Skip to main content
  • Primary research
  • Open access
  • Published:

Evaluation of mutagenesis, necrosis and apoptosis induced by omeprazole in stomach cells of patients with gastritis

Abstract

Background

Gastritis is a superficial and prevalent inflammatory lesion that is considered a public health concern once can cause gastric ulcers and gastric cancer, especially when associated with Helicobacter pylori infection. Proton pump inhibitors, such as omeprazole, are the most widely used drugs to treat this illness. The aim of the study was evaluate cytogenetic effects of omeprazole in stomach epithelial cells of patients with gastritis in presence and absence of H. pylori, through cytogenetic biomarkers and catalse and superoxide dismutase analysis.

Methods

The study included 152 patients from the Gastroenterology Outpatient Clinic of Hospital Getúlio Vargas, Teresina—Brazil, that reported continuous and prolonged omeprazole use in doses of 20, 30 and 40 mg/kg. The participants were divided into groups: (1) patients without gastritis (n = 32); (2) patients without gastritis but with OME use (n = 24); (3) patients with gastritis (n = 26); (4) patients with gastritis undergoing OME therapy (n = 26); (5) patients with gastritis and H. pylori (n = 22) and (6) patients with gastritis and H. pylori on OME therapy (n = 22).

Results

OME induced cytogenetic imbalance in the stomach epithelium through the formation of micronuclei (group 6 > 1, 2, 3, 4, 5; group 5 > 1, 2, 3; group 4 > 1, 2, 3); bridges (groups 4 and 6 > 1, 2, 3, 5 and group 2 > 3, 5); buds (groups 2,4,6 > , 1, 3, 5); binucleated cells (group 6 > 1, 2, 3, 4, 5; group 4 > 1, 2, 3); (groups 2 and 3 > 1); picnoses (group 6 > 1, 2, 3, 4, 5), groups 2 and 5 > 1, 3; group 4 > 1, 2, 3, 5); cariorrexis (groups 6 and 4 > 1, 2, 3, 5; groups 2, 3, 5 > 1) and karyolysis (groups 2, 4, and 6 > 1, 3, 5; groups 3 and 5 > 1). The OME cytogenetic instability was associated with H. pylori infection, indicating clastogenic/aneugenic effects, chromosomes alterations, gene expression changes, cytotoxicity and apoptosis.

Conclusions

The cytogenetic changescan be attributed to several mechanisms that are still unclear, including oxidative damage, as observed by increased catalase and superoxide dismutase expresion. Positive correlations between antioxidant enzymes were found with micronuclei formation, and were negative for picnoses. Thus, the continuous and prolonged omeprazole use induces genetic instability, which can be monitored through cytogenetic analyzes, as precursor for gastric cancer.

Introduction

Gastric lesions, such as gastritis, can damage the gastric wall and/or duodenal epithelium, producing ulcers and even cancer [27, 43, 49]. Other pathological gastric changes may occur due to Helicobacter pylori (H. pylori) co-infection, which increase hydrochloric acid and pepsin secretion by the gastric mucosa [27, 43]. Proton Pump Inhibitors (PPIs) such as Omeprazole (OME), lansoprasol, pantoprazole, esomeprazole, among others are used to suppress gastric acidity [55, 64, 88], and have been used in long-term therapies [16, 17, 46, 70].

PPIs cause several adverse effects, and there are reports that its prolonged use promotes cardiovascular and renal alterations, anemia, thrombocytopenia, gastric polyps and carcinoma [31, 40, 94, 107]. In addition, PPIs can induce DNA fragmentation [29], causing chromosome rearrangements [91], as well as chromosomes breaks, which increase micronuclei formation (MN) [8, 84]. Genotoxic drugs lead to tMN formation [1, 36] and other types of nuclear alterations such as apoptosis, cytoplasmic bridges and nuclear shoots [33, 34]. Therefore, cytogenetic evaluations are important for several human diseases, as well as for therapeutic monitoring of genotoxicity [11, 48, 87].

Drug metabolic products can induce genetic mutations, breaks and/or chromosome rearrangements. There are reports of OME metabolites cytotoxicity, hepatoxicity and carcinogenicity [91]. Cytochrome P450 enzymes (CYPs) act on PPIs metabolism [26], which are considered monoxygenases responsible for the metabolism of several drugs through expodification, hydrolyzation, desulfurization, dealkylation, oxidation or sulfoxidation reactions [6]. OME is biotransformed in the liver by enzymatic activity of CYP2C19 and CYP3A4, producing the 5-hydroxy (5-OH) omeprazole and omeprazole Sulfona metabolites [30, 71].

In non-clinical studies, OME toxicogenic effects were identified in plant cells (Allium cepa) [13], as well as Saccharomyces cerevisiae and murine Sarcoma 180 cells [75]. Thus, based in previous investigations, the present study aimed to evaluate cytogenetic damages in patients whith gastritis undergoing OME therapy, with and without H. pylori co-infection, through micronucleus test and expression of catalase and superoxide dismutase.

Materials and methods

Ethical aspects

The present study was a controlled cross-sectional research, approved by the Research Ethics Committees (CEP) of UNINOVAFAPI (no. 1.521.307), Federal University of Piauí—UFPI (no. 1.607.441) and Ethics Committee of Hospital Getúlio Vargas (No. 1,569,041). All participants agreed to participate voluntarily and signed the Free and Informed Consent Form (FICF), in accordance with resolution 466/12 of the National Health Council.

Study location and sample

One hundred and fifty-two patients from the Gastroenterology Outpatient Clinic of Hospital Getúlio Vargas, Teresina—PI (2017–2019) were enrolled in this study. The participants had been undergone to upper digestive endoscopy and reported prolonged OME use (or not) in doses of 20 and 40 mg/kg. Medical reports about the presence or absence of gastric diseases, including gastritis and H. pylori infection, were examined after endoscopy and urea test. Participants were grouped according to the criteria: WG—patients without gastritis (n = 32); WG + OME—patients without gastritis with OME use (n = 24); G—patients with gastritis (n = 26); G + OME—patients with gastritis and OME use (n = 26); G + HP—patients with gastritis and H. pylori (n = 22) and G + HP + OME—patients with gastritis and H. pylori, using OME (n = 22).

Inclusion and exclusion criteria

The study included patients: (1) with or without gastritis; over 18 years old and legally responsible; (2) that signed the informed consent form; (3) that have prolonged OME useor not, by medical recommendation or self-medication, or reported no OME use;. The following participants were excluded: (1) over 70-year-old; (2) current illnesses that required surgical treatment, chemotherapy or radiotherapy.

Data collection

After Informed Consent Form signed, questionnaires were applied according to Carrano and Natarajam [21] based on the protocol published by the International Commission for Environmental Protection against Mutagens and Carcinogens (ICPEMC), with adaptations for nutritional aspects, socio-cultural and health and lifestyle information.

Exfoliated cells from the oral epithelium of patients undergoing endoscopy were obtained by scraping the inside of the cheek with a cytobrush. Cells of gastric epithelium (region of the body and antrum of the stomach) were collected at the time of endoscopy. All samples were placed in tubes with sodium phosphate buffer (PBS) (50 mM, pH 7.4), properly identified, and transported in dry ice to the Laboratory of Toxicological Genetics of the Federal University of Piauí, for immediately processing and tests. Peripheral blood samples were collected with heparin, and transported similarly to the other samples. The urea test was performed by the hospital’s medical team and the results were released together with the medical report.

Micronucleus test on exfoliated cells of the stomach epithelium

The Micronucleus Test was performed according to Thomas et al. [95], with some adaptations. Samples of the stomach epithelium (antrum and body) were collected during endoscopy. The material collected was placed in identified and previously prepared microtubes, containing 5 mL of saline solution (0.9% NaCl). Immediately after, the samples were sent to the Laboratory of Toxicological Genetics at the Federal University of Piauí in for analysis. To avoid external contamination, cell samples were washed three times before smear preparation. The washing process was carried out in 5 mL saline solution (0.9% NaCl), with centrifugations for 10 min at 1500 rpm, followed by removal of the supernatant and replacement of the solution always in the final volume of 5 mL. Two slides were prepared for each patient. After fixation, with methanol/acetic acid (3:1), the slides were stained with 2% Giemsa. Then, the slides were washed twice in distilled water for 3 min and, finally, dried at room temperature. The incidence of micronuclei, nuclear buds, binucleated cells and nuclear abnormalities that represent cell death, carioretic, pycnotic and karyolitic cells, were observed in 2000 cells per patient with the use of optical microscopy, in the 1000 × amplification.

Profile of patients’ enzymatic antioxidant defenses

From the peripheral blood samples collected, 10% erythrocyte homogenates were prepared (50 mM sodium phosphate buffer pH 7.4), which were centrifuged (800g, 20 min) and the supernatants used for catalase (CAT) activity assay. The reaction medium was prepared with H2O2 (18 mL) plus 1 M Tris HCl Buffer, 5 nM EDTA pH 8.0 (1.0 mL) and H2O (0.8 mL). Then, 980 μL of the reaction medium and 20 μL of the 10% erythrocyte homogenate was placed in the quartz cuvette. Finally, the reading was performed in a spectrophotometer for 6 min at 230 nm. The blank was made by reading the relative absorbance at 230 nm with only 1 mL of the reaction medium [23]. The protein concentration was determined [58]. The results were expressed in mmol/min/mg of protein. The homogenates of the 10% erythrocytes (50 mM sodium phosphate buffer, pH 7.4) were also centrifuged (800g, 20 min) and the supernatants used for superoxide dismutase (SOD) activity assessment. SOD activity was tested using the reduction rate of cytochrome C by superoxide radicals, using the xanthine-xanthine oxidase system as a source of superoxide anion (O2) [5]. The results were expressed in U/mg of protein. One unit (U) of SOD activity corresponds to a 50% inhibition of the reaction of O2 with cytochrome C. For protein concentration, the method of Lowry et al. [58] was used.

Urease test

The evaluation of H. pylori presence was performed by the urea test, according to Uotani and Graham [103]. Samples of gastric epithelial mucosa (antrum and body) were collected by biopsy during endoscopy. The urease test was performed and the result was obtained together with the endoscopy report.

Statistical analysis

The results of the analyzed biomarkers were presented as mean ± the standard deviation from mean. The data obtained were evaluated using Analysis of Variance (ANOVA) followed by the Bonferroni test as a post hoc test. The data were analyzed using the GraphPad Prism 6.0 software (San Diego, CA, USA), the experimental groups were compared with the control group and with each other. Pearson's correlations were performed using the IBM SPSS Statistics 23 statistical program. P < 0.05 was defined as statistically significant.

Results

After applying the questionnaire depicting nutritional aspects, socio-cultural, health and lifestyle information for each patient, the investigated population was characterized (Table 1). The patients were aged 36–53 years old, mostly female, brown colored people, married and lower education level. Most participants were not exposed to potentially mutagenic chemicals, such as pesticides, cleaning materials, dyes and solvents. However, it was observed that 64% of patients with G + OME reported exposure to cleaning products and 44% to pesticides. Patients reported no regular physical exercises, as well low alcohol consumption, smoking, meat and vegetables consumption. In addition, more than 50% of patients with gastritis, including those with H. pylori coinfection, reported family history of cancer and absence of other hereditary diseases.

Table 1 Sociocultural and health characteristics of patients with gastritis and on omeprazole (OME) therapy at Getúlio Vargas Hospital

Regarding the mutagenic evaluation of patients stomach epithelium cells (antrum and body), clastogenic and/or aneugenic effects were observed through MN formation in WG + OME (3.62 ± 1.81) and gastritis (3, 00 ± 1.74), in relation to patients that didn’t have gastritis and were not OME user (1.62 ± 0.83). Patients with G + OME (5.091 ± 1.71) also had more MN than those with gastritis and no OME use. The presence of H. pylori in patients with gastritis (5.09 ± 1.71) also contributed to these effects, which in OME therapy presented increased MN formation (8.22 ± 1.74), in relation to all study groups (Fig. 1). There were no significant differences between patients with gastritis in relation to those without gastritis and in prolonged OME use, as well as among those with G + HP in relation to those with G + OME.

Fig. 1
figure 1

Clastogenic and/or aneugenic effects involved in micronuclei formation in 2000 stomach epithelial cells. A MN, B binucleated, WG (Without Gastritis, n = 32); (Without Gastritis + OME, n = 24), G (Gastritis-n = 26); G + OME (Gastritis + OME, n = 26), G + HP (Gastritis + H. pylori, n = 22); WG + OME G + HP + OME (Gastritis + H. pylori + OME, n = 22). Values represent the mean ± S.D.M. Differences between groups were determined by Analysis of Variance (ANOVA), and Bonferroni’s test (post hoc test). Significances were observed for < 0.05 and p < 0.0001, a in relation to WG, b in relation to SG + OME, c in relation to G, d in relation to G + OME and e in relation to G + HP

OME therapy was able to induce other nuclear alterations such as buds and nucleoplasmic bridges on the stomach epithelium (body and antrum) (Fig. 2). Increased number of buds were observed in patients without gastritis (11.67 ± 4.26) and with gastritis (12.08 ± 4.26) when compared with patients without gastritis and OME use (1.78 ± 1, 09). The presence of H. pylori in patients with gastritis (3.69 ± 2.80) did not increase buds in comparison to those with gastritis without OME use (2.13 ± 1.80), but when in therapy with OME (13.73 ± 2.25), an increase of buds was detected, indicating effects on expression and genes (Fig. 2A).

Fig. 2
figure 2

Buds (A) and Nucleoplasmic bridges (B) formation in 2000 oral epithelial cells. WG (Without Gastritis, n = 32); (Without Gastritis + OME, n = 24), G (Gastritis-n = 26); G + OME (Gastritis + OME, n = 26), G + HP (Gastritis + H. pylori, n = 22); WG + OME G + HP + OME (Gastritis + H. pylori + OME, n = 22). Values represent the mean ± S.E.M. Differences between groups were determined by Analysis of Variance (ANOVA), and Bonferroni’s test (post hoc test). Significances were observed for < 0.05 and p < 0.0001, a, b, c, d, and e in relation to WG, SG + OME, G, and G + OME, and e G + HP, respectively

In relation to nucleoplasmic bridges, OME induced an increase in patients without gastritis (10, 55 ± 2.52) and with gastritis (12.69 ± 2.96) when compared to the group without gastritis and OME use (2.03 ± 1.42). In patients with gastritis (3.73 ± 1.82) and gastritis and H. pylori (3.65 ± 2.01), it was not seen an increase in nucleoplasmic bridges in relation to the group without gastritis and no OME use. However, in patients with H. pylori and OME use (13.59 ± 3.99) increased nuclear abnormalities were detected (Fig. 2B).

The OME use and/or therapy induced cytotoxic effects by increasing binucleated cells in patients without gastritis and in OME user without medical prescription (5.44 ± 2.14). In addition, cytotoxicicity was also observed in patients with gastritis (7.11 ± 3.97), as well as in patients with gastritis and positive for H. pylori (20, 41 ± 3.15), when compared to patients without gastritis and no OME use (2.33 ± 0.91). Moreover, similar results were found in patients with gastritis, H. pylori and no OME use (5.01 ± 1.27). Likewise, in patients with gastritis with/without OME use (4.23 ± 1.25), an increase in binucleated cells was observed (Fig. 3A).

Fig. 3
figure 3

Cytogenetic changes indicative of cytotoxicity in the stomach epithelium of patients under omeprazole (OME) therapy by the formation of A binucleated cells and B pyknosis. WG (Without Gastritis, n = 32); WG + OME (Without Gastritis + OME, n = 24), G (Gastritis-n = 26); G + OME (Gastritis + OME, n = 26), G + HP (Gastritis + H. pylori, n = 22); G + HP + OME (Gastritis + H. pylori + OME, n = 22). Values represent the mean ± S.D.M. The differences between the groups were determined by Analysis of Variance (ANOVA), followed by the Bonferroni test (post hoc test). Significances were observed for p < 0.0001, a, b, c, d, and e in relatated to SG, SG + OME, G, d G + OME, and G + HP, respectively

OME also induced cytotoxic effects by pycnotic cells formationin patients without gastritis and OME use (13.96 ± 5.17) and in patients with gastritis and OME use (18.46 ± 4.32), as well as in patients that are not using OME, but positive for H. pylori (7.84 ± 3.16) and in H. pylori positive patients and OME therapy (23, 36 ± 8.72), when compared to patients without gastritis and are not using OME (1, 61 ± 0.61) (Fig. 3B).

Similar to observed for binucleated and pycnotic cells, OME induced apoptotic effects by nuclear fragmentation (karyorrhexis) in stomach epithelium cells of patients without gastritis (202.20 ± 69.65), and with gastritis and no H. pylori (232, 60 ± 93.63) or with H. pylori (209, 40 ± 78.06), when compared to patients without gastritis and no OME use (150.00 ± 49.02). The presence of H. pylori in patients with gastritis and on OME therapy (291, 10 ± 70.20) also induced an increase in cariorrexis (Fig. 4A). Apoptotic OME effects by nuclear dissolution (karyolysis) were similarly observed in stomach epithelial cells of patients without gastritis (366.30 ± 108.60), with gastritis in OME use (359.00 ± 120.20) and with gastritis, H. pylori and OME use (397.80 ± 140.50) in relation to patients without gastritis and no OME use (149.90 ± 46.32). However, these effects were also observed in patients with gastritis (324.20 ± 179.40) and with gastritis, H. pylori (258.00 ± 122.70) and no OME therapy (Fig. 4B). A photomicrographic profile with nuclear and mutagenic changes can be seen in Fig. 5, emphasizing mainly the groups treated with omeprazole.

Fig. 4
figure 4

Cytogenetic changes indicative of apoptosis in the stomach epithelium of patients undergoing Omeprazole therapy (OME) by the formation of A karyorexis and B karyolysis. WG (Without Gastritis, n = 32); WG + OME (Without Gastritis + OME, n = 24), G (Gastritis-n = 26); G + OME (Gastritis + OME, n = 26), G + HP (Gastritis + H. pylori, n = 22); G + HP + OME (Gastritis + H. pylori + OME, n = 22). Values represent the mean ± S.D.M. Differences between groups were determined by Analysis of Variance (ANOVA, followed by the Bonferroni test (post hoc test). Significances for p < 0.0001. ae in relation to WG, WG + OME, G, G + OME, and G + HP, respectively

Fig. 5
figure 5

Photomicrographic profile (light microscopy-1000 × magnification, Giemsa staining) of cytogenetic analyzes of cells obtained from stomach biopsies of patients with gastritis (1–3); with gastritis treated with omeprazole (4–6); with gastritis and H. pylori on omeprazole treatment (7–9). In slides (1–3) intact chromatin (IC) is observed, but with some nuclear alterations. In slides (4–9) greater amounts of cell death and cytogenetic alterations are observed. Legend: Micronuclei (MN); bridges (B); karyorrhexis (C); karyolysis (CA); Buds (BU); binucleated (BN)

In the study, it was observed that OME induces changes in dosages of antioxidant enzymes such as catalase and superoxide dismutase (Fig. 6). In patients without and with gastritis under OME therapy or were in therapy, as well as in patients with gastritis without/withH. pylori, they presented catalase increases in relation to patients without gastritis and were not taking OME (Fig. 6A). However, patients with gastritis and H. pylori on OME therapy showed significant increase in catalase compared to the other groups. The data for superoxide dismutase were similar to those observed for catalase (Fig. 6B).

Fig. 6
figure 6

Changes in antioxidant enzyme dosages during omeprazole therapy (OME-40 mg) in patients with negative and positive gastritis. A Catalase and B superoxide dismutase. WG (Without Gastritis, n = 46); (Without Gastritis + OME, n = 22), G (Gastritis-n = 48); G + OME (Gastritis + OME, n = 48), G + HP (Gastritis + H. pylori, n = 27); WG + OME G + HP + OME (Gastritis + H. pylori + OME, n = 27). Values represent the mean ± S.D.M. Differences between groups were determined by Analysis of Variance (ANOVA, followed by the Bonferroni test (post hoc test). Significances were observed for p < 0.05 and p < 0.0001, a, b, c, d, and e in relation to WG, G, G + HP, WG + OME, and G + OME, respectively

In patients with gastritis and H. pylori on OME therapy, positive correlations were observed between micronucleus induction with catalase and superoxide dismutase measurements, and negative correlation for picnoses induction (Fig. 7).

Fig. 7
figure 7

Pearson’s statistical correlations between cytogenetic biomarkers and antioxidant enzymes for micronuclei and catalase (A); micronuclei and superoxide dismutase (B); pycnosis and catalase (C) and D pycnosis and superoxide dismutases (D). WG (Without Gastritis, n = 46); (Without Gastritis + OME, n = 22), G (Gastritis-n = 48); G + OME (Gastritis + OME, n = 48), G + HP (Gastritis + H. pylori, n = 27); WG + OME G + HP + OME (Gastritis + H. pylori + OME, n = 27)

Discussion

In the present study, it was observed that long term OMEuse and/or therapy, regardless dose specifications (20, 30 and 40 mg/kg) can cause cytogenetic changes in stomach epithelial cells by aneugenic and/or clastogenic effects through micronuclei formation. The OME effects on micronuclei formation in human cells are still unwell described. However, there are in silico studies suggesting that OME can lead to chromosomal changes, as well as contribute to micronuclei formation [85], also promoting DNA covalent bonds, characterizing its genotoxicity [78].

Investigations in gastric epithelial cells of patients with gastritis and with positive H. pylori infection, indicate a risk of genotoxicity, with greater significance in relation to negative patients [62], as well as was observed in the study in relation to the formation of micronuclei, in epithelial cells of the stomach. H. pylori infections can cause chronic gastritis, peptic and duodenal ulcers, adenocarcinoma and gastric lymphoma [99]. There are reports that OME can induce DNA damage, after its metabolism by N-nitrosamines formation, generating several nuclear alterations such as MN, pycnosis and cariorrexis [69, 96]. MN can also be induced by chromosome breaks [84], internal chromosomes that have been separated from the nucleus [10], double-stranded DNA breaks or as a result of mitotic spindle dysfunction [34, 35]. It is worthynote that DNA breaks-induced apoptosis can also happen due to oxidative stress [32, 79].

According to Amieva and Peek [4], there are two bacterial factors that make it a risk factor for gastric cancer and peptide ulcer: (1) the oncoprotein CagA that stimulates cell proliferation by mitotic signaling; (2) the cytotoxin VacA that alters membrane permeability and causes mitochondrial injury-induced apoptosis. In addition, other virulence mechanisms are pointed out, such as motility (flagella), chemotaxis, urease production (pH neutralization) and adaptive mechanisms such as inflammation activation, immune suppression, E-cadherin cleavage and host cell cholesterol modification [7].

According to Raza et al. [83], the H. pylori virulence is determined by the presence of the cytotoxin associated with the CagA gene, VacA cytotoxin and SabA adhesion proteins. The VacA cytotoxin induces membrane channel formation, cytochrome C release and modulates antigen presentation [39]. SabA acts as a chemical gradient (bicarbonate/CO2 or urea/ammonia) that guides the bacteria to bind the mucosa [76]. SabA expression is associated with intestinal metaplasia development, gastric atrophy and cancer [3, 54, 102].

DNA damage can be induced by OME through its secondary metabolites sulfone, sulfite and hydroxy-omeprazole [28, 85], as also for its electrophilic potential [82] due to covalent DNA bonds [18, 38]. It is observed that OME can increase nuclear cell proliferation antigens (PCNA) [57, 109], by modulating lysosomal transport, with mechanisms of LC3 gene expression associated to autophagy [101].

In addition to micronuclei formation, other cytogenetic cahnges were observed in the stomach epithelial cells of patients with/without gastritis (with/without H. pylori infection, in and/or OME therapy), such as nucleoplasmic buds and bridges. Tthe buds are the result of DNA amplification or repair [35, 60] and the bridges are originated from failures in chromosomal rearrangements or are result from chromosomal ends fusion, telomeres that allow chromatin filaments formation of that link two distinct nuclei [35]. Corroborating the findings in this study, previous reports show that OME can induce changes in chromosomes and micronucleus formation [18, 38, 85].

The OME cytotoxicity has been reported in normal human cells (HEK293 and NIH3T3) [91]. In stomach epithelial cells of patients in OM|Euse or in therapy, cytogenetic changes are indicative of cytotoxicity, due to picnoses and binucleated cells formation, especially in patients with H. pylori infection. Pyknosis occur due to chromatin condensation and dissolution, and binucleated cells result from cytokinesis failures at the end of cell division [86]. H. pylori releases cytotoxins that can induce apoptosis by alterations in cytochrome C release [39], as well as destroy cellular junctions in the gastric epithelium [3], and promote transient increased acid secretion that lead to hypochlorhydria and intestinal metaplasia [99]. These events are linked to increased grastric cancer risk [51, 63, 66, 89, 90, 105].

Moreover, OME has hepatotoxic effects as a result of apoptosis stimulated by tumor necrosis factor alpha (TNF-α), as well as by alterations of liver enzymes AST and ALT [22, 37, 98]. There is also reports of nephrotoxic effects, thrombocytopenia, acute interstitial nephritis, anaphylactic reactions and gynecomastia [22, 61].

In this study, OME induced apoptosis in stomach epithelial cells due to nuclear fragmentation (cariorrexis) and nuclear dissolution (karyolysis), as seen in patients without gastritis and with gastritis in OME use and/or therapy, and also in patients with H. pylori infection, as previously observed in other nuclear alterations. Although apoptosisis one of the mechanisms associated to acute gastric injury [59], OME has apoptotic effects in human gastric cancer cells (HGC-27) [108], colorectal tumor cells [52, 67], and normal human nuclear polymorphic leukocytes [20, 68, 72].

According to previous studies, drugs that induce oxidative damage may increase the levels of endogenous antioxidant enzymes such as catalase and superoxide dismutase [2, 47, 80]. Catalase (CAT) is one of the antioxidant enzymes that participates in H2O2 degradation through dismutation reactions, mainly in peroxisomes, and it has been considered as an important oxidative biomarker [45, 77]. Superoxide dismutase (SOD) converts the oxygen produced during oxidative stress to H2O2. In this regard, to act effectively in maintaining cellular integrity and function, SOD depends on the balance between SOD, GPx and CAT [74, 77].

In this study it was possible to detect an increase in antioxidant defenses for these enzymes, especially in patients with H. pylori infection. H. pylori infection can also increase reactive oxygen and nitrogen species in the stomach [42]. Moreover, gastric lesions can induce oxidative stress, with amplification by OME therapy [53] independently of co-infection with H. pylori, and also increase antioxidant enzymes such as SOD and CAT, and glutathione reductase (GSH) [9, 41]. Drugs may contribute to increase oxidative stress levels [2, 47, 81], due to an imbalance between antioxidant defenses and oxidative stress levels [44] and regulation of lipid peroxidation [104].

OME is one of the drugs that can induce oxidative stress [53], which culminates in cell apoptosis [77, 92, 106]. Free radicals induce gastric lesions [93], and contribute to carcinogenesis [100]. Gastric lesions can produce free radicals, which are controled by SOD and GPx enzymes, which lead to tissue recovery and gastroprotection [24, 25]. OME can also induce lipid peroxidation, and as a cellular response, increased activity of catalase and superoxide dismutase is observed, which make them, important oxidative stress markers [24].

During metabolism, OME can generate sulfone, sulfite and hydroxy-omeprazole, compounds that can generate more oxidative damage [14, 15, 28]. OME increase heme-oxigenease enzyme independently of the aryl hydrocarbon receptor (AhR), which consequently increases peroxide levels [73]. Oxidative damage can be one of OME mechanisms for inducing DNA changes in gastric epithelium cells, as it can produce H2O2 when it binds to protein C283, which contains CACT and C136 for generating beta oxidation of fatty acids [97]. OME can induce oxidative damage in S. cerevisiae, in addition to cytogenetic damage in murine Sarcoma 180 cells [50, 75].

Among these mechanisms, our study points out that cytogenetic changes can be induced by oxidative effects that lead to micronuclei formation and other nuclear alterations indicative of cytotoxicity. Corroborating these analyzes, positive and negative statistical correlations were observed between micronuclei and CAT/SOD concentrations, and between pyknosis, respectively. Proton pump inhibiting drugs (PPIs), such as OME, may have genotoxic and/or carcinogenic effects [28] through several mechanisms including oxidative stress. When substances that induce oxidative stress are in excess and the antioxidant system is unable to neutralize the oxidative process [56], several mechanisms can induce cellular regulation and activation of cell death signaling cascades (apoptosis or necrosis) [106], or, conversely, induce cell proliferation, metastasis, resistance to apoptosis and angiogenesis as a consequence of genetic instability (Fig. 8) [12, 19, 65].

Fig. 8
figure 8

Long-term omeprazole use possible induces mutagenic, apoptoses and necroses effects through oxidative stress induction that can lead to micronuclei formation andother nuclear alterations indicative of cytotoxicity and apoptosis; several cellular mechanisms induce cell regulation and activation of signaling cascades for cell death (apoptosis or necrosis), or, conversely, induce cell proliferation, metastasis, resistance to apoptosis and angiogenesis as a consequence of genetic instability. (ATP-adenosine triphosphate; CAT-catalase; IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; NF-κB-nuclear factor kappa-light-chain-enhancer of activated B cells; OS-oxidative stress; SOD-superoxide dismutase)

Conclusions

In this study, in stomach epithelial cells of patients without gastritis and with gastritis, especially those with H. pylori infection, and OME use and/or therapy, it was possible to point out that OME induces cytogenetic changes due to (1) clastogenic and/or aneugenic effects that induce micronuclei formation; (2) altered gene expression, chromosomal rearrangements and fusion of chromosomal ends; (3) cytotoxicicity by increased picnoses and binucleated cells and (4) apoptosis by increasing karyorexis and karyolysis. Several mechanisms, not yet elucidated, can be attributed to these OME cytogenetic effects, but oxidative effects can also be involved, as observed by increased concentrations of endogenous antioxidant enzymes such as catalse and superoxide dismutase, which have also been associated to increased micronuclei and picnoses. These data point out the risks regarding long term OME use/therapy, as well as the monitoring of cytogenetic changes and oxidative damage, as an important strategy for the genetic instability prevention.

Availability of data and materials

Yes.

References

  1. Adeoye GO, Alimba CG, Oyeleke OB. The genotoxicity and systemic toxicity of a pharmaceutical effluent in Wistar rats may involve oxidative stress induction. Toxicol Rep. 2015;2:1265–72. https://doi.org/10.1016/j.toxrep.2015.09.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Almenara CC, Mill JG, Vassallo DV, Baldo MP, Padilha AS. In vitro fructose exposure overactivates NADPH oxidase and causes oxidative stress in the isolated rat aorta. Toxicol In Vitro. 2015;29:2030–7. https://doi.org/10.1016/j.tiv.2015.08.013.

    Article  CAS  PubMed  Google Scholar 

  3. Alzahrani S, Lina TT, Gonzalez J, Pinchuk IV, Beswick EJ, Reyes VE. Effect of Helicobacter pylori on gastric epithelial cells. World J Gastroenterol. 2014;20:12767–80. https://doi.org/10.3748/wjg.v20.i36.12767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Amieva M, Peek RM. Pathobiology of Helicobacter pylori–induced gastric cancer. Gastroenterology. 2016;150:64–78. https://doi.org/10.1053/j.gastro.2015.09.004.

    Article  CAS  PubMed  Google Scholar 

  5. Arthur JR, Boyne R. Superoxide dismutase and glutathione peroxidase activities in neutrophils from selenium deficient and copper deficient cattle. Life Sci. 1985;36:1569–75. https://doi.org/10.1016/0024-3205(85)90381-9.

    Article  CAS  PubMed  Google Scholar 

  6. Attia TZ, Yamashita T, Tsujino H, Derayea SM, Tsutsumi Y, Uno T. Effect of drug combination on omeprazole metabolism by cytochrome P450 2C19 in Helicobacter pylori eradication therapy. Chem Pharm Bull. 2019;67(8):810–5. https://doi.org/10.1248/cpb.c19-00084.

    Article  CAS  Google Scholar 

  7. Backert S, Tegtmeyer N. Type IV secretion and signal transduction of Helicobacter pylori CagA through interactions with host cell receptors. Toxins. 2017;9:115. https://doi.org/10.3390/toxins9040115.

    Article  CAS  PubMed Central  Google Scholar 

  8. Bhatia A, Kumar Y. Cancer cell micronucleus: an update on clinical and diagnostic applications. Apmis. 2013;121:569–81. https://doi.org/10.1111/apm.12033.

    Article  PubMed  Google Scholar 

  9. Baldissera FG, Cruzat VF. Estresse oxidativo, qualidade de vida e exercício uma abordagem nutricional. Brazil: Atenção Integral à Saúde; 2014.

    Google Scholar 

  10. Balmus G, Karp NA, Ng BL, Jackson SP, Adams DJ, McIntyre RE. A high-throughput in vivo micronucleus assay for genome instability screening in mice. Nat Protoc. 2015;10(1):205–15. https://doi.org/10.1038/nprot.2015.010.

    Article  PubMed  Google Scholar 

  11. Banerjee P, Eckert AO, Schrey AK, Preissner R. ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Res. 2018;46:W257–63. https://doi.org/10.1093/nar/gky318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bellone M, Calcinotto A, Filipazzi P, De Milito A, Fais S, Rivoltini L. The acidity of the tumor microenvironment is a mechanism of immune escape that can be overcome by proton pump inhibitors. Oncoimmunology. 2013;2(1):e22058. https://doi.org/10.4161/onci.22058.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Braga AL, de Meneses AAPM, de Oliveira Santos JV, Dos Reis AC, de Lima RMT, da Mata AMOF, et al. Toxicogenetic study of omeprazole and the modulatory effects of retinol palmitate and ascorbic acid on Allium cepa. Chemosphere. 2018;204:220–6. https://doi.org/10.1016/j.chemosphere.2018.04.021.

    Article  CAS  PubMed  Google Scholar 

  14. Brambilla G, Martelli A. Genotoxicity and carcinogenicity studies of analgesics, anti-inflammatory drugs and antipyretics. Pharmacol Res. 2009;60(1):1–17. https://doi.org/10.1016/j.phrs.2009.03.007.

    Article  CAS  PubMed  Google Scholar 

  15. Brambilla G, Mattioli F, Martelli A. Genotoxic and carcinogenic effects of gastrointestinal drugs. Mutagenesis. 2010;25(4):315–26. https://doi.org/10.1093/mutage/geq025.

    Article  CAS  PubMed  Google Scholar 

  16. Brzozowski T, Ptak-Belowska A, Kwiecien S, Krzysiek-Maczka G, Strzalka M, Drozdowicz D, et al. Novel concept in the mechanism of injury and protection of gastric mucosa: role of renin-angiotensin system and active metabolites of angiotensin. Curr Med Chem. 2012;19(1):55–62. https://doi.org/10.2174/092986712803413953.

    Article  CAS  PubMed  Google Scholar 

  17. Brzozowski T, Konturek PC, Pajdo R, Ptak-Belowska A, Kwiecien S, Pawlik M, et al. Physiological mediators in nonsteroidal anti-inflammatory drugs (NSAIDs)-induced impairment of gastric mucosal defense and adaptation. Focus on nitric oxide and lipoxins. J Physiol Pharmacol. 2008;59:89–102.

    PubMed  Google Scholar 

  18. Burlinson B, Morriss SH, Gatehouse DG, Tweats DJ, Ekman L, Bolcsfoldi G, et al. Genotoxicity studies of gastric acid inhibiting drugs. Lancet. 1990;335(8686):419–20. https://doi.org/10.1016/0140-6736(90)90260-C.

    Article  CAS  PubMed  Google Scholar 

  19. Canitano A, Iessi E, Spugnini EP, Federici C, Fais S. Proton pump inhibitors induce a caspase-independent antitumor effect against human multiple myeloma. Cancer Lett. 2016;376(2):278–83. https://doi.org/10.1016/j.canlet.2016.04.015.

    Article  CAS  PubMed  Google Scholar 

  20. Capodicasa E, Cornacchione P, Natalini B, Bartoli A, Coaccioli S, Marconi P, Scaringi L. Omeprazole induces apoptosis in normal human polymorphonuclear leucocytes. Int J Immunopathol Pharmacol. 2008;21(1):73–85. https://doi.org/10.1177/039463200802100109.

    Article  CAS  PubMed  Google Scholar 

  21. Carrano AV, Natarajan AT. Considerations for population monitoring using cytogenetic techniques. Mutat Res. 1988;204(3):379–406. https://doi.org/10.1016/0165-1218(88)90036-5.

    Article  CAS  PubMed  Google Scholar 

  22. Cesário MR, Barros BS, Courson C, Melo DM, Kiennemann A. Catalytic performances of Ni–CaO–mayenite in CO2 sorption enhanced steam methane reforming. Fuel Process Technol. 2015;131:247–53. https://doi.org/10.1016/j.fuproc.2014.11.028.

    Article  CAS  Google Scholar 

  23. Chance B, Maehly AC. [136] Assay of catalases and peroxidases. Meth Enzymol. 1955;2:764–75. https://doi.org/10.1016/s0076-6879(55)02300-8.

    Article  Google Scholar 

  24. Chen C, Arjomandi M, Qin H, Balmes J, Tager I, Holland N. Cytogenetic damage in buccal epithelia and peripheral lymphocytes of young healthy individuals exposed to ozone. Mutagenesis. 2006;21(2):131–7. https://doi.org/10.1093/mutage/gel007.

    Article  CAS  PubMed  Google Scholar 

  25. Cheng YT, Wu SL, Ho CY, Huang SM, Cheng CL, Yen GC. Beneficial effects of camellia oil (Camellia oleifera Abel.) on ketoprofen-induced gastrointestinal mucosal damage through upregulation of HO-1 and VEGF. J Agric Food Chem. 2014;62(3):642–50. https://doi.org/10.1021/jf404614k.

    Article  CAS  PubMed  Google Scholar 

  26. Denisenko NP, Sychev DA, Sizova ZM, Smirnov VV, Ryzhikova KA, Sozaeva ZA, Grishina EA. Urine metabolic ratio of omeprazole in relation to CYP2C19 polymorphisms in Russian peptic ulcer patients. Curr Pharmacogenomics Person Med. 2017;10:253–9. https://doi.org/10.2147/PGPM.S141935.

    Article  CAS  Google Scholar 

  27. Dias N, Santos P, Pinto M, Fernandes C, Júnior A, Souza S, et al. Análise de prontuários de pacientes com gastrite em um hospital na região oeste ii do estado de Goiás. Rev Eletrôn Fac Montes Belos. 2015;8(1):1–9.

    Google Scholar 

  28. Downes N, Foster J. Regulatory forum opinion piece: carcinogen risk assessment: the move from screens to science. Toxicol Pathol. 2015;43(8):1064–73. https://doi.org/10.1177/0192623315598578.

    Article  CAS  PubMed  Google Scholar 

  29. El-Maraghy SA, Rizk SM, Shahin NN. Gastroprotective effect of crocin in ethanol-induced gastric injury in rats. Chem Biol Interact. 2015;229:26–35. https://doi.org/10.1016/j.cbi.2015.01.015.

    Article  CAS  PubMed  Google Scholar 

  30. Esteva F, Iñarrea P, Cornudella R, Lanas A. Omeprazole does not interfere with the antiplatelet effect of low dose aspirin in man. Gastroenterology. 1998;114:A116. https://doi.org/10.1016/s0016-5085(98)80473-3.

    Article  Google Scholar 

  31. Eusebi LH, Rabitti S, Artesiani ML, Gelli D, Montagnani M, Zagari RM, Bazzoli F. Proton pump inhibitors: risks of long-term use. J Gastroenterol Hepatol. 2017;32(7):1295–302. https://doi.org/10.1111/jgh.13737.

    Article  PubMed  Google Scholar 

  32. Fang HM, Wang JJ, Zhang LJ, Xu JM, Mei Q, Lu CX. Omeprazole and rabeprazole azole effect on rat intestinal P-glycoprotein. Chin J Hosp Pharm. 2013;33:7–11.

    Google Scholar 

  33. Faria LEM, Braga JRM. Aplicação do teste de micronúcleo para avaliação de potencial genotóxico em epitélio oral de estudantes universitários. Rev Eletrôn Atualiza Saúde. 2015;1(1):35–41.

    Google Scholar 

  34. Fenech M. Cytokinesis-block micronucleus cytome assay. Nat Protoc. 2007;2(5):1084. https://doi.org/10.1038/nprot.2007.77.

    Article  CAS  PubMed  Google Scholar 

  35. Fenech M, Kirsch-Volders M, Natarajan AT, Surralles J, Crott JW, Parry J, et al. Molecular mechanisms of micronucleus, nucleoplasmic bridge and nuclear bud formation in mammalian and human cells. Mutagenesis. 2011;26(1):125–32. https://doi.org/10.1093/mutage/geq052.

    Article  CAS  PubMed  Google Scholar 

  36. Fenech M, Knasmueller S, Bolognesi C, Bonassi S, Holland N, Migliore L, et al. Molecular mechanisms by which in vivo exposure to exogenous chemical genotoxic agents can lead to micronucleus formation in lymphocytes in vivo and ex vivo in humans. Mutat Res Rev Mutat Res. 2016;770:12–25. https://doi.org/10.1016/j.mrrev.2016.04.008.

    Article  CAS  PubMed  Google Scholar 

  37. Fontana RJ, Hayashi PH, Gu J, Reddy KR, Barnhart H, Watkins PB, et al. Idiosyncratic drug-induced liver injury is associated with substantial morbidity and mortality within 6 months from onset. Gastroenterol. 2014;147(1):96–108. https://doi.org/10.1053/j.gastro.2014.03.045.

    Article  CAS  Google Scholar 

  38. Furihata C, Hirose K, Matsushima T. Genotoxicity and cell proliferative activity of omeprazole in rat stomach mucosa. Mutat Res Lett. 1991;262(1):73–6. https://doi.org/10.1016/0165-7992(91)90109-H.

    Article  CAS  Google Scholar 

  39. Gajewski A, Mnich E, Szymański K, Hinc K, Obuchowski M, Moran AP, Chmiela M. Helicobacter pylori antigens, acetylsalicylic acid, LDL and 7-ketocholesterol-their potential role in destabilizing the gastric epithelial cell barrier. An in vitro model of Kato III cells. Acta Biochim Pol. 2016;63(1):145–52. https://doi.org/10.18388/abp.2015_1122.

    Article  CAS  PubMed  Google Scholar 

  40. Gesmundo I, Di Blasio L, Banfi D, Villanova T, Fanciulli A, Favaro E, et al. Proton pump inhibitors promote the growth of androgen-sensitive prostate cancer cells through ErbB2, ERK1/2, PI3K/Akt, GSK-3β signaling and inhibition of cellular prostatic acid phosphatase. Cancer lett. 2019;449:252–62. https://doi.org/10.1016/j.canlet.2019.02.028.

    Article  CAS  PubMed  Google Scholar 

  41. Glorieux C, Calderon PB. Catalase, a remarkable enzyme: targeting the oldest antioxidant enzyme to find a new cancer treatment approach. Biol Chem. 2017;398(10):1095–108.

    Article  CAS  PubMed  Google Scholar 

  42. Golbabapour S, Hajrezaie M, Hassandarvish P, Abdul Majid N, Hadi AHA, Nordin N, Abdulla MA. Acute toxicity and gastroprotective role of M. pruriens in ethanol-induced gastric mucosal injuries in rats. Biomed Res Int. 2013. https://doi.org/10.1155/2013/974185.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Graham DY. History of Helicobacter pylori, duodenal ulcer, gastric ulcer and gastric cancer. World J Gastroenterol. 2014;20(18):5191–204. https://doi.org/10.3748/wjg.v20.i18.5191.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Gunasekarana V, Raj GV, Chand P. A comprehensive review on clinical applications of comet assay. J Clin Diagn Res. 2015;9(3):GE01–5. https://doi.org/10.7860/jcdr/2015/12062.5622.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Gupta DK, Inouhe M, Rodríguez-Serrano M, Romero-Puertas MC, Sandalio LM. Oxidative stress and arsenic toxicity: role of NADPH oxidases. Chemosphere. 2013;90(6):1987–96. https://doi.org/10.1016/j.chemosphere.2012.10.066.

    Article  CAS  PubMed  Google Scholar 

  46. Haastrup PF, Thompson W, Søndergaard J, Jarbøl DE. Side effects of long-term proton pump inhibitor use: a review. Basic Clin Pharmacol Toxicol. 2018;123(2):114–21. https://doi.org/10.1111/bcpt.13023.

    Article  CAS  PubMed  Google Scholar 

  47. Herbet M, Izdebska M, Piątkowska-Chmiel I, Poleszak E, Jagiełło-Wójtowicz E. Estimation of oxidative stress parameters in rats after simultaneous administration of rosuvastatin with antidepressants. Pharmacol Rep. 2016;68(1):172–6. https://doi.org/10.1016/j.pharep.2015.08.004.

    Article  CAS  PubMed  Google Scholar 

  48. Holland N, Bolognesi C, Kirsch-Volders M, Bonassi S, Zeiger E, Knasmueller S, Fenech M. The micronucleus assay in human buccal cells as a tool for biomonitoring DNA damage: the HUMN project perspective on current status and knowledge gaps. Mutat Res Rev Mutat Res. 2008;659(1–2):93–108. https://doi.org/10.1016/j.mrrev.2008.03.007.

    Article  CAS  Google Scholar 

  49. Hong F, Wu N, Zhou Y, Ji L, Chen T, Wang L. Gastric toxicity involving alterations of gastritis-related protein expression in mice following long-term exposure to nano TiO2. Food Res Int. 2017;95:38–45. https://doi.org/10.1016/j.foodres.2017.02.013.

    Article  CAS  PubMed  Google Scholar 

  50. Ishiguro T, Ishiguro M, Ishiguro R, Iwai S. Cotreatment with dichloroacetate and omeprazole exhibits a synergistic antiproliferative effect on malignant tumors. Oncology Lett. 2012;3(3):726–8. https://doi.org/10.3892/ol.2012.552.

    Article  CAS  Google Scholar 

  51. Keilberg D, Ottemann KM. How Helicobacter pylori senses, targets and interacts with the gastric epithelium. Environ Microbiol. 2016;18(3):791–806. https://doi.org/10.1111/1462-2920.13222.

    Article  PubMed  Google Scholar 

  52. Kim YJ, Lee JS, Hong KS, Chung JW, Kim JH, Hahm KB. Novel application of proton pump inhibitor for the prevention of colitis-induced colorectal carcinogenesis beyond acid suppression. Cancer Prev Res. 2010;3(8):963–74. https://doi.org/10.1158/1940-6207.CAPR-10-0033.

    Article  CAS  Google Scholar 

  53. Kohler JE, Blass AL, Liu J, Tai K, Soybel DI. Antioxidant pre-treatment prevents omeprazole-induced toxicity in an in vitro model of infectious gastritis. Free Radic Biol Med. 2010;49(5):786–91. https://doi.org/10.1016/j.freeradbiomed.2010.05.034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kondratyev AD, Chung KN, Jung MO. Identification and characterization of a radiation-inducible glycosylated human early-response gene. Cancer res. 1996;56(7):1498–502.

    CAS  PubMed  Google Scholar 

  55. Lahner E, Galli G, Esposito G, Pilozzi E, Corleto VD, Annibale B. Updated features associated with type 1 gastric carcinoids patients: a single-center study. Scand J Gastroenterol. 2014;49(12):1447–55. https://doi.org/10.3109/00365521.2014.968859.

    Article  PubMed  Google Scholar 

  56. Laskoski LM, Dittrich RL, Valadão CAA, Brum JS, Brandão Y, Brito HFV, de Sousa RS. Oxidative stress in hoof laminar tissue of horses with lethal gastrointestinal diseases. Vet Immunol Immunopathol. 2016;171:66–72. https://doi.org/10.1016/j.vetimm.2016.02.008.

    Article  CAS  PubMed  Google Scholar 

  57. Liu X, He Y, Li F, Huang Q, Kato TA, Hall RP, Li CY. Redefinindo os papéis dos fatores apoptóticos na carcinogênese. Oncol Mol Cell. 2016;3:e1054550.

    Article  Google Scholar 

  58. Lowry OH, Rosebrough NJ, Farr AL, Randal RJ. Protein measurement with 665 Follin phenol reagent. J Biol Chem. 1951;193:265–75.

    Article  CAS  PubMed  Google Scholar 

  59. Lou DI, Hussmann JA, McBee RM, Acevedo A, Andino R, Press WH, Sawyer SL. High-throughput DNA sequencing errors are reduced by orders of magnitude using circle sequencing. Proc Natl Acad Sci. 2013;110(49):19872–7. https://doi.org/10.1073/pnas.1319590110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Luzhna L, Kathiria P, Kovalchuk O. Micronuclei in genotoxicity assessment: from genetics to epigenetics and beyond. Front Genet. 2013;4:131. https://doi.org/10.3389/fgene.2013.00131.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Mahmoud-Awny M, Attia AS, Abd-Ellah MF, El-Abhar HS. Mangiferin mitigates gastric ulcer in ischemia/reperfused rats: involvement of PPAR-γ, NF-κB and Nrf2/HO-1 signaling pathways. PLoS ONE. 2015;10(7): e0132497. https://doi.org/10.1371/journal.pone.0132497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Marie MAM, Altahir IE. Relationship between the extent of DNA damage and gastritis in normal and Helicobacter pylori-infected patients. Gut liver. 2011;5(3):315. https://doi.org/10.5009/gnl.2011.5.3.315.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Marino ML, Fais S, Djavaheri-Mergny M, Villa A, Meschini S, Lozupone F, et al. Proton pump inhibition induces autophagy as a survival mechanism following oxidative stress in human melanoma cells. Cell Death Dis. 2010;1(10):e87–e87. https://doi.org/10.1038/cddis.2010.67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Minalyan A, Benhammou JN, Artashesyan A, Lewis MS, Pisegna JR. Autoimmune atrophic gastritis: current perspectives. Clin Exp Gastroenterol. 2017;10:19–27. https://doi.org/10.2147/ceg.s109123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Moloney JN, Cotter TG. ROS signalling in the biology of cancer. Semin Cell Dev Biol. 2018;80:50–64. https://doi.org/10.1016/j.semcdb.2017.05.023.

    Article  CAS  PubMed  Google Scholar 

  66. Morimura T, Fujita K, Akita M, Nagashima M, Satomi A. The proton pump inhibitor inhibits cell growth and induces apoptosis in human hepatoblastoma. Pediatr Surg Int. 2008;24(10):1087–94. https://doi.org/10.1007/s00383-008-2229-2.

    Article  PubMed  Google Scholar 

  67. Müerköster SS, Rausch AV, Isberner A, Minkenberg J, Blaszczuk E, Witt M, et al. The apoptosis-inducing effect of gastrin on colorectal cancer cells relates to an increased IEX-1 expression mediating NF-κ B inhibition. Oncogene. 2008;27(8):1122–34. https://doi.org/10.1038/sj.onc.1210728.

    Article  CAS  Google Scholar 

  68. Müerköster S, Isberner A, Arlt A, Witt M, Reimann B, Blaszczuk E, et al. Gastrin suppresses growth of CCK2 receptor expressing colon cancer cells by inducing apoptosis in vitro and in vivo. Gastroenterology. 2005;129(3):952–68. https://doi.org/10.1053/j.gastro.2005.06.059.

    Article  CAS  PubMed  Google Scholar 

  69. Novotna A, Srovnalova A, Svecarova M, Korhonova M, Bartonkova I, Dvorak Z. Differential effects of omeprazole and lansoprazole enantiomers on aryl hydrocarbon receptor in human hepatocytes and cell lines. PLoS ONE. 2014;9(6): e98711. https://doi.org/10.1371/journal.pone.0098711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Numico G, Fusco V, Franco P, Roila F. Proton pump inhibitors in cancer patients: how useful they are? A review of the most common indications for their use. Crit Rev Oncol Hematol. 2017;111:144–51. https://doi.org/10.1016/j.critrevonc.2017.01.014.

    Article  PubMed  Google Scholar 

  71. Park S, Hyun YJ, Kim YR, Lee JH, Ryu S, Kim JM, et al. Effects of CYP2C19 genetic polymorphisms on PK/PD responses of omeprazole in Korean healthy volunteers. J Korean Med Sci. 2017;32(5):729–36. https://doi.org/10.3346/jkms.2017.32.5.729.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Patel A, Zhang S, Shrestha AK, Maturu P, Moorthy B, Shivanna B. Omeprazole induces heme oxygenase-1 in fetal human pulmonary microvascular endothelial cells via hydrogen peroxide-independent Nrf2 signaling pathway. Toxicol Appl Pharmacol. 2016;311:26–33. https://doi.org/10.1016/j.taap.2016.10.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Patel A, Zhang S, Moorthy B, Shivanna B. Omeprazole does not potentiate acute oxygen toxicity in fetal human pulmonary microvascular endothelial cells exposed to hyperoxia. Pharm Anal Acta. 2015;6:424. https://doi.org/10.4172/2153-2435.1000424.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Patlolla JM, Zhang Y, Li Q, Steele VE, Rao CV. Anti-carcinogenic properties of omeprazole against human colon cancer cells and azoxymethane-induced colonic aberrant crypt foci formation in rats. Int J Oncol. 2012;40(1):170–5. https://doi.org/10.3892/ijo.2011.1214.

    Article  CAS  PubMed  Google Scholar 

  75. Paz MFCJ, Braga AL, de Meneses AAPM, de Carvalho RM, de Aguiar RPS, Cronemberger LAC, et al. Ascorbic acid and retinol palmitate modulatory effect on omeprazole-induced oxidative damage, and the cytogenetic changes in S. cerevisiae and S180 cells. Chem Biol Interact. 2019;311:108776. https://doi.org/10.1016/j.cbi.2019.108776.

    Article  CAS  PubMed  Google Scholar 

  76. Peterson AJ, Menheniott TR, O'Connor L, Walduck AK, Fox JG, Kawakami K, Minamoto P, Ong EK, Wang TC, Judd LM, Giraud AS. Helicobacter pylori infection promotes methylation and silencing of trefoil factor 2, leading to gastric tumor development in mice and humans. Gastroenterology. 2010;139:2005–17. https://doi.org/10.1053/j.gastro.2010.08.043.

    Article  CAS  PubMed  Google Scholar 

  77. Pey A, Zamoum T, Christen R, Merle PL, Furla P. Characterization of glutathione peroxidase diversity in the symbiotic sea anemone Anemonia viridis. Biochimie. 2017;132:94–101. https://doi.org/10.1016/j.biochi.2016.10.016.

    Article  CAS  PubMed  Google Scholar 

  78. Phillips DH, Hewer A, Osborne MR. Interaction of omeprazole with DNA in rat tissues. Mutagenesis. 1992;7(4):277–83. https://doi.org/10.1093/mutage/7.4.277.

    Article  CAS  PubMed  Google Scholar 

  79. Pittaluga M, Sgadari A, Dimauro I, Tavazzi B, Parisi P, Caporossi D. Physical exercise and redox balance in type 2 diabetics: effects of moderate training on biomarkers of oxidative stress and DNA damage evaluated through comet assay. Oxid Med Cell Longev. 2015. https://doi.org/10.1155/2015/981242.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Poprac P, Jomova K, Simunkova M, Kollar V, Rhodes CJ, Valko M. Targeting free radicals in oxidative stress-related human diseases. Trends Pharmacol Sci. 2017;38(7):592–607. https://doi.org/10.1016/j.tips.2017.04.005.

    Article  CAS  PubMed  Google Scholar 

  81. Porto ML, Lírio LM, Dias AT, Batista AT, Campagnaro BP, Mill JG, et al. Increased oxidative stress and apoptosis in peripheral blood mononuclear cells of fructose-fed rats. Toxicol In Vitro. 2015;29(8):1977–81. https://doi.org/10.1016/j.tiv.2015.08.006.

    Article  CAS  PubMed  Google Scholar 

  82. Powers RE, Lawton GP, Modlin IM. Genotoxicity, carcinogenicity and acid-suppressing medications. Pharmacol Ther. 1995;65(3):303–17. https://doi.org/10.1016/0163-7258(95)98596-i.

    Article  CAS  PubMed  Google Scholar 

  83. Raza Y, Khan A, Farooqui A, Mubarak M, Facista A, Akhtar SS, Khan S, Kazi JI, Bernstein C, Kazmi SU. Oxidative DNA damage as a potential early biomarker of Helicobacter pylori associated carcinogenesis. Pathol. Oncol. Res. 2014;20:839–46. https://doi.org/10.1007/s12253-014-9762-1.

    Article  CAS  PubMed  Google Scholar 

  84. Rim KT, Kim SJ. A review on mutagenicity testing for hazard classification of chemicals at work: focusing on in vivo micronucleus test for allyl chloride. Saf Health Work. 2015;6(3):184–91. https://doi.org/10.1016/j.shaw.2015.05.005.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Rosenkranz HS, Klopman G. Omeprazole: an exploration of its reported genotoxicity. Mutagenesis. 1991;6(5):381–4. https://doi.org/10.1093/mutage/6.5.381.

    Article  CAS  PubMed  Google Scholar 

  86. Sabharwal R, Verma P, Syed MA, Sharma T, Subudhi SK, Mohanty S, Gupta S. Emergence of micronuclei as a genomic biomarker. Indian J Med Paediatr Oncol. 2015;36(4):212–8. https://doi.org/10.4103/0971-5851.171541.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Sagari SG, Babannavar R, Lohra A, Kodgi A, Bapure S, Rao Y. Micronuclei frequencies and nuclear abnormalities in oral exfoliated cells of nuclear power plant workers. J Clin Diagn Res. 2014;8(12):ZC15–7. https://doi.org/10.7860/JCDR/2014/9059.5240.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Savarino V, Dulbecco P, De Bortoli N, Ottonello A, Savarino E. The appropriate use of proton pump inhibitors (PPIs): need for a reappraisal. Eur J Intern Med. 2017;37:19–24. https://doi.org/10.1016/j.ejim.2016.10.007.

    Article  CAS  PubMed  Google Scholar 

  89. Scaringi L, Cornacchione P, Ayroldi E, Corazzi L, Capodicasa E, Rossi R, Marconi P. Omeprazole induces apoptosis in jurkat cells. Int J Immunopathol Pharmacol. 2004;17(3):331–42.

    Article  CAS  PubMed  Google Scholar 

  90. Seoane M, Esperanza M, Cid Á. Cytotoxic effects of the proton pump inhibitor omeprazole on the non-target marine microalga Tetraselmis suecica. Aquat Toxicol. 2017;191:62–72. https://doi.org/10.1016/j.aquatox.2017.08.001.

    Article  CAS  PubMed  Google Scholar 

  91. Shankar G, Borkar RM, Udutha S, Kanakaraju M, Charan GS, Misra S, Srinivas R. Identification and structural characterization of the stress degradation products of omeprazole using Q-TOF-LC-ESI-MS/MS and NMR experiments: evaluation of the toxicity of the degradation products. New J Chem. 2019;43(19):7294–306. https://doi.org/10.1039/C9NJ00932A.

    Article  CAS  Google Scholar 

  92. Sies H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: oxidative eustress. Redox Biol. 2017;11:613–9. https://doi.org/10.1016/j.redox.2016.12.035.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Sofidiya MO, Agufobi L, Akindele AJ, Olowe JA, Familoni OB. Effect of Flabellaria paniculata Cav. extracts on gastric ulcer in rats. BMC Complement Altern Med. 2012;12(1):168. https://doi.org/10.1186/1472-6882-12-168.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Strand DS, Kim D, Peura DA. 25 years of proton pump inhibitors: a comprehensive review. Gut liver. 2017;11(1):27–37. https://doi.org/10.5009/gnl15502.

    Article  CAS  PubMed  Google Scholar 

  95. Thomas P, Holland N, Bolognesi C, Kirsch-Volders M, Bonassi S, Zeiger E, et al. Buccal micronucleus cytome assay. Nat Protoc. 2009;4(6):825–37. https://doi.org/10.1038/nprot.2009.53.

    Article  CAS  PubMed  Google Scholar 

  96. Thongon N, Krishnamra N. Omeprazole decreases magnesium transport across Caco-2 monolayers. World J Gastroenterol. 2011;17(12):1574–83. https://doi.org/10.3748/wjg.v17.i12.1574.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Tonazzi A, Console L, Indiveri C. Inhibition of mitochondrial carnitine/acylcarnitine transporter by H2O2: molecular mechanism and possible implication in pathophysiology. Chem Biol Interact. 2013;203(2):423–9. https://doi.org/10.1016/j.cbi.2013.01.006.

    Article  CAS  PubMed  Google Scholar 

  98. Thomas B, Mohamed M, Al Hail M, Awwad FAY, Wahba RM, Hassan SB, et al. A case of probable esomeprazole-induced transient liver injury in a pregnant woman with hyperemesis. Clin Pharmacol. 2016;8:199–202. https://doi.org/10.2147/cpaa.s106234.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Trindade LMDF, de Oliveira Menezes LB, de Souza Neta AM, Rolemberg PCL, Souza LD, de Carvalho Barreto ID, Meurer L. Prevalence of Helicobacter pylori infection in samples of gastric biopsies. Gastroenterol Res. 2017;10(1):33–41. https://doi.org/10.14740/gr785w.

    Article  Google Scholar 

  100. Tsuchiya T, Kijima A, Ishii Y, Takasu S, Yokoo Y, Nishikawa A, et al. Mechanisms of oxidative stress-induced in vivo mutagenicity by potassium bromate and nitrofurantoin. J Toxicol Pathol. 2018;31(3):179–88. https://doi.org/10.1293/tox.2018-0024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Udelnow A, Kreyes A, Ellinger S, Landfester K, Walther P, Klapperstueck T, Wohlrab J, Henne-Bruns D, Knippschild U, Würl P. Omeprazole inhibits proliferation and modulates autophagy in pancreatic cancer cells. PloS one. 2011;6:e20143. https://doi.org/10.1371/journal.pone.0020143.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Xiao X, Wu ZC, Chou KC. A multi-label classifier for predicting the subcellular localization of gram-negative bacterial proteins with both single and multiple sites. PLoS ONE. 2011;6(6): e20592. https://doi.org/10.1371/journal.pone.0020592.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Uotani T, Graham DY. Diagnosis of Helicobacter pylori using the rapid urease test. Ann Transl Med. 2015;3(1):9. https://doi.org/10.3978/j.issn.2305-5839.2014.12.04.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Ward WM, Hoffman JD, Loo G. Genotoxic effect of ethacrynic acid and impact of antioxidants. Toxicol Appl Pharmacol. 2015;286(1):17–26. https://doi.org/10.1016/j.taap.2015.03.016.

    Article  CAS  PubMed  Google Scholar 

  105. Wessler S, Krisch LM, Elmer DP, Aberger F. From inflammation to gastric cancer–the importance of Hedgehog/GLI signaling in Helicobacter pylori-induced chronic inflammatory and neoplastic diseases. Cell Commun Signal. 2017;15(1):1–13. https://doi.org/10.1186/s12964-017-0171-4.

    Article  CAS  Google Scholar 

  106. Woźniak A, Walawender M, Tempka D, Coy E, Załęski K, Grześkowiak BF, Mrówczyński R. In vitro genotoxicity and cytotoxicity of polydopamine-coated magnetic nanostructures. Toxicol In Vitro. 2017;44:256–65. https://doi.org/10.1016/j.tiv.2017.07.022.

    Article  CAS  PubMed  Google Scholar 

  107. Yu LY, Sun LN, Zhang XH, Li YQ, Yu L, Meng L, et al. A review of the novel application and potential adverse effects of proton pump inhibitors. Adv Ther. 2017;34(5):1070–86. https://doi.org/10.1007/s12325-017-0532-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhang JL, Liu M, Yang Q, Lin SY, Shan HB, Wang HY, Xu GL. Effects of omeprazole in improving concurrent chemoradiotherapy efficacy in rectal cancer. World J Gastroenterol. 2017;23(14):2575–84. https://doi.org/10.3748/wjg.v23.i14.2575.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Zheng H, Chen Y, Zhang J, Wang L, Jin Z, Huang H, et al. Evaluation of protective effects of costunolide and dehydrocostuslactone on ethanol-induced gastric ulcer in mice based on multi-pathway regulation. Chem Biol Interact. 2016;250:68–77. https://doi.org/10.1016/j.cbi.2016.03.003.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are owed to RENORBIO and UFPI for funding and hosting, respectively of this project.

Funding

No Funding received but Will Pay the APC.

Author information

Authors and Affiliations

Authors

Contributions

The work was supervised by MTI, JS-R and AAdCMC. Project administration, Final draft of the work, by AMOFdM, MFCJP, A-APMdM, AdR, BdSS, CDdCS, SAM, TSGM, CS, SDD, MA.A, MTI, JS-R, JMdCeS, AAdCMC. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Muhammad Torequl Islam or Javad Sharifi-Rad.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

da Mata, A.M.O.F., Paz, M.F.C.J., de Menezes, AA.P.M. et al. Evaluation of mutagenesis, necrosis and apoptosis induced by omeprazole in stomach cells of patients with gastritis. Cancer Cell Int 22, 154 (2022). https://doi.org/10.1186/s12935-022-02563-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-022-02563-5

Keywords