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

Regulation of inflammatory genes in decidual cells: Involvement of the bromodomain and extra-terminal family proteins

  • Sandeep Ajgaonkar,

    Roles Investigation, Methodology, Visualization, Writing – original draft

    Affiliation College of Health, Medicine, and Wellbeing, University of Newcastle, Callaghan, NSW, Australia

  • Jonathan J. Hirst,

    Roles Supervision, Writing – review & editing

    Affiliations College of Health, Medicine, and Wellbeing, University of Newcastle, Callaghan, NSW, Australia, Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia

  • Mary Norris,

    Roles Supervision, Validation

    Affiliations College of Health, Medicine, and Wellbeing, University of Newcastle, Callaghan, NSW, Australia, Department of Maternity and Gynaecology, John Hunter Hospital, New Lambton Heights, NSW, Australia

  • Tamas Zakar

    Roles Conceptualization, Formal analysis, Funding acquisition, Project administration, Supervision, Writing – review & editing

    Tamas.Zakar@newcastle.edu.au

    Affiliations College of Health, Medicine, and Wellbeing, University of Newcastle, Callaghan, NSW, Australia, Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia, Department of Maternity and Gynaecology, John Hunter Hospital, New Lambton Heights, NSW, Australia

Abstract

The decidua undergoes proinflammatory activation in late pregnancy, promoting labor. Bromodomain and Extra-Terminal (BET) family proteins interact with acetylated histones and may control gene expression in inflammation. Here, we assessed whether BETs are involved in inflammatory gene regulation in human decidual cells. We have treated primary cultures of decidual stromal cells (DSCs) from term pregnancies with endotoxin (LPS) and measured the expression of a panel of pro-and anti-inflammatory genes. BET involvement was assessed using the selective BET inhibitors (+)-JQ1 and I-BET-762 or the negative control compound (-)-JQ1. Histone 3 and -4 acetylation and BETs binding at the target gene promoters were determined to assess whether these processes are involved in the actions of LPS, BETs, and BET inhibitors. LPS increased the expression of the proinflammatory (PTGS2, IL6, CXCL8/IL8, TNF) and the anti-inflammatory (IL10, IDO1) genes of the panel. The constitutively expressed inflammatory genes (PTGS1, PTGES) were unaffected. The BET inhibitors, but not the control compound, reduced the basal and LPS-induced expression of PTGS1, PTGS2, IL6, CXCL8/IL8, IL10, and IDO1. TNF expression was not changed by BET inhibition. The dominant BETs were Bromodomain-containing protein -2 (BRD2) and -4L (BRD4L) in DSCs. LPS increased histone 4 acetylation at the CXCL8/IL8 and TNF promoters and histone 3 and -4 acetylation at the IDO1 promoter, while (+)-JQ1 abrogated histone acetylation at several promoters. Overall, histone acetylation and promoter binding of BETs showed no consistent relationship with gene expression across the gene panel and the treatments. BET proteins, predominantly BRD2 and BRD4L, control critical pro- and anti-inflammatory genes in DSCs. TNF induction exemplifies a BET-independent pathway. Changing histone acetylation at the promoters is not a general obligatory requirement for inflammatory gene expression in response to LPS. BETs likely act at chromatin loci separate from the examined promoters. BET inhibitors may block decidual activation at labor.

Introduction

The decidua is the endometrium of pregnancy, forming the maternal side of the maternal-fetal interface. It plays a crucial role in protecting the semi-allogenic fetus from immune rejection while preserving decidual competence to elicit an immune response against infections ascending from the vagina or originating from peripheral sources. These contrasting roles are performed by tightly controlled pro- and anti-inflammatory, immunosuppressive and immune-promoting responses [1, 2]. Disturbances of this balance are associated with pregnancy disorders such as miscarriage, pre-eclampsia, early membrane rupture, and premature labor.

The decidua is a complex tissue with diverse and dynamic cellular composition. It contains a variety of bone marrow-derived CD45+ leukocytes that include monocytes/macrophages, dendritic cells, granulocytes, NK (natural killer) cells, T and B lymphocytes, and mast cells that change in abundance during gestation [3]. The largest proportion of cells is a resident decidual cell type establishing the stroma of the tissue. Decidual stromal cells (DSCs) exhibit pro- and anti-inflammatory capacities and influence leukocytes locally to adapt to the idiosyncratic needs of pregnancy [1]. DSCs promote Th2 immune bias [4], inhibit natural killer (NK) cell cytotoxicity, and reduce the activity of antigen-presenting (dendritic) cells [5] at the maternal-fetal interface favoring pregnancy maintenance. The pregnancy-protective phenotype of the decidua vanes towards the end of gestation [6, 7], and decidual cells shift to a proinflammatory phenotype [8], which promotes myometrial activation and cervical remodeling. This change is part of the process leading to labor and birth. Pathological activation of the decidua leads to dysfunctional labor, untimely delivery, and newborn complications. The mechanisms that underlie the inflammatory transformation of the decidua in normal or pathological labor are poorly understood. In culture, DSCs respond to LPS (lipopolysaccharide, an inflammatory trigger from Gram-negative bacteria) or cytokines like IL-1 beta and TNF-a by producing a variety of proinflammatory factors, including IL-6, IL-8, PTGS2, PTGES [913]. Anti-inflammatory cytokine production (IL-10) has also been reported after LPS treatment [14], indicating a balanced pro-and anti-inflammatory response.

Inflammatory transformation of cells involves widespread gene expression changes associated with alterations of chromatin structure at promoters, enhancers, and super-enhancers controlling inflammatory genes [15, 16]. The altered acetylation state of histones and non-histone proteins has been recognized as a critical component of the process [17]. One initial event is the binding of BRD4, (bromodomain-containing protein 4) a member of the BET (bromodomain and extra-terminal) family of proteins, to gene regulatory regions in a complex with NFkB (nuclear factor NF-kappa-B), the central regulator of inflammation [18]. BET recruitment is critical for activating super-enhancers and promoters controlling proinflammatory genes [19, 20], while BETs dissociating from other super-enhancers deactivates non-inflammatory genes during the phenotype transition [21]. BET family proteins bind acetyl-lysine residues in acetylated histone and non-histone proteins through their tandem bromodomains and protein interaction domains [22], which is the mechanistic basis of their chromatin interactions. Importantly, BET proteins can be targeted effectively by drugs that disrupt the bromodomain-acetyl lysine binding [23, 24], which abrogates the super-enhancer mediated cell phenotype transitions [19]. BET proteins are emerging central players in inflammation with modifiable activity [25].

The role of BET proteins in inflammatory gene regulation in the decidua is undefined. Here we have addressed this gap of knowledge by determining (1) the effect of BET inhibitors on LPS-induced inflammatory gene expression in primary cultures of DSCs, (2) the expression of BET family members in DSCs, and (3) the effects of LPS and BET inhibitor on histone acetylation and BET protein binding at the promoters of inflammatory genes in the DSCs. The results demonstrate that BETs regulate pro- and anti-inflammatory genes in the decidua, and BET inhibitors abrogate the expression of the LPS-induced pro- and anti-inflammatory genes. The results also suggest that BRD2 and BRD4, the dominant BET proteins in DSCs, interact predominantly with genomic regulatory elements distant from the promoters of the inflammatory genes studied. Nonetheless, BET inhibitors can potentially block decidual inflammation with therapeutic benefits.

Materials and methods

Ethics approval

The study has been approved by the Hunter New England Human Research Ethics Committee (Project ID: 2019/ETH12332 and 2019/ETH12330). All participants donated placental tissue following informed written consent.

Isolation, purification, and culture of decidual stromal cells

Tissue processing.

Decidua tissue was isolated from placentae delivered by elective Cesarean section (in the absence of labor) at term after uncomplicated singleton pregnancies. None of the donors were diagnosed with intrauterine infection, inflammation, or histological chorioamnionitis. The reflected membranes were cut off at approximately 2 cm from the placental disk. The amnion was peeled away, and decidua tissue was scraped off the maternal side with a glass microscope slide. The scrapings (12-15g from one placenta) were washed in DPBS (Dulbecco’s phosphate-buffered saline containing 40 μg/mL Gentamicin sulfate) and suspended in 25 mL digestion buffer (DMEM/F12 containing 20mM HEPES, pH7.4; 60 μg/mL Gentamicin sulfate; and the following enzymes: collagenase, Worthington LK003240, 1500 units; Dispase II, Sigma D4693, 62.5 mg, and DNAse I, Sigma DN25, 1.25 mg. Digestion was performed at 37°C in a shaker water bath until the tissue was disintegrated entirely, which took 2–3 hours. The digest was centrifuged at 800 g for 10 min at room temperature, and the sediment was suspended in 25 mL of DMEM/F12 supplemented with 20 mM HEPES, pH7.4, 20% (v/v) heat-inactivated FBS, and 40 μg/mL Gentamicin sulfate. After centrifugation (600 g, 10 min, room temperature), the pellet was suspended in 10 mL of serum-free DMEM/F12 with 20 mM HEPES, pH 7.4, and 40 μg/mL Gentamicin sulfate, passed through a 120 μm pore size strainer and processed for Percoll density purification.

Percoll purification.

“100% Percoll” was prepared by combining nine parts (vol) of the commercially supplied Percoll suspension (Sigma P1644) with one part of 10 x Hank’s salt solution (Ca++ and Mg++-free) 200 mM HEPES, pH 7.4 and 40 μg/mL Gentamicin sulfate. Discontinuous density gradients of 50% overlaid with 30% Percoll were prepared, and the cell suspension, adjusted to 25% Percoll, was layered on the top. The gradients were centrifuged at 900 g for 40 min at room temperature. Cells were recovered from the 50%/30% boundary and washed to remove residual Percoll.

Immuno-magnetic enrichment of DSCs.

Enrichment was achieved using the EasySep Human CD45+ cell depletion kit (II) from StemCell Technologies (Tullamarine, VIC Australia), following the manufacturer’s instructions. Briefly, cells were suspended at 108 cells/mL concentration in Hank’s salt solution supplemented with 20 mM HEPES, pH 7.4, 2% (v/v) FBS, and 40 μg/mL Gentamicin sulfate. A Depletion Cocktail containing anti-CD45 antibodies was added, and CD45+ cells (mostly leukocytes [26]) were absorbed by adding magnetic RapidSpheres™ to the suspension. RapidSpheres™ with absorbed cells were removed using a strong magnet, and the enriched DSCs were suspended in cell culture medium (DMEM/F12, 20% FBS, 10 nM estradiol, 100 nM medroxyprogesterone acetate, 40 μg/mL Gentamicin sulfate). Cell yields were typically between 4.5–5.5x107 with 80 to 85% viability (Trypan blue exclusion). The absence of CD45+ cells was verified by flow cytometry in preliminary experiments. A representative example of the flow cytometry analysis of freshly isolated decidual cells is presented in S1 Fig, showing that >99% of live single cells were CD45 negative after purification.

Cell culture.

Cells were seeded at 105/cm2 in cell culture flasks with gelatin (Type-2, Sigma G1393)—coated culture surface. Cultures were incubated in a humidified atmosphere of 5% CO2 in air at 37°C. The medium was changed every third day until 10–14 days of culture. Immunocytochemical characterization of the sub-confluent cultures (S2 Fig) showed pervasive expression of vimentin, a marker of stromal cells in the decidua [26], and the absence of CD45+ cell contamination. The cultured DSCs were treated with (+)-JQ1, I-BET-762, or (-)-JQ1 (0.5 μM) for 48 h, followed by stimulation with lipopolysaccharide (LPS, from E. coli 055:B5, 1 μg/mL, for 24 h in the presence of the drugs).

Messenger RNA determination

Total RNA was isolated from cells cultured in T25 flasks using the RNeasy Mini Kit and the RNase-Free DNase Set (Qiagen, Chadstone Centre, VIC Australia) following the manufacturer’s protocols. RNA purity was assessed by UV absorption using a NanoDrop 1000 spectrophotometer, and the Agilent 2100 Bioanalyzer System was used to verify RNA integrity. Purified RNA was spiked with Alien RNA transcript (107 copies/μg, Stratagene, Integrated Sciences, Chatswood NSW Australia), which served as the reference RNA in the qRT-PCR assays.

The RNA was reverse transcribed using the Superscript™ III (Life Technologies, Mulgrave, VIC Australia) with random hexamer primers. Quantitative real-time PCR was performed using the Applied Biosystems™ QuantStudio™ 6-Flex Real-Time PCR system (Thermo Fisher, Scoresby VIC Australia) with SYBR™ Green detection reagents sourced from the manufacturer. Primers have been designed using Primer-BLAST from NCBI, and their sequences are listed in the S1 Table. Primer and template cDNA concentrations were optimized for each mRNA assay [27].

Chromatin immunoprecipitation (ChIP)

Cell fixation.

Cells grown in T75 culture flasks were fixed by a two-step cross-linking procedure that enhances the detection of modified histones and transcription factors bound to the chromatin [28]. Briefly, the adherent cultures were rinsed with PBS (phosphate-buffered saline) and cross-linked by adding 2 mM EGS (Ethylene glycol-bis(succinic acid N-hydroxy-succinimide ester) (Sigma-Aldrich E3257) in PBS for 30 min at room temperature. Next, fresh buffer with 1% formaldehyde was added at room temperature for 10 min to perform the second cross-linking step. Formaldehyde action was quenched with 125 mM glycine in cold PBS. Cells were permeabilized in hypotonic buffer (50 mM Tris-HCl, pH 7.5, 10 mM NaCl, 3 mM MgCl2, 0.15% v/v Igepal Ca-630) supplemented with protease inhibitors (cOmplete™ Mini Protease Inhibitor Cocktail, Roche, and 0.5 mM phenylmethanesulfonyl fluoride, PMS) and histone deacetylase inhibitor (10 mM Na-butyrate). Cells were scraped off the culture surface, and the nuclei were sedimented by centrifugation and stored at -80°C until further processing.

Preparation of chromatin extracts.

Nuclear pellets were suspended in Extraction Buffer (50 mM Tris HCl, pH8.1; 10 mM EDTA; 1% SDS and inhibitors as above) and sonicated using a Misonix XL-2000 microprobe sonifier with the following optimized settings: Ten 10-sec bursts at 8–11 watts and one pulse/sec, separated by 50-sec intervals. Heating was prevented by performing the sonication in a salt-ice bath. Sonicated samples were centrifuged at 13,000 rpm at 4°C for 30 min, and the supernatants were stored frozen until immunoprecipitation.

Immunoprecipitation.

Chromatin extracts were diluted 10-fold with Modified RIPA buffer (1.13% Triton X-100, 0.11% Na-deoxycholate, 1.1 mM EDTA, 0.56 mM EGTA, 158 mM NaCl, 10mM Tris HCl pH 8.1 supplemented with 0.55 mM PMSF, 0.11% BSA, 4.4 μg/mL sonicated salmon sperm DNA, 11.1 mM Na-butyrate, cOmplete protease inhibitors). Immunoprecipitations were performed in 500 μL volume at 4°C overnight using antibodies (3μg/immunoprecipitation) listed in S2 Table. Immuno-complexes were captured by Protein-A/G Magnetic Beads (Pierce™, ChIP-grade, Cat. No. 26162) pre-washed and pre-equilibrated with Modified RIPA diluted with Extraction Buffer (9:1). Beads with attached immunocomplexes were collected in a magnetic stand and washed sequentially with the following solutions: Once with IPW (0.1% SDS, 1% Triton X100, 2 mM EDTA, 20 mM Tris HCl pH 8.1, 150 mM NaCl), once with IPW containing 500 mM NaCl, once with IPWL (0.25M LiCl, 1% NP40, 1% (w/v) Na-deoxycholate, 1mM EDTA, 10mM Tris HCl, pH 8.1) and twice with TE (10mM Tris, pH 8.1, 1 mM EDTA). Beads suspended in TE were transferred to clean Eppendorf LoBind tubes, reducing the background ChIP signal [29].

Chromatin elution, crosslink reversal, and DNA purification.

Chromatin fragments were eluted by incubating the beads in 1% SDS, 0.1 M NaHCO3 at 65°C for 10 min. The bead-free eluates were supplemented with Proteinase K (57 μg/mL) and incubated at 56°C for one hour, followed by 65°C overnight to reverse crosslinks. An aliquot of each sonicated extract was processed for crosslink reversal without immunoprecipitation to determine the input in the ChIP assays.

DNA was purified by phenol-chloroform-isoamyl alcohol extraction (25:24:1 v/v) followed by Na-acetate-ethanol precipitation with 45 μg GlycoBlue™ (Invitrogen) co-precipitant added to each sample. Precipitates were washed 2 x with TE buffer to remove residual SDS and dissolved in TE. DNA content in the input samples was determined by UV absorption using the NanoDrop 1000 spectrophotometer.

Quantitative real-time PCR (qPCR).

The abundance of target gene promoter sequences in the immunoprecipitated DNA was determined by qPCR. Gene-specific oligonucleotide primer sequences and their positions relative to transcription start sites are listed in S1 Table. Oligonucleotide primers were designed and optimized as described above. Triplicate PCR reactions were performed with each chromatin extract and gene from each immunoprecipitation using the QuantStudio™ 6-Flex Real-Time PCR system with SYBR™ Green detection.

Data processing and statistical analysis

Messenger RNA abundance.

Results were calculated relative to the spiked-in exogenous RNA [30] (Alien) by the delta-Ct method [27]. Biological variation was reduced by scaling data over the average of all treatments in each experiment. Data were tested for distribution and transformed to meet the criteria for normal distribution. The data were then analyzed by repeated-measures ANOVA followed by F-tests for individual contrasts with significance adjusted to false discovery rate (FDR, “Q”) using the Benjamini-Hochberg method. In some of the experiments, as indicated in the Figure legends, the non-normally distributed data were evaluated using the non-parametric K-sample-equality-of-medians test followed by Fisher’s exact test, Wilcoxon’s signed-rank test, or Dunn’s test of multiple comparisons using rank sums with Bonferroni-adjustment of significance, as appropriate. Differences at p<0.05 or Q<0.05 were considered statistically significant.

ChIP.

Results were calculated as the recovery of promoter fragments relative to input using delta-Ct values. The results for each gene were scaled over the average of all treatments with each immunoprecipitation (acetyl-histone 3, acetyl-histone 4, BRD2, BRD4, IgG) in each experiment and expressed relative to the negative control, IgG, to reduce biological and technical variation. Data were tested for distribution and transformed to meet normal distribution criteria. The data were then analyzed by repeated-measures ANOVA followed by F-tests for individual contrasts with significance adjusted to false discovery rate (FDR, “Q”) using the Benjamini-Hochberg method. Q<0.05 (5% FDR) was considered statistically significant. Statistical calculations were performed using STATA/IC 15.1 (College Station, TX; RRID: SCR_012763)

Results

BETs are involved in gene expression control in DSCs

Treatments and gene targets.

We have utilized two highly selective BET inhibitors, (+)-JQ1 and I-BET-762 [31, 32], which can serve as chemical probes of functional BET involvement in chromatin regulation and gene expression control [33]. We have also treated cells with (-)-JQ1, the enantiomer of (+)-JQ1, as the inactive negative control compound [31]. Lipopolysaccharide (LPS) was used to model inflammatory stimulation by bacteria. We determined the effects of the treatments on a panel of pro-and anti-inflammatory genes selected for their potential participation in the inflammatory response of DSCs (S3 Fig). Basal levels of these mRNAs varied significantly in vehicle-treated cells (p = 0.001, K-sample-equality-of-medians test), with the constitutively expressed PTGS1 and PTGES exhibiting the highest mRNA abundance. At the same time, the expression of the inducible PTGS2, TNF, and IDO1 genes was low.

The effects of LPS and BET inhibitors on target gene expression.

Fig 1 summarizes the effects of LPS, BET inhibitors, and the combination of LPS and BET inhibitors on target gene expression in a heat map grid format. Significant stimulation and inhibition at 5% FDR are visualized in the orange and blue cells of the grid, respectively, while non-significant contrasts are shown in the uncolored cells. The FDR values were calculated using all contrasts (28 altogether) presented in the heat map in S4 Fig with the uncorrected P-values. Relative mRNA abundances for each gene and responses to treatments are displayed individually in Fig 2 (PTGS1, PTGES, and PTGS2, encoding members of the prostaglandin synthesis pathway), Fig 3 (IL6, CXCL8/IL8, and TNF, encoding key proinflammatory mediators), and Fig 4 (IL10 and IDO1, encoding anti-inflammatory mediators). Significant differences highlighted in the heatmap in Fig 1 are indicated in the figures. LPS significantly increased the mRNA levels (vs. vehicle) of the genes selected for inclusion in the panel, except for PTGS1 and PTGES. The BET inhibitors (+)-JQ1 and I-BET-762, but not the negative control (-)-JQ1, reduced the basal (drugs vs. vehicle) and LPS-stimulated (LPS + drugs vs. LPS) expression of PTGS2, IL6, and IL10. The drugs did not affect the low basal levels of CXCL8/IL8 and IDO1 mRNAs. Still, the LPS-induced expression of these genes was significantly blocked by the BET inhibitors but not the control compound. BET inhibitors did not affect the expression of TNF and PTGES either in the presence or absence of LPS. Finally, the abundance of PTGS1 mRNA was decreased by (+)-JQ1 both in the presence and absence of LPS, while (-)-JQ1 and I-BET-762 had no effect under the treatment conditions employed. The response patterns of PTGS2, IL6, IL10, CXCL8/IL8, and IDO1 to BET inhibitors strongly suggest that BET proteins are involved in the mechanism of LPS stimulation of these genes in the DSCs. The lack of effect of BET inhibitors indicates that TNF and PTGES expression is not BET dependent. Our data suggest some BET involvement in the (constitutive) expression of PTGS1.

thumbnail
Fig 1. Heat map grid summarizing the effects of LPS and BET inhibitors on target gene mRNA expression in DSCs.

Culture and treatment conditions, qRT-PCR assay details, and data processing are described in the Materials and methods section. The grid cells show significance values (Q) adjusted to the false discovery rate (FDR). The significance threshold is 5% FDR (Q<0.05), highlighted in bold. The orange color indicates a significant increase, the blue color indicates a significant decrease regarding the contrasts shown. The gene symbols are in red. The Q values have been calculated by contrasting all treatments with each other (28 contrasts, listed in S4 Fig). The contrasts selected for display in the heatmap indicate LPS effects (LPS vs. Vehicle), drug effects ((+)-JQ1 or (-)-JQ1 or I-BET-762 vs. Vehicle), and drug effects on the LPS response (LPS + (+)-JQ1 or (-)-JQ1 or I-BET-762 vs. LPS).

https://doi.org/10.1371/journal.pone.0280645.g001

thumbnail
Fig 2.

The effect of LPS and BET inhibitors on PTGS1 (Panel A), PTGES (Panel B) and PTGS2 (Panel C) mRNA expression in DSCs. Sub-confluent primary DSC cultures were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, I-BET-762, and their combinations as indicated under the columns. Messenger RNA abundance was determined by qRT-PCR. Further details of the experimental procedures and data analysis are described in the Materials and methods. Columns show the mean ± SEM, n = 4–5 independent experiments. Significant differences (at 5% FDR), highlighted in the heatmap in Fig 1, are indicated by the horizontal orange and blue lines. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.g002

thumbnail
Fig 3.

The effect of LPS and BET inhibitors on IL6 (Panel A), CXCL8/IL8 (Panel B) and TNF (Panel C) mRNA expression in DSCs. Primary DSC cultures were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, I-BET-762, and their combinations as indicated under the columns. Messenger RNA relative abundance was determined by qRT-PCR. Significant differences (at 5% FDR), highlighted in the heatmap in Fig 1, are indicated by the horizontal orange and blue lines. Further details and data presentation are described in Legend to Fig 2.

https://doi.org/10.1371/journal.pone.0280645.g003

thumbnail
Fig 4.

The effect of LPS and BET inhibitors on IL10 (Panel A) and IDO1 (Panel B) mRNA expression in DSCs. Primary DSC cultures were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, I-BET-762, and their combinations as indicated under the columns. Messenger RNA relative abundance was determined by qRT-PCR. Significant differences (at 5% FDR), highlighted in the heatmap in Fig 1, are indicated by the horizontal orange and blue lines. Further details and data presentation are described in Legend to Fig 2.

https://doi.org/10.1371/journal.pone.0280645.g004

BETs are expressed in DSCs

(+)-JQ1 and I-BET-762 are pharmacological probes targeting the tandem bromodomains of the BET family members BRD2, BRD3, BRD4, and BRDT (bromodomain-containing proteins 2, -3, -4, and bromodomain testis-specific protein) [33, 34]. We have determined the expression of these BETs (except for the testis-specific BRDT) in DSCs to delineate the BET inhibitor targets that mediate the effects of the drugs. Data in Fig 5A) shows that BRD2, BRD3, and BRD4 mRNAs are present in DSCs. Both the long (canonical) BRD4 mRNA variant, BRDL, and the truncated BRD4S were detected. Differences in the mRNA levels indicated that BRD2 and BRD4L were the dominant BETs in the DSCs. LPS did not affect BRD2 and BRD4L expression (Fig 5B and 5C). Treatment with (+)-JQ1, but not with (-)-JQ1, significantly increased the level of BRD2 mRNA (Fig 5B), indicating BET-dependent negative feedback. BRD4L mRNA levels were not affected by the drugs (Fig 5C).

thumbnail
Fig 5. Expression of BET family members in DSCs.

Total RNA was extracted from primary DSC cultures and mRNA levels encoding BRD2, BRD3, BRD4S and BRD4L were determined by qRT-PCR. (Panel A) Columns represent the mean ± SEM of n = 6 independent experiments. *, p<0.05 vs. BRD3 and BRD4S, by Dunn’s test (non-parametric) with Bonferroni correction for multiple comparisons. Panels B and C: The effect of (+)-JQ1, (-)-JQ1, and LPS on BRD2 and BRD4L expression, respectively. Cultures were treated with drugs and LPS, as described in the Materials and methods. Messenger RNA levels were determined by qRT-PCR. Columns represent the mean ± SEM of n = 4–9 independent experiments. LPS did not affect BRD mRNA levels (p>0.05, Wilcoxon’s signed-rank test), and drug effects were evaluated after pooling data with LPS- treated and LPS-untreated cells. *, p<0.05 vs. (+)-JQ1 treated cells (Dunn’s test of multiple comparisons using rank sums with Bonferroni adjustment of significance). The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.g005

Histone acetylation and BET presence at target gene promoters

BET proteins bind acetylated histone 3 and -4 (acH3 and acH4) through their bromodomains, which is critical to their actions in chromatin. BRD2 and -4 have been reported to bind and interact with the promoters of genes they control [35, 36]. Based on these observations, we have explored histone acetylation and BRD2 and -4 presence at the promoters of the genes induced by LPS in our gene panel. Chromatin immunoprecipitation (ChIP) was performed using antibodies that recognize acH3, acH4, BRD2, and BRD4, and the enrichment of proximal promoter sequences of the target genes relative to the negative control IgG was determined in the immunoprecipitated chromatin fragments. Enrichment levels were compared among cultures treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations.

Acetyl histone 3.

The heat map grid in Fig 6 summarizes the changes of acH3 levels at the gene promoters in cells treated with LPS, (+)-JQ1 and (-)-JQ1. Significance thresholds were adjusted to 5% FDR (Q) based on all contrasts, shown in S5 Fig, including the unadjusted P values. Figs 7 and 8 display the acH3 ChIP results in each individual gene promoter under each treatment condition and indicate the significant differences highlighted in the heatmap in Fig 6.

thumbnail
Fig 6. Heat map grid summarizing the effects of LPS, (+)-JQ and (-)-JQ1 on histone 3 acetylation at the promoters of IL6, CXCL8/IL8, TNF, IL10 and IDO1.

Culture and treatment conditions, chromatin immunoprecipitation (ChIP) details, and data processing are described in the Materials and methods section. The grid cells show significance values (Q) adjusted to the false discovery rate (FDR). The significance threshold is 5% FDR (Q<0.05), highlighted in bold. The orange color indicates a significant increase, the blue color indicates a significant decrease in the acH3 ChIP signal regarding the contrasts shown. The gene symbols are in red. The Q values have been calculated by contrasting all treatments with each other (21 contrasts, listed in S5 Fig). The contrasts selected for display in the heatmap indicate LPS effects (LPS vs. Vehicle), drug effects ((+)-JQ1 or (-)-JQ1 vs. Vehicle), and drug effects on the LPS response (LPS + (+)-JQ1 or (-)-JQ1 vs. LPS).

https://doi.org/10.1371/journal.pone.0280645.g006

thumbnail
Fig 7.

The effects of LPS, (+)-JQ1 and (-)-JQ1 on histone 3 acetylation at the IL6 (Panel A) CXCL8/IL8 (Panel B) and TNF (Panel C) promoters in DSCs. Primary cultures of DSCs were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations indicated under the columns. Histone 3 acetylation was determined by chromatin immunoprecipitation (ChIP) using a pan-acetyl histone 3 antibody (listed in S2 Table), and the results were expressed relative to the negative control IgG. Further details of the experimental procedures and the data processing are described in the Materials and methods section. Columns show the mean ± SEM, n = 5–6 independent experiments. Significant differences (at 5% FDR), highlighted in the heatmap in Fig 6, are indicated by the horizontal orange and blue lines. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.g007

thumbnail
Fig 8.

The effects of LPS, (+)-JQ1, and (-)-JQ1 on histone 3 acetylation at the IL10 (Panel A) and IDO1 (Panel B) promoters in DSCs. Primary cultures of DSCs were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations as indicated under the columns. Histone 3 acetylation was determined by chromatin immunoprecipitation (ChIP). Columns show the mean ± SEM, n = 5–6 independent experiments. Significant differences (at 5% FDR), highlighted in the heatmap in Fig 6, are indicated by the horizontal orange and blue lines. Further details are described in the legend in Fig 7. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.g008

We have detected histone 3 acetylation at the IL6, CXCL8/IL8, and TNF promoters in unstimulated DSCs (vehicle vs. the ChIP negative control, IgG). Histone 3 acetylation was not detected at the promoters of the anti-inflammatory IL10 and IDO1 genes in unstimulated cells. LPS treatment induced histone 3 acetylation at the IDO1 promoter but not at the other promoters (LPS vs. Vehicle). Treatment with (+)-JQ1 alone resulted in a significant decrease in acH3 level at the IL6 promoter, which was also seen in the presence of the negative control compound, (-)-JQ1 ((+)-JQ1 or (-)-JQ1 vs. Vehicle). Conversely, (+)-JQ1 alone increased histone 3 acetylation at the IL10 and IDO1 promoters, but this effect was not shared by (-)-JQ1. Treatment with (-)-JQ1, but not with (+)-JQ1, reduced acH3 levels at the IL6 and CXCL8/IL8 promoters after LPS exposure (LPS + (+)-JQ1 or (-)JQ1 vs. LPS). Furthermore, (+)-JQ1 increased histone 3 acetylation at the TNF and IL10 promoters in response to LPS, but the negative control drug (-)-JQ1 was ineffective at these sites. At the IDO1 promoter, histone 3 acetylation after LPS treatment was unaffected by the drugs. Overall, the differences in Histone 3 acetylation showed no consistent relationship with the gene expression changes detected in response to the same treatments.

Acetyl histone 4.

The histone 4 acetylation responses to LPS, (+)-JQ1, and (-)-JQ1 are summarized in the heatmap in Fig 9. As before, a heat map containing the full list of contrasts is presented in the S6 Fig. Acetyl-H4 ChIP results in each gene promoter under all treatment conditions are displayed in Figs 10 and 11, and significant differences highlighted in Fig 9 are indicated. In unstimulated cells, we have detected histone 4 acetylation at the IL6 promoter (relative to IgG). After LPS stimulation, the acH4 levels increased significantly at the TNF and the IDO1 promoters relative to the vehicle. Notably, the acH4 level at the CXCL8/IL8 promoter was significant in LPS-treated cells relative to the IgG negative control but not compared to vehicle-treated cells, which had an acH4 level similar to IgG, suggesting indirectly that LPS stimulated histone 4 acetylation at this locus, as well. Treatment with (+)-JQ1 alone increased histone 4 acetylation of the IL10 and the IDO1 promoters relative to the vehicle, while (-)-JQ1 did not show this effect. At the IL6 promoter, (-)-JQ1, but not its active analog, (+)-JQ1, reduced histone 4 acetylation relative to vehicle treatment (Fig 10A). The drugs did not influence histone 4 acetylation at the CXCL8/IL8, IL10, and IDO1 promoters in response to LPS. At the IL6 promoter, treatment with (-)-JQ1, but not with (+)-JQ1, reduced acH4 levels in response to LPS. Like with histone 3, histone 4 acetylation changes at the promoters showed no consistent relationship to the gene expression changes after the same treatments.

thumbnail
Fig 9. Heat map grid summarizing the effects of LPS, (+)-JQ, and (-)-JQ1 on histone 4 acetylation at the promoters of IL6, CXCL8/IL8, TNF, IL10, and IDO1.

Culture and treatment conditions, chromatin immunoprecipitation (ChIP) details, and data processing are described in the Materials and methods section. The grid cells show significance values (Q) adjusted to the false discovery rate (FDR). The significance threshold is 5% FDR (Q<0.05), highlighted in bold. The orange color indicates a significant increase, the blue color indicates a significant decrease in the acH3 ChIP signal regarding the contrasts shown. The gene symbols are in red. The Q values have been calculated by contrasting all treatments (21 contrasts, listed in S6 Fig). The contrasts selected for display in the heatmap indicate LPS effects (LPS vs. Vehicle), drug effects ((+)-JQ1 or (-)-JQ1 vs. Vehicle), and drug effects on the LPS response (LPS + (+)-JQ1 or LPS + (-)-JQ1 vs. LPS).

https://doi.org/10.1371/journal.pone.0280645.g009

thumbnail
Fig 10.

The effects of LPS, (+)-JQ1 and (-)-JQ1 on histone 4 acetylation at the IL6 (Panel A) CXCL8/IL8 (Panel B) and TNF (Panel C) promoters in DSCs. Primary cultures of DSCs were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations indicated under the columns. Chromatin immunoprecipitation (ChIP) was performed using a pan-acetyl histone 4 antibody (S2 Table), and the results were expressed relative to the negative control IgG. Further details of the experimental procedures and the data processing are described in the legend of Fig 7. Columns show the mean ± SEM, n = 5–6 independent experiments. Significant differences (at 5% FDR), highlighted in the heatmap in Fig 9, are indicated by the horizontal orange and blue lines. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.g010

thumbnail
Fig 11.

The effects of LPS, (+)-JQ1 and (-)-JQ1 on histone 4 acetylation at the IL10 (Panel A) and IDO1 (Panel B) promoters in DSCs. Experimental details and data presentation are the same as in Fig 10. Columns show the mean ± SEM, n = 5–6 independent experiments. Significant differences (at 5% FDR), highlighted in the heatmap in Fig 9, are indicated by the horizontal orange and blue lines. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.g011

BRD2 and BRD4.

ChIP analysis with antibodies verified for chromatin immunoprecipitation did not detect significant BRD2 or BRD4 binding to the promoter of the investigated genes at the 5% FDR level under our experimental conditions. The contrast lists of BRD2 and BRD4 ChIP results are provided in S7 and S10 Figs, respectively. The diagrams showing the BRD2 and BRD4 ChIP results in individual gene promoters under all treatment conditions are presented in S8 and S9 Figs for BRD2 and S11 and S12 Figs for BRD4.

Discussion

Pharmacological agents that bind the tandem bromodomains of the BET family transcription factors have been used successfully to determine the involvement of these proteins in regulatory events at the molecular level. Blocking the bromodomains interferes with the binding of BETs to lysine-acetylated histones disrupting their gene regulatory function. Two of the best-characterized members of the BET inhibitors are (+)-JQ1 and I-BET-762. When these compounds are employed at a concentration of ≤ 1 μM, and their effects are contrasted to an analog of a similar structure practically inactive on the target, conclusions about functional BET involvement can be drawn with confidence. The verified control compound we used was (-)-JQ1, the enantiomer of (+)-JQ1 [31, 37]. The pharmacological selectivity of BET inhibitors provided an opportunity to assess the involvement of the BET proteins in the regulation of inflammatory genes in DSCs critical for pregnancy maintenance and the timing of labor [7, 38, 39].

We have established primary cultures of decidual stromal cells isolated from term placentas by adapting published procedures [10]. Cells were purified by removing CD45+ leukocytes and cultured without passage in the presence of estrogen and progestogen for a maximum of 20 days to approach conditions preserving the in vivo DSC phenotype. LPS was employed as the inflammatory stimulus to model infectious insults the decidua may be exposed to under pathological conditions. We chose to determine treatment effects on genes prototypical of pro-and anti-inflammatory responses. The target genes included PTGS1, PTGS2, and PTGES, coding for Prostaglandin G/H Synthase 1, -2; and Prostaglandin E Synthase, which are critical enzymes of the prostaglandin synthesis pathway; IL6 (encoding Interleukin-6, a multifunctional proinflammatory cytokine), CXCL8/IL8 (responsible for producing Interleukin-8, a neutrophil chemokine and proinflammatory factor), and TNF (the gene for Tumor Necrosis Factor, a pleiotropic cytokine critically involved in inflammatory responses). We have also studied genes encoding anti-inflammatory factors such as IL-10 (Interleukin-10) and IDO-1 (Indoleamine 2,3-Dioxygenase 1), a secreted tryptophan metabolizing enzyme with potent immune-mitigating and antimicrobial actions [40].

LPS stimulated the expression of all studied genes except PTGS1 and PTGES, which had constitutively high mRNA levels. Notably, both the pro-inflammatory genes (PTGS2, IL6, CXCL8/IL8, TNF) and the anti-inflammatory genes (IL10 and IDO1) in our panel responded to LPS treatment. The responses were robust, even considering that the primary cells could have been exposed to infectious stimuli in situ before isolation. We took care to process tissues after elective operative (Cesarean) deliveries without clinical or histological signs of intrauterine infection or inflammation to minimize the risk of in situ LPS exposure. Nevertheless, this possibility represents a limitation of the study. The BET inhibitor effects on mRNA levels were consistent with the involvement of BETs in the LPS responses except for TNF, which was not influenced significantly by (+)-JQ1, I-BET-762, or (-)-JQ1. PTGS2, IL6, and IL10 mRNA levels were reduced by (+)-JQ1 and I-BET-734, but not (-)-JQ1, in unstimulated cells, indicating BET involvement in basal expression. The lack of BET inhibitor effect on TNF expression suggests that there is a BET-independent pathway of LPS action in DSCs. TNF-a has been reported to stimulate the IL-6 [41], IL-8 [9], IL-10 [42], and prostaglandin [4245] production of decidual cells or tissue in vitro, and time course and immunoneutralization experiments suggested that TNF-a may mediate or augment the action of LPS on inflammatory mediator release from choriodecidua explants [42]. Moreover, TNF-a has been shown to promote the proinflammatory transformation of endothelial (HUVEC) cells by a mechanism that includes the redistribution of BRD4, one of the BETs dominantly expressed in decidual cells, to super-enhancer chromatin loci controlling inflammatory genes [19]. Our data are compatible with an LPS-induced, TNF-a-mediated, and BRD4 -dependent mechanism of inflammatory transition of DSCs. Firm evidence for this scenario will require integrated genome-wide data of transcriptome changes, chromatin remodeling, and BET localization in response to LPS and other labor-associated stimulants.

The chromatin immunoprecipitation study aimed to gain molecular-level information about BET involvement in LPS actions on our inflammatory gene panel. BET involvement in gene regulation is diverse and multifaceted [22]. We have focused on the proximal promoter regions (within 1kb upstream of the transcription start site) where BRD2 and BRD4 were reported to control many genes [36, 46]. Nonetheless, we detected no significant binding of BRD2 or BRD4 at 5% FDR to the proximal promoter of the investigated genes. However, a low level of binding cannot be excluded because some unadjusted significance values were below the P = 0.05 cut-off (S5 and S7 Figs). The BRD2 and the BRD4 antibodies have been ChIP certified, and we have employed a dual fixation procedure to detect both direct and indirect (protein-to-protein mediated) binding to DNA [47]. We cannot exclude that technical reasons, e.g., epitope masking, may have contributed to the low ChIP signal. Furthermore, we have assayed BET binding after extended treatments optimized for mRNA changes, which could not detect possible early transient BRD binding event(s). Establishing the time course of BET binding to a broader array of candidate genes and regulatory chromatin regions after stimulation, especially enhancers and super-enhancers [21, 22], is a critical next approach to delineating the relationship between chromatin-bound BRDs and target gene expression in DSCs.

Histone 3 acetylation was detected at the promoters of three of the five genes examined (IL6, CXCL8/IL8, TNF). LPS did not affect acH3 levels at these promoters or at the promoter of IL10, which was not acetylated in the unstimulated state. The expression of these genes, however, was significantly enhanced by LPS. In our limited panel of LPS target genes, only IDO1 exhibited a concomitant increase in mRNA expression and promoter histone 3 acetylation after endotoxin treatment. This indicates that histone 3 acetylation at the promoter is not a general obligatory requirement for LPS-induced inflammatory gene expression in DSCs. A similar conclusion can be drawn about histone 4 acetylation, which was found only at the IL6 promoter in the unstimulated state and was induced by LPS only at the TNF and CXCL8/IL8 promoters (vs. the IgG control) despite upregulated expression across the panel of genes analyzed by ChIP.

BET inhibitor treatment abrogated histone acetylation at the inflammatory gene panel. One type of response was observed at the IL10 and IDO1 promoters, where (+)-JQ1 increased H3 and H4 acetylation in unstimulated cells. Treatment with (+)-JQ1 enhanced H3 acetylation at the IL10 and TNF promoters in response to LPS. (-)-JQ1 did not show these effects, arguing for BET involvement, which was likely indirect since no BRD binding was detected at these sites. BRD4 has been reported to possess an intrinsic histone acetyltransferase activity [48] and augment histone acetylation by interacting with p300 and CBP [49], but the mechanisms involved in DSCs remain to be established. Notably, acetyl-H3 and -H4 levels at the promoters showed no consistent relationship with gene expression changes. The second type of response was detected at the IL6, and CXCL8/IL8 promoters, where the negative control compound, (-)-JQ1, but not its active analog, (+)-JQ1, reduced histone acetylation in LPS stimulated cells. Furthermore, both (+)-JQ1 and (-)-JQ1 decreased IL6 promoter acH3 levels in unstimulated cells. These actions of (-)-JQ1, even when copied by (+)-JQ1, may be off-target, not involving BETs. Alternatively, molecular dynamics (MD) simulations assessing the binding energies of (+)-JQ1 and (-)-JQ1 to the BRD4 bromodomain have indicated that both compounds possess similar affinities to BRD4 [50]. The more than 100-fold higher IC50 of (-)-JQ1 compared to its chiral isomer, (+)-JQ1 [31], could be attributed to the faster insertion of (+)-JQ1 into the binding pocket of the bromodomain [50]. Despite the slower binding kinetics (-)-JQ1 might still be able to associate with BRDs after more prolonged exposure, such as 72 h in our experiments (48 h pre-treatment and 24 h co-treatment with LPS), especially since the bromodomain dissociation kinetics of (-)-JQ1 is also slower than that of (+)-JQ1 [50]. The MD simulation results may thus allow BET-mediated actions by (-)-JQ1, depending on experimental conditions. Again, the drug-induced changes in acetyl histone levels at the promoters did not correlate with gene activity.

In summary, our results demonstrate that BET proteins are critically involved in the response of DSCs to inflammatory stimulation. Using a panel of candidate genes, we show that both pro-and anti-inflammatory genes respond in a BET-dependent fashion and BET inhibitor drugs inhibit their responses. A BET-independent pathway also exists, mediating the stimulation of TNF expression by LPS. Analysis of acetylated histone levels at the promoters indicates that enhanced histone acetylation is not involved obligatorily in the transcriptional responses. BETs may regulate gene activity by acting at distant (e.g., enhancer) loci rather than at the proximal promoters. Nevertheless, BET inhibitors may represent a new class of pharmacological agents capable of arresting the inflammatory transformation of the decidua at labor with potential therapeutic benefit.

Supporting information

S1 Fig. Flow cytometry analysis of purified decidual stromal cells.

Decidual cells were isolated and purified by Percoll gradient centrifugation and depleted of CD45+ leukocytes by immunomagnetic selection as described in the Materials and methods section. Cells (5x105) were suspended in 0.1 mL PBS, 1% BSA, and 1 μL of Human BD Fc Block (BD Pharmingen 564219) was added, followed by 2 μL of FITC mouse anti-human CD45 (BD Pharmingen 555482). Washed cells were stained with Propidium Iodide (1 μL of 0.5 mg/mL) and analyzed using a Fortessa (BD Biosciences) flow cytometer at recommended settings. A, Gating strategy to identify live single cells (FCC-A, forward scattering area; PI, Propidium Iodide). B, FITC fluorescence histogram of the gated events (FITC-A::CD45). No FITC-positive event was detected in the region of the expected CD45+ subset.

https://doi.org/10.1371/journal.pone.0280645.s001

(TIF)

S2 Fig. Characterization of decidual stromal cells in culture.

Purified decidual stromal cells were cultured to near confluence as described in the Materials and methods section. Cultures were fixed with paraformaldehyde, permeabilized, and blocked with hydrogen peroxide, followed by antigen retrieval using standard procedures (Citric acid, pH 6.0, 95C for 15 min). Non-specific staining was blocked with 10% goat serum. Immunocytochemistry was performed using the VECTASTAIN ABC kit (PK6102, Vector Laboratories) following the manufacturer’s instructions. The primary antibodies were against CD45 (DAKO IR751) or vimentin (DAKO IR630) used at 1:200 dilution. Control IgG was included in the staining kit. Cells were counterstained with hematoxylin. A, IgG negative control; B, CD45; C: vimentin. Brown staining indicates pervasive expression of the DSC marker vimentin (C), while staining for CD45 is absent (B). Magnification is shown under the panels. Size bars indicate 100 μm.

https://doi.org/10.1371/journal.pone.0280645.s002

(TIF)

S3 Fig. Inflammatory gene expression in primary cultures of decidual stromal cells (DSCs).

Purified stromal cells from term decidua were cultured to near confluence, and PTGS1, PTGS2, PTGES, IL6, CXCL8/IL8, TNF, ILI0, and IDO1 mRNA abundance was determined by qRT-PCR relative to a spiked-in reference RNA (Alien). Values were scaled relative to the average in each experiment. Columns represent the mean + SEM, n = 5 independent experiments. The abundance of the mRNAs is significantly different from each other (p = 0.001, K-sample-equality-of-medians test).

https://doi.org/10.1371/journal.pone.0280645.s003

(TIF)

S4 Fig. Heat map grid showing the effects of LPS and BET inhibitors on inflammatory gene expression in DSCs.

Culture and treatment conditions, qRT-PCR assay details, and data processing are described in the Materials and methods. Genes are indicated on the top. The grid cells show unadjusted (P), and false discovery rate (FDR) adjusted significance values (Q) for all contrasts shown on the left. The significance threshold is 5% FDR (Q<0.05), highlighted in bold. Colors indicate an increase, decrease, or no significant difference (white) based on Q.

https://doi.org/10.1371/journal.pone.0280645.s004

(TIF)

S5 Fig. Heat map grid showing the effects of LPS and BET inhibitors on histone 3 acetylation at inflammatory gene promoters in DSCs.

Culture and treatment conditions, ChIP assay details, and data processing are described in the Materials and methods. Genes are indicated on the top. The grid cells show unadjusted (P), and false discovery rate (FDR) adjusted significance values (Q) for all contrasts shown on the left. The significance threshold is 5% FDR (Q<0.05), highlighted in bold. Colors indicate an increase, decrease, or no significant difference (white) based on Q.

https://doi.org/10.1371/journal.pone.0280645.s005

(TIF)

S6 Fig. Heat map grid showing the effects of LPS and BET inhibitors on histone 4 acetylation at inflammatory gene promoters in DSCs.

Culture and treatment conditions, ChIP assay details, and data processing are described in the Materials and methods. Genes are indicated on the top. The grid cells show unadjusted (P), and false discovery rate (FDR) adjusted significance values (Q) for all contrasts shown on the left. The significance threshold is 5% FDR (Q<0.05), highlighted in bold. Colors indicate an increase, decrease, or no significant difference (white) based on Q.

https://doi.org/10.1371/journal.pone.0280645.s006

(TIF)

S7 Fig. Grid showing the effects of LPS and BET inhibitors on BRD2 binding at inflammatory gene promoters in DSCs.

Culture and treatment conditions, ChIP assay details, and data processing are described in the Materials and methods. Genes are indicated on the top. The grid cells show unadjusted (P), and false discovery rate (FDR) adjusted significance values (Q) for all contrasts shown on the left. None of the contrasts reach the significance threshold of 5% FDR (Q<0.05).

https://doi.org/10.1371/journal.pone.0280645.s007

(TIF)

S8 Fig.

ChIP assays to detect BRD2 binding at the IL6 (Panel A) CXCL8/IL8 (Panel B) and TNF (Panel C) promoters in DSCs treated with LPS, (+)-JQ1 and (-)-JQ1. Primary cultures of DSCs were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations indicated under the columns. Chromatin immunoprecipitation (ChIP) was performed using a ChIP-certified BRD2 antibody (S2 Table), and the results were expressed relative to the negative control IgG. Columns show the mean ± SEM, n = 5 independent experiments. No significant BRD2 binding relative to IgG was detected (at 5% FDR) under any treatment condition. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.s008

(TIF)

S9 Fig.

ChIP assays to detect BRD2 binding at the IL10 (Panel A) and IDO1 (Panel B) promoters in DSCs treated with LPS, (+)-JQ1 and (-)-JQ1. Primary cultures of DSCs were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations indicated under the columns. Chromatin immunoprecipitation (ChIP) was performed using a ChIP-certified BRD2 antibody (S2 Table), and the results were expressed relative to the negative control IgG. Columns show the mean ± SEM, n = 4–5 independent experiments. No significant BRD2 binding relative to IgG was detected (at 5% FDR) under any treatment condition. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.s009

(TIF)

S10 Fig. Grid showing the effects of LPS and BET inhibitors on BRD4 binding at inflammatory gene promoters in DSCs.

Culture and treatment conditions, ChIP assay details, and data processing are described in the Materials and methods. Genes are indicated on the top. The grid cells show unadjusted (P), and false discovery rate (FDR) adjusted significance values (Q) for all contrasts shown on the left. None of the contrasts reach the significance threshold at 5% FDR (Q<0.05).

https://doi.org/10.1371/journal.pone.0280645.s010

(TIF)

S11 Fig.

ChIP assays to detect BRD4 binding at the IL6 (Panel A) CXCL8/IL8 (Panel B) and TNF (Panel C) promoters in DSCs treated with LPS, (+)-JQ1 and (-)-JQ1. Primary cultures of DSCs were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations indicated under the columns. Chromatin immunoprecipitation (ChIP) was performed using a ChIP-certified BRD4 antibody (S2 Table), and the results were expressed relative to the negative control IgG. Columns show the mean ± SEM, n = 5–6 independent experiments. No significant BRD4 binding relative to IgG was detected (at 5% FDR) under any treatment condition. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.s011

(TIF)

S12 Fig.

ChIP assays to detect BRD4 binding at the IL10 (Panel A) and IDO1 (Panel B) promoters in DSCs treated with LPS, (+)-JQ1 and (-)-JQ1. Primary cultures of DSCs were treated with vehicle, LPS, (+)-JQ1, (-)-JQ1, and their combinations indicated under the columns. Chromatin immunoprecipitation (ChIP) was performed using a ChIP-certified BRD4 antibody (S2 Table), and the results were expressed relative to the negative control IgG. Columns show the mean ± SEM, n = 5–6 independent experiments. No significant BRD4 binding relative to IgG was detected (at 5% FDR) under any treatment condition. The results of the individual experiments and summary statistics are presented in the S1 Data.

https://doi.org/10.1371/journal.pone.0280645.s012

(TIF)

S1 Data. Minimal underlying dataset of the study.

https://doi.org/10.1371/journal.pone.0280645.s015

(XLSX)

Acknowledgments

The Authors gratefully acknowledge our Research Nurse, Ms. Anne Wright, the John Hunter Hospital Midwifery Staff, and all tissue donors for their essential contribution to the study.

References

  1. 1. Oreshkova T, Dimitrov R, Mourdjeva M. A cross-talk of decidual stromal cells, trophoblast, and immune cells: a prerequisite for the success of pregnancy. Am J Reprod Immunol. 2012;68(5):366–73. Epub 2012/06/08. pmid:22672047.
  2. 2. Sun F, Wang S, Du M. Functional regulation of decidual macrophages during pregnancy. J Reprod Immunol. 2021;143:103264. Epub 2020/12/29. pmid:33360717.
  3. 3. Gomez-Lopez N, Guilbert LJ, Olson DM. Invasion of the leukocytes into the fetal-maternal interface during pregnancy. J Leukoc Biol. 2010;88(4):625–33. pmid:20519637.
  4. 4. He YY, He XJ, Guo PF, Du MR, Shao J, Li MQ, et al. The decidual stromal cells-secreted CCL2 induces and maintains decidual leukocytes into Th2 bias in human early pregnancy. Clin Immunol. 2012;145(2):161–73. pmid:23069648.
  5. 5. Croxatto D, Vacca P, Canegallo F, Conte R, Venturini PL, Moretta L, et al. Stromal cells from human decidua exert a strong inhibitory effect on NK cell function and dendritic cell differentiation. PLoS One. 2014;9(2):e89006. pmid:24586479; PubMed Central PMCID: PMC3930605.
  6. 6. Norwitz ER, Bonney EA, Snegovskikh VV, Williams MA, Phillippe M, Park JS, et al. Molecular Regulation of Parturition: The Role of the Decidual Clock. Cold Spring Harb Perspect Med. 2015;5(11). pmid:25918180; PubMed Central PMCID: PMC4632866.
  7. 7. Menon R, Bonney EA, Condon J, Mesiano S, Taylor RN. Novel concepts on pregnancy clocks and alarms: redundancy and synergy in human parturition. Hum Reprod Update. 2016;22(5):535–60. Epub 2016/07/02. pmid:27363410; PubMed Central PMCID: PMC5001499.
  8. 8. El-Azzamy H, Balogh A, Romero R, Xu Y, LaJeunesse C, Plazyo O, et al. Characteristic Changes in Decidual Gene Expression Signature in Spontaneous Term Parturition. J Pathol Transl Med. 2017;51(3):264–83. pmid:28226203; PubMed Central PMCID: PMC5445200.
  9. 9. Lockwood CJ, Arcuri F, Toti P, Felice CD, Krikun G, Guller S, et al. Tumor necrosis factor-alpha and interleukin-1beta regulate interleukin-8 expression in third trimester decidual cells: implications for the genesis of chorioamnionitis. Am J Pathol. 2006;169(4):1294–302. pmid:17003486; PubMed Central PMCID: PMC1698845.
  10. 10. Lockwood CJ, Murk WK, Kayisli UA, Buchwalder LF, Huang SJ, Arcuri F, et al. Regulation of interleukin-6 expression in human decidual cells and its potential role in chorioamnionitis. Am J Pathol. 2010;177(4):1755–64. pmid:20724602; PubMed Central PMCID: PMC2947272.
  11. 11. Ibrahim SA, Ackerman WEt, Summerfield TL, Lockwood CJ, Schatz F, Kniss DA. Inflammatory gene networks in term human decidual cells define a potential signature for cytokine-mediated parturition. Am J Obstet Gynecol. 2016;214(2):284 e1- e47. pmid:26348374.
  12. 12. Arntzen KJ, Egeberg K, Rahimipoor S, Vatten L, Austgulen R. LPS mediated production of IL-1, PGE2 and PGF2alpha from term decidua involves tumour necrosis factor and tumour necrosis factor receptor p55. J Reprod Immunol. 1999;45(2):113–25. pmid:10674980.
  13. 13. Dudley DJ, Edwin SS, Van Wagoner J, Augustine NH, Hill HR, Mitchell MD. Regulation of decidual cell chemokine production by group B streptococci and purified bacterial cell wall components. Am J Obstet Gynecol. 1997;177(3):666–72. pmid:9322640.
  14. 14. Dudley DJ, Edwin SS, Dangerfield A, Jackson K, Trautman MS. Regulation of decidual cell and chorion cell production of interleukin-10 by purified bacterial products. Am J Reprod Immunol. 1997;38(4):246–51. pmid:9352010.
  15. 15. Smale ST. Selective Transcription in Response to an Inflammatory Stimulus. Cell. 2010;140(6):833–44. pmid:20303874
  16. 16. Simonatto M, Natoli G. Functional genomics of the inflammatory response: where are we now? Brief Funct Genomics. 2013;12(6):483–8. Epub 2013/07/03. pmid:23814131.
  17. 17. Ghizzoni M, Haisma HJ, Maarsingh H, Dekker FJ. Histone acetyltransferases are crucial regulators in NF-kappaB mediated inflammation. Drug Discov Today. 2011;16(11–12):504–11. Epub 2011/04/12. pmid:21477662; PubMed Central PMCID: PMC5218544.
  18. 18. Huang B, Yang XD, Zhou MM, Ozato K, Chen LF. Brd4 coactivates transcriptional activation of NF-kappaB via specific binding to acetylated RelA. Mol Cell Biol. 2009;29(5):1375–87. Epub 2008/12/24. pmid:19103749; PubMed Central PMCID: PMC2643823.
  19. 19. Brown JD, Lin CY, Duan Q, Griffin G, Federation A, Paranal RM, et al. NF-kappaB directs dynamic super enhancer formation in inflammation and atherogenesis. Mol Cell. 2014;56(2):219–31. Epub 2014/09/30. pmid:25263595; PubMed Central PMCID: PMC4224636.
  20. 20. Belkina AC, Nikolajczyk BS, Denis GV. BET protein function is required for inflammation: Brd2 genetic disruption and BET inhibitor JQ1 impair mouse macrophage inflammatory responses. J Immunol. 2013;190(7):3670–8. Epub 2013/02/20. pmid:23420887; PubMed Central PMCID: PMC3608815.
  21. 21. Amaral PP, Bannister AJ. Re-place your BETs: the dynamics of super enhancers. Mol Cell. 2014;56(2):187–9. Epub 2014/11/07. pmid:25373538.
  22. 22. Cheung KL, Kim C, Zhou MM. The Functions of BET Proteins in Gene Transcription of Biology and Diseases. Front Mol Biosci. 2021;8:728777. Epub 2021/09/21. pmid:34540900; PubMed Central PMCID: PMC8446420.
  23. 23. Noguchi-Yachide T. BET Bromodomain as a Target of Epigenetic Therapy. Chem Pharm Bull (Tokyo). 2016;64(6):540–7. Epub 2016/06/03. pmid:27250788.
  24. 24. Chung CW, Coste H, White JH, Mirguet O, Wilde J, Gosmini RL, et al. Discovery and characterization of small molecule inhibitors of the BET family bromodomains. J Med Chem. 2011;54(11):3827–38. Epub 2011/05/17. pmid:21568322.
  25. 25. Wang N, Wu R, Tang D, Kang R. The BET family in immunity and disease. Signal Transduct Target Ther. 2021;6(1):23. Epub 2021/01/20. pmid:33462181; PubMed Central PMCID: PMC7813845.
  26. 26. Kanematsu D, Shofuda T, Yamamoto A, Ban C, Ueda T, Yamasaki M, et al. Isolation and cellular properties of mesenchymal cells derived from the decidua of human term placenta. Differentiation. 2011;82(2):77–88. Epub 2011/06/21. pmid:21684674.
  27. 27. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8. pmid:11846609.
  28. 28. Nowak DE, Tian B, Brasier AR. Two-step cross-linking method for identification of NF-kappaB gene network by chromatin immunoprecipitation. Biotechniques. 2005;39(5):715–25. pmid:16315372.
  29. 29. Dahl JA, Collas P. Q2ChIP, a quick and quantitative chromatin immunoprecipitation assay, unravels epigenetic dynamics of developmentally regulated genes in human carcinoma cells. Stem Cells. 2007;25(4):1037–46. pmid:17272500.
  30. 30. Smith RD, Brown B, Ikonomi P, Schechter AN. Exogenous reference RNA for normalization of real-time quantitative PCR. Biotechniques. 2003;34(1):88–91. pmid:12545545.
  31. 31. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O, et al. Selective inhibition of BET bromodomains. Nature. 2010;468(7327):1067–73. Epub 2010/09/28. pmid:20871596; PubMed Central PMCID: PMC3010259.
  32. 32. Mirguet O, Gosmini R, Toum J, Clement CA, Barnathan M, Brusq JM, et al. Discovery of epigenetic regulator I-BET762: lead optimization to afford a clinical candidate inhibitor of the BET bromodomains. J Med Chem. 2013;56(19):7501–15. Epub 2013/09/11. pmid:24015967.
  33. 33. Ferri E, Petosa C, McKenna CE. Bromodomains: Structure, function and pharmacology of inhibition. Biochem Pharmacol. 2016;106:1–18. Epub 2015/12/29. pmid:26707800.
  34. 34. Hsu SC, Blobel GA. The Role of Bromodomain and Extraterminal Motif (BET) Proteins in Chromatin Structure. Cold Spring Harb Symp Quant Biol. 2017;82:37–43. Epub 2017/12/03. pmid:29196562.
  35. 35. Jang MK, Mochizuki K, Zhou M, Jeong HS, Brady JN, Ozato K. The bromodomain protein Brd4 is a positive regulatory component of P-TEFb and stimulates RNA polymerase II-dependent transcription. Mol Cell. 2005;19(4):523–34. Epub 2005/08/20. pmid:16109376.
  36. 36. Hnilicova J, Hozeifi S, Stejskalova E, Duskova E, Poser I, Humpolickova J, et al. The C-terminal domain of Brd2 is important for chromatin interaction and regulation of transcription and alternative splicing. Mol Biol Cell. 2013;24(22):3557–68. Epub 2013/09/21. pmid:24048450; PubMed Central PMCID: PMC3826993.
  37. 37. Tang S, Schapira M. Are Enentiomer of Chemical Probes Good Negative Control? [Data set]. ZENODO. 2021;https://doi.org/10.5281/zenodo.5031252.
  38. 38. Wijaya JC, Khanabdali R, Georgiou HM, Kalionis B. Ageing in human parturition: impetus of the gestation clock in the decidua. Biol Reprod. 2020;103(4):695–710. Epub 2020/06/28. pmid:32591788.
  39. 39. Lim R, Nguyen-Ngo C, Lappas M. Targeting bromodomain-containing proteins to prevent spontaneous preterm birth. Clinical Science. 2019;133(23):2379–400. pmid:31750510
  40. 40. Yeung AW, Terentis AC, King NJ, Thomas SR. Role of indoleamine 2,3-dioxygenase in health and disease. Clin Sci (Lond). 2015;129(7):601–72. Epub 2015/07/18. pmid:26186743.
  41. 41. Dudley DJ, Trautman MS, Araneo BA, Edwin SS, Mitchell MD. Decidual cell biosynthesis of interleukin-6: regulation by inflammatory cytokines. J Clin Endocrinol Metab. 1992;74(4):884–9. pmid:1548355
  42. 42. Sato TA, Keelan JA, Mitchell MD. Critical paracrine interactions between TNF-alpha and IL-10 regulate lipopolysaccharide-stimulated human choriodecidual cytokine and prostaglandin E2 production. J Immunol. 2003;170(1):158–66. pmid:12496396.
  43. 43. Kent AS, Sullivan MH, Sun MY, Zosmer A, Elder MG. Effects of interleukin-6 and tumor necrosis factor-alpha on prostaglandin production by cultured human fetal membranes. Prostaglandins. 1993;46(4):351–9. pmid:8248548
  44. 44. Mitchell MD, Edwin S, Romero RJ. Prostaglandin biosynthesis by human decidual cells: effects of inflammatory mediators. Prostaglandins Leukot Essent Fatty Acids. 1990;41(1):35–8. pmid:2251294
  45. 45. Pollard JK, Thai D, Mitchell MD. Mechanism of cytokine stimulation of prostaglandin biosynthesis in human decidua. J Soc Gynecol Investig. 1994;1(1):31–6. pmid:9419743
  46. 46. Yang Z, Yik JH, Chen R, He N, Jang MK, Ozato K, et al. Recruitment of P-TEFb for stimulation of transcriptional elongation by the bromodomain protein Brd4. Mol Cell. 2005;19(4):535–45. Epub 2005/08/20. pmid:16109377.
  47. 47. Zeng PY, Vakoc CR, Chen ZC, Blobel GA, Berger SL. In vivo dual cross-linking for identification of indirect DNA-associated proteins by chromatin immunoprecipitation. Biotechniques. 2006;41(6):694, 6, 8. Epub 2006/12/29. pmid:17191611.
  48. 48. Devaiah BN, Case-Borden C, Gegonne A, Hsu CH, Chen Q, Meerzaman D, et al. BRD4 is a histone acetyltransferase that evicts nucleosomes from chromatin. Nature structural & molecular biology. 2016;23(6):540–8. Epub 2016/05/10. pmid:27159561; PubMed Central PMCID: PMC4899182.
  49. 49. Wu T, Kamikawa YF, Donohoe ME. Brd4’s Bromodomains Mediate Histone H3 Acetylation and Chromatin Remodeling in Pluripotent Cells through P300 and Brg1. Cell Rep. 2018;25(7):1756–71. Epub 2018/11/15. pmid:30428346.
  50. 50. Kuang M, Zhou J, Wang L, Liu Z, Guo J, Wu R. Binding Kinetics versus Affinities in BRD4 Inhibition. Journal of chemical information and modeling. 2015;55(9):1926–35. pmid:26263125