- Research
- Open access
- Published:
Selenium elicited an enhanced anti-inflammatory effect in primary bovine endometrial stromal cells with high cortisol background
BMC Veterinary Research volume 20, Article number: 383 (2024)
Abstract
Background
An elevated endogenous cortisol level due to the peripartum stress is one of the risk factors of postpartum bovine uterine infections. Selenium is a trace element that elicits anti-inflammation and antioxidation properties. This study aimed to reveal the modulatory effect of selenium on the inflammatory response of primary bovine endometrial stromal cells in the presence of high-level cortisol. The cells were subjected to lipopolysaccharide to establish cellular inflammation. The mRNA expression of toll-like receptor 4 (TLR4), proinflammatory factors, and selenoproteins was measured with qPCR. The activation of NF-κB and MAPK signalling pathways was detected with Western blot and immunofluorescence.
Results
The pretreatment with sodium selenite (2 and 4 µΜ) resulted in a down-regulation of TLR4 and genes encoding proinflammatory factors, including interleukin (IL)-1β, IL-6, IL-8, tumour necrosis factor α, cyclooxygenase 2, and inducible nitric oxide synthase. Selenium inhibited the activation of NF-κB and the phosphorylation of mitogen-activated protein kinase kinase, extracellular signal-regulated kinase, p38MAPK and c-Jun N-terminal kinase/stress-activated protein kinase. The suppression of those genes and pathways by selenium was more significant in the presence of high cortisol level (30 ng/mL). Meanwhile the gene expression of glutathione peroxidase 1 and 4 was promoted by selenium, and was even higher in the presence of cortisol and selenium.
Conclusions
The anti-inflammatory action of selenium is probably mediated through NF-κB and MAPK, and is augmented by cortisol in primary bovine endometrial stromal cells.
Background
The postpartum reproductive disorders of dairy cows seriously affect their production performance and cause economic losses [1]. Due to the cervical laxity and the damage of endometrial epithelium after calving, cows are prone to uterine infections, such as metritis and endometritis [2, 3], which can impair the bovine fertility [4]. The tight junction between endometrial epithelial cells prevents pathogens from penetrating into the stroma. However, this protection is lost because of endometrial exfoliation, so that the pathogens are more likely to invade the stroma [5]. Escherichia coli (E. coli) is one of the main pathogenic bacteria found in the infected bovine uterine cavity, and can cause endometrial tissue damage and inflammation through their virulence factor lipopolysaccharide [6, 7].
The endometrium defends itself against pathogen invasion mainly through the innate immunity, in which the inflammatory response predominates [8]. Toll-like Receptors (TLR) are among the most studied families of pattern recognition receptors in mammals [9, 10]. Lipopolysaccharide (LPS) is an exogenous ligand for Toll-like receptor 4 (TLR4), which induces an inflammatory response manifested as the increased release of inflammatory mediators and tissue damage [11]. Both the bovine endometrial epithelial cells (BEEC) and stromal cells (BESC) express TLR [11], and the binding of LPS to TLR4-CD14-MD2 receptor complex activates the MyD88-dependent pathway, which further activates the NF-κB and MAPK intracellular signalling and promotes the transcription of genes encoding inflammatory mediators, such as interleukin 1 (IL1B), interleukin 6 (IL6), C-X-C motif chemokine ligand 8 (CXCL8), and tumour necrosis factor (TNF), and prostaglandin-endoperoxide synthase 2 (PTGS2) and inducible nitric oxide synthase 2 (NOS2) [11,12,13].
Selenium (Se) is a trace element that plays a vital role in animal health and performance [14]. The main source of Se is diet or dietary supplements [15], and the nutritional requirement of Se is 300 µg/kg dry matter (DM) for dairy cows [16]. A marked Se deficiency can be observed when Se content is less than 0.05 mg/kg DM [17]. The accumulation of Se in edible part of plants is directly dependent on soil Se concentration, and the heterogeneous distribution of Se making many parts of the world Se deficient [18, 19]. The decreased dry matter intake and the increased demand for nutrition during peripartum period makes the animal more prone to Se deficiency [20], which has been associated with the reduced fertility, and the incidence of retained placenta, mastitis, and metritis [21, 22]. Exogenous Se supplementation has been reported to reduce the incidence of reproductive diseases [21, 23]. Feeding Se-replete cows with a supranutritional Se-yeast supplement during late gestation improves their postpartum antioxidant status and immune responses [24]. In the form of selenocysteine, Se incorporates in selenoproteins such as glutathione peroxidase (GPX) superfamily. Se supplementation in the diet of calves [25] and heifers [26] is often marked with an elevated serum Se content and GPX activity. The Se-containing GPX 1–4 help protect cells from oxidative stress, inflammation, and oxidant-mediated cell death [14]. Studies have shown that Se alleviated the LPS-induced endometrial damage, inhibited TLR4-mediated activation of NF-κB pathway, and reduced the production of inflammatory mediators in mouse uterus [27]. Our laboratory also found that Se supplement relieved the E. coli-induced goat endometritis with less tissue damage and polymorphonuclear cell count in uterine secretions [28], and that Se suppressed the LPS-induced activation of NF-κB and MAPK signalling and the downstream proinflammatory gene expression in BEEC [29].
Cortisol is a kind of glucocorticoid substance, which is widely used for anti-inflammation and immunosuppression purposes [30]. The periparturient cattle endure a series of stressors and have high endogenous cortisol concentrations [31, 32]. The maternal cortisol level increases 3- to 4-fold during calving, and even 5- to 7-fold in cows under metabolic stress [33]. A comparatively high cortisol concentration suppresses innate immune function and increases the risk of uterine infections [34]. Our laboratory has reported that cortisol inhibited endometrial inflammation through NF-κB and MAPK pathways both in vivo and in vitro [35,36,37]. In the presence of high cortisol, Se has been found to relieve endometrial inflammation and promote tissue repair in E. coli-induced endometritis, and has been reported to ameliorate inflammation in LPS-stimulated BEEC [29, 35, 36]. However, as to the stromal cells with high cortisol background, neither the anti-inflammatory activity of Se nor the mechanism of this process has been reported.
Compared with epithelial cells, the stromal cells are more abundant and closer to the vascular system, so the inflammatory response of stromal cells is no less important than that of epithelial cells [38, 39]. Previously, we have revealed the anti-inflammatory effect of Se in BEEC with high cortisol background [29]. Here we used the primary BESC to determine whether there is a similar effect of Se with the presence of high cortisol. We hypothesized that Se protects BESC from LPS-induced inflammatory response with high cortisol background, and the underlying mechanism involves the NF-κB and MAPK pathways, as well as selenoproteins.
Results
Vimentin identification in primary BESC
As shown in Fig. 1a, the cells were stained positive for vimentin, and were mainly spindle or polygonal in shape. The cytoplasm of the positive cells was dark brown, whereas the nucleus was not stained. The purity of the primary BESC was more than 95%. These cells were confirmed to be the primary BESC and can be used for subsequent experiments.
Se inhibited the LPS-induced TLR4 and proinflammatory gene expression
After LPS stimulation for 12 and 24 h, the gene expression of TNF, IL1B, IL6, CXCL8, NOS2, PTGS2, and TLR4 was upregulated (p < 0.01) in various degrees in BESC (Fig. 2). Except for NOS2 and TLR4, the rises of these genes were greater at 24 h than those at 12 h. Among them, the IL1B and IL6 increased (p < 0.01) about 60-fold at 24 h. Se supplement of both 2 and 4 µM down-regulated (p < 0.01) the LPS-induced expression of these genes. However, no difference was found between the two Se supplement groups. Se alone had no effect (p > 0.01) on these gene expression compared to the blank control.
Se inhibited the LPS-induced MYD88 expression and activation of NF-κB and MAPK pathways
LPS stimulation caused an increase (p < 0.01) in MYD88 protein level (Fig. 3b), as well as the elevated (p < 0.05) phosphorylation levels of P65 and IκBα (Fig. 3c and d), and ERK, JNK, P38, and MEK1/2 (Fig. 3e-i), indicating the MyD88-dependent activations of NF-κB and MAPK. Compared with LPS groups, the phosphorylation levels of MYD88 and the key proteins in NF-κB and MAPK pathway decreased (p < 0.05) in cells cotreated with LPS and Se (2 and 4 µM). No difference was found between the two cotreatment groups, or between the control group and the Se (4 µM) group.
Se inhibited TLR4, MYD88, and proinflammatory genes at high cortisol level
As shown in Fig. 4, LPS stimulated (p < 0.01) the expression of mRNAs encoding TLR4, MYD88, and the inflammatory mediators in BESC. Compared with the LPS group, cotreatment of cortisol and LPS decreased (p < 0.01) these gene expressions. On the basis of cortisol and LPS, Se supplement further down-regulated (p < 0.01) the expression of these genes except IL1B, IL6, and CXCL8 at 12 h, and TLR4 and CXCL8 at 24 h (p > 0.05).
Se inhibited MYD88 expression and the activation of NF-κB and MAPK under high cortisol level
As shown in Fig. 5, cortisol reduced (p < 0.01) the LPS-induced MYD88 protein expression, and the key protein phosphorylation in NF-κB and MAPK pathways. Compared with the LPS and cortisol cotreatment (LPS + COR) group, the addition of Se (2 and 4 µM) further reduced (p < 0.05) the phosphorylation levels of key proteins in both pathways (p < 0.05). The reduction in MYD88 level was observed by Se supplementation (p < 0.01). This result suggested that Se can inhibit MyD88/NF-κB and MAPK pathway in BESC at high cortisol level.
Se inhibited the LPS-induced P65 nuclear translocation at high cortisol level
To verify the inhibition of Se on the LPS-induced NF-κB activation at high cortisol level, we used immunofluorescence staining to detect P65 nuclear translocation. As depicted in Fig. 6, LPS stimulated P65 to enter the nucleus (p < 0.01). Both Se and cortisol reduced (p < 0.05) the amount of nuclear P65 in LPS-stimulated BESC. Compared with LPS + COR group, Se supplement further prevented (p < 0.05) P65 from entering the nucleus.
Se promoted GPX1 and GPX4 expression
Se acts as selenoproteins. Therefore, we examined the effect of Se on the gene expression of GPX1 and GPX4 (Fig. 7). As expected, Se supplement up-regulated (p < 0.05) GPX1 and GPX4 expression with or without the presence of LPS, and the increment was more pronounced at 24 h (5- to 8-fold) than that at 12 h (around 2-fold). The increase amplitude of GPX is more obvious (p < 0.01) in cells treated with 4 µM Se than those treated with 2 µM Se. The presence of LPS seemed to reduce (p < 0.05) GPX4 expression because of the differential expression between the LPS group and the LPS and Se cotreatment (LPS + Se) groups, and between the Se + COR group and the LPS, COR, and Se cotreatment (LPS + COR + Se) groups. It seemed to suggest that LPS negatively impact GPX4 expression. Surprisingly, the GPX1 and GPX4 expression was ever higher (p < 0.05) in Se + COR group than that in Se group, and in the LPS + COR + Se group than that in the LPS + Se group.
Discussion
The innate immunity exerts fast, non-specific defense against unfamiliar pathogens [9]. Cattle with uterine disease present a typical innate immune response with increased expression of genes encoding inflammatory cytokines (IL1A, IL1B, IL6, TNF and IL12A), chemokines (CXCL5 and CXCL8), and prostaglandin synthesis enzymes in the endometrium [8]. LPS has been observed to stimulate the proinflammatory factors including IL-6, TNFα, and nitric oxide in bovine endometrial explants [40], and to induce the degradation of IκB and nuclear translocation of NF-κB P65, as well as the fast phosphorylation of mitogen-activated protein kinas proteins, such as JNK, MEK, ERK1/2, and P38 in bovine endometrial cells [11, 41]. The NF-κB pathway activates downstream inflammatory mediators, including IL-1β, IL-6, TNF-α, COX-2, iNOS and IL-8 [42, 43]. The MAPK pathways have been reported to regulate the expression of TNF-α [44]. Ding et al. investigated the whole-transcriptomic gene changes of stromal cells treated with LPS, and revealed the enrichment of differentially expressed genes in immune-related pathways such as TNF signaling pathway, leukocyte-mediated immunity, IL-1β secretion, and NF-κB signaling [45]. In our experiments, LPS stimulated the expression of TLR4, MYD88, TNF, NOS2, IL1B, IL6, CXCL8, and PTGS2, and the key protein phosphorylation of NF-κB and MAPK signalling pathways in BESC. These results were consistent with the studies described above.
Se has a biphasic dose-response, with toxicity at high doses but favorable properties at very low doses [14]. An adequate supply of Se contributes to the reduced risk of cancer, auto-immune diseases, and subfertility, whereas an over-supply of Se increases the risk of endocrine disruption, mental disorders, and cancer [46]. Supra-nutritional doses of Se-containing compounds can be applied as a chemotherapeutic agent to induce oxidation and apoptosis of cancer cells [47, 48]. Se supplementation is critical to the last 60 days of gestation, when the fetus was completely dependent on the dam for its supply of nutrient [49]. In Se-deficient states, cows sacrifice their available Se to ensure adequate Se intake for their calf [50]. The rate of adequate Se in the blood of cattle is 0.08 ~ 0.16 mg/L [51]. Here the Se concentration in basal medium was measured to be 45.63 µg/L, which was between upper limit of Se deficiency and the lower limit of Se sufficiency [52]. After supplementation with 2 and 4 µM Se, and concentration reached Se sufficient levels [53, 54].
The inflammatory status can be related with Se concentration. According to Prabhu et al., the Se-deficient RAW 264.7 macrophage cell line represented with an upregulation of NF-κB and iNOS. Upon LPS stimulation, these cells showed higher iNOS expression and nitric oxide production than the Se-supplemented cells [55]. In porcine brain, dietary Se deficiency activated the iNOS/NF-κB pathway and the downstream inflammatory cytokines, leading to inflammatory lesions [56]. Se supplement has been widely observed to relieve the inflammatory disease process in various tissue types by inhibiting related pathways such as NF-κB and MAPK [57]. Se attenuated TLR4 and its downstream signaling pathways in mouse endometrium and mouse endometrial epithelial cells under LPS stimulation [58]. These results were similar to what we reported in epithelial cells [29]. Here, we showed that 2 and 4 µM Se down-regulated the LPS-induced expression of TLR4 and MYD88, the activation of NF-κB and MAPK pathway, and the downstream proinflammatory mediators, suggesting that Se inhibited the MyD88/NF-κB and MAPK signalling pathways. Se pretreatment dose-dependently down-regulated the rat alveolar TNF and IL1B expression [59], and the LPS-induced phosphorylation of ERK1/2, P38, and JNK in mouse mammary epithelial cells [60]. However, in our previous study, we did not observe any dose-dependent suppression of Se on inflammatory factors and NF-κB and MAPK pathways in epithelial cells [29]. Similarly in this experiment, there was no difference in Se anti-inflammation between 2 and 4 µM Se, which could be related to the narrow range of Se concentration used in our study. The condition of Se deficiency was not investigated in this experiment.
Being absorbed in small intestine, Se is reduced to hydrogen selenide through a chain of reduction reactions related with glutathione and glutathione reductase, then becomes an active Se source in selenoprotein synthesis [27]. The synthesized selenoproteins are transported through bloodstream to different tissues and organs [61, 62]. The GPX is among the well characterized selenoprotein enzymes related to immune functions. GPX1 is the most abundant and ubiquitously expressed GPX, and GPX4 is well known to protect against lipid peroxidation [63, 64]. GPX1 and GPX4 were found to be increased in human lymphocytes upon supplementation of sodium selenite [65]. Se supplementation, in the form of selenomethionine, has been reported to upregulate GPX1 and GPX4 levels in bovine endometrial cells [66]. Similarly, we observed the increased gene expression of GPX1 and GPX4 in BESC pretreated with Se. The GPX1 and GPX4 activities has been observed to increase with the increasing selenomethionine concentration (0 to 200 nM) [67]. One study detected a dose-dependent upregulation of the GPX1 level after Se nanoparticles treatment [68]. Consistently, we observed a higher expression of GPX in cells treated with 4 µM Se than those treated with 2 µM Se. The cytosolic form of GPX4 has been proved to suppress IL-1-driven NF-κB activation and leukotriene biosynthesis [69]. GPX1 and GPX4 dampen phosphorylation cascades predominantly via prevention of inactivation of phosphatases by H2O2 or lipid hydroperoxides [14]. By controlling NF-κB, GPX indirectly regulates the expression of cyclooxygenases and lipoxygenases via MAPK and PTGS2 [14]. Considering the above reports, we proposed that the anti-inflammatory effect of Se could be mediated through GPX1 and GPX4 in BESC. Our results show that LPS stimulation reduced the expression of GPX4, but had no influence on GPX1. There are in vitro studies reporting no change in GPX1 and GPX4 gene expression, but an upregulation of their protein levels in a bovine endometrial cell line [66] and in mouse macrophages [70] stimulated with LPS. But conversely, LPS has been reported to reduce the expression of GPX1 gene in macrophages [71] and mouse uteri [72] and GPX4 protein in rat myocardial tissue [73]. In our result, it is noteworthy that the GPX1 and GPX4 expression was further upregulated by Se in the presence of cortisol. Whether and how cortisol affects Se or selenoproteins was rarely reported. In zebrafish exposed to predation stress, the GPX1 expression in gut was found to be increased, however, the animal’s cortisol level unchanged [74].
Cortisol dampens NF-κB activation and MAPK phosphorylation, thereby inhibiting inflammation in bovine endometrial epithelial and stromal cells, as well as the goat endometrium [35,36,37]. Previously, we found that in the presence of cortisol, Se exerted a greater inhibition on BEEC inflammation than Se treatment alone [29]. Comparably in stromal cells, the addition of Se further down-regulated the expression of inflammatory genes and reduced the phosphorylation of the key proteins of the two pathways as compared with the LPS + COR cotreatment group. This result suggested that the sites of action of COR and Se, although both related with NF-κB and MAPK, may differ. In addition, given that Se acts through selenoproteins, and that the cotreatment of Se and cortisol further up-regulated GPX1 and GPX4 expression, we speculate that cortisol facilitated Se uptake and utilisation by BESC. However, whether cortisol affect selenoprotein synthesis requires further investigation.
Conclusion
Se inhibited the LPS-induced inflammatory response by suppressing NF-κB and MAPK in BESC, and this inhibition was more apparent in the presence of high-level cortisol. Meanwhile Se promoted the expression of GPX1 and GPX4, and their expression was further enhanced under COR treatment. This anti-inflammatory effect of Se with high cortisol background may be related to GPX1 and GPX4.
Methods
Cell culture
The uteri at ovarian stage I (days 1 ~ 4 of the estrous cycle) were collected from cows at a local abattoir to prepare the primary BESC. One uterus was collected for each batch of the cells, and no fewer than three batches of cells were employed for each experiment. Only the tissue with no obvious pathological change from cows free from reproductive diseases was collected. The collected uterus was rinsed with sterile saline and taken back to the laboratory in an ice box filled with iodophor solution. Then, the uterine tissue was cleaned and opened on a clean bench. The endometrium was clipped and cleaned with phosphate-buffered saline (pH values from 7.2 to 7.4) containing 5% dual antibiotic (50 U/mL penicillin/streptomycin). The endometrium was cut into mincemeat-like pieces then rinsed, and configured with 0.4% collagenase II solution (C6885, Sigma, USA) (the volume ratio of endometrial tissue to solution, 1: 1.5) to digest for 60 min. The digested mixture was filtered and the filtrate was centrifuged at 290×g for 5 min to remove the supernatant. After resuspension with the complete medium containing 3% dual antibiotic, 15% fetal bovine serum (A3161002CC, Gibco, Australia), L-glutamine (G8540-100G, Sigma, USA), DMEM-F12 (D8900, Gibco, USA), the cells were inoculated in the 25cm2 cell culture flasks. The cells were then cultured in a 37 °C, 5% CO2 cell culture incubator (Thermo Fisher, USA) with 12-hourly fluid changes until the cells were fully grown. Compared to the epithelial cells, stromal cells adhere to the wall faster (around 12 h), and are more sensitive to trypsin (0458-50G, Amresco, USA). Therefore, we purified the stromal cells by controlling the digestion time (30 ~ 40 s) of trypsin to obtain primary BESC for subsequent experiments. The stromal cells were identified by detecting intracellular vimentin using immunocytochemistry. The primary and secondary antibodies were the Vimentin Antibody (E-5) (sc-373717, Santa Cruz Biotechnology, Inc., USA) and the mouse IgG1 (SC-3877 Santa Cruz Biotechnology, Inc., USA), respectively.
Treatment design
The effect of sodium selenite (Na2SeO3, S5261, Sigma, USA) on BESC viability was determined by a Cell Counting Kit-8 (A311-02-AA, Dojindo Molecular Technologies, Inc, China). The cytotoxicity of Na2SeO3 was measured by a lactate dehydrogenase assay kit (A020-2-2, Jiancheng Bioengineering Research Institute, China). As a result, the concentrations of 2 and 4 µM Na2SeO3 showed little influence on BESC viability (data unpublished, see Additional file 1), and were used in subsequent experiments.
The current study was divided into two parts. First, we aimed to explore the effect of Se on BESC inflammation by detecting the key proteins of NF-κB and MAPK pathways and the proinflammatory genes. The cells were pretreated with Se for 12 h. Then 1 µg/mL LPS (L2880, O55:B4, Sigma, USA) was added to the medium to stimulate the cells. RNA and protein were extracted to detect the relevant inflammatory genes and the pathway proteins, respectively. Next, we observed the effect of Se on the inflammatory response of BESC at high cortisol (H0888, Sigma, USA) levels. The concentration of 30 ng/mL cortisol were selected to mimic high cortisol background [29, 36]. The cells were pretreated with Se for 12 h, followed by LPS and cortisol treatment. The experimental groupings were as follows: the blank control group, the LPS group, the LPS + COR group, and the LPS + COR + Se groups (2 or 4 µM Se).
RNA extraction and quantitative PCR
The BESC was inoculated in the six-well plates (1 × 106 cells/well), and the above experimental treatments were carried out when the cells reached 60%~70% confluence. The BESC was collected at 12 and 24 h. Total RNA from the cells was extracted using a MagicPure 32 Totai RNA Kit (Z-EC521-S1-32, TransGen Biotech, China), and was quantified using a NanoDrop 2000 (Thermo, USA). The A260/280 ratio of each sample was between 1.8 and 2.1. The obtained RNA was converted to cDNA with a TransScript Uni ALL-in-One First-Strand cDNA Synthesis SuperMix for qPCR (#AU341-02-V2, TransGen Biotech, China), and the reverse transcription system included 4 µL of 5×All Mix, 1 µL of Remover, and 15 µL of RNA + ddH2O, 20 µL in total. QPCR was performed using a CFX 96 Real-Time PCR Detection System (BIO-RAD, USA) with a 20 µL amplification system, containing 4 µL of cDNA, 10 µL SYBR, 5 µL of ddH2O, and 0.5 µL of each primer. The sequences of the primers were presented in Table 1. The expression of each gene was normalized against the housekeeping gene actin-β (ACTB). The 2−△△Ct method was used to calculate the relative expression of the genes.
Protein extraction and western blot
The BESC was inoculated in the six-well plates (1 × 106 cells/well), cell treatment was carried out when the cells grew to 80% confluence. After 45–60 min treatment, cells were lysed with the radioimmunoprecipitation assay buffer (C1053, APPLYGEN, China) containing a mixture of protease inhibitors (P1260, APPLYGEN, China) and protein phosphatase inhibitors (P1265, APPLYGEN, China) to obtain protein samples. The protein concentration was determined by a bicinchoninic acid protein assay kit (P0010, Beyotime, China). Each protein sample was mixed with 1/4 volume of 4×SDS-PAGE loading buffer (P1015, Solarbio, China), followed by a 100 °C water bath for 10 min to obtain the final sample. The protein sample of 20 ~ 30 ng was loaded on 8 ~ 10% SDS-polyacrylamide gels for separation, and was then transferred to the polyvinylidene difluoride membranes (HPVH00010, Millipore, Germany). The membranes were cut prior to hybridisation with antibodies. After being soaked with 5% non-fat milk (0.05% Tween, 20% TBST) for 1.5 h at room temperature to block the non-specific binding, the membrane was incubated with 1:1000 dilution of the primary antibody in dilution buffer (Abs954, Absin, China) overnight at 4 °C. Antibodies specific for MYD88 (# 4283), p-P65 (# 3033), P65 (# 8242), p-IκBα (# 2859), IκBα (# 4812), p-ERK1/2 (# 4370), ERK1/2 (# 4695), p-P38 (# 4511), P38 (# 8690), p-JNK (# 4668), JNK (# 9258), GAPDH (# 8884), and the HRP-conjugated goat anti-rabbit secondary antibody (# 7074) were purchased from Cell Signalling Technology (Danvers, MA, USA). Antibodies for MEK1/2 (AF6385) and p-MEK1/2 (AF8035) were purchased from Affinity Biosciences. Then the membrane was incubated with 1:2000 dilution of HRP-conjugated goat anti-rabbit secondary antibody in 5% non-fat milk for 2 h at room temperature. The chemiluminescent signal was developed using the ECL reagents (1810202, Thermo Scientific, USA), and the image was captured by a ChemiScope5300Pro CCD camera (Clinx Science Instruments, China). The band intensity was quantified by the Quantity One software (Bio-Rad, USA).
Immunofluorescence staining
The cells were inoculated in a 24-well plate (1 × 104 cells/well) containing cover glasses. The cell treatment was carried out when the BESC had grown to 30%~40% confluence. After 45 min treatment, the cells were fixed with 4% paraformaldehyde (BL539A, Biosharp, China) for 15 min at room temperature. Then the sample was incubated with TBST containing 0.4% Triton X-100 (ST797, Beyotime, China) for 15 min at room temperature to permeabilize the cells. After 1.5 h incubation with TBST containing 5% Albumin Bovine V (#A8020, Solarbio, China) at room temperature, the cells were incubated with 1:250 dilution of primary antibody (NF-κB P65) in the antibody diluent overnight at 4°C, and with the subsequent FITC-conjugated secondary antibody (A0423, Beyotime, China) at room temperature for 1.5 h. The cell nuclei were stained using DAPI (SD8495, Beyotime Biotechnology, China) at room temperature for 15 min. Finally, the cell slides were removed and placed on the slide, and the slides were sealed after dropping fluorescent anti-quenching solution. The fluorescent distribution of BESC was visualized and captured using a confocal microscopy (Leica TCS SP8 STED, Leica Corporation, Germany). The immunofluorescence signals were quantified by the Image J software (National Institutes of Health, USA).
Statistical analysis
All experiments were repeated at least three times. The SPSS 25.0 (IBM, USA) program was applied for data analysis. Statistically significant differences were calculated by one-way ANOVA, followed by Dunnett’s test. The result was expressed as the means ± standard error of means (SEM). A two- sided p value of < 0.05 was considered statistically significant.
Data availability
The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.
Abbreviations
- BEEC:
-
Bovine endometrial epithelial cells
- BESC:
-
Bovine endometrial stromal cells
- CXCL8:
-
C-X-C motif chemokine ligand 8
- COR:
-
Cortisol
- DM:
-
Dry matter
- E. coli:
-
Escherichia coli
- GPX:
-
Glutathione peroxidase
- IL1B:
-
Interleukin 1
- IL6:
-
Interleukin 6
- LPS:
-
Lipopolysaccharide
- NOS2:
-
Inducible nitric oxide synthase2
- PTGS2:
-
Prostaglandin-endoperoxide synthase 2
- Se:
-
Selenium
- TLR:
-
Toll-like Receptors
- TLR4:
-
Toll-like receptor 4
- TNF:
-
Tumour necrosis factor
References
Bogado Pascottini O, LeBlanc SJ, Gnemi G, Leroy JLMR, Opsomer G. Genesis of clinical and subclinical endometritis in dairy cows. Reproduction. 2023;166:R15–24. https://doi.org/10.1530/REP-22-0452.
LeBlanc SJ, Review. Postpartum reproductive disease and fertility in dairy cows. Animal. 2023;17. https://doi.org/10.1016/j.animal.2023.100781. Suppl 1:100781.
Dubuc J, Denis-Robichaud J. A dairy herd-level study of postpartum diseases and their association with reproductive performance and culling. J Dairy Sci. 2017;100:3068–78. https://doi.org/10.3168/jds.2016-12144.
Sheldon IM, Cronin JG, Bromfield JJ. Tolerance and innate immunity shape the development of postpartum uterine disease and the impact of endometritis in dairy cattle. Annu Rev Anim Biosci. 2019;7:361–84. https://doi.org/10.1146/annurev-animal-020518-115227.
Gilbert RO, Santos NR. Dynamics of postpartum endometrial cytology and bacteriology and their relationship to fertility in dairy cows. Theriogenology. 2016;85:1367–74. https://doi.org/10.1016/j.theriogenology.2015.10.045.
Carneiro LC, Cronin JG, Sheldon IM. Mechanisms linking bacterial infections of the bovine endometrium to disease and infertility. Reprod Biol. 2016;16:1–7. https://doi.org/10.1016/j.repbio.2015.12.002.
Moresco EMY, LaVine D, Beutler B. Toll-like receptors. Curr Biol CB. 2011;21:R488–493. https://doi.org/10.1016/j.cub.2011.05.039.
Sheldon IM, Cronin JG, Healey GD, Gabler C, Heuwieser W, Streyl D, et al. Innate immunity and inflammation of the bovine female reproductive tract in health and disease. Reproduction. 2014;148:R41–51. https://doi.org/10.1530/REP-14-0163.
Fitzgerald KA, Kagan JC. Toll-like receptors and the control of immunity. Cell. 2020;180:1044–66. https://doi.org/10.1016/j.cell.2020.02.041.
Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–20. https://doi.org/10.1016/j.cell.2010.01.022.
Cronin JG, Turner ML, Goetze L, Bryant CE, Sheldon IM. Toll-like receptor 4 and MYD88-dependent signaling mechanisms of the innate immune system are essential for the response to lipopolysaccharide by epithelial and stromal cells of the bovine endometrium. Biol Reprod. 2012;86:51. https://doi.org/10.1095/biolreprod.111.092718.
Herath S, Fischer DP, Werling D, Williams EJ, Lilly ST, Dobson H, et al. Expression and function of toll-like receptor 4 in the endometrial cells of the uterus. Endocrinology. 2006;147:562–70. https://doi.org/10.1210/en.2005-1113.
Guha M, Mackman N. LPS induction of gene expression in human monocytes. Cell Signal. 2001;13:85–94. https://doi.org/10.1016/s0898-6568(00)00149-2.
Barchielli G, Capperucci A, Tanini D. The role of selenium in pathologies: an updated review. Antioxidants. 2022;11:251. https://doi.org/10.3390/antiox11020251.
Mehdi Y, Hornick JL, Istasse L, Dufrasne I. Selenium in the environment, metabolism and involvement in body functions. Molecules. 2013;18:3292–311. https://doi.org/10.3390/molecules18033292.
Clark JH. Nutrient requirements of dairy cattle. USA: National Academy of Sciences; 2014.
Wendel A. Selenium in biology and medicine. Berlin: Springer-; 1989.
Gao J, Liu Y, Huang Y, Lin Z, Bañuelos GS, Lam MHW, et al. Daily selenium intake in a moderate selenium deficiency area of Suzhou, China. Food Chem. 2011;126:1088–93. https://doi.org/10.1016/j.foodchem.2010.11.137.
Khanal DR, Knight AP, Selenium. Its role in livestock health and productivity. J Agric Environ. 2010;11:101–6. https://doi.org/10.3126/aej.v11i0.3657.
Xiao J, Khan MZ, Ma Y, Alugongo GM, Ma J, Chen T, et al. The antioxidant properties of selenium and vitamin E; their role in periparturient dairy cattle health regulation. Antioxid (Basel). 2021;10:1555. https://doi.org/10.3390/antiox10101555.
Spears JW, Weiss WP. Role of antioxidants and trace elements in health and immunity of transition dairy cows. Vet J. 2008;176:70–6. https://doi.org/10.1016/j.tvjl.2007.12.015.
Sordillo LM. Selenium-dependent regulation of oxidative stress and immunity in periparturient dairy cattle. Vet Med Int. 2013;2013:154045. https://doi.org/10.1155/2013/154045.
Wilde D. Influence of macro and micro minerals in the peri-parturient period on fertility in dairy cattle. Anim Reprod Sci. 2006;96:240–9. https://doi.org/10.1016/j.anireprosci.2006.08.004.
Hall JA, Bobe G, Vorachek WR, Kasper K, Traber MG, Mosher WD, et al. Effect of supranutritional organic selenium supplementation on postpartum blood micronutrients, antioxidants, metabolites, and inflammation biomarkers in selenium-replete dairy cows. Biol Trace Elem Res. 2014;161:272–87. https://doi.org/10.1007/s12011-014-0107-4.
Hall JA, Bobe G, Hunter JK, Vorachek WR, Stewart WC, Vanegas JA, et al. Effect of feeding selenium-fertilized alfalfa hay on performance of weaned beef calves. PLoS ONE. 2013;8:e58188. https://doi.org/10.1371/journal.pone.0058188.
Chorfi Y, Girard V, Fournier A, Couture Y. Effect of subcutaneous selenium injection and supplementary selenium source on blood selenium and glutathione peroxidase in feedlot heifers. Can Vet J. 2011;52:1089–94.
An JK, Chung AS, Churchill DG. Nontoxic levels of Se-containing compounds increase survival by blocking oxidative and inflammatory stresses via signal pathways whereas high levels of Se induce apoptosis. Molecules. 2023;28:5234. https://doi.org/10.3390/molecules28135234.
Li H, Yuan C, Wang H, Cui L, Liu K, Guo L, et al. The effect of selenium on endometrial repair in goats with endometritis at high cortisol levels. Biol Trace Elem Res. 2023;202(6):2564–76. https://doi.org/10.1007/s12011-023-03866-y.
Cui L, Zhang J, Guo J, Zhang M, Li W, Dong J, et al. Selenium suppressed the LPS-induced inflammation of bovine endometrial epithelial cells through NF-κB and MAPK pathways under high cortisol background. J Cell Med. 2023;27:1373–83. https://doi.org/10.1111/jcmm.17738.
Straub RH, Cutolo M. Glucocorticoids and chronic inflammation. Rheumatology. 2016;55:ii6–14. https://doi.org/10.1093/rheumatology/kew348.
Laven RA, Peters AR. Bovine retained placenta: aetiology, pathogenesis and economic loss. Vet Rec. 1996;139:465–71. https://doi.org/10.1136/vr.139.19.465.
Dimri U, Ranjan R, Sharma MC, Varshney VP. Effect of vitamin E and selenium supplementation on oxidative stress indices and cortisol level in blood in water buffaloes during pregnancy and early postpartum period. Trop Anim Health Prod. 2010;42:405–10. https://doi.org/10.1007/s11250-009-9434-4.
Horst RL, Jorgensen NA. Elevated plasma cortisol during induced and spontaneous hypocalcemia in ruminants. J Dairy Sci. 1982;65:2332–7. https://doi.org/10.3168/jds.S0022-0302(82)82505-8.
Dhabhar FS. Effects of stress on immune function: the good, the bad, and the beautiful. Immunol Res. 2014;58:193–210. https://doi.org/10.1007/s12026-014-8517-0.
Fang L, Cui L, Liu K, Shao X, Sun W, Li J, et al. Cortisol inhibits lipopolysaccharide-induced inflammatory response in bovine endometrial stromal cells via NF-κB and MAPK signaling pathways. Dev Comp Immunol. 2022;133:104426. https://doi.org/10.1016/j.dci.2022.104426.
Dong J, Qu Y, Li J, Cui L, Wang Y, Lin J, et al. Cortisol inhibits NF-κB and MAPK pathways in LPS activated bovine endometrial epithelial cells. Int Immunopharmacol. 2018;56:71–7. https://doi.org/10.1016/j.intimp.2018.01.021.
Cui L, Zheng Y, Wang H, Dong J, Li J, Song Q, et al. Cortisol inhibits the Escherichia coli-induced endometrial inflammatory response through NF-κB and MAPK pathways in postpartum goats. Anim Reprod Sci. 2020;215:106333. https://doi.org/10.1016/j.anireprosci.2020.106333.
Chapwanya A, Meade KG, Doherty ML, Callanan JJ, Mee JF, O’Farrelly C. Histopathological and molecular evaluation of Holstein-Friesian cows postpartum: toward an improved understanding of uterine innate immunity. Theriogenology. 2009;71:1396–407. https://doi.org/10.1016/j.theriogenology.2009.01.006.
Wagner WC, Hansel W. Reproductive physiology of the post partum cow. Reproduction. 1969;18:493–500. https://doi.org/10.1530/jrf.0.0180493.
Deng Y, Liu B, Mao W, Shen Y, Fu C, Gao L, et al. Regulatory roles of PGE2 in LPS-induced tissue damage in bovine endometrial explants. Eur J Pharmacol. 2019;852:207–17. https://doi.org/10.1016/j.ejphar.2019.03.044.
Sweet MJ, Hume DA. Endotoxin signal transduction in macrophages. J Leukoc Biol. 1996;60:8–26. https://doi.org/10.1002/jlb.60.1.8.
Regueiro V, Moranta D, Campos MA, Margareto J, Garmendia J, Bengoechea JA. Klebsiella pneumoniae increases the levels of toll-like receptors 2 and 4 in human airway epithelial cells. Infect Immun. 2009;77:714–24. https://doi.org/10.1128/IAI.00852-08.
Lv H, Zhu C, Liao Y, Gao Y, Lu G, Zhong W, et al. Tenuigenin ameliorates acute lung injury by inhibiting NF-κB and MAPK signalling pathways. Respir Physiol Neurobiol. 2015;216:43–51. https://doi.org/10.1016/j.resp.2015.04.010.
Yoon WJ, Lee NH, Hyun CG. Limonene suppresses lipopolysaccharide-induced production of nitric oxide, prostaglandin E2, and pro-inflammatory cytokines in RAW 264.7 macrophages. J Oleo Sci. 2010;59:415–21. https://doi.org/10.5650/jos.59.415.
Ding X, Lv H, Deng L, Hu W, Peng Z, Yan C, et al. Analysis of transcriptomic changes in bovine endometrial stromal cells treated with lipopolysaccharide. Front Vet Sci. 2020;7:575865. https://doi.org/10.3389/fvets.2020.575865.
Navarro-Alarcon M, Cabrera-Vique C. Selenium in food and the human body: a review. Sci Total Environ. 2008;400:115–41. https://doi.org/10.1016/j.scitotenv.2008.06.024.
Wallenberg M, Misra S, Björnstedt M. Selenium cytotoxicity in cancer. Basic Clin Pharmacol Toxicol. 2014;114:377–86. https://doi.org/10.1111/bcpt.12207.
Kuršvietienė L, Mongirdienė A, Bernatonienė J, Šulinskienė J, Stanevičienė I. Selenium anticancer properties and impact on cellular redox status. Antioxid (Basel). 2020;9:80. https://doi.org/10.3390/antiox9010080.
Mehdi Y, Dufrasne I. Selenium in cattle: a review. Molecules. 2016;21:545. https://doi.org/10.3390/molecules21040545.
Hefnawy AEG, Tórtora-Pérez JL. The importance of selenium and the effects of its deficiency in animal health. Small Ruminant Res. 2010;89:185–92. https://doi.org/10.1016/j.smallrumres.2009.12.042.
Dargatz DA, Ross PF. Blood selenium concentrations in cows and heifers on 253 cow-calf operations in 18 states. J Anim Sci. 1996;74:2891–5. https://doi.org/10.2527/1996.74122891x.
Suttle NF. Problems in the diagnosis and anticipation of trace element deficiencies in grazing livestock. Vet Rec. 1986;119:148–52. https://doi.org/10.1136/vr.119.7.148.
Sivertsen T, Øvernes G, Østerås O, Nymoen U, Lunder T. Plasma vitamin E and blood selenium concentrations in Norwegian dairy cows: regional differences and relations to feeding and health. Acta Vet Scand. 2005;46. https://doi.org/10.1186/1751-0147-46-177.
Gerloff BJ. Effect of selenium supplementation on dairy cattle. J Anim Sci. 1992;70:3934–40. https://doi.org/10.2527/1992.70123934x.
Prabhu KS, Zamamiri-Davis F, Stewart JB, Thompson JT, Sordillo LM, Reddy CC. Selenium deficiency increases the expression of inducible nitric oxide synthase in RAW 264.7 macrophages: role of nuclear factor-kappab in up-regulation. Biochem J. 2002;366:203–9. https://doi.org/10.1042/BJ20020256.
Zhang Y, Cui J, Lu Y, Huang C, Liu H, Xu S. Selenium deficiency induces inflammation via the iNOS/NF-κB pathway in the brain of pigs. Biol Trace Elem Res. 2020;196:103–9. https://doi.org/10.1007/s12011-019-01908-y.
Jia W, Ding W, Chen X, Xu Z, Tang Y, Wang M, et al. Selenium-containing compound ameliorates lipopolysaccharide-induced acute lung injury via regulating the MAPK/AP-1 pathway. Inflammation. 2021;44:2518–30. https://doi.org/10.1007/s10753-021-01521-z.
Chen Y, Zhao Y, Yang J, Jing H, Liang W, Chen M, et al. Selenium alleviates lipopolysaccharide-induced endometritis via regulating the recruitment of TLR4 into lipid rafts in mice. Food Funct. 2020;11(1):200–10. https://doi.org/2020.
Liu J, Yang Y, Zeng X, Bo L, Jiang S, Du X, et al. Investigation of selenium pretreatment in the attenuation of lung injury in rats induced by fine particulate matters. Environ Sci Pollut Res Int. 2017;24:4008–17. https://doi.org/10.1007/s11356-016-8173-0.
Zhang W, Zhang R, Wang T, Jiang H, Guo M, Zhou E, et al. Selenium inhibits LPS-induced pro-inflammatory gene expression by modulating MAPK and NF-κB signaling pathways in mouse mammary epithelial cells in primary culture. Inflammation. 2014;37:478–85. https://doi.org/10.1007/s10753-013-9761-5.
Santesmasses D, Mariotti M, Gladyshev VN. Bioinformatics of selenoproteins. Antioxid Redox Signal. 2020;33:525–36. https://doi.org/10.1089/ars.2020.8044.
Bulteau AL, Chavatte L. Update on selenoprotein biosynthesis. Antioxid Redox Signal. 2015;23:775–94. https://doi.org/10.1089/ars.2015.6391.
Brigelius-Flohé R, Maiorino M. Glutathione peroxidases. Biochim Biophys Acta. 2013;1830:3289–303. https://doi.org/10.1016/j.bbagen.2012.11.020.
Lubos E, Loscalzo J, Handy DE. Glutathione peroxidase-1 in health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 2011;15:1957–97. https://doi.org/10.1089/ars.2010.3586.
Broome CS, McArdle F, Kyle JA, Andrews F, Lowe NM, Hart CA, et al. An increase in selenium intake improves immune function and poliovirus handling in adults with marginal selenium status. Am J Clin Nutr. 2004;80:154–62. https://doi.org/10.1093/ajcn/80.1.154.
Adeniran SO, Zheng P, Feng R, Adegoke EO, Huang F, Ma M, et al. The antioxidant role of selenium via GPx1 and GPx4 in LPS-induced oxidative stress in bovine endometrial cells. Biol Trace Elem Res. 2022;200:1140–55. https://doi.org/10.1007/s12011-021-02731-0.
Stolwijk JM, Falls-Hubert KC, Searby CC, Wagner BA, Buettner GR. Simultaneous detection of the enzyme activities of GPx1 and GPx4 guide optimization of selenium in cell biological experiments. Redox Biol. 2020;32:101518. https://doi.org/10.1016/j.redox.2020.101518.
Wang S, Chen Y, Han S, Liu Y, Gao J, Huang Y, et al. Selenium nanoparticles alleviate ischemia reperfusion injury-induced acute kidney injury by modulating GPx-1/NLRP3/Caspase-1 pathway. Theranostics. 2022;12:3882–95. https://doi.org/10.7150/thno.70830.
Papp LV, Holmgren A, Khanna KK. Selenium and selenoproteins in health and disease. Antioxid Redox Signal. 2010;12:793–5. https://doi.org/10.1089/ars.2009.2973.
Carlson BA, Yoo MH, Sano Y, Sengupta A, Kim JY, Irons R, et al. Selenoproteins regulate macrophage invasiveness and extracellular matrix-related gene expression. BMC Immunol. 2009;10:57. https://doi.org/10.1186/1471-2172-10-57.
Wang Y, Xiong Z, Li C, Liu D, Li X, Xu J, et al. Multiple beneficial effects of aloesone from aloe vera on LPS-induced RAW264.7 cells, including the inhibition of oxidative stress, inflammation, M1 polarization, and apoptosis. Molecules. 2023;28:1617. https://doi.org/10.3390/molecules28041617.
Shaukat A, Shaukat I, Rajput SA, Shukat R, Hanif S, Huang S, et al. Icariin alleviates Escherichia coli lipopolysaccharide-mediated endometritis in mice by inhibiting inflammation and oxidative stress. Int J Mol Sci. 2022;23:10219. https://doi.org/10.3390/ijms231810219.
Zhou B, Zhang J, Chen Y, Liu Y, Tang X, Xia P, et al. Puerarin protects against sepsis-induced myocardial injury through AMPK-mediated ferroptosis signaling. Aging. 2022;14:3617–32. https://doi.org/10.18632/aging.204033.
Casetta J, Ribeiro RP, Lewandowski V, Khatlab ADS, De Oliveira Neto AR, Boscolo WR, et al. Expression of the PEPT1, CAT, SOD2 and GPX1 genes in the zebrafish intestine supplemented with methionine dipeptide under predation risk. J Anim Physiol Anim Nutr. 2021;105:1214–25. https://doi.org/10.1111/jpn.13535.
Acknowledgements
Not applicable.
Funding
This work was supported by the National Natural Science Foundation of China (NO: 32072937, 31802253, 32102735), the International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions (No: 8), the Postgraduate Research & Practice Innovation Program of Jiangsu Province (KYCX21_3279), the earmarked fund for Jiangsu Agricultural Industry Technology System (JATS[2023]456), the National Key R&D Program of China (2023YFD1801100), the Natural Science Foundation of Jiangsu Province (NO: BK20210808), the 111 Project (D18007), and the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD). The funding bodies were not involved in the study design, data analysis, interpretation of data, or in writing the manuscript.
Author information
Authors and Affiliations
Contributions
L.C.: funding acquisition, supervision, writing-original draft, writing-review and editing. M.Z.: data curation, formal analysis, methodology, writing-original draft. F.Z., Z.W., S.Q. and X.M.: data curation. CY.: funding acquisition, data curation. J.D. and H.W. : funding acquisition. K.L. and L.G.: methodology. J.L.: conceptualization, funding acquisition, supervision. All authors have read and approved the final manuscript, and agreed to be accountable for its contents.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
12917_2024_4240_MOESM1_ESM.pdf
Supplementary Material 1: The effect of Na2SeO3 on the viability and LDH release of primary bovine endometrial stromal cells
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Cui, L., Zhang, M., Zheng, F. et al. Selenium elicited an enhanced anti-inflammatory effect in primary bovine endometrial stromal cells with high cortisol background. BMC Vet Res 20, 383 (2024). https://doi.org/10.1186/s12917-024-04240-3
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s12917-024-04240-3