We present a protocol to screen anti-hepatitis B virus (HBV) compounds targeting pre- and post-viral entry lifecycle stages, using isothermal titration calorimetry to measure binding affinity (KD) with host sodium taurocholate cotransporting polypeptide. Antiviral efficacy was determined through the suppression of viral lifecycle markers (cccDNA formation, transcription, and viral assembly).
Hepatitis B virus (HBV) infection has been considered a crucial risk factor for hepatocellular carcinoma. Current treatment can only lessen the viral load but not result in complete remission. An efficient hepatocyte model for HBV infection would offer a true-to-life viral life cycle that would be crucial for the screening of therapeutic agents. Most available anti-HBV agents target lifecycle stages post viral entry but not before viral entry. This protocol details the generation of a competent hepatocyte model capable of screening for therapeutic agents targeting pre-viral entry and post viral entry lifecycle stages. This includes the targeting of sodium taurocholate cotransporting polypeptide (NTCP) binding, cccDNA formation, transcription, and viral assembly based on imHC or HepaRG as host cells. Here, the HBV entry inhibition assay used curcumin to inhibit HBV binding and transporting functions via NTCP. The inhibitors were evaluated for binding affinity (KD) with NTCP using isothermal titration calorimetry (ITC)-a universal tool for HBV drug screening based on thermodynamic parameters.
Hepatitis B virus (HBV) infection is considered a life-threatening disease worldwide. Chronic HBV infection is laden with a risk of liver cirrhosis and hepatocellular carcinoma1. Current anti-HBV treatment focuses mostly on post viral entry using nucleos(t)ide analogs (NAs) and interferon-alpha (IFN-α)2,3. The discovery of an HBV entry inhibitor, Myrcludex B, has identified a novel target for anti-HBV agents4. The combination of entry inhibitors and NAs in chronic HBV has significantly lessened the viral load compared to those targeting viral replication alone5,6. However, the classical hepatocyte model for the screening of HBV entry inhibitors is limited by low viral receptor levels (sodium taurocholate cotransporting polypeptide, NTCP). The overexpression of hNTCP in hepatoma cells (i.e., HepG2 and Huh7) improves HBV infectivity7,8. Nevertheless, these cell lines express low levels of phase I and II drug-metabolizing enzymes and exhibit genetic instability9. Hepatocyte models that can help target distinct mechanisms of candidate anti-HBV compounds such as previral entry, NTCP binding, and viral entry would expedite the identification and development of efficacious combination regimens. The study for anti-HBV activity of curcumin has elucidated the inhibition of viral entry as a new mechanism in addition to post viral entry interruption. This protocol details a host model for the screening of anti-HBV entry molecules10.
The goal of this method is to explore candidate anti-HBV compounds for viral entry inhibition, especially blocking NTCP binding and transport. As NTCP expression is a critical factor for HBV entry and infection, we optimized the hepatocyte maturation protocol to maximize NTCP levels11. In addition, this protocol can differentiate the inhibitory effect on HBV entry as inhibition of HBV attachment versus inhibition of internalization. The taurocholic acid (TCA) uptake assay was also modified using an ELISA-based method instead of a radioisotope to represent NTCP transport12,13. The receptor and ligand interaction was confirmed by their 3D structures14,15. The inhibition of NTCP function can be evaluated by measuring TCA uptake activity16. However, this technique did not provide direct evidence of NTCP binding to the candidate inhibitors. Therefore, the binding can be investigated using various techniques, such as surface plasmon resonance17, ELISA, fluorescence-based thermal shift assay (FTSA)18, FRET19, AlphaScreen, and various other methods20. Among these techniques, ITC is a goal standard in binding analysis because it can observe heat absorption or emission in almost every reaction21. The binding affinity (KD) of NTCP and candidate compounds was directly evaluated using ITC; these affinity values were more precise than those obtained using the in silico prediction model22.
This protocol covers techniques in hepatocyte maturation, HBV infection, and screening for HBV entry inhibitor. Briefly, a hepatocyte model was developed based on imHC and HepaRG cell lines. The cultured cells were differentiated into mature hepatocytes within 2 weeks. The upregulation of NTCP levels was detected using real-time PCR, western blot, and flow cytometry11. Hepatitis B virion (HBVcc) was produced and collected from HepG2.2.15. The differentiated imHC or HepaRG (d-imHC, d-HepaRG) was prophylactically treated with the anti-HBV candidates 2 h prior to the inoculation with HBV virion. The expected outcome of the experiment was the identification of the agents that decrease cellular HBV and infectivity. Anti-NTCP activity was evaluated using the TCA uptake assay. NTCP activity could be suppressed by the agents that specifically bound NTCP. The ITC technique was employed to investigate the feasibility of interactive binding that could predict inhibitors and their target proteins, determining the binding affinity (KD) of the ligand for the receptor via non-covalent interactions of the biomolecular complex23,24. For instance, KD ≥ 1 × 103 mM represents weak binding, KD ≥ 1 × 106 µM represents moderate binding, and KD ≤ 1 × 109 nM represents strong binding. The ΔG is directly correlated with binding interactions. In particular, a reaction with negative ΔG is an exergonic reaction, indicating that binding is a spontaneous process. A reaction with a negative ΔH indicates that the binding processes depend on hydrogen bonding and Van der Waals forces. Both TCA uptake and ITC data could be used to screen for anti-HBV entry agents. The outcomes of these protocols can provide a foundation for not only anti-HBV screening but also the interaction with NTCP as assessed through binding affinity and transport function. This paper describes host cell preparation and characterization, experimental design, and evaluation of the anti-HBV entry together with the NTCP binding affinity.
NOTE: The following procedures must be performed in a Class II biological hazard flow hood or a laminar-flow hood. The handling of HBV was ethically approved by the IRB (MURA2020/1545). See the Table of Materials for details about all solutions, reagents, equipment, and cell lines used in this protocol.
1. Preparing host cells (mature hepatocytes)
2. Quantification of cellular NTCP
3. Production of the cell culture-derived HBV particles (HBVcc)
4. Anti-HBV entry assay
5. HBV-cell binding assay
6. Taurocholic acid (TCA) uptake assay
7. Determination of protein-ligand interactions using isothermal titration calorimetry
NOTE: This assay system was developed based on the MicroCal PEAQ-ITC (ITC software).
8. Statistical analysis
Hepatic maturation features were observed, including binucleated cells and polygonal-shaped morphology (Figure 1), especially in the differentiated stage of imHC (Figure 1A). A large increase in NTCP expression was measured in d-HepaRG and d-imHC at 7-fold and 40-fold, respectively (Figure 1B). The highly glycosylated form of NTCP, postulated to confer susceptibility to HBV entry, was detected more in d-imHC than in d-HepaRG (Figure 1C). The differentiated imHC contained 65.9% higher NTCP levels than did the undifferentiated cells (Figure 1D).
Figure 2A shows a summary of the prophylactic treatment. Figure 2B shows HBV immunofluorescence staining performed to evaluate the anti-HBV entry activities on day 7 post infection, while Figure 2C outlines the HBV binding assay protocol. The level of HBV binding on the cell surface receptor was evaluated by real-time PCR (Figure 2D). To determine whether NTCP was the receptor for HBV attachment, TCA uptake was determined for the candidate binding inhibitors (Figure 2E). Based on this model, the putative inhibitory activity of candidate compounds decreased HBV binding through NTCP.
The calorimetric alteration was detected with continuous injections to the sample cell (Figure 3). A standard non-linear least squares regression was plotted based on one binding site fitted well to the data. The solid line indicated the best fit to the experimental values (Figure 3A,B). Table 1 shows thermodynamic parameters correlated with the binding of NTCP with cyclosporine A (CsA) or a candidate compound.
Figure 1: Hepatic maturation of HepaRG and imHC with NTCP characterization. Both cell lines were cultured in hepatic maturation medium for 2 weeks. (A) Hepatocyte characteristics such as binucleated cells and polygonal-shaped morphology were exclusively observed in the maturation stage of imHC. Scale bars = 50 µm. (B) The expression of NTCP was upregulated in both HepaRG and imHC. NTCP normalized with GAPDH was higher in imHC than in HepaRG. (C) A highly glycosylated form of NTCP was observed after maturation. (D) Percentage of NTCP elevation in d-imHC was evaluated using flow cytometry. Data are presented as mean ± SD. *, **, and *** represent statistical difference with p < 0.05, p < 0.01, and p < 0.001, respectively. Abbreviations: NTCP = sodium taurocholate cotransporting polypeptide; GAPDH = glyceraldehyde 3-phosphate dehydrogenase; NTCP-FITC = fluorescein isothiocyanate-labeled NTCP. Please click here to view a larger version of this figure.
Figure 2: Prophylactic CCM treatment decreased viral entry, intracellular HBV DNA, and TCA uptake activity. (A) d-imHC were pretreated with CCM for 2 h prior to the inoculation with HBV. (B) Anti-HBV entry activity was determined by immunofluorescence staining on day 7 post infection. (C) A schematic schedule of HBV binding protocol is presented. (D) The level of bound HBV DNA on hepatocytes was evaluated and compared with 4 µM CsA and the classical HBV entry inhibitor 25 units/mL heparin. (E) The effect of the 10-30 µM CCM on TCA uptake inhibition was mediated through the NTCP receptor. Abbreviations: CCM = curcumin; HBV = hepatitis B virus; TCA = taurocholic acid; d-imHC = differentiated imHC; CsA = cyclosporine A; HP = heparin. Please click here to view a larger version of this figure.
Figure 3: The affinity of NTCP to individual molecules (CsA or candidate compound) was demonstrated using ITC. The ITC profile for the combination of NTCP with either (A) CsA or (B) curcumin was generated from sequential injection of a compound (150 μM CsA or candidate) into the NTCP solution (15 µM of NTCP, pH 7.0, 25 °C). The calorimetric raw data between differential power (µcal/s) and time (min) were plotted. Data were analyzed based on a one-site binding model where the solid lines indicated the best-fit results. During the injection of ligands (CSA or curcumin) into the NTCP solution, the enthalpy (ΔH) changed after an increase in the ligand concentration in the sample cell. These data indicate that the binding reaction occurred. Abbreviations: CsA = cyclosporine A; NTCP = sodium taurocholate cotransporting polypeptide; ITC = isothermal titration calorimetry; DP = differential power. Please click here to view a larger version of this figure.
Protein | Ligand | Binding constant (KD) | Enthalpy change (ΔH) | Entropy change (ΔTΔS) | Gibbs Free Energy change (ΔG) | Type of binding |
(M-1) | (kcal/mol) | (kcal/mol) | (kcal/mol) | |||
Human NTCP (SLA10A1) | CCM | 1.21 ×10-6 | -1 | 0.6 | -0.39 | Hydrogen bond |
Human NTCP (SLA10A1) | TCA | 1 ×10-9 | 80 | -96 | -16 | Hydrophobic interaction |
Table 1: Thermodynamic parameters resulting from the interaction between NTCP and individual compounds (CCM and TCA) using ITC. Abbreviations: NTCP = sodium taurocholate cotransporting polypeptide; CCM = curcumin; TCA = taurocholic acid; ITC = isothermal titration calorimetry.
HBV infection is initiated via low-affinity binding to heparan sulfate proteoglycans (HSPGs) on hepatocytes25, followed by the binding to NTCP with subsequent internalization through endocytosis26. As NTCP is a crucial receptor for HBV entry, targeting HBV entry can be clinically translated to diminish de novo infection, mother-to-child transmission (MTCT), and recurrence after liver transplantation. Interrupting viral entry would be a feasible alternative cure for chronic HBV infection.
Some critical steps in the above-mentioned protocol are summarized here. While preparing host cells, ensure that hepatocytes reach 100% confluency before adding the maturation medium with 2% DMSO. Culturing subconfluent hepatocytes in 2% DMSO will lead to cell death and detachment. The maturation period can be extended up to 4 weeks to maximize NTCP expression27,28, although a 2 week maturation period is recommended.
Three techniques have been proposed for the quantification of cellular NTCP expression. It is recommended that users select the technique they are most familiar with; here, we preferred flow cytometry to western blot analysis because it is convenient, quick, and easy.
For the production of the cell culture-derived HBV particles (HBVcc), HepG2.2.15 was derived from HepG2 by transient transfection with HBV full-length genome together with the geneticin (G418) resistance gene29. The culture medium should contain 380-500 µg/mL geneticin, otherwise the cells will not produce HBVcc. The low-protein-binding, 0.45 µm filter should be used to clarify the supernatant to avoid losing HBV yield. For concentrating HBV, polyethylene glycol (PEG) was used with a standard centrifuge. HBV particles can also be concentrated using a sucrose gradient and ultracentrifugation. Multiple freeze and thaw cycles should be avoided since the infectivity will be decreased.
In the anti-HBV entry assay, the hepatocytes must reach 100% confluence before maturation induction. This protocol was developed based on prophylactic treatment. Host cells were exposed to the candidate compounds for 2 h prior to infection with HBV12. After 7 days post infection, HBV infectivity was evaluated using immunofluorescence. All components, concentrations of the blocking solution, primary and secondary antibodies, and washing steps must be optimized to achieve the best result with minimal non-specific staining. Here, we have provided optimized concentrations and techniques.
The TCA uptake assay protocol describes the gold standard for the inhibition of NTCP transport. We modified the protocol to avoid radioactive TCA. The critical steps for the TCA uptake assay are cell washing and harvesting. All these procedures must be performed on ice all the time to prevent further cellular activity-internalization of the compound. After homogenizing the cells and pelleting cell debris, the supernatant must be gently aspirated without disrupting the pellet. If the user's facility permits the use of the liquid scintillation technique, 3H-taurocholic acid is commonly used in TCA transport and uptake studies. As we validated TCA uptake using ELISA, this alternative technique can substitute the use of the radioactive compound.
ITC is a biophysical method widely used to determine the thermodynamic parameters linked to the interactions between soluble proteins and small molecular weight ligands30,31. This technique can be used to investigate the interaction of various ligands with a small molecule or protein. ITC is a direct and noninvasive method with no additional steps for signal detection, no molecular weight limitations, and no labeling, immobilization, or any other chemical modification. The limitation of ITC is the prerequisite of high concentrations of the interacting materials. Protein and ligand have to be highly purified and sufficiently soluble in water (10-100 µM) at 25 °C for 1 h with stirring. The unstable protein solution can be detected through heat signal change as a function of time.
Human enhanced-NTCP hepatocytes provide an alternative model for the in vitro study of HBV infection but are similar to HepaRG and primary human hepatocytes. These host models are hampered by their limited reliability and susceptibility8,33. The inefficient HBV propagation in HepG2-NTCP or Huh7-NTCP cells was evidenced by low HBV inoculum derived from HBVcc-producing cells. In several studies, HBV was used to infect the target cells at an MOI of 1,00033,34. The subviral particles in HBVcc contain HBV envelope protein (L/M/S) and competitively bind NTCP, resulting in decreased infectivity35. To overcome this limitation, this protocol modified HepaRG or imHC culture with a maturation protocol to maximize NTCP levels. HBVcc derived from HepG2.2.15 was concentrated before being used as inoculum. The HBV infectivity was higher than 80% in differentiated imHC based on the measurement of the HBV core antigen11.
We have described the identification of anti-HBV compounds targeting NTCP to interrupt viral entry and adopted a hepatic maturation procedure to increase NTCP levels. To identify entry inhibitors, hepatocytes were pretreated with candidate compounds for 2 h before the infection with HBV at 4 °C to allow viral binding but not entry. The decrease in HBV infectivity was evaluated on day 7 post infection. The representative data included cyclosporine A, a classical NTCP inhibitor, as a positive control to validate the model.
Since NTCP facilitates both transporter- and receptor-mediated endocytosis functions, HBV entry inhibitors could target at least one of these mechanisms. The inhibition of NTCP transport can be evaluated using a TCA uptake assay based on ELISA, eliminating the radioactive TCA from the classical protocol36. The affinity between NTCP and the entry inhibitor was measured using ITC. This technique provided essential thermodynamic parameters, including the stoichiometry (n), dissociation constant (KD), change in free energy (ΔG), enthalpy (ΔH), entropy (ΔS), and heat capacity of binding (ΔCp). Various anti-HBV agents have been identified by molecular docking, such as quercetin, rutin, hesperidin, and luperol, that could specifically bind HBV reverse transcriptase comparable to lamivudine, a nucleoside analog. The compounds were investigated using ITC and exhibited negative Gibb energy (−ΔG) ranging from −9.3 to −5.2 kcal/mol and KD of 1 × 10-6 to 1 × 10-3 M with HBV polymerase37. Taken together, the data obtained from these aforementioned assays will help screen the antiviral efficacy of candidate compounds that act as HBV entry inhibitors. The established protocol will be useful for discovering novel NTCP antagonists/inhibitors. The suppression of viral entry could be useful in prophylactic and therapeutic treatment.
The authors have nothing to disclose.
This research project is supported by Mahidol University and the Thailand Science Research and Innovation (TSRI) separately awarded to A. Wongkajornsilp and K. Sa-ngiamsuntorn. This work was financially supported by the Office of National Higher Education Science Research and Innovation Policy Council through the Program Management Unit for Competitiveness (grant number C10F630093). A. Wongkajornsilp is a recipient of a Chalermprakiat grant of the Faculty of Medicine Siriraj Hospital, Mahidol University. The authors would like to thank Miss Sawinee Seemakhan (Excellent Center for Drug Discovery, Faculty of Science, Mahidol University) for her assistance with the ITC technique.
Cell lines | |||
HepaRG Cells, Cryopreserved | Thermo Fisher Scientific | HPRGC10 | |
Hep-G2/2.2.15 Human Hepatoblastoma Cell Line | Merck | SCC249 | |
Reagents | |||
4% Paraformadehyde Phosphate Buffer Solution | FUJIFLIM Wako chemical | 163-20145 | |
BD Perm/Wash buffer | BD Biosciences | 554723 | Perm/Wash buffer |
Cyclosporin A | abcam | 59865-13-3 | |
EDTA | Invitrogen | 15575-038 | 8 mM |
G 418 disulfate salt | Merck | 108321-42-2 | |
Halt Protease Inhibitor Cocktail EDTA-free (100x) | Thermo Scientific | 78425 | |
HEPES | Merck | 7365-45-9 | |
illustraTM RNAspin Mini RNA isolation kits | GE Healthcare | 25-0500-71 | |
illustra RNAspin Mini RNA Isolation Kit | GE Healthcare | 25-0500-71 | |
ImProm-II Reverse Transcription System | Promega | A3800 | |
KAPA SYBR FAST qPCR Kit | Kapa Biosystems | KK4600 | |
Lenti-X Concentrator | Takara bio | PT4421-2 | concentrator |
Luminata crescendo Western HRP substrate | Merck | WBLUR0100 | |
Master Mix (2x) Universal | Kapa Biosystems | KK4600 | |
Nucleospin DNA extraction kit | macherey-nagel | 1806/003 | |
Phosphate buffered saline | Merck | P3813 | |
Polyethylene glycol 8000 | Merck | 25322-68-3 | |
ProLong Gold Antifade Mountant | Thermo scientific | P36930 | |
Recombinant NTCP | Cloud-Clone | RPE421Hu02 | |
RIPA Lysis Buffer (10x) | Merck | 20-188 | |
TCA | Sigma | 345909-26-4 | |
TCA Elisa kit | Mybiosource | MB2033685 | |
Triton X-100 | Merck | 9036-19-5 | |
Trypsin-EDTA | Gibco | 25200072 | Dilute to 0.125% |
Antibodies | |||
Anti-NTCP1 antibody | Abcam | ab131084 | 1:100 dilution |
Anti-GAPDH antibody | Thermo Fisher Scientific | AM4300 | 1:200,000 dilution |
HRP-conjugated goat anti-rabbit antibody | Abcam | ab205718 | 1:10,000 dilution |
HRP goat anti-mouse secondary antibody | Abcam | ab97023 | 1:10,000 dilution |
Goat anti-Rabbit IgG Secondary Antibody, Alexa Fluor 488 | Invitrogen | A-11008 | 1:500 dilution |
Reagent composition | |||
1° Antibody dilution buffer | |||
1x TBST | |||
3% BSA | Sigma | A7906-100G | Working concentration: 3% |
Sodium azide | Sigma | 199931 | Working concentration: 0.05% |
Hepatocyte Growth Medium | |||
DME/F12 | Gibco | 12400-024 | |
10% FBS | Sigma Aldrich | F7524 | |
1% Pen/Strep | HyClon | SV30010 | |
1% GlutaMAX | Gibco | 35050-061 | |
Hepatic maturation medium | |||
Williams’ E medium | Sigma Aldrich | W4125-1L | |
10% FBS | Sigma Aldrich | F7524 | |
1% Pen/Strep | HyClon | SV30010 | |
1% GlutaMAX | Gibco | 35050-061 | |
5 µg/mL Insulin | Sigma Aldrich | 91077C-100MG | |
50 µM hydrocotisone | Sigma Aldrich | H0888-1g | |
2% DMSO | PanReac AppliChem | A3672-250ml | |
IF Blocking solution | |||
1x PBS | Gibco | 21300-058 | |
3% BSA | Sigma | A7906-100G | Working concentration: 3% |
0.2% Triton X-100 | Sigma | T8787 | Working concentration: 0.2% |
RIPA Lysis Buffer Solution | Merck | 20-188 | Final concentration: 1X |
Protease Inhibitor Cocktail | Thermo Scientific | 78425 | Final concentration: 1X |
Na3VO4 | Final concentration: 1 mM | ||
PMSF | Final concentration: 1 mM | ||
NaF | Final concentration: 10 mM | ||
Western blot reagent | |||
10x Tris-buffered saline (TBS) | Bio-Rad | 170-6435 | Final concentration: 1X |
Tween 20 | Merck | 9005-64-5 | |
1x TBST | 0.1% Tween 20 | ||
1x PBS | Gibco | 21300-058 | |
Pierce BCA Protein Assay Kit | Thermo Fisher Scientific | A53225 | |
Polyacrylamide gel | Bio-Rad | 161-0183 | |
Ammonium Persulfate (APS) | Bio-Rad | 161-0700 | Final concentration: 0.05% |
TEMED | Bio-Rad | 161-0800 | Stacker gel: 0.1%, Resolver gel: 0.05% |
2x Laemmli Sample Buffer | Bio-Rad | 161-0737 | Final concentration: 1X |
Precision Plus Protein Dual Color Standards | Bio-Rad | 161-0374 | |
WB Blocking solution/ 2° Antibody dilution buffer | |||
1x TBST | |||
5% Skim milk (nonfat dry milk) | Bio-Rad | 170-6404 | Working concentration: 5% |
1x Running buffer 1 L | |||
10x Tris-buffered saline (TBS) | Bio-Rad | 170-6435 | Final concentration: 1X |
Glycine | Sigma | G8898 | 14.4 g |
SDS | Merck | 7910 | Working concentration: 0.1% |
Blot transfer buffer 500 mL | |||
10x Tris-buffered saline (TBS) | Bio-Rad | 170-6435 | Final concentration: 1X |
Glycine | Sigma | G8898 | 7.2 g |
Methanol | Merck | 106009 | 100 mL |
Mild stripping solution 1 L | Adjust pH to 2.2 | ||
Glycine | Sigma | G8898 | 15 g |
SDS | Merck | 7910 | 1 g |
Tween 20 | Merck | 9005-64-5 | 10 mL |
Equipments | |||
15 mL centrifuge tube | Corning | 430052 | |
50 mL centrifuge tube | Corning | 430291 | |
Airstream Class II | Esco | 2010621 | Biological safety cabinet |
CelCulture CO2 Incubator | Esco | 2170002 | Humidified tissue culture incubator |
CFX96 Touch Real-Time PCR Detector | Bio-Rad | 1855196 | |
FACSVerse Flow Cytometer | BD Biosciences | 651154 | |
Graduated pipettes (10 mL) | Jet Biofil | GSP010010 | |
Graduated pipettes (5 mL) | Jet Biofil | GSP010005 | |
MicroCal PEAQ-ITC | Malvern | Isothermal titration calorimeters | |
Mini PROTEAN Tetra Cell | Bio-Rad | 1658004 | Electrophoresis chamber |
Mini Trans-blot absorbent filter paper | Bio-Rad | 1703932 | |
Omega Lum G Imaging System | Aplegen | 8418-10-0005 | |
Pipette controller | Eppendorf | 4430000.018 | Easypet 3 |
PowerPac HC | Bio-Rad | 1645052 | Power supply |
PVDF membrane | Merck | IPVH00010 | |
T-75 A91:D106flask | Corning | 431464U | |
Trans-Blot SD Semi-Dry Transfer Cell | Bio-Rad | 1703940 | Semi-dry transfer cell |
Ultrasonic processor (Vibra-Cell VCX 130) | Sonics & Materials | ||
Versati Tabletop Refrigerated Centrifuge | Esco | T1000R | Centrifuge with swinging bucket rotar |