Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Anti-inflammatory effects of progesterone through NF-κB and MAPK pathway in lipopolysaccharide- or Escherichia coli-stimulated bovine endometrial stromal cells

  • Luying Cui ,

    Contributed equally to this work with: Luying Cui, Xinyu Shao

    Roles Data curation, Funding acquisition, Supervision, Writing – original draft, Writing – review & editing

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

  • Xinyu Shao ,

    Contributed equally to this work with: Luying Cui, Xinyu Shao

    Roles Data curation, Formal analysis, Methodology

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

  • Wenye Sun,

    Roles Methodology

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

  • Fangling Zheng,

    Roles Methodology

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

  • Junsheng Dong,

    Roles Funding acquisition

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

  • Jun Li,

    Roles Supervision

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

  • Heng Wang ,

    Roles Funding acquisition

    sdaulellow@163.com (HW); yzjjli@163.com (JL)

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

  • Jianji Li

    Roles Conceptualization, Funding acquisition, Supervision, Writing – review & editing

    sdaulellow@163.com (HW); yzjjli@163.com (JL)

    Affiliations College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, PR China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, PR China, Joint International Research Laboratory of Agriculture and Agriproduct Safety of the Ministry of Education, Yangzhou, Jiangsu, PR China

Abstract

Postpartum uterine infection in dairy cows is commonly caused by pathogenic bacteria such as Escherichia coli (E. coli). Progesterone elicits immunosuppressive function within bovine endometrium, and has been suggested to be related to postpartum uterine infection. Endometrial stroma is exposed to bacteria due to the disruption of epithelium during parturition, but the effect and mechanism of progesterone on innate immune response of stromal cells has not been reported. This study evaluated the impact of progesterone on inflammatory response of primary endometrial stromal cells stimulated by lipopolysaccharide or heat-killed E. coli. Quantitative PCR analysis revealed that progesterone repressed mRNA induction of IL1B, IL6, TNF, CXCL8, NOS2, and PTGS2 in stromal cells in response to lipopolysaccharide or E. coli challenge. Consistently, Western blot and immunofluorescence staining results showed that progesterone suppressed lipopolysaccharide- or E. coli-induced MAPK and NF-κB activations characterized with decreased phosphorylations of ERK1/2, JNK, P38, IκBα, and P65, and inhibition of P65 nuclear translocation. In unstimulated stromal cells, progesterone alone did not affect the mRNA transcription for IL6, TNF, CXCL8, NOS2, and PTGS2, and the signaling cascade of MAPK and NF-κB, but decreased IL1B mRNA expression. These results revealed that the anti-inflammatory effect of progesterone in lipopolysaccharide- or E. coli-challenged endometrial stromal cells was probably mediated through MAPK and NF-κB pathways.

Introduction

After parturition, the bovine uterine lumen becomes contaminated, and persistence of pathogenic bacteria often leads to uterine infection, such as metritis and endometritis. These uterine impairments are associated with poor reproductive performance even after successful resolution of the disease, and eventually cause huge financial losses [1]. Among various bacteria that contaminate bovine uterus, Escherichia coli (E. coli) are abundant and are the first step in the pathogenesis of postpartum uterine disease in cattle [2].

Classical innate immunity is a principal component of uterine defense and encompasses anatomical, physiological, phagocytic, and inflammatory barriers [1]. The endometrial epithelial barrier is disrupted due to parturition, making the underlying stroma exposed to pathogenic microorganisms. Lipopolysaccharide (LPS) is the main endotoxin of E. coli. Exposure of bovine endometrial cells with LPS or E. coli results in inflammatory response through pattern recognition receptors [2]. Toll-like receptor 4 (TLR4) binds to LPS together with myeloid differentiation factor 2 and CD14 on endometrial cell surface, followed by the activation of nuclear factor-κB (NF-κB) transcription factors and the mitogen-activated protein kinases (MAPK) extracellular signal, including extracellular regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK), and finally results in an enhanced expression of transcripts for cytokines and chemokines such as interleukin 1 beta (IL1β), IL6, tumor necrosis factor-α (TNF-α), IL8, nitric oxide, and prostaglandins [24].

Progesterone has been proved to show immunosuppressive function within the endometrium of ruminants, and the pathogenesis of postpartum uterine infection can be related to progesterone [5]. Normally after parturition, progesterone concentration keeps low or undetectable until after the first ovulation [68]. But an early ovulation could make the cow at a higher risk of uterine infection if uterine involution is incomplete [9]. Spontaneous uterine infection in cattle do not usually develop until after the first postpartum corpus luteum forms and begins producing progesterone [10]. In practice, we found the presence of corpus luteum in about 25% of cows diagnosed with endometritis.

Mechanism underlying the immunosuppressive role of progesterone on uterus of ruminant species was associated with decreased of lymphocyte proliferation, modulation on amount and function of polymorphonuclear leukocyte, and regulation of inflammatory cytokines and chemokines [1013]. Generally, progesterone exhibited an anti-inflammatory property in various cell types [1416], and has been shown to perturbate prostaglandin secretions in bovine endometrial epithelial and stromal cells in response to LPS or E. coli challenge [3]. But the study by Saut et al. found no impact of progesterone on the inflammatory response of bovine endometrial epithelial and stromal cells stimulated by LPS [4]. Previously we verified that progesterone inhibited LPS- or E. coli-induced expression of proinflammatory genes and MAPK and NF-κB pathways in primary bovine endometrial epithelial cells [17]. Compared with epithelial cells, stromal cells are more susceptible to pathogenic E. coli with a stronger binding affinity and a more intense inflammatory response [18]. However, the effect of progesterone on bovine endometrial stromal cells (BESC) remains controversial.

In the present study, we hypothesized that progesterone ameliorates inflammatory response in bovine endometrial stromal cells. Primary BESC was stimulated with LPS or heat-killed E. coli, and was treated with physiological concentrations of progesterone (1, 3, or 5 ng/mL). The changes in mRNA expressions of IL1B, IL6, TNF, C-X-C motif chemokine ligand 8 (CXCL8), nitric oxide synthase 2 (NOS2), and prostaglandin-endoperoxide synthase 2 (PTGS2), and activations of NF-κB and MAPK pathways were determined.

Materials and methods

Cell culture

All experimental procedures were approved by the Animal Care and Use Committee of Yangzhou University (NSFC2020-SYXY24). The bovine uteri without gross evidence of genital disease or infection were collected aseptically at local abattoir and were kept on ice until further processing in the laboratory. The uterine surface was disinfected with iodophor and 75% alcohol and was flushed clean with sterile saline. The uterine horn was dissected longitudinally to expose the endometrium. The epithelium was removed by using a moisturized cotton ball. Then the intercaruncular endometrial stripes were dissected from the myometrial layer with a scalpel blade and were chopped into small pieces. After repeated rinses with phosphate-buffered saline (PBS) supplemented with 100 U/mL penicillin/streptomycin, the minced tissue was digested in DMEM/F12 (D8900, sigma, USA) containing 0.25% collagenase II (C6885-5G, Sigma, USA) at 37°C for 50 min. The cell suspension was filtered through a 450 μm mesh to remove undigested tissue fragments, and the filtrate was washed 3 times by centrifugation (100× g for 5 min) with PBS. The cells were resuspended in DMEM/F12 containing 15% fetal bovine serum and 100 U/mL penicillin/streptomycin and were cultured at 37°C with 5% CO2. The medium was changed 12 h after plating to allow attachment of stromal cells and removal of epithelial cells. The purity of stromal cell population was determined to be more than 95% by the detection of vimentin using immunocytochemistry. The culture media were changed every 24 to 48 h once confluence had been reached approximately 90%.

Experiment design and treatments

Both LPS and heat-killed E. coli were used to induce the inflammatory response in BESC. LPS (L2630) and progesterone (P0130) were purchased from Sigma-Aldrich. LPS was dissolved in DMEM/F12 at a concentration of 1 mg/mL as a stock solution at -20°C, and was diluted to 1 μg/mL with DMEM/F12 during experiment. Progesterone was dissolved and diluted in ethanol to 104 ng/mL as a stock solution at -20°C, and was diluted to 1, 3, or 5 ng/mL with DMEM/F12 as working solutions [17]. The heat-killed E. coli O55:B5 was prepared as previously described [19]. The inactive bacteria were resuspended in DMEM/F12 to a final inoculum of 1×108 CFU/mL. To determine the effect of progesterone on the inflammatory response of BESC, the cells were plated in 6-, 24-, or 96-well plates or 6-cm dishes and were challenged with 1 μg/mL LPS or 1×108 CFU/mL heat-killed E. coli in control medium or medium containing progesterone. One, 3, and 5 ng/mL progesterone were selected because the serum concentration of progesterone normally ranges from 1 to 5 ng/mL in cattle [4]. Based on our previous reports in epithelial cells and a pilot study in BESC, LPS of 1 μg/mL or heat-killed E. coli of 1×108 CFU/mL was able to induce the inflammatory response [19]. The cell viability was determined 24 h after treatment. The relative mRNA abundance of inflammatory genes was detected 12 and 24 h after the cotreatment of LPS and progesterone, or 18 and 24 h after the cotreatment of heat-killed E. coli and progesterone. The changes in key protein levels of NF-κB and MAPK pathways were detected at 45 and 60 min, respectively, in cells with cotreated with LPS and progesterone. The time point for the detection of NF-κB and MAPK pathways was 120 min in cells cotreated with heat-killed E. coli and progesterone. P65 nuclear translocation was detected using immunofluorescence at 45 or 120 min in cells stimulated with LPS or bacteria, respectively. In addition, to observe the impact of progesterone in unstimulated conditions, BESC was treated with progesterone alone for 12 h to detect proinflammatory gene expressions, and for 30 min to detect phosphorylation of key proteins in MAPK and NF-κB pathways. The selection of these time points was based on pre-experimental results.

Cell viability assay

The Cell Counting Kit-8 (CCK-8, Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was used to evaluate the impact of progesterone, LPS, and heat-killed E. coli, either alone or in combination, on the cell viability of BESC. The cells were seeded into wells of a 96-well plate (2×103 cells per well) and grown to 80% confluence. The medium was replaced with DMEM/F12 containing progesterone, LPS and progesterone, or heat-killed E. coli and progesterone. After 24 h treatment, the CCK-8 solution was added to each well, followed by an additional incubation for 2 h. The optical density was read at 450 nm using a microplate reader (Tecan, Austria).

RNA extraction and quantitative PCR

The cells were plated in 6-well plates (2×105 cells per well) and grown to 80% fusion. After the treatment as previously described, the cells were washed with PBS, and the total RNA was subsequently extracted using a Trizol reagent (ET111, TRAN, China) according to the manufacturer’s protocol. The extracted RNA was quantified using a Nanodrop 2000 spectrophotometer (Thermo, USA). The absorption ratio (A260/280) was determined to be between 1.8 and 2.0. The RNA was reverse transcribed into cDNA using the PrimerScript RT regent Kit gDNA Eraser (DRR047A, TaKaRa, Japan). The quantitative PCR was carried out using a CFX 96 Real-Time PCR Detection System (Bio-Rad, USA) as previously described [20]. The 2-△△Ct method was used to analyze the relative mRNA abundance. The actin beta (ACTB) was used as an internal control. A single product was amplified by each primer pair. The products were purified and sequenced (TsingKe Biotech, Beijing, China), and the sequence results were analyzed using BLAST (http://blast.ncbi.nlm.nih.gov/blast.cg) and compared to GenBank database. The sequences of primers were shown in Table 1.

Western blot analysis

The cells were seeded in 6-cm culture dishes (6×105 cells per dish) and grown to 80% confluence. After treatment, the cells were collected and lysed using a RIPA buffer (P0013B, Beyotime, China) containing protease and phosphatase inhibitor cocktail (P1046, Beyotime, China) for routine protein extraction. The total proteins were quantified using a bicinchoninic acid protein assay kit (P0010, Beyotime, China). Cell protein extract (20 to 30 μg) was subjected to a 10% SDS-polyacrylamide gel and was transferred to a polyvinylidene difluoride membrane (Millipore, Germany). The samples were blocked with Tris-buffered saline containing 0.05% Tween 20 and 5% skimmed milk, followed by the incubation with a primary antibody (1:1000 dilution with 5% bovine serum albumin) at 4°C overnight. The primary antibodies specific for β-actin (# 4970), IκBα (# 4812), P-IκBα (# 2859), P65 (# 8242), P-P65 (# 3033), ERK1/2 (# 4695), P-ERK 1/2 (# 4370), JNK (# 9258), P-JNK (# 4668), P38 (# 8690), and P-P38 (# 4511) were purchased from Cell Signaling Technology. The membranes were subsequently incubated with an HRP-conjugated secondary antibody (111-035-003, Jackson ImmunoResearch, USA; 1:10000 dilution with 5% skimmed milk) at room temperature for 1 h. The protein blots were detected and visualized using a chemiluminescence HRP substrate (1810202, Clinx Science Instruments, China) and a ChemiScope 5300 Pro CCD camera (Clinx Science Instruments, China). The blots were quantified by Quantity One software (Bio-Rad, CA, USA).

Immunofluorescence staining

The cells were plated in a 24-well culture plate and were treated according to the experiment design. Then the cells were fixed with 4% paraformaldehyde at room temperature for 12 min. After washing with PBS, the cell membrane was penetrated with 0.4% Triton X-100 (ST797, Beyotime, China) for 15 min. The cells were washed with PBS and were blocked using PBS containing 10% goat serum (ZLI-9021, ZSGB-BIO, China) for 45 min at room temperature. The cells were incubated with a primary antibody specific for NF-κB P65 (#8242, Cell Signaling Technology, USA) with 1:400 dilution at 4°C overnight, and were subsequently incubated with an FITC-conjugated secondary antibody (A11034, Thermo Fisher Scientific, USA) with 1:500 dilution for 1 h at room temperature in dark environment. The nuclei were stained with DAPI (C1005, Beyotime, China). The cells were visualized using a fluorescence microscope (Leica TCS Sp8, Leica company, Germany).

Statistical analysis

All data were presented as the means ± standard error of means (SEM), and were analyzed using the SPSS 26.0 software (IBM, NY, USA). Statistically significant differences were calculated by one-way ANOVA, followed by Least Significant Difference test. A two-sided P-value less than 0.05 was designated as significant. Each experiment was repeated at least three times.

Results

BESC viability

The BESC viability was not influenced (P > 0.05) by the treatment of progesterone, LPS, or heat-killed E. coli individually or in combination (Fig 1).

thumbnail
Fig 1. The changes in cell viability of primary bovine endometrial stromal cells using Cell Counting Kit-8 method.

The cells were treated with 1, 3, or 5 ng/mL progesterone (A), 1 μg/mL lipopolysaccharide and progesterone (B), or 1×108 CFU/mL heat-killed E. coli and progesterone (C) for 24 h. The vehicle is 0.5% alcohol. P4, progesterone. LPS, lipopolysaccharide. All data were presented as means ± SEM (n ≥ 3).

https://doi.org/10.1371/journal.pone.0266144.g001

Proinflammatory gene expression

The impact of progesterone alone on proinflammatory gene expressions was evaluated at 12 h. This time point was selected according to a pre-experimental result (unpublished data, S1 Appendix). The mRNA expressions of IL6, TNF, CXCL8, NOS2, or PTGS2 were unaffected (P > 0.05) by 1, 3, or 5 ng/mL progesterone. However, progesterone decreased (P < 0.05) IL1B mRNA expression (Fig 2A). Based on a pilot study, time points of 12 and 24 h exhibited peak abundance of mRNA transcripts for proinflammatory genes in LPS-treated BESC (unpublished data, S1 Appendix). As shown in Fig 2B, exposure of cells to LPS upregulated (P < 0.05) relative mRNA abundance of IL1B, IL6, TNF, CXCL8, NOS2, and PTGS2. Compared with LPS group, progesterone (1, 3, and 5 ng/mL) decreased (P < 0.05) mRNA expressions of IL1B, IL6, CXCL8, NOS2, and PTGS2 in response to LPS at 12 and 24 h. Down-regulation (P < 0.05) of TNF mRNA expression was only observed in cells cotreated with LPS and 5 ng/mL progesterone at 12 h, and in cells cotreated with LPS and progesterone (1 or 3 ng/mL) at 24 h. The time course experiment showed that heat-killed E. coli induced expression of proinflammatory genes (P < 0.05), and the increase was most prominent at 18 and 24 h (Fig 2C). Progesterone (1, 3, and 5 ng/mL) generally reduced (P < 0.05) IL1B, IL6, TNF, CXCL8, NOS2, and PTGS2 mRNA expression at 18 and 24 h in BESC with bacterial challenge, except (P > 0.05) TNF and NOS2 at 24 h in cells cotreated with E. coli and 5 ng/mL P4 (Fig 2D).

thumbnail
Fig 2. Progesterone treatment attenuated gene expression of inflammatory mediators in primary bovine endometrial stromal cells stimulated with lipopolysaccharide or heat-killed E. coli.

The mRNA expressions of IL1B, IL6, CXCL8, TNF, NOS2, and PTGS2 were assessed by qPCR (normalized to ACTB). A. Unstimulated cells were treated with 1, 3, and 5 ng/mL progesterone for 12 h. B. Cells were cotreated with 1 μg/mL lipopolysaccharide and progesterone (1, 3, and 5 ng/mL) for 12 and 24 h. C. Time-course changes in inflammatory gene expressions in cells treated with 1×108 CFU/mL heat-killed E. coli for 6, 9, 12, 18, and 24 h. D. Cells were cotreated with 1×108 CFU/mL heat-killed E. coli and progesterone (1, 3, and 5 ng/mL) for 18 and 24 h. LPS, lipopolysaccharide. All data were presented as means ± SEM (n ≥ 3). * P < 0.05, difference compared with the control; # P < 0.05, difference compared with the LPS group.

https://doi.org/10.1371/journal.pone.0266144.g002

NF-κB activation

As shown in Fig 3A, progesterone (1, 3, or 5 ng/mL) alone did not influence (P > 0.05) key protein levels in NF-κB pathway. Our preliminary study showed that the phosphorylations of IκBα and P65 elevated at 45 min in response to LPS stimulation (unpublished data, S1 Appendix). The addition of progesterone decreased (P < 0.05) LPS-induced phosphorylations of P65 and IκBα. The heat-killed E. coli resulted in phosphorylations of IκBα and P65 from 30 to 120 min, and was maximal (P < 0.05) at 120 min (Fig 3B). The amount of phosphorylations of IκBα and P65 in cells cotreated with progesterone (1, 3, or 5 ng/mL) and heat-killed E. coli was generally less (P < 0.05) than in cells treated only with heat-killed E. coli.

thumbnail
Fig 3. Progesterone inhibited phosphorylations of IκBα and P65 in primary bovine endometrial stromal cells in response to lipopolysaccharide or heat-killed E. coli stimulation.

The protein level of IκBα, P-IκBα, P65, and P-P65 was determined using Western blot. The protein blots were quantified by Quantity One software. A. Unstimulated cells were treated with 1, 3, and 5 ng/mL progesterone for 30 min. B. The cells were cotreated with 1 μg/mL lipopolysaccharide and progesterone (1, 3, or 5 ng/mL) for 45 min. This time point was based on the results of our previous study. C. Time-course changes in key protein phosphorylation of NF-κB pathway in cells challenged with 1×108 CFU/mL heat-killed E. coli for 30, 60, 90, and 120 min. D. The cells were cotreated with heat-killed E. coli and progesterone for 120 min. LPS, lipopolysaccharide. P4, progesterone. All data were presented as means ± SEM (n ≥ 3). * P < 0.05, difference compared with the control; # P < 0.05, difference compared with the LPS group (B) or the heat-killed E. coli group (D).

https://doi.org/10.1371/journal.pone.0266144.g003

The immunofluorescence results showed translocation of NF-κB P65 into the nucleus in BESC stimulated with LPS and heat-killed E. coli (Fig 4). The addition of 3 ng/mL progesterone reduced the amount of P65 in nucleus in cells challenged with LPS or bacteria.

thumbnail
Fig 4. Progesterone inhibited nuclear translocation of P65 in primary bovine endometrial epithelial cells stimulated with lipopolysaccharide or E. coli.

The cells were cotreated with 1 μg/mL lipopolysaccharide and 3 ng/mL progesterone for 45 min (A), or cotreated with 1×108 CFU/mL heat-killed E. coli and 3 ng/mL progesterone for 120 min (B). LPS, lipopolysaccharide. P4, progesterone.

https://doi.org/10.1371/journal.pone.0266144.g004

MAPK phosphorylation

The cells treated with only progesterone (1, 3, and 5 ng/mL) showed no change (P > 0.05) in key protein levels of MAPK pathway (Fig 5A). Treatment of BESC with 1 μg/mL LPS induced phosphorylations of ERK, JNK, and P38 at 60 min (data unpublished, S1 Appendix). As shown in Fig 5B, compared with LPS group, the ratios of P-ERK/ERK, P-JNK/JNK, and P-P38/P38 were generally lower in cells of cotreatment groups, except the P-JNK/JNK ratio in cells cotreated with LPS and progesterone (1 or 3 ng/mL). Heat-killed E. coli caused elevation (P < 0.05) in the phosphorylations of ERK, JNK, and P38, with peak levels at 120 min (Fig 5C). Similarly, progesterone (1, 3, and 5 ng/mL) treatment reduced (P < 0.05) the phosphorylations of ERK, JNK, and P38 in response to bacterial challenge (Fig 5D).

thumbnail
Fig 5. Progesterone suppressed phosphorylations of ERK, JNK, and P38 in primary bovine endometrial stromal cells in response to lipopolysaccharide or heat-killed E. coli stimulation.

The protein levels of ERK, P-ERK, JNK, P-JNK, P38, and P-P38 was determined using Western blot. The blots were quantified by Quantity One software. A. Unstimulated cells were treated with 1, 3, and 5 ng/mL progesterone for 30 min. B. The cells were cotreated with 1 μg/mL lipopolysaccharide and progesterone (1, 3, or 5 ng/mL) for 60 min. The time point was based on the results of our previous study. C. Time-course changes in key protein phosphorylation of MAPK pathway in cells challenged with 1×108 CFU/mL heat-killed E. coli for 30, 60, 90, and 120 min. D. The cells were cotreated with heat-killed E. coli and progesterone for 120 min. LPS, lipopolysaccharide. P4, progesterone. All data were presented as means ± SEM (n ≥ 3). * P < 0.05, difference compared with the control; # P < 0.05, difference compared with the LPS group (B) or the heat-killed E. coli group (D).

https://doi.org/10.1371/journal.pone.0266144.g005

Discussion

This study evaluated the impact of progesterone on inflammatory response of primary bovine endometrial stromal cells. As expected, treatment of BESC with LPS or heat-killed E. coli resulted in upregulated mRNA expressions of IL1B, IL6, TNF, CXCL8, NOS2, and PTGS2, as well as the phosphorylation of key proteins in MAPK and NF-κB pathways. The addition of progesterone decreased these proinflammatory gene expressions and inhibited activations of MAPK and NF-κB signaling in BESC with inflammatory response. However, in resting state, progesterone did not influence these proinflammatory genes or related pathways except IL1B mRNA expression.

Increased expressions of proinflammatory genes in response to LPS or E. coli in BESC has been reported extensively [2, 2125]. In agreement with these reports, we observed upregulated mRNA encoding IL1B, IL6, TNF, CXCL8, NOS2, and PTGS2. Proinflammatory cytokines such as IL1β, IL6, and TNF participate in the recruitment of white blood cells to inflamed tissue, the production of acute-phase proteins, the cell death of inflamed tissue, and the modification of vascular endothelial permeability [26]. IL8, encoded by CXCL8 gene, is a powerful chemo-attractant that recruits neutrophils and lymphocytes [27]. Nitric oxide and prostaglandins are inflammatory mediators processed through inducible nitric oxide synthase and cyclooxygenase, respectively. Cyclooxygenase-2 (COX-2), encoded by PTGS2 gene, is the primary enzyme involved in the biosynthesis of most prostaglandins in bovine endometrium [28].

Previously we observed inhibited mRNA expressions of IL1B, IL6, TNF, and CXCL8 by progesterone in primary bovine endometrial epithelial cells with inflammatory response [17], which was similar to the result of current study that progesterone decreased IL1B, IL6, TNF, CXCL8, NOS2, and PTGS2 mRNA expressions induced by LPS or heat-killed E. coli in BESC. Bovine endometrial epithelial and stromal cells preincubated with progesterone secreted less prostaglandin after LPS treatment [3]. Progesterone (10−9 to 10−7 M) was found to suppress IL6 and CXCL8 at both mRNA and protein levels in human endometrial epithelial cell line exposed to LPS and high-mobility group box 1 [29]. In the study of human endometriosis, progesterone (10−6 mol/L) was found to be effective in attenuating TNFα/estrogen-induced expression of CXCL8 in endometriotic stromal cells [30]. The suppressed expression or production of inflammatory mediators has also been reported in murine macrophages [31, 32], mature rat dendritic cells [33], human intestinal epithelial cells [34], and human umbilical vein endothelial cells [14] by progesterone or through progesterone receptor (PGR). Contrary to the findings mentioned above, Saut et al. observed that pretreatment of BESC with progesterone (1 to 30 ng/mL) did not modulate IL6 or CXCL8 responses to 100 ng/mL LPS [4]. Such discrepancy is probably due to the differences in LPS concentration, treatment time and detection time. It is noteworthy that progesterone of 5 ng/mL seemed to be not as effective as progesterone of 1 and 3 ng/mL in inhibiting expressions of some proinflammatory genes, such as IL1B, TNF, and NOS2 at 24 h in LPS- or E. coli-stimulated BESC. Such result was not observed in epithelial cells with similar treatment [17], probably indicating the specific mechanism of BESC. Progesterone concentration has been found to influence mRNA expressions of both PGRA and PGRB isoforms and thus regulates their effects within bovine endometrial cells [35]. PGRB is a progesterone-dependent gene activator, whereas PGRA is a weak gene activator and a potent PGRB inhibitor, thereby reducing the effects of progesterone [35]. Therefore, the difference between progesterone of 5 ng/mL and progesterone of 1 and 3 ng/mL in regulating IL1B, TNF, and NOS2 mRNA expressions in BESC may be related to the changes in the relative levels of PGR isoforms, and requires further study.

Inconsistent results have been shown in literature about the effect of progesterone alone on the production or gene expression of inflammatory mediators in vitro. The secretions of IL6, IL8, or prostaglandins were unaffected in bovine endometrial epithelial and stromal cells pretreated with 5 ng/mL progesterone [3, 4]. Mild upregulations of IL1B, IL6, TNF, and CXCL8 mRNA by progesterone (5 ng/mL) in bovine endometrial epithelial cells have been reported by our lab [17]. Increased production of TNFα has been observed in human endometrial epithelial cells treated with 1 μmol/L progesterone [36]. Unchanged productions of IL1β and TNFα were found by Butts et al. in rat dendritic cells treated with progesterone (10−10 and 10−8 mol/L) [33]. A negative regulation of progesterone (10−7 mol/L) on mRNA expressions of IL6 and CXCL8 was demonstrated by Goddard et al. [14]. We observed that progesterone (1, 3, and 5 ng/mL) alone showed no influence on basal IL6, TNF, CXCL8, NOS2, and PTGS2 mRNA transcriptions, except a down-regulation of IL1B mRNA transcript. Signaling cascades of MAPK and NF-κB probably don’t mediate progesterone’s inhibition of basal IL1B transcription because these pathways were unaffected in unstimulated BESC. Regulation of IL1B transcription by progesterone or through PGR was rarely reported in unstimulated cells. Methylation of specific CpG sites in the proximal promoters of IL1B gene correlates with its expression in human chondrocytes [37]. Myeloid cells transcribe IL1B gene constitutively at a low level with an absolute requirement of Spi1 transcription factor [38]. Further investigation could focus on molecular mechanisms at transcriptional level, such as transcription factors or microRNAs that constitutively regulate IL1B gene.

Activation of TLR4 by LPS results in inductions of NF-κB and MAPK signaling cascades, where enhanced cytokine expression is a major response [39]. In unstimulated conditions, NF-κB is sequestered in the cytoplasm by the inhibitor of NF-κB (IκB) family. Upon stimulation, the IκB kinase complex is activated and phosphorylates IκB molecules, targeting them for ubiquitination and subsequent degradation. Degradation of IκB releases NF-κB, which translocates into the nucleus to initiate a transcriptional response [40]. Our result coincided with previous reports that LPS activated NF-κB pathway in BESC [2, 24]. Progesterone has been shown to prevent NF-κB activation and so inhibit proinflammatory mediator production in reproductive tissues and immune cells [13]. Suppression of NF-κB by progesterone has been reported in bovine and human endometrial epithelial cells [17, 29], murine macrophages [31], mouse microglia [15], and human CD4+ T cells [41]. In line with previous studies, we found that progesterone suppressed NF-κB activation in BESC stimulated with LPS or heat-killed E. coli. There was no change in key protein levels of NF-κB pathway in BESC treated with progesterone alone, which was similar to the observations in bovine endometrial epithelial cells [17], murine macrophages [31], and mouse microglia [15].

The well-known MAPK pathway, mainly composed of ERK1/2, JNK, and P38, transduces environmental and developmental signals into adaptive and programmed responses such as survival, proliferation, differentiation, inflammation, and apoptosis [42]. Activation of MAPK has been shown to initiate inflammatory gene expression in LPS-induced bovine endometrial stromal cells [2, 24, 43]. Consistently, we found LPS-induced phosphorylations of ERK1/2, JNK, and P38 in BESC. The involvement of MAPK in anti-inflammatory action of progesterone in vitro has been reported. Activation of ERK1/2, JNK, and p38 induced by LPS or E. coli was abrogated by the addition of progesterone in primary bovine endometrial epithelial cells [17]. Pretreatment of human endometrial stromal cells with 100 ng/mL progesterone for 9 days suppressed activation of p38 MAPK induced by IL1β [44]. Progesterone (10−8 to 10−6 M) attenuated LPS-mediated p38, JNK, and ERK phosphorylations in mouse microglia, and mifepristone reversed progesterone’s effect [15]. Similar to these observations, we found inhibited activations of ERK1/2, JNK, and p38 by progesterone in BESC in response to LPS or heat-killed E. coli. In the study of primary human myometrial cells with inflammatory response, Lei et al. reported that progesterone induced MAPK phosphatase-1, which can reverse phosphorylation events of MAPK, and this anti-inflammatory effect of progesterone was mediated by glucocorticoid receptor rather than progesterone receptor [45], suggesting a more complex mechanism of progesterone antagonism on inflammation. Similar to our previous observation in bovine endometrial epithelial cells [17], progesterone alone did not affect MAPK pathway in BESC, indicating that the molecular mechanism of progesterone is different between the cells in resting state and the cells with inflammatory response. Both MAPK and NF-κB can be directly activated by LPS stimulation [46]. These pathways have been found to synergize through synchronized binding to κB and AP-1 sites found together in the promoters of many genes, including CXCL8 [2]. Furthermore, the production of proinflammatory cytokines, such as IL1β and TNFα, in turn activate MAPK and NF-κB, forming a positive feedback [46]. Due to the complex interactions of these pathways, the exact intracellular signaling mechanism of anti-inflammation by progesterone in bovine endometrial cells deserves further investigation by using pathway inhibitors and gene silencing and overexpression techniques.

Conclusions

Progesterone elicited anti-inflammatory effect in LPS- or heat-killed E. coli-stimulated primary bovine endometrial stromal cells by downregulating gene expressions of IL1B, IL6, TNF, CXCL8, NOS2, and PTGS2 corresponding to inhibitions of NF-κB and MAPK activation. Except the downregulation of basal IL1B transcription, progesterone alone showed no influence on these proinflammatory gene expressions and NF-κB and MAPK signaling cascades in unstimulated BESC.

References

  1. 1. Sheldon IM, Dobson H. Postpartum uterine health in cattle. Anim Reprod Sci. 2004;82–3:295–306. pmid:15271461
  2. 2. 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. pmid:22053092
  3. 3. 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;147:562–570. pmid:16223858
  4. 4. Saut JPE, Healey GD, Borges AM, Sheldon IM. Ovarian steroids do not affect bovine endometrial cytokine or chemokine responses to Escherichia coli or LPS in vitro. Reproduction. 2014;148:593–606. pmid:25246618
  5. 5. Lewis GS. Steroidal regulation of uterine resistance to bacterial infection in livestock. Reprod Biol Endocrinol. 2003;1:117. pmid:14641941
  6. 6. Webb R, Lamming GE, Haynes NB, Foxcroft GR. Plasma progesterone and gonadotrophin concentrations and ovarian activity in post-partum dairy cows. J Reprod Fertil. 1980;59:133–143. pmid:6772775
  7. 7. Stevenson JS, Britt JH. Relationships among luteinizing hormone, estradiol, progesterone, glucocorticoids, milk yield, body weight and postpartum ovarian activity in Holstein cows. J Anim Sci. 1979;48:570–577. pmid:528417
  8. 8. Henricks DM, Dickey JF, Hill JR, Johnston WE. Plasma estrogen and progesterone levels after mating, and during late pregnancy and postpartum in cows. Endocrinology. 1972;90:1336–1342. pmid:5012745
  9. 9. Opsomer G, Grohn YT, Hertl J, Coryn M, Deluyker H, de Kruif A. Risk factors for post partum ovarian dysfunction in high producing dairy cows in Belgium: a field study. Theriogenology. 2000;53:841–857. pmid:10730974
  10. 10. Lewis GS. Steroidal regulation of uterine immune defenses. Anim Reprod Sci. 2004;82–83:281–294. pmid:15271460
  11. 11. Hansen PJ. Regulation of uterine immune function by progesterone—lessons from the sheep. J Reprod Immunol. 1998;40:63–79. pmid:9862257
  12. 12. Dhaliwal GS, Murray RD, Woldehiwet Z. Some aspects of immunology of the bovine uterus related to treatments for endometritis. Anim Reprod Sci. 2001;67:135–152. pmid:11530260
  13. 13. Shah NM, Lai PF, Imami N, Johnson MR. Progesterone-related immune modulation of pregnancy and labor. Front Endocrinol. 2019;10:198. pmid:30984115
  14. 14. Goddard LM, Ton AN, Org T, Mikkola HK, Iruela-Arispe ML. Selective suppression of endothelial cytokine production by progesterone receptor. Vascul Pharmacol. 2013;59:36–43. pmid:23747964
  15. 15. Lei BL, Mace B, Dawson HN, Warner DS, Laskowitz DT, James ML. Anti-inflammatory effects of progesterone in lipopolysaccharide-stimulated BV-2 microglia. PLOS ONE. 2014;9:e103969. pmid:25080336
  16. 16. Okabe H, Makino S, Kato K, Matsuoka K, Seki H, Takeda S. The effect of progesterone on genes involved in preterm labor. J Reprod Immunol. 2014;104–105:80–91. pmid:24933116
  17. 17. Cui LY, Wang H, Lin JQ, Wang YL, Dong JS, Li J, et al. Progesterone inhibits inflammatory response in E.coli- or LPS-stimulated bovine endometrial epithelial cells by NF-κB and MAPK pathways. Dev Comp Immunol. 2020;105:103568. pmid:31821816
  18. 18. Piersanti RL, Zimpel R, Molinari PCC, Dickson MJ, Ma ZX, Jeong KC, et al. A model of clinical endometritis in Holstein heifers using pathogenic Escherichia coli and Trueperella pyogenes. J Dairy Sci. 2019;102:2686–2697. pmid:30692014
  19. 19. Cui LY, Wang YL, Wang H, Dong JS, Li ZX, Li J, et al. Different effects of cortisol on pro-inflammatory gene expressions in LPS-, heat-killed E.coli-, or live E.coli-stimulated bovine endometrial epithelial cells. BMC Vet Res. 2020;16:9. pmid:31918707
  20. 20. Cui LY, Qu Y, Cai HL, Wang H, Dong JS, Li J, et al. Meloxicam inhibited the proliferation of LPS-stimulated bovine endometrial epithelial cells through Wnt/β-catenin and PI3K/AKT pathways. Front Vet Sci. 2021;8:637707. pmid:34307514
  21. 21. Herath S, Lilly ST, Fischer DP, Williams EJ, Dobson H, Bryant CE, et al. Bacterial lipopolysaccharide induces an endocrine switch from prostaglandin F2α to prostaglandin E2 in bovine endometrium. Endocrinology. 2009;150:1912–1920. pmid:19056817
  22. 22. Herath S, Dobson H, Bryant CE, Sheldon IM. Use of the cow as a large animal model of uterine infection and immunity. J Reprod Immunol. 2006;69:13–22. pmid:16386311
  23. 23. Koh YQ, Mitchell MD, Almughlliq FB, Vaswani K, Peiris HN. Regulation of inflammatory mediator expression in bovine endometrial cells: effects of lipopolysaccharide, interleukin 1 beta, and tumor necrosis factor alpha. Physiol Rep. 2018;6:e13676. pmid:29707922
  24. 24. Cui LY, Cai HL, Sun FZ, Wang YL, Qu Y, Dong JS, et al. Beta-endorphin inhibits the inflammatory response of bovine endometrial cells through delta opioid receptor in vitro. Dev Comp Immunol. 2021;121:104074. pmid:33775662
  25. 25. 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. pmid:33324700
  26. 26. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010;140:805–820. pmid:20303872
  27. 27. Foley C, Chapwanya A, Creevey CJ, Narciandi F, Morris D, Kenny EM, et al. Global endometrial transcriptomic profiling: transient immune activation precedes tissue proliferation and repair in healthy beef cows. BMC Genom. 2012;13:489. pmid:22985206
  28. 28. Arosh JA, Parent J, Chapdelaine P, Sirois J, Fortier MA. Expression of cyclooxygenases 1 and 2 and prostaglandin E synthase in bovine endometrial tissue during the estrous cycle. Biol Reprod. 2002;67:161–169. pmid:12080013
  29. 29. Mita S, Shimizu Y, Notsu T, Imada K, Kyo S. Dienogest inhibits Toll-like receptor 4 expression induced by costimulation of lipopolysaccharide and high-mobility group box 1 in endometrial epithelial cells. Fertil Steril. 2011;96:1485–1489. pmid:22014880
  30. 30. Horie S, Harada T, Mitsunari M, Taniguchi F, Iwabe T, Terakawa N. Progesterone and progestational compounds attenuate tumor necrosis factor alpha-induced interleukin-8 production via nuclear factor kappa B inactivation in endometriotic stromal cells. Fertil Steril. 2005;83:1530–1535. pmid:15866594
  31. 31. Su L, Sun YX, Ma F, Lu PP, Huang HF, Zhou J. Progesterone inhibits Toll-like receptor 4-mediated innate immune response in macrophages by suppressing NF-kappa B activation and enhancing SOCS1 expression. Immunol Lett. 2009;125:151–155. pmid:19607861
  32. 32. Menzies FM, Henriquez FL, Alexander J, Roberts CW. Selective inhibition and augmentation of alternative macrophage activation by progesterone. Immunology. 2011;134:281–291. pmid:21977998
  33. 33. Butts CL, Shukair SA, Duncan KM, Bowers E, Horn C, Belyavskaya E, et al. Progesterone inhibits mature rat dendritic cells in a receptor-mediated fashion. Int Immunol. 2007;19:287–296. pmid:17289656
  34. 34. Salzman AL, Linn SC, Szabo C. Progesterone inhibits inducible nitric oxide synthase mRNA expression in human intestinal epithelial cells. Int J Mol Med. 2000;6:209–216. pmid:10891568
  35. 35. Rekawiecki R, Kowalik MK, Kotwica J. Onapristone (ZK299) and mifepristone (RU486) regulate the messenger RNA and protein expression levels of the progesterone receptor isoforms A and B in the bovine endometrium. Theriogenology. 2015;84:348–357. pmid:25976976
  36. 36. Laird SM, Tuckerman EM, Saravelos H, Li TC. The production of tumour necrosis factor alpha (TNF-alpha) by human endometrial cells in culture. Hum Reprod. 1996;11:1318–1323. pmid:8671447
  37. 37. Hashimoto K, Otero M, Imagawa K, de Andres MC, Coico JM, Roach HI, et al. Regulated transcription of human matrix metalloproteinase 13 (MMP13) and interleukin-1beta (IL1B) genes in chondrocytes depends on methylation of specific proximal promoter CpG sites. J Biol Chem. 2013; 288:10061–10072. pmid:23417678
  38. 38. Pulugulla SH, Packard TA, Galloway NLK, Grimmett ZW, Doitsh G, Adamik J, et al. Distinct mechanisms regulate IL1B gene transcription in lymphoid CD4 T cells and monocytes. Cytokine. 2018; 111:378–381. pmid:30300855
  39. 39. Sordillo LM. Nutritional strategies to optimize dairy cattle immunity. J Dairy Sci. 2016;99:4967–4982. pmid:26830740
  40. 40. Bhatt D, Ghosh S. Regulation of the NF-kappaB-mediated transcription of inflammatory genes. Front Immunol. 2014;5:71. pmid:24611065
  41. 41. Hellberg S, Raffetseder J, Rundquist O, Magnusson R, Papapavlou G, Jenmalm MC, et al. Progesterone dampens immune responses in in vitro activated CD4(+) T cells and affects genes associated with autoimmune diseases that improve during pregnancy. Front Immunol. 2021;12:672168. pmid:34054852
  42. 42. Zlobin A, Bloodworth JC, Osipo C. Mitogen-activated protein kinase (MAPK) signaling. In: Badve S, Kumar GL, editors. Predictive biomarkers in oncology. Switzerland AG: Springer Nature; 2019. pp. 213–221.
  43. 43. Yan CB, Lv HM, Peng Z, Yang DX, Shen PX, Yu JC, et al. Analysis of miRNA expression changes in bovine endometrial stromal cells treated with lipopolysaccharide. Theriogenology. 2021;167:85–93. pmid:33784501
  44. 44. Yoshino O, Osuga Y, Hirota Y, Koga K, Hirata T, Yano T, et al. Endometrial stromal cells undergoing decidualization down-regulate their properties to produce proinflammatory cytokines in response to interleukin-1 beta via reduced p38 mitogen-activated protein kinase phosphorylation. J Clin Endocrinol Metab. 2003;88:2236–2241. pmid:12727980
  45. 45. Lei K, Georgiou EX, Chen L, Yulia A, Sooranna SR, Brosens JJ, et al. Progesterone and the repression of myometrial inflammation: the roles of MKP-1 and the AP-1 system. Mol Endocrinol. 2015;29:1454–1467. pmid:26280733
  46. 46. Chen LC, Stephanie AC, Bilal U, Vinay T. Noncoding RNAs: master regulators of inflammatory signaling. Trends Mol Med. 2018;24:66–84. pmid:29246760