Skip to main content

Preliminary study of BF/C2 on immune mechanism of grass carp against GCRV infection

Abstract

BF/C2 is a crucial molecule in the coagulation complement cascade pathway and plays a significant role in the immune response of grass carp through the classical, alternative, and lectin pathways during GCRV infection. In vivo experiments demonstrated that the mRNA expression levels of BF/C2 (A, B) in grass carp positively correlated with GCRV viral replication at various stages of infection. Excessive inflammation leading to death coincided with peak levels of BF/C2 (A, B) mRNA expression and GCRV viral replication. Correspondingly, BF/C2 (A, B) recombinant protein, CIK cells and GCRV co-incubation experiments yielded similar findings. Therefore, 3 h (incubation period) and 9 h (death period) were selected as critical points for this study. Transcriptome sequencing analysis revealed significant differences in the expression of BF/C2A and BF/C2B during different stages of CIK infection with GCRV and compared to the blank control group (PBS). Specifically, the BF/C2A_3 and BF/C2A_9 groups exhibited 2729 and 2228 differentially expressed genes (DEGs), respectively, with 1436 upregulated and 1293 downregulated in the former, and 1324 upregulated and 904 downregulated in the latter. The BF/C2B_3 and BF/C2B_9 groups showed 2303 and 1547 DEGs, respectively, with 1368 upregulated and 935 downregulated in the former, and 818 upregulated and 729 downregulated in the latter. KEGG functional enrichment analysis of these DEGs identified shared pathways between BF/C2A and PBS groups at 3 and 9 h, including the C-type lectin receptor signaling pathway, protein processing in the endoplasmic reticulum, Toll-like receptor signaling pathway, Salmonella infection, apoptosis, tight junction, and adipocytokine signaling pathway. Additionally, the BF/C2B groups at 3 and 9 h shared pathways related to protein processing in the endoplasmic reticulum, glycolysis/gluconeogenesis, and biosynthesis of amino acids. The mRNA levels of these DEGs were validated in cellular models, confirming consistency with the sequencing results. In addition, the mRNA expression levels of these candidate genes (mapk1, il1b, rela, nfkbiab, akt3a, hyou1, hsp90b1, dnajc3a et al.) in the head kidney, kidney, liver and spleen of grass carp immune tissue were significantly different from those of the control group by BF/C2 (A, B) protein injection in vivo. These candidate genes play an important role in the response of BF/C2 (A, B) to GCRV infection and it also further confirmed that BF/C2 (A, B) of grass carp plays an important role in coping with GCRV infection.

Peer Review reports

Introduction

The complement system is a highly conserved part of the innate immune system, encompassing various membrane-bound and soluble components closely related to the adaptive immune system. It is involved in initiating the adaptive immune response and supports numerous host defense mechanisms, including chemotaxis, opsonization, induction of inflammatory responses, and the cleavage of microbes, apoptotic cells, and immune complexes [1]. The system consists of a complex and nuanced cascade of over 50 soluble and cell-binding proteins, primarily found in serum and cell membranes [2,3,4]. It activates through three main pathways (classical, alternative, and lectin) and regulates lysis through one pathway. Each pathway is initiated by distinct pathogen patterns and involves different recognition molecules [5,6,7]. The classical pathway (CP) is triggered by antigen–antibody complexes or C-reactive protein, the lectin pathway (LP) detects mannose on bacterial surfaces, and the alternative pathway (AP) functions continuously at low levels through spontaneous C3 hydrolysis [5, 8]. Though driven by various mechanisms, all pathways converge on the cleavage of key proteins C3 and C5, activating the membrane attack complex (MAC) and inducing the cleavage of target cells [8]. Numerous studies have highlighted the indispensable role of the complement system in defending against pathogens [9].

In this system, BF/C2 is an essential molecule. In mammals, BF and C2 derive from different pathways: BF is crucial in the alternative pathway, while C2 is important in both the classical and lectin pathways. In fish, these components are not distinguishable, leading to the general use of the term BF/C2 [10]. Specifically, in grass carp, BF/C2 encompasses BF/C2A and BF/C2B, which fold into three globular domains similar to those of BF and C2 in humans and mice. The correlation between BF/C2A, BF/C2B, and BF and C2 in fish mirrors that found in bony fish, cartilaginous sharks, and jawless lampreys [11, 12]. The specific role of BF/C2 in the fish complement system remains a key focus of ongoing research. Whether BF/C2 plays the same role in fish and mammals is also the focus of current research.

In mammals, BF and C2 have been identified as interferon-stimulating genes [13,14,15]. Clinical studies using interferon treatments for human viral diseases demonstrated significant induction of BF and C2 expression by interferon. Subsequent research indicated that both α-interferon and γ-interferon could stimulate the production of BF and C2, associated with the presence of interferon-stimulated response elements (ISRE) and γ-interferon activation sequence (GAS) on the promoters of the BF and C2 genes [14, 15]. These elements on the promoter can be recognized and bound by the transcription factors IRFs and STATs, activated by interferon signals, thereby activating the promoter and enhancing the transcriptional expression of BF and C2 [14, 15]. Hence, BF and C2 are considered typical interferon-stimulating genes. Studies have verified that the complement proteins BF and C2 predominantly exert an antiviral effect by activating the complement system. Knockout experiments in mice, lacking the BF or C2 gene, showed that these mice struggled to combat infections such as influenza A virus, West Nile virus, and poxvirus, indicating a reduced activation of C3 and the complement system, increased viral proliferation, and higher susceptibility and mortality [14, 15]. Moreover, the antiviral response of the BF and C2 genes varies among species. For instance, in response to dengue virus infection, the expression level of the mouse BF gene was higher than that in humans, resulting in a more pronounced activation of C3 and the complement system and, consequently, enhanced disease resistance in mice. This difference is attributed to the greater number of ISRE and GAS sites on the mouse BF gene promoter compared to humans [14, 15]. Considering the sequential relationship between promoter activity, gene expression, and disease resistance, increasing evidence suggests that variations in the BF and C2 promoters are closely linked to human resistance to viral diseases. Given this background, the role of BF/C2 in grass carp, particularly whether a similar regulatory mechanism exists, remains an intriguing question for further investigation.

Grass carp (Ctenopharyngodon idella), the highest producing freshwater aquaculture species in our country, reached a production of 590.48 million tons in 2022 [16]. However, the aquaculture industry faces significant challenges due to hemorrhagic disease caused by Grass carp reovirus (GCRV), with the low immunity of first-year grass carp being a primary factor in the high mortality rates among young fish induced by GCRV [17]. In 2012, our team conducted a transcriptomic analysis of the spleen of first-year grass carp before and after GCRV infection. We found that the differences in BF/C2 were particularly pronounced within the complement-coagulation cascade pathway, which exhibited the most significant changes [18]. Additionally, using modern molecular-assisted resistance breeding alongside traditional techniques, our team assessed the activity level of the BF/C2 complement protein in the plasma of grass carp parental stocks from the Xiangjiang and Yangtze Rivers using ELISA. The results indicated that BF/C2 complement protein levels were normally distributed within the grass carp population. Furthermore, in 2014, our research group immunized female grass carp with a GCRV attenuated vaccine, resulting in offspring with significantly enhanced resistance to GCRV [19, 20]. It was observed that the serum of offspring from highly immune-resistant mothers exhibited elevated expressions of complement proteins (BF/C2, C3, etc.) [19, 20]. These findings suggest that BF/C2 may play a crucial role in the resistance of grass carp to GCRV infection. Consequently, this study undertook a transcriptomic analysis of BF/C2 (A, B) proteins (with PBS as a control) during different stages of GCRV infection in CIK cells, aiming to elucidate the immune mechanism of BF/C2 (A, B) in response to GCRV infection in grass carp.

Material and method

BF/C2(A, B) protein-GCRV-CIK cell lines co-incubation experiment

To examine the impact of GCRV on C. idellus kidney cells (CIK) following incubation with BF/C2(A, B) protein, CIK cell lines established by our research group were utilized (They were cultured in M199 medium containing 1% penicillin–streptomycin and 10% fetal bovine serum at 28 ℃ in an incubator containing 5% CO2 and saturated humidity). Initially, well-cultured CIK cells were seeded in six-well plates and allowed to grow overnight until they completely covered the bottom of the plates. Subsequently, BF/C2(A, B) protein [21] was added at a final concentration of 380 ng/ml. After two hours of incubation, GCRV (strain: 1.58 × 10^4 TCID50/mL, GCRV-873 strain) was introduced to the cells. Samples were then collected at various time points: 1 h, 3 h, 6 h, 9 h, 12 h, and 24 h post-infection. Gene expression level of vp7 (GCRV) were analyzed by quantitative PCR (qPCR).

The experiment with grass carp infected with GCRV

Grass carps (5–7 cm in body length) were sourced from Huarong County, Hunan Province, China. The fish underwent a one-week acclimation period in recirculating freshwater tanks maintained at 28 °C and were fed a commercial diet equivalent to 3% of their body weight twice daily before any experimental procedures. The animal experiments were conducted in accordance with the guidelines approved by the Animal Care and Use Committee of Hunan Agricultural University (Changsha, China; Approval Code: 201,903,295; Approval Date: September 13, 2019). A total of 100 grass carps were used for the GCRV challenge experiment and were randomly allocated into two groups. Fish in the experimental group were exposed to the GCRV virus (a type II virus (Huan1307) donated by the Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences) by immersion for 10 min before being returned to the circulation tanks, while the others served as the control group. Grass carps were randomly sampled (five individuals per time point) at five different stages as identified in prior research, including the incubation period (12 h post GCRV challenge, prior to the onset of symptoms), onset period (when symptoms began to emerge), death period (when grass carps started dying), recovering period (when grass carps began to recover), and restored period (when grass carps fully recovered and symptoms had disappeared). After anaesthesia with MS-222 (25 mg/L) (Sigma Aldrich Co., St. Louis, USA) prior to sample collection, liver tissue from each sample was collected and stored at -80 °C until RNA was extracted.

Sample preparation

In this study, CIK cell lines established by our research group were utilized. Initially, well-cultured CIK cells were seeded in six-well plates and allowed to grow overnight until they completely covered the bottom of the plates. Subsequently, BF/C2 (A,B) protein was added to achieve a final concentration of 380 ng/ml. After an incubation period of 2 h, GCRV was introduced to the wells. The cells were then incubated for additional periods of 3 h and 9 h, after which they were harvested using Trizol reagent and promptly stored at -80 °C.

Library construction, and high-throughput sequencing

Total RNA of CIK samples was extracted and evaluated for purity and quantity. Quality assessment was performed according to the RNA quality assessment criteria. After meeting the assessment conditions, RNA libraries were constructed and sequenced, producing 150 bp-long paired-end reads. The above sequencing process and analysis were performed by Novogene Bioinformatics Technology Co. Ltd. (Beijing, China). Please refer to [22] for sequencing standards and specific procedures.

RNA sequencing analysis

Raw RNA data were obtained in FASTQ format and processed using fastp (Version 20.1, length required 50). This involved the removal of reads containing poly-N sequences and low-quality reads to produce clean reads. Adaptor sequences and low-quality sections were also removed before the clean reads were assembled into expressed sequence tag clusters (contigs). These contigs were then subjected to de novo assembly into transcripts using Trinity (Version 2.4, seqType fq, SS_lib_type RF) via the paired-end method. The longest transcript from each assembly was selected as a unigene based on similarity and length for subsequent analyses. Functional annotation of the unigenes was performed using the Swiss-Prot database with the diamond tool, applying a threshold of e < 1 × 10−5. Proteins that showed the highest similarity to the unigenes were used to assign functional annotations. Additionally, the unigenes were mapped against the Kyoto Encyclopedia of Genes and Genomes (KEGG) database to annotate their potential involvement in various metabolic pathways. Each unigene was quantified and its expression level was calculated, and then differential expression unigenes (DEGs) among different groups were identified [22]. This software calculated differences using a negative binomial distribution test to evaluate the significance. Hierarchical cluster analysis of DEGs was conducted using R (version 3.2.0) to visualize the expression patterns of unigenes across different experimental groups and samples. KEGG pathway enrichment analysis of the DEGs was also performed in R based on the hypergeometric distribution, helping to identify significantly impacted metabolic pathways.

qPCR analysis

Total RNA was extracted from the samples with an RNA extraction reagent (e.zn.a. ®Total RNA Kit II (Omega, Norcross, GA, USA)) and RNA quality was determined. cDNA synthesis was conducted utilizing the RevertAid™ First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, Waltham, MA, USA), adhering to the manufacturer's protocol. The cDNA was used as a template for Quantitative PCR (qPCR). The qPCR process was executed on the CFX96 Touch™ Real-Time PCR Detection System (Bio-Rad, Hercules, CA, USA). The comparative threshold cycle method (2−ΔΔCT) was used to analyze the expression levels of target genes with β-actin as the reference gene. Each experiment involved three biological replicates. Table 1 lists the primers used in this study.

Table 1 Nucleotide sequences of the primers

In vivo injection experiments of recombinant protein BF/C2(A, B)

In this experiment, 40 cultured grass carp were selected for recombinant protein injection and randomly divided into two groups. The experimental group was injected with recombinant protein BF/C2(A, B) (concentration 480 ug/ml, injection volume 100 ul/g), and the control group was injected with PBS. After 24 h of injection, five grass carp were randomly selected from the experimental group and the control group. After anaesthesia with MS-222 (25 mg/L) (Sigma Aldrich Co., St. Louis, USA) prior to sample collection. Head-kidney, kidney, liver and spleen were collected and stored at -80 ℃. The total RNA of each tissue was extracted and reverse-transcribed into cDNA. As mentioned above, mRNA expression levels of genes were detected by SYBR green fluorescent qPCR. Table 1 lists the primers used in this study.

Statistical analysis

All data are indicated as mean ± standard deviation (N = 3 or 5) and were analyzed with Statistical Package for Social Sciences Version 25.0 (SPSS Inc., Chicago, IL, USA). A two-sample Student t test was used for the comparisons between groups. Multiple group comparisons were executed by one-way ANOVA and by a Tukey multiple group comparison test. A p value < 0.05 was considered as a statistically significant difference.

Results

BF/C2(A, B) protein-GCRV-CIK cell lines co-incubation experiment

The recombinant proteins BF/C2(A,B) were added to CIK cells respectively for 2 h, and then GCRV virus was added and collected for 1 h, 3 h, 6 h, 9 h, 12 h and 24 h cells and RNA extraction was performed. The results of qPCR showed that the recombinant protein BF/C2(A,B) can suppress the virus GCRV relative expression. At 1 h-6 h, the viral replication of GCRV supplemented with BF/C2A and BF/C2B proteins was not significantly inhibited compared with PBS, and at 6 h-24 h, the viral replication of GCRV supplemented with BF/C2A and BF/C2B proteins was significantly inhibited compared with PBS. Moreover, the viral replication of GCRV with BF/C2A and BF/C2B proteins reached its peak at 9 h (Fig. 1A).

Fig. 1
figure 1

A The mRNA expressions of VP7 relative expression level in CIK. B The mRNA expressions of BF/C2(A, B) and GCRV II in liver tissues of C. idella

The fold changes in BF/C2(A, B) mRNA expression during GCRV infection

To investigate the dynamic changes in BF/C2A and BF/C2B during GCRV infection, its mRNA expression levels in the liver of C. idella after GCRV challenge at incubation period, onset period, death period, recovering period and restored period were characterized by qPCR. Compared with the control group, the mRNA expression of BF/C2A decreased in incubation period, and then showed a trend of first increasing and then decreasing in onset period, death period, recovering period and restored period. It peaked during the death period (Fig. 1B). Compared with the control group, the mRNA expression of BF/C2B decreased in incubation period, and then showed a trend of first increasing and then decreasing in onset period, death period, recovering period and restored period. It peaked during the death period (Fig. 1B). During this process, the virus replication volume of GCRV shows a trend of first increasing and then decreasing from onset period, death period, recovering period and restored period, in which the peak is reached in death period (Fig. 1B). The results showed that the mRNA expression levels of BF/C2A and BF/C2B were the highest when the viral replication of GCRV reached its peak, which further confirmed that BF/C2(A, B) played an important role in the response of grass carp to GCRV. Combined with the results of 2.1 experiment, we believed that 3 h and 9 h of the incubation challenge experiment were equivalent to the incubation period and death period of the live challenge experiment. Therefore, 3 h and 9 h were selected as nodes for subsequent BF/C2(A, B)-treated transcriptome analysis of grass carp kidney cells.

Sequencing data quality assessment

Through the statistical data of sequencing results (BioProject ID: PRJNA1110017, https://www.ncbi.nlm.nih.gov/sra/?term=PRJNA1110017), a total of 848,131,402 raw reads were obtained, and 818,834,098 clean data were screened. The error rate was 0.01. The percentage of Q20 bases and Q30 bases were greater than 98.67% and 96.48%, respectively, and the GC content was 45.37 ~ 47.49% (Table 2). To obtain mapping data (reads), clean data (reads) are compared with reference genomes (NCBI: gcf_019924925_1_hzgc01, Sex: female). The results are shown in Table 3, and the total mapped from 92.02% to 94.25%. The above results show that Illumina sequencing data and reference genome data are authentic and reliable, and can be used for follow-up studies.

Table 2 Statistics reads of transcriptomic sequences
Table 3 Comparison of reads and reference genome

DEGs analysis

Data analysis showed that BF/C2A_3 group and PBS_3 group shared 11,749 genes, BF/C2A_9 group and PBS_9 group shared 11,947 genes, BF/C2A_3 group, PBS_3 group, BF/C2A_9 group and PBS_9 group shared 11,423 genes (Fig. 2A). BF/C2B_3 group shared 11,811 genes with PBS_3 group, BF/C2B_9 group shared 11,922 genes with PBS_9 group, and BF/C2B_3 group, PBS_3, BF/C2B_9 group and PBS_9 group shared 11,467 genes (Fig. 2B). By screening for the DEGs in these shared genes, compared with PBS_3 group, there were 2729 (up:1436, down:1293) DEGs in BF/C2A_3 group and 2303 (up:1368, down: 935) DEGs in BF/C2B_3 group, respectively. Compared with PBS_9 group, BF/C2A_9 group and BF/C2B_9 group had 2228 (up:1324, down: 904) DEGs and 1547 (up:818, down: 729) DEGs, respectively (Fig. 3A, B; Attachments).

Fig. 2
figure 2

Statistical Venn diagram of DEGs between BF/C2 groups and PBS groups. A The BF/C2A group was compared with the PBS group in the three-hour and nine-hour DEGs statistical Venn diagrams; B The BF/C2B group was compared with the PBS group in the three-hour and nine-hour DEGs statistical Venn diagrams. Note: Circles with different colors represent different gene sets, and numerical values represent the number of genes/transcripts common and unique among different gene sets

Fig. 3
figure 3

Volcanic map of DEGs between BF/C2 groups and PBS groups. A BF/C2A_3_VS_PBS_3; B BF/C2A_9_VS_PBS_9; C BF/C2B_3_VS_PBS_3; D BF/C2B_9_VS_PBS_9; Note: Each point in the figure represents a specific gene, red dots and green dots represent significantly up-regulated and down-regulated genes, and gray dots are non-significantly different genes

GO

GO database contains three categories: biological process, cellular component, and molecular function, and each category contains several secondary classifications. The top 10 significantly enriched pathways of each categories were analyzed. BF/C2A_3_VS_PBS_3: biological process (DNA replication, Ras protein signal transduction and so on), cellular component (actin cytoskeleton, cytoskeletal part and so on), molecular function(enzyme binding, growth factor binding and so on) (Fig. 4A). BF/C2A_9_VS_PBS_9: biological process (ATP biosynthetic process, cofactor biosynthetic process and so on), cellular component (chromosomal region, chromosome and so on), molecular function (actin binding, cytokine receptor binding and so on) (Fig. 4B). BF/C2B_3_VS_PBS_3: biological process (coenzyme biosynthetic process, coenzyme metabolic process and so on), cellular component (actin cytoskeleton, cytoskeletal part and so on), molecular function(carbon–carbon lyase activity, coenzyme binding and so on) (Fig. 4C). BF/C2B_9_VS_PBS_9: biological process (carboxylic acid metabolic process, cofactor biosynthetic process and so on), cellular component (actin cytoskeleton, cytoplasm and so on), molecular function (actin binding, coenzyme binding and so on) (Fig. 4D).

Fig. 4
figure 4

GO classification statistics column chart of DEGs between BF/C2 groups and PBS groups. A BF/C2A_3_VS_PBS_3; B BF/C2A_9_VS_PBS_9; C BF/C2B_3 _VS_PBS_3; D BF/C2B_9_VS_PBS_9; Note: Histogram of GO classification statistics (multiple gene sets): in the graph, the abscissa indicates the secondary classification and terminology of GO, the ordinate indicates the number of genes in the secondary classification

It is found that BF/C2A_3_VS_PBS_3 and BF/C2A_9_VS_ PBS_9 share one GO signaling pathway: endoplasmic reticulum (cellular component) and BF/C2B_3_VS_PBS_3 and BF/C2B_9_VS_PBS_9 share GO signaling eight pathways: cofactor biosynthetic process, cofactor metabolic process and monocarboxylic acid metabolic process (biological process), actin cytoskeleton, endomembrane system, extracellular region and myosin complex (cellular component), and coenzyme binding (molecular function) (Fig. 4A, B, C, D).

KEGG functional annotation and enrichment analysis of DEGs

The DEGs were annotated by KEGG signal pathway using KEGG database. Through the analysis of the top 20 pathways that were significantly enriched. BF/C2A_3_VS_PBS_3: Salmonella infection (76 DEGs), C-type lectin receptor signaling pathway (38 DEGs), RIG-I-like receptor signaling pathway (22 DEGs), Protein processing in endoplasmic reticulum (50 DEGs), Apoptosis (45 DEGs), Tight junction (52 DEGs), Focal adhesion (61 DEGs), Toll-like receptor signaling pathway (26 DEGs), Adipocytokine signaling pathway (26 DEGs), Adherens junction (34 DEGs), Phagosome (36 DEGs), Motor proteins (47 DEGs), Gap junction (29 DEGs), NOD-like receptor signaling pathway (39 DEGs), Mitophagy-animal (24 DEGs), TGF-beta signaling pathway (30 DEGs), Ferroptosis (15 DEGs), Ubiquitin mediated proteolysis (35 DEGs), Sulfur metabolism (5 DEGs) and Melanogenesis (29 DEGs) (Fig. 5A). BF/C2A_9_VS_PBS_9: Salmonella infection (65 DEGs), C-type lectin receptor signaling pathway (33 DEGs), Protein processing in endoplasmic reticulum (43 DEGs), Apoptosis (39 DEGs), Toll-like receptor signaling pathway (23 DEGs), Cellular senescence (39 DEGs), Tight junction (44 DEGs), Adipocytokine signaling pathway (22 DEGs), Nucleotide metabolism (23 DEGs), NOD-like receptor signaling pathway (34 DEGs), Motor proteins (43 DEGs), RIG-I-like receptor signaling pathway (15 DEGs), Herpes simplex virus 1 infection (34 DEGs), Glycolysis / Gluconeogenesis (17 DEGs), Biosynthesis of amino acids (18 DEGs), Phagosome (30 DEGs), Mitophagy–animal (19 DEGs), VEGF signaling pathway (19 DEGs), p53 signaling pathway (17 DEGs) and Terpenoid backbone biosynthesis (6 DEGs) (Fig. 5B). BF/C2B_3_VS_PBS_3: Tight junction (49 DEGs), Salmonella infection (62 DEGs), Adherens junction (34 DEGs), Protein processing in endoplasmic reticulum (42 DEGs), Regulation of actin cytoskeleton (58 DEGs), Apelin signaling pathway (38 DEGs), Ferroptosis (16 DEGs), Phagosome (33 DEGs), Mitophagy– animal (23 DEGs), TGF-beta signaling pathway (28 DEGs), Focal adhesion (51 DEGs), Autophagy–animal (38 DEGs), C-type lectin receptor signaling pathway (27 DEGs), Biosynthesis of amino acids (19 DEGs), Gap junction (25 DEGs), Motor proteins (39 DEGs), Melanogenesis (26 DEGs), Toll-like receptor signaling pathway (20 DEGs), Adipocytokine signaling pathway (20 DEGs) and Glycolysis / Gluconeogenesis (16 DEGs) (Fig. 5C). BF/C2B_9_VS_PBS_9: Protein processing in endoplasmic reticulum (40 DEGs), Steroid biosynthesis (8 DEGs), Glycolysis / Gluconeogenesis (18 DEGs), Biosynthesis of amino acids (18 DEGs), Biosynthesis of cofactors (27 DEGs), Carbon metabolism (23 DEGs), Retinol metabolism (12 DEGs), Butanoate metabolism (6 DEGs), beta-Alanine metabolism (8 DEGs), Terpenoid backbone biosynthesis (6 DEGs), Pentose phosphate pathway (8 DEGs), Fatty acid degradation (10 DEGs), TGF-beta signaling pathway (20 DEGs), Phagosome (23 DEGs), Cysteine and methionine metabolism (10 DEGs), Valine, leucine and isoleucine degradation (10 DEGs), Adipocytokine signaling pathway (15 DEGs), Motor proteins (28 DEGs), Propanoate metabolism (7 DEGs) and Tryptophan metabolism (8 DEGs) (Fig. 5D).

Fig. 5
figure 5

KEGG signal pathway enrichment analysis bubble diagram of DEGs between BF/C2 groups and PBS groups. A BF/C2A_3_VS_PBS_3; B BF/C2A_9_VS_PBS_9; C BF/C2B_3_VS_ PBS_3; D BF/C2B_9_VS_PBS_9. Note: The vertical line is KEGG signaling pathway, and horizontal line represents Rich factor. (The ratio of Sample number to Background number in this term. The greater the Rich factor, the greater the degree of enrichment.) The size of the dot indicates the number of genes in this term, and the color of the dot corresponds to different p-adjust

It is found that BF/C2A_3_VS_PBS_3 and BF/C2A_9_VS_ PBS_9 share seven KEGG signaling pathways: C-type lectin receptor signaling pathway, Protein processing in endoplasmic reticulum, Toll-like receptor signaling pathway, Salmonella infection, Apoptosis, Tight junction and Adipocytokine signaling pathway and BF/C2B_3_VS_PBS_3 and BF/C2B_9_VS_PBS_9 share KEGG signaling three pathways: Protein processing in endoplasmic reticulum, Glycolysis / Gluconeogenesis, Biosynthesis of amino acids. BF/C2A_(3, 9)_VS_PBS_(3, 9) and BF/C2B_(3, 9)_VS_PBS_(3, 9) share KEGG signaling pathway: Protein processing in endoplasmic reticulum (Fig. 5).

DEGs screening

According to the results of differential genes and KEGG enrichment signaling pathway, the shared genes in BF/C2A_3_VS_PBS_3 and BF/C2A_9_VS_PBS_9 shared pathways were statistically analyzed. A total of 144 differential genes were screened out, including 24 (up:19, down:5) genes in C-type lectin receptor signaling pathway, 18 (up:15, down:3) genes in Protein processing in endoplasmic reticulum,16 (up:14, down:2) differential genes in Toll-like receptor signaling pathway, 29 (up:12, down:17) differential genes in Salmonella infection, 22 (up:8, down:14) differential genes in Apoptosis, 20 (up:4, down:16) differential genes in Tight junction and 16 (up:10, down:6) differential genes in Adipocytokine signaling pathway. The shared genes in BF/C2B_3_VS_PBS_3 and BF/C2B_9_VS_PBS_9 shared pathways were statistically analyzed. A total of 20 differential genes were screened out, including 20 (up:17, down:3) genes in Protein processing in endoplasmic reticulum (Table 4).

Table 4 Assemble the functional annotation summary of the transcript in the KEGG

qPCR verification

To verify the reliability of high-throughput sequencing results, 25 DEGs were expressed by qPCR. These 25 differential genes include 12 DEGs of BF/C2A_(3, 9)_VS_PBS_(3, 9): mapk1 (mitogen-activated protein kinase 1), il1b (interleukin 1, beta), rela (v-rel avian reticuloendotheliosis viral oncogene homolog A), nfkbiab (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha b), akt3a (v-akt murine thymoma viral oncogene homolog 3a), nfkb1 (nuclear factor of kappa light polypeptide gene enhancer in B-cells 1), nfkbiaa (nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha a), mapk9 (mitogen-activated protein kinase 9), dnajc3b (DnaJ (Hsp40) homolog, subfamily C, member 3b), hmgcs1 (3-hydroxy-3-methylglutaryl-CoA synthase 1), mapkapk3 (mitogen-activated protein kinase 3), hspa4a (heat shock protein 4a). The 8 DEGs of BF/C2B_(3, 9)_VS_PBS_(3, 9): hyou1 (hypoxia up-regulated 1), hsp90b1 (heat shock protein 90, beta (grp94), member 1), dnajc3a (DnaJ (Hsp40) homolog, subfamily C, member 3a), herpud1 (homocysteine-inducible, endoplasmic reticulum stress-inducible, ubiquitin-like domain member 1), erol (endoplasmic reticulum oxidoreductase 1 alpha), atf4 (atf4b—activating transcription factor 4b), hsp40 (heat shock protein 40), derlin (derlin). And the 5 DEGs shared by BF/C2A_(3, 9)_VS_PBS_(3, 9) and BF/C2B_(3, 9)_VS_PBS_(3, 9): C3a anaphylatoxin chemotactic receptor-like, C1r (complement C1r), C1q (complement C1q), hspa5 (heat shock protein 5), hsp90aa1.2 (heat shock protein 90, alpha (cytosolic), class A member 1, tandem duplicate 2), cxcl8a (chemokine (C-X-C motif) ligand 8a) (Table 5). The results showed that the expression levels of these genes were consistent with the transcriptome expression analysis (Fig. 6A,B,C).

Table 5 DEGs information description
Fig. 6
figure 6

Validation of the expression of 25 genes in transcriptome data by qPCR. β-Actin was used as a reference gene. There are three duplicates for each sample. A The 12 DEGs of BFC2A_(3, 9)_VS_PBS_(3, 9); B The 8 DEGs of BFC2B_(3, 9)_VS_PBS_(3, 9); C The 5 DEGs shared by BFC2A_(3, 9)_VS_PBS_(3, 9) and BFC2A_(3, 9)_VS_PBS_(3, 9)

In vivo injection experiments of recombinant protein BF/C2(A, B)

The qPCR analysis was performed on the liver, spleen, kidney and head kidney of the experimental group and the control group. The results showed that the expression trend in these immune tissues was consistent with the trend of transcriptome analysis. This further reflects the importance of these genes (Fig. 7 A,B,C).

Fig. 7
figure 7

BF/C2 (A, B) recombinant protein was injected in vivo for DEGs detection. β-Actin was used as a reference gene. There are three duplicates for each sample A The mRNA expressions of the 12 DEGs of BFC2A_(3, 9)_VS_PBS_(3, 9) in head-kidney, kidney, liver and spleen of C. idella; B The mRNA expressions of the 8 DEGs of BFC2B_(3, 9)_VS_PBS_(3, 9) in head-kidney, kidney spleen and liver of C. idella; C The mRNA expressions of the 4 DEGs of shared by BFC2A_(3, 9)_VS_PBS_(3, 9) and BFC2B_(3, 9)_VS_PBS_(3, 9) in head-kidney, kidney spleen and liver of C. idella

Discussion

BF/C2, a critical pathway molecule within the complement system, is essential for its function. Previous studies have explored the impact of Aeromonas hydrophila on BF/C2 in various fish species, including grass carp and Oryzias latipes [23]. In grass carp, it was observed that BF/C2b transcription is prevalent across different tissues and is induced both in vivo and in vitro by A. hydrophila, as well as by lipopolysaccharide and flagellin stimuli [10]. Overexpression of BF/C2b in cells led to significantly increased transcription levels of all complement components except C5. Following A. hydrophila infection or stimulation, notable upregulation was observed in the levels of BF/C2b, IL1β, TNF-α, IFN, CD59, C5aR1, and ITGβ-2 in grass carps [10, 12]. Conversely, BF/C2b transcription was down-regulated in cells interfered with after A. hydrophila attack, which also induced the NF-κB signaling pathway, underscoring the crucial role of BF/C2b in the innate immunity of grass carp [10, 12]. Additionally, studies have indicated that BF/C2 also significantly contributes to the response of live and kidney cells to GCRV infection in grass carp. There was a notable increase in BF/C2 expression in the kidney, liver, spleen, and head kidney cells following GCRV infection [21]. It was also noted that BF/C2 could activate the complement C3, though the exact mechanisms through which grass carp exerts its effects post-C3 activation by BF/C2 remain unexplored [21]. Similar to mammals, grass carp utilize components C3 through C9 to ultimately form the membrane attack complex (MAC), highlighting functional parallels in immune responses across these species.

CLRs (C-type lectin receptor) as important families of PRRs (pattern recognition receptors), playing essential roles in the innate immunity of fish. They facilitate various immune functions such as microbial agglutination, anti-bacterial or anti-viral responses, cell adhesion, enhanced opsonization, phenoloxidase activation, nodular formation, phagocytosis, and encapsulation [24, 25]. Toll-like receptors (TLRs) are crucial protein molecules in innate immunity, acting as a bridge to adaptive immunity. These single transmembrane non-catalytic proteins recognize conserved molecular structures from microorganisms. Upon breach of physical barriers by pathogens like microorganisms and viruses, TLRs recognize these invaders and trigger immune cell responses [26,27,28]. Similar to many PRRs, C-type lectins can activate the Toll receptor signaling pathway and the immune deficiency signaling pathway by recognizing pathogen-associated molecular patterns (PAMPs). This activation releases antimicrobial peptides, antiviral factors, and other immune-active substances. It also triggers the prophenoloxidase cascade, leading to melanin and active oxide production, promoting cell melanosis and nodule formation, ultimately completing the immune defense against pathogens [29]. Protein processing in the endoplasmic reticulum was notably enriched in both BF/C2A_(3, 9)_VS_PBS_(3, 9) and BF/C2B_(3, 9)_VS_PBS_(3, 9). The endoplasmic reticulum plays a crucial role in protein synthesis, processing, and modification. During viral infections and calcium homeostasis disturbances, misfolded protein accumulation can lead to severe ER stress. As part of the endoplasmic reticulum stress-mediated pathway, protein processing in the endoplasmic reticulum is a principal pathway for cell apoptosis, interacting with the death receptor pathway and the mitochondrial pathway [30]. These findings underscore the importance of protein processing in the endoplasmic reticulum as a critical pathway for future studies to understand the function of BF/C2(A, B) in grass carp.

We identified 12 significantly different genes in the BF/C2A_(3, 9)_VS_PBS_(3, 9) group, and 8 significantly different genes were identified in the BF/C2B_(3, 9)_VS_PBS_(3, 9) group. Among various intracellular signaling pathways, the MAPK cascade is particularly pivotal, with mapk1 and mapkapk3 being crucial components. These proteins are key in translating external stimuli into a broad array of cellular responses, including growth, inflammation, and stress response [31,32,33]. The regulatory function of the MAPK family in human physiology and pathology is a subject of ongoing deep research. Activated mapk1 can migrate from the cytoplasm to the nucleus, where it influences gene transcription and translation by phosphorylating multiple transcription factors, thus propagating upstream extracellular stimuli to various downstream effector molecules in the nucleus [31,32,33]. When grass carp kidney cells incubated with BF/C2A exhibit increased expression of mapk1, further studies are needed to determine if it also undergoes the aforementioned migration and action. Il1b, a member of the interleukin-1 cytokine family, is a key pro-inflammatory factor that plays an important role in the body's immune response. It is mainly secreted by sentinel cells of the innate immune system, such as mononuclear macrophages, etc. [34, 35]. Il1b, part of the interleukin-1 cytokine family, plays a crucial role as a pro-inflammatory factor in the immune response. It is primarily secreted by sentinel cells of the innate immune system, such as mononuclear macrophages [34, 35]. Il1b operates through autocrine, paracrine, and endocrine mechanisms, affecting a range of cells including mononuclear macrophages, fibroblasts, epithelial cells, and endothelial cells [34,35,36,37]. The activation of Il1b occurs when PAMPs or Damage-Associated Molecular Patterns (DAMPs) are recognized by Pattern Recognition Receptors (PRRs), which include TLRs, NOD-like receptors (NODs), retinoic acid-inducible gene I-like receptors (RLRs), CLRs, and various intracellular DNA receptors [38,39,40]. These receptors subsequently activate inflammatory transcription factors upon recognizing these molecular patterns. Research indicates that the absence of Il1b in mice leads to high susceptibility to group B streptococcus, suggesting a role for Il1b in bacterial infection inhibition [41]. Moreover, Il1b knockout mice exhibit a higher viral load in the brain compared to wild-type, indicating a reduced immune response [42]. Additionally, Il1b has been shown to inhibit the replication of the Human Immunodeficiency Virus type-1 [43]. Beyond its immunomodulatory roles, Il1b also promotes cell proliferation and differentiation, as evidenced by studies showing that Il1b can stimulate endothelial progenitor cells to form blood vessels through the activation of the MAPK phosphorylation pathway [44]. Nuclear-factor κB (NF-κB) is a ubiquitous transcription factor with multifaceted regulatory roles in cells, participating in a variety of physiological and pathological processes such as inflammation, immune response, oxidative stress, and apoptosis [45,46,47]. The NF-κB pathway is central to the regulation of various cytokine networks and controls over 200 target genes, most of which are inflammatory genes involved in the inflammatory response. This includes genes for adhesion molecules, interleukins, chemotactic factors, acute phase response genes, and cytokines, such as IL1b and tumor necrosis factor (TNF-α) [45,46,47]. Nfkb1, nfkbiab, nfkbiaa, and rela are critical members of the NF-κB protein family and play significant roles in these responses. For instance, nfkb1 encodes the p105 and p50 subunits of the NF-κB family, where p50/p50 homodimers exhibit anti-inflammatory effects by inhibiting the transcription of inflammatory cytokines like TNF-α and interleukin-12 (IL-12), and promoting the transcription of the anti-inflammatory cytokine interleukin-10 (IL-10) [48,49,50,51]. The nfkbia (a, b) genes encode IκBα, which, in its resting state, binds to p65, masking the nuclear localization signal of the p50 protein and thereby keeping NF-κB inactive in the cytoplasm [48,49,50,51]. Upon stimulation by agents such as lipopolysaccharide, reactive oxygen species, or TNF-α, IκBα becomes phosphorylated and is subsequently ubiquitinated and degraded in the proteasome. This degradation releases the p50/p65 heterodimer, which then rapidly translocates into the nucleus to activate target genes, thereby participating in and regulating a range of physiological and pathological processes [48,49,50,51].

Heat shock proteins (HSPs) are stress proteins produced by the body in response to external stressors such as heat stress, trauma, infection, tumors, and hypoxia. They are also known as molecular chaperone proteins due to their protective roles in cellular physiology [52]. HSPs are categorized based on their molecular weights into six classes: large molecular HSPs (100–110 kDa), HSP90 (83–90 kDa), HSP70 (66–78 kDa), HSP60, HSP40, and small molecular HSPs (15–30 kDa) [53]. HSP90b1, a member of the heat shock protein 90 family, shares 50% homology with HSP90 and is also referred to as Endoplasmin due to its location in the endoplasmic reticulum cavity, where it acts as a potent molecular adjuvant. In protein synthesis, HSP90b1 is involved in the correct folding, stretching, assembly, and transport of proteins. It binds to unfolded proteins, prevents protein aggregation, and inhibits the secretion of misfolded proteins. HSP90b1 has been identified as a potential molecular carrier for tumor antigens, aiding in tumor antigen presentation and activating CD8 + cytotoxic T lymphocytes, which are crucial for anti-tumor specific immune responses. In gastric cancer cells, HSP90b1 interacts with the client protein LRP5 and inhibits the ubiquitin–proteasome degradation pathway of LRP5, thereby influencing the progression of gastric cancer [54]. Additionally, lncRNA-AC245100.4 binds to HSP90, altering its chaperone function, increasing the stability of the client protein IKK, and further promoting the growth of prostate cancer [55].

As a crucial member of the heat shock protein family, the hsp40 gene plays a significant role in physiological and biochemical processes such as protein translation, folding, and translocation by stimulating the adenosine triphosphatase (ATPase) activity of the hsp70 gene [56, 57]. The J domain of the hsp40 gene, represented by dnajc3a and dnajc3b, collaborates with the hsp70 gene to regulate various life processes including apoptosis, cell metabolism, and cell survival. This interaction is vital for cellular immune responses, body growth, and embryonic development [58,59,60]. The hsp40 gene is ubiquitously present across a wide range of biological cells, from chlamydomonas to mammals [61,62,63,64]. In particular, 57 hsp40 family genes have been identified in Channel Catfish [61], 31 in the maternal lineage of Mare [62], 36 in Corhynchus Mykiss [63], and 50 in Japanese Flounder [64]. In aquatic biology research, numerous members of the heat shock protein family have been shown to be involved in the response mechanisms of aquatic organisms to heat stress. For instance, Bai Xueqiu et al. [65] observed that the relative expression levels of the hsp70 and hsp90 genes in various tissues of Echinus intermedius were upregulated following high-temperature induction. Similarly, the relative expressions of hsp40, hsp70, and hsp90 genes in the gill tissue of Japanese shrimp (Marsupenaeus japonicus) were increased after exposure to heat stress at 32℃ [58]. Liu Tianyu et al. [66] performed heat treatments at 28℃ on Chlamys farreri and discovered that such treatments could induce apoptosis in its blood cells, leading to a decrease in the immunity of C. Farreri. however, the relative expression of mRNA for the hsp70 and hsp90 genes in blood cells was upregulated, thereby helping to protect cells and tissues from damage.

Conclusions

In summary, based on the results of in vitro and in vivo experiments, 3 h(incubation period) and 9 h(peak period) were selected as the critical points of this study, and a series of differential genes (mapk1, il1b, rela, nfkbiab, akt3a, hyou1, hsp90b1, dnajc3a et al.) and pathways (C-type lectin receptor signaling pathway, protein processing in the endoplasmic reticulum, Toll-like receptor signaling pathway, Salmonella infection et al.) caused by BF/C2 in response to GCRV infection were analyzed by transcriptome sequencing. By analyzing these differential genes and pathways, the immune molecules that may be involved in BF/C2 response to GCRV infection were screened out. The immune mechanism of BF/C2 against GCRV infection in grass carp was further supported by the mRNA expression changes of these candidate molecules. This study provides a basis for further research on the immune mechanism of BF/C2 in response to GCRV infection. In addition, we also found that BF/C2A and BF/C2B may have different immune mechanisms in response to GCRV infection, and this result will be the focus of future research.

Availability of data and materials

All data generated or analyzed during this study are included in this article, and the raw data can be obtained by contacting the corresponding author. The transcriptome sequencing raw data can be found through the National Center for Biotechnology Information (NCBI) database, entry number: (BioSample) PRJNA1110017. (https://www.ncbi.nlm.nih.gov/sra/?term=PRJNA1110017)

Abbreviations

BF/C2 (A,B):

Complement factor BF/C2(A,B)

qRT-PCR:

Quantitative real-time PCR

CIK:

Kidney cell

GCRV:

Grass carp reovirus

DEGs:

Differentially expressed genes

References

  1. Dodds AW, Matsushita M. The phylogeny of the complement system and the origins of the classical pathway. Immunobiology. 2007;212(4–5):233–43.

    Article  CAS  PubMed  Google Scholar 

  2. Nakao M, Tsujikura M, Ichiki S, Vo TK, Somamoto T. The complement system in teleost fish: progress of post-homolog-hunting researches. Dev Comp Immunol. 2011;35(12):1296–308.

    Article  CAS  PubMed  Google Scholar 

  3. Leonardi L, La Torre F, Soresina A, Federici S, Cancrini C, Castagnoli R, Cardinale F. Inherited defects in the complement system. Pediatr Allergy Immunol. 2022;33:73–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Talaat IM, Elemam NM, Saber-Ayad M. Complement system: an immunotherapy target in colorectal cancer. Front Immunol. 2022;13:810993.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sarma JV, Ward PA. The complement system. Cell Tissue Res. 2011;343(1):227–35.

    Article  CAS  PubMed  Google Scholar 

  6. Noris M, Remuzzi G. Overview of complement activation and regulation. Semin Nephrol. 2013;33(6):479–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nonaka M, Smith SL. Complement system of bony and cartilaginous fish. Fish Shellfish Immunol. 2000;10(3):215–28.

    Article  CAS  PubMed  Google Scholar 

  8. Walport MJ. Complement. Second of two parts. N Engl J Med. 2001;344(15):1140–4.

    Article  CAS  PubMed  Google Scholar 

  9. Orsini F, De Blasio D, Zangari R, Zanier ER, De Simoni MG. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci. 2014;8:380.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Shen Y, Zhang J, Xu X, Fu J, Liu F, Li JL. Molecular cloning, characterization and expression of the complement component Bf/C2 gene in grass carp. Fish Shellfish Immunol. 2012;32(5):789–95.

    Article  CAS  PubMed  Google Scholar 

  11. Nakao M, Fushitani Y, Fujiki K, Nonaka M, Yano T. Two diverged complement factor B/C2-like cDNA sequences from a teleost, the common carp (Cyprinus carpio). J Immunol (Baltimore, Md : 1950). 1998;161(9):4811–8.

    Article  CAS  Google Scholar 

  12. Shin DH, Webb B, Nakao M, Smith SL. Molecular cloning, structural analysis and expression of complement component Bf/C2 genes in the nurse shark Ginglymostoma cirratum. Dev Comp Immunol. 2007;31(11):1168–82.

    Article  CAS  PubMed  Google Scholar 

  13. Brodszki N, Frazer-Abel A, Grumach AS, Kirschfink M, Litzman J, Perez E, Jolles S. European society for immunodeficiencies (ESID) and European reference network on rare primary immunodeficiency, autoinflammatory and autoimmune diseases (ERN RITA) complement guideline: deficiencies, diagnosis, and management. J Clin Immunol. 2020;40(4):576–91.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Huang Y, Krein PM, Winston BW. Characterization of IFN-gamma regulation of the complement factor B gene in macrophages. Eur J Immunol. 2001;31(12):3676–86.

    Article  CAS  PubMed  Google Scholar 

  15. Cabezas-Falcon S, Norbury AJ, Hulme-Jones J, Klebe S, Adamson P, Rudd PA, Carr JM. Changes in complement alternative pathway components, factor B and factor H during dengue virus infection in the AG129 mouse. J Gen Virol. 2021;102(3):001547.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. China Fisheries Statistical Yearbook 2023. China Agricultural Press. 2023;30. https://mp.weixin.qq.com/s/bzXyX7YJEoCief0NwbHYxQ.

  17. Chu P, He L, Huang R, Liao L, Li Y, Zhu Z, Wang Y. Autophagy Inhibits Grass Carp Reovirus (GCRV) Replication and Protects Ctenopharyngodon idella Kidney (CIK) cells from excessive inflammatory responses after GCRV infection. Biomolecules. 2020;10(9):1296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Xu BH, Zhong L, Liu QL, Xiao TY, Su JM, Chen KJ, Chen J. Characterization of grass carp spleen transcriptome during GCRV infection. Genetics and Molecular Research: GMR. 2016;15(2):gmr. 15026650.

  19. Xiao TY, Zhou ZY, Wang RH, Li YG, Jin SZ, Li W, Wang HQ. Intergenerational transmission and expression characteristics of immune factors immunized by GCRV Attenuated vaccine against grass carp mothers. Acta Fisheries Sin. 2017;41(08):1308–18. (Chinese).

    Google Scholar 

  20. Liu Y, Ding CH, Lv Z, Yu JB, Wang HQ, Zhang XW, Shi CB, Ren Y, Wang Q, Lv LG, Liu JK, Li DF, Xiao TY, Liu QL. Study on the annual variation of five immune factors in mother and progeny of grass carp treated with maternal immunization. J Aquat Sci. 2012;46(12):2396–408. (Chinese).

    Google Scholar 

  21. Wei YL, Lv Z, Liu QL, Yu JB, Xiao Y, Du ZJ, Xiao TY. Structural comparison and expression function analysis of BF/C2 in Ctenopharyngodon idella and Squaliobarbus curriculus. Fish Shellfish Immunol. 2023;142:109154.

    Article  CAS  PubMed  Google Scholar 

  22. Liu WX, Li CX, Xie XX, Ge W, Qiao T, Sun XF, Cheng SF. Transcriptomic landscape reveals germline potential of porcine skin-derived multipotent dermal fibroblast progenitors. Cell Mol Life Sci. 2023;80(8):224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kuroda N, Wada H, Naruse K, Simada A, Shima A, Sasaki M, Nonaka M. Molecular cloning and linkage analysis of the Japanese medaka fish complement Bf/C2 gene. Immunogenetics. 1996;44(6):459–67.

    Article  CAS  PubMed  Google Scholar 

  24. Ofek I, Crouch E, Keisari Y. The role of C-type lectins in the innate immunity against pulmonary pathogens. Adv Exp Med Biol. 2000;479:27–36.

    Article  CAS  PubMed  Google Scholar 

  25. Wang XW, Wang JX. Diversity and multiple functions of lectins in shrimp immunity. Dev Comp Immunol. 2013;39(1–2):27–38.

    Article  PubMed  Google Scholar 

  26. Hashimoto C, Hudson KL, Anderson KV. The Toll gene of Drosophila, required for dorsal-ventral embryonic polarity, appears to encode a transmembrane protein. Cell. 1988;52(2):269–79.

    Article  CAS  PubMed  Google Scholar 

  27. Gay NJ, Keith FJ. Drosophila Toll and IL-1 receptor. Nature. 1991;351(6325):355–6.

    Article  CAS  PubMed  Google Scholar 

  28. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86(6):973–83.

    Article  CAS  PubMed  Google Scholar 

  29. Belicky S, Katrlik J, Tkac J. Glycan and lectin biosensors. Essays Biochem. 2016;60(1):37–47.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Jiang S, Shi F, Lin H, Ying Y, Luo L, Huang D, Luo Z. Inonotus obliquus polysaccharides induces apoptosis of lung cancer cells and alters energy metabolism via the LKB1/AMPK axis. Int J Biol Macromol. 2020;151:1277–86.

    Article  CAS  PubMed  Google Scholar 

  31. Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y, Hu LL. ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med. 2020;19(3):1997–2007.

    PubMed  PubMed Central  Google Scholar 

  32. Yue J, Lopez JM. Understanding MAPK signaling pathways in apoptosis. Int J Mol Sci. 2020;21(7):2346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chen S, Zhao L, Sherchan P, Ding Y, Yu J, Nowrangi D, Zhang JH. Activation of melanocortin receptor 4 with RO27–3225 attenuates neuroinflammation through AMPK/JNK/p38 MAPK pathway after intracerebral hemorrhage in mice. J Neuroinflammation. 2018;15:106.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Weber A, Wasiliew P, Kracht M. Interleukin-1 (IL-1) pathway. Sci Signal. 2010;3(105):cm1.

    PubMed  Google Scholar 

  35. Hoffmann E, Thiefes A, Buhrow D, Dittrich-Breiholz O, Schneider H, Resch K, Kracht M. MEK1-dependent delayed expression of Fos-related antigen-1 counteracts c-Fos and p65 NF-kappaB-mediated interleukin-8 transcription in response to cytokines or growth factors. J Biol Chem. 2005;280(10):9706–18.

    Article  CAS  PubMed  Google Scholar 

  36. Bandman O, Coleman RT, Loring JF, Seilhamer JJ, Cocks BG. Complexity of inflammatory responses in endothelial cells and vascular smooth muscle cells determined by microarray analysis. Ann N Y Acad Sci. 2002;975:77–90.

    Article  CAS  PubMed  Google Scholar 

  37. Holzberg D, Knight CG, Dittrich-Breiholz O, Schneider H, Dorrie A, Hoffmann E, Kracht M. Disruption of the c-JUN-JNK complex by a cell-permeable peptide containing the c-JUN delta domain induces apoptosis and affects a distinct set of interleukin-1-induced inflammatory genes. J Biol Chem. 2003;278(41):40213–23.

    Article  CAS  PubMed  Google Scholar 

  38. Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20(2):95–112.

    Article  CAS  PubMed  Google Scholar 

  39. Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in Immunity and Sterile Inflammation. In: Abbas AK, Aster JC, Feany MB, editors. Annual review of pathology: mechanisms of disease, vol. 15. 2020. p. 493–518.

    Google Scholar 

  40. Moriyama K, Nishida O. Targeting cytokines, pathogen-associated molecular patterns, and damage-associated molecular patterns in sepsis via blood purification. Int J Mol Sci. 2021;22(16):8882.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Biondo C, Mancuso G, Midiri A, Signorino G, Domina M, Cariccio VL, Beninati C. The interleukin-1β/CXCL1/2/neutrophil axis mediates host protection against group B streptococcal infection. Infect Immun. 2014;82(11):4508–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sergerie Y, Rivest S, Boivin G. Tumor necrosis factor-α and interleukin-1β play a critical role in the resistance against lethal herpes simplex virus encephalitis. J Infect Dis. 2007;196(6):853–60.

    Article  CAS  PubMed  Google Scholar 

  43. Wang X, Mbondji-Wonje C, Zhao J, Hewlett I. IL-1β and IL-18 inhibition of HIV-1 replication in Jurkat cells and PBMCs. Biochem Biophys Res Commun. 2016;473(4):926–30.

    Article  CAS  PubMed  Google Scholar 

  44. Rosell A, Arai K, Lok J, He T, Guo S, Navarro M, Lo EH. Interleukin-1β augments angiogenic responses of murine endothelial progenitor cells in vitro. J Cereb Blood Flow Metab. 2009;29(5):933–43.

    Article  CAS  PubMed  Google Scholar 

  45. Chen F, Castranova V, Shi X, Demers LM. New insights into the role of nuclear factor-kappaB, a ubiquitous transcription factor in the initiation of diseases. Clin Chem. 1999;45(1):7–17.

    Article  CAS  PubMed  Google Scholar 

  46. Bremner P, Heinrich M. Natural products as targeted modulators of the nuclear factor-kappaB pathway. J Pharm Pharmacol. 2002;54(4):453–72.

    Article  CAS  PubMed  Google Scholar 

  47. Hall G, Hasday JD, Rogers TB. Regulating the regulator: NF-kappaB signaling in heart. J Mol Cell Cardiol. 2006;41(4):580–91.

    Article  CAS  PubMed  Google Scholar 

  48. de Winther MPJ, Kanters E, Kraal G, Hofker MH. Nuclear factor kappaB signaling in atherogenesis. Arterioscler Thromb Vasc Biol. 2005;25(5):904–14.

    Article  PubMed  Google Scholar 

  49. Gareus R, Kotsaki E, Xanthoulea S, van der Made I, Gijbels MJJ, Kardakaris R, Pasparakis M. Endothelial cell-specific NF-κB inhibition protects mice from atherosclerosis. Cell Metab. 2008;8(5):372–83.

    Article  CAS  PubMed  Google Scholar 

  50. Cao S, Zhang X, Edwards JP, Mosser DM. NF-kappaB1 (p50) homodimers differentially regulate pro- and anti-inflammatory cytokines in macrophages. J Biol Chem. 2006;281(36):26041–50.

    Article  CAS  PubMed  Google Scholar 

  51. Dhingra R, Shaw JA, Aviv Y, Kirshenbaum LA. Dichotomous actions of NF-κB signaling pathways in heart. J Cardiovasc Transl Res. 2010;3(4):344–54.

    Article  PubMed  Google Scholar 

  52. Khalil AA, Kabapy NF, Deraz SF, Smith C. Heat shock proteins in oncology: diagnostic biomarkers or therapeutic targets? Biochim Biophys Acta. 2011;1816(2):89–104.

    Article  CAS  Google Scholar 

  53. Wu J, Liu T, Rios Z, Mei Q, Lin X, Cao S. Heat shock proteins and cancer. Trends Pharmacol Sci. 2017;38(3):226–56.

    Article  CAS  PubMed  Google Scholar 

  54. Wang H, Deng G, Ai M, Xu Z, Mou T, Yu J, Li G. Hsp90ab1 stabilizes LRP5 to promote epithelial-mesenchymal transition via activating of AKT and Wnt/β-catenin signaling pathways in gastric cancer progression. Oncogene. 2019;38(9):1489–507.

    Article  CAS  PubMed  Google Scholar 

  55. Cui R, Liu C, Lin P, Xie H, Wang W, Zhao J, Yu X. LncRNA AC245100.4 binds HSP90 to promote the proliferation of prostate cancer. Epigenomics. 2020;12(15):1257–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hill RB, Flanagan JM, Prestegard JH. 1H and 15N magnetic resonance assignments, secondary structure, and tertiary fold of Escherichia coli DnaJ (1–78). Biochemistry. 1995;34(16):5587–96.

    Article  CAS  PubMed  Google Scholar 

  57. Qian YQ, Patel D, Hartl FU, McColl DJ. Nuclear magnetic resonance solution structure of the human Hsp40 (HDJ-1) J-domain. J Mol Biol. 1996;260(2):224–35.

    Article  CAS  PubMed  Google Scholar 

  58. Danwattananusorn T, Fagutao FF, Shitara A, Kondo H, Aoki T, Nozaki R, Hirono I. Molecular characterization and expression analysis of heat shock proteins 40, 70 and 90 from kuruma shrimp Marsupenaeus japonicus. Fish Sci. 2011;77(6):929–37.

    Article  CAS  Google Scholar 

  59. Qiu XB, Shao YM, Miao S, Wang L. The diversity of the DnaJ/Hsp40 family, the crucial partners for Hsp70 chaperones. Cell Mol Life Sci. 2006;63(22):2560–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Pullen MY, Weihl CC, True HL. Client processing is altered by novel myopathy-causing mutations in the HSP40 J domain. Plos One. 2020;15(6):e0234207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Song L, Li C, Xie Y, Liu S, Zhang J, Yao J, Liu Z. Genome-wide identification of Hsp70 genes in channel catfish and their regulated expression after bacterial infection. Fish Shellfish Immunol. 2016;49:154–62.

    Article  CAS  PubMed  Google Scholar 

  62. Wang Q, Wei W, Liu Y, Zheng Z, Du X, Jiao Y. HSP40 gene family in pearl oyster Pinctada fucata martensii: genome-wide identification and function analysis. Fish Shellfish Immunol. 2019;93:904–10.

    Article  CAS  PubMed  Google Scholar 

  63. Ma F, Liu Z, Kang YJ, Quan JQ. Identification of Hsp40 gene in rainbow trout and its expression under heat stress. J Agric Biotechnol. 2019;27(10):1782–92. (Chinese).

    Google Scholar 

  64. Yan W, Qiao Y, Qu J, Liu X, Zhang Q, Wang X. The hsp40 gene family in Japanese flounder: identification, phylogenetic relationships, molecular evolution analysis, and expression patterns. Front Mar Sci. 2021;2021:7.

    Google Scholar 

  65. Bai XP, Pang ZG, Zhang WJ, Chang YQ, Gao XY, Ding J. Study on the expression of hsp70 gene and hsp90 gene induced by high temperature in Echinus intermedius. Oceanol Limnol. 2015;46(05):1034–9. (Chinese).

    CAS  Google Scholar 

  66. Liu TY, Wang Q, Chen MY. Thermal stimulation on the immune function of ZhiKong scallops and the effect of heat shock protein expression. Chin J Ocean Univ (natural science edition). 2017;47(08):31–43.

    CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work is supported by the National Natural Science Foundation of China (3237210588) and the National Key Research and Development Program of China(2023YFD2401602).

Author information

Authors and Affiliations

Authors

Contributions

YLW: Investigation, Visualization, Writing-original draft. YX: Investigation, Participate in work. QLL: Writing-review & editing. ZJDu: Supervision. TYX: Supervision, Conceptualization, Methodology.

Corresponding authors

Correspondence to Zongjun Du or Tiaoyi Xiao.

Ethics declarations

Ethics approval and consent to participate

The animal experiments were conducted in accordance with the guidelines approved by the Animal Care and Use Committee of Hunan Agricultural University (Changsha, China; Approval Code: 201903295; Approval Date: September 13, 2019). All experimental procedures and sample collection methods were performed in accordance with approved guidelines and regulations to ensure animal welfare.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

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

Wei, Y., Xiao, Y., Liu, Q. et al. Preliminary study of BF/C2 on immune mechanism of grass carp against GCRV infection. BMC Genomics 25, 715 (2024). https://doi.org/10.1186/s12864-024-10609-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12864-024-10609-3

Keywords