Scavenger receptor class A member 5 (SCARA5) and high-mobility group box 1 (HMGB1) protein have become increasingly attractive for their critical functions in innate inflammatory reactions and disorders. However, the functional relevance between these two molecules has never been described. This study discovered that SCARA5 is an HMGB1 recognition receptor that is negatively involved in HMGB1-mediated inflammation in pufferfish (Tetraodon nigroviridis) and zebrafish (Danio rerio) models. Hence, SCARA5 is added as a new member to the HMGB1 receptor family. Tetraodon HMGB1 (TnHMGB1) is a trafficking protein that can be secreted from the nucleus to the outside of cells upon CpG-oligodeoxynucleotide (ODN) stimulation. This protein exerts a strong synergistic effect on CpG-ODN–induced inflammation, as determined by the enhanced proinflammatory cytokine expression through coadministration of TnHMGB1 with CpG-ODN and impaired inflammatory responses through TnHMGB1 depletion. Tetraodon SCARA5 (TnSCARA5) is an inducible protein detected upon TnHMGB1 stimulation; this protein plays an inhibitory role in CpG-ODN–induced inflammation because TnSCARA5 overexpression suppresses cell responsiveness to CpG-ODN induction, whereas TnSCARA5 ablation intensifies the inflammatory reactions. TnSCARA5 can strongly associate with TnHMGB1 through the A and B boxes, depending on the redox state of the cysteine residues, but T box inhibits the association. TnSCARA5 mediates the endocytosis of TnHMGB1 into lysosomes. Results suggest that TnSCARA5 inhibits the CpG-ODN–mediated inflammation via the clearance of HMGB1 mediator for CpG-ODN stimulant. The above findings highlight a novel regulatory mechanism underlying innate inflammation and provide new insights into the clinical treatment of HMGB1-mediated diseases.

Scavenger receptors (SRs) represent one group of the most important pattern recognition receptor family members; this group consists of a large subfamily with eight classes (A–H) according to their similarities in multidomain protein structure (1). SRs were initially found to recognize acetylated low-density lipoproteins, participate in lipid metabolism, and contribute to various lipid metabolism–relevant disorders, such as atherosclerosis and Alzheimer’s disease (2). Consequently, SRs have become increasingly attractive for their critical functions in innate immunity, among which class A members have received particular attention (2). Class A members are composed of the macrophage SR, macrophage receptors with long collagenous structure, SR with C-type lectin domain, and SR class A member 5 (SCARA5). These receptors can bind with an array of pathogen-associated molecular patterns (PAMPs), such as LPS, peptidoglycan, lipoteichoic acid, and unmethylated CpG-DNA, which stimulate a number of host innate inflammatory responses (35). Naturally, the host innate inflammatory responses are essential for host defense against pathogen invasions. However, excessive inflammatory responses may result in severe inflammatory disorders (6). Therefore, the innate inflammatory reactions present a double-edged mechanism, which must be tightly regulated to maintain immune homeostasis and thus prevent inflammatory disorders. Apart from the role of PAMPs in the augmentation of inflammation, damage-associated molecule patterns (DAMPs), such as the recently identified high-mobility group box 1 (HMGB1) protein, also endorse key functions to enhance inflammation (7, 8). SRs play crucial anti-inflammatory roles by eliminating PAMPs; however, whether they participate in DAMP-mediated inflammation remains poorly understood.

HMGB1 was first identified as a nonhistone nuclear protein bound to the minor groove of DNA without sequence specificity that stabilizes DNA structure and modulates transcriptional activity (9). Recently, this protein has been defined as a classical proinflammatory mediator in the pathogenesis of both sterile and infectious inflammatory processes outside the cell (10, 11). HMGB1 promotes inflammation through various mechanisms. For example, it acts as a late mediator of endotoxemia to enhance endotoxin lethality and associates with IL-1β, LPS, Pam3CSK4, and CpG-oligodeoxynucleotides (ODNs) to induce the burst of inflammatory cytokines (1214). During bacterial or viral infection, HMGB1 can be secreted by the activated monocytes and macrophages to act as a molecular chaperone for the transplant of LPS and CpG-ODNs to their receptors TLR4 and TLR9 to elicit inflammatory responses (1416). To date, RAGE is identified to be the major potent receptor for HMGB1, although TLR2 and TLR4 are also closely involved in HMGB1 signaling (1719). However, other receptors, specifically the negative regulatory receptor for HMGB1, have never been described.

Our previous study identified a SCARA5 homolog from pufferfish (Tetraodon nigroviridis) model and showed that SCARA5 is a LPS recognition receptor, which negatively regulates NF-κB activation by competing with TNFR-associated factor 2 recruitment into the TNF-α signaling pathway (4). In the present study, we report SCARA5 acts as an HMGB1 receptor that negatively regulates CpG-ODN–triggered inflammation, in which HMGB1 is an essential partner for CpG-ODN transplant to TLR9 in the endosomes (20). To our knowledge, this report is the first to show that SCARA5 is an HMGB1 receptor inhibiting the synergic effect of CpG-ODN with HMGB1 on TLR9-mediated inflammatory response; thus, a new receptor for HMGB1 is added, and new insights into the function of SR family members in host innate inflammatory responses are provided. The present findings also contribute to enhance our understanding about the biology of HMGB1.

Wild-type spotted green pufferfish (T. nigroviridis) and AB zebrafish (Danio rerio) were bred and maintained in a circulating water bath at 26–28°C under standard conditions as previously described (21). The fish were held in the laboratory for at least 2 wk before the experiment to evaluate their overall health. Only healthy fish, as determined by their general appearance and level of activity, were used in the study. Zebrafish embryos were collected at different stages of embryonic development in accordance with previously established protocols (22). Imprinting control region mice (4–8 wk) were obtained from the Laboratory Animal Unit of Zhejiang Academy of Medical Sciences (Hangzhou, China) and housed under standard conditions. All animal experiments were performed in accordance with legal regulations and were approved by a local Ethics Committee.

cDNAs of Tetraodon HMGB1 (TnHMGB1) and TnTLR9 were amplified by RT-PCR by using the homologous sequences predicted from the University of California Santa Cruz and National Center for Biotechnology Information genome databases. Full-length encoding sequence of Tetraodon SCARA5 (TnSCARA5) was generated on the basis of our previously published sequence in the National Center for Biotechnology Information genome database (no. AFX69239.1). The primers used in cloning are listed in Supplemental Table I. PCR products were purified using a gel extraction kit (Qiagen), inserted into the pGEM-T easy vector (Promega), and transformed into competent Escherichia coli TOP10 cells (Invitrogen). Plasmid DNA was purified using a plasmid Miniprep kit (Qiagen) (23). Sequence alignments of TnHMGB1 and TnTLR9 were analyzed using the ClustalW program (version 1.81) (24, 25). Phylogenetic trees were constructed with MEGA 4.0 by using the neighbor-joining method.

Encoding sequences for wild-type and mutant TnHMGB1 proteins with deficiency of A, B, or T boxes were inserted into pET28a to construct prokaryotic expression vectors, namely, pET28a-TnHMGB1, pET28a-TnHMGB1-mutA, pET28a-TnHMGB1-mutB, and pET28a-TnHMGB1-mutT. The encoding sequences of TnHMGB1 and TnTLR9 were subcloned into pDsRED-C1 and pcDNA6/myc-His B (Invitrogen) to obtain the recombinant fusion proteins of TnHMGB1 with enhanced red fluorescent protein and TnTLR9 with Myc-tag at the C-terminals, respectively. The plasmids encoding recombinant fusion proteins of TnSCARA5 with enhanced GFP and Myc/His-tag (p-enhanced GFP [EGFP]-TnSCARA5 and pcDNA6/myc-His-TnSCARA5) were constructed previously in our laboratory (4). NF-κB luciferase and pRL-TK (Renilla luciferase reporter) constructs were purchased from Clontech (Palo Alto, CA) and Promega. The primers used for construct generation are listed in Supplemental Table I. Plasmids for transfection and microinjection were prepared free of endotoxin by using an EZNA plasmid mini kit (Omega Bio-Tek).

Total RNA from leukocytes or selected tissues, including liver, intestine, spleen, kidney, skin, gill, muscle, and brain, in normal or CpG-Singapore grouper iridovirus (SGIV)-ODN–stimulated fish was isolated and transcribed into first-strand cDNA with oligo(dT) (Takara Bio) as previously described (23). The expression levels of inflammatory cytokines IL-1β, IL-6, TNF-α, and IFN-γ were quantified via real-time PCR by using a SYBR Premix Ex Taq kit (Takara Bio) in accordance with the manufacturer’s instructions. The total amount of mRNA was normalized to endogenous β-actin mRNA. Real-time PCR was performed as previously described (26). In all cases, each PCR trial was performed with triplicate samples and repeated at least three times. The primers used in the analysis are listed in Supplemental Table I.

Subcellular localization and induced secretion of TnHMGB1 were examined in human embryonic kidney 293T (HEK293T) cells. For subcellular localization, cells were seeded into multiwell plates (Corning) and cultured in DMEM supplemented with 10% FBS at 37°C in 5% CO2 to allow growth into 70–90% confluence. Subsequently, cells were transiently transfected with pDsRED-C1-TnHMGB1 plasmid by using FuGENE HD (Promega) transfection reagent in accordance with the manufacturer’s protocol (23). At 24 h posttransfection, cells were fixed in 4% paraformaldehyde for 10 min and stained with DAPI at 37°C for 5 min. Red fluorescence images were obtained using a laser scanning confocal microscope (LSM 510; Carl Zeiss) with a ×63 lens. For induced secretion, Tetraodon leukocytes were freshly isolated from the head kidney, and spleen tissues by Lympholyte-H and -M (vol ratio, 3:1; density, 1.081) (Sango), and the interlayer of cell suspensions was collected after centrifugation for 25 min at 2500 rpm (27). Afterward, the separated leukocytes were stimulated with 1 μM CpG-SGIV-ODN6 (Table I) for 8 h, and culture supernatants were collected after ultrafiltration for Western blot analysis.

Table I.
Sequences and backbone of CpG-SGIV-ODN used for stimulation or association
NameSequence (5′→ 3′)Use
CpG-SGIV-ODN1 5′-TCGTCGTACACGCGCGTGTCGTT-3′ Stimulation 
CpG-SGIV-ODN2 5′-TCGTCGAACGAAATCCCTACCGGTATCGCGACGGATTTGTCGTT-3′ Stimulation 
CpG-SGIV-ODN3 5′-AAGGTTTCGTTTCGTTTACGG-3′ Stimulation 
CpG-SGIV-ODN4 5′-TCTATCGATGAGCTGTTGACCGGGAGGGGGGG-3′ Stimulation 
CpG-SGIV-ODN5 5′-TCTATCGATGAGCTGTTGACCGGGAGGGGGGG-3′ (phosphorothioate) Stimulation 
CpG-SGIV-ODN6 5′-TCGTCGTACACGCGCGTGTCGTT-3′ (phosphorothioate) Stimulation 
CpG-SGIV-ODN6 5′-TCGTCGTACACGCGCGTGTCGTT-3′ (phosphodiester) Association 
NameSequence (5′→ 3′)Use
CpG-SGIV-ODN1 5′-TCGTCGTACACGCGCGTGTCGTT-3′ Stimulation 
CpG-SGIV-ODN2 5′-TCGTCGAACGAAATCCCTACCGGTATCGCGACGGATTTGTCGTT-3′ Stimulation 
CpG-SGIV-ODN3 5′-AAGGTTTCGTTTCGTTTACGG-3′ Stimulation 
CpG-SGIV-ODN4 5′-TCTATCGATGAGCTGTTGACCGGGAGGGGGGG-3′ Stimulation 
CpG-SGIV-ODN5 5′-TCTATCGATGAGCTGTTGACCGGGAGGGGGGG-3′ (phosphorothioate) Stimulation 
CpG-SGIV-ODN6 5′-TCGTCGTACACGCGCGTGTCGTT-3′ (phosphorothioate) Stimulation 
CpG-SGIV-ODN6 5′-TCGTCGTACACGCGCGTGTCGTT-3′ (phosphodiester) Association 

Ab against TnHMGB1 was prepared using a novel Ag epitope-based protocol (27). Briefly, the antigenic epitopes of TnHMGB1 were predicted through online software ABCPred (http://www.imtech.res.in/raghava/abcpred/) and BepiPred (http://www.cbs.dtu.dk/services/BepiPred/). The tertiary structure was modeled by the SWISS-MODEL program to detect whether the predicted epitopes were exposed on the surface of the protein, and two typical epitopes (MGKDPKKPRGKMSC and TAADDKQPYEKKAC) were used as alternative Ags to prepare the specific Abs against the TnHMGB1 protein. The epitope peptides were synthesized by InvivoGen. New Zealand White rabbits, aged 4–6 wk old and weighing 2.0–2.5 kg, were immunized with the synthetic peptides (28). Antiserum was collected after the last immunization, and Ab was affinity purified into IgG isotype by a protein A–agarose column (Qiagen) (29). Western blot analysis was performed to evaluate the specificity of the Ab. The Ab against TnSCARA5 was prepared in our previous research with purified TnexSCARA5 protein (4).

Small interfering RNA (siRNA)–encoding lentiviruses (LVs) against TnHMGB1 and TnSCARA5 were produced using a previously described protocol (23, 27). Briefly, DNA oligonucleotides encoding for the selected short hairpin RNAs containing siRNAs or the nonrelated scrambled siRNA were synthesized and constructed into the pSUPER vector downstream of the H1 promoter (30, 31). The generated constructs or control pSUPER, along with pCMV-Tag2B-TnHMGB1 or pcDNA6-TnSCARA5, were cotransfected into HEK293T cells to examine the efficacy of siRNAs via real-time PCR and Western blot analysis. The U6 promoter cassette in lentiviral vector pLB was replaced by the H1-siRNA cassette excised from the siRNA constructs with the highest efficacy to produce pLB-TnHMGB1 and pLB-TnSCARA5 lentiviral vectors. These two vectors were cotransfected with pCMV-dR8.2 and pCMV-VSVG packaging vectors into HEK293T cells by using FuGENE HD (Promega). Lentiviral supernatant was harvested three times within 48–96 h posttransfection and concentrated via ultracentrifugation at 25,000 rpm for 90 min. Viral titers were determined by transduction and FACS analysis of GFP expression in HEK293T cells. After injecting Tetraodon with LVs (5 × 105 transducing units [TU]/fish) once every 24 h for three times, the silencing efficacy of the constructed LVs was examined in PBLs and head kidney leukocytes (HKLs) through real-time PCR.

The prokaryotic expression vectors for wild-type and mutant TnHMGB1 proteins were separately transformed into E. coli Rosetta (DE3) cells. The positive colonies were inoculated into 100 ml of Luria–Bertani medium containing chloramphenicol (100 μg/ml) and kanamycin (50 μg/ml). The inoculated Luria–Bertani medium was incubated in a shaking incubator at 37°C and 250 rpm until the OD600 value reached 0.6. Subsequently, isopropyl β-d-thiogalactoside was added to a final concentration of 1 mM and shaken at 100 rpm and 20°C overnight. The proteins were expressed in soluble forms and determined by 10% SDS-PAGE. The recombinant TnHMGB1 proteins were purified by Ni-NTA agarose affinity chromatography in accordance with the QIAexpressionist manual (Qiagen). The concentration of the purified proteins was measured using the Bradford method (29). To prepare oxidized TnHMGB1 or reduced TnHMGB1, wild-type TnHMGB1 was exposed to 100 mM H2O2 for 1 h or 5 mM DTT for 1 h before adding it into cells for interaction analysis (57).

NF-κB activation was examined by luciferase reporter assays in HEK293T cells and zebrafish embryos as previously described (26). For HEK293T cells, plasmids in different combinations (pcDNA6, pcDNA6-TnTLR9, and pcDNA6-TnTLR9 plus pcDNA6-TnSCARA5), as well as NF-κB luciferase reporter construct (200 ng/ml; Clontech) and pGL-TK Renilla luciferase internal control (20 ng/ml; Clontech), were transfected into cells. After 24 h transfection, CpG-SGIV-ODN6 (5 μg/ml) was added into the supernatant for stimulation for 5 h to evaluate the involvement of TnSCARA5 in CpG-ODN/TnHMGB1–mediated inflammation. For zebrafish embryos, different concentrations (0.25, 0.5, 0.75, and 1.0 μg/μl) of CpG-SGIV-ODN6 or pcDNA6-TnSCARA5 plasmids (50, 75, 100, and 125 μg/μl) under 1.0 μg/μl CpG-SGIV-ODN6 stimulation added with NF-κB and pGL-TK were microinjected into one-cell stage embryos. After 24 h stimulation or postmicroinjection, the firefly and Renilla luciferase activities of the HEK293T cells and embryos per group were measured using a Dual-Luciferase reporter assay system (Promega). The relative luciferase activity unit was determined as the ratio of firefly luciferase activity divided by the Renilla luciferase activity. Each trial was repeated at least three times.

The association of TnSCARA5 with TnHMGB1 was examined by quantitative ELISA (qELISA) and coimmunoprecipitation (CoIP). For ELISA, various concentrations (8–256 nM) of recombinant TnHMGB1 proteins were coated onto microtiter wells overnight at 4°C. After blocking with 2% BSA for 2 h and washing three times, the wells were incubated with different concentrations (36 and 72 nM) of TnSCARA5 for 2 h at 37°C, followed by incubation with rabbit anti-TnSCARA5 Ab for 2 h at 37°C and HRP-conjugated goat anti-rabbit IgG for 30 min. Color was developed by tetramethylbenzidine and H2O2 as substrates, and OD450 was read by a Synergy H1 hybrid reader (BioTek) (29). For CoIP, HEK293T cells were transfected with pcDNA6-TnSCARA5 fused with myc and His tag. After 24 h transfection, the cell supernatant was changed to serum-free buffer, and recombinant wild-type, mutant TnHMGB1 proteins or TnHMGB1 with different redox forms were separately added for incubation for 20 min. Afterward, cells were treated with 450 μl of lysis buffer (1% Triton X-100, 150 mM NaCl, 1 mM EDTA, 0.1% SDS, 1% sodium deoxycholate, 50 mM Tris-HCl [pH 7.4]) with protease inhibitor mixture (Roche) for 30 min at 4°C. After centrifugation for 10 min at 14,000 rpm, the supernatant was collected and incubated at 4°C overnight with mouse anti-myc Ab (Abcam). Up to 50 μl of protein G–agarose bead (Sigma-Aldrich) was then added with shaking for 4 h at 4°C and washed three times with cold PBS. The precipitants were denatured in loading buffer for detection on 12% SDS-PAGE and analysis by Western blot.

The association of CpG-SGIV-ODN6 or the CpG-SGIV-ODN6-TnHMGB1 complex to TnSCARA5, as well as the association of CpG-SGIV-ODN6 to TnHMGB1, was estimated by a streptavidin pulldown assay. Biotin-labeled CpG-SGIV-ODN6 (5 μg) with a phosphorothioate or phosphodiester backbone was immobilized onto 10 μl of streptavidin Sepharose beads (Cell Signaling Technology) and then incubated with the recombinant TnHMGB1 protein or/and TnSCARA5 lysate from HEK293 cells transfected with the TnSCARA5 plasmid pcDNA6-TnSCARA5 for 16 h at 4°C. After washing three times with lysis buffer, the precipitates were analyzed by Western blot.

TnSCARA5-mediated internalization of TnHMGB1 was determined in Tetraodon leukocytes by FACS and ELISA and evaluated by the induced trafficking of TnSCARA5 under TnHMGB1 stimulation in HEK293 cells. For FACS assay, leukocytes (106 cells/ml) were preincubated with anti-TnSCARA5 Ab and normal rabbit IgG as control for 2 h at 28°C. After dealing with cytochalasin B (80 μg/ml; Sigma-Aldrich) for 1 h, the cells were treated with various concentrations (20, 30, and 40 μg) of FITC-conjugated TnHMGB1 protein for 30 min. After washing five times with ice-cold PBS to remove the unbound protein, the internalization signals were determined by a FACScan flow cytometer (BD Biosciences) with emission at 488 nm. For each sample, at least 10,000 individual cells were recorded using a dot plot combination of low-angle forward-scattered and right-angle side-scattered laser light (25). For ELISA, leukocytes were blocked with or without anti-TnSCARA5 Ab for 2 h. After incubating with the recombinant TnHMGB1 protein, the supernatant was collected at different time points and then coated onto microtiter wells (1:10 dilution) overnight at 4°C. OD450 was read as previously described (28). For trafficking analysis, the HEK293 cells transfected with pcDNA6-TnSCARA5 were stimulated with TnHMGB1 for 30 or 60 min and detected via immunofluorescence staining as previously described (22). Briefly, cells were fixed with 4% paraformaldehyde, permeabilized with 0.5% Triton X-100 for 15 min, and blocked with 5% goat serum for 1 h at room temperature. After incubating with rabbit anti-TnSCARA5 Ab (1:400 dilution) at 4°C overnight and washing three times with PBS, the cells were incubated with DyLight 549–conjugated goat anti-rabbit IgG for 1 h and stained with 0.1% DAPI (Invitrogen) for 3–5 min at room temperature. Fluorescence signals were observed under a confocal laser scanning microscope (LSM 510; Carl Zeiss) with a ×63 lens.

Colocalization of TnHMGB1 and TnSCARA5 with lysosome was determined in Tetraodon leukocytes and HEK293 cells. Tetraodon leukocytes were incubated with FITC-conjugated TnHMGB1 (10 μg/ml) at 28°C for 1 h and then stained with DMEM containing 75 nM LysoTracker Red (Beyotime) at 37°C for 1 h. After being incubated with 0.1% DAPI (Invitrogen) for 3–5 min at room temperature, the leukocytes were photomicrographed under a confocal laser scanning microscope. HEK293 cells transfected with pEGFP-TnSCARA5 were fixed with 4% paraformaldehyde at room temperature for 15 min, incubated with LysoTracker Red and DAPI, and then photomicrographed as described above.

A functional blockade assay was performed on mouse leukocytes to evaluate whether SCARA5 also plays an inhibitory role in CpG-ODN–induced inflammation in mammals. Leukocytes were freshly isolated from the spleen by Lympholyte-M (Sango), and the interlayer of cell suspensions was collected after centrifugation at 2200 rpm for 25 min. After blocking with different concentrations (5, 10, 15, and 20 μg/ml) of anti-mouse SCARA5 Ab and stimulation with CpG-SGIV-ODN6 (3 μg/ml), the mRNA levels of inflammatory cytokines (IL-1β, IL-6, IFN-γ, and TNF-α) in leukocytes were quantified by real-time PCR as previously described.

All experiments were replicated at least three times. All data are expressed as means ± SD. Statistical evaluations of the differences between the values of experimental groups were conducted using multiple Student t tests. A p value <0.05 was considered statistically significant.

With human HMGB1 and TLR9 sequences as queries, two homologous genes (TnHMGB1 and TnTLR9) were cloned from a model pufferfish (T. nigroviridis) through computational searching of the Tetraodon genome/EST databases. TnHMGB1 and TnTLR9 genes share overall conserved gene organization and genome synteny to their mammalian counterparts. TnHMGB1 cDNA consists of 649 bp with a 600-bp open reading frame that encodes 199 aa (Supplemental Fig. 1A; GenBank accession no. KU214880). TnHMGB1 protein is predicted to contain three conserved cysteines (Cys22, Cys44, and Cys106), an aspartic acid and glutamic acid–rich T box, and two DNA-binding domains (A and B boxes), all of which are functionally important in mammalian HMGB1 molecules. Multiple alignment analysis showed that TnHMGB1 shares high amino acid identities (74%) with its mammalian homologs (Supplemental Fig. 1A). TnTLR9 cDNA consists of 3175 bp with a 3123-bp open reading frame that encodes 1040 aa. TnTLR9 protein contains typical TLR structural hallmarks completely conserved throughout different species, including several leucine-rich repeat structures, a transmembrane domain, and a Toll/IL-1–resistance intracellular region (Supplemental Fig. 2A; GenBank accession no. KU214881). Phylogenetic analysis showed that TnHMGB1 and TnTLR9 are independently clustered with their homologs with a high bootstrap probability (Supplemental Figs. 1B, 2B). All of these observations suggest that TnHMGB1 and TnTLR9 are highly conserved molecules from fish to mammals throughout the vertebrate evolution. Additionally, TnSCARA5 was also identified as a highly conserved molecule with typical structural hallmarks of the SCARA5 family in our previous study (GenBank accession no. AFX69239.1) (4). Thus, fish is an attractive model for the functional studies of HMGB1, SCARA5, and TLR9 as a whole.

As an endogenous inflammatory factor, HMGB1 can be translocated from nucleus to cytoplasm and secrete from the cells under stimulation with PAMPs, such as LPS and CpG-ODNs in mammals. To investigate whether TnHMGB1 has such a feature, an induced secretion assay was performed by stimulating Tetraodon leukocytes with CpG-ODNs. For this purpose, we synthesized six viral genomic DNA fragments with typical CpG stimuli motifs derived from the SGIV genome (CpG-SGIV-ODN1–6) (Table I). The separated Tetraodon leukocytes were stimulated with these CpG-SGIV-ODNs, and the levels of inflammatory cytokines (IL-1β and IFN-γ) were detected by real-time PCR. Among the six CpG-SGIV-ODNs examined, CpG-SGIV-ODN6 has the strongest stimulatory effect (Fig. 1A). This CpG-SGIV-ODN6 fragment was then used in the subsequent experiments. HEK293T cells were transfected with pDsRed-TnHMGB1, and the subcellular localization of TnHMGB1 was detected by the DsRed fusion protein with red fluorescence. Results showed that TnHMGB1 is distributed in the cell nucleus without stimulation. After stimulation with CpG-SGIV-ODN6 for 8 h, TnHMGB1 migrated into the cytoplasm gradually (Fig. 1D) and secreted into the cultural supernatant as detected by the Western blot analysis (Fig. 1B, 1C).

FIGURE 1.

Translocation and secretion of TnHMGB1 under CpG-SGIV-ODN stimulation. (A) Evaluation of the stimulatory effect of the six synthesized CpG-SGIV-ODN fragments. Leukocytes extracted from the Tetraodon spleen and kidney tissues were stimulated with the six CpG-SGIV-ODNs. Real-time PCR was conducted to detect the upregulated expression of IL-1β and IFN-γ. The data are presented as means ± SD from at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls. (B) Western blot for the detection of the reactivity and specificity of the prepared polyclonal Ab (anti-TnHMGB1) against the TnHMGB1 recombinant protein in the whole protein extract of Escherichia coli induced by isopropyl β-d-thiogalactoside (anti-TnHMGB1 group). The control group (control) was not incubated with any Ab. (C) Western blot for the detection of TnHMGB1 protein secreted outside of the leukocytes upon stimulation with CpG-SGIV-ODN6. Leukocytes isolated from Tetraodon spleen and kidney tissues were stimulated with CpG-SGIV-ODN6 for 8 h, and the culture supernatant was collected and ultrafiltered for analysis. The control group received no stimulation of CpG-SGIV-ODN6. (D) Translocation of TnHMGB1 in HEK293T cells. HEK293T cells were transfected with pDsRED-TnHMGB1 plasmid, fixed, and observed under a laser scanning confocal microscope (LSM 510). Red fluorescence is distributed in the nuclei of the cells that received no stimulation with CpG-SGIV-ODN6 (control) and translocates into the cytoplasm of the cells subjected to CpG-SGIV-ODN6 or LPS (positive control) stimulation for 8 h. Scale bars, 10 μm.

FIGURE 1.

Translocation and secretion of TnHMGB1 under CpG-SGIV-ODN stimulation. (A) Evaluation of the stimulatory effect of the six synthesized CpG-SGIV-ODN fragments. Leukocytes extracted from the Tetraodon spleen and kidney tissues were stimulated with the six CpG-SGIV-ODNs. Real-time PCR was conducted to detect the upregulated expression of IL-1β and IFN-γ. The data are presented as means ± SD from at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls. (B) Western blot for the detection of the reactivity and specificity of the prepared polyclonal Ab (anti-TnHMGB1) against the TnHMGB1 recombinant protein in the whole protein extract of Escherichia coli induced by isopropyl β-d-thiogalactoside (anti-TnHMGB1 group). The control group (control) was not incubated with any Ab. (C) Western blot for the detection of TnHMGB1 protein secreted outside of the leukocytes upon stimulation with CpG-SGIV-ODN6. Leukocytes isolated from Tetraodon spleen and kidney tissues were stimulated with CpG-SGIV-ODN6 for 8 h, and the culture supernatant was collected and ultrafiltered for analysis. The control group received no stimulation of CpG-SGIV-ODN6. (D) Translocation of TnHMGB1 in HEK293T cells. HEK293T cells were transfected with pDsRED-TnHMGB1 plasmid, fixed, and observed under a laser scanning confocal microscope (LSM 510). Red fluorescence is distributed in the nuclei of the cells that received no stimulation with CpG-SGIV-ODN6 (control) and translocates into the cytoplasm of the cells subjected to CpG-SGIV-ODN6 or LPS (positive control) stimulation for 8 h. Scale bars, 10 μm.

Close modal

The induced secretion of TnHMGB1 by CpG-SGIV-ODN6 implies its involvement in CpG-ODN–induced inflammation. Emerging studies have suggested that HMGB1 is an inflammatory accessory molecule that confers stimulatory function associated with various proinflammatory stimulants, such as LPS, IL-1β, Pam3CSK4, and CpG-ODN (14). HMGB1 has also been found as a CpG-ODN-binding protein that initiates TLR9 signaling pathway (20). In the present study, we explored whether TnHMGB1 can act in synergy with CpG-ODN to augment the inflammatory responses. We initially demonstrated the strong ability of TnHMGB1 protein to interact with CpG-SGIV-ODN6 through a streptavidin pulldown assay (see Fig. 9). Leukocytes isolated from Tetraodon spleen and kidney tissues were treated with TnHMGB1 and/or CpG-SGIV-ODN6 in different combinations (TnHMGB1 or CpG-SGIV-ODN6 alone, and TnHMGB1 plus CpG-SGIV-ODN6), and inflammatory cytokines (IL-1β, IL-6, TNF-α, and IFN-γ) were examined after stimulation for 4 h. The expression levels of inflammatory cytokines were significantly (p < 0.01) increased (>5-fold increase for IFN-γ and TNF-α, and >10-fold increase for IL-1β and IL-6) in the TnHMGB1 and CpG-SGIV-ODN6 combined-stimulating group compared with that in the TnHMGB1 or CpG-SGIV-ODN6 mono-stimulating group (Fig. 2). Thus, TnHMGB1 dramatically augmented the CpG-ODN–induced inflammatory responses.

FIGURE 9.

TnSCARA5 preferentially interacts with TnHMGB1 rather than with the TnHMGB1–CpG-ODN complex. HEK293T cells were transfected with TnSCARA5 plasmid and incubated with the biotinylated CpG-SGIV-ODN6–TnHMGB1 complex and with biotinylated CpG-SGIV-ODN6 (as positive control). The association of biotinylated CpG-SGIV-ODN6 with recombinant TnHMGB1 protein was also set as control. CpG-SGIV-ODN6 was synthesized with a phosphodiester backbone. The samples were precipitated with streptavidin beads and analyzed by Western blot. Immunoblots of the pulldowns show that no detectable TnSCARA5 was precipitated in the TnHMGB1CpG-SGIV-ODN6 pulldown assay but in both of the control groups.

FIGURE 9.

TnSCARA5 preferentially interacts with TnHMGB1 rather than with the TnHMGB1–CpG-ODN complex. HEK293T cells were transfected with TnSCARA5 plasmid and incubated with the biotinylated CpG-SGIV-ODN6–TnHMGB1 complex and with biotinylated CpG-SGIV-ODN6 (as positive control). The association of biotinylated CpG-SGIV-ODN6 with recombinant TnHMGB1 protein was also set as control. CpG-SGIV-ODN6 was synthesized with a phosphodiester backbone. The samples were precipitated with streptavidin beads and analyzed by Western blot. Immunoblots of the pulldowns show that no detectable TnSCARA5 was precipitated in the TnHMGB1CpG-SGIV-ODN6 pulldown assay but in both of the control groups.

Close modal
FIGURE 2.

Synergistic effect of TnHMGB1 on CpG-ODN–induced inflammation. Leukocytes isolated from Tetraodon spleen and kidney tissues were stimulated with specified concentrations of TnHMGB1 and CpG-SGIV-ODN6 in different combinations. The proinflammatory cytokines IL-1β, IL-6, TNF-α, and IFN-γ were quantified by real-time PCR after stimulation for 4 h. Expression levels of the proinflammatory cytokines were significantly increased in TnHMGB1 and CpG-SGIV-ODN6 combined-stimulating groups. The data are presented as means ± SD of at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

FIGURE 2.

Synergistic effect of TnHMGB1 on CpG-ODN–induced inflammation. Leukocytes isolated from Tetraodon spleen and kidney tissues were stimulated with specified concentrations of TnHMGB1 and CpG-SGIV-ODN6 in different combinations. The proinflammatory cytokines IL-1β, IL-6, TNF-α, and IFN-γ were quantified by real-time PCR after stimulation for 4 h. Expression levels of the proinflammatory cytokines were significantly increased in TnHMGB1 and CpG-SGIV-ODN6 combined-stimulating groups. The data are presented as means ± SD of at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

Close modal

For further confirmation, an in vivo TnHMGB1 knockdown assay was performed by using an LV-based siRNA delivery system. Three siRNAs targeting different TnHMGB1 regions were selected from an online design program. Of the three generated constructs, pSUPER-TnHMGB1–3 is the most effective (80%) in inducing TnHMGB1 mRNA and protein degradation in HKE293T cells (Fig. 3B). This construct was used to produce the recombinant LV. The generated lentivirus (LV-TnHMGB1) reached a titer of ∼1 × 104 TU/μl by flow cytometry examination (Fig. 3A) and showed a highly infectious efficacy as determined by most GFP+ HEK293T cells (Fig. 3C). To detect the in vivo interference efficiency for TnHMGB1, the LV was administered into Tetraodon for 3 d, and real-time PCR was performed to test the silencing of TnHMGB1 in PBLs and HKLs. The expression of TnSCARA5 was detected to exclude cross-reaction. The results showed that TnHMGB1 expression can be sharply downregulated by 68 and 75% in PBLs and HKLs, respectively (Fig. 3D). Subsequently, CpG-SGIV-ODN6 was administrated into the TnHMGB1-depleted fish, and inflammatory cytokines (IL-1β and IFN-γ) were examined in leukocytes, as well as in spleen and kidney tissues. As expected, TnHMGB1 depletion dramatically (p < 0.01) inhibited the expression of inflammatory cytokines in response to CpG-SGIV-ODN6 stimulation (Fig. 3E), which demonstrates the synergy of TnHMGB1 in CpG-ODN–induced inflammation.

FIGURE 3.

Knockdown of TnHMGB1 exhibits decreased inflammatory response upon CpG-SGIV-ODN6 stimulation. (A) Titer determinations for LV against TnHMGB1 (LV-TnHMGB1). The LV-TnHMGB1 titer was assessed by flow cytometry in accordance with the percentage of GFP+ HEK293T cells after exposure to different dilutions of the virus. The shaded graphs show background fluorescence in the control cells without LV infection. The number above the bracketed line indicates the percentage of GFP+ cells in image. (B) Screening of effective siRNAs against TnHMGB1. Three designed siRNAs targeting different regions of TnHMGB1 mRNA were independently inserted into the pSUPER vector (siRNA1–3). The HEK293T cells were cotransfected with siRNA1–3 or the control plasmid (pSUPER vector harboring the scrambled siRNA) with the overexpression plasmid pCMV-tag2B-TnHMGB1. The efficacy of siRNAs was measured by real-time PCR and Western blot. (C) Detection of the viral infection efficacy in HEK293T cells determined by GFP fluorescence under a fluorescence microscope (Zeiss Axiovert 40 CFL, original magnification ×400). Most HEK293T cells strongly expressed GFP, demonstrating that the constructed LV exhibits a highly infectious efficacy. (D) Evaluation of the silencing efficiency of LV-TnHMGB1 in vivo through real-time PCR. The expression levels of TnHMGB1 in PBLs and HKLs were sharply downregulated. (E) Knockdown of TnHMGB1 exhibiting less proinflammatory cytokines of IL-1β and IFN-γ in response to CpG-SGIV-ODN6 stimulation. The relative gene expression of IL-1β and IFN-γ was quantified by real-time PCR and normalized to β-actin. Results are expressed as the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

FIGURE 3.

Knockdown of TnHMGB1 exhibits decreased inflammatory response upon CpG-SGIV-ODN6 stimulation. (A) Titer determinations for LV against TnHMGB1 (LV-TnHMGB1). The LV-TnHMGB1 titer was assessed by flow cytometry in accordance with the percentage of GFP+ HEK293T cells after exposure to different dilutions of the virus. The shaded graphs show background fluorescence in the control cells without LV infection. The number above the bracketed line indicates the percentage of GFP+ cells in image. (B) Screening of effective siRNAs against TnHMGB1. Three designed siRNAs targeting different regions of TnHMGB1 mRNA were independently inserted into the pSUPER vector (siRNA1–3). The HEK293T cells were cotransfected with siRNA1–3 or the control plasmid (pSUPER vector harboring the scrambled siRNA) with the overexpression plasmid pCMV-tag2B-TnHMGB1. The efficacy of siRNAs was measured by real-time PCR and Western blot. (C) Detection of the viral infection efficacy in HEK293T cells determined by GFP fluorescence under a fluorescence microscope (Zeiss Axiovert 40 CFL, original magnification ×400). Most HEK293T cells strongly expressed GFP, demonstrating that the constructed LV exhibits a highly infectious efficacy. (D) Evaluation of the silencing efficiency of LV-TnHMGB1 in vivo through real-time PCR. The expression levels of TnHMGB1 in PBLs and HKLs were sharply downregulated. (E) Knockdown of TnHMGB1 exhibiting less proinflammatory cytokines of IL-1β and IFN-γ in response to CpG-SGIV-ODN6 stimulation. The relative gene expression of IL-1β and IFN-γ was quantified by real-time PCR and normalized to β-actin. Results are expressed as the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

Close modal

To evaluate whether TnSCARA5 is involved in TnHMGB1/CpG-ODN–mediated inflammatory responses, an induced expression assay of TnSCARA5 in response to TnHMGB1 stimulation was initially performed. Experimental fish were injected with TnHMGB1 protein for 18 h, and the expression of TnSCARA5 was detected by real-time PCR. The results showed that the transcripts of TnSCARA5 were significantly (p < 0.01) upregulated in most tissues examined, with an 8- to 12-fold increase in intestine, kidney, brain, and skin, as well as a 2- to 3-fold increase in gill and muscle (Fig. 4A). Additionally, the expression of TnSCARA5 was also increased in kidney and spleen leukocytes in response to TnHMGB1 stimulation in a dose-dependent manner (Fig. 4B). These observations provide initial insights into the involvement of TnSCARA5 in TnHMGB1/CpG-ODN–mediated inflammation.

FIGURE 4.

Induced expression of TnSCARA5 upon TnHMGB1 stimulation in different tissues and leukocytes of Tetraodon. The expression of TnSCARA5 was induced by TnHMGB1 at 10 μg/fish in different tissues (A) and at 5, 10, and 15 μg/ml in leukocytes (B) and determined by real-time PCR. All data are presented as the normalized mean fold induction ± SD over the unstimulated level from three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

FIGURE 4.

Induced expression of TnSCARA5 upon TnHMGB1 stimulation in different tissues and leukocytes of Tetraodon. The expression of TnSCARA5 was induced by TnHMGB1 at 10 μg/fish in different tissues (A) and at 5, 10, and 15 μg/ml in leukocytes (B) and determined by real-time PCR. All data are presented as the normalized mean fold induction ± SD over the unstimulated level from three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

Close modal

The involvement of TnSCARA5 in TnHMGB1/CpG-ODN–induced inflammation was determined in HEK293T cells and zebrafish embryos. No significant NF-κB activation was observed in HEK293T cells in response to CpG-SGIV-ODN6 stimulation because of the low TLR9 expression in this cell line. With the exotic expression of TnTLR9 in HEK293T cells, the NF-κB activation was significantly (p < 0.05) upregulated in cells under stimulation. However, with the co-overexpression of TnTLR9 and TnSCARA5 for 24 h, the NF-κB activation was significantly (p < 0.05) reduced to the basal level after stimulation for 5 h (Fig. 5A). The zebrafish embryo model showed that the administration of different concentrations (0.25, 0.5, 0.75, and 1 μg/μl) of CpG-SGIV-ODN6 induced NF-κB luciferase activation in a dose-dependent manner (>4- to 10-fold increase) (Fig. 5B). Nevertheless, coinjection of CpG-SGIV-ODN6 (1 μg/μl) with different concentrations (50, 75, 100, and 125 ng/μl) of pCDNA6-TnSCARA5 significantly (p < 0.01) inhibited the NF-κB activation in a dose-dependent manner (>3- to 5-fold decrease) (Fig. 5C). Correspondingly, the expression of inflammatory cytokines (IL-1β, IL-6, IFN-γ, and TNF-α) was significantly (p < 0.05) increased (>4-fold increase) in embryos stimulated with CpG-SGIV-ODN6 (0.5 and 1 μg/μl) (Fig. 5D–G); this increase was dramatically (p < 0.05) inhibited in pCDNA6-TnSCARA5 (50, 75, and 100 ng/μl)–administrated embryos in a dose-dependent manner under stimulation with CpG-SGIV-ODN6 (1 μg/μl) for 16 h (Fig. 5H, 5I).

FIGURE 5.

Negative regulation of TnSCARA5 in TnHMGB1/CpG-ODN–induced inflammation. (A) TnSCARA5 negatively regulates NF-κB activation in vitro. HEK293T cells were transfected with specified plasmids as well as NF-κB luciferase and pGL-TK Renilla luciferase reporter plasmids. Then, 24 h after transfection, the cells were treated with CpG-SGIV-ODN6 (5 μg/ml) for 5 h. The firefly and Renilla luciferase activities were assayed and expressed as the fold induction relative to the control. (B and C) Induced activation of NF-κB and negative regulation of TnSCARA5 for NF-κB activation in vivo. One-cell-stage zebrafish embryos were coinjected with NF-κB luciferase and pRL-TK Renilla luciferase reporter plasmids plus different concentrations of CpG-SGIV-ODN6 (B) or with various concentrations of pcDNA6-TnSCARA5 plasmids under CpG-SGIV-ODN6 (l μg/μl) stimulation (C). After microinjection for 24 h, the luciferase activities were assayed. (DI) Induced expression of proinflammatory cytokines (IL-1β, IL-6, TNF-α, and IFN-γ) and negative regulation of TnSCARA5 for the expression of these cytokines. One-cell-stage zebrafish embryos were injected with CpG-SGIV-ODN6 at 0.5 and 1 μg/μl. After microinjection for different times, mRNA levels of IL-1β (D), IL-6 (E), IFN-γ (F), and TNF-α (G) were quantified. In parallel, one-cell-stage zebrafish embryos were coinjected with various concentrations of pcDNA6-TnSCARA5 under CpG-SGIV-ODN6 (1 μg/μl) stimulation. After microinjection for 16 h, mRNA levels of IL-1β, IL-6, TNF-α, and IFN-γ were quantified (H and I). Results are expressed as mean ± SD of three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

FIGURE 5.

Negative regulation of TnSCARA5 in TnHMGB1/CpG-ODN–induced inflammation. (A) TnSCARA5 negatively regulates NF-κB activation in vitro. HEK293T cells were transfected with specified plasmids as well as NF-κB luciferase and pGL-TK Renilla luciferase reporter plasmids. Then, 24 h after transfection, the cells were treated with CpG-SGIV-ODN6 (5 μg/ml) for 5 h. The firefly and Renilla luciferase activities were assayed and expressed as the fold induction relative to the control. (B and C) Induced activation of NF-κB and negative regulation of TnSCARA5 for NF-κB activation in vivo. One-cell-stage zebrafish embryos were coinjected with NF-κB luciferase and pRL-TK Renilla luciferase reporter plasmids plus different concentrations of CpG-SGIV-ODN6 (B) or with various concentrations of pcDNA6-TnSCARA5 plasmids under CpG-SGIV-ODN6 (l μg/μl) stimulation (C). After microinjection for 24 h, the luciferase activities were assayed. (DI) Induced expression of proinflammatory cytokines (IL-1β, IL-6, TNF-α, and IFN-γ) and negative regulation of TnSCARA5 for the expression of these cytokines. One-cell-stage zebrafish embryos were injected with CpG-SGIV-ODN6 at 0.5 and 1 μg/μl. After microinjection for different times, mRNA levels of IL-1β (D), IL-6 (E), IFN-γ (F), and TNF-α (G) were quantified. In parallel, one-cell-stage zebrafish embryos were coinjected with various concentrations of pcDNA6-TnSCARA5 under CpG-SGIV-ODN6 (1 μg/μl) stimulation. After microinjection for 16 h, mRNA levels of IL-1β, IL-6, TNF-α, and IFN-γ were quantified (H and I). Results are expressed as mean ± SD of three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

Close modal

Furthermore, the function of TnSCARA5 in TnHMGB1/CpG-ODN–mediated inflammation was examined by an in vivo knockdown assay. Among the five constructs (pSUPER-TnSCARA5-1–5) generated with five siRNAs targeting different regions of TnSCARA5, pSUPER-TnSCARA5-2 has the most effective silencing (63%) for TnSCARA5 (Fig. 6B). This construct was further used to produce the recombinant LV (LV-TnSCARA5) with a titer of ∼104 TU/μl (Fig. 6A) and high infectious efficacy in HEK293T cells (Fig. 6C). An in vivo interference assay showed that the TnSCARA5 expression declined by 64 and 60% in PBLs and HKLs, respectively, after the fish received the LV-TnSCARA5 for 3 d (Fig. 6D). Accordingly, the expression of inflammatory cytokines (IL-1β and IFN-γ) in TnSCARA5-depleted leukocytes, kidney, and spleen tissues was overall increased by 38–64% compared with that of the control group under stimulation of CpG-SGIV-ODN6 (Fig. 6E). These findings again demonstrated the inhibitory role of TnSCARA5 in HMGB1/CpG-ODN–elicited inflammation.

FIGURE 6.

Knockdown of TnSCARA5 exhibits stronger inflammatory responses with CpG-SGIV-ODN6 stimulation. (A) Titer determination for LV against TnSCARA5 (LV-TnSCARA5). The LV-TnSCARA5 titer was assessed by flow cytometry on the basis of the percentage of GFP+ HEK293T cells exposed to different dilutions of the virus. The shaded graphs show background fluorescence on control cells without LV infection. The number above the bracketed line indicates the percentage of GFP+ cells in each image. (B) Screening of the most effective siRNAs against TnSCARA5. Five designed siRNAs targeting different regions of TnSCARA5 mRNA were independently inserted into the pSUPER vector (siRNA1–5). The HEK293T cells were cotransfected with siRNA1–5 or the control plasmid (pSUPER vector harboring the scrambled siRNA) with the TnSCARA5 overexpression plasmid. The efficacy of the siRNAs was measured by real-time PCR and Western blot. (C) Detection of the viral infection efficacy in HEK293T cells as indicated by GFP fluorescence under a fluorescence microscope (Zeiss Axiovert 40 CFL, original magnification ×400). Most HEK293T cells strongly expressed GFP, demonstrating that the constructed LV possessed a highly infectious efficacy. (D) Evaluation of the silencing efficiency of LV-TnSCARA5 in vivo through real-time PCR analysis. The expression of TnSCARA5 in PBLs and HKLs was detected by real-time PCR. (E) Knockdown of TnSCARA5 exhibiting increased proinflammatory cytokine levels of IL-1β and IFN-γ with CpG-SGIV-ODN6 stimulation. The relative mRNA expression of IL-1β and IFN-γ was quantified by real-time PCR and normalized to β-actin. Results are expressed as the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

FIGURE 6.

Knockdown of TnSCARA5 exhibits stronger inflammatory responses with CpG-SGIV-ODN6 stimulation. (A) Titer determination for LV against TnSCARA5 (LV-TnSCARA5). The LV-TnSCARA5 titer was assessed by flow cytometry on the basis of the percentage of GFP+ HEK293T cells exposed to different dilutions of the virus. The shaded graphs show background fluorescence on control cells without LV infection. The number above the bracketed line indicates the percentage of GFP+ cells in each image. (B) Screening of the most effective siRNAs against TnSCARA5. Five designed siRNAs targeting different regions of TnSCARA5 mRNA were independently inserted into the pSUPER vector (siRNA1–5). The HEK293T cells were cotransfected with siRNA1–5 or the control plasmid (pSUPER vector harboring the scrambled siRNA) with the TnSCARA5 overexpression plasmid. The efficacy of the siRNAs was measured by real-time PCR and Western blot. (C) Detection of the viral infection efficacy in HEK293T cells as indicated by GFP fluorescence under a fluorescence microscope (Zeiss Axiovert 40 CFL, original magnification ×400). Most HEK293T cells strongly expressed GFP, demonstrating that the constructed LV possessed a highly infectious efficacy. (D) Evaluation of the silencing efficiency of LV-TnSCARA5 in vivo through real-time PCR analysis. The expression of TnSCARA5 in PBLs and HKLs was detected by real-time PCR. (E) Knockdown of TnSCARA5 exhibiting increased proinflammatory cytokine levels of IL-1β and IFN-γ with CpG-SGIV-ODN6 stimulation. The relative mRNA expression of IL-1β and IFN-γ was quantified by real-time PCR and normalized to β-actin. Results are expressed as the mean ± SD of three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

Close modal

To determine whether TnSCARA5 acts as a recognition receptor for TnHMGB1, the association of TnSCARA5 with TnHMGB1 was examined by qELISA and CoIP. For qELISA, recombinant TnSCARA5 and TnHMGB1 proteins were purified by Ni-NTA agarose affinity chromatography (Fig. 7A, 7B), and various concentrations of these two proteins were used in different combinations. As expected, increased binding activities between TnSCARA5 and TnHMGB1 were clearly detected with the increase of TnHMGB1 (from 8 to 32 nM, or from 8 to 64 nM) to a designated concentration of TnSCARA5 at 36 or 72 nM, respectively. In contrast, no association was detected in the control groups, in which TnHMGB1 was replaced by various concentrations of nonrelated BSA proteins (Fig. 7D). These results indicated that TnSCARA5 interacts with TnHMGB1 in a dose-dependent manner and at a ratio of ∼1:1, as estimated by the saturated concentration of TnHMGB1 for the interaction with TnSCARA5 in the binding curves. For CoIP, the pcDNA6-TnSCARA5 construct was transfected into HEK293T cells, and two different concentrations (150 and 300 nM) of TnHMGB1 protein were incubated with the cells for 20 min at 24 h posttransfection. The associated TnSCARA5 and TnHMGB1 proteins were precipitated with Ab against Myc-tag fused to TnSCARA5 and detected by Western blot analysis with Ab against His-tag fused to TnHMGB1. Correspondingly, empty pcDNA6.0 construct–transfected cells incubated with the same amounts of TnHMGB1 protein were used as negative control. As expected, the interaction signals from TnSCARA5 with TnHMGB1 were clearly detected in the experimental groups but not in the controls (Fig. 7E). In agreement with the qELISA observations, the association of TnSCARA5 with TnHMGB1 was significantly enhanced with the increase of the administrated TnHMGB1 proteins (Fig. 7F).

FIGURE 7.

Determination of the association between TnSCARA5 and TnHMGB1. (AC) SDS-PAGE analyses show the recombinant TnSCARA5 (A), TnHMGB1 (B), and TnHMGB1 mutants (C) expressed in E. coli as a soluble protein. The recombinant proteins were purified by Ni-NTA agarose affinity chromatography. (D) In vitro binding of TnSCARA5 to TnHMGB1 determined by quantitative ELISA. Different concentrations of purified TnHMGB1 were coated onto 96-well plates. The recombinant TnSCARA5 protein (36 or 72 nM) was added and probed with secondary Ab conjugated to HRP against anti-TnSCARA5 Ab. (E) Binding of TnSCARA5 to TnHMGB1 demonstrated by CoIP. HEK293T cells were transfected with pcDNA6-Myc/His-TnSCARA5 (or empty control vector) and incubated with TnHMGB1, followed by immunoprecipitation with Myc tag–Ab and detected with His/Myc tag–Ab. TnSCARA5 in the precipitations can be detected by Myc and His tag–Abs, whereas TnHMGB1 can only be detected by the His tag–Ab. The cell lysate before precipitation was employed as positive control. (F) Association of TnSCARA5 with TnHMGB1. Two different concentrations (150 and 300 nM) of TnHMGB1 were incubated to verify the association. (G) Association of TnSCARA5 with TnHMGB1 mutants. Slight interaction of TnSCARA5 with TnHMGB1-mutA or TnHMGB1-mutB was detected; however, the lack of the T box of TnHMGB1 enhanced the association. (H and I) Demonstration of the inhibiting effect of T box by a competitive binding assay. The TnHMGB1-mutT and TnHMGB1 combined (1:1) group (a), the TnHMGB1-mutT group (b), and the TnHMGB1 group (c) were precipitated and detected by Western blot (H). The gray value of signal intensity was calculated and depicted in bar graph (I), showing that the increased binding of TnHMGB1-mutT to TnSCARA5 decreased by replacing 50% of the TnHMGB1-mutT with wild-type TnHMGB1. The data are presented as means ± SD of at least three independent experiments. The black lines indicate where parts of the images were joined.

FIGURE 7.

Determination of the association between TnSCARA5 and TnHMGB1. (AC) SDS-PAGE analyses show the recombinant TnSCARA5 (A), TnHMGB1 (B), and TnHMGB1 mutants (C) expressed in E. coli as a soluble protein. The recombinant proteins were purified by Ni-NTA agarose affinity chromatography. (D) In vitro binding of TnSCARA5 to TnHMGB1 determined by quantitative ELISA. Different concentrations of purified TnHMGB1 were coated onto 96-well plates. The recombinant TnSCARA5 protein (36 or 72 nM) was added and probed with secondary Ab conjugated to HRP against anti-TnSCARA5 Ab. (E) Binding of TnSCARA5 to TnHMGB1 demonstrated by CoIP. HEK293T cells were transfected with pcDNA6-Myc/His-TnSCARA5 (or empty control vector) and incubated with TnHMGB1, followed by immunoprecipitation with Myc tag–Ab and detected with His/Myc tag–Ab. TnSCARA5 in the precipitations can be detected by Myc and His tag–Abs, whereas TnHMGB1 can only be detected by the His tag–Ab. The cell lysate before precipitation was employed as positive control. (F) Association of TnSCARA5 with TnHMGB1. Two different concentrations (150 and 300 nM) of TnHMGB1 were incubated to verify the association. (G) Association of TnSCARA5 with TnHMGB1 mutants. Slight interaction of TnSCARA5 with TnHMGB1-mutA or TnHMGB1-mutB was detected; however, the lack of the T box of TnHMGB1 enhanced the association. (H and I) Demonstration of the inhibiting effect of T box by a competitive binding assay. The TnHMGB1-mutT and TnHMGB1 combined (1:1) group (a), the TnHMGB1-mutT group (b), and the TnHMGB1 group (c) were precipitated and detected by Western blot (H). The gray value of signal intensity was calculated and depicted in bar graph (I), showing that the increased binding of TnHMGB1-mutT to TnSCARA5 decreased by replacing 50% of the TnHMGB1-mutT with wild-type TnHMGB1. The data are presented as means ± SD of at least three independent experiments. The black lines indicate where parts of the images were joined.

Close modal

To further determine which domain of TnHMGB1 is involved in the association of TnSCARA5 with TnHMGB1, three mutant TnHMGB1 proteins with deletion of A, B, and T boxes (named TnHMGB1-mutA, TnHMGB1-mutB, and TnHMGB1-mutT, respectively) were constructed (Fig. 7C), and the interactions of TnSCARA5 with these mutants were examined by CoIP. Minimal interaction of TnSCARA5 with TnHMGB1-mutA or TnHMGB1-mutB was detected compared with that of TnSCARA5 with wild-type TnHMGB1. Notably, the absence of T box in TnHMGB1 did not reduce but instead promoted the association of TnHMGB1 with TnSCARA5 (Fig. 7G). To confirm this observation, a competitive binding assay was performed to detect the decline of the increased binding of TnHMGB1-mutT to TnSCARA5 by replacing 50% of the TnHMGB1-mutT with wild-type TnHMGB1. As expected, the enhanced interaction in the TnHMGB1-mutT plus TnSCARA5 group was dramatically decreased in the TnHMGB1 and TnHMGB1-mutT (1:1) plus TnSCARA5 group (Fig. 7H, 7I). This result indicated that the absence of T box promoted the binding capacity of TnHMGB1 to TnSCARA5. Thus, T box in TnHMGB1 may play a negative regulatory role in the association of TnSCARA5 with TnHMGB1, and both A and B boxes may be necessary for the association.

Three functionally important cysteines exist in the A (Cys22 and Cys44) and B boxes (Cys106). Previous studies have shown that HMGB1 can switch its redox states (from a fully reduced form to a disulfide-bonded form) by forming a reversible intracellular disulfide bond between Cys22 and Cys44, and different redox forms exercise different functions. The three cysteines in HMGB1 can also be terminally oxidized to sulfonates (fully oxidized form) in an irreversible manner by treating the protein with H2O2. The disulfide-bonded form has a more compact folding polypeptide chain than does the fully reduced or fully oxidized form; hence, this form would have an increased electrophoretic mobility under nonreducing conditions (32). To evaluate which redox form was possessed by TnHMGB1, the recombinant TnHMGB1 protein pretreated with or without DTT (5 mM for 1 h) was detected in the presence or absence of 2-ME (0.5 mM for 10 min). The results of Coomassie brilliant blue staining (Fig. 8A) and Western blot analysis (Fig. 8B) clearly showed that the TnHMGB1 protein had an increased electrophoretic mobility under nonreducing conditions. This result suggests the presence of a disulfide bond in the recombinant TnHMGB1. To explore whether the interaction of TnHMGB1 with TnSCARA5 is associated with the redox state (disulfide bonded, fully reduced, or fully oxidized) of TnHMGB1, CoIP assays were carried out to determine the binding efficiency of the three forms of TnHMGB1 to TnSCARA5. The fully reduced and fully oxidized forms of TnHMGB1 were prepared by pretreating the recombinant TnHMGB1 protein (disulfide-bonded form) with DTT and H2O2 as previously described. Results showed that the association signal in the fully oxidized TnHMGB1-administered CoIP sample was significantly weaker (p < 0.05) than that of the two other samples administered with fully reduced TnHMGB1 and untreated TnHMGB1 with a disulfide bond. This result suggests that the fully oxidized TnHMGB1 had a significantly weaker combination with TnSCARA5 than did the two other forms of TnHMGB1. However, no significant difference in association signals was found between the fully reduced and disulfide-bonded TnHMGB1-administered samples. This result suggests similar binding capacities of the two TnHMGB1 forms to TnSCARA5 (Fig. 8C, 8D). The specific redox forms of TnHMGB1 that were used in the binding assays were detected from the CoIP complexes by their electrophoretic patterns under reducing and nonreducing conditions through Western blot analysis. As expected, TnHMGB1 pretreated with DTT (fully reduced) or H2O2 (fully oxidized) migrated as a major band with a lower electrophoretic mobility both under reducing and nonreducing conditions. In contrast, disulfide-bonded TnHMGB1 migrated under nonreducing conditions as a single band with an increased electrophoretic mobility (Fig. 8E) and shifted under reducing conditions to the same position as fully reduced or fully oxidized TnHMGB1 did (Fig. 8C). These results suggest that the different redox forms of TnHMGB1 produced remained specific during the binding assays.

FIGURE 8.

Effect of the redox states of TnHMGB1 on its association to TnSCARA5. Coomassie staining (A) and Western blot analysis (B) showed that the recombinant TnHMGB1 protein pretreated with DTT migrated slower in the presence or absence of 2-ME (β-ME) and that the TnHMGB1 protein without DTT treatment migrated faster in the absence of β-ME, suggesting the presence of a disulfide bond in the recombinant TnHMGB1. (B) is an overlaid picture from the same blot film detected under chemiluminescence and transmitting light, respectively. The black line indicates where part of the image was joined. (CF) Interactions of different redox states of TnHMGB1 with TnSCARA5 were detected by CoIP (C and E). The gray value of the relative signal intensity (TnHMGB1/TnSCARA5) was calculated using ImageJ and is shown in (D) and (F). Oxidized TnHMGB1 had a significantly weaker combination with TnSCARA5 than did the two other forms. Specific redox forms of TnHMGB1 from the CoIP complexes were detected by their electrophoretic patterns under reducing (C) and nonreducing (E) conditions. Disulfide-bonded TnHMGB1 migrated with an increased electrophoretic mobility under nonreducing conditions and shifted to the same position as did fully reduced or fully oxidized TnHMGB1.

FIGURE 8.

Effect of the redox states of TnHMGB1 on its association to TnSCARA5. Coomassie staining (A) and Western blot analysis (B) showed that the recombinant TnHMGB1 protein pretreated with DTT migrated slower in the presence or absence of 2-ME (β-ME) and that the TnHMGB1 protein without DTT treatment migrated faster in the absence of β-ME, suggesting the presence of a disulfide bond in the recombinant TnHMGB1. (B) is an overlaid picture from the same blot film detected under chemiluminescence and transmitting light, respectively. The black line indicates where part of the image was joined. (CF) Interactions of different redox states of TnHMGB1 with TnSCARA5 were detected by CoIP (C and E). The gray value of the relative signal intensity (TnHMGB1/TnSCARA5) was calculated using ImageJ and is shown in (D) and (F). Oxidized TnHMGB1 had a significantly weaker combination with TnSCARA5 than did the two other forms. Specific redox forms of TnHMGB1 from the CoIP complexes were detected by their electrophoretic patterns under reducing (C) and nonreducing (E) conditions. Disulfide-bonded TnHMGB1 migrated with an increased electrophoretic mobility under nonreducing conditions and shifted to the same position as did fully reduced or fully oxidized TnHMGB1.

Close modal

Finally, we explored whether TnSCARA5 possesses the ability to associate with the TnHMGB1–CpG-ODN complex in addition to its interaction with the TnHMGB1 protein. For this purpose, TnSCARA5 was overexpressed in HEK293T cells and then incubated with the TnHMGB1–CpG-SGIV-ODN6 complex (preincubated TnHMGB1 protein with biotin-labeled CpG-SGIV-ODN6 at 4°C for 16 h), biotin-labeled CpG-SGIV-ODN6, and His-tagged TnHMGB1 protein (as positive controls). The TnSCARA5 was then precipitated with streptavidin beads or Abs against the Myc-tag fused to the TnSCARA5 protein. Western blot analysis showed that no detectable TnSCARA5 was precipitated in the TnHMGB1CpG-SGIV-ODN6 pull-down assay. In contrast, obvious TnSCARA5 precipitates were detected in both of the two control assays (Supplemental Fig. 3A). Considering that the CpG-ODNs with a phosphorothioate backbone might have unspecific binding, we synthesized CpG-SGIV-ODN6 with a phosphodiester backbone and verified its binding abilities to TnHMGB1 and TnSCARA5 proteins. Similar to CpG-SGIV-ODN6 with a phosphorothioate backbone, CpG-SGIV-ODN6 with a phosphodiester backbone also showed strong binding abilities to TnHMGB1 and TnSCARA5 proteins. Meanwhile, as with the TnHMGB1–CpG-SGIV-ODN6 (with a phosphorothioate backbone) complex, the TnHMGB1–CpG-SGIV-ODN6 (with a phosphodiester backbone) complex showed no association to TnSCARA5 protein (Fig. 9). The above-mentioned observations suggest that TnSCARA5 associates with TnHMGB1 protein or CpG-ODN but does not do so with the TnHMGB1–CpG-ODN complex.

The aforementioned experiments showed that TnHMGB1 exerts a synergistic effect on CpG-ODN–induced inflammatory responses, and TnSCARA5 serves as a TnHMGB1 receptor with an inhibitory role in TnHMGB1/CpG-ODN–mediated inflammation. Therefore, we supposed that TnSCARA5 exerts its function by the clearance of TnHMGB1 through internalizing exogenous TnHMGB1 protein. To confirm this hypothesis, an internalization assay was performed on Tetraodon leukocytes. For the assay, the recombinant TnHMGB1 protein was conjuncted with FITC in advance. Internalization of FITC-TnHMGB1 was determined by FACS analysis. The performance of the TnSCARA5-mediated internalization of TnHMGB1 was examined by blocking the receptor with anti-TnSCARA5 Ab. Results show that the administration of FITC-TnHMGB1 led to a dose-dependent increase in percentages of leukocytes with fluorescence (from 4.09 to 13.18%) (Fig. 10A–C); in contrast, pretreatment with anti-TnSCARA5 Ab significantly reduced (p < 0.05) the percentages of the fluorescent cells from 13.18 to 8.25% (Fig. 10D). To exclude the adsorption, pretreatment with cytochalasin B (an actin inhibitor that can suppress receptor-mediated endocytosis) was set as control. As expected, the number of fluorescent leukocytes rarely increased in the presence of cytochalasin B (Fig. 10E, 10F). For further confirmation, TnSCARA5-mediated internalization of exogenous TnHMGB1 was determined by the decreased level of extracellular TnHMGB1 at different time points. ELISA results showed that the amount of the recombinant TnHMGB1 protein in the supernatant of the cultural leukocytes decreased after 30 min; however, the TnHMGB1 protein in the control group pretreated with anti-TnSCARA5 Ab remained unchanged (Fig. 11A).

FIGURE 10.

Internalization of exogenous TnHMGB1 by leukocytes via TnSCARA5. Leukocytes isolated from Tetraodon spleen and kidney tissues were incubated with different concentrations of FITC-TnHMGB1 for 1 h and analyzed by FACS. Compared with the control group (A), the FITC-TnHMGB1 incubation groups show increased FITC+ leukocytes (B and C) in a dose-dependent manner. Leukocytes preincubated with anti-TnSCARA5 Ab for 2 h, followed by treating with 15 μg/ml FITC-TnHMGB1, display decreased percentages of leukocytes with fluorescence (D) compared with those of group (C). With cytochalasin B treatment, the internalization of FITC-TnHMGB1 rarely increased (E). The statistical analysis of FITC+ leukocytes in groups (A)–(E) is shown as a bar graph in (F). All data are presented as means ± SD of at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

FIGURE 10.

Internalization of exogenous TnHMGB1 by leukocytes via TnSCARA5. Leukocytes isolated from Tetraodon spleen and kidney tissues were incubated with different concentrations of FITC-TnHMGB1 for 1 h and analyzed by FACS. Compared with the control group (A), the FITC-TnHMGB1 incubation groups show increased FITC+ leukocytes (B and C) in a dose-dependent manner. Leukocytes preincubated with anti-TnSCARA5 Ab for 2 h, followed by treating with 15 μg/ml FITC-TnHMGB1, display decreased percentages of leukocytes with fluorescence (D) compared with those of group (C). With cytochalasin B treatment, the internalization of FITC-TnHMGB1 rarely increased (E). The statistical analysis of FITC+ leukocytes in groups (A)–(E) is shown as a bar graph in (F). All data are presented as means ± SD of at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

Close modal
FIGURE 11.

Evaluation of TnHMGB1 internalization and colocalization with lysome in a TnSCARA5-dependent manner. (A) Tetraodon leukocytes were isolated and blocked with or without anti-TnSCARA5 Ab for 2 h. After incubating with the recombinant TnHMGB1 protein, the supernatant was collected at different time points to measure the amount of extracellular TnHMGB1 by ELISA. Extracellular TnHMGB1 decreased after 30 min but not in the Ab blockade group. (B) Tetraodon leukocytes were treated with 10 μg/ml FITC-TnHMGB1. After washing three times with ice-cold PBS to remove unbound TnHMGB1, the cells were subjected to LysoTracker Red staining. Red fluorescence reveals that TnHMGB1 colocalized well in the lysosome. Scale bars, 5 μm (original magnification ×630).

FIGURE 11.

Evaluation of TnHMGB1 internalization and colocalization with lysome in a TnSCARA5-dependent manner. (A) Tetraodon leukocytes were isolated and blocked with or without anti-TnSCARA5 Ab for 2 h. After incubating with the recombinant TnHMGB1 protein, the supernatant was collected at different time points to measure the amount of extracellular TnHMGB1 by ELISA. Extracellular TnHMGB1 decreased after 30 min but not in the Ab blockade group. (B) Tetraodon leukocytes were treated with 10 μg/ml FITC-TnHMGB1. After washing three times with ice-cold PBS to remove unbound TnHMGB1, the cells were subjected to LysoTracker Red staining. Red fluorescence reveals that TnHMGB1 colocalized well in the lysosome. Scale bars, 5 μm (original magnification ×630).

Close modal

In an attempt to explore whether the inhibitory function of TnSCARA5 is associated with the clearance of TnHMGB1, colocalization of TnHMGB1 with lysosome was detected in Tetraodon leukocytes. As shown in Fig. 11B, FITC-TnHMGB1 was taken up by the leukocytes and colocalized with lysosome in the cytoplasm. Meanwhile, the kinetic subcellular distribution of TnSCARA5 was tracked after TnHMGB1 stimulation. In this step, a Myc-tagged TnSCARA5 construct (pcDNA6-TnSCARA5) was transfected into HEK293 cells. After stimulation with TnHMGB1 (300 nM), the localization signal of TnSCARA5 was detected by immunofluorescence staining with secondary Ab against primary anti-TnSCARA5 Ab with red fluorescence. The result clearly demonstrated that TnSCARA5 was transferred to the surface membrane after stimulation for 30 min and returned to the cytoplasm after 1 h stimulation (Fig. 12A). Moreover, the EGFP-labeled TnSCARA5 in HEK293 cells (transfected with pEGFP-TnSCARA5) showed that the TnSCARA5 can be colocalized with the lysosome tracker (LysoTracker Red) after returning to the cytoplasm (Fig. 12B). These findings showed a kinetic change in the TnSCARA5 subcellular localization from the cytoplasm to the cell surface membrane and return to the lysosomes in the cytoplasm. Hence, TnSCARA5 can be recruited onto the cell membrane upon TnHMGB1 stimulation, which contributes to the clearance of TnHMGB1 by the receptor-mediated internalization of this protein into lysosome.

FIGURE 12.

Kinetic change of the TnSCARA5 subcellular localization with TnHMGB1 stimulation. (A) HEK293 cells transfected with pcDNA6-TnSCARA5 plasmid were stimulated with TnHMGB1 (300 nM) and the localization signal of TnSCARA5 was detected by immunofluorescence staining with red fluorescence–conjugated secondary Ab against anti-TnSCARA5 Abs. The shift of red fluorescence shows that TnSCARA5 transferred to the cell membrane and returned to the cytoplasm near the nucleus after TnHMGB1 stimulation for 30 or 60 min. Scale bars, 5 μm. (B) HEK293 cells were transfected with pEGFP-TnSCARA5, fixed, and stained with the lysosome probe LysoTracker Red. The fluorescence reveals that TnSCARA5 colocalized well in the lysosome with TnHMGB1 stimulation for 60 min. Scale bars, 10 μm (original magnification ×630).

FIGURE 12.

Kinetic change of the TnSCARA5 subcellular localization with TnHMGB1 stimulation. (A) HEK293 cells transfected with pcDNA6-TnSCARA5 plasmid were stimulated with TnHMGB1 (300 nM) and the localization signal of TnSCARA5 was detected by immunofluorescence staining with red fluorescence–conjugated secondary Ab against anti-TnSCARA5 Abs. The shift of red fluorescence shows that TnSCARA5 transferred to the cell membrane and returned to the cytoplasm near the nucleus after TnHMGB1 stimulation for 30 or 60 min. Scale bars, 5 μm. (B) HEK293 cells were transfected with pEGFP-TnSCARA5, fixed, and stained with the lysosome probe LysoTracker Red. The fluorescence reveals that TnSCARA5 colocalized well in the lysosome with TnHMGB1 stimulation for 60 min. Scale bars, 10 μm (original magnification ×630).

Close modal

SCARA5 is an evolutionarily conservative molecule that is present in mammals, birds, and fish. Thus, whether SCARA5 also plays an inhibitory role in CpG-ODN–induced inflammation in mammals was determined. For this, nonactivated splenic leukocytes were freshly isolated from mouse, stimulated with CpG-SGIV-ODN6 (3 μg/ml), and collected at 2, 4, 6, 8, and 10 h after stimulation. Results showed that the expression of inflammatory cytokines (IL-1β, IL-6, TNF-α, and IFN-γ) in the stimulated splenic leukocytes peaked at 6 h (Supplemental Fig. 3B). Then, the expression of these cytokines in response to the stimulation of different concentrations of CpG-SGIV-ODN6 (1–5 μg/ml) was determined at 6 h. CpG-SGIV-ODN6 induced the expression of the inflammatory cytokines in a dose-dependent manner (Supplemental Fig. 3C). Ab blockade assays showed that the expression of IL-1β, IL-6, TNF-α, and IFN-γ in splenic leukocytes treated with different concentrations of anti-mouse SCARA5 Ab (5–20 μg/ml) increased by 26–41% after CpG-SGIV-ODN6 stimulation compared with that of the control cells, which received no Ab treatment (Fig. 13A). Similarly, the expression of inflammatory cytokines increased by 20–70% after splenic leukocytes were treated with anti-mouse SCARA5 Ab (15 μg/ml) and stimulated by different concentrations of CpG-SGIV-ODN6 (1–4 μg/ml) (Fig. 13B). These results suggest that SCARA5 also plays an inhibitory role in CpG-ODN–induced inflammation in mammals. However, the detailed mechanisms underlying SCARA5-mediated regulation need further investigation.

FIGURE 13.

Negative regulation of mouse SCARA5 in CpG-SGIV-ODN–induced inflammation. (A) Nonactivated mouse splenic leukocytes were preincubated with different concentrations of anti-mouse SCARA5 Ab for 1 h at 37°C and stimulated with CpG-SGIV-ODN6 (3 μg/ml) for 6 h. Real-time PCR showed that the expression of inflammatory cytokines in the Ab-blocked groups increased. (B) Nonactivated mouse splenic leukocytes were preincubated with anti-mouse SCARA5 Ab (15 μg/ml) and stimulated with different concentrations of CpG-SGIV-ODN6. The expression levels of inflammatory cytokines increased in different groups. Data are presented as mean ± SD of at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

FIGURE 13.

Negative regulation of mouse SCARA5 in CpG-SGIV-ODN–induced inflammation. (A) Nonactivated mouse splenic leukocytes were preincubated with different concentrations of anti-mouse SCARA5 Ab for 1 h at 37°C and stimulated with CpG-SGIV-ODN6 (3 μg/ml) for 6 h. Real-time PCR showed that the expression of inflammatory cytokines in the Ab-blocked groups increased. (B) Nonactivated mouse splenic leukocytes were preincubated with anti-mouse SCARA5 Ab (15 μg/ml) and stimulated with different concentrations of CpG-SGIV-ODN6. The expression levels of inflammatory cytokines increased in different groups. Data are presented as mean ± SD of at least three independent experiments. *p < 0.05, **p < 0.01 compared with controls.

Close modal

The activation of innate immunity initiated by exogenous PAMPs or endogenous DAMPs through pattern recognition receptors is essential for the establishment of host defense against infections. However, excessive activation results in both infectious and sterile inflammatory diseases and autoimmune disorders, such as sepsis, systemic inflammatory response syndrome (SIRS), systemic lupus erythematosus, ulcerative colitis, and rheumatoid arthritis (29, 33, 34). The LPS and unmethylated CpG-ODNs derived from a range of bacterial and viral genomes and even mitochondrial DNA are the two major stimulators of the occurrence of sepsis, SIRS, and autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis (35, 36). LPS induces septic shock through TLR4 in mammals by associating with CD14, LPS-binding protein, and myeloid differentiation protein 2, because TLR4-deficient (TLR4−/−) mice were found to be tolerant of endotoxin lethality (37). However, the CpG-ODNs induce SIRS by the TLR9 located in the endosomes of macrophages, monocytes, and dendritic cells through the MyD88-dependent NF-κB signaling pathway (38). Mice challenged with excessive amounts of bacterial CpG-ODN were shown to die within 3 d because of the release of abundant proinflammatory cytokines, such as IL-1β, IL-6, and TNF-α (35). Thus, the burst of these cytokines has been used as an indicator for SIRS, which is an inflammatory reaction closely associated with pathogen infections or intense tissue damage (39). Moreover, mitochondrial DNA with unmethylated CpG motifs also activates the TLR9 signaling pathway and contributes to posttraumatic SIRS (36, 40, 41).

Emerging evidence has shown that HMGB1 endorses critical functions in the pathogenesis of both sterile and infectious inflammation outside the cells. In sterile injury, damage to cellular integrity results in a nearly instantaneous, passive release of HMGB1. For instance, HMGB1 release was initiated in ischemia–reperfusion injury within 1 h and remained elevated for 24 h. The protein served as an early mediator that in turn activated TNF-α and other cytokines (42). However, HMGB1 acts as a potent late mediator of infectious inflammation, such as sepsis, SIRS, and other disease models, in which HMGB1 can be secreted by activated macrophages and monocytes exposed to pathogens (12). The role of HMGB1 in sepsis has been extensively investigated. The results showed that HMGB1 was released by macrophages for >8 h and maintained at peak levels from 16 to 32 h after LPS exposure in mouse model, which is different from the TNF-α and IL-1β releases within minutes. The administration of anti–TNF-α Abs did not improve survival, but the administration of neutralizing anti-HMGB1 Abs or recombinant HMGB1 A box protein significantly attenuates endotoxin lethality in mice (12, 43). The recombinant A box protein is a competitive antagonist of HMGB1 that suppresses HMGB1-promoted cellular activation, which further highlights the importance of HMGB1 in sepsis (44). In contrast, the role of HMGB1 in SIRS induced by CpG-ODNs is limited, although HMGB1 has been shown to exhibit great importance in the CpG-ODN-TLR9 signaling pathway (45). Under CpG-ODN stimulation, HMGB1 shuttles in and out of immune cells and regulates inflammatory responses to CpG-DNA. Extracellular HMGB1 executes different roles depending on its redox form. Reduced cysteines render HMGB1 a chemoattractant that promotes the recruitment of inflammatory cells through CXCR4, whereas a disulfide bond renders it a proinflammatory cytokine that accelerates the delivery of CpG-ODNs to its receptor TLR9 through RAGE. Whether using the CpG-ODN-HMGB1 complex or CpG-ODN uptake, RAGE is important in CpG/HMGB1-induced inflammatory responses (46, 47). Intracellular HMGB1 specifically associates with TLR9 in the endoplasmic reticulum–Golgi intermediate compartment before and during the association of CpG-ODN to TLR9. Lack of the intracellular TLR9-associated HMGB1 can be compensated by extracellular HMGB1. Additionally, some earlier investigations suggested that HMGB1 can directly stimulate monocytes to release TNF-α and other proinflammatory cytokines in the late period of inflammation (12, 14). However, recent studies have shown that highly purified HMGB1 cannot induce detectable proinflammatory cytokines on its own. Instead, HMGB1 associates with proinflammatory factors to intensify the inflammatory responses (14, 15). Therefore, HMGB1 may act as a major chaperone mediator in synergy with LPS and CpG-ODNs to promote inflammation in sepsis, SIRS, and various autoimmune disorders.

Animal models are powerful tools for understanding the major immunological events and disease mechanisms difficult to address in humans. Aside from the classical mouse model, fish models, such as zebrafish and pufferfish, have become increasingly attractive in studies of comparative immunology and in modeling human diseases, such as T cell acute lymphoblastic leukemia, diabetes mellitus, Alagille syndrome, alcoholic liver disorder, and hepatocellular carcinoma; fish are suitable models because they exhibit precise and conservative innate immunity during vertebrate evolution (4850). In the present study, we again showed the advantages of fish models in uncovering the involvement of HMGB1 in CpG-ODN–induced inflammation and the regulatory mechanisms underlying the inflammatory reactions. Structurally, the HMGB1 identified from the pufferfish T. nigroviridis (TnHMGB1) is highly homologous to its human counterpart, as evidenced by a high amino acid identity (>74%) and conserved functional domains, including the A box, B box, T box, and Cys residues (10). TnHMGB1 also distributes in the nucleus of the resting cells and can be secreted out of the cells under stimulation of the proinflammatory stimulant CpG-ODN. By stimulating Tetraodon leukocytes with TnHMGB1 and CpG-ODN in different combinations, TnHMGB1 was demonstrated to exhibit a strong synergistic effect on CpG-ODN–induced inflammation. The expression of inflammatory cytokines was dramatically augmented in TnHMGB1 and CpG-ODN combined groups. Furthermore, TnHMGB1-deficient leukocytes exhibited impaired responsiveness to CpG-ODN stimulation, which support the synergy of TnHMGB1 with CpG-ODN stimulant. Additionally, we also found that the inflammatory effect of TnHMGB1 with CpG-ODN is dependent on TLR9, because no detectable inflammatory responses can be induced in HEK293 cells with low expression of TLR9. However, such behavior can be significantly improved by the exogenous expression of TnTLR9. Thus, TnHMGB1 clearly acts as an inflammatory accessory mediator that confers stimulatory function associated with CpG-ODN through TLR9 signaling pathway, which should be highly conserved between fish and mammals. These attributes make fish an attractive model to explore the CpG-ODN/HMGB1–induced inflammatory reactions and relevant disorders, such as SIRS.

As shown by the data, the major recognition receptors for extracellular HMGB1 were identified to be RAGE, TLR2, and TLR4. These receptors are physically potent in activating cellular growth, differentiation, migration, and inflammation in innate immunity through MyD88-dependent NF-κB and MAPK signaling pathways (22, 51). However, the negative regulatory pathways and mechanisms for HMGB1-mediated biological reactions are poorly understood. In the present study, we demonstrated that SCARA5 is an HMGB1 receptor that is negatively involved in HMGB1/CpG-ODN–elicited inflammation. This finding adds a new member to the HMGB1 receptor family and provides new insights into the regulatory role of SCARA5 in innate immunity. A number of experimental lines support this conclusion. Functionally, the overexpression of TnSCARA5 in HEK293T cells and in zebrafish embryos significantly inhibited the NF-κB activation and inflammatory cytokine expressions in response to TnHMGB1/CpG-ODN stimulation. However, the depletion of TnSCARA5 in leukocytes, kidney, and spleen tissues extremely enhanced the inflammatory responses. By qELISA and CoIP assays, TnSCARA5 was demonstrated to exhibit a strong ability to associate with TnHMGB1 in a dose-dependent manner. By constructing mutant TnHMGB1 proteins with deletions of A, B, and T boxes, we showed that both A and B boxes were essential for the interaction of TnHMGB1 with TnSCARA5. However, the T box negatively regulated the association, because the ablation of T box promoted the interaction of the two molecules. To our knowledge, this result is the first observation showing the regulatory function of HMGB1 T box in molecular interactions. The biological significance and mechanism underlying such a process must be further clarified. Alternatively, TnSCARA5 was demonstrated to preferentially interact with TnHMGB1 but not with the TnHMGB1–CpG-ODN complex. From previous studies, we found that A and B boxes are two tandem DNA-binding domains linked by a short, flexible linker that lies in the minor groove and HMG boxes; these domains contribute to DNA bending by interacting with DNA through hydrophobic residues (52). Thus, the A and B boxes are common functional domains for the interactions of HMGB1 with DNA/CpG-ODN and with SCARA5. Hence, explaining why TnSCARA5 preferentially interacts with TnHMGB1 rather than with the TnHMGB1–CpG-ODN complex seems reasonable. In the latter interaction, the A and B boxes might have been spatially occupied by the CpG-ODN, which competitively excludes the association of TnHMGB1 with TnSCARA5 through the A and B boxes simultaneously. However, further investigation is needed to clarify the details of this competitive interaction. The preferential binding of SCARA5 to HMGB1 instead of the HMGB1–CpG-ODN complex may be of great biological significance. As a double-edged sword, HMGB1 released earlier forms complexed with CpG-ODN as a proinflammatory stimulant for TLR9 to initiate inflammation, which is essential for the activation of host innate immunity. However, HMGB1-released later forms complex with CpG-ODN and other stimulants to boost excessive inflammation, which frequently result in inflammatory disorders. Thus, the preference of SCARA5 for HMGB1 might be a later event in the inflammatory reactions that occur to prevent a second inflammatory burst by the clearance of HMGB1 chaperone molecule without disturbing the early activation of innate immunity by HMGB1–CpG-ODN stimulation. This hypothesis was supported by the observation that the expression of TnSCARA5 was significantly induced under TnHMGB1 stimulation. The finding also suggests the existence of a precise feedback regulatory mechanism between SCARA5 and HMGB1. Furthermore, the T box of HMGB1 might act as a switch to turn off the interaction of SCARA5 with HMGB1–CpG-ODN. Hence, a more extended T box might be formed with the binding of CpG-ODN to HMGB1. This binding might make TnSCARA5 more preferable to interact with TnHMGB1 than the TnHMGB1–CpG-ODN complex. The exact mechanisms underlying this process definitely need elucidation.

Structurally, two receptor internalization–related motifs of YXXØ (Ø represents hydrophobic amino acids) exist in the intracellular part close to the transmembrane domain of TnSCARA5. These motifs include Y-A-I-V (residues 61–64) and Y-I-L-V (residues 67–70). The YXXØ motif, characteristic of internalized receptors, is the binding site of clathrin adaptor protein 2 (53). Thus, TnSCARA5 might mediate clathrin-dependent endocytosis by interacting with clathrin/adaptor protein 2. In accordance with this notion, we found that the TnSCARA5 expressed in HEK293T cells can be recruited to the cell membrane and return to the cytoplasm in response to TnHMGB1 stimulation. This finding is consistent with that in our last study, in which LPS drives the recycling process of TnSCARA5 (4). By a further internalization assay, we demonstrated that exogenous TnHMGB1 can be internalized by Tetraodon leukocytes through TnSCARA5-mediated endocytosis in a dose-dependent manner. This observation was indicated by the dose-dependent increase of the fluorescent cells incubated with FITC-TnHMGB1 and the decreased phagocytosis of leukocytes pretreated with anti-TnSCARA5 Ab or cytochalasin B, an inhibitor for actin polymerization essential in receptor-mediated endocytosis (54, 55). After internalization with TnHMGB1 into the cytoplasm, TnSCARA5 and TnHMGB1 were clearly demonstrated to be localized in the lysosomes, strongly suggesting that the inflammatory inhibition of TnSCARA5 may be largely ascribed to its involvement in the clearance of the TnHMGB1 chaperone mediator. Alternatively, the lysosomal acidic environment is well accepted to be strictly maintained by the ATP-dependent proton pump in the lysosome membrane. With CpG motif stimulation, the acidification of CpG-DNA in lysosomes is coupled with the rapid generation of intracellular reactive oxygen species (ROS) (56). Three cysteine residues are present in HMGB1; two residues in the A box form a reversible disulfide bond under mildly oxidizing conditions and can be further oxidized into sulfonates by the increase in ROS (5760). Functional diversity arises depending on the redox of HMGB1, as shown by the facts that reduced cysteines make HMGB1 a chemoattractant, whereas a disulfide bond makes HMGB1 a proinflammatory cytokine (57). These observations imply that a high level of ROS can be generated with the transport of CpG-ODN and HMGB1 into lysosomes by TnSCARA5. This process results in the terminal oxidization of cysteine residues into sulfonates. Such redox of TnHMGB1 might lead to the dissociation of TnHMGB1 from TnSCARA5. To explore this hypothesis, we examined the binding capacity of TnHMGB1 to TnSCARA5 under H2O2 treatment. As expected, the association of TnHMGB1 with TnSCARA5 was significantly decreased after the exposure of TnHMGB1 to H2O2. Thus, HMGB1 potentially disassociates from SCARA5 in the lysosome because of the acidic and oxidative environment under CpG-ODN stimulation. Such conditions may enable TnSCARA5 recycling from the lysosome to the surface membrane. Such notions may explain the ability of TnSCARA5 to interact with CpG-ODN besides its binding to HMGB1, which proves that ROS production by CpG-ODN stimulant is essential for the disassociation of HMGB1 from SCARA5.

In summary, the present study presents pioneering investigations of the role of SCARA5 as an HMGB1 recognition receptor crucial for the inhibition of CpG-ODN/HMGB1–induced inflammation in a pufferfish model. We previously found that TnSCARA5 is an LPS receptor negatively involved in LPS-induced inflammation. Besides this information, we demonstrated that SCARA5 is a multifunctional receptor with broad biological activities in innate immunity. Moreover, we showed that the protein is greatly important in preventing inflammation during the second inflammatory burst. This study will not only enrich current knowledge on the regulatory function of SCARA5 in innate immunity, but also serve useful in guiding the clinical treatment of diseases mediated by TnHMGB1.

This work was supported by National Basic Research Program of China (973) Grants 2012CB114404 and 2012CB114402, Key Program of National Natural Science Foundation of China Grant 31630083, National Natural Science Foundation of China Grants 31172436, 31272691, 31372554, 31472298, and 31572641, National Key Research and Development Program of China Grant 2016YFA0101001, Hi-Tech Research and Development Program of China (863) Grant 2012AA092202, and by Scientific Research Fund of Zhejiang Provincial Science and Technology Department Grant 2016-2020.

The sequences presented in this article have been submitted to GenBank (http://www.ncbi.nlm.nih.gov/genbank/) under accession numbers KU214880 (TnHMGB1) and KU214881 (TnTLR9).

The online version of this article contains supplemental material.

Abbreviations used in this article:

CoIP

coimmunoprecipitation

DAMP

damage-associated molecule pattern

EGFP

enhanced GFP

HEK293T

human emborynic kidney 293T

HKL

head kidney leukocyte

HMGB1

high-mobility group box 1

LV

lentivirus

ODN

oligodeoxynucleotide

PAMP

pathogen-associated molecule pattern

qELISA

quantitative ELISA

ROS

reactive oxygen species

SCARA5

scavenger receptor class A member 5

SGIV

Singapore grouper iridovirus

siRNA

small interfering RNA

SIRS

systemic inflammatory response syndrome

SR

scavenger receptor

Tn-

Tetraodon-

TU

transducing unit.

1
Plüddemann
A.
,
Mukhopadhyay
S.
,
Gordon
S.
.
2006
.
The interaction of macrophage receptors with bacterial ligands.
Expert Rev. Mol. Med.
8
:
1
25
.
2
Plüddemann
A.
,
Neyen
C.
,
Gordon
S.
.
2007
.
Macrophage scavenger receptors and host-derived ligands.
Methods
43
:
207
217
.
3
Hampton
R. Y.
,
Golenbock
D. T.
,
Penman
M.
,
Krieger
M.
,
Raetz
C. R.
.
1991
.
Recognition and plasma clearance of endotoxin by scavenger receptors.
Nature
352
:
342
344
.
4
Meng
Z.
,
Zhang
X. Y.
,
Guo
J.
,
Xiang
L. X.
,
Shao
J. Z.
.
2012
.
Scavenger receptor in fish is a lipopolysaccharide recognition molecule involved in negative regulation of NF-κB activation by competing with TNF receptor-associated factor 2 recruitment into the TNF-α signaling pathway.
J. Immunol.
189
:
4024
4039
.
5
Józefowski
S.
,
Sulahian
T. H.
,
Arredouani
M.
,
Kobzik
L.
.
2006
.
Role of scavenger receptor MARCO in macrophage responses to CpG oligodeoxynucleotides.
J. Leukoc. Biol.
80
:
870
879
.
6
Kong
X.
,
Thimmulappa
R.
,
Kombairaju
P.
,
Biswal
S.
.
2010
.
NADPH oxidase-dependent reactive oxygen species mediate amplified TLR4 signaling and sepsis-induced mortality in Nrf2-deficient mice.
J. Immunol.
185
:
569
577
.
7
Andersson
U.
,
Wang
H.
,
Palmblad
K.
,
Aveberger
A. C.
,
Bloom
O.
,
Erlandsson-Harris
H.
,
Janson
A.
,
Kokkola
R.
,
Zhang
M.
,
Yang
H.
,
Tracey
K. J.
.
2000
.
High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes.
J. Exp. Med.
192
:
565
570
.
8
Rouhiainen
A.
,
Kuja-Panula
J.
,
Wilkman
E.
,
Pakkanen
J.
,
Stenfors
J.
,
Tuominen
R. K.
,
Lepäntalo
M.
,
Carpén
O.
,
Parkkinen
J.
,
Rauvala
H.
.
2004
.
Regulation of monocyte migration by amphoterin (HMGB1).
Blood
104
:
1174
1182
.
9
Campana
L.
,
Bosurgi
L.
,
Rovere-Querini
P.
.
2008
.
HMGB1: a two-headed signal regulating tumor progression and immunity.
Curr. Opin. Immunol.
20
:
518
523
.
10
Andersson
U.
,
Tracey
K. J.
.
2011
.
HMGB1 is a therapeutic target for sterile inflammation and infection.
Annu. Rev. Immunol.
29
:
139
162
.
11
Scaffidi
P.
,
Misteli
T.
,
Bianchi
M. E.
.
2002
.
Release of chromatin protein HMGB1 by necrotic cells triggers inflammation.
Nature
418
:
191
195
.
12
Wang
H.
,
Bloom
O.
,
Zhang
M.
,
Vishnubhakat
J. M.
,
Ombrellino
M.
,
Che
J.
,
Frazier
A.
,
Yang
H.
,
Ivanova
S.
,
Borovikova
L.
, et al
.
1999
.
HMG-1 as a late mediator of endotoxin lethality in mice.
Science
285
:
248
251
.
13
Gardella
S.
,
Andrei
C.
,
Ferrera
D.
,
Lotti
L. V.
,
Torrisi
M. R.
,
Bianchi
M. E.
,
Rubartelli
A.
.
2002
.
The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway.
EMBO Rep.
3
:
995
1001
.
14
Sha
Y.
,
Zmijewski
J.
,
Xu
Z.
,
Abraham
E.
.
2008
.
HMGB1 develops enhanced proinflammatory activity by binding to cytokines.
J. Immunol.
180
:
2531
2537
.
15
Wähämaa
H.
,
Schierbeck
H.
,
Hreggvidsdottir
H. S.
,
Palmblad
K.
,
Aveberger
A. C.
,
Andersson
U.
,
Harris
H. E.
.
2011
.
High mobility group box protein 1 in complex with lipopolysaccharide or IL-1 promotes an increased inflammatory phenotype in synovial fibroblasts.
Arthritis Res. Ther.
13
:
R136
.
16
He
Z.-W.
,
Qin
Y.-H.
,
Wang
Z.-W.
,
Chen
Y.
,
Shen
Q.
,
Dai
S.-M.
.
2013
.
HMGB1 acts in synergy with lipopolysaccharide in activating rheumatoid synovial fibroblasts via p38 MAPK and NF-KB signaling pathways.
Mediat. Inflamm.
2013
:
596716
. doi 10.1155/2013/596716.
17
Yang
D.
,
Chen
Q.
,
Yang
H.
,
Tracey
K. J.
,
Bustin
M.
,
Oppenheim
J. J.
.
2007
.
High mobility group box-1 protein induces the migration and activation of human dendritic cells and acts as an alarmin.
J. Leukoc. Biol.
81
:
59
66
.
18
Dumitriu
I. E.
,
Baruah
P.
,
Valentinis
B.
,
Voll
R. E.
,
Herrmann
M.
,
Nawroth
P. P.
,
Arnold
B.
,
Bianchi
M. E.
,
Manfredi
A. A.
,
Rovere-Querini
P.
.
2005
.
Release of high mobility group box 1 by dendritic cells controls T cell activation via the receptor for advanced glycation end products.
J. Immunol.
174
:
7506
7515
.
19
Park
J. S.
,
Svetkauskaite
D.
,
He
Q.
,
Kim
J. Y.
,
Strassheim
D.
,
Ishizaka
A.
,
Abraham
E.
.
2004
.
Involvement of Toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein.
J. Biol. Chem.
279
:
7370
7377
.
20
Ivanov
S.
,
Dragoi
A. M.
,
Wang
X.
,
Dallacosta
C.
,
Louten
J.
,
Musco
G.
,
Sitia
G.
,
Yap
G. S.
,
Wan
Y.
,
Biron
C. A.
, et al
.
2007
.
A novel role for HMGB1 in TLR9-mediated inflammatory responses to CpG-DNA.
Blood
110
:
1970
1981
.
21
Westerfield
M.
2000
.
The Zebrafish Book: A Guide for the Lab Oratory Use of Zebrafish (Danio rerio).
University of Oregon Press
,
Eugene, OR
.
22
Kimmel
C. B.
,
Ballard
W. W.
,
Kimmel
S. R.
,
Ullmann
B.
,
Schilling
T. F.
.
1995
.
Stages of embryonic development of the zebrafish.
Dev. Dyn.
203
:
253
310
.
23
Gu
Y. F.
,
Fang
Y.
,
Jin
Y.
,
Dong
W. R.
,
Xiang
L. X.
,
Shao
J. Z.
.
2011
.
Discovery of the DIGIRR gene from teleost fish: a novel Toll-IL-1 receptor family member serving as a negative regulator of IL-1 signaling.
J. Immunol.
187
:
2514
2530
.
24
Thompson
J. D.
,
Higgins
D. G.
,
Gibson
T. J.
.
1994
.
CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice.
Nucleic Acids Res.
22
:
4673
4680
.
25
Tamura
K.
,
Peterson
D.
,
Peterson
N.
,
Stecher
G.
,
Nei
M.
,
Kumar
S.
.
2011
.
MEGA5: molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods.
Mol. Biol. Evol.
28
:
2731
2739
.
26
Xiong
R.
,
Nie
L.
,
Xiang
L. X.
,
Shao
J. Z.
.
2012
.
Characterization of a PIAS4 homologue from zebrafish: insights into its conserved negative regulatory mechanism in the TRIF, MAVS, and IFN signaling pathways during vertebrate evolution.
J. Immunol.
188
:
2653
2668
.
27
Zhu
L. Y.
,
Lin
A. F.
,
Shao
T.
,
Nie
L.
,
Dong
W. R.
,
Xiang
L. X.
,
Shao
J. Z.
.
2014
.
B cells in teleost fish act as pivotal initiating APCs in priming adaptive immunity: an evolutionary perspective on the origin of the B-1 cell subset and B7 molecules.
J. Immunol.
192
:
2699
2714
.
28
Hu
Y. X.
,
Guo
J. Y.
,
Shen
L.
,
Chen
Y.
,
Zhang
Z. C.
,
Zhang
Y. L.
.
2002
.
Get effective polyclonal antisera in one month.
Cell Res.
12
:
157
160
.
29
Wang
P.
,
Lu
Y. Q.
,
Wen
Y.
,
Yu
D. Y.
,
Ge
L.
,
Dong
W. R.
,
Xiang
L. X.
,
Shao
J. Z.
.
2013
.
IL-16 induces intestinal inflammation via PepT1 upregulation in a pufferfish model: new insights into the molecular mechanism of inflammatory bowel disease.
J. Immunol.
191
:
1413
1427
.
30
Elbashir
S. M.
,
Harborth
J.
,
Weber
K.
,
Tuschl
T.
.
2002
.
Analysis of gene function in somatic mammalian cells using small interfering RNAs.
Methods
26
:
199
213
.
31
Zhang
R. P.
,
Shao
J. Z.
,
Xiang
L. X.
.
2011
.
GADD45A protein plays an essential role in active DNA demethylation during terminal osteogenic differentiation of adipose-derived mesenchymal stem cells.
J. Biol. Chem.
286
:
41083
41094
.
32
Antoine
D. J.
,
Harris
H. E.
,
Andersson
U.
,
Tracey
K. J.
,
Bianchi
M. E.
.
2014
.
A systematic nomenclature for the redox states of high mobility group box (HMGB) proteins.
Mol. Med.
20
:
135
137
.
33
Wang
H.
,
Ward
M. F.
,
Sama
A. E.
.
2014
.
Targeting HMGB1 in the treatment of sepsis.
Expert Opin. Ther. Targets
18
:
257
268
.
34
Balk
R. A.
2014
.
Systemic inflammatory response syndrome (SIRS): where did it come from and is it still relevant today?
Virulence
5
:
20
26
.
35
Hong
Z.
,
Jiang
Z.
,
Liangxi
W.
,
Guofu
D.
,
Ping
L.
,
Yongling
L.
,
Wendong
P.
,
Minghai
W.
.
2004
.
Chloroquine protects mice from challenge with CpG ODN and LPS by decreasing proinflammatory cytokine release.
Int. Immunopharmacol.
4
:
223
234
.
36
Zhang
J. Z.
,
Liu
Z.
,
Liu
J.
,
Ren
J. X.
,
Sun
T. S.
.
2014
.
Mitochondrial DNA induces inflammation and increases TLR9/NF-κB expression in lung tissue.
Int. J. Mol. Med.
33
:
817
824
.
37
Poltorak
A.
,
He
X.
,
Smirnova
I.
,
Liu
M.-Y.
,
Van Huffel
C.
,
Du
X.
,
Birdwell
D.
,
Alejos
E.
,
Silva
M.
,
Galanos
C.
, et al
.
1998
.
Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene.
Science
282
:
2085
2088
.
38
Hemmi
H.
,
Takeuchi
O.
,
Kawai
T.
,
Kaisho
T.
,
Sato
S.
,
Sanjo
H.
,
Matsumoto
M.
,
Hoshino
K.
,
Wagner
H.
,
Takeda
K.
,
Akira
S.
.
2000
.
A Toll-like receptor recognizes bacterial DNA.
Nature
408
:
740
745
.
39
Dinarello
C. A.
1997
.
Proinflammatory and anti-inflammatory cytokines as mediators in the pathogenesis of septic shock.
Chest
112
(
Suppl.
):
321S
329S
.
40
Kitajima
I.
,
Niimi
H.
.
2012
.
[Establishment of the rapid, hypersensitive testing systems for sepsis/SIRS]
.
Rinsho Byori
60
:
46
51
.
41
Hauser
C. J.
,
Sursal
T.
,
Rodriguez
E. K.
,
Appleton
P. T.
,
Zhang
Q.
,
Itagaki
K.
.
2010
.
Mitochondrial damage associated molecular patterns from femoral reamings activate neutrophils through formyl peptide receptors and P44/42 MAP kinase.
J. Orthop. Trauma
24
:
534
538
.
42
Tsung
A.
,
Sahai
R.
,
Tanaka
H.
,
Nakao
A.
,
Fink
M. P.
,
Lotze
M. T.
,
Yang
H.
,
Li
J.
,
Tracey
K. J.
,
Geller
D. A.
,
Billiar
T. R.
.
2005
.
The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion.
J. Exp. Med.
201
:
1135
1143
.
43
Apetoh
L.
,
Ghiringhelli
F.
,
Tesniere
A.
,
Criollo
A.
,
Ortiz
C.
,
Lidereau
R.
,
Mariette
C.
,
Chaput
N.
,
Mira
J. P.
,
Delaloge
S.
, et al
.
2007
.
The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy.
Immunol. Rev.
220
:
47
59
.
44
Yang
H.
,
Wang
H.
,
Czura
C. J.
,
Tracey
K. J.
.
2002
.
HMGB1 as a cytokine and therapeutic target.
J. Endotoxin Res.
8
:
469
472
.
45
Tian
J.
,
Avalos
A. M.
,
Mao
S. Y.
,
Chen
B.
,
Senthil
K.
,
Wu
H.
,
Parroche
P.
,
Drabic
S.
,
Golenbock
D.
,
Sirois
C.
, et al
.
2007
.
Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE.
Nat. Immunol.
8
:
487
496
.
46
Sirois
C. M.
,
Jin
T.
,
Miller
A. L.
,
Bertheloot
D.
,
Nakamura
H.
,
Horvath
G. L.
,
Mian
A.
,
Jiang
J.
,
Schrum
J.
,
Bossaller
L.
, et al
.
2013
.
RAGE is a nucleic acid receptor that promotes inflammatory responses to DNA. [Published erratum appears in 2014 J. Exp. Med. 5: 1001.]
J. Exp. Med.
210
:
2447
2463
.
47
Hiraku
Y.
,
Guo
F.
,
Ma
N.
,
Yamada
T.
,
Wang
S.
,
Kawanishi
S.
,
Murata
M.
.
2016
.
Multi-walled carbon nanotube induces nitrative DNA damage in human lung epithelial cells via HMGB1-RAGE interaction and Toll-like receptor 9 activation.
Part. Fibre Toxicol.
13
:
16
.
48
Langenau
D. M.
,
Traver
D.
,
Ferrando
A. A.
,
Kutok
J. L.
,
Aster
J. C.
,
Kanki
J. P.
,
Lin
S.
,
Prochownik
E.
,
Trede
N. S.
,
Zon
L. I.
,
Look
A. T.
.
2003
.
Myc-induced T cell leukemia in transgenic zebrafish.
Science
299
:
887
890
.
49
Olsen
A. S.
,
Sarras
M. P.
 Jr.
,
Intine
R. V.
.
2010
.
Limb regeneration is impaired in an adult zebrafish model of diabetes mellitus.
Wound Repair Regen.
18
:
532
542
.
50
Passeri
M. J.
,
Cinaroglu
A.
,
Gao
C.
,
Sadler
K. C.
.
2009
.
Hepatic steatosis in response to acute alcohol exposure in zebrafish requires sterol regulatory element binding protein activation.
Hepatology
49
:
443
452
.
51
Huttunen
H. J.
,
Fages
C.
,
Rauvala
H.
.
1999
.
Receptor for advanced glycation end products (RAGE)-mediated neurite outgrowth and activation of NF-κB require the cytoplasmic domain of the receptor but different downstream signaling pathways.
J. Biol. Chem.
274
:
19919
19924
.
52
Stott
K.
,
Tang
G. S. F.
,
Lee
K.-B.
,
Thomas
J. O.
.
2006
.
Structure of a complex of tandem HMG boxes and DNA.
J. Mol. Biol.
360
:
90
104
.
53
Bonifacino
J. S.
,
Traub
L. M.
.
2003
.
Signals for sorting of transmembrane proteins to endosomes and lysosomes.
Annu. Rev. Biochem.
72
:
395
447
.
54
Maxfield
F. R.
,
McGraw
T. E.
.
2004
.
Endocytic recycling.
Nat. Rev. Mol. Cell Biol.
5
:
121
132
.
55
Ferguson
S. M.
,
De Camilli
P.
.
2012
.
Dynamin, a membrane-remodelling GTPase.
Nat. Rev. Mol. Cell Biol.
13
:
75
88
.
56
Yi
A. K.
,
Tuetken
R.
,
Redford
T.
,
Waldschmidt
M.
,
Kirsch
J.
,
Krieg
A. M.
.
1998
.
CpG motifs in bacterial DNA activate leukocytes through the pH-dependent generation of reactive oxygen species.
J. Immunol.
160
:
4755
4761
.
57
Venereau
E.
,
Casalgrandi
M.
,
Schiraldi
M.
,
Antoine
D. J.
,
Cattaneo
A.
,
De Marchis
F.
,
Liu
J.
,
Antonelli
A.
,
Preti
A.
,
Raeli
L.
, et al
.
2012
.
Mutually exclusive redox forms of HMGB1 promote cell recruitment or proinflammatory cytokine release.
J. Exp. Med.
209
:
1519
1528
.
58
Tang
Y.
,
Zhao
X.
,
Antoine
D.
,
Xiao
X.
,
Wang
H.
,
Andersson
U.
,
Billiar
T. R.
,
Tracey
K. J.
,
Lu
B.
.
2016
.
Regulation of posttranslational modifications of HMGB1 during immune responses.
Antioxid. Redox Signal.
24
:
620
634
.
59
Nyström
S.
,
Antoine
D. J.
,
Lundbäck
P.
,
Lock
J. G.
,
Nita
A. F.
,
Högstrand
K.
,
Grandien
A.
,
Erlandsson-Harris
H.
,
Andersson
U.
,
Applequist
S. E.
.
2013
.
TLR activation regulates damage-associated molecular pattern isoforms released during pyroptosis.
EMBO J.
32
:
86
99
.
60
Balosso
S.
,
Liu
J.
,
Bianchi
M. E.
,
Vezzani
A.
.
2014
.
Disulfide-containing high mobility group box-1 promotes N-methyl-d-aspartate receptor function and excitotoxicity by activating Toll-like receptor 4-dependent signaling in hippocampal neurons.
Antioxid. Redox Signal.
21
:
1726
1740
.

The authors have no financial conflicts of interest.

Supplementary data