Sumit Kumar Mandal†
a,
Mohammed Muzaffar-Ur-Rehman†b,
Sonakshi Puria,
Banoth Karan Kumarb,
Pankaj Kumar Sharmaa,
Murugesan Sankaranarayanan
b and
P. R. Deepa
*a
aBiochemistry and Enzyme Biotechnology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani-333 031, Rajasthan, India. E-mail: p20200425@pilani.bits-pilani.ac.in; pankajsharma@pilani.bits-pilani.ac.in; deepa@pilani.bits-pilani.ac.in
bMedicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani-333 031, Rajasthan, India. E-mail: p20210457@pilani.bits-pilani.ac.in; p20180042@pilani.bits-pilani.ac.in; murugesan@pilani.bits-pilani.ac.in
First published on 4th April 2025
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated nuclear receptors with a crucial regulatory role in carbohydrate and lipid metabolism and are emerging druggable targets in “metabolic syndrome” (MetS) and cancers. However, there is a need to identify ligands that can activate specific PPAR subtypes, particularly PPARβ/δ, which is less studied compared with other PPAR isoforms (α and γ). Herein, using the drug-repurposing approach, the ZINC database of clinically approved drugs was screened to target the PPARβ/δ receptor through high-throughput-virtual-screening, followed by molecular docking and molecular dynamics (MD) simulation. The top-scoring ligands were subjected to drug-likeness analysis. The hit molecule was tested in an in vitro model of NAFLD (non-alcoholic fatty liver disease). The top five ligands with strong binding affinity towards PPARβ/δ were canagliflozin > empagliflozin > lumacaftor > eprosartan > dapagliflozin. RMSD/RMSF analysis demonstrated stable protein–ligand complexation (PLC) by the top-scoring ligands with PPARβ/δ. In silico ADMET prediction analysis revealed favorable pharmacokinetic profiles of these top five ligands. Canagliflozin showed significant (P < 0.001) dose-dependent decrease in lipid accumulation and the associated oxidative stress-inflammatory response, suggesting its promising anti-steatotic potential. These outcomes pave the way for further validation and development of PPAR activity-modulating therapeutics.
The activation mechanism is quite similar among the three PPAR isoforms, yet they show some differences. For example, PPARα is activated by fatty acids, while PPARγ is activated by thiazolidinedione drugs and is reported to be regulated by a natural product, chelerythrine, resulting in an anti-diabetic effect.3,4 PPARβ/δ, alternatively, has broad ligand specificity, including for fatty acids, eicosanoids, and ligands such as leukotriene B4 and prostacyclin.5 When these ligands bind to PPARs' ligand binding domain, a conformational change is induced, enabling the PPARs to interact with co-activators and co-repressors. Once activated, the PPARs bind to specific DNA sequences, known as PPAR response elements (PPREs), within the promotor region of the target gene, resulting in the transactivation of the gene.6 Transrepression is another PPAR mechanism that occurs without a ligand and is involved in the anti-inflammatory effects of PPARs.7
The multi-faceted regulation of lipid metabolism and associated energy mechanisms that have been reported2,8 are compiled in Fig. 1. It shows that activation of PPARβ/δ inhibits the STAT3/SOCS3 pathway and improves insulin sensitivity in adipose tissue.9 A recent study has shown that treatment with leptin increases the expression of FGF21 in visceral adipose tissue by activating PPARβ/δ.10 GW501516, a PPARβ/δ agonist, lowers triglyceride levels in hepatic tissue by restoring hepatic AMPK levels via activation of the PGC-1/α-lipin-1/PPARα pathway in a rat model.11,12 Similarly, GW501516 reduces multiple clinical features of metabolic syndrome by increasing the oxidation of fatty acids in skeletal muscles.13 Metabolic syndrome represents a complement of related disorders that include hyperlipidemia, diabetes mellitus, hypertension, and cardiovascular disorders, which may have the hepatic complication of non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH).
Studies have also shown that lipolytic functions of PPARβ/δ, which have a potentially beneficial effect on fatty liver disease, are mediated by activating the autophagy pathway and attenuating endoplasmic reticulum stress in obese mice.14,15 Other beneficial effects reported with respect to PPARβ/δ agonists are mitigation of oxidative stress,16 inflammation17,18 and mitochondrial biogenesis,19 which make it a promising target for identifying PPARβ/δ agonists that could potentially treat metabolic disorders.20 One of the risks of metabolic syndrome is the development of cancers. In this context, it is important to note that PPARβ/δ has also been evaluated for its potential anti-cancer effects.16,21–23
Recently, using a machine learning approach, newer PPARβ/δ agonists were reported.24 However, there are limited reports on PPARβ/δ agonists, which prompted the present study. Herein, we screened a database with clinically approved drugs as potential pharmacological candidates using high-throughput virtual screening (HTVS) technique, molecular docking/dynamics simulations and pharmacokinetic profiling. This was followed by biochemical testing of a hit-ligand agonist molecule in an in vitro steatotic cell culture model of NAFLD.
HepG2 cells were sourced from the National Centre for Cell Science (NCCS – Pune, India). The minimum essential medium (MEM) was procured from Gibco Life Technologies (Carlsbad, USA), while fetal bovine serum (FBS), oleic acid, and Oil Red O (ORO) powder were obtained from HiMedia (Mumbai, India). All other chemicals were purchased from Sigma-Aldrich (Schnelldorf, Germany) and were of the highest analytical grade.
Prior to docking, proteins must be prepared since their crystal structures might have issues including improper bonding orders, missing side chains, loops, and atoms. The Protein Preparation Wizard was used to create the crystal structure of the two proteins, and the purpose of this process was to examine the protein structures and confirm the assignment of bonds and binding orders, the insertion of hydrogens, the identification of disulfide bonds, the completion of any missing loops or side chains, and the rectification of any incorrectly labelled components. Moreover, the OPLS-2005 force field was used to minimize crystal protein structures. Heavy atoms in the structures were constrained to relieve torsional tension using a harmonic potential of 25 kcal mol−1 throughout the procedure, whereas hydrogen was left un-constrained.27
ΔG = −RT![]() ![]() |
In the equation, ΔG represents the changes in docking binding energy, T represents temperature, R represents the Boltzmann gas constant (R = 1.987 cal mol−1 K−1), and Kd represents dissociation constant. This equation enables the estimation of binding affinity based on the docking binding energy.30
The analysis focused on the ligand molecules that showed significant results. The prepared system underwent energy minimization for 100 picoseconds (ps). The final production run consisted of a 100 ns MD simulation conducted under constant temperature and pressure (NPT ensemble) with a relaxation period of 1 ps and a temperature of 300 K. The Nose–Hoover chain coupling technique was used for temperature control. Bonding interactions were calculated with a time step of 2 fs using the reversible reference system propagator algorithm (RESPA) integrator.37,38 The trajectories obtained from the simulation were examined for the root mean square deviation (RMSD) and root mean square fluctuation (RMSF) of the protein–ligand complex, as well as the RMSF of the ligand itself.
Canagliflozin, identified as the top scoring ligand, was experimentally evaluated to evaluate its effectiveness in reducing steatosis in a cell culture model of non-alcoholic fatty liver disease (NAFLD). The subsequent sections detail the experimental confirmation of the pharmacological efficacy of canagliflozin.
To prepare canagliflozin for experiments, it was initially dissolved in dimethyl sulphoxide (DMSO) to prepare a stock solution (SS) with a concentration of 1 mM. Working concentrations of canagliflozin, ranging from 5–100 μM, were then prepared from the stock solution, ensuring that the final concentration of DMSO in the test dilutions did not exceed 1%. Confluent monolayers of HepG2 cells were grown in 96-well plates for 24 hours. These cells were treated with different concentrations of canagliflozin in triplicate and incubated at 37 °C in a CO2 environment for 24 hours. After the 24 hour treatment period, the supernatants were aspirated from the wells, and 100 μL of MTT solution (2 mg mL−1) was added to each well. The plates were then incubated for 3 hours at 37 °C, followed by the addition of 100 μL DMSO to dissolve the formazan crystals. Optical density was measured at 570 nm using a multi-well spectrophotometer (Multiskan, Thermo Fisher Scientific, Waltham, MA). The safe dose concentration, often considered the concentration at which 80% of cells remain viable, was determined using non-linear regression curve analysis within GraphPad Prism statistical software.27
Following treatment, the cells were treated with lysis buffer, homogenized, and centrifuged at 13000×g for 15 minutes at 4 °C for the lipid peroxidation assay using a modified protocol. The resulting supernatant was collected for subsequent analysis. The cell lysates from each experimental group were adjusted to contain an equal quantity of protein (1 mg mL−1) and were treated with 250 μL of 10% trichloroacetic acid (TCA). Afterward, these lysates were mixed with 375 μL of 1% (w/v) thiobarbituric acid (TBA) under acidic conditions. This solution was heated in a boiling water bath for 15 minutes, promoting the formation of a pink-colored adduct. The absorbance of this resultant adduct was measured spectrophotometrically at 530 nm using the Multiskan FC (Thermo Scientific, DE). The values obtained were expressed as micromoles (μM) of malondialdehyde per milligram (mg) of protein.28
S. no | Ligands | Drug name | PPARβ/δ (kcal mol−1) | MM-GBSA (kcal mol−1) |
---|---|---|---|---|
1 | ZINC000043207238 | Canagliflozin | −13.907 | −73.80 |
2 | ZINC000036520252 | Empagliflozin | −12.756 | −77.09 |
3 | ZINC000064033452 | Lumacaftor | −12.509 | −58.09 |
4 | ZINC000029319828 | Eprosartan | −11.944 | −54.77 |
5 | ZINC000003819138 | Dapagliflozin | −11.86 | −71.22 |
6 | Co-crystal ligand | (2,3-Dimethyl-4-{[2-(prop-2-yn-1-yloxy)-4-{[4-(trifluoromethyl)phenoxy]methyl}phenyl]sulfanyl}phenoxy)acetic acid | −12.74 | −96.96 |
Table 2 summarizes the docking interactions and bond distances of the top five ligands against PPARβ/δ. Canagliflozin, a sodium-glucose transport protein sub-type 2 inhibitor used for the treatment of type 2 diabetes, exhibited significant docking scores against PPARβ/δ (−13.907 kcal mol−1). When canagliflozin was docked against PPARβ/δ, it formed four hydrogen bond interactions with Thr288, Thr289, His449 and Phe282. Empagliflozin, a sodium-glucose co-transporter 2 inhibitor used to improve glycemic control in adults with type 2 diabetes mellitus, showed a similar interaction pattern. It formed hydrogen bonds with Thr288 and halogen bonds with Thr288, His449, Lys367. Lumacaftor, used for the treatment of cystic fibrosis, interacted with PPAR β/δ through hydrogen bonds with His449 and formed one halogen bond with His449. Eprosartan, an angiotensin II receptor inhibitor used for the treatment of high blood pressure, formed two aromatic bond interactions with Cys285 and Phe327. It also exhibited one halogen bond interaction with Thr289. Dapagliflozin, a sodium-glucose transport protein sub-type 2 inhibitor used in type 2 diabetes treatment, formed hydrogen bonds with Thr289 and one aromatic H-bond with Phe327 (Fig. 2). Overall, the docking interactions and bond distances of the top five drugs with different PPARβ/δ molecules are summarized in Table 2.
Ligands | PPARβ/δ | ||
---|---|---|---|
Residues | Type of interactions | Distance (Å) | |
a Abbreviations: PPARβ/δ = peroxisome proliferator activated receptor β/δ; kcal = kilocalorie; mol = mole. | |||
ZINC000043207238 (canagliflozin) | THR288 | Halogen bond | 1.93 |
THR289 | H-Bond | 1.81 | |
HIS449 | Halogen bond | 2.00 | |
HIS449 | Halogen bond | 2.50 | |
PHE282 | Halogen bond | 2.58 | |
PHE282 | Halogen bond | 2.12 | |
PHE282 | Halogen bond | 2.00 | |
PHE282 | Halogen bond | 2.44 | |
ZINC000036520252 (empagliflozin) | THR288 | H-Bond | 2.12 |
THR288 | Halogen bond | 3.06 | |
HIS449 | Halogen bond | 5.71 | |
LYS367 | Halogen bond | 6.68 | |
ZINC000064033452 (lumacaftor) | HIS449 | H-Bond | 1.92 |
HID449 | Halogen bond | 3.73 | |
ZINC000029319828 (eprosartan) | CYS285 | Aromatic H-bond | 2.13 |
PHE327 | Aromatic H-bond | 2.43 | |
THR289 | Halogen bond | 2.07 | |
ZINC000003819138 (dapagliflozin) | PHE327 | Aromatic H-bond | 3.64 |
THR289 | H-Bond | 2.74 | |
Co-crystal ligand | THR288 | H-Bond | 2.00 |
TYR473 | H-Bond | 1.82 | |
HIS449 | H-Bond | 1.80 | |
HIS449 | Pi–Pi | 4.47 | |
THR288 | H-Bond | 2.00 |
![]() | ||
Fig. 2 Representation of 2D and 3D docked poses of canagliflozin, empagliflozin, lumacaftor, eprosartan, and dapagliflozin in the active site of PPARβ/δ (PDB: 3GZ9). The active site residues are represented by coloured discs, while interaction bonds are depicted as dashed lines. |
![]() | ||
Fig. 3 (i) RMSD plot depicting the deviation of the protein and ligands during the simulation. 3GZ9a indicates the protein with the hit compound, while 3GZ9b indicates the protein with the co-crystal ligand, orange line indicates the co-crystal ligand, and yellow indicates the hit compound. (ii) RMSF plot depicting fluctuations of the protein residues during simulation. 3GZ9a indicates the protein with respect to the co-crystal ligand and 3GZ9b with respect to the hit compound. (iii) Depiction of the radius of gyration (Rg) graph of the top-scoring ligand (canagliflozin) complexed with 3GZ9 and co-crystal ligand complexed with 3GZ9 during 100 ns MD Simulation. 3GZ9a indicates the protein with respect to the co-crystal ligand and 3GZ9b with respect to the hit compound. |
The radius of gyration (Rg), which determines the compactness of the protein, shows very less deviation between 5.1 Å and 6.0 Å (Fig. 3(iii)). In the case of the hit compound, the docked complex had only one H-bond interaction with Gln286 with a distance of 1.81 Å. After initiating dynamics, the bond between the 5th hydroxy group of glucopyranose of the hit molecule and Gln286 disappeared, and the interaction of His323 with the 3rd hydroxy group was seen, resulting in a change in conformation of the docked ligand, thereby increasing the RMSD to 2.4 Å (Fig. 3). Furthermore, new interactions were observed with Leu339, Val341, Lys367, His449 and Tyr473 until 40 ns. After 55 ns, two additional residues, Gln286 and His323, were observed to form strong interactions. Among these, residues that contributed to direct interactions with the ligand include Gln286, Tyr473, and His449, with 24%, 11%, and 36%, respectively. The residue His323 contributed to water-mediated interactions (11%). In addition, other residues such as Leu339, Val341, and Lys367 formed water-mediated interactions, and their contributions were less than 10% (Fig. 4(ii)). The RMSF plot (Fig. 3(ii)) also shows very low fluctuations of the residues under interaction with the ligand (<2.0 Å). The deviations in the Rg was <1.5 Å (between 4.0–5.25 Å) (Fig. 3(iii)), and the overall deviations in the RMSD plot were not more than 2.0 Å (1.5–3.2 Å). This indicates that the ligand is stable in the active site pocket and is retained within the pocket till the end of the simulation.
For comparison, the PPARβ/δ protein in its apo-form was subjected to dynamics studies for 100 ns. As depicted in Fig. 3(i), the RMSD of the apo-protein has a deviation ranging from 2.0 Å to 5.0 Å, while the protein when complexed with the co-crystal or the hit compound has a lower deviation (2.0–3.0 Å). This indicates that the presence of ligands in the active site of the protein resulted in stability, as evident from the decreased protein deviation or residual fluctuation observed in the respective RMSD and RMSF plots (Fig. 3(i and ii). We also observed most of the interactions with the co-crystal ligand are due to the acetic acid group, while the hit compound engages via hydroxyl groups on its pyranose rings. The co-crystal and the hit compound showed similar behaviour in their dynamics and interaction pattern, which suggested that the hit compound (canagliflozin) could have agonistic behaviour similar to that of the co-crystal ligand, which encouraged us to further carry out in vitro investigation.
Table 3 presents a summary of the predicted ADMET characteristic results for the top five PPARβ/δ agonists. Based on the comparison between the marginal value and the actual value, ADMET results were interpreted as high Caco-2 permeability predicted value >0.90, intestinal absorption less than 30% is considered to be poorly absorbed, human VDss is low if it is less than 0.71 L kg−1 and high if it is more than 2.81 L kg−1, BBB permeability logBB >0.3 is considered to cross BBB, and log
BB 1 are poorly distributed. Compounds with a log
PS of >2 and <3 have the ability to permeate the central nervous system (CNS). The favourable pharmacokinetic profile of the top scoring ligands, as revealed by the above ADMET analysis, is yet another point for considering these ligands as potential PPARβ/δ agonist drug candidates.
Parameters | Absorption | Distribution | ||||||||
---|---|---|---|---|---|---|---|---|---|---|
Water solubility (log![]() |
Caco2 permeability (log![]() |
Intestinal absorption (% human) | Skin permeability (log![]() |
P-Glycoprotein substrate | P-Glycoprotein inhibitor | VDss (human) (log L kg−1) | Fraction unbound (human) (Fu) | BBB permeability (log![]() |
CNS permeability (log![]() |
|
Canagliflozin | −4.21 | 0.85 | 0.85 | −2.7 | Yes | Yes | −0.71 | 0.08 | −0.99 | −3.02 |
Empagliflozin | −3.56 | −0.02 | 55.87 | −2.7 | Yes | Yes | −0.38 | 0.07 | −1.10 | −3.51 |
Lumacaftor | −3.18 | 0.63 | 78.71 | −2.7 | Yes | No | −1.46 | 0.03 | −0.47 | −2.81 |
Eprosartan | −2.89 | 0.67 | 57.17 | −2.7 | Yes | No | −0.72 | 0.14 | −0.87 | −2.47 |
Dapagliflozin | −3.43 | 0.94 | 55.86 | −2.7 | Yes | Yes | −0.33 | 0.08 | −0.99 | −3.34 |
Parameters | Metabolism | Excretion | Toxicity | Lipinski's rule of five | |||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
CYP substrate | CYP inhibitors | Total clearance (log mL min−1 kg−1) | Renal OCT2 substrate | AMES toxicity | Hepatotoxicity | Skin sensitization | Molecular weight | log![]() |
Rotatable bonds | Acceptors | Donors | Surface area | |
Canagliflozin | No | No | 0.035 | No | No | No | No | 444.52 | 2.97 | 5 | 6 | 4 | 183.56 |
Empagliflozin | No | No | 0.40 | No | No | No | No | 450.91 | 1.613 | 6 | 7 | 4 | 185.31 |
Lumacaftor | No | No | −0.10 | No | No | Yes | No | 452.41 | 4.75 | 5 | 5 | 2 | 186.53 |
Eprosartan | No | No | 0.67 | No | No | Yes | No | 424.52 | 4.74 | 10 | 5 | 2 | 178.70 |
Dapagliflozin | No | No | 0.19 | No | No | No | No | 408.88 | 1.84 | 6 | 6 | 4 | 168.47 |
The ADMET analysis of canagliflozin in our study is in resonance with other research reports. Studies have shown that canagliflozin is rapidly and completely absorbed from the gastrointestinal tract, with a peak plasma concentration reached within 1–2 hours after oral administration.42 Food does not significantly affect its absorption. Canagliflozin is highly protein-bound (99%) in the plasma, mainly to albumin. It has a large volume of distribution, indicating that it gets distributed extensively to tissues.43 Canagliflozin is primarily metabolized in the liver via glucuronidation, a phase II metabolic pathway. The major enzymes involved are UGT1A9 and UGT2B4, whereas CYP3A4, a phase I enzyme, plays a minor role in its metabolism.44 Canagliflozin is mainly excreted in the feces (about 50%) as an unchanged drug and metabolite. About 33% is excreted in the urine as metabolites. The terminal half-life of canagliflozin is about 10–13 hours.45 Overall, the outcomes of ADMET analysis of canagliflozin in the present study and from other reports indicate that it has favorable pharmacokinetic properties, which contribute to its efficacy and safety profile in the treatment of type 2 diabetes and potentially other metabolic disorders.
To induce cellular lipid deposits in HepG2 hepatocytes, we supplemented them with free fatty acids (oleic acid) for 48 hours. Canagliflozin, identified as the top hit, demonstrated a concentration-dependent reduction in lipid accumulation in steatotic cells following a 24 hour treatment, which was evaluated using ORO staining (Fig. 5a and b). This anti-steatotic effect was comparable to that observed with the positive control saroglitazar (10 μM). Specifically, neutral lipids such as triglycerides, stained with ORO, were quantified spectrophotometrically at 492 nm. Fig. 5c illustrates a consistent reduction in triglyceride (TG) accumulation as the concentration of canagliflozin increased (p < 0.05). The doses of 10, 15, and 20 μM resulted in lipid accumulation reductions in the steatotic cells by 29.03%, 37.57%, and 48.45%, respectively, while saroglitazar, the reference anti-steatotic drug control, induced a 33.39% reduction.
Lipotoxicity induced by steatosis resulted in oxidative stress in the hepatocytes, evident from an increase in malondialdehyde levels in the disease (control) group (Fig. 5d). This oxidative stress-induced lipid peroxidation was attenuated in the cells treated with canagliflozin. In this study, we observed a reduction in lipid peroxidation by 27.99%, 33.50%, and 46.10% when treated with 10, 15, and 20 μM of canagliflozin, respectively. This is similar to the oxidative reduction observed when cells were treated with the reference drug saroglitazar at 10 μM.
PPAR | Peroxisome proliferator-activated receptors |
PPREs | Peroxisome proliferator-activated receptor response elements |
FDA | Food and drug administration |
RXR | Retinoid X receptor |
NAFLD | Non-alcoholic fatty liver disease |
NASH | Non-alcoholic steatohepatitis |
HTVS | High throughput virtual screening |
MD | Molecular dynamics |
PDB | Protein Data Bank |
OPLS-2005 | Optimized potentials for liquid simulations 2005 |
SP | Standard precision |
XP | Extra precision |
RMSD | Root mean square deviation |
RMSF | Root mean square fluctuation |
Rg | Radius of gyration |
MM-GBSA | Molecular mechanics with generalised Born and surface area solvation |
RESPA | Reversible reference system propagator algorithms |
VSGB | Variable dielectric generalized Born |
Footnote |
† Authors contributed equally. |
This journal is © The Royal Society of Chemistry 2025 |