1. Introduction
Acute kidney injury (AKI) represents a critical global health challenge, with the Global Burden of Disease (GBD) 2021 report estimating 1.3–2.1 million annual deaths worldwide. The prevalence of AKI is reported to be approximately 10–15% in hospitalized patients and up to 50% in ICU patients [
1,
2]. AKI is characterized as a pathological condition marked by a rapid decline in glomerular filtration rate over a short period, resulting in alterations to kidney physiological function and tissue structure [
3]. The causative factors of AKI are recognized to include ischemia-reperfusion (IR), sepsis, and renal transplantation, along with other injuries, with IR being identified as one of the leading causes of AKI. When IR occurs in the kidneys, the oxygen supply is significantly reduced due to a dramatic decrease in blood flow, which subsequently induces oxidative stress, mitochondrial damage, and the production of large quantities of reactive oxygen species as well as pro-inflammatory cytokines, ultimately leading to renal injury [
4]. However, no specific or effective treatment has been developed to prevent or treat IR-induced AKI; therefore, the identification of safe and effective therapeutic drugs, in addition to the elucidation of their mechanisms, is considered critical.
The natural product oleanolic acid (OA) is a commonly occurring oleanane-type pentacyclic triterpene with a broad spectrum of anti-inflammatory, antioxidant, and hypoglycemic effects [
5,
6]. OA, typically found in its free form or as a glycoside, has been extensively studied for its good biological activity [
7,
8]. Additionally, OA can mitigate acute liver injury induced by chemicals [
9], enhance glutathione-mediated antioxidant mechanisms, and alleviate IR-induced myocardial injury [
10]. Although OA has achieved certain achievements in alleviating diseases, it is difficult to carry out clinical transformations and applications owing to its low solubility and dose-limiting side effects [
11,
12]. Therefore, the development of novel OA derivatives with enhanced efficacy and bioavailability is considered critical. This study developed a novel oleanolic acid derivative, DKS26, via C-12,13 hydrogenation and C-28 esterification [
13]. Meanwhile, the bioavailability of DKS26 was enhanced via lipid nanoparticle carrier technology, increasing its oral bioavailability from 5.81% to 29.47% [
13]. Since the anti-inflammatory and antioxidant effect of it is well-suited to the pathogenesis of IR-mediated AKI, the therapeutic effect was further validated in this model.
Swiss TargetPrediction (
http://swisstargetprediction.ch/ (accessed on 18 May 2023)) indicated that DKS26 may act on vitamin D receptors (VDR). VDR is a nuclear receptor belonging to the ligand-activating transcription factor family, and 1,25-(OH)
2D3 is the main ligand that activates VDR. VDR is expressed in both proximal and distal renal tubular epithelial cells as well as in podocytes. The study found that VDR expression was reduced in both diabetic nephropathy mouse models and renal ischemia-reperfusion rat models, and the administration of VDR activators could alleviate renal ischemia-reperfusion injury in rats [
14,
15]. Recent studies have shown that VDR can inhibit NF-κB-mediated inflammation and plays a protective role in the progression of kidney disease [
16,
17]. Mechanistically, IR-induced tissue damage can lead to increased release of inflammatory factors. Inflammatory factors induce Ikk-mediated phosphorylation of nuclear factor kappa B inhibitor alpha (IκBα) and subsequent proteasomal degradation through activation of the TNFR/TLR receptor, which releases Nuclear Factor Kappa-light-chain-enhancer of Activated B Cells (NF-κB) (P65/p50) for nuclear translocation [
18,
19], and upregulate the expression of pro-inflammatory genes (
TNFα,
CCL2), while VDR exerts its protective effect by inhibiting NF-κB P65 nuclear translocation [
10]. Therefore, an IR-induced AKI mouse model was used to assess the therapeutic value and possible mechanism of action of DKS26 in AKI. In this study, we found that DKS26 could counteract AKI by inhibiting oxidative stress, improving the inflammatory microenvironment, and attenuating tubular epithelial cell apoptosis. The key effect may be a protective role by stabilizing the VDR and inhibiting the inflammatory pathway of NF-κB P65.
3. Discussion
This study investigated the protective effects and underlying mechanisms of DKS26, a novel small-molecule compound, in AKI. Clinical observations indicate that patients with AKI typically exhibit abrupt renal dysfunction marked by elevated serum creatinine levels [
20]. In the experimental model, IR-AKI mice developed extensive tubular infarction, increased serum creatinine (
Figure 1), and significantly upregulated expression of NGAL and KIM1 at both protein and mRNA levels (
Figure 2), confirming renal impairment induced by IR. Treatment with DKS26 markedly reduced tubular injury, and partially recovered renal function (
Figure 1 and
Figure 2), suggesting its therapeutic potential in alleviating IR-induced AKI.
Previous studies indicate that AKI progression involves tubular epithelial cell damage, accompanied by proinflammatory mediator release [
20], macrophage infiltration [
4], apoptosis, and oxidative stress [
21,
22]. While ROS participate in physiological signaling under normal conditions, excessive ROS generation triggers oxidative stress and apoptosis [
22]. MDA, combined with antioxidant enzyme activity detection (T-SOD, CAT, GPx), is a standard method for assessing oxidative stress [
23]. Thus, this study employed these indicators alongside DCFH-DA and MitoSOX Red staining to quantify intracellular ROS. The results demonstrated that IR and H/R promoted ROS production in both kidney tissue and mRTECs. DKS26 reduced mitochondria-derived ROS, partially restored antioxidant capacity, alleviated oxidative stress, and reversed apoptosis (
Figure 3A–F,N–Q). Ischemia/Hypoxia induces mitochondrial damage and activates the PINK1/Parkin-dependent mitochondrial autophagy pathway [
24]. In this study, we also found that H/R induced mitochondrial damage and activated the PINK1/Parkin-dependent mitochondrial autophagy pathway; however, the increase in LC3Ⅱ and P62 indicated an increase in autophagosome formation but impaired lysosomal degradation, resulting in ineffective autophagic substrate removal (
Supplementary Figure S1B). In addition, Mitochondrial ROS overload promotes Cyto-c release and Caspase-3 activation, upregulating Bax and downregulating Bcl2 [
24]. Similarly, this study found that DKS26 may protect the structural and functional integrity of mitochondria, thereby reducing the release of mitochondrial damage signals and preventing cell apoptosis (
Figure 3N–Q).
Excessive ROS accumulation in pathological states directly activates the NF-κB pathway, driving inflammatory cytokine synthesis/secretion, renal inflammatory microenvironment formation, inflammatory cell infiltration, and tubular apoptosis [
25]. NF-κB activation is pivotal to IR-AKI progression. During IR-AKI, NF-κB-driven inflammatory cytokines (e.g., TNFα) activate Caspase-8, triggering the mitochondrial apoptotic pathway via proapoptotic Bax upregulation [
26,
27]. Consequently, we further explored DKS26’s anti-inflammatory and antiapoptotic effects. We demonstrated both in vivo and in vitro that both IR and H/R induced the expression of the inflammatory factors TNFα and MCP-1, the pro-apoptotic protein Bax, whereas DKS26 reduced the synthesis and secretion of inflammatory factors, improved the inflammatory microenvironment, and reduced apoptosis (
Figure 3). Collectively, DKS26 ameliorated IR and/or H/R-induced oxidative stress, inflammation, and apoptosis.
As a nuclear receptor with renoprotective anti-inflammatory properties, VDR modulates disease progression via direct interaction with NF-κB p65 subunits, as evidenced in disuse muscle atrophy models [
14,
28]. In the present study, Co-IP assays demonstrated H/R-induced VDR-
p-NF-κB P65
Ser311 complex formation, which was potentiated by DKS26 treatment (
Figure 4C). Functionally, DKS26 restored VDR expression while suppressing
p-NF-κB P65
Ser311 nuclear translocation and downstream proinflammatory gene transcription and itself (
Figure 4 and
Figure 5D,E). In vitro H/R models confirmed DKS26’s concentration-dependent inhibition of NF-κB p65 and
p-NF-κB P65
Ser311 protein expression (
Figure 5A–C). Structural stabilization assays revealed that DKS26 enhances VDR stability via ligand–protein complex formation (
Figure 5F,G), enabling cytoplasmic sequestration of
p-NF-κB P65
Ser311 and subsequent blockade of NF-κB transcriptional activity.
By comparing the effects of DKS26 with VDR agonists (VitD3) and inhibitors (MeTC7), we further clarified the mechanism through which DKS26 exerts its renal protective effects by targeting VDR. The results demonstrated that inhibiting VDR activity promotes the synthesis and release of NF-κB P65-mediated inflammatory factors, while both DKS26 and calcifediol reduced the nuclear translocation of
p-NF-κB P65
Ser311 and suppressed the transcription and synthesis of inflammatory factors. The above suggests that DKS26 exerts VDR agonist-like effects and reverses the inflammation and cellular damage caused by VDR inhibitors (
Figure 6). The NF-κB signaling pathway is regulated by the IKKβ/IκBα axis, wherein IKKβ activation phosphorylates IκBα, promoting its ubiquitination and degradation, thereby releasing the NF-κB P65/P50 dimer [
29,
30]. We demonstrated that VDR directly interacts with IKKβ (
Figure 7A) and found that DKS26, along with VDR agonists, reduces IKKβ phosphorylation/activity and decreases IκBα phosphorylation, thereby suppressing NF-κB pathway activation. In contrast, VDR inhibitors exhibited opposing effects (
Figure 7B–M). These findings indicate that DKS26 inhibits the activation of the IKKβ/IκBα axis by enhancing the VDR-IKKβ interaction, thereby blocking the transcriptional activity of the NF-κB signaling pathway.
This study systematically elucidates the mechanism by which the novel small-molecule compound DKS26 stabilizes VDR and directly or indirectly inhibits NF-κB nuclear translocation and downstream proinflammatory gene transcription. By orchestrating the inhibition of oxidative stress, inflammation, and apoptosis, DKS26 offers a multi-target therapeutic strategy for IR-AKI. These findings not only highlight the central role of VDR in renal protection but also provide a solid experimental foundation for developing therapies targeting the VDR-NF-κB pathway, demonstrating significant theoretical and translational potential. Although the IR-AKI model is widely used, it may differ from the pathological processes of human AKI, which often involve more comorbidities. DKS26’s efficacy in animal models requires validation in human-relevant systems (e.g., organoids). Future studies should address the lack of pharmacokinetic and safety data by incorporating multiple species, diverse models, and clinical data to fully assess DKS26’s therapeutic potential.
4. Materials and Methods
4.1. Reagents
VDR (1:500; sc-13133, Santa Cruz, CA, USA), NF-κB P65 (1:500; Sc-8008, Santa Cruz, CA, USA), Phospho-NF-κB P65(Ser311) (1:1000; 310299, ZENBIO, Durham, NC, USA), IKKβ (1:1000; 381592, ZEN-BIO, Chengdu, China), Phospho-IKKβ (Tyr188) (1:1000; 347345, ZEN-BIO, Chengdu, China), NFKBIA/IkBα (1:500; Sc-373893, Santa Cruz, CA, USA), Phospho-NFKBIA/IkBα (Ser 32/36) (1:500; Sc-52943, Santa Cruz, CA, USA), KIM 1 (1:1000; AB233720, Abcam, Cambridge, UK), NGAL (1:1000; #44058, Cell Signaling Technology, Danvers, MA, USA), MCP-1 (1:1000; 220691, ZEN-BIO, Chengdu, China), TNFα (1:1000; 60291-1-AP, Proteintech, Wuhan, China), NF-κB P50 (1:1000; 14220-1-AP, Proteintech, Wuhan, China), PARK2/Parkin (1:1000; 14060-1-AP, Proteintech, Wuhan, China), PINK1 (1:1000; 23274-1-AP, Proteintech, Wuhan, China), LC3A/B (1:1000; #12741, Cell Signaling Technology, Danvers, MA, USA), P62,SQSTM1 (1:1000; 18420-1-AP, Proteintech, Wuhan, China), β-actin (1:10000; 66009-1-Ig, Proteintech, Wuhan, China), HRP-IgG (1:5000; K1223, APEXBIO, Houston, TX, USA), YSFluor™ 594 Goat Anti-Mouse lgG(H+L) (1:200; 33212ES60, YESEN, Shanghai, China).
4.2. Experimental Animals
Twenty-four 8-week-old male C57 mice housed in an SPF environment were randomly assigned to four groups: Sham, IR, IR + PBS, and IR + DKS26 (n = 6 per group). The IR-AKI model was induced by clamping the bilateral renal pedicles for 30 min and maintaining ambient temperature at 37 °C. The DKS26 treatment group received DKS26 (25 mg/kg), whereas the PBS control group received an equal volume of PBS, both administered 30 min prior to the clamping procedure. Mice in the Sham group underwent a similar surgical procedure, with kidney exposure but without renal pedicle clamping. The mice were sacrificed 24 h after surgical suture recovery and reperfusion (
Figure 8). All animals were treated humanely, and all animal procedures met the relevant legal and ethical requirements according to the protocols (NO:2200404) approved by the Institutional Animal Care and Use Committee of Guizhou Medical University.
Figure 8.
Schematic representation of the timeline for establishing the IR-AKI mouse model and DKS26 administration. DKS26 (25 mg/kg) was administered intravenously via the tail vein, and a 30 min interval was allowed for metabolic distribution in the mice. As previously described, an IR-induced AKI model was established by clamping the renal arteries of C57BL/6 mice for 30 min. IR, ischemia-reperfusion; AKI, acute kidney injury.
Figure 8.
Schematic representation of the timeline for establishing the IR-AKI mouse model and DKS26 administration. DKS26 (25 mg/kg) was administered intravenously via the tail vein, and a 30 min interval was allowed for metabolic distribution in the mice. As previously described, an IR-induced AKI model was established by clamping the renal arteries of C57BL/6 mice for 30 min. IR, ischemia-reperfusion; AKI, acute kidney injury.
4.3. Blood Collection, Tissue Preparation, and Biochemical Assays
Following euthanasia, whole blood samples were collected from each experimental group (n = 6 per group) via cardiac puncture. The blood samples were immediately processed by centrifugation at 4 °C and 4000 rpm for 10 min to obtain serum fractions. Scr concentrations were quantitatively determined using a commercially available assay kit (C011-2-1, Nanjing Institute of Biological Engineering, Nanjing, China) in strict accordance with the manufacturer’s protocol. Subsequently, bilateral nephrectomy was performed, and the kidneys were immediately harvested for downstream processing. Renal tissues were precisely sectioned into 1 mm-thick coronal slices using a sterile surgical blade. These tissue sections were allocated for multiple analytical procedures: (1) RNA isolation for gene expression analysis (n = 6 per group), (2) protein extraction for Western blot analysis (n = 6 per group), and (3) paraffin embedding for histological examination (n = 6 per group). The remaining renal tissues were cryopreserved at −80 °C for potential subsequent analyses.
4.4. Cell Culture and Drug Intervention
Mouse renal tubular epithelial cells (mRTECs) were purchased from UACC (Tucson, Arizona, USA) and cultured with DMEM (Thermo Fisher Scientific, Waltham, Massachusetts, USA) containing 10% FBS (Thermo Fisher Scientific, Waltham, Massachusetts, USA) in a common incubator with 5% CO2 and 95% air with a fusion degree of 70–80% for cell passage or planting into 6-well plates (NEST Laboratories, Pennsylvania, USA) for treatment. (1) The concentration gradient of DKS26 was divided as follows: DKS26 solution was added to 6-well plates at 10, 50, and 100 μg/mL, transferred to an anoxic incubator (94% N2, 5% CO2, and 1% O2) for anoxic culture for 6 h, and then placed under normal culture conditions for reoxygenation for 24 h, and samples were collected for experiments. (2) VDR agonist (calcifediol) /inhibitor (MeTC7) group: Calcifediol 300 nM (MCE, Shanghai, China) or MeTC7 200 nM (MCE, Shanghai, China) were added to the cells in a 6-well plate for 30 min to undergo H/R, and samples were collected for analysis.
4.5. Western Blotting
High-efficiency RIPA cracking buffer (PBeyotime, Shanghai, China) was added to the kidney tissues or cells, and a high-throughput tissue grinder was used to grind at a vibration frequency of 50 Hz, and the supernatant was retained by centrifugation. Protein concentration was determined using the BCA method (Elabscience, Houston, TX, USA), and the proteins were separated by SDS-PAGE. After the transfer, the membrane was blocked with 5% skim milk (Yili, Shanghai, China) at room temperature for 1 h. The membrane was washed three times with TBST for 5 min each. The primary antibody was diluted according to the manufacturer’s instructions and incubated with the membranes overnight at 4 °C with gentle shaking. The next day, excess unbound primary antibodies were washed off, and the membranes were incubated with HRP-conjugated secondary antibodies at room temperature. Finally, the images were collected using a chemiluminescence instrument for analysis.
4.6. Histopathology and Quantification Analyses
Kidney tissues of 3–4 mm thickness were preserved in 4% paraformaldehyde (Leagene, Beijing, China), dehydrated, embedded, and paraffin sectioned according to staining requirements. Six renal cortical fields were randomly selected for the scoring. The criteria for assessing tubule damage included brush border loss, intraepithelial vacuolar degeneration, cell membrane rupture, nuclear exposure, and an altered tubular structure. Tubular injury was scored as follows: 0, <10% of the tubules were damaged; 1, 10–25% of the tubules were damaged; 2, 25–50% were damaged; 3, 50–75% were damaged; and 4, >75% were damaged.
4.7. Immunofluorescence Staining
Cell crawling tablets were fixed with 4% paraformaldehyde for 15 min, washed three times with PBS (Solarbio, Beijing, China), and permeabilized with 0.1% Triton X-100 (Solarbio, Beijing, China) for 10 min. Sections were blocked with 10% donkey serum (Solarbio, Beijing, China)at room temperature for 1 h and incubated with the corresponding primary antibody overnight. The next day, after three washes with PBS, the secondary antibody was added and incubated at room temperature for 1 h, using a film sealing agent containing DAPI (Solarbio, Beijing, China), and observed under a laser confocal microscope.
4.8. Immunohistochemistry Staining
Kidney paraffin sections were dewaxed and hydrated with xylene, incubated with 3% hydrogen peroxide at room temperature for 10 min, repaired with 0.1 M sodium citrate microwave oven antigen, sealed with 10% donkey serum, incubated with corresponding antibodies, and refrigerated at 4 °C overnight. The following day, the corresponding secondary antibodies were added, incubated at room temperature for 1 h, and then the ABC enhancer was added and incubated for 30 min. The slides were stained with 3,3′-diaminobenzidine (DAB, zsbio, Beijing, China), counterstained with hematoxylin (Solarbio, Beijing, China), dehydrated, and cover-slipped. Assessment of IHC was scored by applying a semi-quantitative immunoreactivity score.
4.9. RT-qPCR
RNA was isolated and extracted from tissues or cells using the TRIzol (Thermo Fisher Scientific, Waltham, Massachusetts, USA) method, and the RNA concentration was determined using a microvolume spectrophotometre. The RNA was reverse transcribed into cDNA using the Fast King gDNA Dispelling RT SuperMix kit (Tiangen, Beijing, China, KR118). cDNA was used as a template for real-time quantitative PCR (RT-qPCR) to detect mRNA expression using the TB Green Premix Ex Taq kit (TaKaRa, Kusatsu, Japan, RR420A). The RT-qPCR reaction protocol was as follows: denaturation at 95 °C for 30 s, annealing at 95 °C for 5 s, and extension at 60 °C for 30 s, repeated for 39 cycles. β-actin was used as the reference gene, and the 2−ΔΔCt method was applied to calculate the relative mRNA expression levels of the target gene.
Table 1.
Target gene primers.
Table 1.
Target gene primers.
Gene | Forward (5′-3′) | Reverse (3′-5′) |
---|
HAVCR1 (KIM 1) | CTGGAATGGCACTGTGACATCC | GCAGATGCCAACATAGAAGCCC |
LCN2 (NGAL) | ATGTCACCTCCATCCTGGTCAG | GCCACTTGCACATTGTAGCTCTG |
TNFα | GGTGCCTATGTCTCAGCCTCTT | GCCATAGAACTGATGAGAGGGAG |
IL-6 | TACCACTTCACAAGTCGGAGGC | CTGCAAGTGCATCATCGTTGTTC |
CCL2 (MCP1) | GCTACAAGAGGATCACCAGCAG | GTCTGGACCCATTCCTTCTTGG |
4.10. Determination of MDA, T-SOD, CAT and GPx Levels
Mouse kidneys were collected and homogenized in PBS (or normal saline), followed by processing according to the manufacturer’s protocol. Absorbance at 450 nm/405 nm/412 nm was measured using a microplate reader, and MDA levels (Elabscience, Wuhan, China) as well as T-SOD, CAT, and GPx (Nanjing Institute of Biological Engineering, Nanjing, China) activities in tissue homogenates were calculated.
4.11. Flow Cytometry
A single-cell suspension was prepared by trypsin digestion of the cells in 6-well plates, and the cell suspension was diluted with 1 × annexin-V binding buffer to a concentration of 1 × 106 cells/mL. Fluorescein isothiocyanate (FITC)-conjugated membrane-associated protein-V and phycoerythrin (PE)-conjugated propidium iodide (PI) (Becton, Dickinson and Company, Franklin Lakes, New Jersey, USA) were added and incubated with the cells at room temperature for 15 min, and dead cells were excluded based on positive PI staining, and live single cells were screened out based on positive FITC staining. The proportion of apoptotic cells relative to the total live cells was calculated, where apoptotic cells included both early and late apoptotic populations.
4.12. JC-10 Staining
mRTECs were incubated with 5 μM JC-10 (Solarbio, Beijing, China) at 37 °C for 60 min. A single-cell suspension was generated through trypsinization of cells cultured in 6-well plates. Following two washes with 1× phosphate-buffered saline (PBS), the cells were resuspended in 500 μL of 1 × PBS for subsequent flow cytometric analysis. The fluorescence intensities of red and green channels were quantified, and the corresponding ratios were computed to assess the experimental outcomes.
4.13. DCFH-DA Staining
The mRTECs were incubated with 10 μM DCFH-DA (Beyotime, Shanghai, China) at 37 °C for 30 min. The stained cells were washed with PBS. Subsequently, the cells were stained with DAPI for 10 min. Images were acquired using confocal microscopy with excitation light at 488 nm/450 nm.
4.14. MitoSOX Staining
The mRTECs were incubated with 5 μM MitoSOX (Thermo Fisher, Shanghai, China) at 37 °C for 45 min. The stained cells were washed three times with PBS. Images were acquired using confocal microscopy with excitation light at 561nm.
4.15. Nucleoplasm Separation
The collected cell precipitate was resuspended in buffer A (BestBio, Shanghai, China) pre-supplemented with protease and phosphatase inhibitors and placed on ice for 30 min with shaking once every 10 min. The supernatant (cytoplasmic part) was collected by centrifugation, and extracts B and C, containing protease inhibitors and phosphatase inhibitors, were added to the precipitate. The extracts were then placed on ice for 30 min and shaken every 10 min. After centrifugation, the supernatant (nuclear portion) was collected and stored at −20 °C.
4.16. Co-Immunoprecipitation (Co-IP) Assay
The interaction between VDR and NF-κB was detected using a protein A/G magnetic bead immunoprecipitation assay (Elabscience, Wuhan, China). The 5 ng VDR antibody was added to a 40 μg protein sample and incubated overnight in a shaker at 4 °C. The following day, A/G magnetic beads were added to the protein–antibody complex and incubated on a shaker at 37 °C for 2 h. The unbound protein–antibody complex was removed by washing; the formed magnetic bead–protein–antibody complex microbeads were re-suspended in PBS, and SDS sample buffer was added to eluate the bound proteins from the magnetic beads. Western blot analysis was performed using anti-p-NF-κB P65Ser311 antibodies.
4.17. Swiss TargetPrediction
The Swiss TargetPrediction website (
http://swisstargetprediction.ch/ (accessed on 18 May 2023) was used after inputting the DKS26 chemical structure, forecasting, screening, and its interaction with proteins.
4.18. Cellular Thermal Shift Assay (CETSA)
The cells were treated with or without DKS26 for 2 h, lysate was added to crack the cells, and the cell lysates were placed at different temperature gradients (37–64 °C) for 3 min, frozen in liquid nitrogen, and heated to 25 °C after removal. The freeze–thaw cycle was repeated three times, the lysates were centrifuged, and the supernatants were separated and analyzed by Western blotting.
4.19. Darts Experiment
The cells were treated for 2 h with or without DKS26, and the cell lysate was diluted with TNC buffer (50 mM Tris-HCl, 50 mM NaCl, 1 mM CaCl2) (1:10). After incubating at room temperature for 30 min, we added 5 μg/mL Pronase and incubated for another 30 min. The reaction was stopped by mixing with loading buffer and analyzed by Western blotting.
4.20. Molecular Docking
Molecular docking simulations were performed using the structural template VDR/calcitriol complex deposited in the Protein Data Bank under the code 1DB1 [
17]. Typically, the co-crystal ligand VDR was extracted from the crystal complex, and a docking grid was constructed using Autogrid. The AutoDock Vina program was used to dock the processed pdbqt files of DKS26 and the protein, according to previously set parameters.
4.21. Statistical Analysis
Statistical analyses were performed using GraphPad Prism v9.0 and IBM SPSS Statistics v24.0. The normality of the data was evaluated using the Shapiro–Wilk test, and normally distributed data are expressed as mean ± standard deviation (SD). Comparisons between two groups were analyzed by an independent samples t-test, while multiple group comparisons were assessed using one-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison test. Non-parametric tests are applied if data are not normally distributed. Statistical significance was defined as p < 0.05, with non-significant differences labeled as “n.s.”.