Skip to main content

Echinococcus granulosus cyst fluid inhibits inflammatory responses through inducing histone demethylase KDM5B in macrophages

Abstract

Background

Echinococcus granulosus cyst fluid (EgCF) weakens macrophage inflammatory responses, thereby enabling the parasite to evade the immune system. However, the role of histone modification in this process remains to be explored.

Methods

The levels of IL-6, TNF-α, IL-10, H3K4me3, and KDM5B were detected using quantitative real-time PCR, ELISA, and Western blotting. The enrichment of H3K4me3 and KDM5B at the promoter of inflammatory factors was detected by chromatin immunoprecipitation.

Results

Based on EgCF-stimulated macrophage models, we found that EgCF significantly inhibited mRNA expression and protein secretion of IL-6 and TNF-α and upregulated mRNA expression of IL-10 under the influence of TLR4. EgCF lowered the level of H3K4me3 and promoted the transcription and protein stability of histone demethylase KDM5B. Chromatin immunoprecipitation analysis revealed that EgCF suppressed the enrichment of H3K4me3 modification at the promoters of TNF-α and IL-6 and downregulated their expression in macrophages. Additionally, the inhibition of KDM5B activity by CPI-455 weakened the anti-inflammatory effect of EgCF.

Conclusions

Our findings demonstrate a novel mechanism through which EgCF promotes KDM5B expression and inhibits the enrichment of H3K4me3 at the promoters of inflammatory cytokines to suppress the inflammatory response.

Graphical Abstract

Introduction

Echinococcosis refers to two serious zoonoses, which are mainly defined as cystic echinococcosis (CE) and alveolar echinococcosis caused by Echinococcus granulosus and E. multilocularis in humans, respectively [1, 2]. Generally, the CE cyst includes the parasite larva which establishes in the intermediate host (mainly domestic ungulates); the definitive host (carnivore) contains the adult worm [3]. Humans may become an intermediate host of Echinococcus granulosus due to inadvertent ingestion of parasite eggs present in contaminated water or food. Established fluid-filled CE cysts are primarily located in the liver and lung [1, 4]. Surgery is the primary option for treating echinococcosis, but surgery can also cause secondary cysts [1, 5]. In addition, early diagnosis of echinococcosis remains difficult, and drugs may cause adverse events [1]. Therefore, it is imperative to explore the pathogenic mechanisms of echinococcosis and develop novel anti-parasitic treatments.

Long-term survival of Echinococcus granulosus in the host suggests that the parasite has evolved certain mechanisms to avoid immune response from the host [4]. Mechanisms have not been understood yet but some larval components are candidates as they showed immunomodulatory effects on innate immune cells [6]. Notably, E. granulosus cyst fluid (EgCF) is a complex mixture of components that could modulate immune responses [7, 8]. EgCF contains many secreted proteins helpful to parasite survival [9]. For instance, glycomolecules in the EgCF can interfere with dendritic cell maturation and activation mediated by the TLR4 signaling pathway [10,11,12].

Macrophages are critical mediators of pathogen-driven escape [13]. Depending on the microenvironment, macrophages can acquire distinct functional phenotypes including classically activated macrophages (M1 macrophages) and alternatively activated macrophages (M2 macrophages) [14]. Under the influence of LPS, macrophages can be polarized to the M1 phenotype and secrete high levels of pro-inflammatory cytokines such as TNF-α and IL-6 [14]. Recent evidence suggests that the function of macrophages can be regulated by histone modifications [15], which are dynamically regulated by histone-modifying enzymes [16]. KDM5B as a histone demethylase belongs to the KDM5 family [17]. It is a JmjC-containing demethylase that selectively targets H3K4me3 [15], a well-known marker of active transcription [18]. KDM5B can inhibit transcription factor NF-κB binding to IL-6 and IL-23a promoters through demethylating H3K4me3 in bone marrow-derived macrophages (BMDMs) [19]. However, the function of KDM5B in macrophages during cystic echinococcosis is poorly understood.

This study investigated the role of histone modification in suppressing macrophage inflammatory responses resulting from EgCF. The current results showed that EgCF promoted the transcription and protein stability of KDM5B, which inhibited the enrichment of H3K4me3 at the promoters of inflammatory factors and suppressed the inflammatory response.

Materials and methods

Cell culture

The mouse macrophage cell line RAW 264.7 and the human monocyte cell line THP-1 were purchased from the Cell Bank of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Wuhan Procell Life Science and Technology Co., Ltd., respectively. RAW 264.7 cells were maintained in DMEM (Gibco) containing 10% heat-inactivated fetal bovine serum (FBS, Gibco) and 1% penicillin-streptomycin (Gibco) in a humidified atmosphere with 5% CO2 at 37 °C following the provider’s instructions. THP-1 cells were cultured in RPMI 1640 (Gibco) supplemented with 10% FBS, 1% penicillin-streptomycin, and 0.05 mM β-mercaptoethanol (Gibco). THP-1 cells were differentiated into macrophages by treating the cells with 100 nM phorbol 12-myristate 13-acetate (PMA) for 24 h (h) [20,21,22,23].

Mouse peritoneal macrophages were isolated from C57BL/6 female mice. Briefly, the mice were killed and intraperitoneally injected with DMEM using a 5-ml syringe. The abdomen was gently massaged for 5 min without removing the needle. Subsequently, the fluid was slowly withdrawn from the abdominal cavity via the syringe. The collected peritoneal fluid was centrifuged at 300 g for 5 min. Next, the precipitated cells were washed and seeded into six-well plates at a density of 1 × 106 cells per well in 2 ml medium (DMEM supplemented with 1% l-glutamine, 10% FBS, and 1% penicillin-streptomycin). Non-adherent cells were removed after 2-h culture and fed with 2 ml of fresh medium. The adherent cells were treated with LPS and EgCF the following day, as previously reported [12, 24].

Preparation of Echinococcus granulosus (sensu lato) cyst fluid

Sheep liver CE cysts were obtained from a slaughterhouse in Shihezi, Xinjiang, China. Briefly, the liver and operating instruments were sterilized using 75% alcohol. The fluid was withdrawn from the cysts using a sterile 50-ml syringe and fully mixed. After standing, the clear and transparent cyst fluid in the upper layer was centrifuged at 1000 g for 10 min and filtered through a 0.22-μM filter. The EgCF employed was a pool generated from different cysts and stored in a − 80 °C freezer after protein concentration had been estimated by NanoDrop (4 mg/ml).

Quantitative real-time PCR

RNA was extracted using a total RNA kit (Omega) and reverse-transcribed using HiFiScript cDNA Synthesis Kit (Cwbio) following the manufacturer’s instructions. Quantitative real-time PCR (qRT-PCR) was conducted utilizing the UltraSYBR One-Step RT-qPCR Kit (Cwbio) with specific primers in a CFX96 Touch detection system (Bio-Rad). Relative expression levels of genes were normalized to β-actin by the 2−ΔΔCt method. Primer sequences are listed in Table 1.

Table 1 Primer sequences for qRT-PCR and ChIP assay

ELISA

RAW 264.7 cells and differentiated THP-1 cells were treated with LPS (1 μg/ml) in the presence or absence of EgCF (50% v/v) for 24 h. The cell culture medium was collected and centrifuged at 1000 g for 10 min. The TNF-α and IL-6 concentrations in the harvested supernatants were measured using ELISA kits (Multi Sciences).

Western blotting

Cells were washed and lysed in the RIPA lysis buffer. Protein lysates were electrophoretically separated on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to PVDF membranes. Five percent bovine serum albumin dissolved in Tris-buffered saline containing 0.1% Tween-20 (TBST) was used to block the membranes for 2 h. Next, incubation was performed at 4 °C overnight with primary antibodies specific to H3K4me3 (CST #9751), H3K9me3 (CST #13969), H3K36me3 (CST #4909), Histone H3 (BOSTER A12477-2), KDM5B (CST #15327), and β-actin (ZSGB-BIO #TA-09). After washing three times with TBST for 10 min, the membranes were further incubated with peroxidase-labeled secondary antibodies for 1 h at room temperature. The immune complexes were visualized with the ECL substrate (Biosharp BL520A).

Chromatin immunoprecipitation

The chromatin immunoprecipitation (ChIP) assay was performed using the SimpleChIP Plus Enzymatic Chromatin IP Kit (Agarose Beads; CST #9002) according to the manufacturer’s instructions. Cells were cross-linked with a final concentration of 1% formaldehyde for 10 min, and the reaction was terminated by adding glycine at room temperature. The cells were washed with PBS and incubated in lysis buffer on ice for 10 min and disrupted using an ultrasonic cell disruptor. After centrifugation, chromatin was prepared by incubating with specific primary Ab and anti-IgG overnight at 4 °C and precipitated using ChIP-Grade Protein G Agarose Beads. Following the reversal of the protein-DNA crosslinking, DNA was purified using DNA Purification Buffers and Spin Columns (CST #14209). The enrichment of specific DNA sequences following immunoprecipitation was determined by semi-quantitative PCR (semi-qPCR) and qRT-PCR with primers listed in Table 1. Data were normalized to input DNA, and IgG was used as a negative control.

Statistical analysis

All the experiments were independently repeated three times. Data were represented as mean + standard error of the mean (SEM). Statistical analysis was performed using one-way ANOVA for comparing differences between groups in GraphPad Prism (version 8). * P < 0.05, ** P < 0.01, and *** P < 0.001 indicate a statistically significant difference.

Results

EgCF inhibited inflammatory response mediated by TLR4 signaling pathway in macrophages

We first checked cell viability after treatment for all cell sources to ensure effects were not due to cell death (Additional file 1: Fig. S1). Subsequently, we detected the mRNA expression level of IL-6, TNF-α, and IL-10 in mouse RAW 264.7 cells stimulated with LPS and EgCF for 6 h. qRT-PCR analysis showed that simultaneous stimulation of EgCF and LPS significantly inhibited the mRNA expression of IL-6 and TNF-α compared with using LPS stimulation alone (Fig. 1A and B). Contrarily, co-stimulation of LPS and EgCF upregulated the mRNA expression of IL-10 (Fig. 1C). Similarly, co-stimulation of EgCF and LPS in human THP-1 cells also markedly suppressed the mRNA expression of IL-6 and TNF-α and promoted the mRNA expression of IL-10 (Fig. 1D–F). Then, we detected IL-6 and TNF-α protein secretion in supernatants of RAW 264.7 cells and THP-1 cells stimulated with EgCF and LPS for 24 h. ELISA analysis demonstrated that compared with using LPS stimulation alone, the protein levels of IL-6 and TNF-α in cell supernatants were reduced after simultaneous stimulation of EgCF and LPS (Fig. 2A–D). These results indicated that EgCF inhibited macrophage inflammatory responses mediated by the TLR4 signaling pathway.

Fig. 1
figure 1

EgCF suppressed the mRNA expression of inflammatory factors in macrophages. RAW 264.7 cells were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h. The mRNA expression levels of IL-6 (A), TNF-α (B), and IL-10 (C) were detected by qRT-PCR. THP-1 cells were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h. The mRNA expression levels of IL-6 (D), TNF-α (E), and IL-10 (F) were detected by qRT-PCR. Data are presented as the mean + SEM of three independent experiments and compared using the one-way ANOVA and Tukey test. Asterisks indicate a significant difference at **P < 0.01 and ***P < 0.001

Fig. 2
figure 2

EgCF inhibited the secretion of inflammatory factors in macrophages. RAW 264.7 cells were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 24 h. Secretion of IL-6 (A) and TNF-α (B) in cell culture supernatants was measured by ELISA. THP-1 cells were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 24 h. Secretion of IL-6 (C) and TNF-α (D) in cell culture supernatants was measured by ELISA. Data are presented as the mean + SEM of three independent experiments and compared using the one-way ANOVA and Tukey test. Asterisks indicate a significant difference at **P < 0.01 and ***P < 0.001

EgCF suppressed H3K4me3 modification

Methylation of H3K4 and H3K36 typically suggests active transcription, whereas H3K9 methylation is associated with a silent chromatin state [25, 26]. THP-1 and RAW 264.7 cells were stimulated with EgCF and LPS. After 6 h of stimulation, the global levels of H3K4me3, H3K9me3, and H3K36me3 were detected by Western blotting. Our results indicated that the co-stimulation of LPS and EgCF inhibited the global levels of H3K4me3 compared with the stimulation of LPS alone, but there was no obvious difference in H3K9me3 or H3K36me3 level (Fig. 3A and  B). We also found a high enrichment of H3K4me3 modification at the promoter of IL-6 and TNF-α in human THP-1 cells and mouse BMDMs based on public chromatin immunoprecipitation and sequencing (ChIP-seq) data (Fig. 3C–F). These results indicated that EgCF interfered with H3K4me3 modification.

Fig. 3
figure 3

EgCF reduced H3K4me3 modification. THP-1 and RAW 264.7 cells were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h. Levels of H3K4me3, H3K9me3, and H3K36me3 were measured by Western blotting (A, B). Enrichment of H3K4me3 at IL-6 and TNF-α promoters in mouse bone marrow-derived macrophages (GSM2419196, GSM2419204, GSM2419211) (C, D) and human THP-1 cells (GSM2108047) (E, F)

EgCF promoted transcription and protein stability of KDM5B

Histone demethylases could modulate macrophage inflammatory responses by removing methyl groups from lysine and arginine residues [15, 27]. To investigate the relationship between histone demethylases and the anti-inflammatory effects of EgCF, we detected the expression of histone demethylase genes (Fig. 4A) and identified the differentially expressed histone demethylase KDM5B (Fig. 4B) using RNA-seq data of mouse peritoneal macrophages treated with LPS and EgCF. To verify the finding, mouse peritoneal macrophages (PM) stimulated with EgCF and LPS for 6 h were assessed by qRT-PCR. The results showed that the EgCF treatment upregulated the mRNA expression of KDM5B and KDM7A but downregulated the mRNA expression of KDM6B (Fig. 4C). As KDM5B is a demethylase specifically demethylating H3K4me3, we speculated that KDM5B might affect the anti-inflammatory effect of EgCF through regulating H3K4me3 modification. RAW 264.7 cells were also stimulated with EgCF and LPS for 6 h to measure KDM5B mRNA and protein levels by qRT-PCR and Western blotting, respectively. The qRT-PCR results showed that the co-stimulation of LPS and EgCF markedly upregulated the mRNA expression of KDM5B compared with using LPS alone (Fig. 4D), which was similar to the results in mouse peritoneal macrophages (Fig. 4C). Western blotting results showed that the protein level of KDM5B was promoted after EgCF and LPS treatment (Fig. 4E). Similarly, the co-stimulation of EgCF and LPS also markedly upregulated the mRNA and protein expression of KDM5B in THP-1 cells (Fig. 4F and G). Simultaneous stimulation of EgCF and LPS upregulated KDM5B protein expression at different time points in THP-1 cells compared with using the stimulation of LPS alone (Fig. 4H). RAW 264.7 cells were treated with cycloheximide (CHX), an inhibitor of protein synthesis, and we found that KDM5B protein expression was not downregulated when cells were co-stimulated with LPS and EgCF ( Fig. 4I), suggesting that EgCF promoted the stability of KDM5B protein. It has been reported that SUMOylation mediated by the activator STAT 4 protein suppressor (PIAS4) could stabilize KDM5B protein under hypoxia [28]. We then treated RAW 264.7 cells with a SUMOylation inhibitor (TAK-981). The results showed that the protein level of KDM5B was lower in the group of EgCF and LPS co-stimulation after TAK-981 treatment (Fig. 4J), suggesting that SUMOylation was involved in regulating KDM5B. Taken together, these results demonstrated that EgCF promoted KDM5B transcription and protein stability.

Fig. 4
figure 4

EgCF promoted transcription and protein stability of KDM5B. The expressions of histone demethylase gene (A) and differentially expressed KDM5B (B) were obtained from RNA-seq data of mouse peritoneal macrophages treated with LPS and EgCF. Mouse peritoneal macrophages were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h, and mRNA expression levels of KDM5B, KDM6B, and KDM7A (C) were detected by qRT-PCR. RAW 264.7 cells were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h, and mRNA levels of KDM5B were measured by qRT-PCR (D). Protein levels of KDM5B were measured by Western blotting (E). THP-1 cells were treated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h. mRNA levels of KDM5B were measured by qRT-PCR (F), and protein levels of KDM5B were measured by Western blotting (G). Protein levels of KDM5B were measured in THP-1 cells stimulated with EgCF (50% v/v) and LPS (1 μg/ml) for 15, 30, 60, 120, and 240 min (H). RAW 264.7 cells were treated with cycloheximide (CHX, 50 μg/ml) in combination with EgCF and LPS for 2, 4, and 6 h. Protein levels of KDM5B were measured by Western blotting (I). RAW 264.7 cells were treated with the SUMOylation inhibitor TAK-981 (0.5 µM and 1.0 µM) in combination with EgCF and LPS for 6 h, and KDM5B protein levels were measured by Western blotting (J). C, D, F Data are presented as the mean + SEM of three independent experiments and compared using the one-way ANOVA and Tukey test. Asterisks indicate a significant difference at *P < 0.05 and **P < 0.01

EgCF reduced H3K4me3 enrichment and promoted KDM5B enrichment at the promoters of inflammatory factors

To investigate whether KDM5B affected the anti-inflammatory effect of EgCF by regulating H3K4me3 modification, chromatin immunoprecipitation (ChIP) assays were performed in RAW 264.7 cells after stimulation of EgCF and LPS for 6 h. The results showed that compared with using LPS stimulation alone, simultaneous stimulation of EgCF and LPS reduced H3K4me3 enrichment at the TNF-α and IL-6 promoters (Fig. 5A–D). However, simultaneous stimulation of EgCF and LPS increased KDM5B enrichment at the TNF-α promoter in macrophages (Fig. 5E and F). These results demonstrated that EgCF inhibited H3K4me3 enrichment at the inflammatory cytokine promoter via KDM5B in macrophages.

Fig. 5
figure 5

EgCF caused lower H3K4me3 enrichment and higher KDM5B enrichment at the promoters of inflammatory factors. Chromatin immunoprecipitation was performed in RAW 264.7 cells treated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h. qRT-PCR and semi-qPCR were used to detect the enrichment levels of H3K4me3 and KDM5B at TNF-α and IL-6 promoters (A–F). Data are presented as the mean + SEM of three replicates, representing one of three independent experiments and compared using the one-way ANOVA and Tukey test. Asterisks indicate a significant difference at **P < 0.01 and ***P < 0.001

Inhibition of KDM5B attenuated the anti-inflammatory response of EgCF

To verify the mechanism that EgCF promoted KDM5B expression to inhibit the enrichment of H3K4me3 at the promoters of inflammatory cytokines, CPI-455 (a pan-KDM5 inhibitor) was used to pre-treat RAW 264.7 cells for 48 h. RAW 264.7 cells were stimulated with EgCF and LPS for 6 h after CPI-455 treatment (Fig. 6A). The qRT-PCR results demonstrated that the mRNA expression of IL-6 and TNF-α was recovered in RAW 264.7 cells treated with the KDM5 inhibitor CPI-455, indicating that the anti-inflammatory effect of EgCF was attenuated (Fig. 6B and C). CPI-455 was also used to inhibit KDM5B in THP-1 cells (Fig. 6D). Similarly, compared with simultaneous stimulation of EgCF and LPS, the mRNA expression of IL-6 and TNF-α was also significantly upregulated in THP-1 cells treated with CPI-455 (Fig. 6E and F). In summary, inhibiting KDM5B could at least partially abolish the anti-inflammatory effect of EgCF.

Fig. 6
figure 6

Inhibiting KDM5B attenuated the anti-inflammatory response of EgCF. RAW 264.7 cells were stimulated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h after pretreatment with CPI-455 (25 μM) for 48 h. Protein levels of KDM5B were measured by Western blotting (A). TNF-α and IL-6 mRNA expression levels (B, C) were measured by qRT-PCR. THP-1 cells pretreated with CPI-455 (25 μM) for 48 h were stimulated with EgCF (50% v/v) and LPS (1 μg/ml) for 6 h. Protein levels of KDM5B were measured by Western blotting (D). TNF-α and IL-6 mRNA expression levels (E, F) were measured by qRT-PCR. Working model for Echinococcus granulosus cyst fluid inhibiting the expression of inflammatory factors in macrophages (G). B, C, E, F Data are presented as the mean + SEM of three replicates, representing one of three independent experiments, and compared using the one-way ANOVA and Tukey test. Asterisks indicate a significant difference at *P < 0.05, **P < 0.01, and ***P < 0.001

Discussion

The ability of Echinococcus granulosus to survive in the host for a long period suggests that the parasite can effectively manage the host's immune response [29]. We have previously shown that EgCF can suppress inflammatory responses by regulating TRAF6 in macrophages [24]. Targeting epigenetic modifiers such as histone demethylases is an effective strategy in drug development. However, the efficacy of histone modification in inhibiting inflammatory response mediated by EgCF in macrophages remains to be explored. In this study, we demonstrated that EgCF promoted the transcription and protein stability of KDM5B, thereby inhibiting the enrichment of H3K4me3 at the promoters of inflammatory factors to suppress inflammatory response (Fig. 6G). Recent evidence also suggests that Leishmania parasites mediate epigenetic alterations in macrophages and suppress host defense, thereby facilitating parasite survival and disease progression [30]. Leishmania donovani could employ specific histone lysine methyltransferases/demethylases to redirect epigenetic programming of macrophage polarization for its successful establishment within the host [27].

This study used two macrophage models (mouse RAW 264.7 and human THP-1) to investigate the anti-inflammatory effect of EgCF. The results showed that EgCF treatment significantly downregulated the expression of pro-inflammatory cytokines IL-6 and TNF-α in macrophages, similar to the cyst fluid of E. multilocularis [31]. Surprisingly, TNF-α in RAW 264.7 cells and IL-6 in THP-1 cells were high in the control group (Fig. 2). Nevertheless, a similar phenomenon has also been observed in other studies [32, 33], and this would not affect the conclusion that the protein levels of TNF-α and IL-6 in cell supernatants were reduced after simultaneous stimulation of EgCF and LPS compared with using LPS stimulation alone.

In general, H3K4me3 is considered a hallmark of active transcription [18]. We found that EgCF suppressed the global levels of H3K4me3 in THP-1 and RAW 264.7 cells induced by LPS, indicating that H3K4me3 was involved in the EgCF regulation of anti-inflammatory responses in macrophages. Based on ChIP-seq data, we also found that H3K4me3 was highly enriched at the pro-inflammatory promoters in human and mouse macrophages, which supported the link between H3K4me3 modification and the anti-inflammatory effect of EgCF.

Subsequently, differentially expressed histone demethylases were analyzed to examine how EgCF downregulated the level of H3K4me3. The results showed that simultaneous stimulation of EgCF and LPS promoted the mRNA and protein expression of KDM5B compared with using LPS stimulation alone [27]. KDM5B is a JmjC-containing demethylase specific to H3K4me3 [34, 35]. In line with our finding, a recent report showed that KDM5B demethylated H3K4me3 at the promoters of IL-12, TNF-α, and arginase-1, thereby suppressing inflammatory responses in a Leishmania-infected macrophage model [27]. KDM5B could also inhibit the binding of NF-κB to IL-6 and IL-23a promoters by downregulating H3K4me3 levels in BMDMs [19]. We speculated that EgCF might suppress the H3K4me3 level by promoting the KDM5B level. To investigate whether KDM5B influenced the anti-inflammatory effect of EgCF by regulating H3K4me3 modification, we analyzed the enrichment of H3K4me3 and KDM5B at the promoters of inflammatory factors using ChIP assay. It was found that KDM5B was significantly enriched at the promoters of inflammatory factors after EgCF treatment, while H3K4me3 was the opposite. Previous studies have shown that CPI-455 could mediate pan-KDM5 inhibition and elevate global levels of H3K4me3 [36, 37]. CPI-455 treatment could at least partially abolish the anti-inflammatory effect of EgCF in RAW 264.7 and THP-1 cells. Further development of more potent and selective KDM5B inhibitors might achieve better effects [38].

However, EgCF is a mixture of carbohydrates, lipids, and proteins. For instance, antigen 5 (EgAg5), an abundant highly immunogenic component, is a thermolabile protein [39]; antigen B (EgAgB), one of the major molecules synthesized by the cyst, is a thermostable lipoprotein [40]. Our previous study showed that modification of carbohydrates, but not heat denaturation of proteins, abolished the ability of EgCF to inhibit LPS-induced inflammatory response [12]. Specific components of EgCF that affected the epigenetic control of host macrophages required further exploration. Investigating whether the activity of EgCF is thermostable could provide clues to understanding possible related molecules.

Overall, our findings demonstrated that E. granulosus promoted KDM5B expression and suppressed H3K4me3 enrichment at the promoter of inflammatory factors, inhibiting macrophage inflammatory responses. Targeting epigenetic modifiers such as histone demethylases may provide new ideas for the treatment of cystic echinococcosis.

Availability of data and materials

Data supporting the conclusions of this article are included within the article.

References

  1. Wen H, Vuitton L, Tuxun T, Li J, Vuitton DA, Zhang W, et al. Echinococcosis: advances in the 21st century. Clin Microbiol Rev. 2019. https://doi.org/10.1128/CMR.00075-18.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Vuitton DA, McManus DP, Rogan MT, Romig T, Gottstein B, Naidich A, et al. International consensus on terminology to be used in the field of echinococcoses. Parasite. 2020;27:41. https://doi.org/10.1051/parasite/2020024.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Zhang W, Li J, McManus DP. Concepts in immunology and diagnosis of hydatid disease. Clin Microbiol Rev. 2003;16:18–36. https://doi.org/10.1128/cmr.16.1.18-36.2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gottstein B, Soboslay P, Ortona E, Wang J, Siracusano A, Vuitton D. Immunology of alveolar and cystic echinococcosis (AE and CE). Adv Parasitol. 2017;96:1–54. https://doi.org/10.1016/bs.apar.2016.09.005.

    Article  CAS  PubMed  Google Scholar 

  5. Brunetti E, Kern P, Vuitton DA, Writing Panel for the W-I. Expert consensus for the diagnosis and treatment of cystic and alveolar echinococcosis in humans. Acta Trop. 2010;114:1–16. https://doi.org/10.1016/j.actatropica.2009.11.001.

    Article  PubMed  Google Scholar 

  6. Zhang W, Ross AG, McManus DP. Mechanisms of immunity in hydatid disease: implications for vaccine development. J Immunol. 2008;181:6679–85. https://doi.org/10.4049/jimmunol.181.10.6679.

    Article  CAS  PubMed  Google Scholar 

  7. Ahn CS, Han X, Bae YA, Ma X, Kim JT, Cai H, et al. Alteration of immunoproteome profile of Echinococcus granulosus hydatid fluid with progression of cystic echinococcosis. Parasit Vectors. 2015;8:10. https://doi.org/10.1186/s13071-014-0610-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yang J, Wu J, Fu Y, Yan L, Li Y, Guo X, et al. Identification of different extracellular vesicles in the hydatid fluid of echinococcus granulosus and immunomodulatory effects of 110 K EVs on sheep PBMCs. Front Immunol. 2021;12:602717. https://doi.org/10.3389/fimmu.2021.602717.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cui SJ, Xu LL, Zhang T, Xu M, Yao J, Fang CY, et al. Proteomic characterization of larval and adult developmental stages in Echinococcus granulosus reveals novel insight into host-parasite interactions. J Proteom. 2013;84:158–75. https://doi.org/10.1016/j.jprot.2013.04.013.

    Article  CAS  Google Scholar 

  10. Wang Y, Wang Q, Lv S, Zhang S. Different protein of Echinococcus granulosus stimulates dendritic induced immune response. Parasitology. 2015;142:879–89. https://doi.org/10.1017/s0031182014002005.

    Article  CAS  PubMed  Google Scholar 

  11. Yin S, Chen X, Zhang J, Xu F, Hou J, Wu X, et al. Initial studies on the role of hydatid fluid in the immune evasion strategies of Echinococcus granulosus. Pak J Zool. 2014;46:1711–8.

    CAS  Google Scholar 

  12. Hou J, Li L, Dong D, Wang L, Wang X, Yang K, et al. Glycomolecules in Echinococcus granulosus cyst fluid inhibit TLR4-mediated inflammatory responses via c-Raf. Cell Mol Immunol. 2020;17:423–5. https://doi.org/10.1038/s41423-019-0314-1.

    Article  CAS  PubMed  Google Scholar 

  13. Muraille E, Leo O, Moser M. TH1/TH2 paradigm extended: macrophage polarization as an unappreciated pathogen-driven escape mechanism? Front Immunol. 2015;5:603. https://doi.org/10.3389/fimmu.2014.00603.

    Article  CAS  Google Scholar 

  14. Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. Eur J Pharmacol. 2020;877:173090. https://doi.org/10.1016/j.ejphar.2020.173090.

    Article  CAS  PubMed  Google Scholar 

  15. Kang MK, Mehrazarin S, Park NH, Wang CY. Epigenetic gene regulation by histone demethylases: emerging role in oncogenesis and inflammation. Oral Dis. 2017;23:709–20. https://doi.org/10.1111/odi.12569.

    Article  CAS  PubMed  Google Scholar 

  16. Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150:12–27. https://doi.org/10.1016/j.cell.2012.06.013.

    Article  CAS  PubMed  Google Scholar 

  17. Kristensen LH, Nielsen AL, Helgstrand C, Lees M, Cloos P, Kastrup JS, et al. Studies of H3K4me3 demethylation by KDM5B/Jarid1B/PLU1 reveals strong substrate recognition in vitro and identifies 2,4-pyridine-dicarboxylic acid as an in vitro and in cell inhibitor. FEBS J. 2012;279:1905–14. https://doi.org/10.1111/j.1742-4658.2012.08567.x.

    Article  CAS  PubMed  Google Scholar 

  18. Kooistra SM, Helin K. Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol. 2012;13:297–311. https://doi.org/10.1038/nrm3327.

    Article  CAS  PubMed  Google Scholar 

  19. Zhao Z, Su Z, Liang P, Liu D, Yang S, Wu Y, et al. USP38 couples histone ubiquitination and methylation via KDM5B to resolve inflammation. Adv Sci. 2020;7:2002680. https://doi.org/10.1002/advs.202002680.

    Article  CAS  Google Scholar 

  20. Chang YY, Lu CW, Jean WH, Shieh JS, Lin TY. Phorbol myristate acetate induces differentiation of THP-1 cells in a nitric oxide-dependent manner. Nitric Oxide. 2021. https://doi.org/10.1016/j.niox.2021.02.002.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Li P, Hao Z, Wu J, Ma C, Xu Y, Li J, et al. Comparative proteomic analysis of polarized human THP-1 and mouse RAW264.7 macrophages. Front Immunol. 2021;12:700009. https://doi.org/10.3389/fimmu.2021.700009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Liu T, Huang T, Li J, Li A, Li C, Huang X, et al. Optimization of differentiation and transcriptomic profile of THP-1 cells into macrophage by PMA. PLoS One. 2023;18:e0286056. https://doi.org/10.1371/journal.pone.0286056.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ozleyen A, Yilmaz YB, Tumer TB. Dataset on the differentiation of THP-1 monocytes to LPS inducible adherent macrophages and their capacity for NO/iNOS signaling. Data Brief. 2021;35:106786. https://doi.org/10.1016/j.dib.2021.106786.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lin K, Zhou D, Li M, Meng J, He F, Yang X, et al. Echinococcus granulosus cyst fluid suppresses inflammatory responses by inhibiting TRAF6 signalling in macrophages. Parasitology. 2021;148:887–94. https://doi.org/10.1017/s0031182021000548.

    Article  CAS  PubMed  Google Scholar 

  25. Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell. 2012;48:491–507. https://doi.org/10.1016/j.molcel.2012.11.006.

    Article  CAS  PubMed  Google Scholar 

  26. Fanucchi S, Domínguez-Andrés J, Joosten LAB, Netea MG, Mhlanga MM. The intersection of epigenetics and metabolism in trained immunity. Immunity. 2021;54:32–43. https://doi.org/10.1016/j.immuni.2020.10.011.

    Article  CAS  PubMed  Google Scholar 

  27. Parmar N, Chandrakar P, Kar S. Leishmania donovani subverts host immune response by epigenetic reprogramming of macrophage M(Lipopolysaccharides + IFN-γ)/M(IL-10) polarization. J Immunol. 2020;204:2762–78. https://doi.org/10.4049/jimmunol.1900251.

    Article  CAS  PubMed  Google Scholar 

  28. Zhou B, Zhu Y, Xu W, Zhou Q, Tan L, Zhu L, et al. Hypoxia Stimulates SUMOylation-Dependent Stabilization of KDM5B. Frontiers Cell Dev Biol. 2021;9:741736. https://doi.org/10.3389/fcell.2021.741736.

    Article  Google Scholar 

  29. Diaz A. Immunology of cystic echinococcosis (hydatid disease). Br Med Bull. 2017;124:121–33. https://doi.org/10.1093/bmb/ldx033.

    Article  CAS  PubMed  Google Scholar 

  30. Marr AK, MacIsaac JL, Jiang R, Airo AM, Kobor MS, McMaster WR. Leishmania donovani infection causes distinct epigenetic DNA methylation changes in host macrophages. PLoS Pathog. 2014;10:e1004419. https://doi.org/10.1371/journal.ppat.1004419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hübner MP, Manfras BJ, Margos MC, Eiffler D, Hoffmann WH, Schulz-Key H, et al. Echinococcus multilocularis metacestodes modulate cellular cytokine and chemokine release by peripheral blood mononuclear cells in alveolar echinococcosis patients. Clin Exp Immunol. 2006;145:243–51. https://doi.org/10.1111/j.1365-2249.2006.03142.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chen B, Li H, Ou G, Ren L, Yang X, Zeng M. Curcumin attenuates MSU crystal-induced inflammation by inhibiting the degradation of IκBα and blocking mitochondrial damage. Arthritis Res Ther. 2019;21:193. https://doi.org/10.1186/s13075-019-1974-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang Y, Ding J, Wang Y, Feng X, Du M, Liu P. Guanxinkang decoction attenuates the inflammation in atherosclerosis by regulating efferocytosis and MAPKs signaling pathway in LDLR(-/-) mice and RAW264.7 cells. Front Pharmacol. 2021;12:731769. https://doi.org/10.3389/fphar.2021.731769.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fu YD, Huang MJ, Guo JW, You YZ, Liu HM, Huang LH, et al. Targeting histone demethylase KDM5B for cancer treatment. European J Med Chem. 2020;208:112760. https://doi.org/10.1016/j.ejmech.2020.112760.

    Article  CAS  Google Scholar 

  35. Zhao B, Liang Q, Ren H, Zhang X, Wu Y, Zhang K, et al. Discovery of pyrazole derivatives as cellular active inhibitors of histone lysine specific demethylase 5B (KDM5B/JARID1B). Eur J Med Chem. 2020;192:112161. https://doi.org/10.1016/j.ejmech.2020.112161.

    Article  CAS  PubMed  Google Scholar 

  36. Vinogradova M, Gehling VS, Gustafson A, Arora S, Tindell CA, Wilson C, et al. An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells. Nat Chem Biol. 2016;12:531–8. https://doi.org/10.1038/nchembio.2085.

    Article  CAS  PubMed  Google Scholar 

  37. Gaillard S, Charasson V, Ribeyre C, Salifou K, Pillaire MJ, Hoffmann JS, et al. KDM5A and KDM5B histone-demethylases contribute to HU-induced replication stress response and tolerance. Biol Open. 2021. https://doi.org/10.1242/bio.057729.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Zheng YC, Chang J, Wang LC, Ren HM, Pang JR, Liu HM. Lysine demethylase 5B (KDM5B): a potential anti-cancer drug target. Eur J Med Chem. 2019;161:131–40. https://doi.org/10.1016/j.ejmech.2018.10.040.

    Article  CAS  PubMed  Google Scholar 

  39. Manzano-Roman R, Sanchez-Ovejero C, Hernandez-Gonzalez A, Casulli A, Siles-Lucas M. Serological diagnosis and follow-up of human cystic echinococcosis: a new hope for the future? Biomed Res Int. 2015;2015:428205. https://doi.org/10.1155/2015/428205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Silva-Alvarez V, Folle AM, Ramos AL, Zamarreno F, Costabel MD, Garcia-Zepeda E, et al. Echinococcus granulosus antigen B: a hydrophobic ligand binding protein at the host-parasite interface. Prostaglandins Leukot Essent Fatty Acids. 2015;93:17–23. https://doi.org/10.1016/j.plefa.2014.09.008.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (82060579 and 82060297), the Bingtuan Science and Technology Program (2022ZD046 and 2021BB006), the Science and Technology Program of Shihezi University (GJHZ202202 and CXPY202003), and the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2020-PT330-003).

Author information

Authors and Affiliations

Authors

Contributions

JH, LHW, and XPW conceived and designed the study. XPW, RLL, CXF, CY, DD, XWW, and XLC conducted data gathering. XPW performed statistical analyses. XPW, LHW, and JH wrote the manuscript.

Corresponding authors

Correspondence to Lianghai Wang or Jun Hou.

Ethics declarations

Ethics approval and consent to participate

All animal procedures were approved by the Animal Experimental Ethical Inspection of First Affiliated Hospital, Shihezi University School of Medicine (A2020-045-01), and performed according to the Guide for the Care and Use of Laboratory Animals.

Consent for publication

Not applicable.

Competing interests

The authors declare there are no competing interests.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1: Figure S1.

Cell viability after treatment for all cell sources was measured at the indicated time points using the Cell Counting Kit 8 assay (CCK8; Dojindo). ns, not significant.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, X., Lin, R., Fu, C. et al. Echinococcus granulosus cyst fluid inhibits inflammatory responses through inducing histone demethylase KDM5B in macrophages. Parasites Vectors 16, 321 (2023). https://doi.org/10.1186/s13071-023-05948-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13071-023-05948-1

Keywords