Skip to main content
Advertisement
  • Loading metrics

ASPSCR-1 and Sirt-5 alleviate Clonorchis liver fluke rCsNOSIP-induced oxidative stress, proliferation, and migration in cholangiocarcinoma cells

  • Meng Bian ,

    Contributed equally to this work with: Meng Bian, Shan Li

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Writing – original draft

    Affiliations Department of Clinical laboratory, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, People’s Republic of China, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, Henan, People’s Republic of China

  • Shan Li ,

    Contributed equally to this work with: Meng Bian, Shan Li

    Roles Data curation, Funding acquisition, Investigation, Methodology, Writing – review & editing

    Affiliation Department of Pathology and Pathophysiology, School of Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, People’s Republic of China

  • Hanzong Zhou,

    Roles Methodology

    Affiliation The fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People’s Hospital), Zhengzhou, Henan, People’s Republic of China

  • Lijun Bi,

    Roles Methodology, Resources

    Affiliation Institute of Biophysics, Chinese Academy of Scineces, Beijing, People’s Republic of China

  • Yong Shen,

    Roles Methodology, Software, Visualization

    Affiliations Department of Clinical laboratory, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, People’s Republic of China, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, Henan, People’s Republic of China

  • Chen Tingjin,

    Roles Funding acquisition, Methodology

    Affiliations Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-sen University, Guangzhou, People’s Republic of China, Department of Pathology, Duke University School of Medicine, Durham, North Carolina, United States of America

  • Xinbing Yu ,

    Roles Funding acquisition, Project administration, Resources, Supervision, Validation

    yuxb@mail.sysu.edu.cn(XY); huang66@mail.sysu.edu.cn(YH); zlyyxuqingxia0575@zzu.edu.cn (QX)

    Affiliation Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-sen University, Guangzhou, People’s Republic of China

  • Yan Huang ,

    Roles Supervision

    yuxb@mail.sysu.edu.cn(XY); huang66@mail.sysu.edu.cn(YH); zlyyxuqingxia0575@zzu.edu.cn (QX)

    Affiliation Key Laboratory for Tropical Diseases Control, Ministry of Education, Sun Yat-sen University, Guangzhou, People’s Republic of China

  • Qingxia Xu

    Roles Conceptualization, Supervision

    yuxb@mail.sysu.edu.cn(XY); huang66@mail.sysu.edu.cn(YH); zlyyxuqingxia0575@zzu.edu.cn (QX)

    Affiliations Department of Clinical laboratory, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, People’s Republic of China, Zhengzhou Key Laboratory of Digestive System Tumor Marker Diagnosis, Zhengzhou, Henan, People’s Republic of China

Abstract

Background

Clonorchiasis, caused by the infection of Clonorchis sinensis (C. sinensis), is a kind of neglected tropical disease, but it is highly related to cholangiocarcinoma. It has been well known that NO from chronic inflammation responses are thought to be a major component of the damage and ultimate carcinogenesis ESPs such as nitric oxide synthase interacting protein (NOSIP) are thought to enhance the damage. The objective of this study was to identify the protein candidates interact with recombinant CsNOSIP (rCsNOSIP) and explore their role involved in CCA development or progression.

Methods

We applied HuProt microarray containing 21,000 probe sets for a systematic identification of rCsNOSIP-binding proteins and grouped binding hits by gene function. Pull-down assays were used to confirm the interaction of rCsNOSIP with alveolar soft part sarcoma (ASPSCR-1) and sirtuins 5 (Sirt-5). ASPSCR-1/Sirt-5 over-expression and siRNA knockdown experiments were employed for obtain of ASPSCR-1/Sirt-5 high or low expression (ASP-oe/Sirt5-oe or ASP-si/Sirt5-si) cholangiocarcinoma cell line (CCLP-1) cells. Nitric oxide (NO) and reactive oxygen species assay (ROS) as well as cell proliferation and wound-healing assays were performed to observe the effect of rCsNOSIP on ASP-oe/Sirt5-oe or ASP-si/Sirt5-si CCLP-1 cells.

Results

Seventy candidate proteins protein "hits" were detected as rCsNOSIP-binding proteins by HuProt microarray and bioinformatics analysis. Pull down assay showed that ASPSCR-1 and Sirt-5 could interact with rCsNOSIP. In addition, endotoxin-free-rCsNOSIP could increase the production of NO and ROS and promote the migration of CCLP-1 cells, while its effect on enhancing cell proliferation was not significant. Furthermore, ROS/NO production, proliferation, or migration were increased in ASP-si or Sirt5-si CCLP-1 cells but decreased in Asp-oe or Sirt5-oe CCLP-1 cells when stimulated with rCsNOSIP.

Conclusions

Our findings suggest that CsNOSIP as a component of CsESPs might promote the development and invasion of CCA and Sirt5/ ASPSCR1 as host molecules might play a novel protective role against adverse stimulus during C. sinensis infection. This work supports the idea that CsESPs induce the occurrence and progression of CCA through ROS/RNS-induced oxidative and nitrative DNA damage.

Author summary

Clonorchis sinensis (C. sinensis) is prevalent in China, Korea, and Vietnam, and 15–20 million people are estimated to be infected by this fluke, creating a socio-economic burden in epidemic regions. It is generally believed that excretory-secretory products from C. sinensis (CsESPs) can directly interact with the biliary epithelium and induce host inflammatory response. Our previous studies have manifested that CsNOSIP as a component of CsESPs can trigger nitrative and oxidative stress by enhancing the release of NO and reactive oxygen species (ROS) in macrophage. In the current study, we identified rCsNOSIP-binding proteins by HuProt microarray. In addition, we demonstrated that ASPSCR-1 and Sirt-5 could interact with rCsNOSIP by pull down assay. Moreover, rCsNOSIP could increase the production of NO and ROS and promote the migration of human CCLP-1 cells by using NO assay, detection of ROS assay as well as wound-healing assays. Our study provided new information about rCsNOSIP in promoting CCA invasion and metastasis as well as a novel protective role of SIRT5/ ASPSCR1 against adverse stimulus and support a basis for further discovery of drug targets for prevention and control of clonorchiasis.

Introduction

Clonorchis sinensis (C. sinensis), the causative agents of clonorchiasis, is an important food-borne parasite endemic throughout Southeast Asia [1,2,3]. Clonorchiasis is prevalent in China, Korea, and Vietnam, and 15–20 million people are estimated to be infected by this fluke, creating a socio-economic burden in epidemic regions [4]. Because of the high correlation between clonorchiasis and cholangiocarcinoma (CCA), C. sinensis was reclassified as a group -I biocarcinogen for CCA by the International Agency for Research on Cancer (IARC) in 2009 [47]. This biocarcinogen has been included in control programs of neglected 57 tropical diseases by WHO [5]. It is generally believed that excretory-secretory products from C. sinensis (CsESPs) can directly interact with the biliary epithelium and induce host inflammatory response [8,9]. During chronic inflammation, excessive nitrogen and oxygen radicals especially nitric oxide (NO) generated by inflammatory and epithelial cells may exert cytotoxic and mutagenic effects through inducing nitrative and oxidative DNA damage [10,11]. NO is accordingly considered as a carcinogen that promotes the occurrence and progression of CCA via chronic inflammation [12]. Certain ESPs, such as NOSIP, are thought to cause further damage through excess NO production. Therefore, understanding the exact function of key molecules in CsESPs that are related to NO production or its reactive intermediates will facilitate illumination of CCA pathogenesis during C. sinensis infection.

NO is produced from a reaction catalyzed by three major isoforms of nitric oxide synthase (NOS) including neuronal (nNOS/NOS1), inducible (iNOS/NOS2), and endothelial (eNOS/NOS3) [13,14]. As an eNOS interaction protein, NO synthase interacting protein (NOSIP) possibly affects eNOS activity and regulates NO production [15,16]. Our previous studies have manifested that C. sinensis NOSIP (CsNOSIP) as a component of CsESPs can trigger nitrative and oxidative stress by enhancing the release of NO and reactive oxygen species (ROS) in macrophage. However, CsNOSIP-mediated molecular mechanism in CCA pathologic process remains as yet unclear. In the present study, HuProt microarray was employed for exploring protein binding partners to recombinant CsNOSIP protein (rCsNOSIP). Potential positive proteins interaction with rCsNOSIP was screened through bioinformatics analysis. The interaction of rCsNOSIP with ASPSCR-1 and Sirt-5, two screened proteins closely related to tumor progression, was confirmed by GST-pull down assay. The influence of rCsNOSIP on proliferation, migration, and NO/ROS production in CCLP1 cells line when over expression and low expression of ASPSCR-1 and Sirt-5 was investigated. Our study will provide new information about parasite-host interaction in CCA pathogenesis and support a basis for further discovery of drug targets for prevention and control of clonorchiasis.

Materials and methods

Protein expression and purification

Recombinant CsNOSIP protein (rCsNOSIP) was produced as previous method with slight modification [17]. Briefly, pET-28a-(+)-CsNOSIP was constructed and transformed into E. coli (DE3) (Promega). Induced by 0.2 mM isopropyl-β-D-thiogalactopyranoside (IPTG), the protein was expressed at 30°C for 12 h in Luria-Bertani medium and purified by Ni-nitrilotriacetic acid (NTA) chromatography. The purified rCsNOSIP was analyzed by 12% SDS-PAGE stained with Coomassie brilliant blue G-250 and the concentration of recombinant soluble protein concentration was detected by the BCA protein assay kit (Novagen, USA). The protein was treated by AffinityPa Detoxi-Ge Endotoxin Removing Gel (Thermo, USA) and Limulus test was used to make sure that the endotoxin had been removed. Then, the concentration of rCsNOSIP was determined by BCA protein after endotoxin cleanup.

Detection of target protein by HuProt microarray assay

The HuProt microarray assay procedures were described previously [18]. Briefly, microarrays were incubated with blocking-buffer (3% BSA in 1× PBS, pH 7.4) for 3 h, shaking at 4°C. Purified CsNOSIP was labeled with Cy3 (GE Healthcare, Little Chalfont, UK) and diluted with binding-buffer (1% BSA in 1×PBS, pH 7.4). The diluted CsNOSIP-Cy3 (15 μg) incubation was performed at 4°C overnight. After washing 3 times with PBST, then twice with ultra-pure water, array was scanned in a LuxScan 10K-A (CapitalBio Corporation, Beijing, China). The scanned fluorescence data was analyzed with GenePix Pro 6.0 software (Molecular Devices). To generate candidate lists, median normalization was performed for each microarray, and the cutoff value was defined as mean+2*SD (95%). Then, the normalized SNRs for CsNOSIP and BSA were set as SNR (+) and SNR (−), and Fold change was defined as SNR (+)/SNR (−). The candidate targets were defined as proteins with SNR (+) ≥5.35 (95%CI) and Fold change ≥1.2 [19].

Bioinformatics analysis

A total of 70 candidate targets were listed for subsequent bioinformatics analysis. GO and the KEGG were used for protein functional annotation. Functional enrichment on the proteins was achieved by the R package cluster Profiler (v4.0.5). KEGG pathway and GO terms with P values < 0.05, were considered significantly enriched based on the hyper geometric distribution.

Cell culture

Human cholangiocarcinoma cell line (CCLP-1) was gifted from Professor Bi’s lab at the Institute of Biophysics, Chinese Academy of Sciences.CCLP-1 cell lines was cultured in DMEM including 1% penicillin, 1% streptomycin and 10% FBS at 37°C under 5% CO2.

Measurement of mRNA expression by quantitative real-time PCR

The extraction of total RNA, complementary DNA synthesis and PCR amplification were carried out as described previously [20]. Briefly, total RNAs from control group (CCLP1 cells without CsNOSIP) and CsNOSIP+ group (CCLP1 cells with 5 μg/ml CsNOSIP for 24 h) were extracted in Trizol reagent (Invitrogen) according to the manufacturer’s instructions. RNA concentrations were detected by nucleic acid/protein analyzer (Beckman Coulter, USA). The preparation of the first-strand cDNA was carried out using RT-PCR Kit (TIANGEN, Beijing, China) with the same quantity of total RNA as the template (1 μg).

Quantitative real-time PCR (qRT-PCR) was employed to study the relative expression of ASPSCR1 and Sirt 5 in CCLP1 induced by CsNOSIP. All PCR was carried out using a Bio-Rad CFX96 real-time PCR system (Bio-Rad Laboratories). The forward and reverse primers for ASPSCR1 were 5’-TGACGCGCCACTCCAAAAACT-3’ and 5’-CCA AATACTCTAAGACCGCCGC-3’, primers for Sirt 5 were 5’-CCCAGAACATCGATGAGC-3’ and 5’-GCCACAACTCCACAAGAGG-3’, respectively [21]. The housekeeping β-actin gene used as a reference for mRNA quantification. The forward and reverse primers for β-actin were 5’- CTTCCTTCCTGGGCATGG-3’ and 5’-GCCGCCAGACAGCACTGT-3’. Quantitative RT-PCR reactions based on SYBR-Green I fluorescence (TIANGEN, Beijing, China) were performed by using Bio-Rad iQ5 instrument (Bio-Rad, USA). The PCR amplification program was 95°C for 30 s, 40 cycles of 95°C for 5 s, and 60°C for 20 s. The iQ5 software was used to analyze the relative quantification according to the 2−ΔΔCt method [22].

Pull-down assays

To investigate the interaction of rCsNOSIP with ASPSCR-1 and Sirt-5, GST tagged rCsNOSIP was prepared for the GST-pull down assay. Expression vectors pET-28a-Flag-ASPSCR-1, pET-28a-Flag-Sirt 5 and pET-28a-His-GST-CsNOSIP were generated by inserting the corresponding tag fusion fragment into pET-28a. Gene fragment synthesis and vector construction were performed by GenScript Biotech Corporation. His-GST-CsNOSIP was purified as described above. 50 μg purified GST-rCsNOSIP was immobilized by GST affinity beads (Roche Applied Science, Indianapolis, IN) and incubated with cells lysate containing ASPSCR-1(or Sirt 5). After shaking at room temperature for 2 h, the beads were pelleted and washed twice with PBS buffer. The bound protein was eluted with 10 mM GSH buffer then resolved by SDS-PAGE and detected by rabbit anti-GST- antibody or mouse anti-Flag. Cell lysate without ASPSCR-1(or Sirt 5) and purified GST protein were used as controls for nonspecific interactions between GST-rCsNOSIP and ASPSCR-1(or Sirt 5).

ASPSCR-1/Sirt 5 over-expression and siRNA knockdown experiments

To investigate whether ASPSCR1 and Sirt 5 protein were involved in the stress response in CCLP 1 cell lines with 5 μg /mL purified CsNOSIP, cell lines of different expression levels of ASPSCR1 and Sirt 5 were generated. Plasmids (pcDNA3.1-ASPSCR-1 and pcDNA3.1-Sirt 5) were from GenScript Biotech Corporation. CCLP-1 cells were transfected with above plasmids using Polyjet reagent (SignaGen) according to the manufacturer’s protocol. After 48 or 72 h, the cells were collected and lysed in lysis buffer, then detected by western blot with anti-ASPSCR1 mouse antibody or anti-Sirt 5 mouse antibody. Alternatively, cells were stimulated by CsNOSIP. For low expression of ASPSCR-1/Sirt 5 in CCLP-1 cell lines, knockdown experiments were performed using gene-specifific siRNAs (siRNA 1 group and siRNA 2 group). siRNAs were chemically synthesised by RIBOBIO (RIBOBIO, Guangzhou, China). Sequences were as follows: ASPSCR1 siRNA 1, 5′-CAAUGCCAAGCUGGAGAUGTT-3′; ASPSCR1 siRNA 2, 5′-CAUCUCCAGCUUGGAUUGTT-3′; Sirt 5 siRNA 1,5′-GGAGAUCCAUGGUAGCUUATT-3′; Sirt 5 siRNA 2,5′- UAAGCUACCAUGGAUCUCCTT-3′. siRNA transfections were performed using Lipofectamine RNAiMax transfection reagent (Life Technologies), according to the manufacturer’s specifications. Whole cell lysate was prepared at 72 h after transfection for western blot analysis. Alternatively, cells were collected for quantitative real-time PCR(qRT-PCR)analysis.

Western blot

Western blot was carried out as described by Xu et al [3]. Briefly, Cells were lysed in RIPA buffer and then loaded onto 12% SDS-polyacrylamide gels for electrophoresis. Proteins on gels were transferred onto a nitrocellulose membrane, and was assayed by Ponceau S staining. Stained membrane was washed with TBST solution (20 mM Tris-HCl, 0.15 M NaCl, 0.05% Tween-20, pH 7.5) immediately and blocked in 5% (w/v) non-fat milk in TBST. The membrane was incubated with corresponding primary antibody (anti-GST- antibody from CoWin Biotech, anti-Flag from YEASEN, anti-ASPSCR1 antibody from Sino Biological, anti-Sirt 5 from Proteintech, anti-β-actin from abcam) at room temperature for 2 h. After 3 times of TBST washing, membrane was incubated with a secondary horseradish peroxidase linked anti-rabbit IgG or anti mouse IgG (1:10000, GE Healthcare). Chemiluminescent substrate (enhanced chemiluminescence -plus substrate, GE Healthcare) was used for detection according to the manufacturer’s instructions.

Quantitative real-time PCR (qRT-PCR)

RNA was extracted from harvest cells using a RNA preppure Kit (TIANGEN, Beijing, China) according to the manufacturer’s instructions. The first-strand cDNA was synthesized from the total RNA using RNA First-Strand cDNA Kit (TIANGEN, Beijing, China). Subsequently, qRT-PCR was conducted using gene-specific primers to determine the expression levels of mRNAs. Following initial denaturation at 95°C for 30 s, 40 cycles of PCR amplification were performed at 95°Cfor 5 s and 60°C for 30 s, with a dissociation curve at the end of the amplification reaction. The qRT-PCR data were calculated by the 2-△△CT method [4] while the expression level of β- actin was used for normalization. The results of qRT-PCR were analyzed using GraphPad Prism (version 7.0). Differences between groups were statistically analyzed using analysis of variance (ANOVA). A P value < 0.05 was considered statistically significant.

NO and ROS measurement

CCLP-1 cells were transfected with pcDNA3.1-ASPSCR-1 or pcDNA3.1-Sirt 5 for 48 h and then stimulated by adding 5 μg/mL purified CsNOSIP for 24 h. Cultured cells and medium were collected separately. NO concentrations were quantified using a nitric oxide assay kit (Beyotime Biotechnology, Shanghai, China). Briefly, cell culture supernatants and standards of NaNO2 (0,1, 2, 5, 10, 20, 40, 60, 100 μM) were added into individual wells of a 96-well plate. Fifty microliters of Griess reagent I and Griess reagent II were then dripped into each well successively. After 3 min incubation, the absorbance was measured at 540 nm utilizing a microplate reader, and the concentration of NO was determined.

Reactive Oxygen Species Assay Kit (2,7-Dichlorodi-hydrofluorescein diacetate, DCFH-DA; Applygen Technologies, Beijing, China) were used to measure the quantity of reactive oxygen species (ROS) in the cholangiocarcinoma cell (CCLP-1). The dye loading was performed by incubating the cells with 10 μM DCFH-DA at 37°C for 60 min. The production of ROS was examined using a Flow cytometer (BD FACS Canto, BD Biosciences USA) by measuring the fluorescence intensity of DCF at an excitation wavelength of 488 nm and an emission wavelength of 525 nm.

Cell proliferation and Wound-healing assays

The proliferation activity of cells was detected by CCK-8 method. Cells were transfected for 48 h, then were seeded in 96-well plates at a density of 5000 cells per well. After cells attachment, the fresh medium with or without 5 μg/mL purified CsNOSIP protein was added into cells, incubating for 24 h. The proliferation activity of cells was detected by Cell Counting Kit-8 (Beyotime, China), according manufacturer’s instructions. In briefly, the medium was removed and replaced with fresh medium containing 10% CCK-8 solution. Cells were incubated for 3 h at 37°C. Then the 450 nm wave-length absorption values were measured using a microplate reader (Multiskan FC, Thermo Scientific, USA). The experiment was performed three times with five replicates for each sample.

Cell migration was analyzed by the wound healing assay [23]. In brief, cells were transfected for 48 h and several wound lines were scratched vertically to the bottom with a 200 ml pipette tip. After being washed with PBS three times, cells were incubated in growth medium containing 5 μg/mL CsNOSIP protein without serum. The wound areas were determined at 24 h with microscope (Nikon, Japan). According to the collected image data, the data analysis was performed by ImageJ (v1.8.0). The calculation formula of the result was descripted by the calculation formula percentage of cell migration = (initial scratch area- final scratch area) /initial scratch area × 100%.

Statistical analysis

Statistical analysis was performed using GraphPad Prism software. All data were presented as mean ± SD. For comparison of groups, one-way ANOVA and Student’s t test were used, and one-way ANOVA using SPSS software for windows (version 17.0; SPSS, Inc., IL). All graphs were performed using GraphPad Prism software. P < 0.05 was taken to be significant.

Results

Expression, endotoxin removal and purification of rCsNOSIP

Recombinant CsNOSIP was expressed in E. coli (DE3) and purified using Ni2+-NTA chromatogr-aphy (S1 Fig). The purified rCsNOSIP of approximately 36 kDa were identified by SDS-PAGE (Fig 1A). The concentration of the recombinant His tag-CsNOSIP fusion protein was about 1.25 mg/ml, while the concentration of endotoxin-free rCsNOSIP was about 645 μg/ml. We applied a HuProt human proteome microarray containing over 21,000 human proteins for a systematic identification of CsNOSIP-binding. Seventy candidate proteins were positived as rCsNOSIP-binding proteins by HuProt microarray and bioinformatics analysis (S1 Table).

thumbnail
Fig 1. SDS-PAGE of purified CsNOSIP and functional enrichment of biological processes of proteins identified by protein array.

(A) SDS-PAGE of purified protein. Lane M, protein marker; lane 1, purified CsNOSIP on Coomassie Brilliant Blue (CBB) stained SDS-PAGE gel. (B) Enrichment of biological processes in terms of gene ontology (GO) categories. (C) Enrichment of Kyoto Encyclopedia of Genes and Genomes (KEGG) categories.

https://doi.org/10.1371/journal.pntd.0011727.g001

Functional enrichment analysis

Go enrichment indicated that these candidate proteins were associated with biological process including protein serine/threonine/tyrosine kinase activity, and regulation of oxidative stress-induced intrinsic apoptotic signaling pathway (Fig 1C). In KEGG pathway, we found that the candidate proteins were associated with various tumors progression, such as thyroid cancer, endometrial cancer, especially highly correlated with cGMP-PKG (Fig 1B).

Transcriptional levels of ASPSCR1 and Sirt 5 in CCLP1 cells stimulated with CsNOSIP by QPCR

The mRNA expression patterns of ASPSCR1 and Sirt 5 in CCLP1 cells were measured by qRT-PCR. As shown in Fig 2, compared with control group, ASPSCR1 and Sirt 5 mRNA expressions were significantly increased (p < 0.05).

thumbnail
Fig 2. mRNA expression of ASPSCR1 and Sirt5 in CCLP-1 cells by qRT-PCR.

Groups: Control: CCLP-1 cells without CsNOSIP; CsNOSIP: CCLP-1 cells with 5 μg/ml CsNOSIP. The transcription levels of ASPSCR1 and Sirt5 are analyzed by means of the 2−ΔΔCt ratio, with human β-actin employing as the transcriptional control. Data are represented as mean ± SD (*p<0.05). Cultures were set up in triplicate and data were displayed as mean ± SD. Statistical significance was analyzed by the Student’s t test (*p < 0.05).

https://doi.org/10.1371/journal.pntd.0011727.g002

CsNOSIP interacts with ASPSCR-1 and Sirt-5

ASPSCR-1 and Sirt-5, which may be closely related to tumor migration had been identified, were selected according to the bioinformatics analysis. Physical interaction between GST-rCsNOSIP and ASPSCR-1/Sirt-5 was confirmed by GST-pull down assay. As shown in Fig 3A, in the test between GST-rCsNOSIP and ASPSCR-1, the two proteins were detected simultaneously. In the test of GST-rCsNOSIP without ASPSCR-1, only rCsNOSIP was detected. In the pull-down test using purified GST protein and ASPSCR-1, the two proteins were not detected in the elution from GST protein coupled affinity beads. In the GST-pull down assay of rCsNOSIP and Sirt-5, the same results were detected (Fig 3B). These results suggested that ASPSCR-1 and Sirt-5 interacted with rCsNOSIP.

thumbnail
Fig 3. CsNOSIP interacts with ASPSCR-1 and Sirt-5.

Western-blot of typical protein-containing fractions collected after pull-down assay of GST-CsNOSIP complexes. (A) GST-pull down assay of CsNOSIP and ASPSCR-1. (B) GST-pull down assay of CsNOSIP and Sirt-5.

https://doi.org/10.1371/journal.pntd.0011727.g003

Expression analysis of ASPSCR-1 and Sirt 5

The over-expression of ASPSCR-1/Sirt 5 was determined by western blot. As shown in S3A Fig, the expression of ASPSCR-1 and Sirt 5 were all increased by transiently transfected an over-expression vector in CCLP-1 cell lines. After 48 hours of transfection, ASPSCR-1 expression level was obvious higher than untreated cells. Compared with ASPSCR-1, over-expression of Sirt 5 needs more time (72 h) after cell transfection. Transfection of cells with specific siRNAs resulted in a large decrease in ASPSCR-1/Sirt 5 expression levels as judged by western-blot (S3B Fig) and qRT-PCR (S3C Fig). In the ASPSCR knockdown experiments, results of western-blot and qRT-PCR were consistent, ASPSCR-1 siRNAs reduced the gene expression and consequently reduced the target protein expression. Sirt 5 siRNAs showed different effects. According to the results of qRT-PCR, two Sirt 5 siRNAs significantly (p<0.001) reduced the target gene expression. While western-blot analysis showed that only Sirt 5 siRNA-1 restricted the expression of the target protein.

Measurement of NO production and ROS level in CCLP-1 cells

NO and ROS have been implicated in the process of CCA by inducing cell damage through the formation of toxic radicals. Hence, we tested the influence of NO by different CCLP-1 cells treated with rCsNOSIP. As shown in Fig 4A, compared to control Cs- group, control Cs+ group significantly increased NO production in CCLP-1 cells (Fig 4A). Interestingly, NO production was increased inASPSCR-1/Sirt-5 low expression (ASP-si or Sirt5-si) CCLP-1 cells (Fig 4A) but decreased in Asp-oe or Sirt5-oe CCLP-1 cells (Fig 4A) when stimulated with rCsNOSIP. In addition, ROS results was shown in Fig 4B, in agreement with NO results, indicated that there were significant increase in ASP-si or Sirt5-si CCLP-1 cells (Fig 4B) but decrease in ASPSCR-1/Sirt-5 over expression (Asp-oe or Sirt5-oe) CCLP-1 cells (Fig 4B) when stimulated with rCsNOSIP (Fig 4B)

thumbnail
Fig 4. Measurement of NO production (A) and ROS (B) in CCLP-1 cells.

Groups: CsNOSIP-: CCLP-1 cells without stimulation; CsNOSIP+: CCLP-1 cells stimulated by CsNOSIP; Asp-si-CsNOSIP+: ASPSCR1 siRNA inhibition CCLP-1 cells stimulated by CsNOSIP; Asp-oe-CsNOSIP+: ASPSCR1 over-expression CCLP-1 cells stimulated by CsNOSIP; Sirt5-si-CsNOSIP+: Sirt5 siRNA inhibition CCLP-1 cells stimulated by CsNOSIP; Sirt5-oe-CsNOSIP+: Sirt5 over-expression CCLP-1 cells stimulated by CsNOSIP. Assays were performed in triplicate and data were displayed as mean ± SD. Statistical significance was analyzed by the one-way ANOVA test (*p <0.05, **p < 0.01, ***p < 0.001).

https://doi.org/10.1371/journal.pntd.0011727.g004

CsNOSIP stimulates proliferation and migration

CCK-8 cell proliferation assay and wound healing assays were performed to investigate the effects of rCsNOSIP on ASP-si/Sirt5-si or ASP-oe/Sirt5-oe CCLP1 cells. As shown in Fig 5C, after stimulation with rCsNOSIP, the proliferation of CCLP1 cells slightly increased but was not changed significantly, while the migration of CCLP1 cells was increased significantly (p<0.001) (Fig 5B). Moreover, the proliferation and migration were increased in ASP-si or Sirt5-si CCLP-1 cells but decreased in Asp-oe or Sirt5-oe CCLP-1 cells when stimulated with rCsNOSIP (Fig 5B and 5C).

thumbnail
Fig 5. The effect of CsNOSIP on CCLP1 cells.

(A) Cell migration of CCLP1 cells shown by wound-healing assays; cells were observed using a light microscope under 10× objective. (B) Migration of different cells treated by CsNOSIP. (C) Cell proliferation level measured by CCK-8 assay. Relative cell migration level was calculated by normalizing to cell migration level at 0 h. Groups: CsNOSIP-: CCLP-1 cells without stimulation; CsNOSIP+: CCLP-1 cells stimulated by CsNOSIP; Asp-si- CsNOSIP+: ASPSCR1 siRNA inhibition CCLP-1 cells stimulated by CsNOSIP; Asp-oe-CsNOSIP+: ASPSCR1 over-expression CCLP-1 cells stimulated by CsNOSIP; Sirt5-si-CsNOSIP+: Sirt5 siRNA inhibition CCLP-1 cells stimulated by CsNOSIP; Sirt5-oe-CsNOSIP+: Sirt5 over-expression CCLP-1 cells stimulated by CsNOSIP. Assays were performed in triplicate and data were displayed as mean ± SD. Statistical significance was analyzed by the one-way ANOVA test (*p< 0.05, **p< 0.01, ***p< 0.001).

https://doi.org/10.1371/journal.pntd.0011727.g005

Discussion

C. sinensis infection is a major risk factor for CCA in Asian countries [7]. C. sinensis-induced CCA is closely linked to oxidative or nitrative stress for creating feasible microenvironment conducive for initiation and promotion of CCA [24]. CsESPs are capable of inducing the generation of ROS/ reactive nitrogen species (RNS) through the activation of NADPH oxidases, xanthine oxidase, and iNOS [12]. Among various components of CsESPs, rCsNOSIP was identified as NOS interacting protein that upregulated the expression of iNOS in macrophage [25]. Accordingly, our results showed that ROS/NO in CCLP-1 cells increased with rCsNOSIP stimulation was speculated to be resulted from the upregulation of catalytic enzymes (Fig 4). In addition to provoke oxidative or nitrative stress, CsESPs have been proven to promote proliferation and suppress apoptosis in different human cells [26,27]. Although rCsNOSIP did not exhibit an obvious effect on enhancing the proliferation of CCLP-1 cells, its effect on enhancing the migration of CCLP-1 cells was similar to that of C. sinensis granulin, another ingredient of CsESPs. This indicates that CsNOSIP might primarily participated in promoting CCA invasion and metastasis (Fig 5) [28].

Given the importance of CsNOSIP as a key molecule in CsESPs [17,25], we identified 70 potential rCsNOSIP-binding proteins (Supporting Information), which predominantly enriched in protein serine/threonine kinase activity, regulating oxidative stress-induced intrinsic apoptotic signaling pathway, and GMP-PKG signaling pathway (Fig 1B). During infection, oxidative or nitrative stress will be triggered from host cells for generating large amounts of nitrogen and oxygen radicals to eliminate pathogen. For example, NO as the production of nitrative stress can activate its downstream cGMP-dependent protein kinase G (PKG) and participate in a protective response against environmental injury [29]. Since CsNOSIP can stimulate ROS/NO production (Fig 4), these rCsNOSIP-binding proteins may be involved in a host defense mechanism against C. sinensis infection. Of these proteins, SIRT5 is a member of sirtuins (SIRTs) family, which are highly conserved nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases with the ability to deacetylate histone and nonhistone targets [30,31], while no researches have reported the function of alveolar soft part sarcoma chromosomal region candidate gene 1 protein (ASPSCR-1). We firstly observed that ROS/NO production, proliferation, or migration were increased in ASP-si or Sirt5-si CCLP-1 cells but decreased in Asp-oe or Sirt5-oe CCLP-1 cells when stimulated with rCsNOSIP (Figs 4 and 5). Excessive NO and other reactive oxygen intermediates will lead to fatal pathogenic consequences, such as oxidative DNA or protein damage and even cancerization [24]. Recently researches revealed a ROS elimination role of SIRT5 through activating superoxide dismutase 1 and inhibiting peroxisome [32,33]. Thus, the interaction of Sirt-5 and ASPSCR-1 with rCsNOSIP implied the protective role of these two proteins in response to pathological stimulus and oxidative or nitrative damage (S3 Fig).

Collectively, our results uncovered a previously unknown function of rCsNOSIP in promoting CCA invasion and metastasis as well as a novel protective role of SIRT5/ ASPSCR1 against adverse stimulus, which partly support the acceptable theory that CsESPs induce the occurrence and progression of CCA through ROS/RNS-induced oxidative and nitrative DNA damage [34]. Further investigations are required to clarify the precise mechanisms for providing therapeutic strategies of clonorchiasis.

Supporting information

S1 Fig. Ni2+-NTA chromatographyl. UV280 nm cure(blue), Programmed %B cure(green) and conductivity cure(red) were show in the graph.

https://doi.org/10.1371/journal.pntd.0011727.s001

(TIF)

S2 Fig. Identification of rCsNOSIP-binding proteins.

(A) The whole protein chip scanning result comprising 21,000 probes. (B) Enlarged piture of one block in this chip assay. Red arrow indicated the positive control. Blue arrow indicated the negative control. Yellow arrow indicated the exhibition spot of positive protein.

https://doi.org/10.1371/journal.pntd.0011727.s002

(TIF)

S3 Fig. Expression analysis of ASPSCR-1 and Sirt-5 in different condition.

(A) Western blot to examine over-expression ASPSCR-1/Sirt-5 in CCLP-1 cells. Lane 1: cells without transfection; Lane 2: cells transfected with an over-expression vector and cultured for 48 h; Lane 3: cell transfected with an over-expression vector and cultured for 72 h. (B) Western blot to examine expression ASPSCR-1/Sirt-5 in CCLP-1 cells. Cells were collected at 72 h after transfected with siRNA. Lane 4: ASPSCR-1/Sirt-5 expression interfered by siRNA 1 group; lane 5: ASPSCR-1/Sirt-5 expression interfered by siRNA 2 group; Lane 6: ASPSCR-1/Sirt-5 expression without siRNA. QPCR to examine expression ASPSCR-1/Sirt-5 in CCLP-1 cells. Cells were collected at 72 h after transfected with siRNA. (C) Results of ASPSCR-1 gene expression treated by siRNAs. (D) Results of Sirt-5 gene expression treated by siRNAs. Assays were performed in triplicate and data were displayed as mean ± SD. Statistical significance was analyzed by the Student’s t test (***p < 0.001).

https://doi.org/10.1371/journal.pntd.0011727.s003

(TIF)

S1 Table. Potencial proteins interaction with CsNOSIP.

https://doi.org/10.1371/journal.pntd.0011727.s004

(DOCX)

References

  1. 1. Lun ZR, Gasser RB, Lai DH, Li AX, Zhu XQ, Yu XB, et al. Clonorchiasis: a key foodborne zoonosis in China. Lancet Infect Dis. 2005; 5:31–41.
  2. 2. Li S, Chen X, Zhou J, Xie Z, Shang M, He L, et al. Amino acids serve as an important energy source for adult flukes of Clonorchis sinensis. PLoS Negl Trop Dis 2020;14: e0008287.
  3. 3. Chen X, Li S, He L, Wang X, Liang P, Chen W, et al. Molecular characterization of severin from Clonorchis sinensis excretory/secretory products and its potential anti-apoptotic role in hepatocarcinoma PLC cells. PloS Negl Trop Dis. 2013;7: e2606.
  4. 4. Young ND, Jex AR, Cantacessi C, Campbell BE, Laha T, Sohn WM, et al. Progress on the transcriptomics of carcinogenic liver flukes of humans–unique biological and biotechnological prospects. Biotechnol Adv 2010;28:859–870. pmid:20673796
  5. 5. Shin HR, Oh JK, Masuyer E, Curado MP, Bouvard V, Fang YY, et al. Epidemiology of cholangiocarcinoma: an update focusing on risk factors. Cancer Sci 2010;101:579–585. pmid:20085587
  6. 6. Lim MK, Ju YH, Franceschi S, Oh JK, Kong HJ, Hwang SS, et al. Clonorchis sinensis infection and increasing risk of cholangiocarcinoma in the Republic of Korea. Am J Trop Med Hyg 2006;75:93–6.
  7. 7. Chen T, Yu J, Tang Z, Xie Z, Lin Z, Sun H, et al. Advanced enzymology, expression profile and immune response of Clonorchis sinensis hexokinase show its application potential for prevention and control of clonorchiasis. PloS Negl Trop Dis. 2015; 9:e0003641.
  8. 8. Sripa B, Kaewkes S, Sithithaworn P, Mairiang E, Laha T, Smout M, et al. Liver fluke induces cholangiocarcinoma. PloS Med 2007;4: e201. pmid:17622191
  9. 9. Shi M, Zhou L, Zhao L, Shang M, He T, Tang Z, et al. Csseverin inhibits apoptosis through mitochondria-mediated pathways triggered by Ca2 + dyshomeostasis in hepatocarcinoma PLC cells. PloS Negl Trop Dis. 2017;11: e000 6074. pmid:29125839
  10. 10. Kawanishi S, Ohnishi S, Ma N, Hiraku Y, Oikawa S, Murata M. Nitrative and oxidative DNA damage in infection-related carcinogenesis in relation to cancer stem cells. Genes Environ. 2017;1:38:26. pmid:28050219
  11. 11. Kawanishi S, Hiraku Y. Oxidative and nitrative DNA damage as biomarker for carcinogenesis with special reference to inflammation. Antioxid Redox Signal. 2006;8(5–6):1047–58. pmid:16771694
  12. 12. Nam JH, Moon JH, Kim IK, Lee MR, Hong SJ, Ahn JH, et al. Free radicals enzymatically triggered by Clonorchis sinensis excretory-secretory products cause NF-kB-mediated inflammation in human cholangiocarcinoma cells. Int J Parasitol 2012; 42:103–113.
  13. 13. Stuehr DJ, Haque MM. Nitric oxide synthase enzymology in the 20 years after the Nobel Prize. Br J Pharmacol. 2019; 176:177–188. pmid:30402946
  14. 14. Bateman RM, Sharpe MD, Ellis CG. Bench-to-bedside review: microvascular dysfunction in sepsis—hemodynamics, oxygen transport, and nitric oxide. Crit Care. 2003;7:359–73. pmid:12974969
  15. 15. König P, Dedio J, Oess S, Papadakis T, Fischer A, Müller-Esterl W, et al. NOSIP and its interacting protein, eNOS, in the rat trachea and lung. Journal of Histochemistry & Cytochemistry 2005;53: 155–164.
  16. 16. Sadaf S, Nagarkoti S, Awasthi D, Singh AK, Srivastava RN, Kumar S, et al. nNOS induction and NOSIP interaction impact granulopoiesis and neutrophil differentiation by modulating nitric oxide generation. Biochim Biophys Acta Mol Cell Res. 2021;186:119018. pmid:33771575
  17. 17. Bian M, Li S, Wang X, Xu Y, Chen W, Zhou C, et al. Identification, immunolocalization, and immunological characterization of nitric oxide synthase-interacting protein from Clonorchis sinensis. Parasitol Res. 2014;113:1749–57. pmid:24604383
  18. 18. Ou J, Peng Y, Yang W, Zhang Y, Hao J, Li F, et al. ABHD5 blunts the sensitivity of colorectal cancer to fluorouracil via promoting autophagic uracil yield. Nat Commun. 2019;10: 1078. pmid:30842415
  19. 19. Cui C, Duan Y, Qiu C, Wang P, Sun G, Ye H, et al. Identification of Novel Autoantibodies Based on the Human Proteomic Chips and Evaluation of Their Performance in the Detection of Gastric Cancer. Front Oncol. 2021; 11:637871. pmid:33718231
  20. 20. Huang L, Hu Y, Huang Y, Fang H, Li R, Hu D, et al. Gene/protein expression level, immunolocalization and binding characteristics of fatty acid binding protein from Clonorchis sinensis (CsFABP). Mol Cell Biochem. 2012;363(1–2):367–76.
  21. 21. González-Fernández R, Martín-Ramírez R, Rotoli D, Hernández J, Naftolin F, Martín-Vasallo P, et al. Granulosa-Lutein Cell Sirtuin Gene Expression Profiles Differ between Normal Donors and Infertile Women. Int J Mol Sci. 2019;21:295. pmid:31906251
  22. 22. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29:e45. pmid:11328886
  23. 23. Chen W, Ning D, Wang X, Chen T, Lv X, Sun J, et al. Identification and characterization of Clonorchis sinensis cathepsin B proteases in the pathogenesis of clonorchiasis. Parasit Vectors. 2015;8:647.
  24. 24. Prueksapanich P, Piyachaturawat P, Aumpansub P, Ridtitid W, Chaiteerakij R, Rerknimitr R. Liver Fluke-Associated Biliary Tract Cancer. Gut Liver.2018;12:236–245. pmid:28783896
  25. 25. Bian M, Xu Q, Xu Y, Li S, Wang X, Sheng J, et al. Investigation on oxidative stress of nitric oxide synthase interacting protein from Clonorchis sinensis. Parasitol Res. 2016;115:77–83.
  26. 26. Kim YJ, Choi MH, Hong ST, Bae YM. Resistance of cholangiocarcinoma cells to parthenolide-induced apoptosis by the excretory-secretory products of Clonorchis sinensis. Parasitol Res. 2009;104:1011–6.
  27. 27. Kim YJ, Choi MH, Hong ST, Bae YM. Proliferative effects of excretory/secretory products from Clonorchis sinensis on the human epithelial cell line HEK293 via regulation of the transcription factor E2F1. Parasitol Res. 2008;102:411–7.
  28. 28. Wang C, Lei H, Tian Y, Shang M, Wu Y, Li Y, et al. Clonorchis sinensis granulin: identification, immunolocalization, and function in promoting the metastasis of cholangiocarcinoma and hepatocellular carcinoma. Parasit Vectors. 2017;10:262.
  29. 29. Mahneva O, Caplan SL, Ivko P, Dawson-Scully K, Milton SL. NO/cGMP/PKG activation protects Drosophila cells subjected to hypoxic stress. Comp Biochem Physiol C Toxicol Pharmacol. 2019;223:106–114. pmid:31150868
  30. 30. Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid Redox Signal. 2018;28:643–661. pmid:28891317
  31. 31. Matsushima S, Sadoshima J. The role of sirtuins in cardiac disease. Am J Physiol Heart Circ Physiol. 2015;309:H1375–89. pmid:26232232
  32. 32. Lin ZF, Xu HB, Wang JY, Lin Q, Ruan Z, Liu FB, et al. SIRT5 desuccinylates and activates SOD1 to eliminate ROS. Biochem Biophys Res Commun. 2013;441:191–5. pmid:24140062
  33. 33. Chen XF, Tian MX, Sun RQ, Zhang ML, Zhou LS, Jin L, et al. SIRT5 inhibits peroxisomal ACOX1 to prevent oxidative damage and is downregulated in liver cancer. EMBO Rep. 2018;19: e45124. pmid:29491006
  34. 34. Kim TS, Pak JH, Kim JB, Bahk YY. Clonorchis sinensis, an oriental liver fluke, as a human biological agent of cholangiocarcinoma: a brief review. BMB Rep. 2016;49:590–597.