Skip to main content
Springer logoLink to Springer
. 2025 Oct 9;41(4):643–665. doi: 10.1007/s12550-025-00607-z

Comparative proteomic analysis indicates differential responses to fumonisin B1 (FB1) and hydrolysed fumonisin B1 (HFB1) in IPEC-J2 porcine epithelial cells in vitro

Nabeela Gamiet 1,2, Nashia Deepnarain 1, Stefan Abel 1, Hester-Mari Burger 3, Elisabeth Mayer 4, Mariska Lilly 1,
PMCID: PMC12612002  PMID: 41068355

Abstract

The intestinal epithelium is frequently exposed to environmental contaminants such as fumonisins, mycotoxins implicated in the development of mycotoxicosis across various mammalian species, with fumonisin B1 (FB1) being the most prevalent and toxic congener. Fumonisin B1 (FB1) can be enzymatically hydrolysed to produce hydrolysed fumonisin B1 (HFB1) that displays reduced inhibitory activity toward ceramide synthase. Given the central role of ceramide synthase in sphingolipid metabolism and cellular homeostasis, the reduced inhibitory activity of HFB1 is considered toxicologically favourable, as it is less likely to disrupt membrane integrity and critical signalling pathways. However, the toxicity of HFB1 remains variable across different in vitro and in vivo models. In this study, we evaluated the impact of FB1 and HFB1 on cell viability, apoptosis, and proliferation in the porcine intestinal cell line (IPEC-J2), including inflammatory responses through interleukin 8 (IL-8). Molecular mechanisms and pathways influenced by FB1 and HFB1 exposure were investigated through proteomic and bioinformatic analyses. Differentially abundant proteins (DAPs) were identified and functionally characterised using Gene Ontology analysis based on the Sus scrofa (domestic pig) database, revealing 52 significant DAPs between FB1 and HFB1 treatments compared to the control. Fibronectin 1 (FN1), an adhesive glycoprotein of the intestine, was consistently detected as a DAP in cells exposed to FB1 and HFB1. FB1 upregulates FN1, while HFB1 downregulates it, leading to different oncogenic pathways revealed by STRING enrichment analysis. Proteomic analysis further revealed distinct DAPs following FB1 and HFB1 exposure, implicating alterations in immune modulation (e.g. differential regulation of CD276), iron homeostasis (upregulation of FTL and FTH1), epithelial integrity (downregulation of NTN4, ST14), extracellular matrix remodelling (reduced SPARC), and angiogenesis-related pathways (decreased TINAGL1, FBLN2, SDC4) suggesting early changes in cellular signalling, stress response, and structural regulation that may be relevant to cancer biology and warrant further investigation. These findings also demonstrate that HFB1 activates distinct cancer-related pathways in vitro compared to FB1, with in vivo studies suggesting divergent mechanisms. HFB1 also induces more extensive protein expression changes in IPEC-J2 cells, as reflected by the greater number of DAPs and the complexity of enriched pathways. However, further investigation is needed to determine whether these changes directly contribute to cytotoxicity or represent compensatory cellular responses.

Supplementary Information

The online version contains supplementary material available at 10.1007/s12550-025-00607-z.

Keywords: Fumonisin B1, Hydrolysed fumonisin B1, Porcine intestinal epithelial cells, Proteomics

Introduction

The intestinal epithelium is comprised of interlinked columnar epithelial cells, such as enterocytes, which play critical roles in the absorptive and digestive functions of the intestine, alongside maintaining its barrier and immune functions (Subramanian et al. 2020). The epithelial tissue is constantly renewed every 3–5 days through a complex interplay of cell proliferation and apoptosis (Pan et al. 2018). The intestinal barrier is vital in protecting the body from harmful external agents, such as bacterial, fungal, or viral microorganisms (MacDonald and Monteleone 2005). Pathological invasions, such as microbial infections, trigger stress responses and induce cellular apoptosis, compromising barrier integrity (Apidianakis et al. 2009). Such disruptions lead to dysregulation of immune and inflammatory signalling, resulting in impaired epithelial renewal and barrier function (Zhou et al. 2017). Hence, maintaining intestinal homeostasis is crucial for optimal gut health and function.

The gastrointestinal tract serves as the first line of defence against ingested dietary substances (Szabó et al. 2023). Intestinal epithelial cells regulate the intestinal barrier function and are constantly exposed to large quantities of contaminants and toxins such as fumonisin B1 (FB1) and hydrolysed fumonisin B1 (HFB1) with subsequent negative health effects (Bouhet and Oswald 2007). FB1 is a low-molecular-weight secondary metabolite produced by Fusarium species and is well-known for causing mycotoxicosis in several mammalian species through ceramide synthase inhibition (Bertero et al. 2018). FB1 exposure in pigs induces toxic effects in organs such as the lungs, liver, and kidneys (Haschek et al. 2001). Pigs are highly susceptible to FB1 toxicity, exhibiting nephrotoxicity, hepatotoxicity, immunotoxicity, and disruption of the intestinal barrier function (Devriendt et al. 2009; Halloy et al. 2005; Knutsen et al. 2018a, 2018b; Loiseau et al. 2007; Terciolo et al. 2019). Previous in vivo studies conducted in piglets have suggested that hydrolysed fumonisins are less toxic than fumonisins (Grenier et al. 2012). Mammalian in vivo studies have also supported this finding (Grenier et al. 2012; Hahn et al. 2015; Howard et al. 2002). For example, Flynn et al. (1997) demonstrated that HFB1 is approximately 100-fold less toxic than FB1 in cultured rat embryos. The differences in membrane permeability (LogP) and metabolic pathways between FB1 and HFB1 elucidate the variations in their toxic effects. FB1’s higher lipophilicity facilitates greater tissue accumulation and pronounced toxicity, while the hydrolysis to HFB1 significantly mitigates these effects, making HFB1 a less toxic compound. The potential for further metabolic conversion of HFB1 to more toxic derivatives underscore the complexity of its toxicological profile (Schertz et al. 2018; Antonissen et al. 2020; Neckermann et al. 2021).

Studies by Gelderblom et al. (1993) demonstrate that FB1 promotes liver cancer initiation in rats in vivo, whereas HFB1 lacks this effect. Conversely, HFB1 exhibits greater cytotoxicity than FB1 in primary hepatocytes isolated from male Fischer rats, suggesting that HFB1 may exert enhanced cytotoxic effects under specific in vitro conditions (Gelderblom et al. 2001). Furthermore, when HFB1 is exposed to palmitoyl-CoA, it becomes acylated, forming N-palmitoyl HFB1, which is 10 times more toxic than FB1 in an in vitro model (Abou-Karam et al., 2004) . At the molecular level, changes in DNA (genomics), RNA (transcriptomics), proteins (proteomics), and small metabolites (metabolomics) can be observed, providing valuable insights into the mechanisms of toxicity (Eshelli et al. 2018; González-López et al. 2021). FB1 can be enzymatically converted to HFB1, a less powerful ceramide synthase inhibitor, which has shown varying toxicity across different in vitro and in vivo models (Humpf et al. 1998; Schelstraete et al. 2020). However, the mechanisms behind these differing results remain poorly understood (Caloni et al. 2005; Dellafiora et al. 2018; Hartl and Humpf 2000; Wang et al. 2016). Studies comparing the toxicological effects of HFB1 and FB1 in mammalian models are limited, and the data thus far are inconsistent (Abbax et al. 1993; Caloni et al. 2002; Dombrink-Kurtzman 2003). To better understand the comparative cellular responses and potential cytotoxic effects, this study aimed to optimise the in vitro model by exposing porcine IPEC-J2 cells to varying concentrations of FB1 and HFB1 to assess their impact on cellular intestinal viability.

Omics studies are essential for identifying, characterising, and quantifying these biological markers to understand the mechanisms of toxicity that may harm both animals and humans (Cimbalo et al. 2022). Proteomics involves studying the collection of proteins in cells and analysing their abundance to deepen our understanding of cellular responses to toxins (Aslam et al. 2016; Piñeiro et al. 2015). By examining the different signalling pathways activated through protein analysis and pathway enrichment, we gain better insights into the mechanisms of toxicity (Kan et al. 2017). While numerous studies have investigated the effects of FB1 and HFB1 separately, few have compared their impacts at the proteome level in vitro. This study, therefore, aims to evaluate the comparative effects of FB1 and HFB1 on the porcine intestinal cell line IPEC-J2 through proteomic analysis.

Materials and methods

Cell culture

Non-transformed, secondary intestinal porcine enterocytes (IPEC-J2) were obtained from dsm-firmenich, Tulln, Austria. Cells were inoculated and maintained in DMEM/Ham’s F-12 medium supplemented with heat-inactivated foetal bovine serum (FBS), 1% insulin-transferrin-selenite (ITS), epidermal growth factor (EGF) (Gibco-Life Technologies, Paisley, UK), L-glutamine, and 2-[4-(2-hydroxyethyl)piperazin-1-yl] ethane sulfonic acid (HEPES) buffer (Lonza, Basel, Switzerland). Cells were passaged twice a week at a split ratio of 1:3 and incubated at 37 ℃ in humidified air containing 5% carbon dioxide (CO2)/95% air. For the cell optimisation model, upon reaching 70–80% confluency, cells were washed with Hank’s balanced salt solution (HBSS), trypsinated (Lonza, Basel, Switzerland), and seeded in supplemented maintenance medium at the density of 3 × 104 cells per well in 96-well microtitre plates. Cultured cells were incubated for a minimum of 24 h at 37 ℃ in 5% CO2/95% air before medium was discarded. Thereafter, cells were exposed to FB1 and HFB1 concentrations (0.98 µM, 1.95 µM, 3.91 µM, 7.81 µM, 15.63 µM, 31.25 µM, 62.5 µM, 125 µM, 250 µM, and 500 µM) for 6 and 24 h (made up in supplemented DMEM/Ham’s F-12 medium with 0.5% FBS and 1% dimethyl sulfoxide (DMSO)) (Sigma-Aldrich, MO, USA). The control well contained only supplemented DMEM/Ham’s F-12 medium with 0.5% FBS and 1% DMSO (Fig. 1). Each experiment was conducted in five independent experiments and repeated twice.

Fig. 1.

Fig. 1

Experimental layout for FB1 and HFB1 exposure and downstream analysis. The schematic illustrates the experimental design used to assess the effects of FB1 and HFB1 exposure in IPEC-J2 cell line. Cells were treated with defined concentrations of FB1 and HFB1, followed by a series of downstream analyses

For the proteomics analysis, cells were seeded in supplemented DMEM/Ham’s F-12 with 10% FBS at the density of 1.5 × 106 cells per dish in 60-mm clear Petri dishes. Thereafter, cells were exposed to FB1 and HFB1 concentrations (7.81 µM and 15.63 µM) for a period of 24 h under standard incubation conditions (Fig. 1), as described above. Eight replicates were prepared per treatment, resulting in a total of 40 samples. Protein extraction and LC–MS/MS analysis were performed as described below, at the Centre for Proteomic and Genomic Research (CPGR, Cape Town, South Africa).

FB1 and HFB1 stock solution preparations

Purified and extracted FB1 (> 95%) and HFB1 (> 98%) (AMHBI, CPUT, SA) were dissolved in DMSO forming a stock solution of 1 mg/mL, respectively. The optimised model development of FB1 and HFB1 included concentrations from 0.98 to 500 µM, in supplemented DMEM/Ham’s F-12 medium with 0.5% FBS and 1% DMSO.

For proteomics, a 31.25 μM stock concentration of each toxin was made up in supplemented DMEM/Ham’s F-12 medium with 0.5% FBS and 1% DMSO. The respective stock solutions were further diluted into various concentrations, selected from the optimised model development (7.81 μM and 15.63 μM FB1; 7.81 μM and 15.63 μM HFB1). The cells were treated with these sub-cytotoxic levels, insignificantly impairing the overall cell viability (maximum decrease was 20%).

Cell viability indices and inflammation biomarkers

Determination of cell viability (ATP production)

The ATP content of the IPEC-J2 cell line was determined after FB1 and HFB1 exposure, using the CellTiter-Glo luminescent cell viability kit according to the manufacturer’s instructions (Promega Corporation, Madison, USA). This assay quantifies viable cells based on the amount of intracellular ATP, which is a marker of metabolically active cells. The reagent lyses cells and produces a luminescent signal proportional to ATP levels, indicating cell viability. The signal (expressed as relative light units (RLU)) was recorded using the Veritas microplate luminometer (Turner Biosystems, CA, USA), and data were expressed as a percentage of ATP content of control cells.

Determination of apoptosis

After FB1 and HFB1 exposure, cells were lysed with 100 μL of filtered 0.5% Triton X-100 (Roche, Mannheim, Germany) made up in Dulbecco’s phosphate buffered saline (DPBS) (Lonza, Basel, Switzerland) and stored at − 80 °C until analysis. Caspase-3 and caspase-7 activity was measured using the Caspase-Glo assay kit (Promega Corporation, Madison, USA) according to the manufacturer’s instructions. This luminescent assay detects caspase activity by using a proluminescent substrate that is cleaved by active caspase-3 and caspase-7, releasing a luciferase substrate and generating a luminescent signal proportional to enzyme activity, which indicates apoptosis. The luminescent signal (RLU) was recorded using the Veritas microplate luminometer (Turner Biosystems, CA, USA), and data was expressed as a fold increase relative to the control.

Determination of cell proliferation

The cell proliferation ELISA, 5′bromo-2′-deoxyuridine BrdU chemiluminescent assay (Roche, Mannheim, Germany), was used to observe the effect of FB1 and HFB1 on cell proliferation, according to the assay specifications. This assay measures DNA synthesis by detecting the incorporation of BrdU, a thymidine analogue, into newly synthesised DNA of proliferating cells using an anti-BrdU antibody and a chemiluminescent signal. In a solid black 96-well microtitre plate after FB1 and HFB1 exposure, chemiluminescence signal (RLU) was detected using the Veritas Microplate Luminometer (Turner Biosystems, CA, USA), and data expressed as a percentage of cell proliferation over controlled cells.

Porcine interleukin-8 ELISA

The IL-8 content in the cell supernatant was determined using a porcine IL-8/CXCL8 ELISA kit (R&D Systems, Minneapolis, USA) according to the manufacturer’s instructions. This enzyme-linked immunosorbent assay quantifies IL-8 levels based on the specific binding of antibodies to IL-8, followed by a colorimetric reaction proportional to the cytokine concentration in the sample. After FB1 and HFB1exposure, cell supernatants (100 μL) were transferred to clear 96-well microtitre plates and stored at − 80 °C until analysis. Absorbance was measured at 450 nm with the Synergy 2 Multimode Microplate Reader (Biotek, Vermont, USA) and data were analysed using the standard curve generated using Gen5 Data Analysis Software (Version 2 for Windows). Extracellular porcine IL-8 was expressed as pg/mL of cell supernatant and as the fold increase relative to the untreated control.

Statistical analyses

For the statistical analysis of FB1 and HFB1 concentrations, a one-way analysis of variance (ANOVA) was conducted to determine whether there were statistically significant differences among the experimental groups. This method assessed the overall group effects by comparing the means of each group. The data analysis was performed using GraphPad Prism (version 10.4). In order to further investigate which specific group differences contributed to any significant effects identified by the ANOVA, a post hoc Tukey–Kramer multiple comparison test was applied. This test allowed for pairwise comparisons between group means while controlling for type I error. Statistical significance was considered at a threshold of p < 0.05, indicating that differences between groups were deemed significant if the probability of such differences arising by chance was less than 5%.

Proteomics sample preparation, protein extraction and clean-up

Cells were prepared and treated as described above and harvested after 24-h exposure to FB1 and HFB1. Medium was discarded and cells were washed with HBSS, trypsinated, and transferred to Eppendorf tubes. The samples were centrifuged for 5 min (500 × g), and the supernatant was discarded. Subsequently, the pellet was washed, centrifuged, the supernatant discarded, and the final pellet placed on ice.

A SIMPLEX protein extraction method by Coman et al. (2016) was modified and used to extract proteins prior to proteomic analysis. Modifications include the addition of acetone (4:1, v/v) overnight instead of methanol (Labchem, Gauteng, SA) for 1 h to the remaining lower phase protein precipitation. Furthermore, protein samples were dissolved in 100 mM tetraethylammonium bromide buffer (TEAB) containing 4 M guanidine hydrochloride (Gu-HCL) and 1% octyl β-D-glucopyranoside (OGP) (Merk/Sigma-Aldrich, Johannesburg, SA). The samples were then placed in a sonicating water bath for 5 min at 4 °C. Cold methanol (800 μL) was added and vortexed for 20 s. This was followed by the addition of chloroform (200 μL) (Merk/Sigma-Aldrich, Johannesburg, SA) and molecular-grade water (600 μL). The samples were vortexed for 20 s after each solvent was added. Samples were then centrifuged at 12,000 × g for 10 min at 4 °C and the top aqueous layer was discarded. An additional 800 μL of methanol was added to the protein layer and vortexed. The samples were then centrifuged for 10 min (12,000 × g) at 4 °C, and the supernatant, discarded. The pellet was left to air dry on ice for 30 min, and samples were stored at − 20 °C until analysis.

Proteomic analysis

Protein solubilisation and quantification

Proteomics was conducted at D-CYPHR, an amalgamation of the proteomics facilitates from the CPGR and UCT IDM, supported by DIPLOMICS. The proteins were solubilised by adding 4% sodium dodecyl sulphate (SDS), 100 mM TEAB and heated to 95 °C for 10 min followed by centrifugation at 10,000 × g for 10 min and the supernatant transferred to a new tube. A sub-volume was removed for protein quantification using the bicinchoninic acid assay (BCA assay) (Merck/Sigma-Aldrich, Johannesburg, SA) according to the manufacturer’s instructions.

On-bead HILIC digest

The HILIC magnetic bead workflow was prepared by aliquoting HILIC beads (ReSyn Biosciences, Gauteng, SA) into a new tube and removing the shipping solution. The beads were then washed with 250 μL wash buffer containing 15% acetonitrile (ACN) (Burdick & Jackson, MI, USA) and 100 mM ammonium acetate (pH 4.5) (Merck/Sigma-Aldrich, Johannesburg, SA) for 1 min. The beads were washed again for a total of two washes and resuspended in a loading buffer consisting of 30% ACN and 200 mM ammonium acetate (pH 4.5), to a concentration of 2.5 mg/mL. A total of 20 μg from each sample was transferred to a protein LoBind plate. The protein was then reduced and alkylated by adding 20 mM dithiothreitol (DTT) and 30 mM iodoacetamide (IAA) (Merck/Sigma-Aldrich, Johannesburg, SA), which was followed by a 10-min incubation at 95 °C. Subsequently, HILIC magnetic beads were added at an equal volume to that of the sample at a ratio of 5:1 total protein. The plate was then incubated on a plate shaker (900 × g) for 30 min at room temperature to bind the proteins to the beads. Thereafter, the beads were washed with 500 μL 95% ACN for 1 min, four times. The protein was digested via the addition of trypsin made up in 50 mM TEAB at a ratio of 1:20 total protein and LysC protease (Pierce, MA, USA), added at a ratio of 1:250 total protein. The plate was then incubated on a plate shaker at 45 °C for 2 h. Subsequently, the supernatant containing peptides was removed and dried, and samples were then resuspended in liquid chromatography loading buffer containing 0.1% formic acid (FA) (Merck/Sigma-Aldrich, Johannesburg, SA) and 2.5% ACN.

Liquid chromatography-mass spectrometry

Liquid chromatography-mass spectrometry (LC–MS) analysis was conducted with a Q-Exacive quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific, MA, USA) coupled with a Dionex Ultimate 3000 nano-ultra-high performance liquid chromatography (UPLC) system. Mass spectrometry was performed using a data-dependent acquisition (DDA) method, specifically a Top10 approach (as indicated by the loop count of 10). The total cycle time was approximately 0.6 s, comprising a 100-ms injection time for the MS1 scan and ten MS2 scans at 50 ms each. Fragmentation was carried out using higher-energy collisional dissociation (HCD), with an AGC target value of 1e5. Additional MS/MS parameters included a resolution of 17,500 (at m/z 200), an isolation window width of 3 m/z, and a normalised collision energy (NCE) of 27%. Data-dependent settings included an underfill ratio of 1%, exclusion of unassigned charges and those with charge states 1, 7, 8, and > 8, peptide match set to ‘preferred’, isotope exclusion enabled, and a dynamic exclusion window of 60 s. The data was acquired using Xcalibur (version 4.1.31.9), Chromeleon (version 6.8- SR13), Orbitrap MS (version 2.9 and build 2926), and Thermo Foundations (version 3.1- SP4). The peptides were dissolved in 0.1% FA and 2% ACN. Thereafter, approximately 400 ng of peptide (injected per sample) was loaded on a C18 trap column (PepMap 100, 300 μM × 5 mm × 5 μM). The samples were then trapped into the column and washed for 3 min with 0.1% formic acid (FA) in LC–MS grade water before the valve was switched and peptides eluted into the analytical column. Chromatographic separation was performed with a Waters nanoEase (Zenfit) m/z peptide charged surface hybrid (CSH) C18 column (75 μM × 25 cm × 1.7 μM). The solvent system used was solvent A consisting of liquid chromatography water and 0.1% FA, and solvent B consisting of ACN and 0.1% FA. The multi-step gradient for peptide separation was generated at 300 nL/min as follows: time change 5 min, gradient change 2–5% solvent B, time change 40 min, gradient change 5–18% solvent B, time change 10 min, gradient change 18–30% solvent B, time change 2 min, gradient change 30–80% solvent B. The gradient was then held at 80% solvent B for 10 min before returning it to 2% solvent B for 15 min. All data acquisition was obtained using Proxeon (Theromo Fisher Scientific, MA, USA) stainless steel emitters. The mass spectrometer was operated in positive ion mode with a capillary temperature of 320 °C and the applied electrospray voltage was 1.95 kV.

Data analysis for protein identification

The raw data generated was searched against a porcine reference proteome (Pig_Refprot_49792_UP000008227_061221.fasta), downloaded from UniprotKB on 06/12/2021. The raw files were processed using Progenesis QI for Proteomics (Non-linear Dynamics, Newcastle upon Tyne, UK) software and the valid proteins (filtered results to remove reverse hits, common contaminants, and singly charged ions) containing at least two unique peptides were reported. A list of regulated proteins was generated using the same criteria as for valid proteins, with the additional requirement that their q-value be less than 0.05. Relative quantification was conducted using Progenesis QI for Proteomics (version 2.0.5556.29015) (Nonlinear Dynamics, Newcastle upon Tyne, UK). The data processing included peak picking, run alignment, and normalisation (singly charged spectra were removed from the processing pipeline). Label-free quantification (LFQ) was performed using Progenesis QI for Proteomics software (Version 2.0, Nonlinear Dynamics, UK) based on data-dependent acquisition (DDA) LC–MS/MS data. In DDA mode, the instrument acquires a full MS1 scan followed by MS/MS (MS2) scans of the most intense precursor ions selected in real time, enabling the identification of a broad range of peptides within a sample. To correct for run-to-run variability, normalisation was applied across samples, and only peptides with high-confidence identifications were included in the quantification process (Li et al. 2009). The data processing steps included peak picking, run alignment, and normalisation. Singly charged ions were excluded to minimise background noise and improve quantification accuracy. Protein quantification was performed using the ‘relative quantitation using non-conflicting peptides’ method, ensuring that only uniquely assigned peptides contributed to the protein-level measurements. The resulting normalised LFQ intensity values were subsequently used for downstream bioinformatics analyses (Cox et al. 2014). Database interrogation was performed with Byonic Software (version 3.8.13) (Protein Metrics, Cupertino, USA) using the Sus scrofa (pig) reference proteome mentioned above.

Bioinformatics analysis

The proteomic meta data obtained were further analysed by bioinformatics analysis to obtain valid proteins and pathways affected by FB1 and HFB1 exposure. The statistical analyses were performed in R (version 4.2.2) to assess the significance of the observed fold changes between FB1 and HFB1 exposures, and the control groups. Proteins were ranked by raw p-value, which was then adjusted for multiple comparisons using the Benjamini–Hochberg (BH) method. The BH procedure was applied to control the false discovery rate (FDR) across multiple hypothesis tests (Benjamini and Hochberg 1995). Two-sample t-tests were performed to compare protein abundance between each treatment and control. Significant proteins with adjusted p-values < 0.1 and fold-changes ≥ 1.3 or ≤ 0.7 were classified as differentially abundant proteins (DAPs). To visualise the DAPs, volcano plots with log2fold change values on the x-axis and − Log10 (BH adjusted p-values) on the y-axis were plotted using the Enhanced Volcano package in R (version 4.2.2).

To assess relative changes in protein abundance between treatment and control conditions, log₂ fold change (log₂FC) was calculated in R (version 2.2) using the formula: log2FC <—log2(Treatment_Mean/Control_Mean). This log transformation provides a symmetric scale for interpreting both upregulated and downregulated proteins, with positive values indicating upregulation and negative values indicating downregulation. Log₂FC values were then visualised using volcano plots together with BH adjusted p-values < 0.1 to identify statistically significant changes. A threshold of ± 0.405 (corresponding to a fold change 1.3) was applied to highlight biologically meaningful differences in either direction. For ease of interpretation, Log₂FC values were also converted back to linear fold change in R, using the inverse function: FoldChange <—2^(log2FC). This transformation allowed DAPs between treatment and control conditions to be expressed in intuitive fold-change terms, complementing the symmetric representation of the log₂ scale. The corresponding log₂FC values are presented in the supplementary Tables S1S3, and the actual fold changes are discussed in the main text. These steps supported data visualisation and downstream filtering, while preserving the biological interpretability of the observed changes.

Functional annotation and pathway analyses

Functional annotations for DAPs were carried out by Gene Ontology (GO) to assess the biological processes in R, while using the following packages: DESeq2 and AnnotationDbi packages which ran in the background, clusterProfiler tool package that performed the GO analysis, the org.Ss.eg.db package was used as the porcine database, specific to a certain type of genomic annotation or identifier mapping for Sus scrofa, the scientific name for the domestic pig.

GO and KEGG pathway analysis

Gene Ontology (GO) analysis was performed on the Cytoscape software for further evaluation of the functional enrichment attributes, to annotate the identified proteins in terms of biological process (BP), molecular function (MF), and cellular component (CP) of the identified DAPs. Similarly, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was performed to predict pathways based on the KEGG database. The GO and KEGG pathways were considered significantly enriched if BH adjusted p-values were < 0.05.

Analysis of the protein–protein interaction networks

The STRING (Search Tool for the Retrieval of Interacting Genes/Proteins) database (https://string-db.org) was utilised to retrieve the functional protein–protein interaction (PPI) networks of the identified significant DAPs from each data set separated into specific exposure concentrations (Szklarczyk et al., 2019). All significant DAPs were tested for functional enrichment and network analysis using STRINGApp (via Cytoscape) (Doncheva et al., 2018). Critically, the background set for enrichment was defined as all proteins quantified in the IPEC-J2 proteomic analysis (i.e. the complete list of detected proteins). STRING enrichment uses Fisher’s-exact against the custom input-derived background, which calculates the p-value and adjusted p-value using the BH method for correction of multiple hypotheses testing. The function categories and pathways with p-value < 0.05 were considered significant. Furthermore, additional customisation of the PPI network was developed in the Cytoscape software (version 3.10, http://www.cytoscape.org/) to recognise the target proteins by fold change and clustering methods. The top 50 DAPs with significant PPI were selected for further assessment.

Protein–protein interaction (PPI) networks were constructed using the STRINGApp (STRING database) within Cytoscape. Protein symbols corresponding to differentially abundant proteins (DAPs), meeting criteria of BH adjusted p-value < 0.1 and fold-change ≥ 1.3 or ≤ 0.7, were uploaded directly as input (Supplementary Tables S1S3). A STRING: protein query was used to request the networks, with a confidence (interaction score) cutoff ≥ 0.7. Resulting networks include both experimentally validated and predicted interactions, which were visualised and annotated in Cytoscape with interaction scores and fold-change mapping. Visualisation was customised based on fold-change values (node colour intensity) and connectivity (degree centrality), allowing identification of key hubs or clusters of proteins potentially responsible for cellular responses to FB1 and HFB1 exposure.

Results and discussion

The effect of FB1 and HFB1 exposure on cell viability

To establish a concentration-dependent response and select a sub-toxic dose for downstream proteomic analysis, this study first evaluated the effects of FB1 and HFB1 on IPEC-J2 cell viability across a range of concentrations and time points. This study demonstrated that exposure to FB1 at concentrations up to 500 µM did not result in a significant decrease in cell viability in IPEC-J2 cells after 6 (Fig. S1A and S1C) or 24 h (Figs. S1B and 2 A). Notably, FB1 at 1.95 µM, 15.63 µM, and 31.25 µM induced a slight but statistically significant increase in cell viability at 6 h (Fig. S1C). After 24 h, exposure to 0.98 µM and 7.81 µM FB1 caused a significant decrease in cell viability (Fig. 2A); however, viability remained above 80%. Studies performed by Chen et al. (2019) and Wan et al. (2013) showed that IPEC-J2 cells exposed to FB1 for 48 h experienced a significant decline in cell viability at concentrations above 25 µM and 40 µM. Other intestinal cell lines such as the porcine iliac artery endothelial cells were also sensitive to FB1 exposure and resulted in a significant decrease in cell viability after 48 h at 50 µg/mL (69 µM) FB1 exposure (Yuan et al. 2019). However, 24 h of exposure to various concentrations (1–70 µM) of FB1 did not affect cell viability in HT-29 cells and only exhibited a slight decline after 72 h at 70 µM FB1 exposure (Minervini et al. 2014). Similarly, many of the mentioned above studies also showed no effect on cell viability after 24 h of FB1 exposure. This suggests that the effect of FB1 on intestinal cells becomes more cytotoxic dose dependent as time progresses, consistent with findings by Wang et al. (2022), who reported no significant changes in cell viability after 24 h of exposure to 10, 20, and 40 µg/mL FB1, but observed a reduction in viability to 95.14% and 83.66% at 20 and 40 µg/mL, respectively, after 48 h.

Fig. 2.

Fig. 2

The effect of fumonisin B1 (FB1) and hydrolysed fumonisin B1 (HFB1) on (A) cell viability (ATP production); (B) apoptosis (caspase 3/7-fold increase); (C) cell proliferation (BrdU incorporation); and (D) an inflammatory biomarker (IL-8: interleukin 8 Conc: concentration) at 24-h incubation periods in IPEC-J2 porcine intestinal cells. ATP-adenosine triphosphate and BrdU production was calculated as a percentage and at a statistical significance of p < 0.05. Concentrations include a control and various concentrations of FB1 and HFB1 (ranging from 0.98 to 500 µM). Statistically significant differences (p < 0.05) are shown as lowercase letters above each of the graph bars where each concentration of FB1 and HFB1 are compared to the control

After 6 h of HFB1 exposure, cell viability was significantly increased at concentrations of 0.98 µM, 7.81 µM, 15.63 µM, 31.25 µM, and 250 µM (up to 140%), with a significant decrease observed only at the highest concentration of 500 µM; however, viability remained above 80% (Fig. S1A and S1C). After 24 h of HFB1 exposure, a significant decrease in cell viability was observed across concentrations from 0.98 to 62.5 µM, with no significant effect at 125 µM, a significant increase at 250 µM, and a pronounced decrease at 500 µM, where viability was reduced to approximately 40% (Figs. S1B and 2 A). The increase in cell viability at certain concentrations relates to a phenomenon whereby stressed or dying cells release or expose molecules called damage-associated molecular patterns (DAMPs), such as ATP and amphotericin on their surface. In return, it signals the innate immune system and increases inflammation. Extracellular ATP specifically is often released from apoptotic cells where the secretion is associated with the apoptotic stage and type of stress or cell death stimulus (Krysko et al. 2012).

The effect of FB1 and HFB1 exposure on apoptosis

Apoptosis was evaluated in response to FB1 and HFB1 exposure to investigate its potential role in mediating cytotoxic effects and cellular damage, as programmed cell death is a key mechanism underlying toxin-induced tissue injury. An increase in apoptosis was observed in IPEC-J2 cells exposed to 125 µM, 250 µM, and 500 µM FB1 after both 6 and 24 h (Fig. S2A and S2B). At lower concentrations, no significant effect on apoptosis was detected after 6 h; however, after 24 h, a decrease in apoptosis was noted at 0.98 µM, 1.95 µM, 3.91 µM, and 7.81 µM compared to control cells (Figs. S2C and 2B). This suggests that cells were undergoing apoptosis from 125 µM exposure independent of the exposure period. Wang et al. (2022) observed a similar finding in IPEC-J2 cells that displayed caspase-3 and caspase-9 cleavage and expression after 48 h from 10 µM up to 40 µg/mL (55 µM) exposure. After 6 h of HFB1 exposure, a reduction in apoptosis was observed at 0.98 µM and 3.91 µM compared to control cells, while significant induction of apoptosis occurred at higher concentrations ranging from 62.5 to 500 µM. (Fig. S2A and S2C). The apoptotic response after 24 h of 31.25, 62.51, 125, and 250 µM HFB1 exposure exceeded that of 6 h (Figs. S2B and 2B). The rapid decrease in apoptosis at concentrations following 250 µM HFB1 after 24 h (Fig. S2B) could be attributed to depleted cell populations in advanced stages of apoptosis. Additionally, cells may also be undergoing a rapid onset of cellular necrosis after a high concentration of HFB1 exposure and may not have enough time or energy to initiate apoptotic mechanisms which will therefore not express apoptotic indicators, resulting in lowered apoptosis (Istifli et al. 2019).

The effect of FB1 and HFB1 exposure on cell proliferation

To further assess the impact of FB1 and HFB1 on cellular growth, the effect of exposure to these compounds on cell proliferation was investigated in IPEC-J2 cells. Cell proliferation was not significantly affected after 6 h of exposure to most FB1 concentrations, except for a slight inhibitory effect observed at 0.98, 15.36, and 62.5 µM (Fig. S3A and S3C). After 24 h of FB1 exposure, concentrations above 31.25 µM resulted in a dose-dependent inhibition of cell proliferation, with greater than 50% inhibition observed at 500 µM (Figs. 2C and S3B). Cell proliferation significantly declined at HFB1 concentrations above 62.5 µM following 6 h of exposure (Fig. S3A and S3C), and approximately 90% inhibition at 250 µM and 500 µM. After 24 h of HFB1 exposure, cell proliferation significantly declined with an initial decrease observed at 1.98 µM, further inhibition from 15.63 µM, and approximately 90% or greater inhibition at concentrations of 31.25 µM and above (Figs. 2C and S3B). This suggests that, like FB1, prolonged exposure of HFB1 promotes dose- and time-dependent cytotoxicity. It was observed that after 48-h exposure to FB1 concentrations of 25 µg/mL (35 µM) and above, elicits injury to porcine epithelial cells by impeding cell proliferation (Chen et al. 2019). HT-29 cells exposed to FB1 concentrations of 8.6 µM and higher also induced a dose-dependent inhibition of cell proliferation after 72 h of exposure (Minervini et al. 2014).

The effect of FB1 and HFB1 exposure on IL-8 concentration

The effect of FB1 and HFB1 exposure on IL-8 concentration was investigated to assess the inflammatory response elicited by these mycotoxins in IPEC-J2 cells. No significant effect on IL-8 concentration was observed at any FB1 concentration following 6 h of exposure (Fig. S4A and S4C). A significant decrease in IL-8 concentration was observed following 125 µM, 250 µM, and 500 µM HFB1 exposure after 6 h (Fig. S4A and S4C). A significant decrease in IL-8 concentration was observed following FB1 exposure at concentrations ranging from 0.98 to 31.25 µM after 24-h exposure, whereas no significant difference was detected at 62.5 µM and higher (Figs. 2D and S4B). HFB1 exposure, on the other hand, showed no significant effect on IL-8 concentration at lower concentrations (0.98 to 7.81 µM) after 24 h, whereas concentrations of 15.63 µM and above induced a significant decrease (Figs. 2D and S4B). The decrease in IL-8 correlates with a decrease in cell viability and cell proliferation, and an increase in apoptosis. Interleukin-8 (IL-8) is known as a cell proliferation enhancing cytokine, whereby inflammation is promoted and the control of repair processes during intestinal mucosal injury or cytotoxic stress occurs (Wilson et al. 1999). A study performed using IPEC-1 cells observed a dose-dependent decrease in IL-8 concentration at mRNA and protein levels after 4 days with FB1 concentrations up to 72.2 µM (Bouhet et al. 2006). Another study observed no IL-8 response in HT-29 cells after 48-h FB1 exposure (Minervini et al. 2014). The current study shows a decrease of IL-8 at all concentrations up to 31.25 µM FB1 exposure, which was also observed following 62.5 µM FB1 exposure after 24 h (Fig. 2D). These observations coupled with cell proliferation depict that IL-8 was secreted upon FB1 exposure beyond 31.25 µM (24 h), affecting an inflammatory response. In an in vivo study performed by Grenier et al. (2012), pigs were fed a HFB1-containing feed for 14 days and the study reported that the proximal section of the intestine (where IPEC-J2 cells originate from) produced a higher concentration of IL-8 compared to FB1. In the current study, a similar trend was observed at concentrations up to 7.81 µM HFB1 after 24 h of exposure (Fig. 2D). However, an increase in IL-8 promotes a decrease in the cell survival rate (Qazi et al. 2011). This suggests that HFB1 may be metabolised differently in the gut in vivo compared to in vitro, where systemic factors such as host enzyme activity or gut microbiota could modify HFB1’s biological activity, resulting in immune responses that are not replicated in isolated epithelial cell models.

Identification of differentially abundant proteins by liquid chromatography-mass spectrometry

To investigate the molecular mechanisms and pathways of proteins influenced by FB1 and HFB1 on the IPEC-J2 cell line, a comparative proteomic analysis was performed. Proteins are the foundation of cellular functions, whereby alterations of key proteins would directly affect cellular outputs (McArdle and Menikou 2020). Cells were exposed to 7.81 µM and 15.63 µM FB1 and HFB1, respectively, for proteomic analysis. These concentrations were selected as they are sub-cytotoxic, as demonstrated by prior assessments of cell viability (Fig. 2A) and caspase-3/7 activity (Fig. 2B), which showed no significant loss in viability or induction of apoptosis at these levels. Specifically, these concentrations represent the highest doses at which no statistically significant cytotoxic or apoptotic effects were observed, thereby allowing for the detection of early molecular responses without confounding cell death-related effects. This approach ensures that the observed proteomic alterations are reflective of subtle cellular stress or early-stage responses to FB1 and HFB1 exposure, rather than downstream effects of overt toxicity. The intracellular proteins of cells exposed to FB1 and HFB1 as well as the untreated control group were quantified by LC–MS/MS methods. The proteins with a fold change ≥ 1.3 and ≤ 0.7 (mean value of all compared groups) and p-value (t-test of all comparison groups) < 0.1 by comparing each treated group to the control group were defined as the significantly differentially abundant proteins (DAPs) (Table 1). Differential expression analysis of the protein datasets generated the lists of significant DAPs, fulfilling the selection criteria of fold change ≥ 1.3 and ≤ 0.7, and BH adjusted p-value < 0.1. Fold change thresholds ≥ 1.3 and ≤ 0.7 for the selected upregulated and downregulated proteins, respectively, were selected in this study because the more commonly applied FC threshold of 1.5-fold was not sensitive enough. The application of FC threshold for this study permitted identification of a larger number of proteins whose differential expression from controls was statistically significant, allowing a better opportunity to identify enriched pathways. The selected threshold is less stringent and may include moderate changes; however, this cut-off was chosen based on precedent in similar proteomic analyses where moderate changes were also of interest, to balance sensitivity and specificity in detecting biologically relevant protein alterations (Black et al. 2021; Li et al. 2024).

Table 1.

The top 52 DAPs between FB1 and HFB1 compared to the control. Fold change (FC) values in bold represent upregulated proteins > 1.3; values ≥ 1.3 indicate upregulation; and values ≤ 0.7 indicate downregulation

Protein symbol Protein name HFB1 (7.81 µM) HFB1 (15.63 µM) FB1 (15.63 µM)
Adjusted p-value FC Adjusted p-value FC Adjusted p-value FC
1 CDK6 Cyclin-dependent kinase 6 0.056 1.637
2 ARFGEF1 ADP ribosylation factor guanine nucleotide 0.047 1.581
3 ACTN2 Actinin alpha 2 0.081 1.531
4 FTL Ferritin light chain 0.015 1.500 0.003 1.496
5 CEP192 Centrosomal protein 0.057 1.458
6 VEZT Vezatin, adherens junctions transmembrane protein 0.075 1.424
7 MRPL18 Mitochondrial ribosomal protein 0.066 1.416
8 TBC1D23 TBC1 domain 0.077 1.377
9 GCA Grancalcin 0.059 1.370
10 PGPEP1 Pyroglutamyl-peptidase I 0.046 1.359
11 GOLT1B Golgi transport 1B 0.083 1.355
12 FTH1 Ferritin heavy chain 0.031 1.350 0.004 1.535
13 PDCD10 Programmed cell death protein 0.047 1.347
14 MRPL28 Mitochondrial ribosomal protein 0.049 1.343
15 ABRACL ABRA C-terminal like 0.072 1.337
16 RPS21 40S ribosomal protein S21 0.057 0.754
17 CYB5B Cytochrome b5 heme-binding 0.071 0.752
18 NFU1 NFU1 iron-sulphur cluster 0.058 0.745
19 SKP1 S-phase kinase 0.082 0.740
20 CD44 CD44 molecule 0.058 0.732
21 WDR48 WD repeat domain 0.045 0.724
22 TINAGL1 Tubulointerstitial nephritis antigen like 1 0.004 0.716 0.032 0.739
23 AGRN Agrin 0.055 0.699
24 NTN4 Netrin 4 0.048 0.698 0.001 0.549
25 ATP5F1D ATP synthase subunit delta 0.045 0.694
26 RPS28 40S ribosomal protein 0.028 0.690
27 S100A11 Protein S100-A11 0.050 0.683
28 SRBD1 S1 RNA binding domain 1 0.032 0.677
29 SH3BGRL3 SH3 domain-binding glutamic acid 0.048 0.663
30 FN1 Fibronectin 1 0.003 0.647 0.000 0.585 0.049 1.371
31 PPP1R2 Protein phosphatase 1 regulatory inhibitor subunit 0.087 0.642
32 INTS3 Integrator complex subunit 3 0.094 0.639
33 SDC4 Syndecan-4 0.005 0.629 0.001 0.623
34 ST14 Suppressor of tumorigenicity 14 0.006 0.613 0.004 0.586
35 CLNS1A Chloride nucleotide-sensitive 0.044 0.607
36 SPARC Secreted protein acidic and cysteine rich 0.054 0.605 0.093 0.575
37 CD276 CD276 molecule 0.073 0.599 0.052 1.469
38 FBLN2 Fibulin-2 0.003 0.573 0.000 0.547
39 CHCHD2 Coiled-coil-helix 0.055 0.469
40 CCN3 Cellular communication 0.078 0.433
41 RBP4 Retinol-binding protein 0.056 1.707
42 TGFBR2 TGF-beta receptor type-2 0.053 1.378
43 EHD2 GTPase superfamily 0.006 1.335
44 ATF3 Cyclic AMP-dependent transcription factor ATF-3 0.001 1.368
45 LAMC1 Laminin subunit gamma-1 0.032 0.753
46 CXADR CXADR Ig-like cell adhesion molecule 0.013 0.750
47 QSOX1 Quiescin sulfhydryl oxidase 0.000 0.727
48 CCN1 Cellular communication 0.006 0.723
49 MET Hepatocyte growth factor receptor 0.053 0.672
50 ITM2B Integral membrane protein 2B 0.033 0.666
51 TFRC Transferrin 0.000 0.628
52 CDH6 Cadherin 6 0.070 0.447

The list of total DAPs along with the FC values and BH-adjusted p-values are provided in Table 1. These DAPs for all datasets are graphically represented using the volcano plots in Fig. 3A–D.

Fig. 3.

Fig. 3

Fig. 3

Effect of HFB1 (A, B) and FB1 (C, D) on DAPs, both at the different concentrations 7.81 µM (represented as HFB1_7 and FB1_7) and 15.63 µM (represented as HFB1_15 and FB1_15), in the intestinal porcine cell line IPEC-J2, respectively. The colours indicate threshold of significant proteins which contain BH adjusted p-values ≤ 0.1, and fold-change ≥ 1.3 or ≤ 0.7. Significant downregulated proteins are shown in blue, upregulated are shown in red, whereas non-significant proteins are shown in grey

A total of 52 DAPs were generated from FB1- and HFB1-exposed cells compared to the control (unexposed cells) (Table 1). A total of 40 DAPs were identified after 7.81 µM HFB1 exposure, comprising 15 upregulated and 25 downregulated proteins. In contrast, exposure to 15.63 µM HFB1 resulted in the upregulation of 3 proteins and downregulation of 15 proteins (Table 1; Fig. 3A and 3B). Cells that were exposed to 15.63 µM FB1 resulted in five upregulated DAPs and none that were considered significant following 7.81 µM FB1 exposure (Table 1; Fig. 3C and 3D).

The Venn diagram (Fig. 4) illustrates the overlap and uniqueness of DAPs identified under three exposure conditions: 7.81 µM HFB1, 15.63 µM HFB1, and 15.63 µM FB1. A total of 40, 18, and 5 DAPs were identified in the 7.81 µM HFB1, 15.63 µM HFB1, and 15.63 µM FB1 groups, respectively. FN1 was the only DAP common to all three conditions, suggesting its potential role as a universal biomarker or core responder to both compounds. Eight additional proteins (FBLN2, TINAGL1, SDC4, ST14, FTL, FTH1, NTN4, and SPARC) were shared between the two HFB1 concentrations, indicating a conserved proteomic response specific to HFB1 exposure. In contrast, FB1 showed minimal overlap with HFB1, sharing only FN1 and CD276, and displayed a largely distinct DAP profile. The highest number of unique proteins was observed in the 7.81 µM HFB1 group, suggesting a broader cellular response at this concentration.

Fig. 4.

Fig. 4

Venn diagram of differentially abundant proteins (DAPs) identified in 7.81 µM HFB1 (represented as HFB1_7), 15.63 µM HFB1, and 15.63 µM FB1 represented as HFB1_15 and FB1_15) exposure groups

Overall, HFB1 exposure resulted in a more extensive alteration of the cellular proteome compared to FB1 under the tested in vitro conditions. A subset of proteins commonly regulated by both FB1 and HFB1 will be discussed in detail, while unique proteins associated with specific concentrations will be briefly highlighted.

Functional enrichment analysis of DAPs in FB1 and HFB1 related to Gene Ontology

The functional enrichment analysis of differentially abundant proteins (DAPs), performed using the porcine genome annotation database, identified distinct biological processes that appear to be altered in response to HFB1 exposure. The STRING enrichment analysis was performed using the entire Sus scrofa proteome as the background database. While this is the standard approach in many studies, it may introduce bias due to differences between the specific cell line proteome and the broader species proteome. Ideally, a cell line–specific proteome should be used as a control to better reflect the expressed protein background and improve the accuracy of enrichment results. However, such a database was not available for the cell line used in this study. Therefore, the identified enrichments should be interpreted with caution, as some observed alterations may reflect general differences between the cell line and the species-level proteome rather than specific biological changes. No significantly downregulated DAPs were found following exposure to 15.63 µM FB1, and no significant Gene Ontology (GO) terms were enriched for FB1-treated cells at either concentration. In contrast, exposure to 7.81 µM HFB1 led to the downregulation of proteins associated with Golgi apparatus function and ribosomal processes, while 15.63 µM HFB1 downregulated proteins involved in ribonucleoside monophosphate biosynthesis, metabolism, and negative regulation of the ERK1/ERK2 cascade (Supplementary Fig. S5). These processes are fundamental to protein synthesis, intracellular transport, and signalling regulation, suggesting that HFB1 impairs essential cellular functions. Furthermore, HFB1 exposure at both concentrations upregulated proteins associated with extracellular matrix (ECM) binding, protein complex binding, and morphogenesis of branching epithelium. These GO terms are typically linked to tissue remodelling and structural reorganisation. Based on these associations, the enrichment of such processes points towards indicative of an adaptive cellular response to HFB1-induced stress, potentially involving cytoskeletal rearrangement or ECM remodelling. The absence of enriched GO terms in the FB1 groups emphasises a comparatively lower degree of pathway-level disruption under the tested conditions.

Functional enrichment analysis of DAPs in FB1 and HFB1 related to KEGG pathways

The KEGG pathway enrichment analysis (Fig. 5) illustrates the most significantly affected pathways in response to different treatments based on adjusted p-values.

Fig. 5.

Fig. 5

Enrichment analyses of KEGG pathways of the significant enriched proteins associated with 15.63 µM FB1, 15.63 µM HFB1 (represented as FB1_15 and HFB1_15) and 7.81 µM HFB1 (represented as HFB1_7) exposed IPEC-J2 cells. All terms are ranked by adjusted p-value

KEGG pathway enrichment analysis revealed differential modulation of several signalling pathways across treatment groups. In the FB1 (15.63 μM) exposure group, pathways associated with inflammation, cell adhesion, and cancer progression, such as ECM-receptor interaction, proteoglycans in cancer, and the AGE-RAGE signalling pathway, were significantly enriched (green bars). The HFB1 (15.63 μM) exposure also showed enrichment in cancer-related and immune signalling pathways, including the PI3K-Akt signalling pathway, focal adhesion, and phagosome pathways (blue bars). Notably, the HFB1 (7.81 μM) exposed group demonstrated a more focused enrichment pattern, with significant involvement of ribosomal activity and cancer-associated ECM-receptor interactions (pink bars). These findings suggest dose- and formulation-dependent modulation of key cellular pathways involved in inflammation, adhesion, and tumorigenesis.

Shared DAPs in IPEC-J2 cells after FB1 and HFB1 exposure

Among all concentrations, Fibronectin 1 (FN1) was the only protein consistently affected by exposure to 15.63 µM FB1 and both concentrations of HFB1(Table 1; Fig. 4). FN1 is crucial for cellular integrity, wound healing, and intestinal epithelial response to injury (Kolachala et al. 2007; Niederlechner et al. 2012; Sun et al. 2020). In previous studies, FN1 levels were reduced during intestinal injury, correlating with increased apoptosis (Niederlechner et al. 2012). Similarly, in our study, FB1 upregulated FN1, while HFB1 downregulated it, indicating that HFB1 exposure may induce more severe intestinal injury. FN1 has also been linked to tumour progression and poor prognosis in various cancers (Cai et al. 2018; Lou et al. 2013; Nakagawa et al. 2014; Sponziello et al. 2016; Sun et al. 2020; Waalkes et al. 2010; Wang et al. 2017; Xiao et al. 2018), and its upregulation in FB1-exposed cells may explain the carcinogenic effects observed in animal models (Gelderblom et al. 1993). Our findings align with previous reports demonstrating the oncogenic role of FN1, which was shown to be upregulated in gastric cancer (GC) tissues and cell lines, correlating with increased tumour invasion, advanced TNM stage, lymph node metastasis, and poor patient prognosis (Zhou et al. 2020). In this study, FB1 exposure led to an upregulation of FN1, potentially supporting its pro-tumorigenic effect, while HFB1 exposure resulted in FN1 downregulation, suggesting a divergent and possibly less oncogenic cellular response. This contrasting regulation highlights a key molecular difference between FB1 and HFB1 that may underlie their distinct biological effects.

Proteomic analysis additionally revealed one shared DAP following FB1 and 7.81 µM HFB1 exposure that are known to be involved in cellular processes relevant to cancer biology. CD276 (B7-H3), an immune checkpoint molecule, was upregulated following 15.63 µM FB1 exposure (Table 1; Fig. 4). Elevated CD276 expression has been associated with immune evasion and poor cancer prognosis (Liu et al. 2021), suggesting that FB1 may influence immune modulation. Conversely, CD276 was downregulated following 7.81 µM HFB1 exposure, potentially indicating a different cellular response.

Distinct upregulated and downregulated DAPs in FB1 and HFB1 exposure

Despite the two shared DAPs, several distinct DAPs were uniquely affected by either FB1 or HFB1, indicating compound-specific proteomic responses. The distinct upregulated proteins at 15.63 µM FB1 included retinol-binding protein 4 (RBP4), TGF-β receptor type 2 (TGFBR2), and GTPase superfamily (EHD2) (Table 1; Fig. 4). RBP4, a protein involved in vitamin A transport (Perduca et al. 2018), was significantly upregulated following FB1 exposure. Dysregulation of RBP4 has been linked to metabolic and inflammatory disturbances in the liver, pancreas, and vascular system, which may relate to the known adverse effects of FB1 on these organs. TGFBR2, a key receptor in the TGF-β signalling pathway that regulates cell proliferation and apoptosis (Fynan and Reiss 1993; Kim 2001; Markowitz and Roberts 1996), was upregulated by FB1. While normally acting as a tumour suppressor, dysregulated TGFBR2 expression in hepatocellular carcinoma (HCC) has been associated with enhanced tumour progression through mechanisms such as epithelial-to-mesenchymal transition and metastasis (Hao et al. 2019). Similarly, EHD2, involved in membrane remodelling and lipid homeostasis (Shah et al. 2014; Simone et al. 2014; Zhang et al. 2022), was also upregulated by FB1, potentially contributing to HCC development by promoting altered lipid metabolism and membrane dynamics that facilitate cancer cell survival, motility, and invasiveness. Together, the increased expression of TGFBR2 and EHD2 may reflect pathways implicated in HCC pathogenesis.

Between 7.81 µM and 15.63 µM HFB1, a total of eight DAPs were identified, of which two were upregulated and six were downregulated (Table 1; Fig. 4). The upregulation of ferritin light (FTL) and heavy chains (FTH1) after exposure to both concentrations of HFB1 suggests altered iron homeostasis, which has been linked to increased oxidative stress and tumour-promoting environments in some contexts (Grazi et al. 1995). The downregulation of netrin-4 (NTN4), a molecule implicated in cell adhesion and migration (Esseghir et al. 2007; Latil et al. 2003), and ST14, a serine protease with known tumour suppressor functions in the gastrointestinal tract (Kosa et al. 2012; Danielsen et al. 2018), further suggests HFB1 may affect pathways involved in epithelial integrity and regulation. The downregulation of SPARC by HFB1, a matricellular protein associated with tissue remodelling and tumour progression (Brabender et al. 2003; Framson and Sage 2004; Schiemann et al. 2003; Takemasa et al. 2001; Wang et al. 2004), may reflect disrupted extracellular matrix dynamics. Similarly, decreased expression of TINAGL1, FBLN2, and SDC4 suggests potential perturbations of HFB1 in angiogenesis and wound-healing pathways (Bernfield et al. 1992; Pap and Bertrand 2013). These protein changes do not imply direct oncogenic effects of HFB1, but may represent early alterations in cell signalling, stress response, or extracellular matrix interaction that warrant further investigation.

Exposure to 73.81 µM HFB1 resulted in 30 distinct significantly regulated DAPs, including both upregulated and downregulated proteins (Table 1; Fig. 4). The most upregulated proteins following exposure to 7.81 µM HFB1 were CDK6, ARFGEF1, and ACTN2, suggesting an early cellular stress response affecting cell cycle regulation, membrane trafficking, and cytoskeletal dynamics. CDK6, a key regulator of the G1 phase of the cell cycle, has been implicated in DNA damage and apoptotic signalling (Chu et al. 2020; Scheicher et al. 2015). The upregulation of ARFGEF1, a protein involved in maintaining Golgi structure and vesicle trafficking, may reflect disruptions in intracellular transport and neuronal function (Shen et al. 2007; Xu et al. 2022; Zhou et al. 2013). ACTN2, primarily expressed in muscle tissue but also linked to integrin-mediated signalling pathways, is involved in actin filament organisation and cell motility (Beggs et al. 1992; Burridge and McCullough 1980; Lindholm et al. 2021). Its increased expression may indicate cytoskeletal rearrangement and integrin activation. Collectively, the upregulation of these proteins may represent a coordinated cellular response to HFB1 exposure, potentially involving alterations in cell cycle control, membrane integrity, and cytoskeletal function. During exposure to 15.63 µM HFB1, ATF3 was significantly upregulated, suggesting activation in response to cellular stress signals such as DNA damage and oxidative stress, which may contribute to the observed increase in cell death and inflammation (Hai et al. 1999, 2010; Zhou et al. 2017).

At 7.81 µM HFB1, significantly downregulated proteins included CCN3 and CHCHD2 (Table 1; Fig. 4). CCN3 is involved in angiogenesis and inflammation (Jun and Lau 2011), while CHCHD2 regulates mitochondrial function and apoptosis, potentially contributing to the observed increase in apoptotic activity (Akiyama et al. 2017; Liu et al. 2015). At 15.63 µM HFB1, CDH6, TFRC, and ITM2B were significantly downregulated. CDH6, a cadherin glycoprotein involved in morphogenesis of the CNS and kidneys (Cho et al. 1998; Zhao et al. 2021), may be linked to tumour progression when dysregulated. ITM2B, known for its role in apoptosis and neurodegenerative disorders, may reflect impaired apoptotic signalling or cancer-related pathways (Baron and Pytel 2017; Xian et al. 2024). Downregulation of TFRC, essential for iron uptake and DNA synthesis, suggests disrupted iron homeostasis, which has been associated with enhanced tumour progression and altered cell metabolism (Gammella et al. 2017; Forciniti et al. 2020; Marchese et al. 2016). Together, these changes in protein expression indicate that HFB1 may disrupt key cellular processes, including apoptosis, iron regulation, and developmental signalling, that contribute to cytotoxicity and potentially initiate or promote tumourigenic pathways.

Protein–protein interaction (PPI) network and pathway analysis of DAPs in FB1 and HFB1

To identify key proteins involved in the cellular response to FB1 and HFB1 exposure, protein–protein interaction (PPI) networks were constructed using the STRING database based on the DAPs identified following exposure of the cells. A high-confidence threshold (score ≥ 0.7) was applied to select PPIs, and the data were exported to Cytoscape for further analysis. The selection criteria were based on datasets originating from Sus scrofa, the porcine model, which is relevant to the context of the study.

By identifying common proteins across the different exposure concentrations, we were able to highlight both shared and unique pathways affected by FB1 and HFB1. In particular, FN1 was upregulated following exposure to 15.63 µM FB1. Using the STRING database, it was observed that FN1 has strong predicted interactions with several integrin subunits, including integrin subunit alpha 5 (ITGA5), integrin subunit alpha V (ITGAV), integrin subunit beta 1 (ITGB1), and integrin subunit beta 3 (ITGB3) (Fig. 6). Although these interactions were not directly identified in our experimental dataset, they provide mechanistic context to the observed FN1 upregulation. This supports a possible involvement of integrins in promoting cellular adhesion, migration, and differentiation. Furthermore, this aligns with previous studies reporting that integrin-FN1 signalling plays a pivotal role in tumour progression, particularly in epithelial cancers (Deng et al. 2019; Li et al. 2014; Desgrosellier and Cheresh 2010).

Fig. 6.

Fig. 6

PPI network of 15.63 µM FB1 to determine the signalling pathways of the five significant upregulated proteins in darker green to purple. The PPI network consisted of 15 nodes and 32 edges

FB1 exposure enriched cancer-related pathways, such as proteoglycans in cancer and ECM-receptor interaction, which are implicated in cellular signalling, adhesion, and tissue remodelling (Desgrosellier and Cheresh 2010). The integrin-related pathways induced by FB1 exposure suggest its potential role in tumour progression, angiogenesis, and cell migration via transforming growth factor-beta (TGF-β) signalling (Mostafavi-Pour et al. 2018; Ren et al. 2009). Furthermore, our results are consistent with previous studies indicating that FB1 exposure affects the matrix cell organisation, as observed in the jejunum through integrin and actin pathways (Dopavogui et al. 2022). The findings suggest that FB1 exposure contributes to tumour progression by promoting these key cellular processes, including inflammation and the suppression of cell proliferation.

In contrast, HFB1 exposure at 7.81 µM resulted in the downregulation of FN1 and revealed strong interactions with various syndecan and collagen pathways. Specifically, proteins such as syndecan-1 (SDC1), syndecan-2 (SDC2), syndecan-4 (SDC4), and collagen type 1 alpha 1 chain (COL1A1) were influenced by HFB1 exposure, which enriched the proteoglycans in cancer and ECM-receptor interaction pathways (Fig. 7). Syndecans are heparan sulphate proteoglycans involved in cellular adhesion, migration, and proliferation, and they are associated with colorectal cancer progression (Han et al. 2004; Park et al. 2002). In particular, SDC4 has been linked to ERK pathway activation, driving cellular proliferation (Chua et al. 2004; Corti et al. 2013). Collagen proteins, including COL1A1, are often mutated or altered during tumour progression to facilitate cellular invasion and metastasis (Xu et al. 2019). The disruption of collagen and syndecan pathways suggests that HFB1 exposure promotes cancer progression by modulating these pathways, particularly during cellular proliferation, migration, and adhesion processes. Although only a limited number of the identified DAPs were central within the PPI network, enrichment analysis revealed associations with cancer-related pathways such as proteoglycans in cancer and ECM-receptor interaction. Proteins like FN1 and COL1A1, despite representing a subset of the network, are known to play critical roles in cell adhesion, migration, and extracellular matrix remodelling, processes relevant to tumour progression. These findings suggest that even modest perturbations in key ECM-associated proteins may influence cancer-related signalling events.

Fig. 7.

Fig. 7

To determine the signalling pathways of the identified 40 DAPs: PPI network of 7.81 µM HFB1. The PPI network was constructed using string database and customised in Cytoscape and, where each node represents a protein, and edges represent the interaction between proteins. The PPI network consisted of 38 nodes and 85 edges. Downregulated DAPs in Sus scrofa intestinal cell line (IPEC-J2) are shown in yellow whereas the upregulated are shown in dark green-purple, other predicted proteins shown in grey. Interactions were predicted with a high confidence level of 0.700 and enrichment p-value < 0.05 was included in the analyses

Unlike FB1, which primarily affects integrin pathways, HFB1 exposure engages alternative cancer-promoting mechanisms, potentially explaining the differential toxicity observed in vitro versus in vivo studies. The present study suggests that HFB1 toxicity may vary depending on its concentration and the specific pathways it influences. At the higher concentration (15.63 µM), HFB1 exposure led to a combination of integrin and syndecan pathway interactions, resulting in a broader range of cancer-promoting effects. Cytoscape analysis revealed interactions between FN1 and integrins (ITGA5, ITGB1), syndecans (SDC4), fibulin (FBLN2), and laminin subunit gamma 1 (LAMC1), all of which are involved in potent cancer-promoting pathways (Fig. 8). This enrichment pointed to the PI3K-Akt signalling pathway, wound healing, and additional cancer-related pathways, highlighting the complex and multifaceted impact of HFB1 at higher concentrations.

Fig. 8.

Fig. 8

PPI network of 15.63 µM HFB1, to determine the signalling pathways of the 18 significant proteins identified; significant upregulated proteins in darker purple; whilst downregulated proteins shown in yellow. The PPI network consisted of 23 nodes and 26 edges

FBLN2, which is overexpressed in lung adenocarcinoma cell lines (Baird et al. 2013), interacts with the ECM through integrin, collagen, and laminin pathways to support cellular proliferation. The increased expression of LAMC1, associated with TGF-β signalling, has been linked to tumour progression and inflammatory microenvironments (Fang et al. 2021; Taniguchi and Karin 2018). These findings provide further evidence of the enhanced toxicity of HFB1 at higher concentrations, as it influences a broader array of cancer-related pathways compared to FB1.

Conclusion

In summary, exposure to different concentrations of mycotoxins resulted in distinct protein interactions and enriched pathways, shedding light on the mechanistic differences between FB1 and HFB1 toxicity. The identification of FN1 regulation, along with the associated cancer-related pathways, offers potential mechanistic insights into the cellular responses to FB1 and HFB1. However, these findings cannot fully explain the differences reported between in vitro and in vivo outcomes, and further studies using more physiologically relevant models are needed to confirm these effects. The results from both the cell optimisation model and the proteomics analysis indicate that HFB1 exerts greater toxicity on the IPEC-J2 cell line, owing to the increased number of affected proteins and enriched pathways. This underscores the importance of considering mycotoxin concentration and the variety of molecular pathways involved when assessing their potential toxicity in intestinal cells. This study provides mechanistic insight into the cellular responses induced by FB1 and HFB1 exposure; however, the limitations of the in vitro model must be acknowledged. The absence of systemic interactions, metabolic activity, and immune modulation inherent to in vivo systems may influence the physiological relevance of the observed effects. Consequently, the findings should be interpreted within the context of the simplified cellular environment. Further in vivo investigations or advanced in vitro models (e.g. co-culture or organ-on-chip systems) are warranted to validate these results and better reflect the biological complexity of whole-organism exposure.

Supplementary Information

Below is the link to the electronic supplementary material.

ESM1 (453.9KB, docx)

(DOCX 453 KB)

Acknowledgements

This research was supported by funding from the Cape Peninsula University of Technology (CPUT). Additionally, we acknowledge the National Research Foundation (NRF) for providing a Postgraduate bursary to support this study. We gratefully acknowledge Dr Elisabeth Mayer (dsm-firmenich; Austria) for providing the IPEC-J2 cell line. We also extend our appreciation to D-CYPHR, an amalgamation of the proteomics facilitates from the CPGR and UCT IDM, supported by DIPLOMICS.

Author contributions

Nabeela Gamiet: Conceptualisation; Data curation; Formal analysis; Investigation; Methodology; Software; Validation; Visualisation; Writing—original draft; and Writing—review & editing. Nashia Deepnarain: Data curation; Formal analysis; Investigation; Methodology; Software; Supervision; Validation; Visualisation; Writing—original draft; and Writing—review & editing. Stefan Abel: Conceptualisation; Formal analysis; Investigation; Methodology; Software; Supervision; Validation; Visualisation; Writing—original draft; and Writing—review & editing. Hester-Mari Burger: Conceptualisation; Supervision; Writing—review & editing. Elisabeth Mayer: Conceptualisation; Methodology; Writing—review & editing. Mariska Lilly: Conceptualisation; Data curation; Formal analysis; Funding acquisition; Investigation; Methodology; Project administration; Resources; Software; Supervision; Validation; Visualisation; Roles/Writing—original draft; and Writing—review & editing.

Funding

Open access funding provided by Cape Peninsula University of Technology. This research was supported by funding from the Cape Peninsula University of Technology (CPUT). Additionally, we acknowledge the National Research Foundation (NRF) for providing a Postgraduate bursary to support this study.

Data availability

No datasets were generated or analysed during the current study.

Declarations

Ethics approval

This article does not contain any studies involving human participants or animals performed by any of the authors.

Competing interests

No authors declare competing of interests.

Footnotes

Publisher's Note

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

References

  1. Abbax H, Gelderblom W, Cawood M, Shier W (1993) Biological activities of fumonisins, mycotoxins from Fusarium moniliforme, in jimsonweed (Datura stramonium L.) and mammalian cell cultures. Toxicon 31:345–353 [DOI] [PubMed] [Google Scholar]
  2. Abou-Karam M, Abbas HK, Shier WT (2004) N-fatty acylation of hydrolyzed fumonisin B1, but not of intact fumonisin B1, strongly enhances in vitro mammalian toxicity. J Toxicol Toxin Rev 23:123–151 [Google Scholar]
  3. Akiyama S, Mochizuki W, NIbe Y, Matsumoto Y, Sakamoto K, Oshima S, Watanabe M, Nakamura T (2017) Ccn3 expression marks a sulfomucin-nonproducing unique subset of colonic goblet cells in mice. Acta Histochem Cytochem 50:159–168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Antonissen G, De Baere S, Novak B, Schatzmayr D, den Hollander D, Devreese M, Croubels S (2020) Toxicokinetics of hydrolyzed fumonisin B1 after single oral or intravenous bolus to broiler chickens fed a control or a fumonisins-contaminated diet. Toxins 12(6):413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Apidianakis Y, Pitsouli C, Perrimon N, Rahme L (2009) Synergy between bacterial infection and genetic predisposition in intestinal dysplasia. Proc Natl Acad Sci U S A 106:20883–20888 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Aslam B, Basit M, Nisar MA, Khurshid M, Rasool MH (2016) Proteomics: technologies and their applications. J Chromatogr Sci. 10.1093/chromsci/bmw167 [DOI] [PubMed] [Google Scholar]
  7. Baird BN, Schliekelman MJ, Ahn Y-H, Chen Y, Roybal JD, Gill BJ, Mishra DK, Erez B, O’Reilly M, Yang Y (2013) Fibulin-2 is a driver of malignant progression in lung adenocarcinoma. PLoS ONE 8:e67054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Baron BW, Pytel P (2017) Expression pattern of the BCL6 and ITM2B proteins in normal human brains and in Alzheimer disease. Appl Immunohistochem Mol Morphol 25:489–496 [DOI] [PubMed] [Google Scholar]
  9. Beggs AH, Byers T, Knoll J, Boyce F, Bruns G, Kunkel L (1992) Cloning and characterization of two human skeletal muscle alpha-actinin genes located on chromosomes 1 and 11. J Biol Chem 267:9281–9288 [PubMed] [Google Scholar]
  10. Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc B 57:289–300 [Google Scholar]
  11. Bernfield M, Kokenyesi R, Kato M, Hinkes MT, Spring J, Gallo RL, Lose EJ (1992) Biology of the syndecans: a family of transmembrane heparan sulfate proteoglycans. Annu Rev Cell Biol 8:365–393 [DOI] [PubMed] [Google Scholar]
  12. Bertero A, Moretti A, Spicer LJ, Caloni F (2018) Fusarium molds and mycotoxins: potential species-specific effects. Toxins 10:244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Black MB, Andersen ME, Pendse SN, Borghoff SJ, Streicker M, McMullen PD (2021) RNA-sequencing (transcriptomic) data collected in liver and lung of male and female B6C3F1 mice exposed to various dose levels of 4-methylimidazole for 2, 5, or 28 days. Data Brief 38:107420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bouhet S, Oswald IP (2007) The intestine as a possible target for fumonisin toxicity. Mol Nutr Food Res 51:925–931 [DOI] [PubMed] [Google Scholar]
  15. Bouhet S, Le Dorze E, Peres S, Fairbrother JM, Oswald IP (2006) Mycotoxin fumonisin B1 selectively down-regulates the basal IL-8 expression in pig intestine: in vivo and in vitro studies. Food Chem Toxicol 44:1768–1773 [DOI] [PubMed] [Google Scholar]
  16. Brabender J, Lord R, Metzger R, Park J, Salonga D, Danenberg K, Danenberg P, Hölscher A, Schneider P (2003) Differential SPARC mRNA expression in Barrett’s oesophagus. Br J Cancer 89:1508–1512 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Burridge K, McCullough L (1980) The association of α-actinin with the plasma membrane. J Supramol Struct 13:53–65 [DOI] [PubMed] [Google Scholar]
  18. Cai X, Liu C, Zhang TN, Zhu YW, Dong X, Xue P (2018) Down-regulation of FN1 inhibits colorectal carcinogenesis by suppressing proliferation, migration, and invasion. J Cell Biochem 119:4717–4728 [DOI] [PubMed] [Google Scholar]
  19. Caloni F, Spotti M, Pompa G, Zucco F, Stammati A, De Angelis I (2002) Evaluation of fumonisin B1 and its metabolites absorption and toxicity on intestinal cells line Caco-2. Toxicon 40:1181–1188 [DOI] [PubMed] [Google Scholar]
  20. Caloni F, Stammati A, Raimondi F, De Angelis I (2005) In vitro study with Caco-2 cells on fumonisin B1: aminopentol intestinal passage and role of P-glycoprotein. Vet Res Commun 29:285–287 [DOI] [PubMed] [Google Scholar]
  21. Chen Z, Chen H, Li X, Yuan Q, Su J, Yang L, Ning L, Lei H (2019) Fumonisin B1 damages the barrier functions of porcine intestinal epithelial cells in vitro. J Biochem Mol Toxicol 33:e22397 [DOI] [PubMed] [Google Scholar]
  22. Cho EA, Patterson LT, Brookhiser WT, Mah S, Kintner C, Dressler GR (1998) Differential expression and function of cadherin-6 during renal epithelium development. Development 125:803–812 [DOI] [PubMed] [Google Scholar]
  23. Chu X, Zheng W, Wang J, Zhang J, Pan Y, Shao C (2020) CDK6 inhibition targeted by miR-378a-3p protects against intestinal injury induced by ionizing radiation. Biochem Biophys Res Commun 531:328–334 [DOI] [PubMed] [Google Scholar]
  24. Chua CC, Rahimi N, Forsten-Williams K, Nugent MA (2004) Heparan sulfate proteoglycans function as receptors for fibroblast growth factor-2 activation of extracellular signal-regulated kinases 1 and 2. Circ Res 94:316–323 [DOI] [PubMed] [Google Scholar]
  25. Cimbalo A, Frangiamone M, Font G, Manyes L (2022) The importance of transcriptomics and proteomics for studying molecular mechanisms of mycotoxin exposure: a review. Food Chem Toxicol. 10.1016/j.fct.2022.113396 [DOI] [PubMed] [Google Scholar]
  26. Coman C, Solari FA, Hentschel A, Sickmann A, Zahedi RP, Ahrends R (2016) Simultaneous metabolite, protein, lipid extraction (SIMPLEX): a combinatorial multimolecular omics approach for systems biology. Mol Cell Proteomics 15:1435–1466 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Corti F, Finetti F, Ziche M, Simons M (2013) The syndecan-4/protein kinase Cα pathway mediates prostaglandin E2-induced extracellular regulated kinase (ERK) activation in endothelial cells and angiogenesis in vivo. J Biol Chem 288:12712–12721 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Cox J, Hein MY, Luber CA, Paron I, Nagaraj N, Mann M (2014) Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol Cell Prot 13:2513–2526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Danielsen ET, Olsen AK, Coskun M, Nonboe AW, Larsen S, Dahlgaard K, Bennett EP, Mitchelmore C, Vogel LK, Troelsen JT (2018) Intestinal regulation of suppression of tumorigenicity 14 (ST14) and serine peptidase inhibitor, Kunitz type-1 (SPINT1) by transcription factor CDX2. Sci Rep 8:11813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Dellafiora L, Galaverna G, Dall’Asta C (2018) Mechanisms of fumonisin B1 toxicity: a computational perspective beyond the ceramide synthases inhibition. Chem Res Toxicol 31:1203–1212 [DOI] [PubMed] [Google Scholar]
  31. Deng Y, Wan Q, Yan W (2019) Integrin α5/ITGA5 promotes the proliferation, migration, invasion, and progression of oral squamous carcinoma by epithelial–mesenchymal transition. Cancer Manag Res. 10.2147/CMAR.S223201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Desgrosellier JS, Cheresh DA (2010) Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer 10:9–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Devriendt B, Gallois M, Verdonck F, Waché Y, Bimczok D, Oswald IP, Goddeeris BM, Cox E (2009) The food contaminant fumonisin B1 reduces the maturation of porcine CD11R1+ intestinal antigen-presenting cells and antigen-specific immune responses, leading to a prolonged intestinal ETEC infection. Vet Res. 10.1051/vetres/2009023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Dombrink-Kurtzman MA (2003) Fumonisin and beauvericin induce apoptosis in turkey peripheral blood lymphocytes. Mycopathologia 156:357–364 [DOI] [PubMed] [Google Scholar]
  35. Doncheva NT, Morris JH, Gorodkin J, Jensen LJ (2018) Cytoscape StringApp: network analysis and visualization of proteomics data. J Proteome Res 18:623–632 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Dopavogui L, Polizzi A, Fougerat A, Gourbeyre P, Terciolo C, Klement W, Pinton P, Laffite J, Cossalter A-M, Bailly J-D (2022) Tissular genomic responses to oral FB1 exposure in pigs. Toxins 14:83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Eshelli M, Qader MM, Jambi EJ, Hursthouse AS, Rateb ME (2018) Current status and future opportunities of omics tools in mycotoxin research. Toxins 10:433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Esseghir S, Kennedy A, Seedhar P, Nerurkar A, Poulsom R, Reis-Filho JS, Isacke CM (2007) Identification of NTN4, TRA1, and STC2 as prognostic markers in breast cancer in a screen for signal sequence encoding proteins. Clin Cancer Res 13:3164–3173 [DOI] [PubMed] [Google Scholar]
  39. Fang L, Che Y, Zhang C, Huang J, Lei Y, Lu Z, Sun N, He J (2021) LAMC1 upregulation via TGFβ induces inflammatory cancer-associated fibroblasts in esophageal squamous cell carcinoma via NF-κB–CXCL1–STAT3. Mol Oncol 15:3125–3146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Flynn T, Stack M, Troy A, Chirtel S (1997) Assessment of the embryotoxic potential of the total hydrolysis product of fumonisin B1 using cultured organogenesis-staged rat embryos. Food Chem Toxicol 35:1135–1141 [DOI] [PubMed] [Google Scholar]
  41. Forciniti S, Greco L, Grizzi F, Malesci A, Laghi L (2020) Iron metabolism in cancer progression. Int J Mol Sci 21:2257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Framson PE, Sage EH (2004) SPARC and tumor growth: where the seed meets the soil? J Cell Biochem 92:679–690 [DOI] [PubMed] [Google Scholar]
  43. Fynan T, Reiss M (1993) Resistance to inhibition of cell growth by transforming growth factor-beta and its role in oncogenesis. Crit Rev Oncog 4:493–540 [PubMed] [Google Scholar]
  44. Gammella E, Buratti P, Cairo G, Recalcati S (2017) The transferrin receptor: the cellular iron gate. Metallomics 9:1367–1375 [DOI] [PubMed] [Google Scholar]
  45. Gelderblom W, Cawood M, Snyman S, Vleggaar R, Marasas W (1993) Structure-activity relationships of fumonisins in short-term carcinogenesis and cytotoxicity assays. Food Chem Toxicol 31:407–414 [DOI] [PubMed] [Google Scholar]
  46. Gelderblom W, Abel S, Smuts CM, Marnewick J, Marasas W, Lemmer ER, Ramljak D (2001) Fumonisin-induced hepatocarcinogenesis: mechanisms related to cancer initiation and promotion. Environ Health Perspect 109:291–300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. González-López NM, Huertas-Ortiz KA, Leguizamon-Guerrero JE, Arias-Cortés MM, Tere-Peña CP, García-Castañeda JE, Rivera-Monroy ZJ (2021) Omics in the detection and identification of biosynthetic pathways related to mycotoxin synthesis. Anal Methods 13:4038–4054 [DOI] [PubMed] [Google Scholar]
  48. Grazi GL, Mazziotti A, Legnani C, Jovine E, Miniero R, Gallucci A, Palareti G, Gozzetti G (1995) The role of tumor markers in the diagnosis of hepatocellular carcinoma, with special reference to the des-gamma-carboxy prothrombin. Liver Transpl Surg 1:249–255 [DOI] [PubMed] [Google Scholar]
  49. Grenier B, Bracarense APF, Schwartz HE, Trumel C, Cossalter AM, Schatzmayr G, Kolf-Clauw M, Moll WD, Oswald IP (2012) The low intestinal and hepatic toxicity of hydrolyzed fumonisin B1 correlates with its inability to alter the metabolism of sphingolipids. Biochem Pharmacol 83:1465–1473 [DOI] [PubMed] [Google Scholar]
  50. Hahn I, Nagl V, Schwartz-Zimmermann HE, Varga E, Schwarz C, Slavik V, Reisinger N, Malachová A, Cirlini M, Generotti S (2015) Effects of orally administered fumonisin B1 (FB1), partially hydrolysed FB1, hydrolysed FB1 and N-(1-deoxy-D-fructos-1-yl) FB1 on the sphingolipid metabolism in rats. Food Chem Toxicol 76:11–18 [DOI] [PubMed] [Google Scholar]
  51. Hai T, Wolfgang CD, Marsee DK, Allen AE, Sivaprasad U (1999) ATF3 and stress responses. Gene Expr 7:321–335 [PMC free article] [PubMed] [Google Scholar]
  52. Hai T, Wolford CC, Chang YS (2010) ATF3, a hub of the cellular adaptive-response network, in the pathogenesis of diseases: is modulation of inflammation a unifying component? Gene Expr 15:1–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Halloy DJ, Gustin PG, Bouhet S, Oswald IP (2005) Oral exposure to culture material extract containing fumonisins predisposes swine to the development of pneumonitis caused by Pasteurella multocida. Toxicology 213:34–44 [DOI] [PubMed] [Google Scholar]
  54. Han I, Park H, Oh ES (2004) New insights into syndecan-2 expression and tumourigenic activity in colon carcinoma cells. J Mol Histol 35:319–326 [DOI] [PubMed] [Google Scholar]
  55. Hao Y, Baker D, Ten Dijke P (2019) TGF-β-mediated epithelial-mesenchymal transition and cancer metastasis. Int J Mol Sci 20:2767 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hartl M, Humpf HU (2000) Toxicity assessment of fumonisins using the brine shrimp (Artemia salina) bioassay. Food Chem Toxicol 38:1097–1102 [DOI] [PubMed] [Google Scholar]
  57. Haschek WM, Gumprecht LA, Smith G, Tumbleson ME, Constable PD (2001) Fumonisin toxicosis in swine: an overview of porcine pulmonary edema and current perspectives. Environ Health Perspect 109:251–257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Howard PC, Couch LH, Patton RE, Eppley RM, Doerge DR, Churchwell MI, Marques MM, Okerberg CV (2002) Comparison of the toxicity of several fumonisin derivatives in a 28-day feeding study with female B6C3F1 mice. Toxicol Appl Pharmacol 185:153–165 [DOI] [PubMed] [Google Scholar]
  59. Humpf HU, Schmelz EM, Meredith FI, Vesper H, Vales TR, Wang E, Menaldino DS, Liotta DC, Merrill AH (1998) Acylation of naturally occurring and synthetic 1-deoxysphinganines by ceramide synthase: formation of N-palmitoyl-aminopentol produces a toxic metabolite of hydrolyzed fumonisin, AP1, and a new category of ceramide synthase inhibitor. J Biol Chem 273:19060–19064 [DOI] [PubMed] [Google Scholar]
  60. Istifli ES, Hüsunet MT, Ila HB (2019) Cell division, cytotoxicity, and the assays used in the detection of cytotoxicity. Cytotoxicity-definition, identification, and cytotoxic compounds
  61. Jun JI, Lau LF (2011) Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat Rev Drug Discov 10:945–963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kan F, Ye L, Yan T, Cao J, Zheng J, Li W (2017) Proteomic and transcriptomic studies of HBV-associated liver fibrosis of an AAV-HBV-infected mouse model. BMC Genomics 18:1–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Kim S (2001) Molecular mechanism of inactivation of TGF-beta receptors during carcinogenesis. Tanpakushitsu Kakusan Koso 46:111–116 [PubMed] [Google Scholar]
  64. Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Dall’Asta C, Eriksen GS, Taranu I, Altieri A, Roldán-Torres R, Oswald IP (2018) Risks for animal health related to the presence of fumonisins, their modified forms and hidden forms in feed. EFSA J 16:e05242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Knutsen HK, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Dall’Asta C, Gutleb AC, Humpf HU, Galli C, Metzler M, Oswald IP, Parent-Massin D, Binaglia M, Steinkellner H, Alexander J (2018b) Appropriateness to set a group health-based guidance value for fumonisins and their modified forms. EFSA J 16:e05172 [DOI] [PMC free article] [PubMed]
  66. Kolachala VL, Bajaj R, Wang L, Yan Y, Ritzenthaler JD, Gewirtz AT, Roman J, Merlin D, Sitaraman SV (2007) Epithelial-derived fibronectin expression, signaling, and function in intestinal inflammation. J Biol Chem 282:32965–32973 [DOI] [PubMed] [Google Scholar]
  67. Kosa P, Szabo R, Molinolo AA, Bugge TH (2012) Suppression of tumorigenicity-14, encoding matriptase, is a critical suppressor of colitis and colitis-associated colon carcinogenesis. Oncogene 31:3679–3695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P (2012) Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer 12:860–875 [DOI] [PubMed] [Google Scholar]
  69. Latil A, Chêne L, Cochant-Priollet B, Mangin P, Fournier G, Berthon P, Cussenot O (2003) Quantification of expression of netrins, slits and their receptors in human prostate tumors. Int J Cancer 103:306–315 [DOI] [PubMed] [Google Scholar]
  70. Li GZ, Vissers JP, Silva JC, Golick D, Gorenstein MV, Geromanos SJ (2009) Database searching and accounting of multiplexed precursor and product ion spectra from the data independent analysis of simple and complex peptide mixtures. Proteomics 9:1696–1719 [DOI] [PubMed] [Google Scholar]
  71. Li J, Chen Z, Tian L, Zhou C, He MY, Gao Y, Wang S, Zhou F, Shi S, Feng X (2014) LncRNA profile study reveals a three-lncrna signature associated with the survival of patients with oesophageal squamous cell carcinoma. Gut 63:1700–1710 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Li Y, Hou J, Yang L, Zhang T, Jiang Y, Du Z, Ma H, Li G, Zhu J, Chen P (2024) ITRAQ based proteomics reveals the potential mechanism of placental injury induced by prenatal stress. Int J Mol Sci 25:9978 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lindholm ME, Jimenez-Morales D, Zhu H, Seo K, Amar D, Zhao C, Raja A, Madhvani R, Abramowitz S, Espenel C (2021) Mono- and biallelic protein-truncating variants in alpha-actinin 2 cause cardiomyopathy through distinct mechanisms. Circ Genom Precis Med 14:e003419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Liu Y, Clegg H, Leslie P, Di J, Tollini L, He Y, Kim T, Jin A, Graves L, Zheng J (2015) CHCHD2 inhibits apoptosis by interacting with Bcl-xL to regulate Bax activation. Cell Death Differ 22:1035–1046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Liu S, Liang J, Liu Z, Zhang C, Wang Y, Watson AH, Zhou C, Zhang F, Wu K, Zhang F (2021) The role of CD276 in cancers. Front Oncol 11:654684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Loiseau N, Debrauwer L, Sambou T, Bouhet S, Miller JD, Martin PG, Viadère JL, Pinton P, Puel O, Pineau T (2007) Fumonisin B1 exposure and its selective effect on porcine jejunal segment: sphingolipids, glycolipids and trans-epithelial passage disturbance. Biochem Pharmacol 74:144–152 [DOI] [PubMed] [Google Scholar]
  77. Lou X, Han X, Jin C, Tian W, Yu W, Ding D, Cheng L, Huang B, Jiang H, Lin B (2013) SOX2 targets fibronectin 1 to promote cell migration and invasion in ovarian cancer: new molecular leads for therapeutic intervention. OMICS 17:510–518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. MacDonald TT, Monteleone G (2005) Immunity, inflammation, and allergy in the gut. Science 307:1920–1925 [DOI] [PubMed] [Google Scholar]
  79. Marchese D, de Groot NS, Lorenzo Gotor N, Livi CM, Tartaglia GG (2016) Advances in the characterization of RNA-binding proteins. WIREs RNA 7:793–810 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Markowitz SD, Roberts AB (1996) Tumor suppressor activity of the TGF-β pathway in human cancers. Cytokine Growth Factor Rev 7:93–102 [DOI] [PubMed] [Google Scholar]
  81. McArdle AJ, Menikou S (2020) What is proteomics? Arch Dis Child Educ Pract Ed. 10.1136/archdischild-2019-317434 [DOI] [PubMed] [Google Scholar]
  82. Minervini F, Garbetta A, D’Antuono I, Cardinali A, Martino NA, Debellis L, Visconti A (2014) Toxic mechanisms induced by fumonisin B1 mycotoxin on human intestinal cell line. Arch Environ Contam Toxicol 67:115–123 [DOI] [PubMed] [Google Scholar]
  83. Mostafavi-Pour Z, Ashrafi MR, Talaei-Khozani T (2018) Downregulation of ITGA4 and ITGA5 genes after formation of 3d spherules by human Wharton’s jelly stem cells (hWJSCs). Mol Biol Rep 45:245–252 [DOI] [PubMed] [Google Scholar]
  84. Nakagawa Y, Nakayama H, Nagata M, Yoshida R, Kawahara K, Hirosue A, Tanaka T, Yuno A, Matsuoka Y, Kojima T (2014) Overexpression of fibronectin confers cell adhesion-mediated drug resistance (CAM-DR) against 5-FU in oral squamous cell carcinoma cells. Int J Oncol 44:1376–1384 [DOI] [PubMed] [Google Scholar]
  85. Neckermann K, Claus G, De Baere S, Antonissen G, Lebrun S, Gemmi C, Taminiau B, Douny C, Scippo ML, Schatzmayr D, Gathumbi J (2021) The efficacy and effect on gut microbiota of an aflatoxin binder and a fumonisin esterase using an in vitro simulator of the human intestinal microbial ecosystem (SHIME®). Food Res Int 145:110395 [DOI] [PubMed] [Google Scholar]
  86. Niederlechner S, Klawitter J, Baird C, Kallweit AR, Christians U, Wischmeyer PE (2012) Fibronectin-integrin signaling is required for L-glutamine’s protection against gut injury. PLoS ONE 7:e50185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Pan L, Zhao Y, Farouk MH, Bao N, Wang T, Qin G (2018) Integrins were involved in soybean agglutinin induced cell apoptosis in IPEC-J2. Int J Mol Sci 19:587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Pap T, Bertrand J (2013) Syndecans in cartilage breakdown and synovial inflammation. Nat Rev Rheumatol 9:43–55 [DOI] [PubMed] [Google Scholar]
  89. Park H, Kim Y, Lim Y, Han I, Oh ES (2002) Syndecan-2 mediates adhesion and proliferation of colon carcinoma cells. J Biol Chem 277:29730–29736 [DOI] [PubMed] [Google Scholar]
  90. Perduca M, Nicolis S, Mannucci B, Galliano M, Monaco HL (2018) Human plasma retinol-binding protein (RBP4) is also a fatty acid-binding protein. Biochim Biophys Acta Mol Cell Biol Lipids 1863:458–466 [DOI] [PubMed] [Google Scholar]
  91. Piñeiro C, Carrera M, Cañas B, Lekube X, Martinez I (2015) Proteomics and food analysis: principles, techniques and applications. Handbook of food analysis-two volume set; CRC Press: Boca Raton, FL, USA 10:393–416
  92. Qazi BS, Tang K, Qazi A (2011) Recent advances in underlying pathologies provide insight into interleukin-8 expression-mediated inflammation and angiogenesis. Int J Inflamm 2011(1):908468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Ren JS, Kamangar F, Qiao YL, Taylor PR, Liang H, Dawsey SM, Liu B, Fan JH, Abnet CC (2009) Serum pepsinogens and risk of gastric and oesophageal cancers in the general population nutrition intervention trial cohort. Gut 58:636–642 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Scheicher R, Hoelbl-Kovacic A, Bellutti F, Tigan AS, Prchal-Murphy M, Heller G, Schneckenleithner C, Salazar-Roa M, Zöchbauer-Müller S, Zuber J (2015) CDK6 as a key regulator of hematopoietic and leukemic stem cell activation. Blood 125:90–101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Schelstraete W, Devreese M, Croubels S (2020) Comparative toxicokinetics of Fusarium mycotoxins in pigs and humans. Food Chem Toxicol 137:111140 [DOI] [PubMed] [Google Scholar]
  96. Schertz H, Kluess J, Frahm J, Schatzmayr D, Dohnal I, Bichl G, Schwartz-Zimmermann H, Breves G, Dänicke S (2018) Oral and intravenous fumonisin exposure in pigs—a single-dose treatment experiment evaluating toxicokinetics and detoxification. Toxins 10(4):150 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Schiemann BJ, Neil JR, Schiemann WP (2003) SPARC inhibits epithelial cell proliferation in part through stimulation of the transforming growth factor-β-signaling system. Mol Biol Cell 14:3977–3988 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Shah C, Hegde BG, Morén B, Behrmann E, Mielke T, Moenke G, Spahn CM, Lundmark R, Daumke O, Langen R (2014) Structural insights into membrane interaction and caveolar targeting of dynamin-like EHD2. Structure 22:409–420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Shen X, Hong MS, Moss J, Vaughan M (2007) BIG1, a brefeldin A-inhibited guanine nucleotide-exchange protein, is required for correct glycosylation and function of integrin β1. Proc Natl Acad Sci U S A 104:1230–1235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Simone LC, Naslavsky N, Caplan S (2014) Scratching the surface: actin and other roles for the C-terminal Eps15 homology domain protein, EHD2. Histol Histopathol 29:285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Sponziello M, Rosignolo F, Celano M, Maggisano V, Pecce V, De Rose RF, Lombardo GE, Durante C, Filetti S, Damante G (2016) Fibronectin-1 expression is increased in aggressive thyroid cancer and favors the migration and invasion of cancer cells. Mol Cell Endocrinol 431:123–132 [DOI] [PubMed] [Google Scholar]
  102. Subramanian S, Geng H, Tan XD (2020) Cell death of intestinal epithelial cells in intestinal diseases. Sheng Li Xue Bao 72:308 [PMC free article] [PubMed] [Google Scholar]
  103. Sun Y, Zhao C, Ye Y, Wang Z, He Y, Li Y, Mao H (2020) High expression of fibronectin 1 indicates poor prognosis in gastric cancer. Oncol Lett 19:93–102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Szabó C, Kachungwa Lugata J, Ortega ADSV (2023) Gut health and influencing factors in pigs. Animals 13:1350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, Jensen LJ (2019) STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47:607–613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Takemasa I, Higuchi H, Yamamoto H, Sekimoto M, Tomita N, Nakamori S, Matoba R, Monden M, Matsubara K (2001) Construction of preferential cDNA microarray specialized for human colorectal carcinoma: molecular sketch of colorectal cancer. Biochem Biophys Res Commun 285:1244–1249 [DOI] [PubMed] [Google Scholar]
  107. Taniguchi K, Karin M (2018) NF-κB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol 18:309–324 [DOI] [PubMed] [Google Scholar]
  108. Terciolo C, Bracarense AP, Souto PC, Cossalter AM, Dopavogui L, Loiseau N, Oliveira CA, Pinton P, Oswald IP (2019) Fumonisins at doses below EU regulatory limits induce histological alterations in piglets. Toxins 11:548 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Waalkes S, Atschekzei F, Kramer MW, Hennenlotter J, Vetter G, Becker JU, Stenzl A, Merseburger AS, Schrader AJ, Kuczyk MA (2010) Fibronectin 1 mRNA expression correlates with advanced disease in renal cancer. BMC Cancer 10:1–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Wan LYM, Turner PC, El-Nezami H (2013) Individual and combined cytotoxic effects of Fusarium toxins (deoxynivalenol, nivalenol, zearalenone and fumonisins B1) on swine jejunal epithelial cells. Food Chem Toxicol 57:276–283 [DOI] [PubMed] [Google Scholar]
  111. Wang C, Lin K, Chen S, Chan Y, Hsueh S (2004) Overexpression of SPARC gene in human gastric carcinoma and its clinic–pathologic significance. Br J Cancer 91:1924–1930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Wang R, Wang X, Ni B, Huan CC, Wu JQ, Wen LB, Liao Y, Tong GZ, Ding C, Fan HJ (2016) Syndecan-4, a PRRSV attachment factor, mediates PRRSV entry through its interaction with EGFR. Biochem Biophys Res Commun 475:230–237 [DOI] [PubMed] [Google Scholar]
  113. Wang J, Deng L, Huang J, Cai R, Zhu X, Liu F, Wang Q, Zhang J, Zheng Y (2017) High expression of fibronectin 1 suppresses apoptosis through the NF-κB pathway and is associated with migration in nasopharyngeal carcinoma. Am J Transl Res 9:4502 [PMC free article] [PubMed] [Google Scholar]
  114. Wang T, Lei H, Zhou L, Tang M, Liu Q, Long F, Li Q, Su J (2022) Effect of fumonisin B1 on proliferation and apoptosis of intestinal porcine epithelial cells. Toxins 14:471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Wilson AJ, Byron K, Gibson PR (1999) Interleukin-8 stimulates the migration of human colonic epithelial cells in vitro. Clin Sci 97:385–390 [PubMed] [Google Scholar]
  116. Xian D, Yang S, Liu Y, Liu Q, Huang D, Wu Y (2024) Microrna-196a-5p facilitates the onset and progression via targeting ITM2B in esophageal squamous cell carcinoma. Pathol Int. 10.1111/pin.13408 [DOI] [PubMed] [Google Scholar]
  117. Xiao J, Yang W, Xu B, Zhu H, Zou J, Su C, Rong J, Wang T, Chen Z (2018) Expression of fibronectin in esophageal squamous cell carcinoma and its role in migration. BMC Cancer 18:1–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Xu S, Xu H, Wang W, Li S, Li H, Li T, Zhang W, Yu X, Liu L (2019) The role of collagen in cancer: from bench to bedside. J Transl Med 17:1–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Xu L, Zhou Y, Ren X, Xu C, Ren R, Yan X, Li X, Yang H, Xu X, Guo X (2022) Expanding the phenotypic and genotypic spectrum of ARFGEF1-related neurodevelopmental disorder. Front Mol Neurosci 15:862096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Yuan Q, Jiang Y, Fan Y, Ma Y, Lei H, Su J (2019) Fumonisin B1 induces oxidative stress and breaks barrier functions in pig iliac endothelium cells. Toxins 11:387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Zhang MS, Cui JD, Lee D, Yuen VW-H, Chiu DK-C, Goh CC, Cheu JW-S, Tse AP-W, Bao MH-R, Wong BPY (2022) Hypoxia-induced macropinocytosis represents a metabolic route for liver cancer. Nat Commun 13:954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Zhao Z, Li S, Li S, Wang J, Lin H, Fu W (2021) High expression of oncogene Cadherin-6 correlates with tumor progression and a poor prognosis in gastric cancer. Cancer Cell Int 21:1–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Zhou C, Li C, Li D, Wang Y, Shao W, You Y, Peng J, Zhang X, Lu L, Shen X (2013) BIG1, a brefeldin a-inhibited guanine nucleotide-exchange protein, regulates neurite development via PI3K–AKT and ERK signaling pathways. Neuroscience 254:361–368 [DOI] [PubMed] [Google Scholar]
  124. Zhou J, Edgar BA, Boutros M (2017) Atf3 acts as a rheostat to control JNK signaling during intestinal regeneration. Nat Commun 8:14289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Zhou Y, Cao G, Cai H, Huang ZhuX (2020) The effect and clinical significance of FN1 expression on biological functions of gastric cancer cells. Cell Mol Biol 66:191–198 [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ESM1 (453.9KB, docx)

(DOCX 453 KB)

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Mycotoxin Research are provided here courtesy of Springer

RESOURCES