Next Article in Journal
Assessment of Metal Contamination in Water of Freshwater Aquaculture Farms from a South Asian Tropical Coastal Area
Previous Article in Journal
Performance and Mechanism of As(III/V) Removal from Aqueous Solution by Fe3O4-Sunflower Straw Biochar
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Deoxynivalenol Induces Apoptosis via FOXO3a-Signaling Pathway in Small-Intestinal Cells in Pig

1
Department of Animal Science and Biotechnology, Kyungpook National University, Sangju-si 37224, Korea
2
Department of Bio Life Sciences, Shingu College, Seongnam-si 13174, Korea
*
Author to whom correspondence should be addressed.
Toxics 2022, 10(9), 535; https://doi.org/10.3390/toxics10090535
Submission received: 5 August 2022 / Revised: 7 September 2022 / Accepted: 7 September 2022 / Published: 13 September 2022
(This article belongs to the Section Toxicology)

Abstract

:
Deoxynivalenol (DON) is a mycotoxin that is found in feed ingredients derived from grains such as corn and wheat. Consumption of DON-contaminated feed has been shown to cause damage to the intestine, kidneys, and liver. However, the molecular mechanism by which DON exerts its effect in the small intestine is not completely understood. As a result, we profiled gene expression in intestinal epithelial cells treated with DON and examined the molecular function in vitro. We hypothesized that DON could induce apoptosis via the FOXO3a-signaling pathway in intestinal epithelial cells based on these findings. DON induced the apoptosis and the translocation of FOXO3a into the nucleus. Moreover, the inhibiting of FOXO3a alleviated the apoptosis and expression of apoptosis-related genes (TRAL, BCL-6, CASP8, and CASP3). ERK1/2 inhibitor treatment suppressed the translocation of FOXO3a into the nucleus. Our discovery suggests that DON induces apoptosis in intestinal epithelial cells through the FOXO3a-signaling pathway.

1. Introduction

Deoxynivalenol (DON), a Fusarium mycotoxin, is a risk factor for human and farm animal health, and the consumption of cereal-based foods can result in its exposure [1,2]. It has a high detection rate worldwide and is frequently present in feedstuffs [3]. DON can also enter the respiratory tract during baking, grinding, or brewing, posing a health risk to workers [4]. Porcine livestock are particularly susceptible to DON exposure due to their cereal-rich diets and rapid absorption [5,6]. Additionally, DON exposure can induce the expression of cytokines, chemokines, and inflammatory genes in vitro, while oral exposure can result in gastroenteritis, diarrhea, vomiting, anorexia, malabsorption, and weight loss [7]. According to adverse studies, DON causes oxidative stress, DNA damage, cell cycle arrest in the G2/M cell cycle phase, deregulation of cell signaling, intestinal barrier disorder, and apoptosis [8,9].
The intestinal epithelial cells (IECs) consist of a continuous single-layered sheet that serves as a barrier between the internal and external environments [10]. IECs are primarily responsible for transporting nutrients, electrolytes, and digestive enzymes, as well as acting as a protective barrier against pathogens and toxins [11]. The porcine intestinal epithelial cells (IPEC-J2) are critical for studying the transport and barrier properties of the porcine intestine at the subcellular or molecular level, thereby assisting in the reduction in animal usage [12]. Additionally, IPEC-J2 studies have been conducted in various fields, including cell viability, cell cycle, nutrient-transport genes, and mitochondrial-related genes [13]. However, the study of the apoptosis-related signaling pathways in IPEC-J2 following DON treatment is lacking.
Apoptosis, one of the cell death processes, is a highly regulated process of cell death that is triggered by a variety of signaling cascades and that ultimately results in death. Essentially, apoptosis is a defense mechanism that eliminates unnecessary cells [14,15]. On the other hand, excessive apoptosis has been linked to a variety of diseases, including neurological disorders, cardiovascular disorders, autoimmune disease, Crohn’s disease, and chronic inflammation [16,17]. Similarly, excessive cell death in IECs can disrupt the internal integrity of the intestinal barrier, resulting in inflammatory disease [18]. One of the apoptosis markers, caspases, are a critical regulator of cell death, and their activated form, cleaved caspase, has been shown to induce apoptotic cell death [19]. Numerous studies indicate that DON induces apoptosis [20,21].
The Forkhead box O (FOXO) transcription factor is involved in autophagy, apoptosis, cell cycle, differentiation, DNA damage repair, and oxidative stress [22]. In comparison to the other FOXO family members, FOXO3a is the most important functional transcription factor in cell death, which is associated with apoptosis and cell cycle inhibition [23]. FOXO3a induces transcription of apoptotic-associated genes such as TRAIL, BCL-6, PUMA, and BIM, resulting in apoptotic processes [24]. The FOXO3a transcription factor is also referred to as a tumor suppressor, and it has been implicated in a variety of signaling pathways, including PI3K/AKT and ER-α [23]. However, the molecular mechanism by which DON induces apoptosis in the small intestine is unknown and requires further investigation.
In this study, we aimed to identify the molecular mechanism on deoxynivalenol-mediated injuries in the porcine small-intestinal epithelium. Accordingly, gene-expression profiling was performed, and based on this data, genes differentially expressed by DON were identified, and the damage mechanism was additionally investigated.

2. Materials and Methods

2.1. Cell Culture and Treatment

IPEC-J2 (DSMZ, Braunschweig, Germany), which was originally isolated from a piglet’s jejunal epithelium, was cultured in a CO2 incubator using Dulbecco’s Modified Eagle Medium (DMEM) (Thermo Fisher Scientific, Wilmington, DE, USA) supplemented with 10% fetal bovine serum and 1% penicillin–streptomycin.

2.2. Cell Viability

Cell viability was determined using a microplate reader set to 450 nm absorbance after 2 h of treatment with WST-1 (Roche Diagnostics GmbH, Mannheim, Germany). Following the IPEC-J2 culture, cells were seeded at a density of 1 × 104 per 100 μL in 96-well plates for 32 h and then incubated overnight in DMEM. For 15 h, DON (Sigma–Aldrich, St. Louis, MO, USA) treatments were performed at concentrations of 0.25 μg/mL, 0.5 μg/mL, 1 μg/mL, 2 μg/mL, 4 μg/mL, 10 μg/mL, 20 μg/mL, and 50 μg/mL.

2.3. Gene-Expression Profiling

IPEC-J2 cells were seeded at a density of 4 × 105 in 6-well plates, incubated in DMEM overnight, and treated with DON 2 μg/mL. TRIzol reagent (Thermo Fisher Scientific, Wilmington, DE, USA) was used to extract total RNA. The Agilent 2100 bioanalyzer was used in conjunction with the RNA 6000 Nano Chip (Agilent Technologies, Amstelveen, The Netherlands) and the Thermo Inc. ND-2000 Spectrophotometer (Thermo, Wilmington, DE, USA) to determine the quality and quantity of RNA. The QuantSeq 3′ mRNA-Seq Library Prop kit was used to construct the library according to the manufacturer’s instructions for the controlling and testing of RNAs. To summarize, 500 ng total RNA was prepared and then hybridized to an oligo-dT primer containing an Illumina-compatible sequence at its 5′ end followed by reverse transcription. Following degradation of the RNA template, second-strand synthesis was initiated using a random primer with an Illumina-compatible linker sequence at its 5′ end. The double-strand library was purified using magnetic beads and amplified to incorporate all of the required adapter sequences for cluster generation. The completed library was purified from PCR components. Sequencing at a high-throughput single-end 75 sequencing was carried out using NextSeq 500 system (Illumina, Inc., San Diego, CA, USA). Gene-expression-profiling data were identified via Excel-based DEGs (differentially expressed genes) analysis. DEGs were analyzed using a GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) mapper for annotation, integration, and visualization. The DEGs were adjusted up- and downregulated by at least 2-fold.

2.4. Annexin-V and Propidium Iodide Staining

After 24 h of DON treatment, the IPEC-J2 cells were harvested and subsequently washed in PBS. Following centrifugation, the supernatants were discarded and treated with 1× Annexin binding buffer, and 5 μL Alexa Fluor 488 Annexin-V (Thermo Fisher Scientific, Wilmington, DE, USA) and 1 μL 100 mg/mL PI working solution were added. Following that, the cells were incubated at room temperature in the dark for 15 min. Subsequently, cells were stained with DAPI (Vector Laboratories, Burlingame, CA, USA) and mounted on slides using coverslips. The photographs were taken with a fluorescence microscope (Korealabtech, Seongnam-si, Gyeonggi-do, Korea).

2.5. Extraction of Cytoplasmic and Nuclear Proteins

Cytoplasmic and nuclear proteins were extracted according to the manufacturer’s instructions using NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo, Wilmington, DE, USA). The cells were washed with chilled PBS and centrifuged for 3 min at 500× g. After discarding the supernatant, CERI (cytoplasmic extraction reagent) was added to the cell pellet and vortexed for 15 s. The suspension was then incubated on ice for 10 min. After adding the CERII in suspension and vortexing for 15 s, the mixture was incubated for 1 min. The suspension was centrifuged at 16,000× g for 5 min, and the cytoplasmic extraction suspension was transferred to a pre-chilled tube. The insoluble pellet was added with nuclear extraction reagent and vortexed for 15 s every 10 min for a total of 40 min. Following the ten-minute centrifugation at 16,000× g, the supernatant was transferred to a pre-chilled tube. The extracted protein was used in subsequent experiments.

2.6. RT-PCR and Western Blotting

The AccuPreP Universal RNA Extraction kit was used to extract total RNA (BioNEER, Daejeon, Korea). The cDNA was used to generate total RNA (1 μg) using the DiaStar™ RT Kit (SolGent, Daejeon, Korea). Primer 3 (http://frodo.wi.mit.edu) was used to design the primers for the target genes for qPCR. The following conditions were used for the qRT-PCR: 95 °C for 3 min, followed by 40 cycles at 95 °C for 15 s, 56–58 °C for 15 s, and 72 °C for 15 s. Normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH), target gene levels were calculated using the 2−ΔΔCt method. The primer sequence of genes were shown in Table 1.
Protein was extracted for Western blotting using a lysis buffer containing a protease inhibitor cocktail. A Pierce BCA Protein Assay kit (Thermo Fisher Scientific, Waltham, MA, USA) was used to determine the protein concentration. After electrophoresis in 9% polyacrylamide gel for 1 h at 100 volts, the protein sample was transferred to a membrane and washed three times with Tris-buffered saline containing 0.1% Tween-20. After 1 h of blocking, the membrane was processed overnight with the primary antibodies, including anti-FOXO3a (Novus Biologicals, Centennial, CO, USA) and anti-ERK1/2 (Cell Signaling Technology, Danvers, MA, USA). After 1 h of secondary antibody treatment, the membrane was visualized for immunoreactivity using the ECL reagent. The ChemiDoc imaging system was used to obtain images of protein bands.

2.7. Gene Silencing of FOXO3a with Small Interfering RNA

To inhibit FOXO3a expression, small interfering RNA (siRNA) was synthesized (Bioneer). The siFOXO3a-518 target sequence was 5′–GACAAACGGCUCACUCUGU-3′, the siFOXO3a-1774 target sequence was 5′-CUCAACGAGUGCGAACCUU-3′, and the siFOXO3a-1895 target sequence was 5′-GAUGCUGACGGGUUGGAUU-3′. Lipofectamine RNAi Reagent was used to transfect siRNA according to the manufacturer’s instructions (Life Technologies, Carlsbad, CA, USA). The IPEC-J2 Cells were incubated for 24 h after siRNA and DON treatments.

2.8. Immunofluorescence Staining of Cells

Cell monolayers on coverslips treated with DON were disposed of in 4% paraformaldehyde for 15 min at room temperature and then washed with PBS for 5 min. To begin, the IPEC-J2 was blocked for 1 h. Following that, Rabbit anti-FOXO3a IgG was treated in a 1:200 antibody solution and incubated in cells overnight at 4 °C. After 5 min of washing with PBS, goat anti-Rabbit (488) (Thermo Fisher Scientific, Carlsbad, CA, USA) was added at a ratio of 1:500 to an antibody solution and incubated in the dark for 1 h. Following that, coverslips were adhered to slides and stained with DAPI (Vector Laboratories, Burlingame, CA, USA). The photographs were taken with a fluorescence microscope.

2.9. Statistics

To determine the significance of the control and treatment groups, data analysis was performed using the general linear model (PROC-GLM) and T-test of SAS. The error bars represent the standard errors of triplicate analysis. In comparison to the control group, a significant difference in the treatment group was denoted by the following symbols: * p < 0.05, ** p < 0.01. The difference was evaluated using the Duncan multiple range tests.

3. Results

3.1. DON Decreased the Viability of IPEC-J2

To determine the viability of IPEC-J2 cells treated with DON, a WST-1 cell proliferation assay was used. IPEC-J2 cells were treated with varying concentrations of DON (0 μg/mL, 0.25 μg/mL, 0.5 μg/mL, 1 μg/mL, 2 μg/mL, 4 μg/mL, 10 μg/mL, 20 μg/mL, and 50 μg/mL) (Figure 1). The inhibitory concentration at half-maximum (IC50) was 2.284 μg/mL. In subsequent experiments, IPEC-J2 cells were treated with DON at a concentration of 2 μg/mL for 24 h to elucidate the molecular mechanism.

3.2. Identification and Validation of DEGs

We identified 837 differentially expressed genes (DEGs) after analyzing the gene-expression profiles, including 411 upregulated and 426 downregulated genes (Figure 2A). Additionally, we confirmed the expression of the TOP 5 genes associated with upregulated DEGs using RT-PCR (Figure 2B). “Poly (A) RNA binding”, “ATP binding”, “zinc ion binding”, and “nucleotide binding” were included in the “molecular function” of upregulated DEGs identified through Gene Ontology (GO) analysis. The term “cellular component” was used interchangeably with “cytoplasm” and “nucleus.” The terms “microtubule-based movement” and “mRNA splicing via spliceosome” were used to describe “biological processes” (Figure 2C, Tables S1–S3). In comparison, the downregulated DEG demonstrated that the “molecular function” was associated with “structural constituent of the ribosome”, “poly (A) RNA binding”, “calcium ion binding”, and “NAD binding.” The terms “extracellular exosome”, “cytoplasm”, “focal adhesion”, and “cell surface” included the term “cellular component.” The terms “translation”, “oxidation-reduction process”, “cell adhesion”, “cell-matrix adhesion”, and “positive regulation of the apoptotic process” were used to describe “biological processes” (Figure 2D, Tables S4–S6). The major upregulated signaling pathways identified in the KEGG analysis were the “FOXO signaling pathway”, “MAPK signaling pathway”, and “p53 signaling pathway” (Figure 2E, Table S7). However, the major downregulated signaling pathways discussed include the metabolic pathway, antibiotic biosynthesis, and ribosome biosynthesis (Figure 2E, Table S8).

3.3. DON Induces Apoptosis in IPEC-J2

To determine whether DON induces apoptosis in IPECs, we performed double staining with Annexin-V and propidium iodide at various concentrations of DON (1 μg/mL, 2 μg/mL) (Figure 3A). Apoptosis was significantly increased in cells treated with DON 1, 2 μg/mL compared to untreated cells. Additionally, we confirmed the mRNA-expression level of apoptosis-related genes such as TRAIL, BCL-6, CASP3, and CASP8. These were significantly increased when compared to the untreated control (Figure 3B).

3.4. DON Triggers Translocation of FOXO3a to the Nucleus

We used quantitative real-time PCR, Western blotting, and immunocytochemistry to determine the expression of FOXO3a. At 1 ug/mL DON, the level of FOXO3a mRNA expression was not significantly different from the control. However, when DON 2 μg/mL was used, the level of FOXO3a mRNA expression was significantly increased compared to the control (Figure 4A). There was no difference in the level of FOXO3a protein expression (Figure 4B). Immunocytochemistry analysis revealed that DON induces FOXO3a nuclear translocation (Figure 4C). Additionally, at a DON 2 μg/mL concentration, FOXO3a expression was significantly decreased in the cytoplasm and significantly increased in the nucleus (Figure 4D). Following these findings, subsequent experiments were conducted at a DON concentration of 2 μg/mL.

3.5. Knockdown of FOXO3a Reduces Apoptosis in Porcine Intestinal Epithelial Cells

We investigated whether silencing FOXO3a affects apoptosis. We confirmed the levels of mRNA and protein expression in three distinct sequences using quantitative real-time PCR and Western blotting (Figure 5A). In light of these findings, we proceeded with subsequent experiments using siFOXO3a-1774. After treatment with siFOXO3a, we analyzed the mRNA-expression levels of apoptosis-related genes (TRAIL, BCL-6, CASP3, and CASP8). These results were significantly reduced when compared to the control group (Figure 5B). Additionally, when cells were treated with siRNA, apoptosis was significantly reduced (Figure 5C).

3.6. Inhibition of ERK1/2 Represses Apoptosis and Translocation of FOXO3a

On IPEC-J2 treated with DON, we examine the phosphorylation of ERK1/2. DON stimulated ERK1/2 phosphorylation in a time-dependent manner (1 h, 2 h, 3 h, 4 h, and 5 h) (Figure 6A), whereas ERK1/2 inhibitor PD98059 inhibited ERK1/2 phosphorylation (Figure 6B). Additionally, inhibition of ERK1/2 phosphorylation decreased the expression of genes associated with apoptosis when compared to untreated control (Figure 6C). Following that, we examined the effect of inhibiting ERK1/2 phosphorylation on FOXO3a translocation. When cells were treated with PD98059 and DON, the level of FOXO3a protein expression in the cytoplasm was increased compared to untreated controls, whereas when compared to untreated controls, the level of FOXO3a protein expression in the nucleus was decreased (Figure 6D).

4. Discussion

Fusarium produces DON during the growth and storage of grains. DON-contaminated feed is found worldwide and is also referred to as vomitoxin since high concentrations cause vomiting [25]. Consumption of feed contaminated with DON results in decreased feed intake, delayed growth, and weight loss [26,27]. Additionally, DON alters the intestine’s morphology and impairs nutrient absorption [28]. As a result of ingesting DON, livestock productivity can be reduced, resulting in economic loss. Porcine is one of the most susceptible species to DON, which is associated with a high grain intake [29,30]. At the cellular level, DON toxicity causes mitochondrial dysfunction, which results in the accumulation of reactive oxygen species (ROS), apoptosis, and inflammation, as well as inhibits protein synthesis via binding to the ribosome [31,32]. However, the effect of DON on porcine intestinal epithelial cells is unknown. As a result, we profiled the gene expression of IPEC-J2 cells following DON treatment. Our study sought to confirm DON-induced apoptosis in porcine intestinal epithelial cells via the FOXO3a-signaling pathway.
The molecular mechanism by which DON induces IPEC-J2 requires additional research. As a result, we profiled the expression of 837 DEGs in porcine intestinal epithelial cells. Cell adhesion and cell-matrix adhesion were identified as the main GO terms, and the FOXO-signaling pathway, MAPK-signaling pathway, and p53-signaling pathway were identified as the primary pathways. FOXO3a, a member of the Forkhead transcription factor FOXO subfamily, has been implicated in processes such as apoptosis, metabolism, stress response, and proliferation [33]. FOXO3a, in particular, acts as an apoptosis regulator [34]. As a result of these findings, we hypothesized that DON could induce apoptosis via the FOXO-signaling pathway.
To confirm DON-induced apoptosis in the small intestine, we performed real-time PCR analysis on the expression of apoptotic genes. FOXO3a has been known to regulate the expression of genes involved in cell death, including tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL), Bcl-2 interacting mediator of cell death (BIM), B-cell CLL/lymphoma 6 (BCL-6), and Fas ligand (FASLG) [35]. TRAIL induces apoptosis by activating death receptors, which activate caspase-8 and result in apoptosis. BCL-6 indirectly inhibits the pro-survival BCL-2 family, resulting in an increase in mitochondrial permeability and activation of caspase-3, which induces apoptosis [36]. Our study established that DON induces apoptosis in IPEC-J2 cells via apoptosis associated with genes such as TRAIL, BCL-6, CASP3, and CASP8 and that DON induces the translocation of FOXO3a into the nucleus. Numerous additional studies indicate that DON induces apoptosis in porcine intestinal epithelial cells via diverse pathway [13,21,37]. However, studies of DON-induced apoptosis via FOXO3a-signaling pathway do not yet exist.
Additionally, we confirmed that DON-induced ERK1/2 phosphorylation. Additionally, to determine whether DON translocates FOXO3a to the nucleus and induces apoptosis, we used siRNA and the ERK1/2 inhibitor PD98059 to induce FOXO3a knockdown. ERK1/2 are protein serin/threonine kinase and become involved in the Ras-Raf-MEK-ERK signal-transduction cascade that is related to various processes such as cell cycle, cell adhesion, migration, differentiation, metabolism, proliferation, transcription, and cell survival. ERK1/2 cascade is also associated with FOXO, which regulates apoptosis and cell cycle arrest [38,39]. Protein kinase B (PKB), ERK, serum- and glucocorticoid-inducible kinases, and IǩB kinase isoform β were previously identified as regulating FOXO3a translocation [40]. The phosphorylation of ERK1/2 by DON has been shown in previous studies [41]. Our study established that DON induces ERK1/2 in a time-dependent manner. Following that, when IPEC-J2 cells were treated with the PD98059 and DON, the treatment decreased the expression of apoptosis-related genes and inhibited the translocation of FOXO3a to the nucleus. Recent research indicates that the silencing of FOXO3a inhibited apoptosis in human gastric mucosa cell lines treated with DON [42]. We examined the effect of FOXO3a silencing on DON-induced apoptosis. This finding demonstrates that DON can induce apoptosis in porcine intestinal epithelial cells via the FOXO3a-signaling pathway.
These results established that DON can induce apoptosis in porcine intestinal epithelial cells via the FOXO3a-signaling pathway (Figure 7). This study demonstrated that DON-contaminated feedstuffs can deteriorate pig small-intestine health, resulting in decreased productivity.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/toxics10090535/s1, Additional information about Gene Ontology and KEGG pathway analysis to differentially expressed genes (PDF), Table S1: biological process of upregulated differentially expressed genes, Table S2: cellular component of upregulated differentially expressed genes, Table S3: molecular function of upregulated differentially expressed genes, Table S4: biological process of downregulated differentially expressed genes, Table S5: cellular component of downregulated differentially expressed genes, Table S6: molecular function of downregulated differentially expressed genes, Table S7: KEGG pathway of upregulated differentially expressed genes, Table S8: KEGG pathway of downregulated differentially expressed genes.

Author Contributions

T.H.K., formal analysis, writing—original draft preparation, validation and software; K.S.K., writing—reviewing and editing; S.I.L., conceptualization, writing—reviewing and editing, supervision, data curation. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2022R1I1A3070740).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pestka, J.J. Deoxynivalenol-induced proinflammatory gene expression: Mechanisms and pathological sequelae. Toxins 2010, 2, 1300–1317. [Google Scholar] [CrossRef] [PubMed]
  2. Pestka, J.J. Deoxynivalenol: Mechanisms of action, human exposure, and toxicological relevance. Arch. Toxicol. 2010, 84, 663–679. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, X.C.; Zhang, Y.F.; Cao, L.; Zhu, L.; Huang, Y.Y.; Chen, X.F.; Chu, X.Y.; Zhu, D.F.; Ur Rahman, S.; Feng, S.B.; et al. Deoxynivalenol Induces Intestinal Damage and Inflammatory Response through the Nuclear Factor-kappaB Signaling Pathway in Piglets. Toxins 2019, 11, 663. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, X.; Fan, M.; Chu, X.; Zhang, Y.; Rahman, S.U.; Jiang, Y.; Chen, X.; Zhu, D.; Feng, S.; Li, Y.; et al. Deoxynivalenol induces toxicity and apoptosis in piglet hippocampal nerve cells via the MAPK signaling pathway. Toxicon 2018, 155, 1–8. [Google Scholar] [CrossRef] [PubMed]
  5. Vatzia, E.; Pierron, A.; Saalmuller, A.; Mayer, E.; Gerner, W. Deoxynivalenol Affects Proliferation and Expression of Activation-Related Molecules in Major Porcine T-Cell Subsets. Toxins 2019, 11, 644. [Google Scholar] [CrossRef]
  6. Przybylska-Gornowicz, B.; Lewczuk, B.; Prusik, M.; Hanuszewska, M.; Petrusewicz-Kosinska, M.; Gajecka, M.; Zielonka, L.; Gajecki, M. The Effects of Deoxynivalenol and Zearalenone on the Pig Large Intestine. A Light and Electron Microscopy Study. Toxins 2018, 10, 148. [Google Scholar] [CrossRef]
  7. Waskiewicz, A.; Beszterda, M.; Kostecki, M.; Zielonka, L.; Golinski, P.; Gajecki, M. Deoxynivalenol in the gastrointestinal tract of immature gilts under per os toxin application. Toxins 2014, 6, 973–987. [Google Scholar] [CrossRef]
  8. Kowalska, K.; Koziel, M.J.; Habrowska-Gorczynska, D.E.; Urbanek, K.A.; Dominska, K.; Piastowska-Ciesielska, A.W. Deoxynivalenol induces apoptosis and autophagy in human prostate epithelial cells via PI3K/Akt signaling pathway. Arch. Toxicol. 2022, 96, 231–241. [Google Scholar] [CrossRef]
  9. Xue, D.F.; Yang, P.; Yang, Y.Y.; Wang, Y.A.; Wu, K.T.; Qi, D.S.; Wang, S. Deoxynivalenol triggers porcine intestinal tight junction disorder through hijacking SLC5A1 and PGC1 alpha-mediated mitochondrial function. Food Chem. Toxicol. 2022, 163, 112921. [Google Scholar] [CrossRef]
  10. Schneeberger, K.; Roth, S.; Nieuwenhuis, E.E.S.; Middendorp, S. Intestinal epithelial cell polarity defects in disease: Lessons from microvillus inclusion disease. Dis. Model. Mech. 2018, 11, dmm031088. [Google Scholar] [CrossRef] [Green Version]
  11. Hegyi, P.; Maleth, J.; Walters, J.R.; Hofmann, A.F.; Keely, S.J. Guts and Gall: Bile Acids in Regulation of Intestinal Epithelial Function in Health and Disease. Physiol. Rev. 2018, 98, 1983–2023. [Google Scholar] [CrossRef] [PubMed]
  12. Zakrzewski, S.S.; Richter, J.F.; Krug, S.M.; Jebautzke, B.; Lee, I.F.; Rieger, J.; Sachtleben, M.; Bondzio, A.; Schulzke, J.D.; Fromm, M.; et al. Improved cell line IPEC-J2, characterized as a model for porcine jejunal epithelium. PLoS ONE 2013, 8, e79643. [Google Scholar] [CrossRef] [PubMed]
  13. Liao, P.; Liao, M.; Li, L.; Tan, B.; Yin, Y. Effect of deoxynivalenol on apoptosis, barrier function, and expression levels of genes involved in nutrient transport, mitochondrial biogenesis and function in IPEC-J2 cells. Toxicol. Res. 2017, 6, 866–877. [Google Scholar] [CrossRef]
  14. Xu, X.; Lai, Y.; Hua, Z.C. Apoptosis and apoptotic body: Disease message and therapeutic target potentials. Biosci. Rep. 2019, 39, BSR20180992. [Google Scholar] [CrossRef] [PubMed]
  15. Charununtakorn, S.T.; Shinlapawittayatorn, K.; Chattipakorn, S.C.; Chattipakorn, N. Potential Roles of Humanin on Apoptosis in the Heart. Cardiovasc. Ther. 2016, 34, 107–114. [Google Scholar] [CrossRef] [PubMed]
  16. Favaloro, B.; Allocati, N.; Graziano, V.; Di Ilio, C.; De Laurenzi, V. Role of apoptosis in disease. Aging 2012, 4, 330–349. [Google Scholar] [CrossRef] [PubMed]
  17. Gunther, C.; Neumann, H.; Neurath, M.F.; Becker, C. Apoptosis, necrosis and necroptosis: Cell death regulation in the intestinal epithelium. Gut 2013, 62, 1062–1071. [Google Scholar] [CrossRef] [PubMed]
  18. Subramanian, S.; Geng, H.; Tan, X.D. Cell death of intestinal epithelial cells in intestinal diseases. Sheng Li Xue Bao 2020, 72, 308–324. [Google Scholar]
  19. Kuranaga, E. Beyond apoptosis: Caspase regulatory mechanisms and functions in vivo. Genes Cells 2012, 17, 83–97. [Google Scholar] [CrossRef]
  20. Xue, R.; Li, S.; Zou, H.; Ji, D.; Lv, M.; Zhou, P.; Wei, Z.; Zhang, Z.; Cao, Y. Melatonin alleviates deoxynivalenol-induced apoptosis of human granulosa cells by reducing mutually accentuated FOXO1 and ER stressdouble dagger. Biol. Reprod. 2021, 105, 554–566. [Google Scholar] [CrossRef]
  21. Kang, R.; Li, R.; Dai, P.; Li, Z.; Li, Y.; Li, C. Deoxynivalenol induced apoptosis and inflammation of IPEC-J2 cells by promoting ROS production. Environ. Pollut. 2019, 251, 689–698. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, X.; Liu, W.; Xiao, J.; Zhang, Y.; Chen, Y.; Luo, C.; Huang, Q.; Peng, F.; Gong, W.; Li, S.; et al. FOXO3a accumulation and activation accelerate oxidative stress-induced podocyte injury. FASEB J. 2020, 34, 13300–13316. [Google Scholar] [CrossRef] [PubMed]
  23. Long, J.; Yang, C.S.; He, J.L.; Liu, X.Q.; Ding, Y.B.; Chen, X.M.; Tong, C.; Peng, C.; Wang, Y.X.; Gao, R.F. FOXO3a is essential for murine endometrial decidualization through cell apoptosis during early pregnancy. J. Cell Physiol. 2019, 234, 4154–4166. [Google Scholar] [CrossRef] [PubMed]
  24. Lee, N.; Pun, N.T.; Jang, W.J.; Bae, J.W.; Jeong, C.H. Pitavastatin induces apoptosis in oral squamous cell carcinoma through activation of FOXO3a. J. Cell Mol. Med. 2020, 24, 7055–7066. [Google Scholar] [CrossRef]
  25. Savard, C.; Gagnon, C.A.; Chorfi, Y. Deoxynivalenol (DON) naturally contaminated feed impairs the immune response induced by porcine reproductive and respiratory syndrome virus (PRRSV) live attenuated vaccine. Vaccine 2015, 33, 3881–3886. [Google Scholar] [CrossRef]
  26. Pinton, P.; Tsybulskyy, D.; Lucioli, J.; Laffitte, J.; Callu, P.; Lyazhri, F.; Grosjean, F.; Bracarense, A.P.; Kolf-Clauw, M.; Oswald, I.P. Toxicity of deoxynivalenol and its acetylated derivatives on the intestine: Differential effects on morphology, barrier function, tight junction proteins, and mitogen-activated protein kinases. Toxicol. Sci. 2012, 130, 180–190. [Google Scholar] [CrossRef]
  27. Payros, D.; Alassane-Kpembi, I.; Pierron, A.; Loiseau, N.; Pinton, P.; Oswald, I.P. Toxicology of deoxynivalenol and its acetylated and modified forms. Arch. Toxicol. 2016, 90, 2931–2957. [Google Scholar] [CrossRef]
  28. Awad, W.A.; Ruhnau, D.; Hess, C.; Doupovec, B.; Schatzmayr, D.; Hess, M. Feeding of deoxynivalenol increases the intestinal paracellular permeability of broiler chickens. Arch. Toxicol. 2019, 93, 2057–2064. [Google Scholar] [CrossRef]
  29. Han, J.; Wang, Q.C.; Zhu, C.C.; Liu, J.; Zhang, Y.; Cui, X.S.; Kim, N.H.; Sun, S.C. Deoxynivalenol exposure induces autophagy/apoptosis and epigenetic modification changes during porcine oocyte maturation. Toxicol. Appl. Pharmacol. 2016, 300, 70–76. [Google Scholar] [CrossRef]
  30. Pierron, A.; Alassane-Kpembi, I.; Oswald, I.P. Impact of two mycotoxins deoxynivalenol and fumonisin on pig intestinal health. Porc. Health Manag. 2016, 2, 21. [Google Scholar] [CrossRef]
  31. Wang, S.; Wu, K.; Xue, D.; Zhang, C.; Rajput, S.A.; Qi, D. Mechanism of deoxynivalenol mediated gastrointestinal toxicity: Insights from mitochondrial dysfunction. Food Chem. Toxicol. 2021, 153, 112214. [Google Scholar] [CrossRef] [PubMed]
  32. Hooft, J.M.; Bureau, D.P. Deoxynivalenol: Mechanisms of action and its effects on various terrestrial and aquatic species. Food Chem. Toxicol. 2021, 157, 112616. [Google Scholar] [CrossRef] [PubMed]
  33. Nho, R.S.; Hergert, P. FoxO3a and disease progression. World J. Biol. Chem. 2014, 5, 346–354. [Google Scholar] [CrossRef] [PubMed]
  34. Parody, J.P.; Ceballos, M.P.; Quiroga, A.D.; Frances, D.E.; Carnovale, C.E.; Pisani, G.B.; Alvarez, M.L.; Carrillo, M.C. FoxO3a modulation and promotion of apoptosis by interferon-alpha2b in rat preneoplastic liver. Liver Int. 2014, 34, 1566–1577. [Google Scholar] [CrossRef]
  35. Das, T.P.; Suman, S.; Alatassi, H.; Ankem, M.K.; Damodaran, C. Inhibition of AKT promotes FOXO3a-dependent apoptosis in prostate cancer. Cell Death Dis. 2016, 7, e2111. [Google Scholar] [CrossRef]
  36. Fu, Z.; Tindall, D.J. FOXOs, cancer and regulation of apoptosis. Oncogene 2008, 27, 2312–2319. [Google Scholar] [CrossRef]
  37. Gu, X.; Guo, W.; Zhao, Y.; Liu, G.; Wu, J.; Chang, C. Deoxynivalenol-Induced Cytotoxicity and Apoptosis in IPEC-J2 Cells Through the Activation of Autophagy by Inhibiting PI3K-AKT-mTOR Signaling Pathway. ACS Omega 2019, 4, 18478–18486. [Google Scholar] [CrossRef]
  38. Zhang, T.; Yang, D.; Fan, Y.; Xie, P.; Li, H. Epigallocatechin-3-gallate enhances ischemia/reperfusion-induced apoptosis in human umbilical vein endothelial cells via AKT and MAPK pathways. Apoptosis 2009, 14, 1245–1254. [Google Scholar] [CrossRef]
  39. Roskoski, R., Jr. ERK1/2 MAP kinases: Structure, function, and regulation. Pharmacol. Res. 2012, 66, 105–143. [Google Scholar] [CrossRef]
  40. Liu, Y.; Ao, X.; Ding, W.; Ponnusamy, M.; Wu, W.; Hao, X.; Yu, W.; Wang, Y.; Li, P.; Wang, J. Critical role of FOXO3a in carcinogenesis. Mol. Cancer 2018, 17, 104. [Google Scholar] [CrossRef]
  41. Zhang, H.; Deng, X.; Zhou, C.; Wu, W.; Zhang, H. Deoxynivalenol Induces Inflammation in IPEC-J2 Cells by Activating P38 Mapk And Erk1/2. Toxins 2020, 12, 180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Yang, Y.X.; Yu, S.; Liu, N.; Xu, H.B.; Gong, Y.Y.; Wu, Y.N.; Wang, P.L.; Su, X.O.; Liao, Y.C.; De Saeger, S.; et al. Transcription Factor FOXO3a Is a Negative Regulator of Cytotoxicity of Fusarium mycotoxin in GES-1 Cells. Toxicol. Sci. 2018, 166, 370–381. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Deoxynivalenol decreased viability in IPEC-J2. Cell viability was determined by WST-1 assay. IPEC-J2 cells were incubated with different concentrations of DON (0.25, 0.5, 1, 2, 4, 10, 20, 50 μg/mL) for 24 h. Cell viability was expressed by the half-maximal inhibitory concentration (IC50).
Figure 1. Deoxynivalenol decreased viability in IPEC-J2. Cell viability was determined by WST-1 assay. IPEC-J2 cells were incubated with different concentrations of DON (0.25, 0.5, 1, 2, 4, 10, 20, 50 μg/mL) for 24 h. Cell viability was expressed by the half-maximal inhibitory concentration (IC50).
Toxics 10 00535 g001
Figure 2. Gene-expression profiling in DON-treated porcine intestinal epithelial cells. (A) Venn diagram of genes up or downregulated 2-folds compared with control after DON treatment. (B) The TOP5 of upregulated differentially expressed genes (DEGs) data were validated by real-time quantitative PCR (n = 3). (C) Gene Ontology enrichment analysis of upregulated genes (p < 0.01). (D) Gene Ontology enrichment analysis of downregulated genes (p < 0.01). (E) Kyoto Encyclopedia of Genes and Genomes pathway classification enrichment analysis of differentially expressed genes (p < 0.05). * p < 0.05, ** p < 0.01, *** p < 0.001.
Figure 2. Gene-expression profiling in DON-treated porcine intestinal epithelial cells. (A) Venn diagram of genes up or downregulated 2-folds compared with control after DON treatment. (B) The TOP5 of upregulated differentially expressed genes (DEGs) data were validated by real-time quantitative PCR (n = 3). (C) Gene Ontology enrichment analysis of upregulated genes (p < 0.01). (D) Gene Ontology enrichment analysis of downregulated genes (p < 0.01). (E) Kyoto Encyclopedia of Genes and Genomes pathway classification enrichment analysis of differentially expressed genes (p < 0.05). * p < 0.05, ** p < 0.01, *** p < 0.001.
Toxics 10 00535 g002
Figure 3. Deoxynivalenol induces apoptosis in porcine intestinal epithelial cells. (A) IPEC-J2 cells were treated at different concentrations of DON 1, 2 μg/mL for 24 h. Cell staining was performed through single-cell staining (Annexin-V; green, Propidium Iodide; red). Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue). (Scale bar means 20 μm.) (B) The mRNA levels of apoptosis-related genes (TRAIL, BCL-6, CASP8, CASP3) at DON 2 μg/mL concentration was compared with control (n = 3). Error bars indicated standard errors (SEs) of triplicate analysis. The arrow indicates Annexin-V positive-cell. The triangle indicates Annexin-V negative-cell. * p < 0.05, ** p < 0.01.
Figure 3. Deoxynivalenol induces apoptosis in porcine intestinal epithelial cells. (A) IPEC-J2 cells were treated at different concentrations of DON 1, 2 μg/mL for 24 h. Cell staining was performed through single-cell staining (Annexin-V; green, Propidium Iodide; red). Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue). (Scale bar means 20 μm.) (B) The mRNA levels of apoptosis-related genes (TRAIL, BCL-6, CASP8, CASP3) at DON 2 μg/mL concentration was compared with control (n = 3). Error bars indicated standard errors (SEs) of triplicate analysis. The arrow indicates Annexin-V positive-cell. The triangle indicates Annexin-V negative-cell. * p < 0.05, ** p < 0.01.
Toxics 10 00535 g003
Figure 4. Deoxynivalenol mediates FOXO3a expression and leads to translocation. (A,B) The expression levels of FOXO3a on the different concentrations of DON 1, 2 μg/mL were compared with the control (n = 3). Error bars indicated standard errors (SEs) of triplicate analysis. Significant differences between control and treatment groups are indicated as a,b. (C,D) FOXO3a translocation was evaluated using Western blotting and immunocytochemistry. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue). (Scale bar means 40 μm).
Figure 4. Deoxynivalenol mediates FOXO3a expression and leads to translocation. (A,B) The expression levels of FOXO3a on the different concentrations of DON 1, 2 μg/mL were compared with the control (n = 3). Error bars indicated standard errors (SEs) of triplicate analysis. Significant differences between control and treatment groups are indicated as a,b. (C,D) FOXO3a translocation was evaluated using Western blotting and immunocytochemistry. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue). (Scale bar means 40 μm).
Toxics 10 00535 g004
Figure 5. Knockdown of FOXO3a attenuates apoptosis. (A) After siFOXO3a treatment, levels of FOXO3a protein expression and mRNA expression. (B) After DON and siFOXO3a treatment, mRNA-expression level of apoptosis-related genes (TRAIL, BCL-6, CASP3, and CASP8) were compared with the negative control. (C) Apoptosis analysis was performed through single-cell staining using the Annexin-V (green) and propidium (red) iodide. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue). (Scale bar means 40 μm). * p < 0.05, ** p < 0.01, *** p < 0.001.
Figure 5. Knockdown of FOXO3a attenuates apoptosis. (A) After siFOXO3a treatment, levels of FOXO3a protein expression and mRNA expression. (B) After DON and siFOXO3a treatment, mRNA-expression level of apoptosis-related genes (TRAIL, BCL-6, CASP3, and CASP8) were compared with the negative control. (C) Apoptosis analysis was performed through single-cell staining using the Annexin-V (green) and propidium (red) iodide. Nuclei were stained with 4′,6-diamidino-2-phenylindole (DAPI; blue). (Scale bar means 40 μm). * p < 0.05, ** p < 0.01, *** p < 0.001.
Toxics 10 00535 g005
Figure 6. Doexynivalenol increased ERK1/2 phosphorylation, and inhibition of ERK1/2 suppresses a translocation of FOXO3a. (A) ERK1/2 phosphorylation level in IPEC-J2 cells after DON treatment in concentrations of 2 μg/mL for (1 h, 2 h, 3 h, 4 h, 5 h). (B) Level of phosphorylation (0 h, 1 h, and 2 h) of ERK1/2 by PD98059 in cells treated with DON at a concentration of 2 μg/mL. (C) The mRNA-expression levels of apoptosis-related genes (TRAIL, BCL-6, CASP3, and CASP8) on PD98059-treated IPEC-J2. (D) Western blotting analysis show inhibition of FOXO3a translocate after PD98059 treatment. Lowercase letters (a and b) mean significant differences between groups based on Duncan’s multi-range test.
Figure 6. Doexynivalenol increased ERK1/2 phosphorylation, and inhibition of ERK1/2 suppresses a translocation of FOXO3a. (A) ERK1/2 phosphorylation level in IPEC-J2 cells after DON treatment in concentrations of 2 μg/mL for (1 h, 2 h, 3 h, 4 h, 5 h). (B) Level of phosphorylation (0 h, 1 h, and 2 h) of ERK1/2 by PD98059 in cells treated with DON at a concentration of 2 μg/mL. (C) The mRNA-expression levels of apoptosis-related genes (TRAIL, BCL-6, CASP3, and CASP8) on PD98059-treated IPEC-J2. (D) Western blotting analysis show inhibition of FOXO3a translocate after PD98059 treatment. Lowercase letters (a and b) mean significant differences between groups based on Duncan’s multi-range test.
Toxics 10 00535 g006
Figure 7. Schematic illustrating of current working hypothesis toward DON-mediated apoptosis via the FOXO3a-signaling pathway. DON-induced translocation of FOXO3a to nucleus, which regulated TRAIL and BCL-6 expression and lead to activation of CAPS8 and CASP3. Finally, CASP8- and CASP3-induced apoptosis on porcine intestinal epithelial cells via the ERK1/2-signaling pathway.
Figure 7. Schematic illustrating of current working hypothesis toward DON-mediated apoptosis via the FOXO3a-signaling pathway. DON-induced translocation of FOXO3a to nucleus, which regulated TRAIL and BCL-6 expression and lead to activation of CAPS8 and CASP3. Finally, CASP8- and CASP3-induced apoptosis on porcine intestinal epithelial cells via the ERK1/2-signaling pathway.
Toxics 10 00535 g007
Table 1. List of PCR primers.
Table 1. List of PCR primers.
GenesDescriptionAccession No. Sequence (5′–3′)
TRAILTumor-necrosis-factor-related apoptosis-inducing ligandNM_001024696ForwardGCA GAC CTG TGT GTT GAT CC
ReverseGGG ATC CCA GAA ACT GTC AT
BCL-6B-cell CLL/lymphoma 6XM_005657112ForwardGTG TCC TAC GGT GCC TTT TT
ReverseTGA CGC AGA ATG TGA TGA GA
FOXO3Fork headbox O3NM_001135959ForwardTCA GCC AGT CTA TGC AAA CC
ReverseCCA TGA GTT CGC TAC GGA TA
CASP3Caspase3NM_214131ForwardCTC AGG GAG ACC TTC ACA AC
ReverseGCA CGC AAA TAA AAC TGC TC
CASP8Caspase8NM_001031779ForwardGCC GAC TGG ATG TGA TTT TAT
ReverseTGT TTC TGA GGT TGC TGG TC
GAPDHGlyceraldehyde-3-phosphate dehydrogenaseNM_001206359ForwardACA CCG AGC ATC TCC TGA CT
ReverseGAC GAG GCA GGT CTC CCT AA
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kang, T.H.; Kang, K.S.; Lee, S.I. Deoxynivalenol Induces Apoptosis via FOXO3a-Signaling Pathway in Small-Intestinal Cells in Pig. Toxics 2022, 10, 535. https://doi.org/10.3390/toxics10090535

AMA Style

Kang TH, Kang KS, Lee SI. Deoxynivalenol Induces Apoptosis via FOXO3a-Signaling Pathway in Small-Intestinal Cells in Pig. Toxics. 2022; 10(9):535. https://doi.org/10.3390/toxics10090535

Chicago/Turabian Style

Kang, Tae Hong, Kyung Soo Kang, and Sang In Lee. 2022. "Deoxynivalenol Induces Apoptosis via FOXO3a-Signaling Pathway in Small-Intestinal Cells in Pig" Toxics 10, no. 9: 535. https://doi.org/10.3390/toxics10090535

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