Screening Method for the Identification of Compounds That Activate Pregnane X Receptor

Caitlin Lynch, Caitlin Lynch, Srilatha Sakamuru, Srilatha Sakamuru, Menghang Xia, Menghang Xia

Published: 2022-12-05 DOI: 10.1002/cpz1.615

Abstract

The pregnane X receptor (PXR) is a nuclear receptor found mainly in the liver and intestine, whose main function is to regulate the expression of drug-metabolizing enzymes and transporters. Recently, it has been noted that PXR plays critical roles in energy homeostasis, immune response, and cancer. Therefore, identifying chemicals or compounds that can modulate PXR is of great interest, as these can result in downstream toxicity or, alternatively, may have therapeutic potential. Testing one compound at a time for PXR activity would be inefficient and take thousands of hours for large compound libraries. Here, we describe a high-throughput screening method that encompasses plating and treating HepG2-CYP3A4-hPXR cells in a 1536-well plate, as well as reading and interpreting assay (e.g., luciferase reporter gene activity) endpoints. These cells are stably transfected with a human PXR expression vector and CYP3A4 -promoter-driven luciferase reporter vector, allowing the identification of compounds that activate PXR through cytochrome 450 3A4. We also describe how to analyze the data from each assay and explain follow-up steps, namely pharmacological characterization and quantitative polymerase chain reaction (qPCR) assays, which can be performed to confirm results from the original screen. These methods can be used to identify and confirm hPXR activators after completion of a compound screening. Published 2022. This article is a U.S. Government work and is in the public domain in the USA. Current Protocols published by Wiley Periodicals LLC.

Basic Protocol 1 : Establishment of a high-throughput assay to identify hPXR activators

Basic Protocol 2 : Quantitative high-throughput screening a compound library to classify hPXR activators

Basic Protocol 3 : Performing pharmacological characterization and qPCR assays to confirm hPXR activators

INTRODUCTION

Nuclear receptors belong to an important family of proteins that act as transcription factors regulating the expression of genes involved in metabolism, cell growth, and development (Schulman, 2010). In particular, the pregnane X receptor (PXR) controls drug-metabolizing enzymes (DMEs) and plays roles in energy homeostasis, inflammation, and cell proliferation (Kliewer et al., 1998; Kumar & Surapaneni, 2001; Sueyoshi & Negishi, 2001). Given the increasing amounts of chemicals being released into the atmosphere each year, alongside the novel drugs being produced, the development of quick and easy approaches to identify toxic and organism-interacting compounds is becoming increasingly relevant. As such, it has become of great interest to identify compounds that can modulate the induction or activity of PXR.

Low-throughput assays that test only a few compounds at a time are currently being used. It is relatively easy and accurate to transfect PXR expression and CYP3A4 promoter–luciferase vectors into each well of a 6-well, 12-well, or 24-well plate. However, without a high-throughput screening method, it would take years to test every chemical for PXR activation. Recently, a quantitative high-throughput method to identify chemicals that can act as human PXR activators has been reported (Lynch et al., 2021). This assay utilizes a double-stable cell line including the previously mentioned vectors for PXR and CYP3A4. The HepG2-CYP3A4-hPXR cell line (Lin et al., 2008) generates an easy-to-work with and ideal model for screening compounds in a high-throughput manner, producing the results in days, not years.

In this article, we describe how to set up a high-throughput screen, using the previously reported method (Lynch et al., 2021), so that a quick and accurate classification of PXR activators can be made. We also provide steps to perform follow-up studies to ascertain the validity of the high-throughput screening data. By identifying PXR activators, therapeutic applications, potential drug-drug interactions, or other toxic implications can be identified.

To identify and confirm a compound as a PXR activator, the following procedure (Fig. 1) should be followed. Basic Protocol 1 explains how to optimize a high-throughput cell-based luciferase reporter gene assay to identify PXR activators from a large library of compounds. The next logical step is to execute the high-throughput screen itself (Basic Protocol 2). This can be done using 96-, 384-, or 1536-well plates so that many compounds can be thoroughly examined in one efficient assay. Finally, Basic Protocol 3 portrays two different follow-up studies, including a pharmacological assay that can be used to measure the shift in potency, after the addition of a selective PXR antagonist, of the studied compound and a real-time quantitative polymerase chain reaction (RT-qPCR) to measure the induction of PXR's most important drug metabolizing enzyme, CYP3A4. These two assays are performed in order to confirm the compounds identified in Basic Protocol 2 are true PXR activators.

Overview of the protocol steps described throughout this article for the identification of hPXR activators. Words in blue denote action steps, while the words to the right of each down arrow indicate endpoints used for narrowing down the number of compounds in each step. NR, nuclear receptor. This image was partially created with BioRender (biorender.com).
Overview of the protocol steps described throughout this article for the identification of hPXR activators. Words in blue denote action steps, while the words to the right of each down arrow indicate endpoints used for narrowing down the number of compounds in each step. NR, nuclear receptor. This image was partially created with BioRender (biorender.com).

Basic Protocol 1: ESTABLISHMENT OF A HIGH-THROUGHPUT ASSAY TO IDENTIFY hPXR ACTIVATORS

Optimization of a new assay can be time-consuming and sometimes challenging; to overcome these difficulties, the following protocol describes a step-by-step methodology for generating an in vitro high-throughput assay, employing luciferase reporter gene technology, to identify PXR activators. The first steps describe thawing and culturing a double-stable cell line that includes an hPXR expression vector and CYP3A4-luciferase plasmid (Lin et al., 2008). A double-stable cell line is preferred so that transient transfection does not need to be performed; eliminating this decreases the experiment time and creates more consistent data by generating a uniform population of cells containing both vectors in each well of the assay plate. Every laboratory needs to optimize the assay by testing a different number of cells/well, positive control concentration, and/or equipment settings. Therefore, an optimization step to address each of these items is necessary. Here, we show how to test multiple concentrations of cells/well and explain how to determine the optimal conditions for the assay. Once the optimization is complete, the assay will be ready for use in a high-throughput manner and ready to screen hundreds or thousands of compounds in one experiment.

NOTE : This protocol must be completed in a biological safety cabinet, to keep contamination to a minimum.

Materials

  • Eagle's Minimum Essential Medium (EMEM; ATCC, cat. no. 30-2003)
  • HyClone Characterized Fetal Bovine Serum (FBS; Cytiva, cat. no. SH30071.03)
  • 10,000 U/ml Penicillin-Streptomycin (Pen-Strep; ThermoFisher Scientific, cat. no. 15140122)
  • HepG2-CYP3A4-hPXR stable cell line (Dr. Taosheng Chen, Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital; Lin et al., 2008)
  • 70% (v/v) ethanol
  • PXR culture medium (see recipe), prewarmed to 37°C
  • 0.25% trypsin-EDTA with phenol red (ThermoFisher Scientific, cat. no. 25200056)
  • Phosphate-buffered saline (PBS), pH 7.4, calcium chloride and magnesium chloride free (ThermoFisher Scientific, cat. no. 10010072)
  • PXR assay medium (see recipe), prewarmed to 37°C
  • Dimethyl sulfoxide (DMSO; Millipore Sigma, cat. no. 472301, CAS no. 67-68-5)
  • Rifampicin (MilliporeSigma, cat. no. R3501, CAS no. 13292-46-1)
  • Tetraoctylammonium bromide (Millipore Sigma, cat. no. 294136, CAS no. 14866-33-2)
  • ONE-Glo Luciferase Assay System (Promega, cat. no. E6130)
  • Biological safety cabinet (Labconco, cat. no. 302610101, or equivalent)
  • Vacuum filter system, 500 ml (Corning Life Sciences, cat. no. 431205)
  • Isotemp water bath (Fisher Scientific, cat. no. FSGPD20) or equivalent
  • 15-ml tubes (ThermoFisher Scientific, cat. no. 339651, or equivalent)
  • 200-µl micropipettor (Rainin, cat. no. 17008652, or equivalent)
  • 200-µl pipet tips (Rainin, cat. no. 30389268)
  • 10-ml pipets (Corning Life Sciences, cat. no. 357551, or equivalent)
  • Centrifuge (ThermoFisher Scientific, cat. no. 75004521, or equivalent)
  • Pipettor (Integra Biosciences, cat. no. 155019, or equivalent)
  • 225-cm2 (T-225) cell culture flasks (Corning Life Sciences, cat. no. 431082), Millicell HY 5-layer T-1000 cell culture flasks (Millipore Sigma, cat. no. PFHYS1008), or 175-cm2 collagen-I-coated flasks (ThermoFisher Scientific, cat. no. 132708)
  • CO2 incubator (ThermoFisher Scientific, cat. no. 3310, or equivalent)
  • Tissue culture microscope (Carl Zeiss Microscopy, LLC, cat. no. 491237-0013-000, or equivalent)
  • Cell counter (Nexcelom Biosciences, cat. no. Cellometer Auto T4-IQOQ, or equivalent)
  • White/solid 1536-well flat-bottom cell-culture-treated plate (Greiner Bio-One, cat. no. 789173-F)
  • BioRAPTR2 Flying Reagent Liquid Dispenser (Let's Go Robotics, Inc., model no. BR2) or equivalent
  • Microplate lids (Wako Automation, cat. no. 08-3241-0005)
  • Clear 1536-well compound plates (Greiner Bio-One, cat. no. 789270-C)
  • Pintool Station (Wako Automation, cat. no. Pintool1144-3100 PLUS VELOCITY) or equivalent
  • ViewLux uHTS Microplate Imager 1430-0010A (Perkin Elmer, cat. no. 26149) or equivalent
  • GraphPad Prism Software (or equivalent)

Thawing HepG2-CYP3A4-hPXR cells

1.In the biological safety cabinet, set up a 500-ml vacuum filter system. Prepare thaw medium by combining 500 ml EMEM, 50 ml FBS, and 5 ml Pen-Strep and filter the liquid.

Note
Thaw medium can be stored up to 1 month at 4°C.

Note
Every item going in and out of the biological safety cabinet should be wiped down with 70% ethanol to minimize contamination.

2.Warm thaw medium to 37°C using a water bath.

3.In the biological safety cabinet, put 9 ml warmed thaw medium into a 15-ml tube.

Note
A different amount of medium can be used as long as you later balance that amount in the centrifuge.

4.Take a single vial of frozen HepG2-CYP3A4-hPXR cells and place in a 37°C water bath for ∼3 min or until the mix starts to thaw.

Note
The abrupt temperature shift from −150°C to 37°C can be damaging to some of the cells. Limiting the thaw time in the water bath can help alleviate this effect. If using a 175-cm2 flask or larger, ensure that at least 10 million cells are being thawed.

5.Wipe the vial with 70% ethanol and then, in the safety cabinet, pour the cell solution into the 9 ml of warmed thaw medium. Remove any remaining liquid in the vial and transfer into the 15-ml tube, using a 200-µl micropipettor or similar.

Note
Any small pipet will suffice as long as the tip is small enough to get into the bottom of the vial.

6.Pipet up and down five to ten times with a 10-ml pipet to break up clumps of cells.

7.Centrifuge cells 4 min at 900 rpm (100 × g), room temperature.

Note
Because high speeds are being used, remember to balance the centrifuge properly.

8.Pour off supernatant from cell pellet into a waste bin and add 10 ml fresh prewarmed thaw medium. Pipet cells up and down at least ten times using a 10-ml pipet. Pour this cell suspension into a 175-cm2 collagen-I-coated flask and swirl around slightly.

Note
A collagen-coated flask is used for more optimal attachment of cells and can even improve end results.

9.Incubate at 37°C, 5% CO2, for a few days, or until cells are at least 80% confluent.

Culturing HepG2-CYP3A4-hPXR cells

10.Warm up PXR culture medium in a 37°C water bath. Let trypsin solution warm up to room temperature.

11.Pour off used medium from the HepG2-CYP3A4-hPXR cells flask into a waste bin.

12.Add 10 ml PBS to the flask and swirl around. Pour off PBS into waste bin. Repeat two times.

Note
This is a very important step. All the debris needs to be washed away with PBS that does not contain calcium chloride (CaCl2) or magnesium chloride (MgCl2) before the trypsin is added. If no PBS is used, or PBS with CaCl2 and MgCl2 is used, the cells will not detach from the flask properly.

13.Add 6 ml trypsin solution to the flask and swirl around to cover the entire flask. Place the flask in the incubator for 3-5 min until cells start to detach from flask.

14.Add 4 ml prewarmed PXR culture medium to the flask. Swirl or pipet liquid around to release and gather as many cells as possible. Suck up cells and medium and transfer into a 50-ml tube.

15.Centrifuge 4 min at 900 rpm (100 × g), room temperature.

16.Pour off supernatant and resuspend cells in 10 ml fresh prewarmed PXR culture medium. Pipet up and down at least ten times to break up clumps.

17.Acquire a fresh 175-cm2 collagen-I-coated flask and pour 40 ml fresh prewarmed PXR culture medium into the flask.

18.Count the cells and transfer ∼20 million cells into the flask. Place flask in the 37°C, 5% CO2 incubator and incubate until the cells become ∼80% confluent.

Note
There may be cell debris over the next few days as the selection antibiotic starts to work and kill the cells lacking the correct plasmid. If there appear to be a lot of floating cells, it may be prudent to pour off the medium and add fresh culture medium to the flask.

19.Culture cells for at least another passage by repeating steps 11-19.This is to ensure that the highest number of the correct cells are present.

Note
Usually, these cells need to be subcultured every 3-4 days. Once they are 80% confluent, they should be used for the assay or passaged again.

Performing optimization assay

20.Warm assay medium to 37°C using a water bath.

21.Repeat steps 12-17 using prewarmed PXR assay medium instead of culture medium. Count the newly made cell suspension.

22.Generate 5.0 × 105 cells/ml and 9.0 × 105 cells/ml solutions using PXR assay medium.

23.Plate 5 µl of each suspension into the first four columns of white 1536-well plates using a liquid dispenser.

Note
In this example, the number of cells in plate 1 should be ∼2500 cells/well, while plate 2 should have 4500 cells/well. This cell number is one of the parameters which will be optimized to fit the needs of the experiment. These assays can also be adapted for plates with fewer wells; an optimization and scale-up will need to be performed.

24.Place a microplate lid on top of each assay plate and incubate the plates at 37°C, 5% CO2 for ∼4-5 hr.

Note
This is done so that the cells have adequate time to attach to the bottom of each well.

25.Create a positive control compound plate for the assay using DMSO, rifampicin, and tetraoctylammonium bromide in the first four columns of a fresh clear compound plate. Store at −20°C when not in use.

Note
An example of this can be seen in Figure 2 (top left plate) and would contain a duplicate 1:2 dilution of rifampicin starting at 20 mM (final concentration of 92 µM) in column 1; the upper half of column 2 then contains a single dose of rifampicin at 10 mM (final concentration of 46 µM), the lower half of column 2 contains a single dose of rifampicin at 5 mM (final concentration of 23 µM), the upper half of column 3 and all of column 4 contain DMSO, and the lower half of column 3 contains tetraoctylammonium bromide at 20 mM (final concentration of 92 µM). The tetraocylammonium bromide only needs to be added to the control plate if a cytotoxicity assay is being performed in tandem, but can be omitted if this control plate will not be used for another assay. Either of the single dose concentrations of rifampicin can be used as the positive control—whichever concentration gives the highest signal. These two concentrations can be optimized for each laboratory's own use.

Compound plate map for treating cells with controls. When combining a positive control and cells into one assay plate, the pintool is used to automatically add 23 nl of positive control liquid into each well already containing cells. The light pink wells in the assay plate (top right) represent plated cells which have already attached to the bottom. The dark red wells in column 1 of the control compound plate (top left) represent the dose-response (DR) dilution series of rifampicin, the positive control. The dark pink and medium pink wells in column 2 represent the single concentrations of rifampicin used in the assay—10 mM and 5 mM, respectively. The black wells in the bottom of column 3 represent 20 mM tetraoctylammonium bromide, while the green wells in columns 3 and 4 represent the DMSO-only negative control wells. A picture of the pintool station is also included in the middle of the figure. This image was created with BioRender (biorender.com).
Compound plate map for treating cells with controls. When combining a positive control and cells into one assay plate, the pintool is used to automatically add 23 nl of positive control liquid into each well already containing cells. The light pink wells in the assay plate (top right) represent plated cells which have already attached to the bottom. The dark red wells in column 1 of the control compound plate (top left) represent the dose-response (DR) dilution series of rifampicin, the positive control. The dark pink and medium pink wells in column 2 represent the single concentrations of rifampicin used in the assay—10 mM and 5 mM, respectively. The black wells in the bottom of column 3 represent 20 mM tetraoctylammonium bromide, while the green wells in columns 3 and 4 represent the DMSO-only negative control wells. A picture of the pintool station is also included in the middle of the figure. This image was created with BioRender (biorender.com).

26.Transfer 23 nl from each well of the positive control plate to the cell plate using a Wako Pintool station. Incubate at 37°C, 5% CO2 for 24 hr.

Note
Figure 3 shows the proper confluency and healthiness of this cell line, plated in a 1536-well plate after a 23-hr treatment with DMSO.

Morphology of healthy HepG2-hPXR-CYP3A4 cells. HepG2 cells were plated in assay medium at 2500 cells/well in a 1536-well plate and allowed to attach overnight. The microscope picture was taken at 10×.
Morphology of healthy HepG2-hPXR-CYP3A4 cells. HepG2 cells were plated in assay medium at 2500 cells/well in a 1536-well plate and allowed to attach overnight. The microscope picture was taken at 10×.

27.Add 4 µl ONE-Glo reagent to each well including cells (first four columns of plates 1 and 2). Incubate at room temperature for 30 min.

Note
We assessed both ONE-Glo and Reagent 2 (from a different kit) luminescent reagents to determine which exhibited optimal performance in our conditions (Fig. 4). Different luciferase reagents can be studied to establish which works best in each laboratory.

Comparison of luminescent reagents. HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of rifampicin, and luminescence values were identified after the addition of either ONE-Glo or Reagent 2. Each curve represents the mean ± SD of duplicate wells. M, molar concentration.
Comparison of luminescent reagents. HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of rifampicin, and luminescence values were identified after the addition of either ONE-Glo or Reagent 2. Each curve represents the mean ± SD of duplicate wells. M, molar concentration.

28.Acquire the luminescence signal from the plate using a plate reader.

Note
If using a ViewLux IMAGER, example settings are as follow: 60 s readout, low speed, high gain, 2× binning. These numbers will need to be optimized depending on the specific assay and instrument used.

29.Analyze the data using the equation below, and generate the % activity curve using GraphPad (Fig. 5).

Note
Use the following equation to analyze the concentration response curves: % activity = [(Vcompound – VDMSO)/(Vrifampicin – VDMSO)]*100, where Vcompound equals the well values, Vrifampicin denotes the median value of the 16 wells of rifampicin at one concentration, and VDMSO represents the median values of the DMSO-only wells. Remember to transform the concentrations (x axis) into log(x) values to have correct graphical representation. To do this in GraphPad, once the molar concentrations are typed in with their respective % activity data, press the analyze button and click transform. When the new window pops up, click the box marked “Transform X values using.” Toggle down to X = Log(X) and click ok.

Optimization of cell number per well in 1536-well plates. HepG2-hPXR-CYP3A4 cells were plated with 2500 cells/well or 4500 cells/well. Luminescence values were determined after the rifampicin treatment, and the % activity was calculated by comparing each data point to that of the positive control, RIF (100% = RIF activity). Each curve represents the mean ± SD of triplicate wells. M, molar concentration.
Optimization of cell number per well in 1536-well plates. HepG2-hPXR-CYP3A4 cells were plated with 2500 cells/well or 4500 cells/well. Luminescence values were determined after the rifampicin treatment, and the % activity was calculated by comparing each data point to that of the positive control, RIF (100% = RIF activity). Each curve represents the mean ± SD of triplicate wells. M, molar concentration.

30.Acquire an EC50 for all compounds.

Note
Once % activity values are put into GraphPad and the concentrations are transformed into log values, click analyze again. Under the XY analyses topic, click “Nonlinear regression (curve fit).” When the new window pops up, use the “log(agonist) versus response – Variable slope (four parameters)” and click ok. Once the analysis comes up, copy the EC50 values onto your graph. Data should be in a sigmoidal shape with an actual number as an EC50, not an ambiguous one (∼). When analyzing the curve for the positive control, the top of the curve should have a value of ∼100% because the positive-control dose-response numbers are being compared to a single concentration of the same compound (rifampicin).

31.Choose the optimum cell number, based on the % activity curve and EC50.

Note
Based on Figure 5, 2500 cells/well gives an EC50 of 2.437 µM while 4500 cells/well gives an ambiguous ∼190.4 M EC50. The curvature is also sigmoidal for the 2500 cells/well but not for the 4500 cells/well. Therefore, the cell number that should be used for Basic Protocol 2 is 2500 cells/well.

Basic Protocol 2: QUANTITATIVE HIGH-THROUGHPUT SCREENING OF A COMPOUND LIBRARY TO CLASSIFY hPXR ACTIVATORS

Once the assay has been optimized according to Basic Protocol 1, a high-throughput screen can be conducted. Profiling thousands of compounds at one time will reduce total experiment time and reagent supplies. In essence, Basic Protocol 2 is the same assay as that completed in Basic Protocol 1, just on a larger scale. This protocol describes the steps in performing a high-throughput luciferase reporter gene assay to identify hPXR activators from a large library of compounds. After the screening is complete, the potency and efficacy of each compound tested can be calculated. The potential PXR agonists identified from the screen can be further examined in the confirmation assays (covered in Basic Protocol 3). This protocol will also explain how to identify the most potent and efficacious compounds by calculating the maximum response and EC50 values; however, there is a need to account for the shape of the concentration curve as well, previously explained in a methods chapter (Huang, 2016).

NOTE : This protocol must be completed in a biological safety cabinet, to keep contamination to a minimum.

Materials

  • HepG2-CYP3A4-hPXR stable cell line (Dr. Taosheng Chen, Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital; Lin et al., 2008)
  • PXR assay medium (see recipe)
  • Phenol-red-free Dulbecco's Modified Eagle's Medium (DMEM; ThermoFisher Scientific, cat. no. 31053028)
  • FBS, charcoal stripped (ThermoFisher Scientific, cat. no. 12676029)
  • 100 mM sodium pyruvate (ThermoFisher Scientific, cat. no. 11360070)
  • 200 mM l-glutamine (ThermoFisher Scientific, cat. no. 25030081)
  • Positive control plate (Basic Protocol 1, step 26)
  • Compound library plates: e.g., Tox21 10K compound library plates (Attene-Ramos et al., 2013)
  • ONE-Glo Luciferase Assay System (Promega, cat. no. E6130)
  • Biological safety cabinet (Labconco, cat. no. 302610101, or equivalent)
  • Isotemp water bath (Fisher Scientific, cat. no. FSGPD20) or equivalent
  • CO2 incubator (ThermoFisher Scientific, cat. no. 3310, or equivalent)
  • 50-ml tubes (Corning Life Sciences, cat. no. 352070, or equivalent)
  • Centrifuge (ThermoFisher Scientific, cat. no. 75004521, or equivalent)
  • 225-cm2 (T-225) cell culture flasks (Corning Life Sciences, cat. no. 431082), Millicell HY 5-layer T-1000 cell culture flasks (Millipore Sigma, cat. no. PFHYS1008), or 175-cm2 collagen-I-coated flasks (ThermoFisher Scientific, cat. no. 132708)
  • 10-ml pipets (Corning Life Sciences, cat. no. 357551, or equivalent)
  • Pipettor (Integra Biosciences, cat. no. 155019, or equivalent)
  • Tissue culture microscope (Carl Zeiss Microscopy, LLC, cat. no. 491237-0013-000, or equivalent)
  • Cell counter (Nexcelom Biosciences, cat. no. Cellometer Auto T4-IQOQ, or equivalent)
  • White/solid 1536-well flat-bottom cell-culture-treated plate (Greiner Bio-One, cat. no. 789173-F)
  • Multidrop™ Combi Reagent Dispenser (ThermoFisher Scientific, cat. no. 5840300), or equivalent
  • Microplate lids (Wako Automation, cat. no. 08-3241-0005)
  • Pintool Station (Wako Automation, cat. no. Pintool1144-3100 PLUS VELOCITY), or equivalent
  • ViewLux Plate Reader (Perkin Elmer, cat. no. 1430-0010A, or equivalent)
  • GraphPad Prism Software (or equivalent)

1.Warm assay medium to 37°C using a water bath. Let trypsin warm up to room temperature.

2.Perform steps 12-17 of Basic Protocol 1, using assay medium instead of culture medium. Count the newly made cell suspension.

3.Generate a 5.0 × 105 cell/ml solution. Plate 5 µl of cell suspension into every well of solid white 1536-well plates using a multidrop liquid dispenser. Place a microplate lid on top of each plate, place the plates in a 37°C, 5% CO2 incubator, and incubate ∼4-5 hr.

Note
Plate as many plates as the chosen library requires. Perform triplicate experiments for each compound plate when possible so that proper statistics and more robust data can be attained.

4.Transfer 23 nl of the previously generated positive control plate from Basic Protocol 1, step 26, as well as 23 nl from the compound library plates, to the cell plate using a Wako Pintool station (Fig. 6).

Note
Library plates should have compounds in columns 5-48 only. The assay plates should contain compounds from the control plates in columns 1-4 and compounds from the library plates in columns 5-48. If performing concentration response curves, the first library plate should have the lowest concentration of each compound, the second plate should have the next highest concentration, and so forth. When triplicate experiments are performed, a plate with only DMSO-treated cells (23 nl, equaling a final value of 0.46% DMSO in the well) should be placed in between replicates to ensure that there is no crossover of compound before the next batch is started; this will produce more accurate data.

Flow chart for compound treatment. A pintool station combines 23 nl of liquid from the first four columns of the positive control plate (top left) and columns 5-48 (top right) of the compound plate and transfers it into the assay plate (middle), which already has attached cells inside, to generate a combined assay/compound plate (bottom). This image was created with BioRender (biorender.com).
Flow chart for compound treatment. A pintool station combines 23 nl of liquid from the first four columns of the positive control plate (top left) and columns 5-48 (top right) of the compound plate and transfers it into the assay plate (middle), which already has attached cells inside, to generate a combined assay/compound plate (bottom). This image was created with BioRender (biorender.com).

5.Incubate at 37°C, 5% CO2 for 24 hr.

6.Follow steps 28-31 of Basic Protocol 1.

Note
Hundreds or thousands of compounds can be assayed in one experiment if a robot is available. Lack of access to a robot will be a rate-limiting step in many laboratories. The data should generate curves, as in Figure 5, for each compound screened.

7.Select positive compounds as well as a few negative compounds. Repeat assay with newly formed compound plates comprising those selected compounds.

Note
Identify compounds with high efficacy (maximum response) and low potency (EC50) values (Huang, 2016). In GraphPad, the maximum response value is listed as “Top.” An example of this narrowing down is to use an efficacy value of >80% of rifampicin's value (positive control) with a potency value of <10 µM. However, these values can be adjusted based on the results of each assay. It is important to include some negative test compounds as well to confirm the validity of the assay.

8.Proceed to Basic Protocol 3 to complete follow-up studies on a small collection of the most promising compounds.

Note
The number of selected compounds should relate to the ability of the laboratory to complete each follow-up assay. The selection of compounds should be based on highest efficacy, lowest potency, pricing, commercial availability, and practical or clinical importance of compounds.

Basic Protocol 3: PERFORMING PHARMACOLOGICAL CHARACTERIZATION AND qPCR ASSAYS TO CONFIRM hPXR ACTIVATORS

Once a manageable group of compounds has been chosen after a primary screening, such as in Basic Protocol 2, additional studies can be conducted to further confirm compound modulation of PXR. Here we first discuss how to perform a pharmacological study, which will display a shift in EC50 values and concentration response curves if the compound's activity is due to PXR activation. If, after the addition of increasing amounts of PXR antagonist (SPA70; Lin et al., 2008), a compound's EC50 value becomes higher in conjunction with a right-shifting concentration response curve, the compound is further confirmed as a PXR activator. The second section demonstrates how to perform an RT-qPCR assay to identify the induction or inhibition of the mRNA expression of a particular gene. In this case, we are looking at the main DME that PXR regulates, CYP3A4.If a compound activates PXR, it should also increase the expression level of CYP3A4. The combination of these assays should corroborate the previous data, from Basic Protocol 2, and a safe conclusion can be made to identify a compound as a true PXR activator.

NOTE : This protocol must be completed in a biological safety cabinet, to keep contamination to a minimum.

Materials

  • Candidate compounds (Basic Protocol 2, step 7)
  • HepG2-CYP3A4-hPXR stable cell line (Dr. Taosheng Chen, Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital; Lin et al., 2008)
  • PXR assay medium (see recipe)
  • Dimethyl sulfoxide (DMSO; Millipore Sigma, cat. no. 472301, CAS no. 67-68-5)
  • SPA70 (Millipore Sigma, cat. no. SML2662, CAS no. 931314-31-7)
  • ONE-Glo Luciferase Assay System (Promega, cat. no. E6130)
  • William's E Medium, phenol red free (ThermoFisher Scientific, cat. no. A1217601)
  • HepaRG™ Thawing and Plating Medium Supplement (Lonza Bioscience, cat. no. MHTAP)
  • 10,000 U/ml Penicillin-Streptomycin (Pen-Strep; ThermoFisher Scientific, cat. no. 15140122)
  • 70% (v/v) ethanol
  • NoSpin™ HepaRG™ cells (Lonza Bioscience, cat. no. NSHPRG)
  • PBS, pH 7.4, calcium chloride and magnesium chloride free (ThermoFisher Scientific, cat. no. 10010072)
  • RNeasy Midi Kit (Qiagen, cat. no. 75144)
  • High-Capacity RNA-to-cDNA™ Kit (ThermoFisher Scientific, cat. no. 4387406)
  • TaqMan™ Gene Expression Master Mix (ThermoFisher Scientific, cat. no. 4370074)
  • UltraPure™ DNase/RNase-Free Distilled Water (ThermoFisher Scientific, 10977023)
  • Human GAPDH Endogenous Control (FAM™/MGB probe, non-primer limited; ThermoFisher Scientific, cat. no. 4352934E, Assay ID: Hs99999905_m1)
  • Human CYP3A4 (FAM™/MGB probe; ThermoFisher Scientific, cat. no. 4331182, Assay ID: Hs00604506_m1)
  • Clear 1536-well compound plates (Greiner Bio-One, cat. no. 789270-C)
  • Biological safety cabinet (Labconco, cat. no. 302610101, or equivalent)
  • Tissue culture microscope (Carl Zeiss Microscopy, LLC, cat. no. 491237-0013-000, or equivalent)
  • 50-ml tubes (Corning Life Sciences, cat. no. 352070, or equivalent)
  • 200-µl micropipettor (Rainin, cat. no. 17008652, or equivalent)
  • 200-µl pipet tips (Rainin, cat. no. 30389268)
  • White/solid 1536-well flat-bottom cell-culture-treated plate (Greiner Bio-One, cat. no. 789173-F)
  • BioRAPTR2 Flying Reagent Liquid Dispenser (Let's Go Robotics, Inc., Model No. BR2), or equivalent
  • Microplate lids (Wako Automation, cat. no. 08-3241-0005)
  • CO2 incubator (ThermoFisher Scientific, cat. no. 3310, or equivalent)
  • Pintool Station (Wako Automation, cat. no. Pintool1144-3100 PLUS VELOCITY) or equivalent
  • ViewLux Plate Reader (Perkin Elmer, cat. no. 1430-0010A) or equivalent
  • GraphPad Prism Software (or equivalent)
  • Isotemp water bath (Fisher Scientific, cat. no. FSGPD20) or equivalent
  • 10-ml pipets (Corning Life Sciences, cat. no. 357551, or equivalent)
  • Pipettor (Integra Biosciences, cat. no. 155019, or equivalent)
  • Centrifuge (ThermoFisher Scientific, cat. no. 75004521, or equivalent)
  • BioCoat® Collagen-1-coated 12-well plate (Corning Life Sciences, cat. no. 354500)
  • Nanodrop spectrophotometer (DeNovix DS-11+) or equivalent
  • MicroAmp® Optical 384-Well Reaction Plate with Barcode (ThermoFisher Scientific, cat. no. 4309849)
  • QuantStudio 5 Real-Time PCR System (ThermoFisher Scientific, ref. no. A28135) or equivalent
  • MicroAmp® Optical Adhesive Film (ThermoFisher Scientific, cat. no. 4311971)
  • Sealing roller
  • Cell counter (Nexcelom Biosciences, cat. no. Cellometer Auto T4-IQOQ or equivalent)
  • Thermal Cycler Model 2720 (Applied Biosystems, cat. no. 4359659) or equivalent

Executing the pharmacologic study

1.Aliquot the promising compounds chosen, from Basic Protocol 2, step 7, into clear 1536-well compound plates.

Note
Set up the plates so that a concentration response curve can be acquired from them, remembering to use only columns 5-48 of the compound plate so that the previously made positive control plate (columns 1-4) can still be used. Therefore, use a 1:3 dilution starting with 20 mM stock compound in DMSO. Each compound should have quadruplicate amounts of each concentration. For example, the first compound chosen should have a 20 mM concentration in wells 5A, 6A, 5B, and 6B. This compound should then have a 6.67 mM concentration in wells 5C, 6C, 5D, and 6D. This should be repeated until 5AF and 6AF are filled. Columns 7 and 8 should contain the next chosen compound, and so on.

2.Generate a HepG2-CYP3A4-hPXR cell suspension, using assay medium from the cells still being passaged from Basic Protocol 1, step 20, with a concentration of 6.25 × 105 cells/ml. Plate 4 µl cell suspension into every well of white/solid 1536-well plates using a liquid dispenser. Place a lid on top of each plate, place the plates in a 37°C, 5% CO2 incubator, and incubate ∼4-5 hr.

Note
Because only 4 µl of cells can be plated, the concentration of cell suspension needs to be 6.25 × 105 cells/ml instead of the 5 × 105 cells/ml used previously in Basic Protocols 1 and 2. Plate three times as many plates as there are compound plates made in step 2. For example, if there was one plate of follow-up compounds generated, plate three plates full of cells. Perform this experiment three times when possible so that proper statistics and robust data can be attained. Leave the plates in the incubator until the cells are attached.

3.Transfer 23 nl from each well of the previously generated positive control plate from Basic Protocol 1, step 26, as well as 23 nl from each well of the compound plates produced in step 2, to each cell plate using a Wako Pintool station (Fig. 6).

4.Using assay medium, make 2.5 ml (per plate) of co-treatment (2.5 µM SPA70, 3.75 µM SPA70, and DMSO):

  • a.Add 6.25 µl of 1 mM SPA70 stock (in DMSO) to 2.5 ml of assay medium to generate 2.5 µM SPA70.
  • b.Add 9.38 µl of 1 mM SPA70 stock (in DMSO) to 2.5 ml of assay medium to generate 3.75 µM SPA70.
  • c.

Add 9.38 µl of DMSO to 2.5 ml of assay medium to represent the negative control.

Always use the same amount of DMSO as the highest amount of compound added. SPA70 is a known selective PXR antagonist (Lin et al., 2008). The concentrations of SPA70 chosen here were identified using optimization assays. However, the concentrations may vary depending on the performance of the assay in a different laboratory.

5.In every well of the respective plates, add 1 µl of DMSO co-treatment to plate 1, 2.5 µM SPA70 (final concentration of 0.5 µM) co-treatment to plate 2, and 3.75 µM SPA70 (final concentration of 0.75 µM) co-treatment to plate 3.

6.Follow steps 28-30 of Basic Protocol 1 to perform the luciferase reporter gene experiment on these plates.

Note
When the three curves from each compound are compiled into one graph, a clear right shift should be seen from DMSO to increasing SPA70 concentrations for each positive compound, as shown in Figure 7.

Pharmacological characterization of compound A. In this pharmacological assay, HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of compound A and co-treated with 0, 0.5, or 0.75 µM of SPA70, a known PXR inhibitor. The % activity was calculated by comparing each data point to the positive control, RIF (100% = RIF activity). Data are expressed as the mean ± SD of triplicate experiments. M, molar concentration.
Pharmacological characterization of compound A. In this pharmacological assay, HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of compound A and co-treated with 0, 0.5, or 0.75 µM of SPA70, a known PXR inhibitor. The % activity was calculated by comparing each data point to the positive control, RIF (100% = RIF activity). Data are expressed as the mean ± SD of triplicate experiments. M, molar concentration.

Quantitative real-time polymerase chain reaction

7.Make HepaRG™ culture medium by combining 200 ml of William's E Medium, 23.6 ml of HepaRG™ Thawing and Plating Medium Supplement (MHTAP), and 0.5 ml (25 U/ml) of Pen-Strep.

Note
HepaRG™ culture medium can be stored for up to 1 month at 4°C.

Note
Every item going in and out of the biological safety cabinet should be wiped down with 70% ethanol solution to minimize contamination. The supplements used may require another optimization step, for optimal data; it is recommended that the manufacturer's instructions for other supplements be checked if the experiment does not appear to work.

8.Warm the HepaRG™ culture medium in a 37°C water bath.

9.Thaw a frozen vial of HepaRG™ cells in a 37°C water bath until only a small piece of ice remains (∼90 sec).

Note
Do not fully submerge the vial into the water bath, as this may cause contamination; only submerge enough of the vial in water to cover the frozen cells.

10.Add the entire vial of cells into 9 ml of the prewarmed HepaRG™ culture medium.

Note
Use a small pipet to ensure the entire amount of liquid is transferred from the vial of cells. A 15- or 50-ml conical tube may be used for this step.

11.Centrifuge 5 min at 900 rpm (100 × g), room temperature.

Note
The manufacturer recommends skipping the centrifugation step when plating these cells; however, in our hands, a spin step generates a clean pellet free from any freezing chemicals with no harsh ramifications.

12.Remove the supernatant and resuspend the pellet using 10 ml fresh prewarmed HepaRG™ culture medium.

13.Plate HepaRG™ cells in a 12-well collagen-I-coated plate at a concentration of 8 × 105 cells/well (Day 0).

Note
Cells should be plated in a confluent manner as these are fully differentiated and will not be passaged further.

14.Incubate the plates at 37°C, 5% CO2.

15.Acquire the EC70 for each selected compound using GraphPad software.

Note
Once the % activity values from the primary high-throughput screen are input into GraphPad and the concentrations are transformed into log values, click analyze again. Under the XY analyses topic, click “Nonlinear regression (curve fit).” When the new window pops up, go to the section labeled “Dose Response – Special” and click the “log(agonist) versus response – Find ECanything.” Next, click on the Constrain tab and put “70” into the value box next to the constant F (where F = fraction of maximal response). Click ok. Once the analysis comes up, copy the EC70 values onto your graph.

16.On day 3, after determining that the cells are healthy by assessment under a microscope, exchange the old medium with 1 ml fresh medium per well.

Note
See Figure 8A for an image of what healthy cells look like under 10× magnification. If the cells get contaminated or look unhealthy (Fig. 8B), they should be discarded, and the experiment started over.

Morphology of healthy and toxicity-affected HepaRG cells. HepaRG cells were treated for 23 hr with DMSO (A) and a toxic compound (B), visually representing proper healthy cells and damaged cells, respectively. The microscope pictures were taken at 10×.
Morphology of healthy and toxicity-affected HepaRG cells. HepaRG cells were treated for 23 hr with DMSO (A) and a toxic compound (B), visually representing proper healthy cells and damaged cells, respectively. The microscope pictures were taken at 10×.

17.Treat the cells with each of the promising compounds (one well per treatment) by adding the correct amount of stock solution to the fresh medium.

Note
Using the EC70 from the concentration response curve, as the treatment concentration, generated in step 15, is a good starting point. The goal is to use a concentration with a high efficacy but low potency value, so that the cells will remain healthy throughout the experiment. If the EC70 value generates toxicity, lower the concentration in the next experiment. Make sure to also include a negative solvent control of the liquid used to dissolve your compounds (e.g., DMSO) and a positive control (e.g., rifampicin, 10 µM) to ensure the assay worked. Given the expense of metabolically competent cells, we recommend starting with one concentration for each compound. If possible, we also suggest making a 1000× stock solution of the compounds being used so that the user can add 1 µl of stock solution to the 1 ml of fresh medium. For example, generate a 10 mM rifampicin stock solution so that 1 µl will be added to the cells, generating a final treatment concentration of 10 µM.

18.Gently shake the plate vertically, as well as horizontally, four times. This will ensure that the compound is spread around the well and into the correct homogenous concentration.

19.Replace the treated plate in the 37°C, 5% CO2 incubator and incubate 24 hr.

20.After the 24-hr treatment, examine each well under a microscope.

Note
Take note of any toxicity, contamination, or abnormal appearance. This will help make sense of the data if a number seems incorrect after the experiment.

21.Suck out the medium and wash each well with 1 ml PBS two times.

22.Harvest the cells by adding 350 µl lysis buffer to each well.

Note
We used Buffer RLT from the Qiagen RNeasy midi-prep kit for this protocol. However, other lysis buffers can be used and should be optimized for each respective laboratory. The amount of lysis buffer was based on the manufacturer's recommendation.

Note
If necessary, the protocol may be paused here by putting the plate into a −80°C freezer until needed. If no kit is available, TRIzol™ may be used instead for the isolation of RNA; follow the manufacturer's instructions carefully as using this chemical involves many more steps, which can generate errors.

23.Perform RNA extraction using the RNeasy kit, following the kit instructions precisely.

Note
For the final step of this protocol, 30 µl RNase-free water is used to elute the RNA off the column and into a tube. Again, the protocol may be paused here by placing the samples in a −80°C freezer.

24.Determine the RNA concentration of each sample using a spectrophotometer.

Note
The A260/A280 ratio should be ∼2.0 if your RNA is pure; when there is a protein, phenol, or other contaminant, the ratio will be much lower than 2.0 and the sample will not provide high-quality data. Also, the A260/A230 ratio should be between 2.0 and 2.2; a higher value indicates that the sample may be contaminated with unwanted organic compounds.

25.Using 1 µg cDNA, perform reverse transcriptase using a high-capacity RNA-to-cDNA kit according to the manufacturer's instructions.

Note
cDNA samples may be stored for up to 1 month at −20°C.

Note
This step should be performed on ice up to the point when the samples are put in the thermal cycler. Be sure to vortex and then spin down the samples before placing them into the thermal cycler (step 32).

26.Add 80 µl DNase/RNase-free distilled water to each sample.

Note
This will work for most samples as they are very concentrated. However, it may be necessary to use a smaller dilution factor if the RT-qPCR data cannot be quantified in the final steps (no CT value is detected).

27.Calculate how many wells, in a 384-well PCR assay plate, are needed for each gene. Figure 9 shows an example of a plate setup for five cDNA samples in triplicate.

Note
A 96-well assay plate may also be used depending on the block in the specific assay machine used for this procedure. Each sample should be tested in triplicate for each gene. There should also always be extra made (e.g., for three extra wells) as some loss can be expected from the pipet tips.

Illustrative display of an RT-qPCR plate setup. The blue circles represent wells with a GAPDH probe while the pink circles represent wells including a CYP3A4 probe. The red box encircling Row A indicates samples treated with DMSO (three each blue and pink), while the blue box (Row C) defines samples treated with rifampicin, the positive control. The lavender boxes (rows E, G, and I) identify three compounds being tested for identification of modulation of GAPDH and CYP3A4. This image was created with BioRender (biorender.com).
Illustrative display of an RT-qPCR plate setup. The blue circles represent wells with a GAPDH probe while the pink circles represent wells including a CYP3A4 probe. The red box encircling Row A indicates samples treated with DMSO (three each blue and pink), while the blue box (Row C) defines samples treated with rifampicin, the positive control. The lavender boxes (rows E, G, and I) identify three compounds being tested for identification of modulation of GAPDH and CYP3A4. This image was created with BioRender (biorender.com).

28.Generate gene mixes according to Table 1 using TaqMan™ Master Mix and the appropriate gene probes.

Note
SYBR Green can also be used here; however, it is a less specific probe and may result in more variable data. TaqMan acquires cleaner, more reproducible data but is much more expensive, so each laboratory must optimize according to their needs. When selecting the correct primers/probes, always include an endogenous control that is stably expressed in the cell line to use as a reference gene (e.g., GAPDH, β-actin, etc.). For all nuclear receptors, commonly regulated genes should be chosen; for example, in this assay, use CYP3A4 to represent modulation of PXR.

Table 1. An Example of PXR Gene Expression Mixes for RT-qPCR
1 well GAPDH (18 wells) CYP3A4 (18 wells)
2× master mix (µl) 10 180 180
Probe (µl) 1 18 18
Water (µl) 5 90 90

29.Add 16 µl of each mix to the corresponding wells being assayed for that gene.

30.Add 4 µl of each diluted cDNA sample into the correct wells, creating a total of 20 µl per well.

Note
A no-template control should also be tested to ensure that the DNA is not contaminated. To do this, combine 16 µl of each one of the mixes with 4 µl water. The amplification plot for each no-template control mix should display higher CT values than the control or sample dilutions. If they are not, your sample may include extraneous nucleic acid contamination and the assay will need to be completed again with extra precautions against contamination.

31.Add an adhesive seal firmly onto the plate using a sealing roller, and ensure all sides are correctly stuck.

32.Mix and spin down the plate, using a vortex (for a few seconds at maximum power) and centrifuge (for 1 min at 1000 rpm, or 163 × g), respectively, to ensure there is a homogenous liquid completely spun into the bottom of the well.

33.Place the plate into the corresponding block of a PCR machine (thermal cycler).

34.Perform PCR using a protocol that best fits the primers bought, according to the manufacturer's instructions.

35.Analyze the data according to the fold = 2−ΔΔCt method, where ΔCt represents the differences in cycle threshold numbers between CYP3A4 (or the genes in question) and GAPDH, and ΔΔCt represents the relative change in these differences between negative control and treatment groups (Livak & Schmittgen, 2001).

REAGENTS AND SOLUTIONS

PXR assay medium

  • 920 ml phenol-red-free Dulbecco's Modified Eagle's Medium (DMEM; ThermoFisher Scientific, cat. no. 31053028)
  • 50 ml charcoal-stripped FBS (ThermoFisher Scientific, cat. no. 12676029; 5% v/v)
  • 10 ml 10,000 U/ml Penicillin-Streptomycin (Pen-Strep; ThermoFisher Scientific, cat. no. 15140122; 1% v/v)
  • 10 ml of 100 mM sodium pyruvate (ThermoFisher Scientific, cat. no. 11360070; 1 mM)
  • 10 ml of 200 mM l-glutamine (ThermoFisher Scientific, cat. no. 25030081; 2 mM)
  • Filter under biological safety cabinet conditions
  • Store up to 1 month at 4°C.

PXR culture medium

  • 880 ml of Eagle's Minimum Essential Medium (EMEM; ATCC, cat. no. 30-2003)
  • 100 ml Characterized Fetal Bovine Serum (FBS; Cytiva, cat. no. SH30071.03; 10% v/v)
  • 10 ml Pen-Strep (ThermoFisher Scientific, cat. no. 15140122; 1% v/v)
  • 10 ml geneticin (ThermoFisher Scientific, cat. no. 10131027; 500 µg/ml
  • Filter under biological safety cabinet conditions
  • Store up to 1 month at 4°C.

COMMENTARY

Background Information

Identification and characterization of potential novel modulators of nuclear receptors are important steps when identifying toxic drug-drug interactions, determining toxicity, or assessing potential therapeutic benefits. Previously, this process has been low throughput and, therefore, time consuming and slow (Attene-Ramos et al., 2013). The ethics of utilizing in vivo animal models, when other options are available, has also become a global concern (Hsu et al., 2016). However, there have been recent strides in generating a high-throughput method that can be used for different nuclear receptors (Lynch, Mackowiak, et al., 2019; Lynch et al., 2021; Lynch et al., 2017; Lynch, Zhao et al., 2019). When screening in a high-throughput manner, thousands of compounds can be assayed in one experiment, leading to a quick and reliable technique. One of the main disadvantages to these methods is the lack of an efficient metabolic capability. When a drug is ingested, it often undergoes metabolism, and these high-throughput assays do not often account for this natural process. Immortalized cell lines are frequently used in high-throughput screens due to the ease of working with them and their availability, cost, and durability. However, these cells do not usually contain all necessary metabolic components and may not represent human conditions. Therefore, follow-up studies are always an essential part of validating compounds identified from a high-throughput screen.

The detailed protocols described above were finalized after many different optimization steps, including examining various reagents, machines, and equipment settings. However, different machines may require different settings and reagents. For instance, Figure 4 represents the difference between the use of ONE-Glo and Reagent 2, clearly identifying ONE-Glo as the superior reagent in this laboratory for this assay. The cell-type is another aspect that should be addressed when utilizing a luciferase reporter gene assay; transient transfection can be used with low- or medium-throughput assays but will be a disadvantage in a high-throughput assay. When performing a transient transfection, each well must include cells containing both plasmids (i.e., PXR and CYP3A4-luc); the transiently transfected cells often contain heterogenic cell populations, causing a high level of variability in the assay. Therefore, it is crucial to have a stable cell line when performing a high-throughput nuclear receptor luciferase reporter gene assay.

Pharmacological assays employing in vivo animal models are often used to assess a drug's activity (Drug Discovery and Evaluation: Pharmacological Assays, 2016). However, as previously noted, it has become imperative to decrease animal usage in the scientific community. The pharmacological assay described above allows the determination of PXR agonistic activity without the use of animals. Once again, however, a metabolic component is missing from this assay, and further experiments are necessary to verify the activity. In the assay described here, SPA70, a known PXR antagonist, is used as the inhibitor of PXR. Before this compound was identified as a selective and potent PXR antagonist, ketoconazole and sulforaphane were the best options. However, they have shown to be ineffective in vivo, potentially due to toxicity (Lin et al., 2017). Therefore, it is advisable to ensure that the pharmacological assay can be completed using varying concentrations of SPA70.

Quantifying mRNA levels, here of a specific gene being regulated by a nuclear receptor, is a common technique when studying chemical activation. Historically, northern blot analysis, introduced in 1977, was the standard method to detect a change in gene expression (Alwine, Kemp, & Stark, 1977). Utilizing electrophoresis, separated bands of mRNA are transferred from an agarose gel to a membrane. The membrane is then probed and signal is detected. The recently preferred and more precise method, RT-qPCR, is a quicker and more sensitive method that requires a significantly smaller amount of mRNA for detection (Bustin, 2000). However, it also poses some drawbacks, including inappropriate results when the slightest contamination occurs. Therefore, it is imperative to work in as aseptic an area as possible when performing this assay.

Critical Parameters

Basic Protocol 1 needs to be completed within a biological safety cabinet. Many specific steps in an experiment need to be optimized so that the assay will work to its optimum level. Cell type, cell number per assay well, reagents used, treatment time, plate type, and signal output reader are all aspects that must be optimized for this assay. Another vital issue is the positive control plate. As all compounds are compared to the positive and negative controls, it is crucial that these compounds be at their optimal concentration and purity. It is also pivotal to keep the assay plates inside the incubator, at 37°C and 5% CO2, throughout the experiment for as long as possible so that the cells can achieve their healthiest forms.

The main goal of high-throughput screening is to acquire reliable, reproducible data efficiently and quickly. Therefore, the core attribute necessary for Basic Protocol 2 is to generate an assay that is easily duplicated without high levels of human error. Utilizing a robot is the simplest way to produce this type of experiment as it increases screening speed and minimizes mistakes. Because this experiment is a high-throughput version of Basic Protocol 1, many of the same critical parameters apply here as well. However, having access to a double-stable cell line is very important to this assay, as transient transfection may not guarantee assay reproducibility.

One of the most crucial aspects of Basic Protocol 3 is the selection of promising compounds using intuitive parameters (potency, efficacy, curve quality, and reproducibility). For example, a compound may have an efficacy of 100% of the positive control, rifampicin, due to an outlier. In this instance, the concentration curve may be flat with one data point up very high (Fig. 10) generating a false positive; in this case, the compound should not be selected for further, more in-depth review. The pharmacological study has many of the same parameters as Basic Protocol 1, as it is just an addendum to that experiment. However, one thing to be mindful of is selecting a known nuclear receptor inhibitor with good potency and efficacy, such as SPA70 for this specific protocol. When performing experiments requiring the use of mRNA, as when executing RT-qPCR, it is imperative to always use RNase-free water and be mindful of possible contaminants, such as oil from ungloved hands or droplets from talking or coughing near the sample. The cells chosen for the RT-qPCR assay should already have the nuclear receptors and target genes inside the system so that chemical induction can be seen. This experiment will determine how much RNA is being generated, and therefore human primary hepatocytes or HepaRG cells are the optimal cell models for this study. Another point of contention for the RT-qPCR assay is which type of dye to use, e.g., TaqMan versus SYBR Green. This will depend on the primers or probes bought and the specific machine's capability; therefore, it is always necessary to make sure those aspects match up.

Activity response curve of an efficacious but not selectable compound. HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of a test compound for 24 hr during the Tox21 10,000 compound high-throughput screen. The % activity was calculated by comparing each data point to the positive control, RIF (100% = RIF activity).
Activity response curve of an efficacious but not selectable compound. HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of a test compound for 24 hr during the Tox21 10,000 compound high-throughput screen. The % activity was calculated by comparing each data point to the positive control, RIF (100% = RIF activity).

Troubleshooting

Once optimized, the screening protocol should be easy to repeat quickly and efficiently. However, optimization and follow-up steps, such as in these protocols, are nuanced and subject to many potential complications. Table 2 lists some ways in which to avoid these problems by explaining solutions in a clear and precise manner.

Table 2. Troubleshooting Guide for Identifying PXR Activators
Problem Possible cause Solution
Cells won't detach from flask Cells not washed with PBS free of CaCl2 and MgCl2 Suck out trypsin, wash the flask twice with CaCl2- and MgCl2-free PBS, and add fresh trypsin.
Overexposure when reading luminescence Overly long exposure Decrease exposure time.
Too many cells in each well Replate cells at a lower density.
Low signal when reading luminescence Insufficiently long exposure Increase exposure time.
Not enough cells Replate cells at a higher density.
Positive control not at proper concentration Redo positive control plate using a higher concentration.
Reagent not strong enough Acquire a different luminescent reagent and try the assay again.
Low RNA concentration Low amount of RNA harvested Plate more wells and pool samples together.
Ethanol not added to Buffer RPE (Qiagen RNeasy Midi Kit) Check that ethanol was added to RPE. If samples have already been extracted without the added ethanol, the experiment will need to be begun again and repeated.
Low or no amplification during PCR Low purity Re-purify or consult purification kit manual.
Not enough cDNA Increase amount of DNA added or number of PCR cycles if suitable.
Deficient primers Check the primer design to ensure they are target specific and complementary.
PCR conditions are not ideal Check the manufacturers’ instructions (for machine, primers, master mix, etc.) and adjust protocol as needed.

Understanding Results

When performing a luciferase reporter gene assay, the concentration response curve of a compound is the main result that needs to be understood; the potency and efficacy calculated based on this curve using any plotting software (e.g., GraphPad) will characterize whether the compound is a modulator of the nuclear receptor. Comparing a positive control compound to test compounds is an optimal way to analyze the screened data.

Using the results of the equation from Basic Protocol 1, step 30, any compound producing reporter gene activity >100% is more efficacious than the positive control. However, even if a chemical has an efficacy of 100% that of rifampicin, if that potency is seen at a concentration that is not clinically relevant, that compound will not be the best choice to select for further review when assessing the data in humans. Figure 11 displays the concentration response curves from a structural cluster of compounds identified in the PXR activator Tox21 10,000 compound screen. Here, the most potent and efficacious compound, compound A, was chosen for further study (Lynch et al., 2021). Compound C could have also been selected; however, there were other compounds showing similar results and we were interested in identifying as many compounds from different structural clusters as possible. Compound B also had a good curve, but had only a 50% efficacy and a lower potency, ruling it out among other, more qualified compounds. These data represent the expected type of result when performing nuclear receptor luciferase reporter gene assays.

Activity response curves from a group of structurally similar compounds. HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of three test compounds from the same structural cluster (compounds A, B, and C) for 24 hr during the Tox21 10,000 compound high-throughput screen. Data are expressed as the mean ± SD of triplicate assays. M, molar concentration.
Activity response curves from a group of structurally similar compounds. HepG2-hPXR-CYP3A4 cells were treated with multiple concentrations of three test compounds from the same structural cluster (compounds A, B, and C) for 24 hr during the Tox21 10,000 compound high-throughput screen. Data are expressed as the mean ± SD of triplicate assays. M, molar concentration.

The more in-depth and complicated data to decipher with these methods are those from Basic Protocol 3. The pharmacological assay is similar to the first two basic protocols, in that its endpoint is still concentration response curves. However, when an inhibitor is added to the treatment condition, a positive compound will generate a curve shifted to the right as compared to that without inhibitor addition (Fig. 7). This right-shift means that the EC50 will be higher, equaling a reduced potency. Compound A, the previously chosen compound, shows a right-shifted curve in an inhibitor-concentration-dependent manner.

The RT-qPCR experiment normalizes treatment groups to a negative control, DMSO, to generate a xenobiotic-induced fold induction. The value for the DMSO group should always equal 1. After acquiring the gene fold induction for each treatment, a statistical analysis needs to be performed in order to capture any significant change. Because the data include triplicate values for a control and more than one treatment, also in triplicate, the statistical comparison tool that should be used is one-way analysis of variance (ANOVA) with post-hoc Dunnett's analysis. The easiest way to perform this analysis is to input the data into GraphPad Prism software and utilize the correct functions. Figure 12 gives an example of what these data might look like for a test of six compounds, including compound A. RIF is the positive control of induction for CYP3A4 through PXR modulation, and compounds A, 3, 4, and 5 statistically significantly induce CYP3A4 activity. Once a compound is identified as statistically different from the negative control, it is defined as an actual inducer of the nuclear receptor being tested. In this example, four of the six compounds tested appear to be PXR activators, through the induction of CYP3A4. However, other nuclear receptors also modulate CYP3A4, which suggests that more studies need to be conducted, potentially using PXR knockout cells, to confirm their true activities.

CYP3A4 mRNA expression in HepaRG cells. HepaRG cells were treated with the vehicle control (0.1% DMSO), positive control (10 µM RIF), compound A, or compounds 1, 2, 3, 4, or 5. Real-time PCR was used to analyze the mRNA expression of CYP3A4. Each bar represents the mean ± SD in triplicate wells. *, p < .05; **, p < .01; ***, p < .001 using ANOVA post-hoc Dunnett's analysis.
CYP3A4 mRNA expression in HepaRG cells. HepaRG cells were treated with the vehicle control (0.1% DMSO), positive control (10 µM RIF), compound A, or compounds 1, 2, 3, 4, or 5. Real-time PCR was used to analyze the mRNA expression of CYP3A4. Each bar represents the mean ± SD in triplicate wells. *, p < .05; **, p < .01; ***, p < .001 using ANOVA post-hoc Dunnett's analysis.

Time Considerations

Depending on the assay conditions in a protocol, it could take anywhere from a few weeks to a few months to optimize this assay. When thawing frozen cells, we suggest allowing at least one cell passage before using them in an assay. For example, thaw cells on Monday, passage cells on Thursday, and the cells will be ready for use on the following Monday. Each time you perform the nuclear receptor luciferase reporter gene assay, it should take about the same amount of time. After the cells are plated and allowed to attach to the bottom of the assay well (3-5 hr), the cells are treated for an optimal amount of time; we suggest between 15 and 24 hr depending on the type of assay. Adding detection reagents, incubating the assay plate, and reading the plates generally takes ∼2 hr. Therefore, altogether, one complete assay will take between 20 and 31 hr. Once the reagents, treatment timing, cells, and machine settings are optimized, a larger screen can take place. The timing for this screen will depend on how many compounds and plates you are screening at once, as well as the machinery available. If no robot is available, we suggest working with ∼10-15 plates at a time due to hands-on restrictions. Using more than 15 plates at a time may get confusing and lead to timing issues as well as an increased chance of human error. For the pharmacological study in Basic Protocol 3, the time expectance is the same (between 20 and 31 hr) as for the initial optimization assay because it is a repeat experiment and adding an inhibitor adds no extra time.

Regarding the RT-qPCR assay, there are various steps which take varying times depending on experience; there are also multiple points at which the experiment can be paused and put in the −80°C or −20°C freezer. Therefore, the assay can take 1 day from harvest to PCR analysis, or it can take however long is necessary given time constraints. Depending on the cell type used, plating of the cells and the time before treatment vary. In this specific protocol, using HepaRG cells, the time from plating to harvesting is 4 days because there is a 24-hr treatment. Once harvested, the lysates can be kept at −80°C until ready to use. The next step is to isolate the RNA, using a kit or TRIzol™, which will take ∼2-4 hr depending on the method and number of samples generated. After the RNA is extracted, the protocol can once again be paused, if desired, and samples kept at −80°C before proceeding. Measuring the RNA concentration and performing reverse transcriptase takes ∼2 hr total, and once again the samples can be stored in a −20°C before proceeding with the next step. Setting up and performing the qRT-PCR will take at least 2 hr depending on the number of samples and genes being measured, the experience of the scientist, and the type of master mix used. With an experienced scientist, one 384-well plate can be fully filled and completed in ∼2.5 hr if using the regular Taqman master mix. If using the fast Taqman master mix, the PCR setup and assay can be completed in ∼1 hr 45 min. Therefore, to complete a qRT-PCR assay once the samples are harvested will take at least 6 hr if there are only a small number of samples and genes to measure. However, depending on whether the researcher stopped at any of various different steps, the assay can be completed weeks or even months later.

Acknowledgments

This work was supported in part by the Intramural Research Program of the National Center for Advancing Translational Sciences (NCATS), National Institutes of Health. The views expressed in this article are those of the authors and do not necessarily reflect the statements, opinions, views, conclusions, or policies of the NIH. Mention of trade names or commercial products does not constitute endorsement or recommendation for use.

Author Contributions

Caitlin Lynch : Data curation, investigation, methodology, validation, writing—original draft, writing—review and editing**; Srilatha Sakamuru** : Data curation, methodology, writing—review and editing**; Menghang Xia** : Conceptualization, investigation, methodology, supervision, writing—review and editing.

Conflict of Interest

The authors report no conflict of interest. All authors have read and approved the final manuscript.

Open Research

Data Availability Statement

The data, tools, and material (or their source) that support Basic Protocol 1 and 3 are available from the corresponding author upon reasonable request. The data that support Basic Protocol 2 are openly available in the Tox21 Gateway at https://tripod.nih.gov//tox21/pubdata/, reference assay tox21-pxr-p1.

Literature Cited

  • Alwine, J. C., Kemp, D. J., & Stark, G. R. (1977). Method for detection of specific RNAs in agarose gels by transfer to diazobenzyloxymethyl-paper and hybridization with DNA probes. Proceedings of the National Academy of Sciences of the United States of America , 74(12), 5350–5354. doi: 10.1073/pnas.74.12.5350
  • Attene-Ramos, M. S., Miller, N., Huang, R., Michael, S., Itkin, M., Kavlock, R. J., … Xia, M. (2013). The Tox21 robotic platform for the assessment of environmental chemicals—from vision to reality. Drug Discovery Today , 18(15-16), 716–723. doi: 10.1016/j.drudis.2013.05.015
  • Bustin, S. A. (2000). Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays. Journal of Molecular Endocrinology , 25(2), 169–193. doi: 10.1677/jme.0.0250169
  • F. J. Hock (Ed.). (2016). Drug Discovery and Evaluation: Pharmacological Assays ( 4 ed.). Cham, Switzerland: Springer.
  • Huang, R. (2016). A quantitative high-throughput screening data analysis pipeline for activity profiling. Methods in Molecular Biology , 1473, 111–122. doi: 10.1007/978-1-4939-6346-1_12
  • Hsu, C.-W., Huang, R., Attene Ramos, M., Austin, C., Simeonov, A., & Xia, M. (2016). Advances in high-throughput screening technology for toxicology. International Journal of Risk Assessment and Management , 20(1-3), 109–135. doi: 10.1504/IJRAM.2017.082562
  • Kliewer, S. A., Moore, J. T., Wade, L., Staudinger, J. L., Watson, M. A., Jones, S. A., … Lehmann, J. M. (1998). An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell , 92(1), 73–82. doi: 10.1016/s0092-8674(00)80900-9
  • Kumar, G. N., & Surapaneni, S. (2001). Role of drug metabolism in drug discovery and development. Medicinal Research Reviews , 21(5), 397–411. doi: 10.1002/med.1016
  • Lin, W., Wang, Y. M., Chai, S. C., Lv, L., Zheng, J., Wu, J., … Chen, T. (2017). SPA70 is a potent antagonist of human pregnane X receptor. Nature Communications , 8(1), 741. doi: 10.1038/s41467-017-00780-5
  • Lin, W., Wu, J., Dong, H., Bouck, D., Zeng, F. Y., & Chen, T. (2008). Cyclin-dependent kinase 2 negatively regulates human pregnane X receptor-mediated CYP3A4 gene expression in HepG2 liver carcinoma cells. Journal of Biological Chemistry , 283(45), 30650–30657. doi: 10.1074/jbc.M806132200
  • Livak, K. J., & Schmittgen, T. D. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods (San Diego, Calif.) , 25(4), 402–408. doi: 10.1006/meth.2001.1262
  • Lynch, C., Mackowiak, B., Huang, R., Li, L., Heyward, S., Sakamuru, S., … Xia, M. (2019). Identification of modulators that activate the constitutive androstane receptor from the Tox21 10K compound library. Toxicological Sciences , 167(1), 282–292. doi: 10.1093/toxsci/kfy242
  • Lynch, C., Sakamuru, S., Huang, R., Niebler, J., Ferguson, S. S., & Xia, M. (2021). Characterization of human pregnane X receptor activators identified from a screening of the Tox21 compound library. Biochemical Pharmacology , 184, 114368. doi: 10.1016/j.bcp.2020.114368
  • Lynch, C., Sakamuru, S., Huang, R., Stavreva, D. A., Varticovski, L., Hager, G. L., … Xia, M. (2017). Identifying environmental chemicals as agonists of the androgen receptor by using a quantitative high-throughput screening platform. Toxicology , 385, 48–58. doi: 10.1016/j.tox.2017.05.001
  • Lynch, C., Zhao, J., Sakamuru, S., Zhang, L., Huang, R., Witt, K. L., … Xia, M. (2019). Identification of compounds that inhibit estrogen-related receptor alpha signaling using high-throughput screening assays. Molecules (Basel, Switzerland) , 24(5), 841. doi: 10.3390/molecules24050841
  • Schulman, I. G. (2010). Nuclear receptors as drug targets for metabolic disease. Advanced Drug Delivery Reviews , 62(13), 1307–1315. doi: 10.1016/j.addr.2010.07.002
  • Sueyoshi, T., & Negishi, M. (2001). Phenobarbital response elements of cytochrome P450 genes and nuclear receptors. Annual Review of Pharmacology and Toxicology , 41, 123–143. doi: 10.1146/annurev.pharmtox.41.1.123

推荐阅读

Nature Protocols
Protocols IO
Current Protocols