macromolecular Synthesis Section at St. Jude Children’s Research Hospital). The PXR LBD/peptide mixture was concentrated to ≤10 mL in a Amicon Ultra-15 centrifugal filter unit with 10 kDa cutoff (MilliporeSigma), filtered through a 0.22 μm syringe filter, and loaded onto a HiLoad 26/600 Superdex 200 pg size exclusion column (Cytiva) equilibrated with storage buffer [20 mM Tris (pH 7.8), 200 mM NaCl, 5% (v/v) glycerol, 5 mM DTT, 2.5 mM EDTA]. Elution fractions were collected and analyzed by SDS-PAGE, pure fractions were pooled, and a 2:1 molar ratio of SRC-1 peptide was again added. The PXR LBD/peptide mixture was concentrated to 3 mg/mL, aliquoted, flash frozen in liquid nitrogen, and stored at −80°C.
PXR LBD (3 mg/mL, 83 μM) was mixed with 2 mM SJPYT-318 and incubated for 1 h at room temperature. This mixture contained 2% DMSO from the compound dilution. Hanging drop trays were set with 1 μL protein-ligand complex and 1 μL reservoir solutions containing 50 mM imidazole (pH 6.8-7.8) and 8-14% isopropanol. Crystals grew in various conditions within 1-3 days and were cryoprotected in respective mother liquors containing 1 mM SJPYT-318, 1% DMSO, and 40% ethylene glycol. The data presented were collected from a single crystal grown in 50 mM imidazole (pH 6.8) with 10% isopropanol.
X-ray diffraction data were collected to a resolution of 2.2 Å at Southeast Regional Collaborative Access Team (SER-CAT) 22-ID beamline at the Advanced Photon Source, Argonne National Laboratory. Frames were processed with XDS.48 The crystals belonged to space group P43212 with one molecule in the asymmetric unit. The structure was solved by molecular replacement in Phaser49 using PDB ID 1M13 as the search model.50 The search model was stripped of solvent and ligand prior to molecular replacement. Iterative cycles of model building and refinement were performed in Coot51 and Phenix.52 The data collection and refinement statistics are shown in Table 3. All crystallographic Figures were made in PyMOL (Schrodinger). The structure is deposited as PDB ID 8SZV.
QUANTIFICATION AND STATISTICAL ANALYSIS
All plots were made in GraphPad Prism 9. Results are expressed as the mean ± standard deviation from at least three independent experiments, and basic dose response curves were fitted as needed. To appropriately reflect both potency and efficacy, area under the curve (AUC) values were calculated in GraphPad Prism.
Supplementary Material
| REAGENT or RESOURCE | SOURCE | IDENTIFIER |
|---|---|---|
| Antibodies | ||
| LanthaScreen Tb-anti-GST Antibody | Thermo Fisher Scientific | Cat#PV3550 |
| Bacterial and virus strains | ||
| TurboCells BL21 (DE3) Competent E. coli | Genlantis | Cat#C302020 |
| Chemicals, peptides, and recombinant proteins | ||
| Dimethyl sulfoxide | Thermo Fisher Scientific | Cat#BP231-100 |
| T0901317 | Cayman Chemical | Cat#71810 |
| Rosiglitazone | MedChemExpress | Cat#HY-17386 |
| GW0742 | MedChemExpress | Cat#HY-13928 |
| SJPYT-299 | This paper | N/A |
| SJPYT-300 | This paper | N/A |
| SJPYT-301 | This paper | N/A |
| SJPYT-302 | Lin et al.27 | N/A |
| SJPYT-303 | This paper | N/A |
| SJPYT-304 | This paper | N/A |
| SJPYT-305 | This paper | N/A |
| SJPYT-315 | Lin et al.27 | N/A |
| SJPYT-316 | Lin et al.27 | N/A |
| SJPYT-317 | Lin et al.27 | N/A |
| SJPYT-318 | This paper | N/A |
| SJPYT-319 | Lin et al.27 | N/A |
| SJPYT-320 | This paper | N/A |
| Dulbecco's Modified Eagle's Medium (DMEM) | ATCC | Cat#30-2002 |
| RPMI-1640 Medium | ATCC | Cat#30-2001 |
| Leibovitz's L-15 Medium | ATCC | Cat#30-2008 |
| DMEM, high glucose, HEPES, no phenol red | Thermo Fisher Scientific | Cat#21063029 |
| Fetal bovine serum (FBS), Canadian Origin | Cytiva | Cat#SH30396.03 |
| HyClone Charcoal/Dextran treated FBS, US Origin | Cytiva | Cat#SH30068.03 |
| Hygromycin B (50 mg/mL) | Thermo Fisher Scientific | Cat#10687010 |
| Zeocin Selection Reagent | Thermo Fisher Scientific | Cat#R25001 |
| Geneticin Selective Antibiotic (G418 Sulfate) (50 mg/mL) | Thermo Fisher Scientific | Cat#10131027 |
| Matrigel Growth Factor Reduced Basement Membrane Matrix | Corning | Cat#356230 |
| Sodium Pyruvate (100 mM) | Thermo Fisher Scientific | Cat#11360070 |
| MEM Non-Essential Amino Acids Solution (100X) | Thermo Fisher Scientific | Cat#11140050 |
| NEBuilder HiFi DNA Assembly Master Mix | New England Biolabs | Cat#E2621S |
| KpnI-HF | New England Biolabs | Cat#R3142S |
| FAM-SRC1-B Peptide: FAM-(PEG)5-CPSSHSSLTERHKILHRLLQEGSPS-NH2 | Lin et al.46 | N/A |
| Magnesium chloride, 1M aq. soln., sterile-filtered | Thermo Fisher Scientific | Cat#J62411AD |
| Bovine Serum Albumin | Sigma-Aldrich | Cat#A3294 |
| Ethylenediamine Tetraacetic Acid (EDTA) | Thermo Fisher Scientific | E478-500 |
| Isopropanol | Thermo Fisher Scientific | Cat#A416-4 |
| Ethylene glycol | Sigma-Aldrich | Cat#102466-500ML |
| Pluronic F-127, 0.2 μm filtered (10% Solution in Water) | Thermo Fisher Scientific | Cat#P6866 |
| Lysozyme, Egg White | Gold Biotechnology | Cat#12650-88-3 |
| Terrific Broth, Modified, Granulated, 5 Kilograms | Research Products International | Cat#T15100-5000.0 |
| D-(+)-Glucose | Sigma-Aldrich | Cat#G8270 |
| SIGMAFAST Protease Inhibitor Cocktail Tablets, EDTA-Free | Sigma-Aldrich | Cat#S8830 |
| IPTG | Gold Biotechnology | Cat#I2481C100 |
| Ampicillin (Sodium), USP Grade | Gold Biotechnology | Cat#A-301-100 |
| DTT (Dithiothreitol) (> 99% pure) Protease free | Gold Biotechnology | Cat#DTT100 |
| Tris Base Ultra Pure [Tris (Hydroxymethyl) Aminomethane] | Research Products International | Cat#T60040-1000.0 |
| Sodium Chloride | Research Products International | Cat#S23020-500.0 |
| Glycerol, 99+%, extra pure | Thermo Fisher Scientific | Cat#158920025 |
| Imidazole, 99% | Thermo Fisher Scientific | Cat#122025000 |
| HisTrap FF | Cytiva | Cat#17525501 |
| HiLoad 26/600 Superdex 200 pg | Cytiva | Cat#28989336 |
| FuGENE HD Transfection Reagent | Promega | Cat#E2311 |
| ProxiPlate-384 Plus F, Black 384-shallow well Microplate | PerkinElmer | Cat#6008260 |
| CulturPlate-384, White Opaque 384-well Microplate, Sterile and Tissue Culture Treated | PerkinElmer | Cat#6007680 |
| PXR (SXR) Ligand Binding Domain Protein | Thermo Fisher Scientific | Cat#PV4841 |
| LXR-a Recombinant Human Protein, Ligand Binding Domain, GST-Tagged | Thermo Fisher Scientific | Cat#PV4657 |
| LXR-b Recombinant Human Protein, Ligand Binding Domain, GST-Tagged | Thermo Fisher Scientific | Cat#PV4660 |
| His-tagged PXR LBD | This paper | N/A |
| Unlabeled SRC-1 Peptide: N-CPSSHSSLTERHKILHRLLQEGSPS-C | This paper | N/A |
| Critical commercial assays | ||
| CellTiter-Glo Luminescent Cell Viability Assay | Promega | Cat#G7572 |
| steadylite plus Reporter Gene Assay System | PerkinElmer | Cat#6066751 |
| LiveBLAzer FRET-B/G Loading Kit with CCF4-AM | Thermo Fisher Scientific | Cat#K1096 |
| Maxwell 16 LEV simplyRNA Tissue Kit | Promega | Cat#AS1280 |
| SuperScript VILO cDNA Synthesis Kit | Thermo Fisher Scientific | Cat#11754050 |
| TaqMan Fast Advanced Master Mix | Thermo Fisher Scientific | Cat#4444557 |
| TaqMan gene expression assay for CYP3A4 | Thermo Fisher Scientific | Cat#Hs00604506_m1 |
| TaqMan gene expression assay for ABCA1 | Thermo Fisher Scientific | Cat#Hs01059118_m1 |
| TaqMan gene expression assay for GAPDH | Thermo Fisher Scientific | Cat#Hs03929097_g1 |
| TaqMan gene expression assay for RNA18S | Thermo Fisher Scientific | Cat#Hs03928990_g1 |
| Deposited data | ||
| Crystal structure of PXR LBD bound to SJPYT-318 | This paper | PDB ID 8SZV |
| Experimental models: Cell lines | ||
| 293T/17 Cells | ATCC | Cat#CRL-11268 |
| GeneBLAzer LXRα-UAS-bla HEK 293T Cells | Thermo Fisher Scientific | Cat#K1692 |
| GeneBLAzer LXRβ-UAS-bla HEK 293T Cells | Thermo Fisher Scientific | Cat#K1699 |
| GeneBLAzer PPARδ-UAS-bla HEK 293T Cells | Thermo Fisher Scientific | Cat#K1690 |
| GeneBLAzer PPARγ-UAS-bla HEK 293H Cells | Thermo Fisher Scientific | Cat#K1701 |
| SNU-C4 Cells | Korean Cell Line Bank | Cat#0000C4 |
| SNU81 Cells | Korean Cell Line Bank | Cat#00081 |
| SNU719 Cells | Korean Cell Line Bank | Cat#00719 |
| LS1034 Cells | ATCC | Cat#CRL-2158 |
| SNU620 Cells | Korean Cell Line Bank | Cat#00620 |
| SW948 Cells | ATCC | Cat#CCL-237 |
| SW1417 Cells | ATCC | Cat#CCL-238 |
| SW1463 Cells | ATCC | Cat#CCL-234 |
| Oligonucleotides | ||
| 5’-TCTAGAGCGGCCGCAGGTACCAGTGAACGGACAGGGACTC-3’ | Integrated DNA Technologies | N/A |
| 5’-GGCCTTAGTTATTCAGGTACCTCAGCTACCTGTGATGCC-3’ | Integrated DNA Technologies | N/A |
| Recombinant DNA | ||
| pG5luc | Promega | Cat#E2440 |
| pBIND | Promega | Cat#E2440 |
| pETDuet-1-PXR LBD-mSRC-1 | Lin et al.32 | N/A |
| pcDNA3-FLAG-PXR | Lin et al.45 | N/A |
| pBIND-PXR LBD | This paper | N/A |
| pBIND-FXR LBD | This paper | N/A |
| pBIND-RORγ LBD | This paper | N/A |
| Software and algorithms | ||
| XDS | Kabsch48 | https://xds.mr.mpg.de/ |
| Phenix | Liebschner et al.52 | https://phenix-online.org/ |
| Coot | Emsley et al.51 | http://www2.mrc-lmb.cam.ac.uk/personal/pemsley/coot/ |
| PyMOL | Schrödinger | https://pymol.org/2/ |
| Prism 9 | Graphpad Software | https://www.graphpad.com/features |
Highlights.
Analogs of a promiscuous nuclear receptor ligand are selective PXR agonists
Selectivity is achieved by utilizing PXR’s large spherical ligand binding pocket
Selective ligand binds PXR in new mode compared to parental compound
ACKNOWLEDGMENTS
Research reported in this publication was supported by ALSAC and NIH National Institute of General Medical Sciences [Grant R35GM118041]. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. X-ray crystallographic data were collected at Southeast Regional Collaborative Access Team (SER-CAT) 22-ID beamline at the Advanced Photon Source, Argonne National Laboratory. SER-CAT is supported by its member institutions, and equipment grants (S10_RR25528, S10_RR028976 and S10_OD027000) from the National Institutes of Health. Use of the Advanced Photon Source was supported by the U. S. Department of Energy, Office of Science, Office of Basic Energy Sciences, under Contract No. W-31-109-Eng-38.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
DECLARATION OF INTERESTS
The authors declare no competing interests.
REFERENCES
- 1.Lazar MA (2017). Maturing of the nuclear receptor family. J Clin Invest 127, 1123–1125. 10.1172/JCI92949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Chen T. (2008). Nuclear receptor drug discovery. Curr Opin Chem Biol 12, 418–426. 10.1016/j.cbpa.2008.07.001. [DOI] [PubMed] [Google Scholar]
- 3.Watson PA, Arora VK, and Sawyers CL (2015). Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer 15, 701–711. 10.1038/nrc4016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Weikum ER, Liu X, and Ortlund EA (2018). The nuclear receptor superfamily: A structural perspective. Protein Sci 27, 1876–1892. 10.1002/pro.3496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Moras D, and Gronemeyer H (1998). The nuclear receptor ligand-binding domain: structure and function. Curr Opin Cell Biol 10, 384–391. 10.1016/s0955-0674(98)80015-x. [DOI] [PubMed] [Google Scholar]
- 6.Hermanson O, Glass CK, and Rosenfeld MG (2002). Nuclear receptor coregulators: multiple modes of modification. Trends Endocrinol Metab 13, 55–60. 10.1016/s1043-2760(01)00527-6. [DOI] [PubMed] [Google Scholar]
- 7.Young CY, Montgomery BT, Andrews PE, Qui SD, Bilhartz DL, and Tindall DJ (1991). Hormonal regulation of prostate-specific antigen messenger RNA in human prostatic adenocarcinoma cell line LNCaP. Cancer Res 51, 3748–3752. [PubMed] [Google Scholar]
- 8.Flower RJ (1988). Eleventh Gaddum memorial lecture. Lipocortin and the mechanism of action of the glucocorticoids. Br J Pharmacol 94, 987–1015. 10.1111/j.1476-5381.1988.tb11614.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Blumberg B, Sabbagh W Jr., Juguilon H, Bolado J Jr., van Meter CM, Ong ES, and Evans RM (1998). SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Genes Dev 12, 3195–3205. 10.1101/gad.12.20.3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kliewer SA, Moore JT, Wade L, Staudinger JL, Watson MA, Jones SA, McKee DD, Oliver BB, Willson TM, Zetterstrom RH, et al. (1998). An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 92, 73–82. 10.1016/s0092-8674(00)80900-9. [DOI] [PubMed] [Google Scholar]
- 11.Lehmann JM, McKee DD, Watson MA, Willson TM, Moore JT, and Kliewer SA (1998). The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J Clin Invest 102, 1016–1023. 10.1172/JCI3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bertilsson G, Heidrich J, Svensson K, Asman M, Jendeberg L, Sydow-Backman M, Ohlsson R, Postlind H, Blomquist P, and Berkenstam A (1998). Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. Proc Natl Acad Sci U S A 95, 12208–12213. 10.1073/pnas.95.21.12208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hyder SM, Stancel GM, Nawaz Z, McDonnell DP, and Loose-Mitchell DS (1992). Identification of an estrogen response element in the 3'-flanking region of the murine c-fos protooncogene. J Biol Chem 267, 18047–18054. [PubMed] [Google Scholar]
- 14.Henley MJ, and Koehler AN (2021). Advances in targeting 'undruggable' transcription factors with small molecules. Nat Rev Drug Discov 20, 669–688. 10.1038/s41573-021-00199-0. [DOI] [PubMed] [Google Scholar]
- 15.Lambert SA, Jolma A, Campitelli LF, Das PK, Yin Y, Albu M, Chen X, Taipale J, Hughes TR, and Weirauch MT (2018). The Human Transcription Factors. Cell 172, 650–665. 10.1016/j.cell.2018.01.029. [DOI] [PubMed] [Google Scholar]
- 16.Davies M, Nowotka M, Papadatos G, Dedman N, Gaulton A, Atkinson F, Bellis L, and Overington JP (2015). ChEMBL web services: streamlining access to drug discovery data and utilities. Nucleic Acids Res 43, W612–620. 10.1093/nar/gkv352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mendez D, Gaulton A, Bento AP, Chambers J, De Veij M, Felix E, Magarinos MP, Mosquera JF, Mutowo P, Nowotka M, et al. (2019). ChEMBL: towards direct deposition of bioassay data. Nucleic Acids Res 47, D930–D940. 10.1093/nar/gky1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Nagy L, and Schwabe JW (2004). Mechanism of the nuclear receptor molecular switch. Trends Biochem Sci 29, 317–324. 10.1016/j.tibs.2004.04.006. [DOI] [PubMed] [Google Scholar]
- 19.Schultz JR, Tu H, Luk A, Repa JJ, Medina JC, Li L, Schwendner S, Wang S, Thoolen M, Mangelsdorf DJ, et al. (2000). Role of LXRs in control of lipogenesis. Genes Dev 14, 2831–2838. 10.1101/gad.850400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kumar N, Solt LA, Conkright JJ, Wang Y, Istrate MA, Busby SA, Garcia-Ordonez RD, Burris TP, and Griffin PR (2010). The benzenesulfoamide T0901317 [N-(2,2,2-trifluoroethyl)-N-[4-[2,2,2-trifluoro-1-hydroxy-1-(trifluoromethyl)ethyl]phenyl]-benzenesulfonamide] is a novel retinoic acid receptor-related orphan receptor-alpha/gamma inverse agonist. Mol Pharmacol 77, 228–236. 10.1124/mol.109.060905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mitro N, Vargas L, Romeo R, Koder A, and Saez E (2007). T0901317 is a potent PXR ligand: implications for the biology ascribed to LXR. FEBS Lett 581, 1721–1726. 10.1016/j.febslet.2007.03.047. [DOI] [PubMed] [Google Scholar]
- 22.Cheng J, Shah YM, and Gonzalez FJ (2012). Pregnane X receptor as a target for treatment of inflammatory bowel disorders. Trends Pharmacol Sci 33, 323–330. 10.1016/j.tips.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wang B, and Tontonoz P (2018). Liver X receptors in lipid signalling and membrane homeostasis. Nat Rev Endocrinol 14, 452–463. 10.1038/s41574-018-0037-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Poudel S, Huber AD, and Chen T (2023). Regulation of Nuclear Receptors PXR and CAR by Small Molecules and Signal Crosstalk: Roles in Drug Metabolism and Beyond. Drug Metab Dispos 51, 228–236. 10.1124/dmd.122.000858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cheng J, Shah YM, Ma X, Pang X, Tanaka T, Kodama T, Krausz KW, and Gonzalez FJ (2010). Therapeutic role of rifaximin in inflammatory bowel disease: clinical implication of human pregnane X receptor activation. J Pharmacol Exp Ther 335, 32–41. 10.1124/jpet.110.170225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sartor RB (2016). Review article: the potential mechanisms of action of rifaximin in the management of inflammatory bowel diseases. Aliment Pharmacol Ther 43 Suppl 1, 27–36. 10.1111/apt.13436. [DOI] [PubMed] [Google Scholar]
- 27.Lin W, Huber AD, Poudel S, Li Y, Seetharaman J, Miller DJ, and Chen T (2023). Structure-guided approach to modulate small molecule binding to a promiscuous ligand-activated protein. Proc Natl Acad Sci U S A 120, e2217804120. 10.1073/pnas.2217804120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Buchman CD, Chai SC, and Chen T (2018). A current structural perspective on PXR and CAR in drug metabolism. Expert Opin Drug Metab Toxicol 14, 635–647. 10.1080/17425255.2018.1476488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Svensson S, Ostberg T, Jacobsson M, Norstrom C, Stefansson K, Hallen D, Johansson IC, Zachrisson K, Ogg D, and Jendeberg L (2003). Crystal structure of the heterodimeric complex of LXRalpha and RXRbeta ligand-binding domains in a fully agonistic conformation. EMBO J 22, 4625–4633. 10.1093/emboj/cdg456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Williams S, Bledsoe RK, Collins JL, Boggs S, Lambert MH, Miller AB, Moore J, McKee DD, Moore L, Nichols J, et al. (2003). X-ray crystal structure of the liver X receptor beta ligand binding domain: regulation by a histidine-tryptophan switch. J Biol Chem 278, 27138–27143. 10.1074/jbc.M302260200. [DOI] [PubMed] [Google Scholar]
- 31.Ghandi M, Huang FW, Jane-Valbuena J, Kryukov GV, Lo CC, McDonald ER 3rd, Barretina J, Gelfand ET, Bielski CM, Li H, et al. (2019). Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508. 10.1038/s41586-019-1186-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lin W, Wang YM, Chai SC, Lv L, Zheng J, Wu J, Zhang Q, Wang YD, Griffin PR, and Chen T (2017). SPA70 is a potent antagonist of human pregnane X receptor. Nat Commun 8, 741. 10.1038/s41467-017-00780-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Huber AD, Li Y, Lin W, Galbraith AN, Mishra A, Porter SN, Wu J, Florke Gee RR, Zhuang W, Pruett-Miller SM, et al. (2022). SJPYT-195: A Designed Nuclear Receptor Degrader That Functions as a Molecular Glue Degrader of GSPT1. ACS Med Chem Lett 13, 1311–1320. 10.1021/acsmedchemlett.2c00223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Trabert B, Sherman ME, Kannan N, and Stanczyk FZ (2020). Progesterone and Breast Cancer. Endocr Rev 41, 320–344. 10.1210/endrev/bnz001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Blakely B, Shin S, and Jin K (2023). Overview of the therapeutic strategies for ER positive breast cancer. Biochem Pharmacol 212, 115552. 10.1016/j.bcp.2023.115552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Xie Y, Xu M, Deng M, Li Z, Wang P, Ren S, Guo Y, Ma X, Fan J, Billiar TR, and Xie W (2019). Activation of Pregnane X Receptor Sensitizes Mice to Hemorrhagic Shock-Induced Liver Injury. Hepatology 70, 995–1010. 10.1002/hep.30691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang J, Bwayi M, Gee RRF, and Chen T (2020). PXR-mediated idiosyncratic drug-induced liver injury: mechanistic insights and targeting approaches. Expert Opin Drug Metab Toxicol 16, 711–722. 10.1080/17425255.2020.1779701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, Qiu Z, Maher L, Redinbo MR, Phillips RS, et al. (2014). Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity 41, 296–310. 10.1016/j.immuni.2014.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shah YM, Ma X, Morimura K, Kim I, and Gonzalez FJ (2007). Pregnane X receptor activation ameliorates DSS-induced inflammatory bowel disease via inhibition of NF-kappaB target gene expression. Am J Physiol Gastrointest Liver Physiol 292, G1114–1122. 10.1152/ajpgi.00528.2006. [DOI] [PubMed] [Google Scholar]
- 40.Kopecky DJ, Jiao XY, Fisher B, McKendry S, Labelle M, Piper DE, Coward P, Shiau AK, Escaron P, Danao J, et al. (2012). Discovery of a new binding mode for a series of liver X receptor agonists. Bioorg Med Chem Lett 22, 2407–2410. 10.1016/j.bmcl.2012.02.028. [DOI] [PubMed] [Google Scholar]
- 41.Fauber BP, de Leon Boenig G, Burton B, Eidenschenk C, Everett C, Gobbi A, Hymowitz SG, Johnson AR, Liimatta M, Lockey P, et al. (2013). Structure-based design of substituted hexafluoroisopropanol-arylsulfonamides as modulators of RORc. Bioorg Med Chem Lett 23, 6604–6609. 10.1016/j.bmcl.2013.10.054. [DOI] [PubMed] [Google Scholar]
- 42.Hall A, Chanteux H, Menochet K, Ledecq M, and Schulze MED (2021). Designing Out PXR Activity on Drug Discovery Projects: A Review of Structure-Based Methods, Empirical and Computational Approaches. J Med Chem 64, 6413–6522. 10.1021/acs.jmedchem.0c02245. [DOI] [PubMed] [Google Scholar]
- 43.Xue Y, Chao E, Zuercher WJ, Willson TM, Collins JL, and Redinbo MR (2007). Crystal structure of the PXR-T1317 complex provides a scaffold to examine the potential for receptor antagonism. Bioorg Med Chem 15, 2156–2166. 10.1016/j.bmc.2006.12.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Farnegardh M, Bonn T, Sun S, Ljunggren J, Ahola H, Wilhelmsson A, Gustafsson JA, and Carlquist M (2003). The three-dimensional structure of the liver X receptor beta reveals a flexible ligand-binding pocket that can accommodate fundamentally different ligands. J Biol Chem 278, 38821–38828. 10.1074/jbc.M304842200. [DOI] [PubMed] [Google Scholar]
- 45.Lin W, Wu J, Dong H, Bouck D, Zeng FY, and Chen T (2008). Cyclin-dependent kinase 2 negatively regulates human pregnane X receptor-mediated CYP3A4 gene expression in HepG2 liver carcinoma cells. J Biol Chem 283, 30650–30657. 10.1074/jbc.M806132200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lin W, and Chen T (2018). Using TR-FRET to Investigate Protein-Protein Interactions: A Case Study of PXR-Coregulator Interaction. Adv Protein Chem Struct Biol 110, 31–63. 10.1016/bs.apcsb.2017.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Livak KJ, and Schmittgen TD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408. 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]
- 48.Kabsch W (2010). Xds. Acta Crystallogr D Biol Crystallogr 66, 125–132. 10.1107/S0907444909047337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.McCoy AJ, Grosse-Kunstleve RW, Adams PD, Winn MD, Storoni LC, and Read RJ (2007). Phaser crystallographic software. J Appl Crystallogr 40, 658–674. 10.1107/S0021889807021206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Watkins RE, Maglich JM, Moore LB, Wisely GB, Noble SM, Davis-Searles PR, Lambert MH, Kliewer SA, and Redinbo MR (2003). 2.1 A crystal structure of human PXR in complex with the St. John's wort compound hyperforin. Biochemistry 42, 1430–1438. 10.1021/bi0268753. [DOI] [PubMed] [Google Scholar]
- 51.Emsley P, Lohkamp B, Scott WG, and Cowtan K (2010). Features and development of Coot. Acta Crystallogr D Biol Crystallogr 66, 486–501. 10.1107/S0907444910007493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liebschner D, Afonine PV, Baker ML, Bunkoczi G, Chen VB, Croll TI, Hintze B, Hung LW, Jain S, McCoy AJ, et al. (2019). Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr D Struct Biol 75, 861–877. 10.1107/S2059798319011471. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The structure of SJPYT-318-bound PXR LBD was deposited in the Protein Data Bank (PDB) under accession code 8SZV.
This paper does not report original code.
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.






