Next Article in Journal
High-Fat-Diet-Induced Kidney Injury in Rats: The Role of Tart Cherry Supplementation
Previous Article in Journal
A Nanoformulation of Ubiquinol and Selenium Promotes Proliferation of Human Induced Pluripotent Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Vitamin D Protects Pancreatic Cancer (PC) Cells from Death and DNA Damage Induced by Oxidative Stress

by
Izabela Szymczak-Pajor
1,*,
Egle Morta Antanaviciute
2,
Józef Drzewoski
3,
Ireneusz Majsterek
4 and
Agnieszka Śliwińska
1
1
Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
2
Centre for Cellular Microenvironments, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow G12 8QQ, UK
3
Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
4
Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 5 Mazowiecka Str., 92-215 Lodz, Poland
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(9), 1101; https://doi.org/10.3390/antiox14091101
Submission received: 9 June 2025 / Revised: 28 August 2025 / Accepted: 4 September 2025 / Published: 10 September 2025
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)

Abstract

In addition to its well-recognized roles in immunomodulation and calcium phosphate homeostasis, growing evidence shows that Vitamin D (Vit. D) presents a wide range of other properties, including antioxidant and anticancer effects. However, the action of Vit. D is not fully recognized in pancreatic cancer (PC) cells exposed to oxidative stress. Therefore, the aim of the present study was to investigate whether vitamin D3 (Vit. D3) protects PC cells from death induced by oxidative stress. PC cells are suggested to be resistant to oxidative stress since they demonstrate overexpression of superoxide dismutase (SOD) 1–3. The study measured PC cell viability, DNA damage level, the mRNA and protein expression of antioxidant enzymes, reactive oxygen species (ROS) level and activity of antioxidant enzymes after exposure to H2O2, Vit. D3 and their combinations. N-Acetyl-L-Cysteine (NAC), a well-known direct ROS scavenger, was used as a positive control. Vit. D3 exposure alone had no effect on PC cell viability, ROS level and DNA damage. Its impact on the mRNA and protein expression of antioxidant enzymes was also scarce. However, Vit. D3 protected PC cells against H2O2-induced death, similarly to NAC. It also diminished the increase in ROS and DNA damage caused by H2O2. In addition, Vit. D3 enhanced the mRNA expression of catalase (CAT), SOD 1–3 and glutathione peroxidase (Gpx)3, but did not affect their protein levels in PC cells exposed to oxidative stress. Interestingly, Vit. D3 increased CAT activity after 24 h in 1.2B4 cells and elevated the activity of both CAT and Gpx after 2 h in PANC-1 cells, which could contribute to the observed reduction of H2O2-induced ROS level. To conclude, our findings show that antioxidant properties of Vit. D3 may protect PC cells from oxidative stress-induced death. Therefore, further studies are needed to understand the action of Vit. D3 in PC cells.

1. Introduction

Pancreatic cancer (PC) is characterized by its high aggressiveness, mortality and resistance to drugs, especially those acting via DNA damage-induced apoptosis [1,2,3,4]. Recent epidemiological data indicate that the median survival for PC patients is approximately 4 months, with a 5-year survival rate of only 13%. The poor prognosis for PC is compounded by late-stage diagnosis. Due to the limited effectiveness of current treatments, there is a continued focus on supplementing standard therapy with novel anticancer substances [4,5].
Calcitriol, an active form of Vitamin D (Vit. D), binds to the cytosolic vitamin D receptor (VDR). The VDR then interacts with the retinoid X receptor (RXR), leading to the regulation of gene expression through Vit. D response elements (VDRE) [6,7,8,9,10,11]. This genomic action controls numerous processes, including calcium phosphate metabolism, proliferation, differentiation, angiogenesis, and immunomodulation. In addition, Vit. D exerts its non-genomic action by binding to membrane VDR, and initiating signal transduction via numerous cell signaling pathways [12,13] such as Ca2+/calmodulin-dependent protein kinase (PKCaMII) and mitogen-activated protein kinases (MAPKs). Thus, Vit. D exhibits a broad biological effect, regulating numerous intracellular processes and the functioning of various systems and organs [11,14,15,16,17,18,19].
PC cells demonstrate increased NADPH oxidase activity, which generates additional reactive oxygen species (ROS) [20,21]. An excess of ROS is harmful because these highly reactive molecules can damage cellular macromolecules such as lipids and DNA. Moreover, ROS can trigger the production of several toxic and highly mutagenic metabolites such as 4-hydroxy-2-nonenal (4-HNE) and malonyldialdehyde (MDA) which promote the transformation of cells into a malignant phenotype. If unrepaired, ROS-induced DNA damage leads to genomic instability and tumorigenesis. In addition, ROS promote molecular pathways (i.e., ERK1/2, NF-ĸB, c-SRC, PIK3/AKT, MMP, and RHO-RAC) are involved in tumor aggressiveness and progression by affecting the regulation of apoptosis, In addition, ROS promote molecular pathways (i.e., ERK1/2, NF-ĸB, c-SRC, PIK3/AKT, MMP, and RHO-RAC) involved in tumor aggressiveness and progression by affecting the regulation of apoptosis, and the proliferation and invasion of tumor cells [22]. Elevated ROS production in PC cells contributes to their enhanced metabolic adaptation, proliferation, survival and angiogenic potential as well as providing protection against apoptosis [23,24]. Vaquero et al. proposed that PC is so aggressive and poorly responsive to treatment due to its resistance to apoptosis [25]. To protect against the damaging effects of ROS, cells have evolved a balanced system that utilizes both enzymatic and non-enzymatic antioxidants. These endogenous and exogenous antioxidants prevent and repair damage caused by ROS, thereby decreasing the risk of numerous diseases, including cancer [26]. The most important intracellular antioxidant defenses comprise the enzymes catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (Gpx) [27] which are known as the first-line antioxidant defense [28]. SOD includes three isoforms in mammals: cytoplasmic Cu/ZnSOD (SOD1), mitochondrial matrix MnSOD (SOD2) and extracellular Cu/ZnSOD (SOD3) [29]. The primary function of SOD is the dismutation of the superoxide radical or singlet oxygen—species generated during tissue metabolism—into H2O2 and O2. The accumulation of H2O2 is toxic to cells and tissues because it is rapidly converted to harmful hydroxyl radicals via the Fenton reaction in the presence of transitional metals such as Fe2+. To prevent this, CAT, which is abundant in peroxisomes, breaks H2O2 down into water and molecular oxygen, thereby limiting free radical-induced damage. In the mitochondria, where CAT is not present, Gpx is responsible for reducing H2O2 to water and lipid peroxides to their respective alcohols [30]. Insufficient neutralization of ROS or unrepaired cellular damage results in oxidative stress [26,31]. Oxidative stress is considered to be one of the major risk factors for cancer, including PC [32,33]. Therefore, maintaining redox homeostasis is essential to prevent the initiation of oxidative stress and the development of related diseases.
A growing body of evidence indicates that Vit. D has anticancer potential [34]. Although the effect of Vit. D on the efficacy of PC therapy has been investigated, clinical trials have shown that Vit. D or its analogs do not significantly improve the effectiveness of current anticancer treatments for PC [35,36]. The mechanisms of Vit. D’s action in cancer cells are still poorly understood, particularly in PC where recent studies have shown that cells overexpress the VDR [37]. Through its genomic activity, Vit. D regulates the expression of multiple target genes leading to a wide range of anticancer effects in various malignant cells, including anti-proliferation, anti-inflammation, induction of apoptosis, stimulation of differentiation and the inhibition of invasion, metastasis and angiogenesis [38]. Hummel et al. have suggested that the Vit. D system is deregulated in pancreatic diseases. While the levels of CYP24A1—an enzyme responsible for calcitriol degradation—are decreased in the endocrine islets during PC development, the CYP24A1 protein is accumulated in pancreatic ducts during malignant transformation. Furthermore, tumors with CYP24A1 overexpression are highly proliferative [39]. Research indicates that Vit. D has an antioxidative effect [40,41,42,43]. In turn, PC cells also exhibit overexpression of SOD1, SOD2 and SOD3, as well as lowered superoxide levels and increased H2O2 levels [44]. The antioxidant defense system is a key player in sustaining redox balance and preventing the accumulation of oxidative damage, including DNA damage. Given the broad range of Vit. D’s cellular actions and the resistance of PC cells to damaging factors (i.e., H2O2), investigating the influence of Vit. D on DNA damage and antioxidant defense in PC cells could significantly contribute to understanding of the mechanisms of its high aggressiveness and treatment resistance. The aim of the present study was to determine if Vit. D3 protects PC cells from death induced by oxidative stress. To achieve this goal, PC cells were exposed to H2O2 in the presence and absence of Vit. D3. We then measured cell survival, ROS level, DNA damage level and the activity, as well as gene and protein expression, of antioxidant enzymes.

2. Materials and Methods

2.1. Cell Culture and Treatment

The 1.2B4 cell line is a human hybrid cell line obtained by the fusion of a human pancreatic carcinoma cell line (HuP-T3) with a primary culture of human pancreatic islets. The cells were purchased from the European Collection of Authenticated Cell Cultures (ECACC, Salisbury, UK). PANC-1 (a pancreatic duct epithelioid carcinoma cell line) was purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). Both 1.2B4 and PANC-1 cells were cultured as a monolayer in the standard conditions: 37 °C, 95% air and 5% CO2 and 100% humidity. 1.2B4 cells were grown in RPMI 1640 supplemented with 10% fetal calf serum and 50 IU/mL Penicillin/Streptomycin (Gibco, Life Technologies, Carlsbad, CA, USA). PANC-1 cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with 10% fetal bovine serum and 50 IU/mL Penicillin/Streptomycin (Gibco, Life Technologies, Carlsbad, CA, USA). The cells were used for experiments while in the logarithmic growth phase, after the third to fifth passage. Trypsin-EDTA solution was used to detach the cells. The living cells were counted using trypan blue staining.
Vit. D3, hydrogen peroxide (H2O2) and N-Acetyl-L-Cysteine (NAC) were purchased from Sigma Aldrich (Saint Louis, MO, USA). NAC is a known antioxidant (a free radical scavenger) and was employed as a positive control. A stock solution of Vit. D3 was prepared in 96% ethanol. NAC was diluted in ultrapure water. The cells were exposed to Vit. D3 and H2O2; a combination of Vit. D3 with H2O2 and NAC; and a combination of NAC with H2O2 for 24, 48 and 72 h. As a vehicle, control cells were treated with ethanol. Ethanol 1 (Et-OH1) corresponds to 75 nM Vit. D3, whereas Ethanol 2 (Et-OH2) corresponds to 100 nM Vit. D3.

2.2. Cell Viability Determination—MTT Assay

The impact of the tested compounds on the viability of PANC-1 and 1.2B4 cells was evaluated by the MTT [3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. PANC-1 and 1.2B4 cells were seeded in 96-well plates at a density of 2500 cells per well. After an overnight incubation, the cells achieved a logarithmic growth phase. Next, the tested compounds were added for 24, 48 and 72 h. The following concentrations were employed: Vit. D3 (5–100 nM), H2O2 (50–750 µM), NAC (0.001–30 mM), a combination of Vit. D3 (5–100 nM) with H2O2 as well as combination of NAC (0.001–30 mM) with H2O2. After the completion of exposure, 20 µL of MTT solution (5 g/L) was added to each well for an additional four-hour incubation. Subsequently, the medium was removed and 100 µL dimethyl sulfoxide (DMSO) was added to each well. The absorbance of the dissolved formazan crystals was measured at 570 nm using a SpectrostarNano microplate reader (LMG Biotech, Ortenberg, Germany). The cell viability was indicated as a percentage of control values and calculated from the mean value of three independent experiments. The results for the control group (non-treated cells) were considered as 100%.
Based on the determined viability of the tested PC cells, the following concentrations of the test compounds were used for further experiments: 75 nM and 100 nM Vit. D3 and 3 mM NAC for 1.2B4 and PANC-1 cells; 400 µM H2O2 for 1.2B4 cells; 300 µM H2O2 for PANC-1 cells.

2.3. Evaluation of DNA Damage—The Alkaline Comet Assay

One result of oxidative stress is an increase in the level of DNA damage that was detected by the alkaline version of the comet assay, performed according to Singh et al. [45] with some modifications [46,47]. The alkaline conditions are required to measure the alkaline labile sites and single and double strand breaks. PANC-1 and 1.2B4 cells were seeded in 24-well plates at a density of 105 cells per well.
After 24, 48 or 72 h treatment with the tested compounds, the 1.2B4 and PANC-1 cells were washed two times with cold PBS, suspended in low-melting-point agarose (0.75%) and spread into normal-melting-point agarose (0.5%) pre-coated microscope slides. Subsequently, the cells were lysed by incubation in lysis buffer at 4 °C for 1 h. The lysis buffer was composed of NaCl, 2.5 M; EDTA, 100 mM; TritonX100, 1%; and Tris, 10 mM; pH 10. After lysis, the slides were suspended in unwinding buffer containing NaOH, 30 mM, and EDTA, 1 mM, in pH > 13 at 4 °C for 20 min.
Electrophoresis was conducted at 0.73 V/cm (28 mA) for 20 min. Then, the slides were washed three times with distilled water and drained. Finally, the slides were stained with 2 mg/mL 4′.6-diamidino-2-phenylindole dihydrochloride (DAPI) under dark conditions at a temperature of 4 °C for 30 min. The comets were observed under a fluorescence microscope (Nikon, Tokyo, Japan) at a magnification of ×200 with a video camera and ultraviolet (UV1), a filter block and personal computer equipped with the LuciaComet v. 4.51 analysis software (Laboratory Imaging, Prague, Czech Republic). DNA damage was reflected as the percentage of DNA in the tail of the observed comet and measured for 50 cells in each treatment sample.

2.4. mRNA Expression—qRT-PCR

1.2B4 and PANC-1 cells were seeded on 12-well plates at a density of 106 and incubated with the tested compounds for 24, 48 and 72 h. After completion of treatment with the tested compounds, total RNA was isolated from using a Total RNA Mini Kit (A&A Biotechnology, Gdynia, Poland). Both the quality and quantity of the isolated RNA were assessed with a Nanodrop 2000 reader (Thermofisher Scientific Inc., Waltham, MA, USA). An amount of 1 μg of total isolated RNA was taken for cDNA synthesis. The reaction of reverse transcription was conducted using a High Capacity cDNA Reverse Transcription Kit (Thermofisher Scientific Inc., Waltham, MA, USA), according to the manufacturer’s protocol. cDNA was employed to run qRT-PCR by using the TaqMan assays targeting the studied genes (ID: Hs00156308_m1 for CAT, Hs00533490_m1 for SOD1, Hs00167309_m1 for SOD2, Hs00162090_m1 for SOD3, Hs00173566_m1 for Gpx3, Hs99999905_m1 for GAPDH) (Life Technologies, Carlsbad, CA, USA) and TaqMan Universal Master Mix (Life Technologies, Carlsbad, CA, USA).
Each sample was subjected to qPCR in duplicate. The expression of the studied genes was calculated as the mean results of three independent experiments. The levels of expression of the studied genes were normalized to an endogenous control, GAPDH. Fold change was calculated by the comparative Ct (ΔΔCt) method using the following equation: Relative Quantity (RQ): (2−ΔΔCt) [48].

2.5. Protein Expression Analysis—Western Blotting

The 1.2B4 and PANC-1 cells were incubated with the tested compounds for 24, 48 and 72 h. Following this, total protein was isolated from PANC-1 and 1.2B4 cells with RIPA lysis buffer composed of 50 mM Tris-HCl, 150 mM NaCl, 1% sodium deoxycholate, 0.1% SDS and 2 mM EDTA and supplemented with a protease inhibitor cocktail (Thermofisher Scientific Inc, Waltham, MA, USA) according to the manufacturer’s instruction. Next, the concentration of protein samples was assessed by Micro BCATM Protein Assay Kit (Life Technologies, Carlsbad, CA, USA). In total, 10 µg of protein of each sample were analyzed by immunoblotting and separated by electrophoresis in denaturing polyacrylamide 4–20% Mini Protean TGX Stain-Free Gel (BioRad, Hercules, CA, USA) at 200 V and 31 mA, for 40 min.
After electrophoresis, the separated proteins were transferred from the gel to a polyvinylidene fluoride (PVDF) membrane using the Trans-Blot Turbo Transfer System (BioRad, Hercules, CA, USA). Then, the PVDF membranes were blocked in 5% nonfat milk diluted in a Tris-buffered saline [TBST] buffer with 0.1% Tween 20. After blocking, the membranes were incubated with rabbit primary anti-CAT antibody (FNab01302), anti-SOD1 antibody (FNab08103), anti-SOD2 (FNab08104) antibody, anti-SOD3 antibody (FNab08105), anti-Gpx3 (FNab03621) antibody (Fine-Test, Wuhan, China) and anti-GAPDH (ab9485) antibody (Abcam, Cambridge, UK) overnight at 4 °C. Then, the blots were washed in TBST buffer (3 times for 15 min) and incubated with anti-rabbit secondary antibodies (ab205718) (Abcam, Cambridge, UK) for 4 h at 4 °C. After washing in TBST buffer (3 times for 15 min), the bands were visualized with ECL Blotting Substrate (BioRad, Hercules, CA, USA) using ChemiDoc MP Imaging System (BioRad, Hercules, CA, USA). Densitometric analysis of visualized protein level was carried out by using Image J 1.34s software (Wayne Rasband, National Institutes of Health, Bethesda, MD, USA). GAPDH was employed as a reference protein standard. The protein expression was calculated as the mean of the value of three independent experiments.

2.6. Determination of ROS Level

The level of intracellular ROS production was measured directly in the cell monolayer in black 96-well flat-bottom microtiter plates with a Fluoroskan Ascent FL microplate reader (Thermo Fisher Scientific, Waltham, MA, USA). The cells were seeded at a density of 104 per well and incubated with tested compounds for 2 h and 24 h after reaching the logarithmic growth phase. Then, the medium was removed and cells were incubated with 5 μM DCFH2-DA at 37 °C for 30 min. After the completion of incubation, the fluorescence of DCF was measured at 530 nm after excitation at 485 nm. Inside the cells, DCFH2-DA after deacetylation to DCFH2 is oxidized by ROS to DCF (fluorescent derivative of DCFH2). The fluorescence intensity of DCF was used to determine the level of ROS. The ROS level was calculated as the mean of value of three independent experiments.

2.7. Determination of Antioxidant Enzymes’ Activity

1.2B4 and PANC-1 cells were seeded on 12-well plates at a density of 106 per well. After achieving the logarithmic growth phase by overnight incubation, the cells were exposed to the tested compounds for 2 h and 24 h. After the completion of incubation, the cells were collected using a rubber policeman and centrifuged. Collected cell pellets were homogenized in cold buffers that contain: 50 mM potassium phosphate, pH 7.0, containing 1 mM EDTA for CAT; 20 mM HEPES, pH 7.2, containing 1 mM EGTA, 210 mannitol and 70 mM sucrose for SOD; 50 mM Tris-HCl, pH 7.5, 5 mM EDTA and 1 mM DTT for Gpx. The obtained homogenates were centrifuged. The supernatant samples were used for determination of the enzymatic activity of antioxidant enzymes. We employed the following kits: Catalase Assay Kit, Superoxide Dysmutase Assay Kit and Glutathione Peroxidase Assay Kit (Cayman Chemical, Ann Arbor, MI, USA) to determine the enzymatic activity of CAT, SOD and Gpx.
CAT activity measurement includes the reaction of the enzyme with methanol which takes place in the presence of H2O2. The produced formaldehyde was determined colorimetrically with chromogen (4-amino-3-hydrozino-5-mercapto-1,2,4-triazole). The chromogen reacts with formaldehyde leading to formation of bicyclic heterocycle that changes from colorless to a purple color upon oxidation. The absorbance was measured at 540 nm.
The SOD activity measurement employs a tetrazolium salt for detection of superoxide radicals formed by hypoxantine and xantine oxidase. The measured change in absorbance per minute (25 °C; pH = 8.0), defined as one unit of SOD, indicates the amount of this enzyme needed to perform 50% dismutation of the superoxygen radical. The absorbance was measured at 450 nm.
The principle of the Gpx activity assay involves the conversion of oxidized glutathione (glutathione disulfide, GSSG) to its reduced form by glutathione reductase (GR) and NADPH. The oxidation of NADPH to NADP+ is related to decrease in absorbance. In turn, the decrease in absorbance measured over time is directly proportional to Gpx activity since it is rate limiting. The absorbance was measured ten times (once per minute) at 340 nm.

2.8. Statistical Analysis

Statistical analysis was carried out using GraphPad Prism 6.0 (San Diego, CA, USA). The differences between two groups were evaluated by using Student’s t-test and Mann–Whitney U-test after testing for normality. To determine the differences between three or more groups, ANOVA with Tukey’s multiple comparison test or the non-parametric Kruskal–Wallis test with Dunn’s multiple comparison test were employed, according to data distribution and homogeneity of variance. The data are presented as mean ± standard deviation (SD) from three independent experiments. The p-value < 0.05 was considered to be statistically significant.

3. Results

3.1. Vit. D3 Protects 1.2B4 and PANC-1 Cells from the Cytotoxic Effect of H2O2

The effect of Vit. D3, H2O2, NAC, combination of Vit. D3 with H2O2, and NAC with H2O2 (Figure 1) on the viability of PC cells was determined via MTT assay. In the tested concentration range, Vit. D3 did not affect the viability of 1.2B4 cells. In turn, H2O2 significantly reduced 1.2B4 cell viability in a dose-dependent manner. To examine how Vit. D3 affects H2O2-induced cell death, 1.2 B4 cells were exposed to 400 µM H2O2 and Vit. D3 (5–100 nM). Vit. D3 decreased H2O2-induced cytotoxic effect toward 1.2B4 cells in a dose-dependent manner. The lowest protective potential was demonstrated by 5 nM of Vit. D3. NAC did not affect 1.2B4 cell viability in the concentration range 0.001–3 mM, but it increased 1.2B4 cell viability at concentrations above 3 mM. The combination of NAC with H2O2 protected 1.2B4 cells against the cytotoxic effect of H2O2.
In case of PANC-1 cells, Vit. D3 also did not significantly alter cell viability. As expected, H2O2 exposure resulted in a pronounced increase in the number of dead PANC-1 cells in a dose-dependent manner. The combined exposure to 300 µM H2O2 and Vit. D3 reduced the cytotoxic effect of H2O2 toward PANC-1 cells. This protective effect increased with the concentration of Vit. D3 in all tested incubation times. NAC did not affect PANC-1 cell viability in the tested concentration range, but showed an increase in PANC-1 cell viability at concentrations above 3 mM. The combination of NAC with H2O2 protected PANC-1 cells against the cytotoxic effect of H2O2.
Taken together, high IC50 values for the cytotoxic H2O2 observed in both cell lines suggest the resistance of PC cells to oxidative stress. The 1.2B4 and PANC-1 cells had a similar response to H2O2, although 1.2B4 cells were less sensitive to H2O2 than PANC-1cells. In our study, we used high concentrations of H2O2 due to the resistance of PC cells to oxidative stress associated with resistance to damaging agents, cytostatics and chemotherapy.
Both cell lines responded similarly to NAC alone and Vit. D3 alone, as well as to combined exposure (Vit. D3 and H2O2, and NAC and H2O2). Thus, one can see that Vit. D3 exerted coherent cytoprotective effects similar to the well-known antioxidant NAC.

3.2. Vit. D3 Exerts a Protective Effect Against H2O2-Induced DNA Damage in PC Cells, but Less than NAC

H2O2 is a well-known inducer of oxidative stress that evokes various forms of DNA damage such as oxidative modifications to bases, as well as single- and double-strand breaks and alkaline-labile sites. Therefore, the next stage of the study examined how Vit. D3 influences the DNA damage induced by H2O2. The representative photos are given in Supplementary Figure S3. The 1.2B4 (Figure 2a) and PANC-1 (Figure 2b) cells were exposed to Vit. D3, H2O2, Vit. D3 combined with H2O2 and NAC combined with H2O2. The resulting level of DNA damage was evaluated by alkaline comet assay. As expected, H2O2 evoked a time-dependent increase in the level of DNA damage in 1.2B4 and PANC-1 cells. In contrast, in both cell lines, exposure to Vit. D3 and NAC alone did not elevate DNA damage. Vit. D3 slightly reduced the level of H2O2-induced DNA damage, but a significant reduction was observed only after 72 h in both cell lines. Moreover, the decrease of H2O2-evoked DNA damage by NAC was pronouncedly higher than that detected for Vit. D3 and observed for 24, 48 and 72 h of exposure. To conclude, although Vit. D3 was able to diminish the level of DNA damage caused by oxidative stress in both PC cell lines, its actions seemed to be less intensive than those of NAC. This finding suggests that Vit. D3 and NAC may act through different mechanisms. It is possible that NAC, being a free radical scavenger, removes ROS and thereby prevents DNA from damage evoked by H2O2 earlier [49].

3.3. Vit. D3 Is More Effective than NAC at Increasing the mRNA Expression of Antioxidant Enzymes Lowered by H2O2 in PC Cells

Since Vit. D3 does not directly act on ROS level, the study examined its action on the expression of the CAT, SOD1, SOD2, SOD3 and Gpx3 antioxidant defense system genes. The effects of Vit. D3, H2O2, NAC, Vit. D3 combined with H2O2 and NAC combined with H2O2 on mRNA expression, as determined by RT-qPCR, are shown in Figure 3 (for 1.2B4 cells) and in Figure 4 (for PANC-1 cells).
In the 1.2B4 cells, H2O2 treatment resulted in a time-dependent decrease in CAT mRNA expression. Exposure to Vit. D3 increased CAT expression, but markedly only after 24 and 72 h. Treatment with Vit. D3 + H2O2 increased CAT expression in comparison to H2O2 exposure alone. Higher upregulation was evoked by 100 nM Vit. D3, especially after 24 and 72 h. In turn, exposure of 1.2 B4 cells to the combination of NAC and H2O2 significantly upregulated CAT mRNA expression after 48 and 72 h.
H2O2 had a slight influence on SOD1 expression, causing a significant downregulation only after 48 h. Exposure to Vit. D3 reduced SOD1 expression after 48 h but increased it after 72 h. Treatment with Vit. D3 + H2O2 resulted in higher SOD1 expression compared to H2O2 alone, especially for 100 nM Vit. D3.
SOD2 expression was not altered by H2O2 treatment. Vit. D3 significantly upregulated SOD2 expression after 48 and 72 h in a dose-dependent manner, with a more pronounced effect observed after 48 h. Treatment with 100 nM Vit. D3 and H2O2 significantly increased SOD2 expression compared to H2O2 alone, but only after 72 h.
SOD3 expression did not change after exposure to H2O2. Vit. D3 treatment decreased SOD3 expression after 24 h, but a dose-dependent increase was reported after 48 and 72 h. In turn, 48 h NAC exposure evoked a significant elevation in SOD3 expression. The 100 nM Vit. D3 + H2O2 treatment pronouncedly increased SOD3 expression compared to H2O2 after 48 and 72 h. The NAC + H2O2 treatment markedly increased SOD3 expression in relation to H2O2 after 48 h.
Gpx3 expression was reduced after 24 and 72 h treatment with H2O2. Exposure to 75 nM Vit. D3 significantly reduced Gpx3 expression after 24 h, while both doses of Vit. D3 markedly downregulated it after 48 h. Incubation with Vit. D3 + H2O2 caused a pronounced increase in Gpx3 expression compared to H2O2 alone; this effect was marked after 72 h at the higher dose of Vit. D3 (100 nM).
In PANC-1 cells, H2O2 exposure decreased CAT expression at 24, 48 and 72 h. Vit. D3 treatment did not affect CAT expression. Treatment with Vit. D3 + H2O2 increased CAT expression; however, the elevation was more pronounced for 100 nM Vit. D3 as compared to H2O2. Exposure to NAC and H2O2 increased CAT expression in relation to H2O2 alone after 48 h.
Similarly to CAT expression, H2O2 evoked a decrease in SOD1 expression at all studied time points in the PANC-1 cells. Vit. D3 increased SOD1 expression in a dose-dependent manner after 24 h and 48 h, but decreased it after 72 h. NAC significantly lowered SOD1 expression after 72 h. Exposure to Vit. D3 + H2O2 resulted in a dose-dependent SOD1 mRNA increase in relation to H2O2 alone. NAC and H2O2 treatment increased SOD1 expression after 24 and 48 h compared to H2O2 alone.
H2O2 significantly decreased SOD2 expression after 24, 48 and 72 h. Vit. D3 had varied effects on SOD2 expression depending on its concentration and incubation periods: 75 nM Vit. D3 decreased SOD2 expression at all time points, while 100 nM Vit. D3 lowered expression after 72 h. NAC upregulated SOD2 expression after 24 h but downregulated it after 72 h. Treatment with 100 nM Vit. D3 and H2O2 increased SOD2 expression at all time points compared to H2O2 alone. NAC + H2O2 treatment increased SOD2 expression compared to H2O2 alone but only after 24 h.
H2O2 treatment resulted in a time-dependent decrease in SOD3 expression. Both Vit. D3 concentrations decreased expression of SOD3 after 72 h. NAC treatment lowered SOD3 expression at all time points. Vit. D3 + H2O2 increased SOD3 expression compared to H2O2 exposure after 24, 48 and 72 h. Treatment with NAC + H2O2 did not significantly alter SOD3 expression compared to H2O2 alone.
As before, H2O2 lowered Gpx3 expression at all studied time points. Incubation with 100 nM Vit. D3 increased Gpx3 expression after 24 h. Both Vit. D3 concentrations diminished significantly Gpx3 expression after 48 and 72 h. Treatment with 100 nM Vit. D3 + H2O2 evoked a significant increase in Gpx3 expression compared to H2O2 alone for 48 h. NAC + H2O2 significantly increased Gpx3 expression compared to H2O2, but only after 24 h.
In conclusion, H2O2 decreased the expression of most examined antioxidant enzyme genes in both the 1.2B4 and PANC-1 cells. In the 1.2B4 cells, exposure to Vit. D3 alone slightly increased the expression of CAT and SOD1–3, but did not alter Gpx3 expression. In turn, in PANC-1 cells weak influence of Vit. D3 on the mRNA expression of antioxidant enzymes was not unidirectional, as we observed both down- and upregulation of particular isoforms. In 1.2B4 cells NAC exposure did not affect the mRNA expression of antioxidant enzymes; however, in PANC-1 cells its effect was diverse. Thus, one can notice that there is a difference in the mRNA expression of antioxidant enzymes in response to the Vit. D3 alone or NAC alone treatments between 1.2B4 and PANC-1 cells. As is further underlined by the differences in response to NAC + H2O2 in relation to H2O2. Namely in PANC-1 cells treated with NAC + H2O2, the mRNA expression of the majority of the antioxidant enzymes slightly upregulated, in contrast to 1.2B4 cells. Both PC cell lines exposed to H2O2 and Vit. D3 exhibited the significantly upregulated expression of antioxidant enzymes, indicating that Vit. D3 may have antioxidant potential under oxidative stress conditions.

3.4. Vit. D3 Elevates CAT Protein Expression During H2O2-Induced Oxidative Stress in 1.2B4 Cells

To further explore the antioxidant action of Vit. D3, the next part of the study examined its effect on the protein expression of CAT, SOD1, SOD2, SOD3 and Gpx3. The effects of treatment with Vit. D3, H2O2, NAC and combinations of Vit. D3 with H2O2 and NAC with H2O2 on protein expression are given in Supplementary Figure S1 (for 1.2B4 cells) and in Supplementary Figure S2 (for PANC-1 cells); the data was obtained by Western blotting.
The impact of the tested compounds alone and in combinations on protein levels of antioxidant enzymes was slight. In 1.2B4 cells we observed an increase only in CAT protein expression after exposure to 75 nM Vit. D3 and H2O2 after 72 h. A pronounced increase in SOD3 protein expression was detected after exposure to 75 nM Vit. D3 for 48 h. None of the applied treatments affected the protein expression of SOD1, SOD2 and Gpx3 in 1.2B4 cells.
In the case of PANC-1 cells, only NAC with H2O2 significantly increased CAT protein expression compared to H2O2 after 24 h. Exposure to NAC alone markedly upregulated the protein expression of Gpx3 protein after 24 h. To conclude, Vit. D3 alone, or in the presence of oxidative stress, did not significantly affect the level of CAT, SOD1, SOD2, SOD3 or Gpx3 in either 1.2B4 or PANC-1 cells.

3.5. Vit. D3 Increases Activity of CAT After 24 h in 1.2B4 Cells and Elevates Activity of CAT and Gpx After 2 h in PANC-1 Cells Leading to Reduction of H2O2-Induced Level of ROS

To examine how Vit. D3 protects against H2O2-induced cell death, the study evaluated its antioxidant potential through the determination of ROS levels (Figure 5) and antioxidant enzymes activity (Figure 6 and Figure 7). As expected, treatment with 400 µM H2O2 for 2 and 24 h increased the level of ROS in 1.2B4 cells. In turn, Vit. D3 did not exert any effect on ROS generation, similarly to NAC. In 1.2B4 cells exposed to Vit. D3 + H2O2 the ROS level pronouncedly decreased when compared to cells treated with H2O2 for 24 h. The ROS level also diminished after 24 h of incubation with combined NAC + H2O2.
As expected, the ROS level increased in PANC-1 cells after exposure to H2O2; however, this increase was more visible after 2 h than 24 h. Vit. D3 and NAC did not influence the ROS level. However, when we compare the level of ROS after H2O2 treatment and the combination of Vit. D3 + H2O2, one can see that the Vit. D3 treatment markedly reduced the ROS level after 2 h, but not after 24 h. The level of ROS after exposure to NAC + H2O2 also decreased after 2 h, but surprisingly increased after 24 h.
To sum up, Vit. D3, similarly to NAC, reduced the ROS level elevated by H2O2 in both 1.2B4 and PANC-1 cell lines. However, this effect was present after 24 h for 1.2B4 cells and after 2 h for PANC-1 cells. This subtle difference between 1.2 B4 and PANC-1 cells requires further exploration.
In 1.2B4 cells, H2O2 treatment for 2 h and 24 h resulted in increased CAT activity compared to the control. Vit. D3 and NAC alone did not affect CAT activity after 2 h and 24 h of incubation. After 2 h of incubation with 75 nM Vit. D3 + H2O2 as well as NAC + H2O2 we found decreased CAT activity in comparison to H2O2 exposure, whereas the incubation with 100 nM Vit. D3 + H2O2 evoked CAT activity. In turn, after 24 h, increased CAT activity was observed only for 100 nM Vit. D3 + H2O2, while NAC + H2O2 reduced CAT activity compared to H2O2 exposure.
Exposure of 1.2B4 cells to H2O2 alone, Vit. D3 alone and NAC alone did not significantly affect SOD activity both after 2 h and 24 h exposure (compared to the control). The treatments with Vit. D3 + H2O2 and NAC + H2O2 did not change SOD activity in relation to H2O2 exposure.
Gpx activity was markedly increased after 24 h treatment with H2O2 as compared to the control. The 2 h exposure to 100 nM Vit. D3 alone significantly increased Gpx activity, whereas NAC had the opposite effect compared to the control. Interestingly, 2 h treatemnt with NAC + H2O2 diminished Gpx activity in relation to the control. The Gpx activity was unchanged after 24 h of exposure to Vit. D3 + H2O2 in comparison to H2O2. Conversely, 24 h treatment with NAC + H2O2 markedly decreased Gpx activity in relation to H2O2 exposure.
In PANC-1 cells, 2 h and 24 h of H2O2 exposure increased CAT activity as compared to the control, whereas Vit. D3 alone did not affect CAT activity. The CAT activity after 2 h of treatment with NAC was markedly reduced, whereas after 24 h it was elevated in relation to the control. The treatment with 75 nM Vit. D3 + H2O2 and NAC + H2O2 for 2 h markedly decreased CAT activity as compared to H2O2. The exposure to 100 nM Vit. D3 + H2O2 for 2 h increased CAT activity as compared to H2O2. In turn, the treatment with 75 nM Vit. D3 + H2O2 significantly decreased CAT activity as compared to H2O2 after 24 h.
Similarly to 1.2B4 cells, SOD activity in PANC-1 cells was not affected by H2O2, Vit. D3 alone or NAC alone. The combined treatment with Vit. D3 + H2O2 and NAC + H2O2 did not affect SOD activity in comparison to H2O2 exposure. Only, the exposure to 75 nM Vit. D3 + H2O2 markedly decreased SOD activity as compared to H2O2 after 2 h.
In PANC-1 cells, H2O2 treatment for 2 h and 24 h resulted in a significant increase in Gpx activity as compared to control. Vit. D3 exposure for 2 and 24 h did not affect the Gpx activity as compared to the control. The 24 h treatment with NAC significantly decreased Gpx activity in relation to the control. The exposure to Vit. D3 + H2O2 markedly increased Gpx activity compared to H2O2 after 2 h. However, Vit. D3 + H2O2 incubation for 24 h did not affect the Gpx activity as compared to H2O2. Conversely, NAC + H2O2 treatment for 24 h caused a significant decrease in Gpx activity as compared to H2O2 exposure.
To sum up, H2O2-induced CAT activity was further increased by 100 nM Vit. D3 after 24 h which was accompanied by a reduction in ROS levels in 1.2B4 cells. The total activity of SOD and Gpx in 1.2B4 cells seemed to be unaffected by Vit. D3. In turn, NAC significantly abrogated H2O2-induced CAT and Gpx activity in 1.2B4 cells.
In the case of PANC-1 cells, the increases in CAT and SOD activity evoked by H2O2 were diminished by 75 nM Vit. D3. Interestingly, the H2O2-mediated elevations of CAT and Gpx activity were further increased by 100 nM Vit., while 75 nM Vit. D3 only elevated the activity of Gpx in PANC-1 cells. The action of NAC was the opposite to Vit. D3 since we observed that the H2O2-induced activity of CAT and Gpx was pronouncedly lowered in response to NAC. However both Vit. D3 and NAC were unable to efficiently decrease the level of ROS elevated by H2O2 in PANC-1 cells. NAC directly and immediately scavenges ROS and its action does not lead to the activation of antioxidant systems. In turn, Vit. D3 shows antioxidant action through its influence on antioxidant enzymes.

4. Discussion

PC, which is often diagnosed at an advanced stage, which causes a poor prognosis, is highly aggressive and resistant to treatment, which is why only 13% of patients survive for 5 years after the diagnosis [50,51]. The effect of Vit. D on the intensity of oxidative stress in cancer cells seems to be insufficiently studied. While studies on breast cancer and normal human bone cells have demonstrated that Vit. D3 acts as a prooxidant agent [52,53], other research involving hyperlipidemic patients with type 2 diabetes mellitus and rats with alloxan-induced diabetes have revealed its antioxidant properties [54,55]. Given these conflicting findings, the present study seeks to determine whether the reduction in oxidative stress is partially responsible for the cytoprotective effect of Vit. D3 in PC cells.
The first stage of the study examined the effect of H2O2 on the viability of 1.2B4 and PANC-1 cells in the presence or absence of Vit. D3 or NAC. It was found that H2O2 decreased the viability of both cell lines, which is consistent with previous studies showing that H2O2 reduces both cancer and normal cell viability [56,57,58,59,60,61,62,63,64]. Notably, analysis of IC50 values revealed that PC cells exhibit resistance to this agent. It seems that the resistance of PC cells to oxidative stress may be related to the overexpression of SOD 1–3 [44] and the fact that PC cells are resistant to damaging agents, e.g., cytostatics. Our results showed that Vit. D3 did not appear to influence the viability of 1.2B4 and PANC-1 cells, aligning with previous observations that low doses of Vit. D (<100 nM) do not affect the viability of cancer cells [65,66]. In contrast, some studies indicate that higher concentrations of Vit. D (>100 nM) can reduce the viability of both normal and cancer cells [67,68,69,70,71]. Regarding the effect of NAC on PC viability, our results are consistent with previous studies, showing that NAC does not influence on cell viability within the concentration range of 0.001–3 mM. Some studies also indicate that NAC may increase cell viability [72,73,74]. It is worth noting, however, that some studies have found NAC to decrease the viability of both normal and cancer cells [75,76,77,78,79].
The results of our study revealed that both Vit D and NAC reversed the toxic effect of H2O2 on PC cells. Vit. D3 decreased the percentage of dead 1.2B4 and PANC-1 cells in a time- and dose-dependent manner following H2O2 treatment. Similarly, a previous study found that the viability of human retinal pigment epithelial cells (ARPE-19) markedly increased after exposure to both H2O2 and 50 nM Vit. D3 compared to cells treated with H2O2 alone [71]. Likewise, Tohari et al. observed that low doses of Vit. D3 notably increased the viability of mouse cone 661W cells compared to H2O2 alone, while higher concentrations of Vit. D3 decreased cell viability [64]. However, another study found calcitriol and its analogs enhanced the cytotoxic effect of H2O2 on human keratinocyte cells (HaCaT) [80]. The results of our series of experiments showed that NAC protects PC cells from the cytotoxic effects of H2O2. These observations are consistent with previous studies indicating that NAC reduces the cytotoxic effect of H2O2 on normal cells [81,82,83,84,85,86]. Taken together, our results suggest that in PC cells, Vit. D3 possesses cytoprotective potential against H2O2-induced oxidative stress, in a similar manner to NAC.
The further step of our study evaluated the effect of Vit. D3 on DNA damage induced by H2O2 in PC cells. While H2O2 increased DNA damage, Vit. D3 was found to reduce it, though the effect was only significant after 72 h. The beneficial effect of NAC was observed already after 24 h and lasted for 72 h. These findings suggest that Vit. D3 employs mechanisms distinct from those of NAC. Vit. D3 may protect against the ROS generation caused by H2O2 and further reduce oxidative stress-induced DNA damage by enhancing the antioxidant defense system [87,88]. It has been noted that Vit. D increases the expression of NRF-2 and Klotho, which contributes to the upregulation of antioxidant enzyme gene expression [87,89,90]. Klotho elevates the expression of antioxidant enzymes such as CAT, peroxiredoxin (PRX)-2, PRX-3, SOD2 and thioredoxin reductase 1 (Trxrd-1). In turn, NRF2 upregulates CAT, Gpx, glutathione reductase (GR), SOD ½, thioredoxin (TRX) and thioredoxin reductase (TR) [87]. Klotho has also been observed to activate NRF2, leading to a decrease in oxidative damage [91]. Indeed, DNA damage caused by oxidative stress has been shown to increase in the colonic epithelial cells of mice lacking a VDR [92]. It was also found that the level of oxidative DNA damage was pronouncedly decreased in normal colorectal mucosa of patients supplemented with 800 IU of Vit. D per day [93]. Wenclewska et al. showed that supplementation with 2000 IU Vit. D per day for three months markedly reduced the level of DNA damage in the lymphocytes of subjects with dysglycemia and/or dyslipidemia; furthermore, the level of oxidative DNA damage was also diminished in the subgroup with T2DM [94]. Graziano et al. have proposed that the Vit. D–Vit. D receptor axis regulates DNA repair during oncogene-induced senescence [95]. It should be further investigated whether the reduction in H2O2-induced DNA damage by Vit. D3 may result from its effect on the expression of DNA repair genes, particularly those involved in homologous recombination (HR) and non-homologous end-joining (NHEJ), which remove oxidative stress-induced DNA damage. To conclude, Vit. D presents different mechanism of protection against the formation and accumulation of DNA damage than NAC. Since Vit. D enhances antioxidant defense and DNA repair—which are multi-stage processes that require more time [96]—the reduction of H2O2-induced DNA damage by Vit. D is only visible after 72 h.
In the next stage of the study, we assessed the effect of Vit. D3 on mRNA expression of CAT, SOD 1–3 and Gpx 3 in PC cells exposed to H2O2. Little is known about the effect of Vit. D on the expression of genes encoding antioxidant enzymes in PC cells. Our results indicate that H2O2 decreased the expression of antioxidant enzymes, while Vit. D3 enhanced their expression alone or in combination with H2O2. Lisse et al. reported a decrease in CAT and SOD1 mRNA expression and an increase in SOD2 mRNA expression after 24 h of incubation with 10 nM Vit. D in human MG-63 osteosarcoma cells. They also noted an eightfold increase in SOD2 mRNA expression after 48 h of incubation with Vit. D3 [68]. Interestingly, it has also been shown that 1,25(OH)2D3 or its analogs induce the expression of thioredoxin reductase 1 (TXNRD1). TXNRD1 is an enzyme that converts thioredoxin into its reduced form, which has antioxidant activity [97,98]. Treatment with 50 nM 1,25(OH)2D3 increased the production of SOD1 and SOD2 in prostate epithelial cells (PEC) and androgen-sensitive prostate cancer cells (LNCaP) [97,99]. Therefore, the effect of Vit D3 on antioxidant mRNA expression seems to be dependent on its concentration. The cytoprotective action of Vit. D3 is associated with the expression of antioxidant defense genes. Furthermore, our data shows that the protective effect of NAC against oxidative stress is only weakly connected with the mRNA expression of antioxidant genes, highlighting a difference in the mechanism of action between Vit D3 and NAC. Berridge et al., found that the genomic action of Vit. D3 has been linked to the induction of NFR2 mRNA expression. NFR2 is a key transcription factor that acts as a master regulator of antioxidant enzyme expression; it is possible that Vit. D may support the antioxidant defense system by targeting NRF2 [87].
It should be mentioned that the mRNA expression of antioxidant enzymes may also be controlled by numerous microRNAs, such as hsa-miR-181b-5p, hsa-miR-155-5p, hsa-miR-342-3p, hsa-miR-106b-5p, hsa-miR-92b-3p, hsa-miR-505-5p, hsa-miR-30a-5p, hsa-miR-181c-3p and hsa-miR-634 [100]. Zhao et al. concluded not only that microRNAs influence Vit. D signaling, but also that Vit. D regulates microRNA networks [101]. To the best of our knowledge, there is currently no data reporting how Vit. D3 regulates the expression of microRNAs targeting particular antioxidant enzymes. Upregulation of miR-200a induces nuclear translocation of the NRF2 signaling pathway, contributing to a decrease in ROS production and apoptosis of primary human osteoblasts [102]. Moreover, miR200a inhibits KEAP1, which leads to the stimulation of NRF2 [103]. Under physiological conditions, KEAP1 binds to the NRF2-ECH2 homologous domain (Neh2), which suppresses NRF2 translocation to the nucleus [104]. However, under oxidative stress, KEAP1 does not interact with NRF2, triggering its separation from NRF2. Then, NRF2 translocates to the nucleus, where it binds to the antioxidant response element and stimulates the expression of antioxidant genes [105,106]. However, the influence of Vit. D on miR200a expression is not yet known.
Since we found that Vit. D3 affects the expression of antioxidant enzyme genes, we decided to perform an analysis of the protein levels of the tested antioxidant enzymes. We observed that the levels of these antioxidant enzymes were merely decreased after exposure to H2O2. Treatment with H2O2 in the presence of Vit. D3 or NAC did not exert a significant impact on the protein expression of the studied antioxidant proteins in PC cells. We only observed an increase in CAT protein expression in response to the 75 nM D3 + H2O2 treatment after 72 h in 1.2B4 cells. Data from the literature on the influence of Vit. D3 on the protein expression of antioxidant enzymes in PC cells is scarce. However, Middleton et al. report that calcitriol (10−7 and 10−9 M) and its analog seocalcitol (10−9 M) increased CAT protein expression in canine bladder transitional cell carcinoma (cbTCC), with no effect on SOD2 or SOD1 protein expression [107].
The next part of the study tested the effect of Vit. D3 on the ROS level after exposure to H2O2. As expected, H2O2 increased the ROS level, while combined incubation with H2O2 and Vit. D3 or NAC lowered it. These findings suggest that the cytoprotective effect of Vit. D3 might be a result of its antioxidant properties. We also observed that the antioxidant action of Vit. D3 depended on its concentration and exposure time. Consistent with our results, Tohari et al. observed that the combination of Vit. D3 with H2O2 reduced the ROS level by 31% after 6 h and 41% after 24 h [64]. It was also reported that administration of calcitriol to rats was associated with a significant reduction in the levels of malondialdehyde (MDA), a well-known oxidative stress marker, and the end product of lipid peroxidation [108].
Finally, we assessed how Vit. D3 affects the activity of antioxidant enzymes in the presence or absence of oxidative stress. It can be observed that Vit. D3 elevated the activity of CAT after 24 h, which was accompanied by the reduction in the H2O2-induced level of ROS in 1.2B4 cells. We also reported an increase in CAT protein expression in response to 72 h of treatment with 75 nM D3 + H2O2 in 1.2B4 cells. In turn, in PANC-1 cells, Vit. D3 elevated the activity of CAT and Gpx, which contributed to a decrease in the ROS level after 2 h. These results are consistent with findings from experiments investigating DNA damage and the mRNA expression of antioxidant enzymes. We did not show any significant effect of Vit. D3 on SOD activity in the presence of oxidative stress. Only a few studies have investigated the effect of Vit. D3 on the activity of antioxidant enzymes. In contrast to our results, AlJorhi et al. observed reduced activity of CAT after the exposure of primary cortical neuronal cells to H2O2 (0.5 mM) with a high dose of Vit. D3 (250 ng/mL), compared to H2O2 alone. Thus, Vit. D3 protected neurons against H2O2-induced oxidative stress, which was accompanied by a decrease in CAT activity [109]. Bhat et al. have demonstrated that Vit. D deficiency contributes to mild oxidative stress, which is accompanied by an increase in the activities of glutathione-dependent enzymes and a decrease in SOD and CAT activities in rat muscles. In turn, supplementation with Vit. D reversed Vit. D deficiency-related changes in all of the antioxidant enzymes tested [110]. Interestingly, Saif-Elnasr et al. did not observe any statistically significant correlation between serum Vit. D levels and the activity of SOD and GPx in T2DM patients and healthy controls. Therefore, they concluded that Vit. D supplementation does not correlate with the activity of antioxidant enzymes [111].
To conclude, H2O2-induced CAT activity was further increased by 100 nM Vit. D3 after 24 h, which was accompanied by a decrease in ROS levels in 1.2B4 cells. Of note, the result of Western blotting analysis also showed an elevation in CAT protein expression in response to the 75D3 + H2O2 treatment after 72 h in 1.2B4 cells. Vit. D3 did not affect the total activity of SOD and Gpx in 1.2 B4 cells. NAC markedly reduced the total activity of CAT and Gpx induced by H2O2 in 1.2B4 cells. The effect of NAC on antioxidant enzyme activity was opposite to that of Vit. D3 since we reported that a H2O2-induced elevation in the activity of CAT and Gpx was significantly abrogated. However, neither Vit. D3 nor NAC efficiently decreased the level of ROS elevated by H2O2 in PANC-1 cells. Interestingly, the results of some studies indicate that higher calcium levels stimulate the activity of antioxidants enzymes, such as CAT [112,113]. In turn, Vit. D treatment elevates intracellular calcium levels and increases the expression of calcium channel genes [114,115]. Alatavi et al. showed that the oral administration of Vit. D with calcium to diabetic rats led to a significant increase in the activities of SOD, GPO and CAT compared with the untreated group [116]. Thus, Vit. D may stimulate the activity of antioxidant enzymes by increasing intracellular calcium levels.
Recently, the influence of Vit. D on the tumor environment has attracted the attention of researchers. It has been observed that crosstalk between the components of the tumor microenvironment and cancer cells may play an important role in the process of both the development and progression of breast cancer. The components of this tumor microenvironment include immune cells, endothelial cells, stromal cells, factors secreted by these cells and the extracellular matrix. Cancer-associated fibroblasts are the most abundant cell type in both the breast cancer and PC microenvironments [117,118]. Łabędź et al. investigated the influence of Vit. D on breast cancer-associated fibroblasts and suggested that calcitrol may alter their immunosuppressive or procancer properties. The observed anticancer polarization of these cancer-associated fibroblasts in response to treatment may be related to a reduction in CCL2, TNC, MMP9 and MMP-2. Conversely, the opposite action may be associated with elevated PDPN, TIMP1 and SPP1. However, cancer-associated fibroblast-conditioned media from nonmetastatic and postmenopausal patients incubated ex vivo with calcitriol reduced the migration of MCF-7 cells [119]. The influence of Vit. D, which has anti-inflammatory and immunomodulatory properties, on the microenvironment of PC is not yet known and is an excellent direction for further research on this tumor. Therefore, the effect of Vit. D on fibroblasts, immune cells and cancer cells in the tumor microenvironment may be different. Our study concerned only cancer cells. The obtained results showed that Vit. D protects PC cells from the harmful effects of H2O2. However, at this stage, it remains unclear how cancer cells communicate with normal cells within the tumor. We also do not know what effect Vit. D would have in the tumor on normal cells such as fibroblasts or immune cells.
The results of clinical trials are still inconclusive; it is not clear whether patients with PC can benefit from Vit. D supplementation or not. Interestingly, Wang et al. reported that VDR is highly expressed in PC cells [37]. Moreover, it was also suggested that the expression level of VDR may be a potential prognostic factor for PC patients [37]. A recent study carried out by Li et al. showed that PC patients with VDR overexpression in pancreatic tumor cells had significantly decreased overall survival. This overexpression of VDR in PC cells promotes the recruitment and polarization of macrophages into M2 macrophage phenotype via the secretion of CCL20, which leads to the activation of tumor progression [120]. Further clinical trials based on combined therapies using Vit. D are necessary in PC patients, both with and without overexpressed VDR in PC cells, to observe their impact on survival/extension of life and the delay of further progression of PC.
It should also be noted that our study has several limitations. Firstly, it would be beneficial to determine the level of oxidative DNA damage using a comet assay with formamidopyrimidine DNA glycosylase (Fpg) and endonuclease III (Nth). Secondly, the PANC-1 and 1.2B4 cell lines have different origins, which may be responsible for some of the variation observed in the results. PANC-1 is a pancreatic adenocarcinoma [121], whereas 1.2B4 is a human hybrid cell line of HuP3 (adenocarcinoma) cells and the primary culture of human pancreatic islets. Thirdly, while the action of Vit. D seems to be well-characterized in numerous normal cell lines, its effects on normal pancreatic duct cells, the origin of most pancreatic adenocarcinomas, remain poorly understood. Fourth, it is also important to note that the MTT assay used for assessing cell viability is non-specific. Therefore, the increase in formazan production observed at NAC concentrations above 3 mM may result from factors other than increased cell viability, such as enhanced cell growth or mitochondrial activity.
To summarize, our series of experiments showed that PC cells are resistant to DNA damaging effects caused by a high concentration of H2O2. Moreover, the results obtained indicate that Vit. D3 protects PC cells against death induced by H2O2. This effect was accompanied by a reduction in ROS level and DNA damage, as well as the mRNA upregulation of CAT, SOD 1-3 and Gpx3. Interestingly, 100 nM Vit. D3 increased, while 75 nM Vit. D3 reduced, the activity of CAT in 1.2B4 and PANC-1 cells, as well as SOD in PANC-1 cells exposed to H2O2-induced oxidative stress. Our findings suggest that Vit. D3 may exert antioxidant activity but is not a free radical scavenger like NAC. However, other possible molecular mechanisms should also be explored to identify how Vit. D3 supports PC cell viability and protects against DNA damage induced by oxidative stress.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antiox14091101/s1.

Author Contributions

Conceptualization, A.Ś. and I.S.-P.; methodology, I.S.-P. and E.M.A.; software, I.S.-P. and I.M.; validation, A.Ś. and I.S.-P.; formal analysis, A.Ś.; investigation, I.S.-P. and E.M.A.; writing—original draft preparation, I.S.-P.; writing—review and editing, A.Ś., J.D. and I.M.; visualization, I.S.-P.; supervision A.Ś.; funding acquisition, A.Ś. All authors have read and agreed to the published version of the manuscript.

Funding

This study and paper were supported by the grant from the Medical University of Lodz (No. 503/1-159-01/503-21-001) and the Polish Society of Metabolic Disorders.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zeng, S.; Pöttler, M.; Lan, B.; Grützmann, R.; Pilarsky, C.; Yang, H. Chemoresistance in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 4504. [Google Scholar] [CrossRef] [PubMed]
  2. Espona-Fiedler, M.; Patthey, C.; Lindblad, S.; Sarró, I.; Öhlund, D. Overcoming Therapy Resistance in Pancreatic Cancer: New Insights and Future Directions. Biochem. Pharmacol. 2024, 229, 116492. [Google Scholar] [CrossRef]
  3. Jiang, H.; Zuo, J.; Li, B.; Chen, R.; Luo, K.; Xiang, X.; Lu, S.; Huang, C.; Liu, L.; Tang, J.; et al. Drug-Induced Oxidative Stress in Cancer Treatments: Angel or Devil? Redox Biol. 2023, 63, 102754. [Google Scholar] [CrossRef] [PubMed]
  4. Stoop, T.F.; Javed, A.A.; Oba, A.; Koerkamp, B.G.; Seufferlein, T.; Wilmink, J.W.; Besselink, M.G. Pancreatic Cancer. Lancet Lond. Engl. 2025, 405, 1182–1202. [Google Scholar] [CrossRef]
  5. Lencioni, G.; Gregori, A.; Toledo, B.; Rebelo, R.; Immordino, B.; Amrutkar, M.; Xavier, C.P.R.; Kocijančič, A.; Pandey, D.P.; Perán, M.; et al. Unravelling the Complexities of Resistance Mechanism in Pancreatic Cancer: Insights from in Vitro and Ex-Vivo Model Systems. Semin. Cancer Biol. 2024, 106–107, 217–233. [Google Scholar] [CrossRef]
  6. Haussler, M.R.; Whitfield, G.K.; Kaneko, I.; Haussler, C.A.; Hsieh, D.; Hsieh, J.-C.; Jurutka, P.W. Molecular Mechanisms of Vitamin D Action. Calcif. Tissue Int. 2013, 92, 77–98. [Google Scholar] [CrossRef]
  7. Christakos, S.; Dhawan, P.; Verstuyf, A.; Verlinden, L.; Carmeliet, G. Vitamin D: Metabolism, Molecular Mechanism of Action, and Pleiotropic Effects. Physiol. Rev. 2016, 96, 365–408. [Google Scholar] [CrossRef] [PubMed]
  8. Pike, J.W.; Meyer, M.B. Fundamentals of Vitamin D Hormone-Regulated Gene Expression. J. Steroid Biochem. Mol. Biol. 2014, 144, 5–11. [Google Scholar] [CrossRef]
  9. Voltan, G.; Cannito, M.; Ferrarese, M.; Ceccato, F.; Camozzi, V. Vitamin D: An Overview of Gene Regulation, Ranging from Metabolism to Genomic Effects. Genes 2023, 14, 1691. [Google Scholar] [CrossRef]
  10. Carlberg, C. Vitamin D and Its Target Genes. Nutrients 2022, 14, 1354. [Google Scholar] [CrossRef]
  11. Warwick, T.; Schulz, M.H.; Günther, S.; Gilsbach, R.; Neme, A.; Carlberg, C.; Brandes, R.P.; Seuter, S. A Hierarchical Regulatory Network Analysis of the Vitamin D Induced Transcriptome Reveals Novel Regulators and Complete VDR Dependency in Monocytes. Sci. Rep. 2021, 11, 6518. [Google Scholar] [CrossRef]
  12. Hii, C.S.; Ferrante, A. The Non-Genomic Actions of Vitamin D. Nutrients 2016, 8, 135. [Google Scholar] [CrossRef] [PubMed]
  13. Żmijewski, M.A. Nongenomic Activities of Vitamin D. Nutrients 2022, 14, 5104. [Google Scholar] [CrossRef] [PubMed]
  14. Zmijewski, M.A. Vitamin D and Human Health. Int. J. Mol. Sci. 2019, 20, 145. [Google Scholar] [CrossRef]
  15. Bikle, D.D. Extraskeletal Actions of Vitamin D. Ann. N. Y. Acad. Sci. 2016, 1376, 29–52. [Google Scholar] [CrossRef]
  16. Silva, I.C.J.; Lazaretti-Castro, M. Vitamin D Metabolism and Extraskeletal Outcomes: An Update. Arch. Endocrinol. Metab. 2022, 66, 748–755. [Google Scholar] [CrossRef]
  17. Liu, W.; Zhang, L.; Xu, H.-J.; Li, Y.; Hu, C.-M.; Yang, J.-Y.; Sun, M.-Y. The Anti-Inflammatory Effects of Vitamin D in Tumorigenesis. Int. J. Mol. Sci. 2018, 19, 2736. [Google Scholar] [CrossRef]
  18. Krajewska, M.; Witkowska-Sędek, E.; Rumińska, M.; Stelmaszczyk-Emmel, A.; Sobol, M.; Majcher, A.; Pyrżak, B. Vitamin D Effects on Selected Anti-Inflammatory and Pro-Inflammatory Markers of Obesity-Related Chronic Inflammation. Front. Endocrinol. 2022, 13, 920340. [Google Scholar] [CrossRef] [PubMed]
  19. Sassi, F.; Tamone, C.; D’Amelio, P. Vitamin D: Nutrient, Hormone, and Immunomodulator. Nutrients 2018, 10, 1656. [Google Scholar] [CrossRef]
  20. Weydert, C.; Roling, B.; Liu, J.; Hinkhouse, M.M.; Ritchie, J.M.; Oberley, L.W.; Cullen, J.J. Suppression of the Malignant Phenotype in Human Pancreatic Cancer Cells by the Overexpression of Manganese Superoxide Dismutase. Mol. Cancer Ther. 2003, 2, 361–369. [Google Scholar]
  21. Cullen, J.J.; Weydert, C.; Hinkhouse, M.M.; Ritchie, J.; Domann, F.E.; Spitz, D.; Oberley, L.W. The Role of Manganese Superoxide Dismutase in the Growth of Pancreatic Adenocarcinoma. Cancer Res. 2003, 63, 1297–1303. [Google Scholar]
  22. Martinez-Useros, J.; Li, W.; Cabeza-Morales, M.; Garcia-Foncillas, J. Oxidative Stress: A New Target for Pancreatic Cancer Prognosis and Treatment. J. Clin. Med. 2017, 6, 29. [Google Scholar] [CrossRef]
  23. Zhang, L.; Li, J.; Zong, L.; Chen, X.; Chen, K.; Jiang, Z.; Nan, L.; Li, X.; Li, W.; Shan, T.; et al. Reactive Oxygen Species and Targeted Therapy for Pancreatic Cancer. Oxid. Med. Cell. Longev. 2016, 2016, 1616781. [Google Scholar] [CrossRef]
  24. Afanas’ev, I. Reactive Oxygen Species Signaling in Cancer: Comparison with Aging. Aging Dis. 2011, 2, 219–230. [Google Scholar]
  25. Vaquero, E.C.; Edderkaoui, M.; Pandol, S.J.; Gukovsky, I.; Gukovskaya, A.S. Reactive Oxygen Species Produced by NAD(P)H Oxidase Inhibit Apoptosis in Pancreatic Cancer Cells. J. Biol. Chem. 2004, 279, 34643–34654. [Google Scholar] [CrossRef]
  26. Jena, A.B.; Samal, R.R.; Bhol, N.K.; Duttaroy, A.K. Cellular Red-Ox System in Health and Disease: The Latest Update. Biomed. Pharmacother. 2023, 162, 114606. [Google Scholar] [CrossRef] [PubMed]
  27. Lei, X.G.; Vatamaniuk, M.Z. Two Tales of Antioxidant Enzymes on β Cells and Diabetes. Antioxid. Redox Signal. 2011, 14, 489–503. [Google Scholar] [CrossRef] [PubMed]
  28. Ighodaro, O.M.; Akinloye, O.A. First Line Defence Antioxidants-Superoxide Dismutase (SOD), Catalase (CAT) and Glutathione Peroxidase (GPX): Their Fundamental Role in the Entire Antioxidant Defence Grid. Alex. J. Med. 2018, 54, 287–293. [Google Scholar] [CrossRef]
  29. Fukai, T.; Ushio-Fukai, M. Superoxide Dismutases: Role in Redox Signaling, Vascular Function, and Diseases. Antioxid. Redox Signal. 2011, 15, 1583–1606. [Google Scholar] [CrossRef]
  30. Pei, J.; Pan, X.; Wei, G.; Hua, Y. Research Progress of Glutathione Peroxidase Family (GPX) in Redoxidation. Front. Pharmacol. 2023, 14, 1147414. [Google Scholar] [CrossRef]
  31. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  32. Li, L.; Leung, P.S. Gracia-Sancho, J., Salvadó, J., Eds.; Chapter 13-Pancreatic Cancer, Pancreatitis, and Oxidative Stress. In Gastrointestinal Tissue; Academic Press: Cambridge, MA, USA, 2017; pp. 173–186. ISBN 978-0-12-805377-5. [Google Scholar]
  33. Weeden, C.E.; Hill, W.; Lim, E.L.; Grönroos, E.; Swanton, C. Impact of Risk Factors on Early Cancer Evolution. Cell 2023, 186, 1541–1563. [Google Scholar] [CrossRef] [PubMed]
  34. Seraphin, G.; Rieger, S.; Hewison, M.; Capobianco, E.; Lisse, T.S. The Impact of Vitamin D on Cancer: A Mini Review. J. Steroid Biochem. Mol. Biol. 2023, 231, 106308. [Google Scholar] [CrossRef]
  35. Evans, T.R.J.; Colston, K.W.; Lofts, F.J.; Cunningham, D.; Anthoney, D.A.; Gogas, H.; de Bono, J.S.; Hamberg, K.J.; Skov, T.; Mansi, J.L. A Phase II Trial of the Vitamin D Analogue Seocalcitol (EB1089) in Patients with Inoperable Pancreatic Cancer. Br. J. Cancer 2002, 86, 680–685. [Google Scholar] [CrossRef]
  36. Blanke, C.D.; Beer, T.M.; Todd, K.; Mori, M.; Stone, M.; Lopez, C. Phase II Study of Calcitriol-Enhanced Docetaxel in Patients with Previously Untreated Metastatic or Locally Advanced Pancreatic Cancer. Investig. New Drugs 2009, 27, 374–378. [Google Scholar] [CrossRef]
  37. Wang, K.; Dong, M.; Sheng, W.; Liu, Q.; Yu, D.; Dong, Q.; Li, Q.; Wang, J. Expression of Vitamin D Receptor as a Potential Prognostic Factor and Therapeutic Target in Pancreatic Cancer. Histopathology 2015, 67, 386–397. [Google Scholar] [CrossRef] [PubMed]
  38. Zhang, X.; Harbeck, N.; Jeschke, U.; Doisneau-Sixou, S. Influence of Vitamin D Signaling on Hormone Receptor Status and HER2 Expression in Breast Cancer. J. Cancer Res. Clin. Oncol. 2017, 143, 1107–1122. [Google Scholar] [CrossRef]
  39. Hummel, D.; Aggarwal, A.; Borka, K.; Bajna, E.; Kállay, E.; Horváth, H.C. The Vitamin D System Is Deregulated in Pancreatic Diseases. J. Steroid Biochem. Mol. Biol. 2014, 144, 402–409. [Google Scholar] [CrossRef]
  40. Almeida Moreira Leal, L.K.; Lima, L.A.; Alexandre de Aquino, P.E.; Costa de Sousa, J.A.; Jataí Gadelha, C.V.; Felício Calou, I.B.; Pereira Lopes, M.J.; Viana Lima, F.A.; Tavares Neves, K.R.; Matos de Andrade, G.; et al. Vitamin D (VD3) Antioxidative and Anti-Inflammatory Activities: Peripheral and Central Effects. Eur. J. Pharmacol. 2020, 879, 173099. [Google Scholar] [CrossRef]
  41. Sepidarkish, M.; Farsi, F.; Akbari-Fakhrabadi, M.; Namazi, N.; Almasi-Hashiani, A.; Maleki Hagiagha, A.; Heshmati, J. The Effect of Vitamin D Supplementation on Oxidative Stress Parameters: A Systematic Review and Meta-Analysis of Clinical Trials. Pharmacol. Res. 2019, 139, 141–152. [Google Scholar] [CrossRef] [PubMed]
  42. Della Nera, G.; Sabatino, L.; Gaggini, M.; Gorini, F.; Vassalle, C. Vitamin D Determinants, Status, and Antioxidant/Anti-Inflammatory-Related Effects in Cardiovascular Risk and Disease: Not the Last Word in the Controversy. Antioxidants 2023, 12, 948. [Google Scholar] [CrossRef]
  43. Russo, C.; Valle, M.S.; Malaguarnera, L. Antioxidative Effects of Vitamin D in Muscle Dysfunction. Redox Exp. Med. 2023, 2023, e230013. [Google Scholar] [CrossRef]
  44. Teoh, M.L.T.; Sun, W.; Smith, B.J.; Oberley, L.W.; Cullen, J.J. Modulation of Reactive Oxygen Species in Pancreatic Cancer. Clin. Cancer Res. 2007, 13, 7441–7450. [Google Scholar] [CrossRef]
  45. Singh, N.P.; McCoy, M.T.; Tice, R.R.; Schneider, E.L. A Simple Technique for Quantitation of Low Levels of DNA Damage in Individual Cells. Exp. Cell Res. 1988, 175, 184–191. [Google Scholar] [CrossRef] [PubMed]
  46. Klaude, M.; Eriksson, S.; Nygren, J.; Ahnström, G. The Comet Assay: Mechanisms and Technical Considerations. Mutat. Res. 1996, 363, 89–96. [Google Scholar] [CrossRef]
  47. Blasiak, J.; Gloc, E.; Drzewoski, J.; Wozniak, K.; Zadrozny, M.; Skórski, T.; Pertynski, T. Free Radical Scavengers Can Differentially Modulate the Genotoxicity of Amsacrine in Normal and Cancer Cells. Mutat. Res. 2003, 535, 25–34. [Google Scholar] [CrossRef] [PubMed]
  48. Livak, K.J.; Schmittgen, T.D. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef] [PubMed]
  49. Pedre, B.; Barayeu, U.; Ezeriņa, D.; Dick, T.P. The Mechanism of Action of N-Acetylcysteine (NAC): The Emerging Role of H2S and Sulfane Sulfur Species. Pharmacol. Ther. 2021, 228, 107916. [Google Scholar] [CrossRef]
  50. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef]
  51. Yu, B.; Shao, S.; Ma, W. Frontiers in Pancreatic Cancer on Biomarkers, Microenvironment, and Immunotherapy. Cancer Lett. 2025, 610, 217350. [Google Scholar] [CrossRef]
  52. Koren, R.; Hadari-Naor, I.; Zuck, E.; Rotem, C.; Liberman, U.A.; Ravid, A. Vitamin D Is a Prooxidant in Breast Cancer Cells. Cancer Res. 2001, 61, 1439–1444. [Google Scholar]
  53. Somjen, D.; Katzburg, S.; Knoll, E.; Sharon, O.; Posner, G.H.; Stern, N. Vitamin D Analogs Induce Lipoxygenase mRNA Expression and Activity as Well as Reactive Oxygen Species (ROS) Production in Human Bone Cells. J. Steroid Biochem. Mol. Biol. 2010, 121, 265–267. [Google Scholar] [CrossRef]
  54. Eftekhari, M.H.; Akbarzadeh, M.; Dabbaghmanesh, M.H.; Hassanzadeh, J. The Effect of Calcitriol on Lipid Profile and Oxidative Stress in Hyperlipidemic Patients with Type 2 Diabetes Mellitus. ARYA Atheroscler. 2014, 10, 82–88. [Google Scholar] [PubMed]
  55. Hamden, K.; Carreau, S.; Jamoussi, K.; Miladi, S.; Lajmi, S.; Aloulou, D.; Ayadi, F.; Elfeki, A. 1Alpha,25 Dihydroxyvitamin D3: Therapeutic and Preventive Effects against Oxidative Stress, Hepatic, Pancreatic and Renal Injury in Alloxan-Induced Diabetes in Rats. J. Nutr. Sci. Vitaminol. 2009, 55, 215–222. [Google Scholar] [CrossRef] [PubMed]
  56. Chwa, M.; Shao, Z.; Atilano, S.; Park, J.Y.; Karageozian, H.; Karageozian, V.; Kenney, M.C. Cell Viability and Transcriptome Changes Associated with Hydrogen Peroxide and Risuteganib Exposure in Human Retinal Cells In Vitro. Investig. Ophthalmol. Vis. Sci. 2021, 62, 3000. [Google Scholar]
  57. Park, W.H. H2O2 Inhibits the Growth of Human Pulmonary Fibroblast Cells by Inducing Cell Death, GSH Depletion and G1 Phase Arrest. Mol. Med. Rep. 2013, 7, 1235–1240. [Google Scholar] [CrossRef]
  58. Park, W.H. The Effects of Exogenous H2O2 on Cell Death, Reactive Oxygen Species and Glutathione Levels in Calf Pulmonary Artery and Human Umbilical Vein Endothelial Cells. Int. J. Mol. Med. 2013, 31, 471–476. [Google Scholar] [CrossRef]
  59. Konyalilar, N.; Yıldız, A.B.; Yazıcı, D.; Bayram, H. Effects of Hydrogen Peroxide and Butyrate on A549 Cell Viability and Permeability. Eur. Respir. J. 2019, 54, PA2402. [Google Scholar] [CrossRef]
  60. Anasooya Shaji, C.; Robinson, B.D.; Yeager, A.; Beeram, M.R.; Davis, M.L.; Isbell, C.L.; Huang, J.H.; Tharakan, B. The Tri-Phasic Role of Hydrogen Peroxide in Blood-Brain Barrier Endothelial Cells. Sci. Rep. 2019, 9, 133. [Google Scholar] [CrossRef]
  61. Luo, Y.; Wen, X.; Wang, L.; Gao, J.; Wang, Z.; Zhang, C.; Zhang, P.; Lu, C.; Duan, L.; Tian, Y. Identification of MicroRNAs Involved in Growth Arrest and Apoptosis in Hydrogen Peroxide-Treated Human Hepatocellular Carcinoma Cell Line HepG2. Oxid. Med. Cell. Longev. 2016, 2016, 7530853. [Google Scholar] [CrossRef]
  62. Nogueira-Pedro, A.; Cesário, T.A.M.; Dias, C.C.; Origassa, C.S.T.; Eça, L.P.M.; Paredes-Gamero, E.J.; Ferreira, A.T. Hydrogen Peroxide (H2O2) Induces Leukemic but Not Normal Hematopoietic Cell Death in a Dose-Dependent Manner. Cancer Cell Int. 2013, 13, 123. [Google Scholar] [CrossRef] [PubMed]
  63. Sliwinska, A.; Rogalska, A.; Szwed, M.; Kasznicki, J.; Jozwiak, Z.; Drzewoski, J. Gliclazide May Have an Antiapoptotic Effect Related to Its Antioxidant Properties in Human Normal and Cancer Cells. Mol. Biol. Rep. 2012, 39, 5253–5267. [Google Scholar] [CrossRef]
  64. Tohari, A.M.; Zhou, X.; Shu, X. Protection against Oxidative Stress by Vitamin D in Cone Cells. Cell Biochem. Funct. 2016, 34, 82–94. [Google Scholar] [CrossRef] [PubMed]
  65. Rogers, C.S.; Yedjou, C.G.; Sutton, D.J.; Tchounwou, P.B. Vitamin D3 Potentiates the Antitumorigenic Effects of Arsenic Trioxide in Human Leukemia (HL-60) Cells. Exp. Hematol. Oncol. 2014, 3, 9. [Google Scholar] [CrossRef]
  66. Piatek, K.; Kutner, A.; Cacsire Castillo-Tong, D.; Manhardt, T.; Kupper, N.; Nowak, U.; Chodyński, M.; Marcinkowska, E.; Kallay, E.; Schepelmann, M. Vitamin D Analogs Regulate the Vitamin D System and Cell Viability in Ovarian Cancer Cells. Int. J. Mol. Sci. 2021, 23, 172. [Google Scholar] [CrossRef]
  67. Bajbouj, K.; Sahnoon, L.; Shafarin, J.; Al-Ali, A.; Muhammad, J.S.; Karim, A.; Guraya, S.Y.; Hamad, M. Vitamin D-Mediated Anti-Cancer Activity Involves Iron Homeostatic Balance Disruption and Oxidative Stress Induction in Breast Cancer. Front. Cell Dev. Biol. 2021, 9, 766978. [Google Scholar] [CrossRef]
  68. Lisse, T.S. Vitamin D Regulation of a SOD1-to-SOD2 Antioxidative Switch to Prevent Bone Cancer. Appl. Sci. 2020, 10, 2554. [Google Scholar] [CrossRef]
  69. Varghese, J.E.; Shanmugam, V.; Rengarajan, R.L.; Meyyazhagan, A.; Arumugam, V.A.; Al-Misned, F.A.; El-Serehy, H.A. Role of Vitamin D3 on Apoptosis and Inflammatory-Associated Gene in Colorectal Cancer: An in Vitro Approach. J. King Saud Univ. -Sci. 2020, 32, 2786–2789. [Google Scholar] [CrossRef]
  70. Cataldi, S.; Ceccarini, M.R.; Patria, F.; Beccari, T.; Mandarano, M.; Ferri, I.; Lazzarini, A.; Curcio, F.; Albi, E. The Effect of Vitamin D3 and Silver Nanoparticles on HaCaT Cell Viability. Int. J. Mol. Sci. 2022, 23, 1410. [Google Scholar] [CrossRef]
  71. Tohari, A.M.; Alhasani, R.H.; Biswas, L.; Patnaik, S.R.; Reilly, J.; Zeng, Z.; Shu, X. Vitamin D Attenuates Oxidative Damage and Inflammation in Retinal Pigment Epithelial Cells. Antioxidants 2019, 8, 341. [Google Scholar] [CrossRef]
  72. Abdul Wahab, S.M.; Abd Hamid, Z.; Mathialagan, R.D.; Taib, I.S. Effect of N-Acetylcysteine Supplementation on Oxidative Stress-Mediated Cryoinjury of Bone Marrow Derived-Hematopoietic Stem Cells. Sains Malays. 2019, 48, 1937–1946. [Google Scholar] [CrossRef]
  73. Kim, G.-H.; Song, D.-K.; Cho, C.-H.; Yoo, S.K.; Kim, D.-K.; Park, G.-Y.; Suh, S.; Jang, B.-C.; Lim, J.-G. Muscular Cell Proliferative and Protective Effects of N-Acetylcysteine by Modulating Activity of Extracellular Signal-Regulated Protein Kinase. Life Sci. 2006, 79, 622–628. [Google Scholar] [CrossRef] [PubMed]
  74. Calvo, R.; Espinosa, M.; Figueroa, D.; Pozo, L.M.; Conget, P. Assessment of Cell Viability of Fresh Osteochondral Allografts in N-Acetylcysteine-Enriched Medium. Cartilage 2020, 11, 117–121. [Google Scholar] [CrossRef]
  75. Yu, Z.; Yu, K.; Wu, S.; Zhao, Q.; Guo, Y.; Liu, H.; Huang, X. Two Contradictory Facades of N-Acetylcysteine Activity towards Renal Carcinoma Cells. J. Taibah Univ. Sci. 2022, 16, 423–431. [Google Scholar] [CrossRef]
  76. Liu, Y.; Liu, K.; Wang, N.; Zhang, H. N-acetylcysteine Induces Apoptosis via the Mitochondria-dependent Pathway but Not via Endoplasmic Reticulum Stress in H9c2 Cells. Mol. Med. Rep. 2017, 16, 6626–6633. [Google Scholar] [CrossRef]
  77. Jurkowska, H.; Wróbel, M. Inhibition of Human Neuroblastoma Cell Proliferation by N-Acetyl-L-Cysteine as a Result of Increased Sulfane Sulfur Level. Anticancer Res. 2018, 38, 5109–5113. [Google Scholar] [CrossRef]
  78. Mitsopoulos, P.; Suntres, Z.E. Protective Effects of Liposomal N-Acetylcysteine against Paraquat-Induced Cytotoxicity and Gene Expression. J. Toxicol. 2011, 2011, 808967. [Google Scholar] [CrossRef]
  79. Liao, C.-Y.; Wu, T.-C.; Yang, S.-F.; Chang, J.T. Effects of NAC and Gallic Acid on the Proliferation Inhibition and Induced Death of Lung Cancer Cells with Different Antioxidant Capacities. Molecules 2021, 27, 75. [Google Scholar] [CrossRef]
  80. Piotrowska, A.; Wierzbicka, J.; Ślebioda, T.; Woźniak, M.; Tuckey, R.C.; Slominski, A.T.; Żmijewski, M.A. Vitamin D Derivatives Enhance Cytotoxic Effects of H2O2 or Cisplatin on Human Keratinocytes. Steroids 2016, 110, 49–61. [Google Scholar] [CrossRef] [PubMed]
  81. Tosi, G.M.; Giustarini, D.; Franci, L.; Minetti, A.; Imperatore, F.; Caldi, E.; Fiorenzani, P.; Aloisi, A.M.; Sparatore, A.; Rossi, R.; et al. Superior Properties of N-Acetylcysteine Ethyl Ester over N-Acetyl Cysteine to Prevent Retinal Pigment Epithelial Cells Oxidative Damage. Int. J. Mol. Sci. 2021, 22, 600. [Google Scholar] [CrossRef]
  82. Liu, X.; Wang, L.; Cai, J.; Liu, K.; Liu, M.; Wang, H.; Zhang, H. N-Acetylcysteine Alleviates H2O2-Induced Damage via Regulating the Redox Status of Intracellular Antioxidants in H9c2 Cells. Int. J. Mol. Med. 2019, 43, 199–208. [Google Scholar] [CrossRef]
  83. Ali, F.; Khan, M.; Khan, S.N.; Riazuddin, S. N-Acetyl Cysteine Protects Diabetic Mouse Derived Mesenchymal Stem Cells from Hydrogen-Peroxide-Induced Injury: A Novel Hypothesis for Autologous Stem Cell Transplantation. J. Chin. Med. Assoc. JCMA 2016, 79, 122–129. [Google Scholar] [CrossRef] [PubMed]
  84. Zhou, J.; Terluk, M.R.; Basso, L.; Mishra, U.R.; Orchard, P.J.; Cloyd, J.C.; Schröder, H.; Kartha, R.V. N-Acetylcysteine Provides Cytoprotection in Murine Oligodendrocytes through Heme Oxygenase-1 Activity. Biomedicines 2020, 8, 240. [Google Scholar] [CrossRef]
  85. Terluk, M.R.; Ebeling, M.C.; Fisher, C.R.; Kapphahn, R.J.; Yuan, C.; Kartha, R.V.; Montezuma, S.R.; Ferrington, D.A. N-Acetyl-L-Cysteine Protects Human Retinal Pigment Epithelial Cells from Oxidative Damage: Implications for Age-Related Macular Degeneration. Oxid. Med. Cell. Longev. 2019, 2019, 5174957. [Google Scholar] [CrossRef]
  86. Wu, W.; Liu, B.-H.; Xie, C.-L.; Xia, X.-D.; Zhang, Y.-M. Neuroprotective Effects of N-Acetyl Cysteine on Primary Hippocampus Neurons against Hydrogen Peroxide-Induced Injury Are Mediated via Inhibition of Mitogen-Activated Protein Kinases Signal Transduction and Antioxidative Action. Mol. Med. Rep. 2018, 17, 6647–6654. [Google Scholar] [CrossRef]
  87. Berridge, M.J. Vitamin D Cell Signalling in Health and Disease. Biochem. Biophys. Res. Commun. 2015, 460, 53–71. [Google Scholar] [CrossRef] [PubMed]
  88. Nair-Shalliker, V.; Armstrong, B.K.; Fenech, M. Does Vitamin D Protect against DNA Damage? Mutat. Res. 2012, 733, 50–57. [Google Scholar] [CrossRef] [PubMed]
  89. Nakai, K.; Fujii, H.; Kono, K.; Goto, S.; Kitazawa, R.; Kitazawa, S.; Hirata, M.; Shinohara, M.; Fukagawa, M.; Nishi, S. Vitamin D Activates the Nrf2-Keap1 Antioxidant Pathway and Ameliorates Nephropathy in Diabetic Rats. Am. J. Hypertens. 2014, 27, 586–595. [Google Scholar] [CrossRef]
  90. Vázquez-Lorente, H.; Herrera-Quintana, L.; Jiménez-Sánchez, L.; Fernández-Perea, B.; Plaza-Diaz, J. Antioxidant Functions of Vitamin D and CYP11A1-Derived Vitamin D, Tachysterol, and Lumisterol Metabolites: Mechanisms, Clinical Implications, and Future Directions. Antioxidants 2024, 13, 996. [Google Scholar] [CrossRef]
  91. Maltese, G.; Psefteli, P.-M.; Rizzo, B.; Srivastava, S.; Gnudi, L.; Mann, G.E.; Siow, R.C.M. The Anti-Ageing Hormone Klotho Induces Nrf2-Mediated Antioxidant Defences in Human Aortic Smooth Muscle Cells. J. Cell. Mol. Med. 2017, 21, 621–627. [Google Scholar] [CrossRef] [PubMed]
  92. Kállay, E.; Bareis, P.; Bajna, E.; Kriwanek, S.; Bonner, E.; Toyokuni, S.; Cross, H.S. Vitamin D Receptor Activity and Prevention of Colonic Hyperproliferation and Oxidative Stress. Food Chem. Toxicol. 2002, 40, 1191–1196. [Google Scholar] [CrossRef]
  93. Fedirko, V.; Bostick, R.M.; Long, Q.; Flanders, W.D.; McCullough, M.L.; Sidelnikov, E.; Daniel, C.R.; Rutherford, R.E.; Shaukat, A. Effects of Supplemental Vitamin D and Calcium on Oxidative DNA Damage Marker in Normal Colorectal Mucosa: A Randomized Clinical Trial. Cancer Epidemiol. Biomark. Prev. 2010, 19, 280–291. [Google Scholar] [CrossRef]
  94. Wenclewska, S.; Szymczak-Pajor, I.; Drzewoski, J.; Bunk, M.; Śliwińska, A. Vitamin D Supplementation Reduces Both Oxidative DNA Damage and Insulin Resistance in the Elderly with Metabolic Disorders. Int. J. Mol. Sci. 2019, 20, 2891. [Google Scholar] [CrossRef]
  95. Graziano, S.; Johnston, R.; Deng, O.; Zhang, J.; Gonzalo, S. Vitamin D/Vitamin D Receptor Axis Regulates DNA Repair during Oncogene-Induced Senescence. Oncogene 2016, 35, 5362–5376. [Google Scholar] [CrossRef]
  96. Zentout, S.; Smith, R.; Jacquier, M.; Huet, S. New Methodologies to Study DNA Repair Processes in Space and Time Within Living Cells. Front. Cell Dev. Biol. 2021, 9, 730998. [Google Scholar] [CrossRef] [PubMed]
  97. Peehl, D.M.; Shinghal, R.; Nonn, L.; Seto, E.; Krishnan, A.V.; Brooks, J.D.; Feldman, D. Molecular Activity of 1,25-Dihydroxyvitamin D3 in Primary Cultures of Human Prostatic Epithelial Cells Revealed by cDNA Microarray Analysis. J. Steroid Biochem. Mol. Biol. 2004, 92, 131–141. [Google Scholar] [CrossRef] [PubMed]
  98. Swami, S.; Raghavachari, N.; Muller, U.R.; Bao, Y.P.; Feldman, D. Vitamin D Growth Inhibition of Breast Cancer Cells: Gene Expression Patterns Assessed by cDNA Microarray. Breast Cancer Res. Treat. 2003, 80, 49–62. [Google Scholar] [CrossRef]
  99. Lambert, J.R.; Kelly, J.A.; Shim, M.; Huffer, W.E.; Nordeen, S.K.; Baek, S.J.; Eling, T.E.; Lucia, M.S. Prostate Derived Factor in Human Prostate Cancer Cells: Gene Induction by Vitamin D via a P53-Dependent Mechanism and Inhibition of Prostate Cancer Cell Growth. J. Cell. Physiol. 2006, 208, 566–574. [Google Scholar] [CrossRef]
  100. Rathor, R.; Suryakumar, G. Exploring miRNA Function in Maintaining Redox Mechanism of High Altitude Hypoxia Associated Maladies: An Evidence Based Study. Adv. Redox Res. 2024, 11, 100103. [Google Scholar] [CrossRef]
  101. Zhao, H.; Forcellati, M.; Buschittari, D.; Heckel, J.E.; Machado, C.J.; Ramulu, N.; Pullagura, S.; Lisse, T.S. Hewison, M., Bouillon, R., Giovannucci, E., Goltzman, D., Meyer, M., Welsh, J., Eds.; Chapter 14-Vitamin D and microRNAs. In Feldman and Pike’ s Vitamin D, 5th ed.; Academic Press: Cambridge, MA, USA, 2024; pp. 261–290. ISBN 978-0-323-91386-7. [Google Scholar]
  102. Zhao, S.; Mao, L.; Wang, S.-G.; Chen, F.-L.; Ji, F.; Fei, H.-D. MicroRNA-200a Activates Nrf2 Signaling to Protect Osteoblasts from Dexamethasone. Oncotarget 2017, 8, 104867–104876. [Google Scholar] [CrossRef]
  103. Ashrafizadeh, M.; Ahmadi, Z.; Samarghandian, S.; Mohammadinejad, R.; Yaribeygi, H.; Sathyapalan, T.; Sahebkar, A. MicroRNA-Mediated Regulation of Nrf2 Signaling Pathway: Implications in Disease Therapy and Protection against Oxidative Stress. Life Sci. 2020, 244, 117329. [Google Scholar] [CrossRef]
  104. Kobayashi, A.; Kang, M.-I.; Okawa, H.; Ohtsuji, M.; Zenke, Y.; Chiba, T.; Igarashi, K.; Yamamoto, M. Oxidative Stress Sensor Keap1 Functions as an Adaptor for Cul3-Based E3 Ligase to Regulate Proteasomal Degradation of Nrf2. Mol. Cell. Biol. 2004, 24, 7130–7139. [Google Scholar] [CrossRef]
  105. Zhang, M.; An, C.; Gao, Y.; Leak, R.K.; Chen, J.; Zhang, F. Emerging Roles of Nrf2 and Phase II Antioxidant Enzymes in Neuroprotection. Prog. Neurobiol. 2013, 100, 30–47. [Google Scholar] [CrossRef]
  106. Batliwala, S.; Xavier, C.; Liu, Y.; Wu, H.; Pang, I.-H. Involvement of Nrf2 in Ocular Diseases. Oxid. Med. Cell. Longev. 2017, 2017, 1703810. [Google Scholar] [CrossRef] [PubMed]
  107. Middleton, R.P.; Nelson, R.; Li, Q.; Blanton, A.; Labuda, J.A.; Vitt, J.; Inpanbutr, N. 1,25-Dihydroxyvitamin D3 and Its Analogues Increase Catalase at the mRNA, Protein and Activity Level in a Canine Transitional Carcinoma Cell Line. Vet. Comp. Oncol. 2015, 13, 452–463. [Google Scholar] [CrossRef] [PubMed]
  108. Banakar, M.C.; Paramasivan, S.K.; Chattopadhyay, M.B.; Datta, S.; Chakraborty, P.; Chatterjee, M.; Kannan, K.; Thygarajan, E. 1alpha, 25-Dihydroxyvitamin D3 Prevents DNA Damage and Restores Antioxidant Enzymes in Rat Hepatocarcinogenesis Induced by Diethylnitrosamine and Promoted by Phenobarbital. World J. Gastroenterol. 2004, 10, 1268–1275. [Google Scholar] [CrossRef]
  109. AlJohri, R.; AlOkail, M.; Haq, S.H. Neuroprotective Role of Vitamin D in Primary Neuronal Cortical Culture. eNeurologicalSci 2019, 14, 43–48. [Google Scholar] [CrossRef]
  110. Bhat, M.; Ismail, A. Vitamin D Treatment Protects against and Reverses Oxidative Stress Induced Muscle Proteolysis. J. Steroid Biochem. Mol. Biol. 2015, 152, 171–179. [Google Scholar] [CrossRef] [PubMed]
  111. Saif-Elnasr, M.; Ibrahim, I.M.; Alkady, M.M. Role of Vitamin D on Glycemic Control and Oxidative Stress in Type 2 Diabetes Mellitus. J. Res. Med. Sci. 2017, 22, 22. [Google Scholar] [CrossRef]
  112. Sánchez-Virosta, P.; Espín, S.; Ruiz, S.; Stauffer, J.; Kanerva, M.; García-Fernández, A.J.; Eeva, T. Effects of Calcium Supplementation on Oxidative Status and Oxidative Damage in Great Tit Nestlings Inhabiting a Metal-Polluted Area. Environ. Res. 2019, 171, 484–492. [Google Scholar] [CrossRef] [PubMed]
  113. Szlacheta, Z.; Wąsik, M.; Machoń-Grecka, A.; Kasperczyk, A.; Dobrakowski, M.; Bellanti, F.; Szlacheta, P.; Kasperczyk, S. Potential Antioxidant Activity of Calcium and Selected Oxidative Stress Markers in Lead- and Cadmium-Exposed Workers. Oxid. Med. Cell. Longev. 2020, 2020, 8035631. [Google Scholar] [CrossRef] [PubMed]
  114. Taneera, J.; Yaseen, D.; Youssef, M.; Khalique, A.; Al Shehadat, O.S.; Mohammed, A.K.; Bustanji, Y.; Madkour, M.I.; El-Huneidi, W. Vitamin D Augments Insulin Secretion via Calcium Influx and Upregulation of Voltage Calcium Channels: Findings from INS-1 Cells and Human Islets. Mol. Cell. Endocrinol. 2025, 599, 112472. [Google Scholar] [CrossRef]
  115. Bittiner, B.; Bleehen, S.S.; Macneil, S. 1α,25(OH)2 Vitamin D3 Increases Intracellular Calcium in Human Keratinocytes. Br. J. Dermatol. 1991, 124, 230–235. [Google Scholar] [CrossRef]
  116. Alatawi, F.S.; Faridi, U.A.; Alatawi, M.S. Effect of Treatment with Vitamin D plus Calcium on Oxidative Stress in Streptozotocin-Induced Diabetic Rats. Saudi Pharm. J. 2018, 26, 1208–1213. [Google Scholar] [CrossRef]
  117. Giorello, M.B.; Borzone, F.R.; Labovsky, V.; Piccioni, F.V.; Chasseing, N.A. Cancer-Associated Fibroblasts in the Breast Tumor Microenvironment. J. Mammary Gland Biol. Neoplasia 2021, 26, 135–155. [Google Scholar] [CrossRef]
  118. Brichkina, A.; Polo, P.; Sharma, S.D.; Visestamkul, N.; Lauth, M. A Quick Guide to CAF Subtypes in Pancreatic Cancer. Cancers 2023, 15, 2614. [Google Scholar] [CrossRef]
  119. Łabędź, N.; Anisiewicz, A.; Stachowicz-Suhs, M.; Banach, J.; Kłopotowska, D.; Maciejczyk, A.; Gazińska, P.; Piotrowska, A.; Dzięgiel, P.; Matkowski, R.; et al. Dual Effect of Vitamin D3 on Breast Cancer-Associated Fibroblasts. BMC Cancer 2024, 24, 209. [Google Scholar] [CrossRef]
  120. Li, H.; Ruan, Y.; Liu, C.; Fan, X.; Yao, Y.; Dai, Y.; Song, Y.; Jiang, D.; Sun, N.; Jiao, G.; et al. VDR Promotes Pancreatic Cancer Progression in Vivo by Activating CCL20-Mediated M2 Polarization of Tumor Associated Macrophage. Cell Commun. Signal. CCS 2024, 22, 224. [Google Scholar] [CrossRef] [PubMed]
  121. Gradiz, R.; Silva, H.C.; Carvalho, L.; Botelho, M.F.; Mota-Pinto, A. MIA PaCa-2 and PANC-1 - Pancreas Ductal Adenocarcinoma Cell Lines with Neuroendocrine Differentiation and Somatostatin Receptors. Sci. Rep. 2016, 6, 21648. [Google Scholar] [CrossRef]
Figure 1. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), a combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and NAC with H2O2 (NAC + H2O2) on the viability of (a) 1.2B4 cells and (b) PANC-1 cells after 24 h (violet lines), 48 h (blue lines) and 72 h (orange lines) of exposure. Cell viability was determined by MTT assay. The PC cells were treated with Vit D3 (5–100 nM), H2O2 (50–750 µM), Vit. D3 (5–100 nM) with H2O2 (400 µM for 1.2B4 cells or 300 µM for PANC-1 cells), NAC (0.001–30 mM), NAC (0.001–30 mM) with H2O2 (400 µM for 1.2B4 cells or 300 µM for PANC-1 cells) for 24 h, 48 h and 72 h. After treatment, MTT was added for 4 h. Then, DMSO was added to dissolve formazan crystals and absorbance was measured at 570 nm with a microplate reader. The cell viability (the data) was determined as a percentage of control. The values for the control group (non-treated cells) were considered to be 100%. The data are expressed as the mean ± standard deviation (SD) of the three independent experiments. * p < 0.05, ** p < 0.01 and *** p < 0.001. vs. control (non-treated cells for Vit. D3, H2O2, NAC or H2O2–treated cells for Vit. D3 + H2O2, NAC + H2O2). Violet stars refer to 24 h; blue stars to 48 h; orange stars to 72 h.
Figure 1. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), a combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and NAC with H2O2 (NAC + H2O2) on the viability of (a) 1.2B4 cells and (b) PANC-1 cells after 24 h (violet lines), 48 h (blue lines) and 72 h (orange lines) of exposure. Cell viability was determined by MTT assay. The PC cells were treated with Vit D3 (5–100 nM), H2O2 (50–750 µM), Vit. D3 (5–100 nM) with H2O2 (400 µM for 1.2B4 cells or 300 µM for PANC-1 cells), NAC (0.001–30 mM), NAC (0.001–30 mM) with H2O2 (400 µM for 1.2B4 cells or 300 µM for PANC-1 cells) for 24 h, 48 h and 72 h. After treatment, MTT was added for 4 h. Then, DMSO was added to dissolve formazan crystals and absorbance was measured at 570 nm with a microplate reader. The cell viability (the data) was determined as a percentage of control. The values for the control group (non-treated cells) were considered to be 100%. The data are expressed as the mean ± standard deviation (SD) of the three independent experiments. * p < 0.05, ** p < 0.01 and *** p < 0.001. vs. control (non-treated cells for Vit. D3, H2O2, NAC or H2O2–treated cells for Vit. D3 + H2O2, NAC + H2O2). Violet stars refer to 24 h; blue stars to 48 h; orange stars to 72 h.
Antioxidants 14 01101 g001
Figure 2. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine -acetylcysteine (NAC) and the combinations of Vit. D3 with H2O2 (Vit. D3 + H2O2) and NAC with H2O2 (NAC + H2O2) on DNA damage level in pancreatic cancer (PC) cells. The 1.2B4 cells (a) and PANC-1 (b) cells were non-treated (C, black bars) and treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, orange bars), NAC (3 mM, blue bars), NAC (3 mM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, violet bars) for 24, 48 and 72 h. The data are expressed as a % DNA in the tail of the comet counted from 50 cells. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. aaa p < 0.001 vs. C; ddd p < 0.001 vs. H2O2; eee p < 0.001 vs. 75D3; fff p < 0.001 vs. 100D3; ggg p < 0.01 vs. 75D3 + H2O2.
Figure 2. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine -acetylcysteine (NAC) and the combinations of Vit. D3 with H2O2 (Vit. D3 + H2O2) and NAC with H2O2 (NAC + H2O2) on DNA damage level in pancreatic cancer (PC) cells. The 1.2B4 cells (a) and PANC-1 (b) cells were non-treated (C, black bars) and treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, orange bars), NAC (3 mM, blue bars), NAC (3 mM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, violet bars) for 24, 48 and 72 h. The data are expressed as a % DNA in the tail of the comet counted from 50 cells. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. aaa p < 0.001 vs. C; ddd p < 0.001 vs. H2O2; eee p < 0.001 vs. 75D3; fff p < 0.001 vs. 100D3; ggg p < 0.01 vs. 75D3 + H2O2.
Antioxidants 14 01101 g002
Figure 3. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on mRNA expression of the antioxidant enzymes (a) catalase (CAT), (b) sodium dismutase 1 (SOD1), (c) sodium dismutase 2 (SOD2), (d) sodium dismutase 3 (SOD3) and (e) glutathione peroxidase 3 (Gpx3) in 1.2B4 cells. The 1.2B4 cells were non-treated (C, black bars) and treated with Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM, orange bars), NAC (3 mM, blue bars), NAC (3 mM) with H2O2 (400 µM, violet bars) for 24, 48 and 72 h. Data are expressed as the mean ± standard deviation (SD) of the fold change of three independent experiments in relation to the untreated control. GAPDH was used as a reference gene. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05; bb p < 0.01; bbb p < 0.001 vs. H2O2; c p < 0.05; cc p < 0.01 vs. 75D3; d p < 0.05; dd p < 0.01 vs. 100D3; e p < 0.05; ee p < 0.01 vs. 75D3 + H2O2; f p < 0.05 vs. 100D3 + H2O2.
Figure 3. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on mRNA expression of the antioxidant enzymes (a) catalase (CAT), (b) sodium dismutase 1 (SOD1), (c) sodium dismutase 2 (SOD2), (d) sodium dismutase 3 (SOD3) and (e) glutathione peroxidase 3 (Gpx3) in 1.2B4 cells. The 1.2B4 cells were non-treated (C, black bars) and treated with Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM, orange bars), NAC (3 mM, blue bars), NAC (3 mM) with H2O2 (400 µM, violet bars) for 24, 48 and 72 h. Data are expressed as the mean ± standard deviation (SD) of the fold change of three independent experiments in relation to the untreated control. GAPDH was used as a reference gene. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05; bb p < 0.01; bbb p < 0.001 vs. H2O2; c p < 0.05; cc p < 0.01 vs. 75D3; d p < 0.05; dd p < 0.01 vs. 100D3; e p < 0.05; ee p < 0.01 vs. 75D3 + H2O2; f p < 0.05 vs. 100D3 + H2O2.
Antioxidants 14 01101 g003
Figure 4. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the mRNA expression of the antioxidant enzymes (a) catalase (CAT), (b) sodium dismutase 1 (SOD1), (c) sodium dismutase 2 (SOD2), (d) sodium dismutase 3 (SOD3), (e) glutathione peroxidase 3 (Gpx3) in PANC-1 cells. The PANC-1 cells were non-treated (C, black bars) and treated with Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (300 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (300 µM, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (300, violet bars) for 24, 48 and 72 h. Data are expressed as the mean ± standard deviation (SD) of the fold change of three independent experiments in relation to the untreated control. GAPDH was used as a reference gene. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05; bb p < 0.01; bbb p < 0.001 vs. H2O2; c p < 0.05; cc p < 0.01 vs. 75D3; d p < 0.05; dd p < 0.01; ddd p < 0.001 vs. 100D3; e p < 0.05; ee p < 0.01 vs. 75D3 + H2O2; f p < 0.05; ff p < 0.01 vs. 100D3 + H2O2; g p < 0.05; gg p < 0.01 vs. NAC.
Figure 4. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the mRNA expression of the antioxidant enzymes (a) catalase (CAT), (b) sodium dismutase 1 (SOD1), (c) sodium dismutase 2 (SOD2), (d) sodium dismutase 3 (SOD3), (e) glutathione peroxidase 3 (Gpx3) in PANC-1 cells. The PANC-1 cells were non-treated (C, black bars) and treated with Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (300 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (300 µM, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (300, violet bars) for 24, 48 and 72 h. Data are expressed as the mean ± standard deviation (SD) of the fold change of three independent experiments in relation to the untreated control. GAPDH was used as a reference gene. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05; bb p < 0.01; bbb p < 0.001 vs. H2O2; c p < 0.05; cc p < 0.01 vs. 75D3; d p < 0.05; dd p < 0.01; ddd p < 0.001 vs. 100D3; e p < 0.05; ee p < 0.01 vs. 75D3 + H2O2; f p < 0.05; ff p < 0.01 vs. 100D3 + H2O2; g p < 0.05; gg p < 0.01 vs. NAC.
Antioxidants 14 01101 g004
Figure 5. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the reactive oxygen species (ROS) level in pancreatic cancer (PC) cells. The 1.2B4 cells (a) and PANC-1 (b) cells were treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, violet bars) for 2 and 24 h. After the completion of treatment, DCFH2-DA was added and incubated for 30 min. Then, the fluorescence of DCF was measured using microplate reader at a 530 nm excitation at 485 nm. The data are expressed as a percentage of the control. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. c p < 0.05; cc p < 0.01; ccc p < 0.001 vs. H2O2; dd p < 0.01 vs. 75D3; ee p < 0.01 vs. 100D3; f p < 0.05; fff p < 0.001 vs. NAC.
Figure 5. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the reactive oxygen species (ROS) level in pancreatic cancer (PC) cells. The 1.2B4 cells (a) and PANC-1 (b) cells were treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (400 µM for 1.2B4 cells; 300 µM for PANC-1 cells, violet bars) for 2 and 24 h. After the completion of treatment, DCFH2-DA was added and incubated for 30 min. Then, the fluorescence of DCF was measured using microplate reader at a 530 nm excitation at 485 nm. The data are expressed as a percentage of the control. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. c p < 0.05; cc p < 0.01; ccc p < 0.001 vs. H2O2; dd p < 0.01 vs. 75D3; ee p < 0.01 vs. 100D3; f p < 0.05; fff p < 0.001 vs. NAC.
Antioxidants 14 01101 g005
Figure 6. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the activity of the antioxidant enzymes catalase (CAT) (a), sodium dismutase (SOD) (b) and glutathione peroxidase (Gpx) (c) in 1.2B4 cells. The cells were non-treated (C, black bars) and treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (400 µM, violet bars) for 2 and 24 h. After the completion of the treatment, the cells were homogenized in cold buffer to determine CAT, SOD and Gpx activity, respectively. The absorbance was measured at 540 nm for CAT, 450 nm for SOD and 340 nm for Gpx. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05 vs. Et-OH1; cc p < 0.01; ccc p < 0.001 vs. Et-OH2; d p < 0.05; dd p < 0.01; ddd p < 0.001 vs. H2O2.
Figure 6. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the activity of the antioxidant enzymes catalase (CAT) (a), sodium dismutase (SOD) (b) and glutathione peroxidase (Gpx) (c) in 1.2B4 cells. The cells were non-treated (C, black bars) and treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (400 µM, violet bars) for 2 and 24 h. After the completion of the treatment, the cells were homogenized in cold buffer to determine CAT, SOD and Gpx activity, respectively. The absorbance was measured at 540 nm for CAT, 450 nm for SOD and 340 nm for Gpx. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05 vs. Et-OH1; cc p < 0.01; ccc p < 0.001 vs. Et-OH2; d p < 0.05; dd p < 0.01; ddd p < 0.001 vs. H2O2.
Antioxidants 14 01101 g006
Figure 7. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the activity of antioxidant enzymes catalase (CAT) (a), sodium dismutase (SOD) (b) and glutathione peroxidase (Gpx) (c) in PANC-1 cells. The cells were non-treated (C, black bars) and treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (400 µM, violet bars) for 2 and 24 h. After the completion of the treatment, the cells were homogenized in cold buffer to determine CAT, SOD and Gpx activity, respectively. The absorbance was measured at 540 nm for CAT, 450 nm for SOD and 340 nm for Gpx. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05; vs. Et-OH1; c p < 0.05 vs. Et-OH2; d p < 0.05; dd p < 0.01; ddd p < 0.001 vs. H2O2.
Figure 7. The effect of vitamin D3 (Vit. D3), hydrogen peroxide (H2O2), N-Acetyl-L-Cysteine (NAC), the combination of Vit. D3 with H2O2 (Vit. D3 + H2O2) and the combination of NAC with H2O2 (NAC + H2O2) on the activity of antioxidant enzymes catalase (CAT) (a), sodium dismutase (SOD) (b) and glutathione peroxidase (Gpx) (c) in PANC-1 cells. The cells were non-treated (C, black bars) and treated with ethanol (Et-OH, gray bars), Vit D3 (75 nM; 100 nM, yellow bars), H2O2 (400 µM, brown bars), Vit. D3 (75 nM; 100 nM) with H2O2 (400 µM, orange bars), NAC (3 mM, blue bars) and NAC (3 mM) with H2O2 (400 µM, violet bars) for 2 and 24 h. After the completion of the treatment, the cells were homogenized in cold buffer to determine CAT, SOD and Gpx activity, respectively. The absorbance was measured at 540 nm for CAT, 450 nm for SOD and 340 nm for Gpx. The data are presented as the mean ± standard deviation (SD) of the three independent experiments. a p < 0.05; aa p < 0.01; aaa p < 0.001 vs. C; b p < 0.05; vs. Et-OH1; c p < 0.05 vs. Et-OH2; d p < 0.05; dd p < 0.01; ddd p < 0.001 vs. H2O2.
Antioxidants 14 01101 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Szymczak-Pajor, I.; Antanaviciute, E.M.; Drzewoski, J.; Majsterek, I.; Śliwińska, A. Vitamin D Protects Pancreatic Cancer (PC) Cells from Death and DNA Damage Induced by Oxidative Stress. Antioxidants 2025, 14, 1101. https://doi.org/10.3390/antiox14091101

AMA Style

Szymczak-Pajor I, Antanaviciute EM, Drzewoski J, Majsterek I, Śliwińska A. Vitamin D Protects Pancreatic Cancer (PC) Cells from Death and DNA Damage Induced by Oxidative Stress. Antioxidants. 2025; 14(9):1101. https://doi.org/10.3390/antiox14091101

Chicago/Turabian Style

Szymczak-Pajor, Izabela, Egle Morta Antanaviciute, Józef Drzewoski, Ireneusz Majsterek, and Agnieszka Śliwińska. 2025. "Vitamin D Protects Pancreatic Cancer (PC) Cells from Death and DNA Damage Induced by Oxidative Stress" Antioxidants 14, no. 9: 1101. https://doi.org/10.3390/antiox14091101

APA Style

Szymczak-Pajor, I., Antanaviciute, E. M., Drzewoski, J., Majsterek, I., & Śliwińska, A. (2025). Vitamin D Protects Pancreatic Cancer (PC) Cells from Death and DNA Damage Induced by Oxidative Stress. Antioxidants, 14(9), 1101. https://doi.org/10.3390/antiox14091101

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop