You are on page 1of 13

Ecotoxicology and Environmental Safety 224 (2021) 112637

Contents lists available at ScienceDirect

Ecotoxicology and Environmental Safety


journal homepage: www.elsevier.com/locate/ecoenv

Whole transcriptome-based ceRNA network analysis revealed ochratoxin


A-induced compromised intestinal tight junction proteins through WNT/
Ca2+ signaling pathway
Xue Yang a, b, c, d, 1, Yanan Gao a, b, c, d, 1, Shengnan Huang a, b, c, d, Chuanyou Su a, b, c, d,
Jiaqi Wang a, b, c, d, Nan Zheng a, b, c, d, *
a
Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of
Agricultural Sciences, Beijing 100193, China
b
Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of
Agricultural Sciences, Beijing 100193, China
c
Milk and Dairy Product Inspection Center of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing
100193, China
d
State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China

A R T I C L E I N F O A B S T R A C T

Edited by: Dr Yong Liang Ochratoxin A (OTA) is a widespread environmental pollutant that is a threat to humans and livestock and re­
mains a global concern to public health. It has negative effects on both humans and animals that are in a
Keywords: continuously exposed environment. The compromised intestinal barrier caused by OTA has aroused widespread
Ochratoxin A concern. This study aimed to investigate the mechanism of OTA-induced tight junction (TJ) protein damage and
Whole transcriptome
the relevant components of the intestinal barrier through in vivo whole transcriptome analysis combined with in
CeRNA regulatory network
vitro functional verification. Bioinformatics analysis in OTA-treated Balb/c mice demonstrated that regulated TJ
Tight junction proteins
Intestinal barrier protein related mRNAs were perturbed, and activated the WNT/Ca2+ signaling pathway possibly regulated by
key lncRNAs and miRNAs. Competing endogenous RNA (ceRNA) network analysis revealed that lncRNA Zeb1
regulated FZD4 binding with WNT5a to release Ca2+ by targeting miR-1258-x and reduced the expression of TJ
proteins, thus damaging the function of the intestinal barrier. An in vitro experiment with Caco-2 cells verified
that an increase in Ca2+ level was involved in OTA-induced decreases in the expression of TJ proteins. Taken
together, these results will help to identify targets in the intestinal barrier that are compromised by OTA, and will
provide the basis for preventing the associated hazard and risk.

1. Introduction as a widespread environmental contaminant, OTA is found to exist in the


middle environment of dust carried by raw materials, workers, and
The fungal toxin ochratoxin A (OTA) is a commonly found envi­ processing sites in the process of food production (Fog Nielsen, 2003;
ronmental contaminant that is produced by certain strains of fungi Halstensen et al., 2004). In addition, because of its nephrotoxicity,
belonging to Aspergillus and Penicillium, for example, P. verrucosum, P. hepatotoxicity, and carcinogenicity, OTA has been classified as a 2B
carbonarius, A. niger, and A. ochraceus (Creppy, 2002). Recently, the high carcinogen by the International Agency for Research on Cancer (Akbari
pollution rate of OTA in agricultural products (natural grains and food et al., 2017; Marin et al., 2019). A provisional tolerable weekly intake of
products) has caused a significant threat to the food industry worldwide 100 ng/kg.bw/week for OTA has been established by the Joint
(Tang et al., 2019; Wei et al., 2020). Various environments, including FAO/WHO Expert Committee on Food Additives (Bondy and Pestka,
abnormal weather, improper harvesting, and storage, could increase the 2000). In addition to nephrotoxicity and hepatotoxicity, the intestinal
levels of OTA in crops (Jaksic et al., 2020; Raiola et al., 2015). Moreover, toxicity of OTA has attracted more and more attention (Wang et al.,

* Corresponding author at: Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal
Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
E-mail address: zhengnan@caas.cn (N. Zheng).
1
These authors contributed equally.

https://doi.org/10.1016/j.ecoenv.2021.112637
Received 22 April 2021; Received in revised form 19 July 2021; Accepted 11 August 2021
Available online 20 August 2021
0147-6513/© 2021 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

2018; Ying et al., 2019). There is an urgent need to find the mechanism 2. Materials and methods
of intestinal toxicity induced by OTA.
The intestinal barrier is the first barrier exposed to food pollutants, of 2.1. Materials
which mycotoxins are among the most universal contaminants (Gal­
arza-Seeber et al., 2016; Gambacorta et al., 2016). Tight junction (TJ) OTA powder was purchased from Pribolab (Qingdao, China), and
proteins, which could block the intercellular space and prevent bacteria then dissolved in 1% dimethyl sulfoxide (DMSO) corn oil solution
and toxins from entering the blood circulation, are of great significance (Sigma-Aldrich, St. Louis, MO, USA). The solution was freshly prepared
in maintaining the intestinal barrier function (Al-Sadi et al., 2011; just before use. Mouse anti-occludin, mouse anti-ZO-1, and rabbit anti-
Marchiando et al., 2011; Yu et al., 2010). They are composed of several claudin-3 were purchased from Proteintech (Chicago, IL, USA). Rabbit
multi-protein complexes, including transmembrane proteins (tricellulin, anti-claudin-4 was obtained from Thermo Scientific (Waltham, MA,
occludin, claudins, and junction adhesion molecules), and cytoplasmic USA). Goat anti-rabbit/mouse IgG conjugated to horseradish peroxidase
scaffold proteins zona occludens (ZOs) (Steed et al., 2010). Evidence and β-actin were from Bioss (Beijing, China). Mouse serum diamine
from previous studies suggested that the impairment of TJ proteins may oxidase (DAO), D-lactic acid (D-lactate), and serum intestinal fatty acid
be closely related to various intestinal pathogenesis (Arrieta et al., 2006; binding protein (I-FABP) enzyme-linked immunosorbent assay (ELISA)
Turner, 2009). By using in vitro experiments in intestinal Caco-2 cells, kits were obtained from Sinogene Scientific (Beijing, China). Trizol re­
OTA was found to induce intestinal barrier impairment through agent kit was obtained from Invitrogen (Carlsbad, CA, USA). Trypsin
increasing its permeability and decreasing the expression of TJ proteins (2.5%), antibiotics (100 units/mL penicillin, 100 μg/mL streptomycin),
(Gao et al., 2017; Maresca et al., 2008; Peng et al., 2019). OTA exposure Fluo-4 AM, BCA protein assay kit, Radio-Immunoprecipitation Assay
down-regulated the expression of TJ proteins claudin-1, claudin-3, and (RIPA) lysate, and phosphate buffer saline (PBS) were purchased from
claudin-4 in Caco-2 cells (Alizadeh et al., 2019). Studies have suggested Beyotime (Shanghai, China). Fetal bovine serum, nonessential amino
that OTA decreased the levels of TJ proteins in IPEC-J2 cells (Wang acids, and Dulbecco’s Modified Eagle’s Medium (DMEM) were from
et al., 2018). The nontoxic concentration of OTA aggravates the intes­ Gibco (Carlsbad, CA, USA). Primers and prime script II 1st strand cDNA
tinal barrier dysfunction of IPEC-J2 cells, accompanied by claudin-3 and synthesis kits were from Sangon Biotech (Shanghai, China). BAPTA-AM
claudin-4 disruption and decreases in expression (Ying et al., 2019). In (Ca2+ inhibitor) was obtained from Sigma-Aldrich (St. Louis, MO, USA)
addition, OTA significantly decreased the mRNA expression of ZO-1, and dissolved in DMEM.
occludin, claudin-c, f, 7a,7b, 11,12, 15a,15b in the intestinal of Juve­
nile Grass Carp (Liu et al., 2020). Despite these effects, the mechanisms
2.2. Experimental animals
underlying OTA-induced TJ protein disruption are still not clear, espe­
cially in vivo.
Thirty-six male healthy Balb/c mice in a good mental state were
Based on mRNA sequencing, the whole transcriptome constructs two
purchased from Beijing Vital River (Beijing, China). The mice were SPF
types of library (small RNA library and ribosome-specific library), which
clean grade aged 6–8 weeks, and the body weight was in the range of 20
can analyze the data of four different RNAs: long non-coding (lnc)RNA,
± 2 g. They were raised in an SPF barrier system at the animal experi­
circle (circ)RNA, micro (mi)RNA and mRNA, simultaneously (Zheng
ment platform of Beijing Vital River Laboratory Animal Technology Co.,
et al., 2019). LncRNA is an RNA molecule with a length of more than 200
Ltd. The whole feeding process strictly followed regulations on animal
nucleotides that cannot encode proteins but is involved in transcrip­
feeding and management with the permission code P2020055. The
tional regulation by binding with miRNA (Guttman et al., 2009; Wang
temperature of the feeding environment was controlled at 20–26 ◦ C and
et al., 2019a; Yi et al., 2019). CircRNA is a class of conserved and
humidity was within the range of 40–70% for 12 h of light and 12 h of
structurally stable non-coding (nc)RNA, which can act as a miRNA
darkness per day.
sponge to regulate mRNA expression, cell migration, and invasion.
MiRNA is a small ncRNA molecule, which can silence RNA and regulate
the expression of post-transcriptional genes (Bartel, 2004). Through 2.3. Gavage experiment and sample collection in mice
miRNA and the competitive binding of three kinds of RNA (lncRNA,
circRNA, and mRNA), a competing endogenous RNA (ceRNA) network After the mice were transferred to the barrier system, they were first
can be established, which provides a way to find the key RNAs that affect adapted to the environment for 7 days with sterile feed and water pro­
biological processes (Li et al., 2018; Taborda et al., 2017; Wang et al., vided freely. Then the mice were randomly divided into three groups
2020; Jiang et al., 2015). In recent years, analyzing the whole tran­ with 12 mice in each group. A subacute treatment consisted of 1/5–1/20
scriptome has gradually become a trend for studying the mechanism of of the half lethal dose (LD50) given each day for about 15–28 days
mycotoxins (Wang et al., 2019b). It was used to explore the molecular (OECD/OCDE 407, 2008). In the early stage of the experiment, ac­
mechanism of the toxicological effect of zearalenone on porcine gran­ cording to the acute oral toxicity experiment, the LD50 range of OTA was
ulosa cells, but it has not indicated how ncRNA regulated the expression estimated at 60 mg/kg b.w., which was estimated through a preliminary
of the cell cycle-related genes CDK1, CCNB1, and CHEK1 (Li et al., acute oral toxicity experiment based on the standard of GB/T
2020). To date, few examples of in vitro verification have been found in 21826–2008. With reference to the results of previous studies (Gao
whole transcriptome studies (Huang et al., 2019b). et al., 2021) and OECD guides (OECD/OCDE 407, 2008), 1/20 of the
In this study, the whole transcriptome combined with in vitro veri­ LD50 were selected as the dose value of intragastric administration. Each
fication was performed to profile the mechanism underlying intestinal mouse in the control group (CTL) was fed with normal saline per day,
toxicity in Balb/c mice exposed to OTA. We constructed a lncRNA- and each mouse in the solvent control group (DMSO) was fed with 1.0%
miRNA-mRNA ceRNA network to reveal OTA compromised TJ pro­ DMSO corn oil solution. The concentration of OTA was 3 mg/kg b.w.
teins through the activation of the WNT/Ca2+ signaling pathway. and each animal received a volume of 200 μL once a day at a fixed time
Verification by Caco-2 cells in vitro showed that OTA increased the level (9–10 AM) for 28 days. The mice drank and ate freely during intragastric
of Ca2+, inducing the decreased expression of TJ proteins. These results administration. Health status and abnormal activities were observed and
may provide new evidence for studying the molecular basis of intestinal recorded every day. The body weight of mice was weighed and recorded
barrier dysfunction caused by OTA. every 3 days. After the last intragastric administration, the mice were
fasted for 24 h. Then the mice were euthanized with carbon monoxide to
obtain the blood from the heart and intestinal jejunal tissue. The intes­
tinal contents were quickly rinsed with precooled PBS, frozen in liquid
nitrogen for 10 min, and stored at − 80 ◦ C.

2
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

2.4. Caco-2 cell culture 2.9. Immunofluorescence assay for TJ protein localization

Caco-2 cells were purchased from the American Type Culture Slides incubated with antibodies were placed in PBS, then shaken
Collection (Manassas, VA, USA), and the cells were cultured as before and washed on the decolorizing shaker three times, 5 min each time.
(Yang et al., 2019). When cell confluence reached 90%, they were After the slides were slightly dried, 4′ , 6-diamidin-2-phenylindol (DAPI)
seeded in 96-well plates, and then Ca2+ levels were measured. After they staining solution was dripped into the circle and incubated at room
were cultured and differentiated in 6-well plates for 14 d, Caco-2 cells temperature without light for 10 min. The glass slides were placed in
undergo spontaneous differentiation resulting in polarization and for­ PBS and then shaken and washed. An autofluorescence quenching agent
mation of the TJ proteins between adjacent cells. Then the differentiated was added to the ring for 5 min, and slides were rinsed for 10 min with
Caco-2 cells were used for TJ protein expression. In this study, the running water. After slides were slightly dried, they were sealed with
concentration of OTA adopted was according to the study by Gao et al. anti-fluorescence quenching tablets. The sections were observed under a
(2018). OTA (2000 μM) was acquired as stock by dissolving in methanol fluorescence microscope and the images were collected. The nucleus
and then diluted with DMEM to the concentration of the working solu­ stained by DAPI is blue under UV excitation, and the positive expression
tion (20 μM). After incubation with 20 μM OTA for 48 h, Caco-2 cells is red or green light labeled with corresponding fluorescein.
were collected and processed for Ca2+ levels. For TJ protein expression,
the differentiated Caco-2 cells were pretreated with 10 μM BAPTA-AM
for 2 h before incubating with 20 μM OTA for 48 h. 2.10. MiRNA sequence analysis

2.5. Determination of blood biochemical indexes and data analysis A trizol reagent kit was used to extract total RNA from mice jejunum
tissue according to the manufacturer’s protocol. The RNA was
Whole blood was collected and coagulated after 1.5 h at 37 ◦ C sequenced using Illumina HiSeq Xten by Gene Denovo Biotechnology
(without anticoagulant). Precipitated blood was placed in a centrifuge at Co. (Guangzhou, China). All of the clean tags were aligned with small
4 ◦ C for 10 min at 4000 rpm. The supernatant was taken as the serum, RNAs in the GeneBank (Release 209.0) and Rfam (Release 11.0) data­
transferred to a clean centrifuge tube, and packed into small parts for bases to identify and remove rRNA, scRNA, snRNA, snoRNA, and tRNA.
standby. Serum biochemical indexes were determined by automatic To identify and remove rRNA, scRNA, snRNA, snoRNA, and tRNA, all
biochemical analyzer HITACH17080 (Hitachi, Ltd., Tokyo, Japan) and the clean tags were aligned with small RNAs in the GeneBank (Release
ELISA kit, including DAO, D-lactate, and I-FABP. ELISA procedures were 209.0) and Rfam (Release 11.0) databases. Differential expression (DE)
followed according to kit instructions. miRNA analysis was performed by edgeR software between two
different groups. We identified miRNAs with a fold change (FC) ≥ 2 and
2.6. Observation of villous structure of jejunum in mice P value < 0.05 as DEmiRNAs. Two software, Miranda (Version 3.3a) and
TargetScan (Version 7.0), were used to predict targets. The Gene
Jejunal tissues were fixed with 4% paraformaldehyde for 36 h, Ontology (GO) (http://www.geneontology.org/) and Kyoto Encyclo­
dehydrated with ethanol, removed with xylene, and embedded in pedia of Genes and Genomes (KEGG) (https://www.kegg.jp) databases
paraffin. Paraffin-embedded tissues were sectioned at 4 µm and stored at were used for the functional annotation of target mRNAs (Kanehisa
room temperature. After hematoxylin-eosin (HE) staining, the sections et al., 2008). GO terms or pathways with P < 0.05 were defined as
were examined by light microscope (eclipse CI; Nikon, Tokyo, Japan). significantly enriched GO terms or pathways.

2.7. Goblet cell count in the jejunum


2.11. The mRNA, lncRNA, and circRNA sequence analysis
The number of intestinal jejunal goblet cells was counted by periodic
acid-Schiff (PAS) staining. The above sections were stained. Goblet cells Total RNA was obtained from mice jejunum tissue, RNA quality was
were purplish-red and the nuclei were blue under the microscope and assessed on an Agilent 2100 Bioanalyzer (Agilent Technologies, Palo
used for image acquisition and analysis. The jejunal tissue sections were Alto, CA, USA) and checked using RNase-free agarose gel electropho­
taken from six mice in each group, five villi were taken from each sec­ resis. To obtain high quality clean reads, the reads were further filtered
tion. The number of goblet cells was counted, and the average value was by fastp (version 0.18.0). The short reads alignment tool Bowtie2
calculated. (version 2.2.8) was used for mapping reads to the rRNA database. The
coding ability of the new transcripts was predicted by two software, CPC
2.8. Western blot for TJ proteins expression and CNCI. Differential expression analysis of mRNAs and ncRNAs was
determined using DESeq2 software between two different groups and by
Jejunum tissue preserved at − 80 ◦ C was weighed and a proportion of edgeR between two samples. The mRNAs/ncRNAs with false discovery
RIPA lysate (RIPA+ phenylmethylsulfonyl fluoride) was added (1 rate (FDR) < 0.05/absolute FC ≥ 1.5 and P value < 0.05/FC ≥ 2 were
mg:10 μL). The tissue was ground for 40 s with a homogenizer, then considered DEmRNAs/DEncRNAs, respectively. DEmRNAs were then
cracked on ice for 15 min, three times. After centrifugation at 4 ◦ C/ subjected to enrichment analysis of GO function and KEGG pathways.
12,000 rpm for 15 min, intestinal proteins were obtained from the su­ GO and KEGG analysis was performed on DEncRNAs after the prediction
pernatant. The differentiated Caco-2 cells collected above were used to of target genes.
extract proteins by RIPA lysate.
The tissue obtained above and cell protein concentration was
determined by a BCA protein assay kit method. Then the protein was 2.12. The ceRNA regulation analysis
denatured at 100 ◦ C for 10 min with 5 × loading buffer and stored at
− 20 ◦ C for western blot analysis, remaining denatured total protein was Spearman Rank Correlation Coefficient (SRCC) was used to evaluate
stored at − 80 ◦ C. Western blot analysis was used to assess TJ protein the expression correlation between mRNA-miRNA or lncRNA-miRNA.
(claudin-3, claudin-4, occludin, and ZO-1) expression. Equal amounts of Pairs with SRCC < − 0.7 were chosen as co-expressed negative pairs,
protein were electrophoresed on sodium dodecyl sulfate (SDS)-poly­ both DEmRNA and DElncRNA were DEmiRNA target genes. All the co-
acrylamide gels and electroblotted onto polyvinylidene difluoride expression competitive triplets identified above were assembled into a
membranes. Western blot analysis in vivo and in vitro was performed as lncRNA-miRNA-mRNA network and visualized using Cytoscape soft­
described previously (Yang et al., 2019). ware (v3.6.0) (http://www.cytoscape.org/).

3
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

2.13. Validation of mRNA, miRNA, and lncRNA expression by 3. Results


quantitative reverse transcription polymerase chain reaction (RT-qPCR)
3.1. OTA damaged intestinal barrier
A Prime Script II 1st strand cDNA synthesis kit and synthesis of
miRNA first-strand cDNA (stem-loop method) were used to reverse Starting from the first day of intragastric administration, the body
transcribe cDNAs. The RT primers of miRNA and the primer sequences weight of mice was weighed and recorded every 3 days. As depicted in
of all RNAs are shown in Table S1 and Table S2. The GAPDH gene was Fig. 1a, the average final body weight of mice was ranked as follows:
chosen as an internal control for mRNA, lncRNA, and circRNA, U6 was CTL>DMSO>OTA. The body weight of Balb/c mice decreased signifi­
used for miRNA (Ma et al., 2018; Zhang et al., 2019). The relative cantly from the 7th day to the 29th day after OTA gavage (P < 0.05).
expression of genes was calculated by the 2− ∆∆Ct method, and the mean The indexes of impaired intestinal barrier function (DAO, D-lactate,
and standard deviation of three biological replicates are shown as the and I-FABP) were measured by ELISA. As the results showed, OTA ten­
results. ded to increase the level of DAO (P > 0.05), yet, the concentrate of D-
lactate and I-FABP were significantly increased by OTA (P < 0.05)
2.14. Measurement of c(Ca2+) (Fig. 1b). At the end of the experiment, although there was no significant
difference in jejunal crypt depth between the CTL and OTA groups
Caco-2 cells were treated with 20 μM OTA for 48 h. The level of Ca2+ (P > 0.05), the length of jejunum villi and the ratio of jejunal villi length
was measured by a Fluo-4AM kit. An appropriate amount of Fluo-4AM to crypt depth in the OTA group was significantly lower than in the CTL
stock solution was taken and diluted to 5 μM by PBS. Then Caco-2 group (P < 0.05) (Fig. 1c, d, e). In addition, the number of mice jejunal
cells were incubated with Fluo-4 AM for 30 min in the dark to load goblet cells in the OTA group was significantly lower than in the CTL
the fluorescent probe. Fluorescence intensity before and after treatment groups (P < 0.05) (Fig. 1 f, g).
was detected with an excitation wavelength of 488 nm and an emission
wavelength of 520 nm. 3.2. Effect of OTA on the expression and localization of intestinal TJ
proteins
2.15. Statistical analysis
As the western blot results showed, OTA significantly decreased
GraphPad Prism 8.0 (GraphPad Software Inc., San Diego, CA, USA) claudin-3, claudin-4, occludin, and ZO-1 protein expression levels in vivo
was used for statistical analysis. One-way ANOVA and Tukey’s multiple (P < 0.05) (Fig. 2a, b). To expound the effect of OTA on the TJ structure
comparison were applied to test statistical differences between the in mice, the TJ proteins (claudin-3, occludin, and ZO-1) in jejunum
treatment groups and the CTL group. P value < 0.05 was considered epithelial cells of different groups were labeled with fluorescence.
statistically significant. Fig. 2c showed the localization of TJ proteins in the jejunum epithelial
cells of the CTL and treatment groups. Compared with the CTL group,
the distribution of TJ proteins, especially for claudin-3, on the lateral
membrane of adjacent cells was significantly reduced in the OTA group,

Fig. 1. The effect of 3 mg/kg b.w. OTA on Balb/c mice jejunum structure. (a) Changes in the body weight of Balb/c mice after OTA treatment. (b) Effect of OTA
treatment on blood biochemical indexes level. (c) Effects of OTA on histological structures of jejunum villi. (d, e) Effect of OTA on villi length and crypt depth of mice
intestinal jejunum, V and C represent villus height and crypt depth, respectively. (f, g) The number of goblet cells in CTL, DMSO, and OTA groups. * P < 0.05
represents a significant difference, and ns represents no significant difference.

4
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

Fig. 2. The changes of TJ proteins induced by OTA. (a) Jejunum tissue protein (30 μg) was immunoblotted to detect TJ proteins, (b) Quantified analysis of TJ protein
expression. (c) Effect of OTA on the localization of intestinal TJ proteins in mice. Nuclei are indicated in blue, fluorescent-labeled TJ proteins are red. *P < 0.05
represents a significant difference.

and the cell gap was not clear. DEmRNAs were found to be enriched in the regulation of actin, focal
adhesion, PI3K-Akt signaling pathway, and WNT signaling pathway
3.3. OTA exposure changed mRNA expression (Fig. 3d). According to the KEGG key pathways, the key genes were
selected to predict protein-protein interactions (PPI) through STRING
Whole transcriptome analysis was performed to understand the analysis. As the PPI results depicted, the DEmRNAs CCND1, WNT5a,
molecule mechanism of OTA-induced intestinal toxicity. In general, 760 FZD4, Axin2, Cacan1d, and Cav1 were target genes (Fig. 3e). The
DEmRNAs were obtained under the screening conditions. These were STRING analysis indicated that these key genes were tightly related with
the sum of DEmRNAs in the three groups (CTL, DMSO, and OTA) after the WNT signaling pathway. Moreover, the key DEmRNAs Cav1 and
comparison between every two groups (Fig. 3a). Venn analysis found Cacan1d related to the Ca2+ signal cascade were also found in the PPI
that the genes from the OTA/CTL and OTA/DMSO groups but not the network. Four DEmRNAs were randomly selected to determine relative
DMSO/CTL groups were the key DEmRNAs in OTA-treated samples expression. As the RT-qPCR results depicted, CCND1 and CLDN8 were
(Fig. 3b). As depicted by the Venn, 564 DEmRNAs were considered to be down-regulated and OAS2 and LPAR2 were upregulated in the OTA
the key genes in the OTA-treated group. group compared with the CTL group (Fig. 3f). All these results were
GO annotation demonstrated that terms in the lipid metabolism consistent with RNA sequencing.
process, extracellular region part, response to external stimulus, and
catalytic activity were enriched (Fig. 3c). KEGG analysis revealed that

5
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

Fig. 3. The overview of the DEmRNAs. (a) The number of DEmRNAs in different groups. (b) Venn analysis of the three groups. (c,d) The GO and KEGG annotation of
the target gene cluster. (e) PPI analysis of 43 key genes. The size of the node indicates the strength of the connectivity between them, the different colors shown in the
node represent different pathways. (f) The mRNA expression results (OTA/CTL) compared with RNA sequencing.

6
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

3.4. OTA treatment altered miRNA expression 2 cells (Fig. 7b). When Ca2 + inhibitor BAPTA-AM was added, the
expression level of the TJ protein was significantly higher than that of
A total of 93 DEmiRNAs were identified by pairwise comparison OTA alone (P < 0.05, Fig. 7c). All of the results suggested that the
among the three groups (CTL, DMSO, and OTA) (Fig. 4a). Next, Venn change of Ca2+ level caused the change of TJ protein expression.
analysis was used to screen DEmiRNAs caused by OTA rather than
DMSO (Fig. 4b). 4. Discussion
To understand the function of the DEmiRNAs, we performed the GO
and KEGG analysis of DEmiRNA target genes. The GO results showed As an environmental pollutant, OTA is a common mycotoxin in food
that the target genes mostly focused on biological process (BP), such as and feed that is almost impossible to avoid and can cause health issues
response to external stimulus, growth, and regulation of cytokine pro­ and economic loss (Chen et al., 2018; Lan et al., 2020). The intestine is
duction (Fig. 4c). For KEGG analysis, circadian rhythm, WNT signaling the first tract to interact with mycotoxins, and the intestinal toxicity of
pathway, and MAPK signaling pathway were most enriched after OTA mycotoxins has attracted attention. Many studies have identified
treatment (Fig. 4d). According to the function prediction and target of OTA-induced intestinal toxicity in vitro (Peng et al., 2019; Wang et al.,
key DEmRNAs, miR-1258-x (target FZD4, Cav1, CCND1, Rasgrp3, 2018), but there were few findings in vivo (Tong et al., 2020). Conse­
Foxp1), mmu-miR-1258–3p (target Plau, E2f5, Hspb1), mmu-miR- quently, in this study, we reported for the first time that mRNA and
122–5p (target WNT5a), miR-205-z (target Cav1, ccm2), mmu-miR- ncRNA (lncRNA, miRNA, and circRNA) were related to OTA-induced TJ
1981–5p (target WNT5a), mmu-miR-1943–5p (target Igf2, Axin2, Pkd1), protein disruption in the jejunum of Balb/c mice by using whole
and mmu-miR-146b-5p (target ccl8, Lgr6) were possibly key DEmiRNAs transcriptome.
related to intestinal barrier function especially for TJ proteins (Fig. 4e). In general, our results have confirmed that OTA damaged the in­
According to the results by RT-qPCR, mmu-miR-203b-3p, mmu-miR- testinal barrier function of Balb/c mice (Fig. 1). The concentrates of D-
28c, mmu-miR-196a-5p, and mmu-miR-216b-5p were all consistent lactate and I-FABP were significantly increased by OTA (P < 0.05). Once
with RNA sequencing results (Fig. 4f). the intestinal barrier injury occurred, a large amount of D-lactate and I-
FABP will be released into the blood, which can increase blood levels (Ji
3.5. OTA exposure altered lncRNA expression et al., 2018). The epithelial monolayer along with the microstructure of
villi and crypts can control the entry of pathogens and exogenous sub­
We identified 640 DElncRNAs in all the three groups (CTL, DMSO, stances (Sumigray et al., 2018). In the present study, we observed that
and OTA) after pairwise comparison (Fig. 5a). To obtain the DElncRNAs OTA injured the structure of villus and crypts, which was consistent with
induced by OTA, Venn analysis was performed to eliminate the influ­ Ruan’s results. The results indicated that the intestinal villi of ducks
ence of DMSO, OTA/CTL and OTA/DMSO but not DMSO/CTL were the became blunt, the epithelium exfoliated, the length of villi decreased,
target DElncRNAs of OTA-treated samples (Fig. 5b). and the depth of crypts was increased by OTA (Ruan et al., 2019). Mucin
Function prediction of the DElncRNAs obtained above was per­ is produced and secreted by goblet cells, OTA reduced the number of
formed on the annotation of their target mRNAs. For GO annotation, goblet cells, suggesting that OTA impaired intestinal function through
most of the DElncRNAs were enriched in cellular component (CC) and decreasing goblet cells, which was consistent with previous studies
BP, shown in extracellular space, extracellular region, immune response, (Feng et al., 2019; Gao et al., 2017; Huang et al., 2019a; Wu et al., 2019).
and response to external stimulus (Fig. 5c). The DElncRNAs target bar­ TJ proteins are an important part of the intestinal barrier, and play a
plot shows that the cGMP-PKG signaling pathway, MAPK signaling barrier role in structure and function to prevent the paracellular
pathway, cAMP signaling pathway, and gap junction were found to be permeation of luminal substances (Tsukita et al., 2001; Gao et al., 2020).
enriched (Fig. 5d). After function prediction of target DEmRNAs, the In our study, TJ proteins deleteriously affected (expression and locali­
lncRNAs Zeb1, Stk36, Phkb, Prss23, Inpp5e, and Gm28588 were tightly zation) by OTA, such as claudin-4, occludin, and ZO-1 (Fig. 2), which
associated with intestinal TJ proteins (Fig. 5e). According to the RT- was consistent with previous experimental results (Gao et al., 2017).
qPCR results, the expression levels of Gm5614 and Srsf5 were Compared with the control group, the protein expression of occludin and
increased, yet Npr3 and Mirt2 were decreased after OTA treatment. All TJ protein 1 in ducks fed with OTA were significantly decreased, and the
these were consistent with RNA sequencing results (Fig. 5f). intestinal barrier was damaged (Ruan et al., 2019). Previous studies
have shown that OTA resulted in intestinal barrier dysfunction, though
3.6. The ceRNA regulation network of TJ proteins the elaborate mechanism is still unknown. In the present study, a whole
transcriptome was first used to investigate the molecular mechanism of
Using the targets pairs DEmiRNA-DEmRNA and DElncRNA- OTA intestinal toxicity on Balb/c mice. Forty-three key genes were used
DEmRNA, ceRNA analysis was performed to obtain further insights for PPI analysis (Fig. 3), most key genes were tightly shared in the WNT
into the role of ncRNAs and mRNAs in intestinal TJ proteins. The rela­ signaling pathway. Yet, although Axin2, Lamc2, and Plau in the PPI
tionship between DEmRNAs and DEncRNAs related to TJ proteins was network did not belong to the WNT signaling pathway, they were also
depicted in Fig. 6. The analysis of key DEmRNAs showed that the WNT/ WNT target genes (Tanigawa et al., 2011). The WNT signal is a chief
Ca2+ signal cascade played a key role in damaging TJ proteins induced switch of self-renewal tissue homeostasis, responsible for sustaining
by OTA. KEGG analysis indicated that FZD4, WNT5a, and Cav1 partic­ intestinal homeostasis, and intestinal epithelial cells (IECs) proliferation
ipated in regulating the levels of Ca2+. Through ceRNA, the upstream and migration (Mah et al., 2016). WNT ligand binding to the FZD re­
target DEmiRNAs and DElncRNAs involved in the regulation of Ca2+ ceptor can activate a variety of intracellular transduction molecules,
signal was found. It was found that lncRNA Zeb1 regulated the expres­ including β-catenin and Ca2+, which were activated through canonical
sion of FZD4 binding with WNT5a to activate the Ca2+ signal cascade by or non-canonical signaling pathways, respectively (Bhanot et al., 1996;
targeting miR-1258-x. Moreover, Cav1 and several lncRNAs combined Slusarski et al., 1997). Additionally, researchers have shown that the
competitively with miR-205-z to regulate the release of Ca2+. WNT signal was also related with the TJ protein (Xing et al., 2020; Yang
et al., 2021). In our study, Cacna1d and Cav1, found in the PPI network,
3.7. Verification that OTA induces TJ protein damage through the Ca2+ shared the release of Ca2+. We speculated that OTA regulated TJ protein
signal cascade in vitro expression through the WNT/Ca2+ signaling pathway activated by the
binding of FZD4 and WNT5a. Wang et al. (2019c) showed that the
To further verify the effect of Ca2+ on the expression of TJ proteins, non-canonical WNT (WNT/Ca2+) signaling pathway mediated the
we used an in vitro cell model. OTA increased the level of Ca2+ (Fig. 7a) regulation of TJ proteins. Likewise, a previous study has also shown that
and decreased the expression level of TJ proteins in differentiated Caco- deoxynivalenol induced intestinal integrity damage and intestinal

7
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

Fig. 4. The overview of the DEmiRNAs. (a) OTA changed the number of DEmiRNAs. (b) Venn analysis between different treatments of DEmiRNAs. (c,d) The GO and
KEGG annotation of the target genes. (e) Sankey diagram of key DEmiRNAs and target key DEmRNA network. (f) The DEmiRNA expression results (OTA/CTL)
compared with RNA sequencing.

8
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

Fig. 5. The analysis of DElncRNAs. (a) The number of DElncRNAs after OTA treatment. (b) The Venn analysis of three comparison groups. (c,d) The GO annotation
and KEGG analysis of DElncRNA target mRNAs. (e) Sankey diagram of key DElncRNAs and the target key DEmRNA networks. (f) The DElncRNA expression results
(OTA/CTL) compared with RNA sequencing.

9
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

Fig. 6. LncRNA-miRNA-mRNA ceRNA network of TJ protein in Balb/c mice. The ellipse represents mRNA, V shape represents miRNA, and the diamond repre­
sents lncRNA.

Fig. 7. The verification of Ca2+ in Caco-2 cells. (a) The changes of Ca2+ level after OTA treatment measured by calcium fluorescent probe Fluo-4 AM. (b,c) The effects
of OTA and BAPTA-AM on claudin-4/occludin expression by western blot analysis and quantification. Different letters (a-c) indicate significant differences
(P < 0.05). BA represents the BAPTA-AM.

10
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

barrier dysfunction by activating the WNT β-catenin signal pathway distribution induced by OTA (Wang et al., 2018).
(Zhou et al., 2020). Based on the in vivo whole transcriptome and in vitro verification, we
MiRNA was considered to be a key element in maintaining and profiled an mRNA-miRNA-lncRNA ceRNA network to elucidate the
improving intestinal barrier function (Lee et al., 2015). In our results, OTA-induced intestinal toxicity in Balb/c mice. This ceRNA network
miR-122, miR-146b, miR-203, and miR-1258 were found to regulate TJ revealed that OTA disrupted TJ proteins through the WNT/Ca2+
protein expression and intestinal barrier function (Fig. 4). Ye et al. signaling pathway. We found that there were two upstream mecha­
(2011) identified that miRNA-122 had a share in regulating intestinal nisms, (i) lncRNA Zeb1 regulates FZD4 binding with WNT5a to release
permeability. Another study indicated that miR-122a induced specific the Ca2+ by targeting miR-1258-x, and (ii) miRNA-1258-x regulated the
mRNA degradation by binding to the nc region of occludin mRNA, expression of Cav1, which controlled extracellular Ca2+ entry.
leading to occludin protein depletion (Zhang et al., 2017). MiR-146b has In conclusion, this study explored the molecular mechanism of OTA-
been shown to regulate the growth and development of the intestinal induced disruption of intestinal TJ proteins in Balb/c mice by using the
tract and IECs (Liao et al., 2013). This can improve the barrier function whole transcriptome combined with in vitro verification. OTA exposure
of colonic midgut epithelium in dextran sodium sulfate treated mice regulated the expression of mRNAs/ncRNAs in many aspects, resulting
(Nata et al., 2013). MiRNA analysis revealed that miR-203 is one of the in compromised TJ proteins and impairment of intestinal barrier func­
differentially regulated miRNAs targeting TJ proteins (Veltman et al., tion. The wide distribution of OTA exposure in the environment is a
2012). Compared with NCM460 normal colon epithelial cells, miR-1258 serious threat to animals and human health. Our results will provide
was down-regulated in tumor tissues and colorectal cancer cell lines and strong data support for elucidating the intestinal toxicological mecha­
regulated the expression of CCND1 (Zhang et al., 2018). nism of OTA and reducing the threat induced by it. These results may
LncRNA also played a vital role in the intestinal barrier, especially provide new evidence for improving our understanding of the hazards
for constituent IECs (Jiang and Zhang, 2020). In our study, OTA induced and risks that OTA poses to public health and the environment.
the changes of lncRNAs expression, including Zeb1, Phkb, Micu1, Mirt2,
Gm28588, and so on (Fig. 5). Micu1 maintained intestinal barrier CRediT authorship contribution statement
integrity by regulating oxidative stress and cell proliferation. After
Micu1 silencing, the integrity of the Caco-2 cell barrier was destroyed Xue Yang: Conceptualization, Methodology, Software, Investiga­
with increased cell bypass permeability and the decreased expression of tion, Writing – original draft, Data curation, Writing – review & editing.
TJ proteins (claudin-1, occludin, and ZO-1) (Leng et al., 2016). Mirt2 has Gao Yanan: Conceptualization, Writing – review & editing, Validation.
a protective effect on endotoxemia-induced mouse death by regulating Huang Shengnan: Investigation, Software. Su Chuanyou: Methodol­
inflammation and recovering intestinal barrier function (Du et al., ogy, Visualization. Zheng Nan: Conceptualization, Supervision, Project
2017). In this literature, lncRNA Micu1 regulated the Mapk4, lncRNA administration, Writing – review & editing, Validation. Wang Jiaqi:
Mirt2 targeted to LPAR2, in which LPAR2 bore a role in the regulation of Conceptualization, Supervision.
the TJ protein (Shukla et al., 2020).
With circRNA/lncRNA as a decoy, miRNA as the center, and mRNA
Declaration of Competing Interest
as a target, there were two ceRNA regulation mechanisms, one for
circRNA-miRNA-mRNA and the other for lncRNA-miRNA-mRNA (Zhang
The authors declare that they have no known competing financial
et al., 2019). In the present study, we did not find the key circRNA
interests or personal relationships that could have appeared to influence
related to TJ proteins and intestinal barrier (Fig S1), so we found the
the work reported in this paper.
related DElncRNAs and DEmiRNAs by constructing a
lncRNA-miRNA-mRNA ceRNA network (Fig. 6). Previous results showed
that the WNT signaling pathway played a key role in TJ protein Acknowledgements
compromise induced by OTA. As the target gene of the WNT signal,
Axin2 was regulated by lncRNA Eya3, Tafld, C4a, and Tomm401 via This work was supported by the National Natural Science Foundation
binding with mmu-miR-1981–5p and mmu-miR-1943–5p. CCND1, also of China (31972190), the Modern Agro-Industry Technology Research
as the target gene of the WNT signal cascade, regulated by lncRNA phkb System of the PR China (CARS-36); the Scientific Research Project for
targeted the miR-1258-x as a ceRNA, which was consistent with Zhang Major Achievements of Agricultural Science and Technology Innovation
et al. (2018). MiRNA lin-4-z, also found in the ceRNA network, con­ Program (CAAS-ZDXT2019004); and the Agricultural Science and
tained many lncRNA targeting sites, including Stk36, Trim65, Technology Innovation Program (ASTIP-IAS12).
MSTRG.10317.1, Mir22hg, Ubxn8, MSTRG.12356.1, Ly6m, Gm43189,
MSTRG12259.3, and Rab11flip3. Lamc2 may be the downstream target Appendix A. Supporting information
of all these DElncRNAs. Interestingly, Lamc2 was also the target of the
WNT signal, which was shown to be related to intestinal mucosal Supplementary data associated with this article can be found in the
inflammation (Coskun et al., 2017). As the ligand and receptor of the online version at doi:10.1016/j.ecoenv.2021.112637.
WNT pathway, the combination of WNT5a and FZD4 will release Ca2+ to
activate the WNT/Ca2+ signal cascade reaction. Cav1, one of the classic References
Ca2+ channels, has a role in the calcium-dependent signal transduction
Akbari, P., Braber, S., Varasteh, S., Alizadeh, A., Garssen, J., Fink-Gremmels, J., 2017.
pathway (Wheeler et al., 2012). It can target miR-205-z and miR-1258-x
The intestinal barrier as an emerging target in the toxicological assessment of
by competing with lncRNA Gm28588, Tut4, and MSTRG.12902.1. mycotoxins. Arch. Toxicol. 91, 1007–1029.
Ca2+ could be a common step in developing OTA-induced toxicity Alizadeh, A., Akbari, P., Varasteh, S., Braber, S., Malekinejad, H., Fink-Gremmels, J.,
2019. Ochratoxin A challenges the intestinal epithelial cell integrity: results obtained
(Ringot et al., 2006; Wang et al., 2018). Increasing the Ca2+ level was
in model experiments with Caco-2 cells. World Mycotoxin J. 12 (4), 1–10.
related to the redistribution of occludin and ZO-1 from the intercellular Al-Sadi, R., Khatib, K., Guo, S., Ye, D., Youssef, M., Ma, T., 2011. Occludin regulates
junctions of colonic epithelium, TJ protein disruption was in parallel to macromolecule flux across the intestinal epithelial tight junction barrier. Am. J.
the elevation of Ca2+ (Gangwar et al., 2017; Wang et al., 2018). In this Physiol. Gastrointest. Liver Physiol. 300, 1054–1064.
Arrieta, M.C., Bistritz, L., Meddings, J.B., 2006. Alterations in intestinal permeability.
study, the results suggested the increase of Ca2+ level was involved in Gut 55, 1512–1520.
decreasing the expression of the TJ protein (Fig. 7). Several studies have Bartel, D.P., 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116,
demonstrated that the inhibitor of Ca2+ BAPTA-AM significantly 281–297.
Bhanot, P., Brink, M., Samos, C.H., Hsieh, J.C., Wang, Y., Macke, J.P., Andrew, D.,
reversed the decrease of TEER, the increase of permeability, the decrease Nathans, J., Nusse, R., 1996. A new member of the frizzled family from Drosophila
of ZO-1 expression, and the interference of ZO-1 and occludin functions as a Wingless receptor. Nature 382, 225–230.

11
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

Bondy, G.S., Pestka, J.J., 2000. Immunomodulation by fungal toxins. J. Toxicol. Environ. Liao, Y., Zhang, M., Lönnerdal, B., 2013. Growth factor TGF-beta induces intestinal
Health B Crit. Rev. 3, 109–143. epithelial cell (IEC-6) differentiation: miR-146b as a regulatory component in the
Chen, W., Li, C., Zhang, B., Zhou, Z., Shen, Y., Liao, X., Yang, J., Wang, Y., Li, X., Li, Y., negative feedback loop. Genes Nutr. 8, 69–78.
Shen, X.L., 2018. Advances in biodetoxification of Ochratoxin A-a review of the past Liu, X., Wu, P., Jiang, W.D., Liu, Y., Jiang, J., Kuang, S.Y., Tang, L., Zhou, X.Q., Feng, L.,
five decades. Front. Microbiol. 9, 1386. 2020. Effects of dietary ochratoxin a on growth performance and intestinal apical
Coskun, M., Soendergaard, C., Joergensen, S., Dahlgaard, K., Riis, L.B., Nielsen, O.H., junctional complex of Juvenile Grass Carp (Ctenopharyngodon idella). Toxins 13 (1),
Sandelin, A., Troelsen, J.T., 2017. Regulation of laminin γ2 expression by CDX2 in 11.
colonic epithelial cells is impaired during active inflammation. J. Cell. Biochem. 118, Ma, X., Zhao, X., Zhang, Z., Guo, J., Guan, L., Li, J., Mi, M., Huang, Y., Tong, D., 2018.
298–307. Differentially expressed non-coding RNAs induced by transmissible gastroenteritis
Creppy, E., 2002. Update of survey, regulation and toxic effects of mycotoxins in Europe. virus potentially regulate inflammation and NF-κB pathway in porcine intestinal
Toxicol. Lett. 127, 19–28. epithelial cell line. BMC Genom. 19, 747.
Du, M., Yuan, L., Tan, X., Huang, D., Wang, X., Zheng, Z., Mao, X., Li, X., Yang, L., Mah, A.T., Yan, K.S., Kuo, C.J., 2016. Wnt pathway regulation of intestinal stem cells.
Huang, K., Zhang, F., Wang, Y., Luo, X., Huang, D., Huang, K., 2017. The LPS- J. Physiol. 594, 4837–4847.
inducible lncRNA Mirt2 is a negative regulator of inflammation. Nat. Commun. 8, Marchiando, A.M., Shen, L., Graham, W.V., Edelblum, K.L., Duckworth, C.A., Guan, Y.,
2049. Montrose, M.H., Turner, J.R., Watson, A.J., 2011. The epithelial barrier is
Feng, L., Chen, S., Zhang, L., Qu, W., Chen, Z., 2019. Bisphenol A increases intestinal maintained by in vivo tight junction expansion during pathologic intestinal
permeability through disrupting intestinal barrier function in mice. Environ. Pollut. epithelial shedding. Gastroenterology 140, 1208–1218.
254, 112960. Maresca, M., Yahi, N., Younès-Sakr, L., Boyron, M., Caporiccio, B., Fantini, J., 2008. Both
Fog Nielsen, K., 2003. Mycotoxin production by indoor molds. Fungal Genet. Biol. 39, direct and indirect effects account for the pro-inflammatory activity of
103–117. enteropathogenic mycotoxins on the human intestinal epithelium: Stimulation of
Galarza-Seeber, R., Latorre, J.D., Bielke, L.R., Kuttappan, V.A., Wolfenden, A.D., interleukin-8 secretion, potentiation of interleukin-1β effect and increase in the
Hernandez-Velasco, X., Merino-Guzman, R., Vicente, J.L., Donoghue, A., Cross, D., transepithelial passage of commensal bacteria. Toxicol. Appl. Pharmacol. 228,
Hargis, B.M., Tellez, G., 2016. Leaky gut and mycotoxins: aflatoxin B1 does not 84–92.
increase gut permeability in broiler chickens. Front. Vet. Sci. 3, 10. Marin, D.E., Braicu, C., Dumitrescu, G., Pistol, G.C., Cojocneanu, R., Neagoe, I.B.,
Gambacorta, L., Pinton, P., Avantaggiato, G., Oswald, I.P., Solfrizzo, M., 2016. Grape Taranu, I., 2019. MicroRNA profiling in kidney in pigs fed ochratoxin A
pomace, an agricultural byproduct reducing mycotoxin absorption: in vivo contaminated diet. Ecotoxicol. Environ. Saf. 184, 109637.
assessment in pig using urinary biomarkers. J. Agric. Food Chem. 64, 6762–6771. Nata, T., Fujiya, M., Ueno, N., Moriichi, K., Konishi, H., Tanabe, H., Ohtake, T., Ikuta, K.,
Gangwar, R., Meena, A.S., Shukla, P.K., Nagaraja, A.S., Dorniak, P.L., Pallikuth, S., Kohgo, Y., 2013. MicroRNA-146b improves intestinal injury in mouse colitis by
Waters, C.M., Sood, A., Rao, R., 2017. Calcium-mediated oxidative stress: a common activating NF-κB and improving epithelial barrier function. J. Gene Med. 15,
mechanism in tight junction disruption by different types of cellular stress. Biochem. 249–260.
J. 474, 731–749. Peng, M., Liu, J., Liang, Z., 2019. Probiotic Bacillus subtilis CW14 reduces disruption of
Gao, Y., Li, S., Wang, J., Luo, C., Zhao, S., Zheng, N., 2017. Modulation of intestinal the epithelial barrier and toxicity of ochratoxin A to Caco-2 cells. Food Chem.
epithelial permeability in differentiated Caco-2 cells exposed to aflatoxin M1 and Toxicol. 126, 25–33.
Ochratoxin A individually or collectively. Toxins 10, 13. Raiola, A., Tenore, G.C., Manyes, L., Meca, G., Ritieni, A., 2015. Risk analysis of main
Gao, Y., Li, S., Wang, J., Luo, C., Zhao, S., Zheng, N., 2018. Modulation of intestinal mycotoxins occurring in food for children: an overview. Food Chem. Toxicol. 84,
epithelial permeability in differentiated Caco-2 cells exposed to aflatoxin M1 and 169–180.
Ochratoxin A individually or collectively. Toxins 10, 13. Ringot, D., Chango, A., Schneider, Y.J., Larondelle, Y., 2006. Toxicokinetics and
Gao, Y., Meng, L., Liu, H., Wang, J., Zheng, N., 2020. The compromised intestinal barrier toxicodynamics of ochratoxin A, an update. Chem. Biol. Inter. 159, 18–46.
induced by mycotoxins. Toxins 12 (10), 619. Ruan, D., Wang, W.C., Lin, C.X., Fouad, A.M., Chen, W., Xia, W.G., Wang, S., Luo, X.,
Gao, Y., Bao, X., Meng, L., Liu, H., Wang, J., Zheng, N., 2021. Aflatoxin B1 and aflatoxin Zhang, W.H., Yan, S.J., Zheng, C.T., Yang, L., 2019. Effects of curcumin on
M1 induce compromised intestinal integrity through clathrin-mediated endocytosis. performance, antioxidation, intestinal barrier and mitochondrial function in ducks
Toxins 13, 3. fed corn contaminated with ochratoxin A. Animal 13, 42–52.
Guttman, M., Amit, I., Garber, M., French, C., Lin, M.F., Feldser, D., Huarte, M., Zuk, O., Shukla, P.K., Meena, A.S., Gangwar, R., Szabo, E., Balogh, A., Chin Lee, S.,
Carey, B.W., Cassady, J.P., Cabili, M.N., Jaenisch, R., Mikkelsen, T.S., Jacks, T., Vandewalle, A., Tigyi, G., Rao, R., 2020. LPAR2 receptor activation attenuates
Hacohen, N., Bernstein, B.E., Kellis, M., Regev, A., Rinn, J.L., Lander, E.S., 2009. radiation-induced disruption of apical junctional complexes and mucosal barrier
Chromatin signature reveals over a thousand highly conserved large non-coding dysfunction in mouse colon. FASEB J. 34, 11641–11657.
RNAs in mammals. Nature 458, 223–227. Slusarski, D.C., Corces, V.G., Moon, R.T., 1997. Interaction of Wnt and a Frizzled
Halstensen, A.S., Nordby, K.C., Elen, O., Eduard, W., 2004. Ochratoxin A in grain dust– homologue triggers G-protein-linked phosphatidylinositol signalling. Nature 390,
estimated exposure and relations to agricultural practices in grain production. Ann. 410–413.
Agric. Environ. Med. 11, 245–254. Steed, E., Balda, M.S., Matter, K., 2010. Dynamics and functions of tight junctions.
Huang, X., Gao, Y., Li, S., Wu, C., Wang, J., Zheng, N., 2019a. Modulation of mucin Trends Cell Biol. 20, 142–149.
(MUC2, MUC5AC and MUC5B) mRNA expression and protein production and Sumigray, K.D., Terwilliger, M., Lechler, T., 2018. Morphogenesis and
secretion in Caco-2/HT29-MTX co-cultures following exposure to individual and compartmentalization of the intestinal crypt. Dev. Cell 45 (2), 183–197.
combined aflatoxin M1 and Ochratoxin A. Toxins 11, 132. Taborda, M.I., Ramírez, S., Bernal, G., 2017. Circular RNAs in colorectal cancer: Possible
Huang, Y., Xu, Y., Lu, Y., Zhu, S., Guo, Y., Sun, C., Xu, L., Chen, X., Zhao, Y., Yu, B., roles in regulation of cancer cells. World J. Gastrointest. Oncol. 9, 62–69.
Yang, Y., Wang, Z., 2019b. lncRNA Gm10451 regulates PTIP to facilitate iPSCs- Tang, Z., Liu, X., Wang, Y., Chen, Q., Hammock, B.D., Xu, Y., 2019. Nanobody-based
derived β-like cell differentiation by targeting miR-338-3p as a ceRNA. Biomaterials fluorescence resonance energy transfer immunoassay for noncompetitive and
216, 119266. simultaneous detection of ochratoxin a and ochratoxin B. Environ. Pollut. 251,
Jaksic, D., et al., 2020. Post-flood impacts on occurrence and distribution of mycotoxin- 238–245.
producing Aspergilli from the sections Circumdati, Flavi, and Nigri in indoor Tanigawa, S., Wang, H., Yang, Y., Sharma, N., Tarasova, N., Ajima, R., Yamaguchi, T.P.,
environment. J. Fungi 6. Rodriguez, L.G., Perantoni, A.O., 2011. Wnt4 induces nephronic tubules in
Ji, J., Gu, Z., Li, H., Su, L., Liu, Z., 2018. Cryptdin-2 predicts intestinal injury during metanephric mesenchyme by a non-canonical mechanism. Dev. Biol. 352, 58–69.
heatstroke in mice. Int J. Mol. Med. 41, 137–146. Tong, C., Li, P., Yu, L.H., Li, L., Li, K., Chen, Y., Yang, S.H., Long, M., 2020. Selenium-rich
Jiang, Z., Zhou, X., Li, R., Michal, J.J., Zhang, S., Dodson, M.V., Zhang, Z., Harland, R.M., yeast attenuates ochratoxin A-induced small intestinal injury in broiler chickens by
2015. Whole transcriptome analysis with sequencing: methods, challenges and activating the Nrf2 pathway and inhibiting NF-KB activation. J. Funct. Foods 66,
potential solutions. Cell Mol. Life Sci. 72, 3425–3439. 103784.
Jiang, Z.-F., Zhang, L., 2021. LncRNA: a potential research direction in intestinal barrier Tsukita, S., Furuse, M., Itoh, M., 2001. Multifunctional strands in tight junctions. Nat.
function. Dig. Dis. Sci. 66, 1400–1408. Rev. Mol. Cell Biol. 2 (4), 285–293.
Kanehisa, M., Araki, M., Goto, S., Hattori, M., Hirakawa, M., Itoh, M., Katayama, T., Turner, J.R., 2009. Intestinal mucosal barrier function in health and disease. Nat. Rev.
Kawashima, S., Okuda, S., Tokimatsu, T., Yamanishi, Y., 2008. KEGG for linking Immunol. 9, 799–809.
genomes to life and the environment. Nucleic Acids Res. 36, 480–484. Veltman, K., Hummel, S., Cichon, C., Sonnenborn, U., Schmidt, M.A., 2012.
Lan, M., Zhang, Y., Wan, X., Pan, M.H., Xu, Y., Sun, S.C., 2020. Melatonin ameliorates Identification of specific miRNAs targeting proteins of the apical junctional complex
Ochratoxin A-induced oxidative stress and apoptosis in porcine oocytes. Environ. that simulate the probiotic effect of E. coli Nissle 1917 on T84 epithelial cells. Int. J.
Pollut. 256, 113374. Biochem Cell Biol. 44, 341–349.
Lee, J., Park, E.J., Yuki, Y., Ahmad, S., Mizuguchi, K., Ishii, K.J., Shimaoka, M., Wang, C., Yang, Y., Zhang, G., Li, J., Wu, X., Ma, X., Shan, G., Mei, Y., 2019a. Long
Kiyono, H., 2015. Profiles of microRNA networks in intestinal epithelial cells in a noncoding RNA EMS connects c-Myc to cell cycle control and tumorigenesis. Proc.
mouse model of colitis. Sci. Rep. 5, 18174. Natl. Acad. Sci. U.S.A. 116, 14620–14629.
Leng, Y., Ge, Q., Zhao, Z., Wang, K., Yao, G., 2016. MICU1 may be a promising Wang, C., Feng, Y., Li, B., Zhou, D., Ma, J., Chen, G., Li, N., 2021. CircRNAsin lung-
intervention target for gut-derived sepsis induced by intra-abdominal hypertension. intestinal axis cancer. Curr. Mol. Med. 21, 291–299.
Cell Death Discov. 2, 16080. Wang, H., Zhai, N., Chen, Y., Fu, C., Huang, K., 2018. OTA induces intestinal epithelial
Li, N., Liu, X.L., Zhang, F.L., Tian, Y., Zhu, M., Meng, L.Y., Dyce, P.W., Shen, W., Li, L., barrier dysfunction and tight junction disruption in IPEC-J2 cells through ROS/Ca2+-
2020. Whole-transcriptome analysis of the toxic effects of zearalenone exposure on mediated MLCK activation. Environ. Pollut. 242, 106–112.
ceRNA networks in porcine granulosa cells. Environ. Pollut. 261, 114007. Wang, H., Jin, J., Wu, J., Qu, H., Wu, S., Bao, W., 2019b. Transcriptome and chromatin
Li, X., Yang, L., Chen, L.L., 2018. The biogenesis, functions, and challenges of circular accessibility in porcine intestinal epithelial cells upon Zearalenone exposure. Sci.
RNAs. Mol. Cell 71, 428–442. Data 6, 298.

12
X. Yang et al. Ecotoxicology and Environmental Safety 224 (2021) 112637

Wang, X., Adegoke, E.O., Ma, M., Huang, F., Zhang, H., Adeniran, S.O., Zheng, P., Yi, F., Zhang, P., Wang, Y., Xu, Y., Zhang, Z., Ma, W., Xu, B., Xia, Q., Du, Q., 2019. Long
Zhang, G., 2019c. Influence of Wilms’ tumor suppressor gene WT1 on bovine Sertoli non-coding RNA slincRAD functions in methylation regulation during the early stage
cells polarity and tight junctions via non-canonical WNT signaling pathway. of mouse adipogenesis. RNA Biol. 16, 1401–1413.
Theriogenology 138, 84–93. Ying, C., Hong, W., Nianhui, Z., Chunlei, W., Kehe, H., Cuiling, P., 2019. Nontoxic
Wei, W., Qian, Y., Wu, Y., Chen, Y., Peng, C., Luo, M., Xu, J., Zhou, Y., 2020. concentrations of OTA aggravate DON-induced intestinal barrier dysfunction in
Detoxification of ochratoxin A by Lysobacter sp. CW239 and characteristics of a IPEC-J2 cells via activation of NF-κB signaling pathway. Toxicol. Lett. 311, 114–124.
novel degrading gene carboxypeptidase cp4. Environ. Pollut. 258, 113677. Yu, D., Marchiando, A.M., Weber, C.R., Raleigh, D.R., Wang, Y., Shen, L., Turner, J.R.,
Wheeler, D.G., Groth, R.D., Ma, H., Barrett, C.F., Owen, S.F., Safa, P., Tsien, R.W., 2012. 2010. MLCK-dependent exchange and actin binding region-dependent anchoring of
Ca(V)1 and Ca(V)2 channels engage distinct modes of Ca2+ signaling to control ZO-1 regulate tight junction barrier function. Proc. Natl. Acad. Sci. U.S.A. 107,
CREB-dependent gene expression. Cell 149, 1112–1124. 8237–8241.
Wu, C., Gao, Y., Li, S., Huang, X., Bao, X., Wang, J., Zheng, N., 2019. Modulation of Zhang, B., Tian, Y., Jiang, P., Jiang, Y., Li, C., Liu, T., Zhou, R., Yang, N., Zhou, X., Liu, Z.,
intestinal epithelial permeability and mucin mRNA (MUC2, MUC5AC, and MUC5B) 2017. MicroRNA-122a regulates zonulin by targeting EGFR in intestinal epithelial
expression and protein secretion in Caco-2/HT29-MTX co-cultures exposed to dysfunction. Cell Physiol. Biochem. 42, 848–858.
aflatoxin M1, Ochratoxin A, and zearalenone individually or collectively. Toxicol. Zhang, Z., Li, J., Huang, Y., Peng, W., Qian, W., Gu, J., Wang, Q., Hu, T., Ji, D., Ji, B.,
Lett. 309, 1–9. Zhang, Y., Wang, S., Sun, Y., 2018. Upregulated miR-1258 regulates cell cycle and
Xing, T., Benderman, L.J., Sabu, S., Parker, J., Yang, J., Lu, Q., Ding, L., Chen, Y.H., inhibits cell proliferation by directly targeting E2F8 in CRC. Cell Prolif. 51, e12505.
2020. Tight junction protein claudin-7 is essential for intestinal epithelial stem cell Zhang, Z., Li, B., Xu, P., Yang, B., 2019. Integrated whole transcriptome profiling and
self-renewal and differentiation. Cell Mol. Gastroenterol. Hepatol. 9, 641–659. bioinformatics analysis for revealing regulatory pathways associated with quercetin-
Yang, K., Zhu, J., Wu, J., Zhong, Y., Shen, X., Petrov, B., Cai, W., 2021. Maternal vitamin induced apoptosis in HCT-116 cells. Front. Pharm. 10, 798.
D deficiency increases intestinal permeability and programs Wnt/beta-catenin Zheng, W., Fan, W., Feng, N., Lu, N., Zou, H., Gu, J., Yuan, Y., Liu, X., Bai, J., Bian, J.,
pathway in BALB/C mice. JPEN J. Parent. Enter. Nutr. 45, 102–114. Liu, Z., 2019. The role of miRNAs in Zearalenone-promotion of TM3 cell
Yang, X., Gao, Y., Yan, Q., Bao, X., Zhao, S., Wang, J., Zheng, N., 2019. Transcriptome proliferation. Int. J. Environ. Res. Public Health 16, 16.
analysis of ochratoxin A-induced apoptosis in differentiated Caco-2 cells. Toxins 12, Zhou, J.Y., Lin, H.L., Wang, Z., Zhang, S.W., Huang, D.G., Gao, C.Q., Yan, H.C., Wang, X.
12. Q., 2020. Zinc L-Aspartate enhances intestinal stem cell activity to protect the
Ye, D., Guo, S., Al-Sadi, R., Ma, T.Y., 2011. MicroRNA regulation of intestinal epithelial integrity of the intestinal mucosa against deoxynivalenol through activation of the
tight junction permeability. Gastroenterology 141, 1323–1333. Wnt/β-catenin signaling pathway. Environ. Pollut. 262, 114290.

13

You might also like