Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Laminin Receptor in Shrimp Is a Cellular Attachment Receptor for White Spot Syndrome Virus

  • Wang-Jing Liu ,

    liuwjing@utaipei.edu.tw

    Affiliation Department of Earth and Life Science, College of Science, University of Taipei, Taipei, Taiwan

  • Yi-Chieh Li,

    Affiliation Department of Earth and Life Science, College of Science, University of Taipei, Taipei, Taiwan

  • Guang-Hsiung Kou,

    Affiliation Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan

  • Chu-Fang Lo

    Affiliation Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan

Abstract

White spot syndrome virus (WSSV, genus Whispovirus, family Nimaviridae) is causing huge economic losses in global shrimp farming, but there is no effective control. Shrimp cell laminin receptor (Lamr) may have a role in WSSV infection. The objective was to characterize interactions between Penaeus monodon Lamr (PmLamr) and WSSV structural proteins. In this study, PmLamr interacted with nine WSSV structural proteins (based on yeast two-hybrid screening), of which one (VP31) was characterized. Protein pull-down assay confirmed the interaction between PmLamr and VP31; the latter was an envelope protein exposed outside the WSSV virion (based on membrane topology assays). Furthermore, similar to mammalian Lamr, there were two major protein bands in shrimp cells. Cellular localization assay demonstrated VP31 co-localized with PmLamr on transfected cells. Enzyme-link immunosorbent assay (ELISA) and competitive ELISA demonstrated binding of VP31 on PmLamr was dose-dependent; however, addition of WSSV virion competed for binding affinity. Furthermore, based on an in vivo neutralization assay, both VP31 and PmLamr delayed mortality in shrimp challenged with WSSV. We concluded Lamr was an important receptor for WSSV infection and the viral envelope protein VP31 may have a role in host cell recognition and binding. These data contributed to elucidating pathogenesis of WSSV infection and may help in controlling this disease.

Introduction

White spot syndrome virus (WSSV; family Nimaviridae, genus Whispovirus [1]) is a double-stranded DNA virus, which contains a large genome (~307 kbp). This virus affects most cultured shrimp with cumulative morality approaching 100% within 3 to 7 d after disease onset [25]. WSSV has an extensive host range (> 93 species of arthropods are known hosts or carriers [5]) and based on wide tissue tropism, the cellular receptor for WSSV is predicted to be conserved and ubiquitous [6]. Various proteins have been suspected to mediate WSSV infection, including Penaeus monodon Rab7 (PmRab7) [7], P. monodon chitin-binding protein (PmCBP) [8], beta-integrin [6], F1 ATP synthase beta subunit [9], or glucose transporter 1 [10, 11].

Laminin receptor (Lamr), which is a cell surface receptor, is notable because it mediates high-affinity interactions between laminin and the cell. Lamr has a predicted molecular mass of 32 kDa. However, when in SDS-polyacrylamide gels it is found to have an apparent electrophoretic mobility of ~37 kDa, and further processed into a 67-kDa protein [12]. Based on its molecular weight and function, Lamr has been designated 37/67-kDa laminin receptor (37LR, 67LR, LAMR1), 32 kDa laminin binding protein (LBP), 32 kDa laminin binding protein precursor (LBP-32, 37LRP), p40 and ribosomal protein SA (RPSA) [13]. Furthermore, Lamr has been also recognized as a multifunctional protein involved in not just cell adhesion, but also a wide range of biological processes, such as cell development, mobility and differentiation [14]. Moreover, it has also been reported that Lamr acts as a receptor for several exogenous agents, including prion proteins, viruses and bacteria [13]. In shrimp, Lamr was first identified as a receptor protein for Taura syndrome virus (TSV) [15]. It was subsequently reported to act as a binding protein for two additional shrimp RNA viruses (infectious myonecrosis virus [IMNV] and yellow head virus [YHV]) [16] and was also implicated in hemocyte homeostasis for white shrimp, Litopenaeus vannamei [17]. In the present research, the potential role of shrimp Lamr in WSSV infection was investigated. Results indicated that PmLamr may act as a host cellular receptor which bound to the WSSV envelope protein VP31 and mediated WSSV infection.

Materials and Methods

PmLamr cloning and expression in yeast

Protein-protein interaction assays were performed by using the Matchmaker Gold yeast two-hybrid system (Clontech). These assays were carried out in order to identify candidates of WSSV structural proteins that could interact with P. monodon Lamr (PmLamr). By cloning the PCR-amplified cDNA fragment encoding PmLamr (Genbank accession number DT044263) into the pGBKT7 vector (Clontech) in frame with the GAL4 DNA binding domain (DBD), the bait plasmid pGBKT7-PmLamr was produced. The resulting bait plasmid was then transformed into yeast (Saccharomyces cerevisiae) strain Y2HGold. The expression of GAL4 DBD-PmLamr fusion bait protein was confirmed by Western blotting with an anti-c-Myc antibody (Abcam). The autoactivation and toxicity of the bait protein was tested before two-hybrid screening. Sequences of primers used for pGBKT7-PmLamr construction are listed (Table 1).

thumbnail
Table 1. Primers used for generating plasmids used in this study.

https://doi.org/10.1371/journal.pone.0156375.t001

Yeast two-hybrid assay

By cloning the 49 WSSV structural protein and three P. monodon cellular protein genes, respectively, into the vector pGADT7 (Clontech) and then transforming the resulting plasmids into the S. cerevisiae host strain Y187, a prey library was produced [18]. In order to identify PmLamr interaction candidate proteins, the prey library clones were mated with the bait (i.e. the pGBKT7-PmLamr-transformed Y2HGold). Both positive and negative controls were made, and this was done by mating pGADT7-T-transformed Y187 prey with pGBKT7-53- or pGBKT7-Lam-transformed bait, respectively (the manufacturer provided the corresponding plasmids). A minimal synthetically defined (SD) double-dropout (DDO; SD medium lacking Leu and Trp [SD/-Leu/-Trp]) medium supplemented with 5-bromo-4-chloro-3-indolyl-α-D-galactopyranoside (X-α-Gal) and Aureobasidin A (DDO/X/A) was used to select positive clones which expressed prey proteins that interacted with PmLamr (bait). Blue colonies that were found to be growing on a DDO/X/A medium were subsequently patched onto higher stringency quadruple-dropout (QDO; SD medium without Ade, His, Leu, and Trp [SD/-Ade/-His/-Leu/-Trp]) plates containing X-α-Gal and Aureobasidin A (QDO/X/A).

Expression and purification of native PmLamr-His and antibody production

The cDNA fragment encoding PmLamr was cloned into the pET-28b(+) vector (Novagen), resulting in plasmid pET28b/PmLamr-His. Sequences of primers used for pET28b/PmLamr-His construction are listed (Table 1). To express a His-tagged-PmLamr (PmLamr-His) fusion protein, pET28b/PmLamr-His was transformed into Escherichia coli strain BL21-CodonPlus (DE3)-RIL (Stratagene), and protein expression induced with 0.1 mM isopropylthiogalactoside (IPTG) overnight at 16°C. Following centrifugation (6,000 ×g for 10 min), E. coli cell pellets were re-suspended in lysis buffer (50 mM Tris-HCl [pH 7.4], 300 mM NaCl, 20 mM imidazole, 1 mM PMSF, 0.25 mg/ml lysozyme) and sonicated on ice. Cellular debris was removed by centrifugation (10,000 ×g for 30 min). Ninitriloacetic acid (Ni-NTA) beads (Qiagen) were added to the supernatant and rotating mixing at 4°C for 2 h, then washed with washing buffer (50 mM Tris-HCl [pH 7.4], 300 mM NaCl, and 100 mM imidazole). Thereafter, PmLamr-His was eluted with elution buffer (50 mM Tris-HCl [pH 7.4], 300 mM NaCl and 250 mM immidazole). Fractions of PmLamr-His were collected, pooled and dialyzed against 1× PBS and used in antibody preparation, pull-down assays, enzyme-link immunosorbent assays (ELISA) and in vivo neutralization assays. For polyclonal antibody production, the fusion protein–PmLamr-His was subjected to SDS-PAGE; protein bands containing fusion proteins were sliced from the gel, minced, mixed with Freund’s complete adjuvant, and inoculated into rabbits.

Expression and purification of native MBP or MBP-VP31

An MBP tagged VP31 (WSSV T-1, GenBank accession number AF440570) expression construct–pMAL/VP31 was formed by digesting the amplified VP31 gene fragment using EcoRI and SacII and ligating it into the corresponding restriction enzyme cutting sites of pMAL-c5X (NEB). Sequences of primers used for pMAL/VP31 construction are listed (Table 1). An excess of recombinant MBP or recombinant MBP-VP31 was produced in E. coli strain BL21-CodonPlus (DE3)-RIL with 0.1 mM IPTG incubated overnight at 16°C. Following centrifugation (6,000 ×g for 10 min), E. coli cell pellets were re-suspended in lysis buffer (20 mM Tris-HCl [pH 7.4], 200 mM NaCl, 1 mM EDTA, 1 mM DTT, 1 mM PMSF, 0.25 mg/ml lysozyme, and 0.1% Triton X-100), kept on ice and sonicated. Cellular debris was removed by centrifugation (10,000 ×g for 30 min). Then the supernatant was added to amylose resins (NEB) and rotating mixing at 4°C for 2 h and washed with washing buffer (20 mM Tris-HCl [pH 7.4], 200 mM NaCl, 1 mM EDTA, 1 mM DTT). Finally, MBP and MBP-VP31 were eluted with elution buffer (washing buffer with 10 mM maltose). Fractions were collected, pooled and dialyzed against 1× PBS. These proteins were used for the following pull-down, ELISA, and in vivo neutralization assays.

Interaction of MBP-VP31 with PmLamr-His (protein pull-down assay)

Approximately 500 ng of PmLamr-His was incubated with 1 μg of either MBP or MBP-VP31 bound to amylose resins in 150 μl TBST buffer (10 mM Tris-HCl [pH 7.2], 150 mM NaCl, 1% Triton X-100, 10 mM EDTA, and a protease inhibitor cocktail tablet [Roche]) at 4°C for 3 h. After seven 10-min washes with TBST buffer, proteins that bound to the beads were resolved by 12% SDS-PAGE followed by Western blot analysis. The presence of PmLamr-His was detected using anti-PmLamr polyclonal antibody and goat anti-rabbit IgG-horseradish peroxidase (HRP) conjugate secondary antibody (Sigma). Protein band signals were visualized using a chemiluminescence reagent ECL (Perkin-Elmer).

Membrane topology of VP31

The membrane topology of VP31 predictions were predicted with TMHMM (http://www.cbs.dtu.dk/services/TMHMM-2.0/; [19]) and HMMTOP (http://www.enzim.hu/hmmtop/) [20, 21] based on the deduced aa sequence of VP31. Both methods were used in single-sequence mode, with all user-adjustable parameters set at default values. To further investigate the membrane topology of VP31 in the WSSV virion, experiments were done as described [22]. For a duration of 2 h at 37°C, aliquots (5 μg of total protein) of purified virions were treated with 5 μg/ml trypsin (Promega) in 100 μl of buffer (50 mM Tris-HCl [pH 7.5], 1 mM CaCl2, 100 mM NaCl). Triton X-100 (final concentration, 1%) was added in some samples prior to trypsin digestion, and this was done in order to dissolve the viral envelope and expose internal structures to trypsin. For the purpose of analyzing samples, Western blotting using antibodies against VP31 [23] was implemented, and with the aim of facilitating comparisons it was also implemented against tegument protein VP26 and the envelope protein VP28. The present study involves a WSSV virion; healthy crayfish (Procambarus clarkii) were challenged with WSSV-T1 strain and virus purified for the purpose of preparing the WSSV virion. Please see the description in Xie and Yang [24].

Protein lysates preparation and Western blot analysis

Shrimp stomach tissues were lysed in 0.33× PBS with protein inhibitor cocktail, centrifuged at 12,000 ×g for 5 min, and supernatant retained for Western blot analysis. To prepare PmLamr recombinant proteins from Drosophila S2 cells, the full-length PmLamr coding region was inserted into a heat-inducible Drosophila heat shock protein 70 promoter (pDHsp/V5-His [25]) by PCR cloning (using primers listed in Table 1 to generate pDHsp/PmLamr-V5-His). For DNA transfection, Drosophila S2 cells were seeded onto 24-well plates (8 × 105 cells/well) and grown in Schneider’s medium (Invitrogen) supplemented with 10% fetal calf serum at 28°C. After 1 h, the pDHsp/PmLamr-V5-His plasmid (200 ng of plasmid DNA per well) was transfected into the S2 cells with Effectene reagent (Qiagen) for 16 to 18 h, followed by heat shock (37°C in a water bath for 30 min). At 72 h after transfection, S2 cells were lysed in 25 μl NP-40 lysis buffer (50 mM Tris-HCl [pH 8.0], 150 mM NaCl and 1% NP-40). Cell lysates were separated by 12% SDS-PAGE, followed by Western blotting. PmLamr proteins were detected using an anti-PmLamr polyclonal antibody.

Confocal microscopy and co-localization

Drosophila S2 cells were seeded onto glass coverslips placed in the wells of a 24-well plate and transfected with pDHsp/PmLamr-V5-His plasmid, as described above. After 48 h, S2 cells were incubated with 1 μg of either MBP or MBP-VP31 for 2 h at 28°C. Attached cells were washed twice with PBS, fixed with freshly prepared 4% formaldehyde in PBS for 10 min and then washed with PBS two times. After blocking with PBS containing 5% bovine serum albumin and 2% normal goat serum for a duration of 16 h at 4°C, the cells were then incubated for a total of 3 h at room temperature (RT) with either the 1:500 PBS-diluted rabbit anti-PmLamr antibody, the 1:500 PBS-diluted mouse anti-MBP antibody (NEB), or with both antibodies. Next, the cells were washed a total of three times with PBST (PBS containing 0.2% Tween-20), and then reacted for 2 h at RT with either 1:1000 PBS-diluted carboxymethylindocyanine (Cy3) dye-conjugated goat anti-rabbit IgG antibody (Sigma), 1:1000 PBS-diluted Alexa Fluor® 488 dye-conjugated goat anti-mouse IgG antibody (Invitrogen), or with both. A confocal microscope (Leica TCS SP5) was used to detect fluorescence signals.

Binding specificity of PmLamr-His to MBP-VP31 (ELISA and competitive ELISA)

For ELISA, flat-bottomed 96-well ELISA plates (Corning Costar) were coated with 2 μg PmLamr-His (in 0.1 M bicarbonate/carbonate buffer [pH 9.6]; 100 μl per well) and incubated overnight at 4°C. Plates were then washed three times with washing buffer (1× PBS containing 0.05% Tween-20), and blocking buffer (washing buffer with additional 1% BSA) was added, followed by incubation for 2 h at RT. After washing with washing buffer, various dilutions of MBP or MBP-VP31 (0 to 400 ng) in blocking buffer were added to plates, followed by incubation for 2 h at RT. Plates were washed and a blocking buffer diluted detection antibody (mouse anti-MBP antibody) was added (100 μl per well), followed by incubation for 1 h at RT. After washing, HRP-labeled goat anti-mouse antibody (Santa Cruz) was added to plates, and reaction mixtures incubated for an additional 1 h at RT. The reaction was visualized using the HRP substrate TMB (3,3’,5,5’-tetramethylbenzidine; eBioscience), stopped by adding 100 μl 1 N H2SO4. An ELISA reader (biochrom) was used to read the absorbance immediately at 450 nm. For competitive ELISA, 96-well ELISA plates were first coated with 2 μg PmLamr-His, followed by washing and blocking. Thereafter, 200 ng of MBP-VP31 mixed with various concentrations (0 to 1200 ng) of WSSV viral proteins or BSA were added to plates, and bound MBP-VP31 detected with anti-MBP antibody (as described above). Each experiment was done twice (three wells each time).

In vivo neutralization of WSSV with the MBP-VP31 and PmLamr-His

Batches of L. vannamei (~3 g of body weight) were used for in vivo neutralization assays. The L. vannamei were bought from National Taiwan Ocean University’s Aquatic Animal Center, and then before experiments were started for at least 3 to 5 d the shrimp were acclimatized in water tanks with a salinity of 33 ± 1 ppt at 25 ± 1°C. The inoculum used was prepared from WSSV-T1 strain, as described [26]. For in vivo neutralization, shrimp were allocated into five groups (20 shrimp in each group). First, a constant volume (50 μl) of 10−7 dilution of original WSSV inoculum was mixed with 4 μg of MBP-VP31, PmLamr-His, or MBP, for 1 h on ice. These mixtures were used to challenge L. vannamei (intramuscular injections). Groups of shrimp injected with WSSV (10−7 viral diluent) mixed with MBP or 1× PBS buffer served as positive and negative controls, respectively. Shrimp were examined twice daily for 11 d and mortalities recorded. The Kaplan-Meier plot (log-rank test) was used to compare mortality.

Results

PmLamr interacted with nine WSSV structural proteins

An auto-activation test for GAL4 DBD fused PmLamr found that in the absence of prey protein, reporter genes in Y2HGold did not autonomously activate. GAL4 DBD fused PmLamr was also not found to be toxic to yeast cells (data not shown). Therefore, it was used as bait to screen the prey library. Yeast two-hybrid mating detected nine WSSV structural proteins (Fig 1) that interacted with PmLamr, including VP16, VP19, VP31, VP38B, VP41A, VP41B, VP52B, VP150-N (N-terminal fragment [aa 1–651] of VP150), VP150-C (C-terminal fragment [aa 641–1302] of VP150), VP187-N (N-terminal fragment [aa 1–804] of VP187), and VP187-C (C-terminal fragment [aa 810–1607] of VP187).

thumbnail
Fig 1. Yeast two-hybrid screening results for interactions between PmLamr and nine WSSV structural proteins.

(A) to (K) PmLamr’s interactions with VP16, VP19, VP31, VP38B, VP41A, VP41B, VP52B, VP150-N, VP150-C, VP187-N, and VP187-C, respectively. (a) Yeast grows on types of medium that lack both Leu and Trp (DDO), and this reveals the presence of each respective pair of plasmids. (b) and (c) indicate that yeast grows on low stringency (DDO/X/A) as well as high stringency (QDO/X/A) medium. Blue signals are seen in both (b) and (c) because of the presence of X-α-Gal. Protein-protein interactions are represented by yeast growth, whereas numbers around plates indicate the transformed yeast’s bait and prey plasmids: 1, pGBKT7-53/pGADT7-T; 2, pGBKT7-Lam/pGADT7-T; 3, pGBKT7-PmLamr/pGADT7-VPxx; 4, pGBKT7-PmLamr/pGADT7; 5, pGBKT7/pGADT7-VPxx; 6, pGBKT7/pGADT7, with VPxx respectively, representing each of the WSSV structural proteins listed above.

https://doi.org/10.1371/journal.pone.0156375.g001

PmLamr interacted with VP31 in a protein pull-down assay

One of the PmLamr interaction proteins identified by yeast two-hybrid screening was VP31, reported as having an important role in interactions with host cells and contributing to viral pathogenesis [2729]. Furthermore, it was also involved in recognition of integrin [30] and chitin-binding protein [31]. To further confirm interactions between PmLamr and VP31, a pull-down assay followed by Western blotting analysis using anti-PmLamr antibody was performed. All recombinant proteins for pull-down assays were successfully expressed (Fig 2A). The PmLamr-His was pulled down by MBP-VP31, but not by amylose resins nor MBP (Fig 2B; compare Lane 3 with Lanes 1 and 2), confirming that VP31 bound to shrimp receptor Lamr.

thumbnail
Fig 2. PmLamr interacted with WSSV VP31.

Protein pull-down assays of PmLamr-His with MBP-VP31. (A) SDS-PAGE of purified MBP, MBP-VP31 and PmLamr-His proteins. (B) PmLamr-His was incubated with amylose resins (Lane 1), MBP- (Lane 2), or MBP-VP31- (Lane 3) conjugated to amylose resins, pelleted, washed, and detected by immunoblotting (anti-PmLamr antibody).

https://doi.org/10.1371/journal.pone.0156375.g002

Membrane topology of VP31

Two approaches (TMHMM and HMMTOP) to predict VP31 membrane topology yielded very similar results: VP31 was predicted to lack transmembrane helices and to be located outside the envelope. To confirm the predictions concerning the topology of VP31, the WSSV virions were subjected to trypsin digestion in order to distinguish proteins that were accessible to proteolysis and those that were protected from digestion because of the lipid bilayer. The WSSV virions were either left untreated or treated with trypsin in the presence or absence of the detergent Triton X-100. Then, the digested products were analyzed with Western blotting, which used antibodies against VP31, VP26, or VP28. As anticipated, in untreated virions, anti-VP31 antibody recognized the VP31 proteins (Fig 3, Lane 1). However, the 31 kDa protein was no longer detected after trypsin digestion, regardless of whether it was in the presence or absence of Triton X-100 (Fig 3, Lanes 2 and 3). For comparison purposes, in order to detect the VP26 (a tegument protein, which is protected from trypsin digestion by the envelope) and the VP28 (an envelope protein with a portion of its C-terminal exposed outside the virion), the treated virions were also subjected to Western blotting. In the absence of Triton X-100, VP28 was digested into two bands, and in the presence of Triton X-100 it was completely digested. The small band was the transmembrane region of VP28 that was protected by envelope from trypsin digestion (Fig 3, Lanes 4–6). Furthermore, VP26 was digested only in the presence of Triton X-100, as it was not digested in the absence of Triton X-100 (Fig 3, Lanes 7–9).

thumbnail
Fig 3. Membrane topology of WSSV VP31.

Western blot analysis using corresponding antibodies showing trypsin digestion of VP31 (Lanes 1–3), and two other proteins are compared: VP28 (an envelope protein where most of its C-terminal is located on the envelope’s exterior; Lanes 4–6) and VP26 (a tegument protein; Lanes 7–9) in both intact (Lanes 2, 5 and 8) and detergent-treated virions (Lanes 3, 6 and 9). Lanes 1, 4 and 7: intact virion with neither trypsin nor Triton-X100 treatment.

https://doi.org/10.1371/journal.pone.0156375.g003

Molecular mass of PmLamr proteins

Using an antibody derived from the full-length PmLamr coding region, Western blot analysis was performed on the shrimp tissue proteins and recombinant proteins of E. coli and insect cells. The molecular mass of PmLamr overexpressed in the E. coli and in Drosophila S2 cells was ~51 kDa (Fig 4, Lanes 2 and 4). The Lamr between P. monodon and L. vannamei shared 97% identity in amino acid sequences [32]. Western blot analysis also successfully detected two major bands (37 and 44 kDa, respectively) in L. vannamei stomach tissue lysates, using antibody derived from recombinant PmLamr (Fig 4, Lane 5).

thumbnail
Fig 4. Molecular mass of shrimp Lamr and recombinant PmLamrs expressed in various expression systems.

Western blot analysis of lysates of pET-28b/PmLamr-His transformed E. coli cells, without or with induction of protein expression (Lanes 1 and 2, respectively), pDHsp-V5-His (empty vector) or pDHsp/PmLamr-V5-His transfected Drosophila S2 cells (Lanes 3 and 4, respectively), and L. vannamei stomach tissue (Lane 5), using anti-PmLamr antibody.

https://doi.org/10.1371/journal.pone.0156375.g004

Co-localization of PmLamr with VP31 on the surface of Drosophila S2 cells

Immunofluorescence analysis of non-permeabilized Drosophila S2 cells employing PmLamr- (Fig 5A) and MBP-specific (for detection of MBP-VP31; Fig 5B) antibodies demonstrated that PmLamr and VP31 co-localized on the surface of these cells (Fig 5D). When recombinant protein MBP was added to PmLamr-transfected Drosophila S2 cells, no Alexa Fluor® 488 dye signal was produced (control; data not shown).

thumbnail
Fig 5. Immunofluorescence images; note colocalization of PmLamr-V5-His with MBP-VP31 on cells.

Drosophila S2 cells were transfected with pDHsp/PmLamr-V5-His. At 48 h after transfection, 1 μg of MBP-VP31 was added. The presence of (A) PmLamr-V5-His (red) is visualized by rabbit anti-PmLamr antibody and Cy3 dye-conjugated goat anti-rabbit IgG antibody. As for (B) MBP-VP31 (green), mouse anti-MBP antibody and Alexa Fluor® 488 dye-conjugated goat anti-mouse IgG antibody were used. (C) The nuclear DNA was counterstained by DAPI. (D) Merged Cy3, Alexa Fluor® 488, and DAPI signals. Scale bar = 10 μm.

https://doi.org/10.1371/journal.pone.0156375.g005

VP31 and PmLamr binding specificity determined by ELISA and competitive ELISA

Increasing MBP-VP31 protein concentration in ELISAs enhanced ability to bind to PmLamr-His (Fig 6A), although there was no binding between MBP and PmLamr-His (control; Fig 6A). Therefore, MBP-VP31 binding to the PmLamr-His was specific and dose-dependent. In order to further clarify the relationship between VP31 and WSSV virion binding to PmLamr, a competitive ELISA assay was performed with MBP-VP31 and purified WSSV. When MBP-VP31 was kept constant (200 ng/100 μl), the OD value decreased as concentrations of WSSV viral proteins increased (from 10 to 1200 ng), whereas for control protein (BSA), OD value was approximately constant when BSA concentrations increased (Fig 6B). Therefore, the WSSV virion specifically competed with VP31 to bind PmLamr.

thumbnail
Fig 6. ELISA and competitive ELISA assays demonstrated WSSV VP31 binding to the PmLamr was specific.

(A) ELISA analysis of MBP-VP31 binding to the PmLamr-His in a dose-dependent manner. PmLamr-His (2 μg/ well) was coated to a 96-well plate and incubated with various amounts of purified MBP-VP31. MBP was added as negative control. (B) In competitive ELISA analysis, binding of MBP-VP31 and PmLamr-His were competed by WSSV virion proteins. PmLamr-His (2 μg/well) was coated to a 96-well plate. MBP-VP31 (200 ng) with various amounts of purified WSSV virion proteins (10–1200 ng) were added to wells and incubated. Addition of BSA (10–1200 ng) to wells served as a negative control. Mean (± SD) from three replicates.

https://doi.org/10.1371/journal.pone.0156375.g006

In vivo neutralization

For in vivo neutralization experiments using shrimp, with the exception of the PBS vehicle injection group, mortalities in the other three groups of shrimp increased steadily after 72 h post injection (hpi). Furthermore, 100% mortality occurred within 240 hpi in a positive control group (shrimp challenged with inoculum containing WSSV mixed with MBP [WSSV+MBP]). Notwithstanding, cumulative motilities were lower (* P<0.05) than in the positive control group at 240 and 264 hpi for shrimp injected with inoculum containing WSSV mixed with PmLamr-His (WSSV+ PmLamr-His), and at each time point from 144 to 264 hpi (** P<0.01) of shrimp injected with inoculum containing WSSV mixed with MBP-VP31 (WSSV+MBP-VP31; Fig 7). No shrimp mortality occurred in the PBS vehicle injected group, whereas all dead shrimp were WSSV-positive on PCR. Therefore, recombinant WSSV VP31 and PmLamr proteins delayed WSSV infection in shrimp in vivo.

thumbnail
Fig 7. Cumulative post-challenge shrimp mortality.

Shrimp were challenged with inoculum containing WSSV mixed with PmLamr-His (WSSV+PmLamr-His), or inoculum containing WSSV mixed with MBP-VP31 (WSSV+MBP-VP31). Asterisks indicated differences between positive and experimental groups (**P<0.01, *P<0.05). Shrimp injected with inoculum containing WSSV mixed with MBP (WSSV+MBP), or injected with PBS only, were positive and negative controls, respectively.

https://doi.org/10.1371/journal.pone.0156375.g007

Discussion

Based on outcomes of the present study, we concluded that PmLamr was a receptor for VP31. In that regard, Lamr is a multifunctional protein that acts as a receptor for viruses that cause infection in addition to being involved in a wide range of other biological processes [33]. Furthermore, Lamr of P. monodon (PmLamr) has been designated as a receptor for three viruses, namely TSV, IMNV and YHV [15, 16]. However, relationships between shrimp Lamr and WSSV, had not been adequately characterized. In the present study, PmLamr interacted with WSSV and mediated infection. In addition, the receptor’s interactions with viral compartments were also dissected. It was noteworthy that PmLamr interacted (yeast two-hybrid screening) with at least nine WSSV structural proteins. Nearly all these interactions were confirmed by co-immunoprecipitation (data not shown), although VP31 was not confirmed, as it was not expressed in insect cells. Regardless, the interaction between VP31 and PmLamr was also confirmed (pull-down assay). Based on neutralization experiments, VP31 was suggested as an important molecule in WSSV infection [27]. Consequently, VP31 was studied in detail.

It was reported that WSSV VP31 protein was an envelope protein (based on virion protein fractionation and immunoelectron microscopy; [22, 23, 26]. The present study confirmed VP31 is an envelope protein. After comparing both information and results concerning the membrane topology of VP31, we inferred that VP31 was an envelope protein, consistent with predictions of TMHMM and HMMTOP software. In the absence of typical transmembrane domains (predicted configurations and the trypsin digestion assay), VP31 may associate with other viral structural protein(s) by forming a complex to anchor on the envelope. Furthermore, interactions with WSSV envelope proteins VP24, VP32, and VP53 may cause VP31 to locate on the virion [31].

The corresponding gene of PmLamr encoded a protein of ~34.1 kDa. In the present study, there were two protein bands (recognized as PmLamr) in shrimp tissue which migrated on SDS-PAGE with a molecular weight of 37 and 44 kDa, respectively. Although the 37 kDa PmLamr band was not yet been reported in shrimp, the 44 kDa band was similar that detected in P. monodon and L vannamei muscle lysates [32]. A similar molecular mass discrepancy was reported in 37LRP/67LR, which was attributed to post-translational covalent modifications or formation of very tightly associated homo- or heterodimers [12, 34], although this is not well characterized.

In addition to yeast two-hybrid and pull-down assays, interactions between VP31 and PmLamr were also identified in cells. For example, in cells that overexpressed PmLamr-V5-His and treated with MBP-VP31, the latter co-localized with PmLamr-V5-His. In vitro binding assays further confirmed specificity of interactions between VP31 and PmLamr, whereas recombinant PmLamr and VP31 delayed or neutralized WSSV’s activity, thereby reducing mortality (in vivo neutralization assays). Collectively, based on these outcomes, we inferred that Lamr and VP31 had important roles in WSSV infection, namely that PmLamr was a cellular receptor for WSSV attachment and VP31 was involved in recognition of this receptor during virus infection.

It has been reported that Lamr most likely is unable to act as a singular receptor for infection. For example, adeno-associated virus serotype 2 (AAV2) appeared to rely on HSPG for cell attachment. As for efficient internalization and transduction, AAV2 requires Lamr interaction. The idea that there is a synergistic function of the two molecules for AAV2 transduction is further substantiated by evidence that shows Lamr and HSPG can form a complex on the cell surface [35]. In addition, WSSV VP31 was previously suggested as an important molecule involved in WSSV infection [27]. VP31 contained an integrin recognition RGD (conserved Arg-Gly-Asp sequence) motif; this motif was reported to be involved in host cell adhesion and might be associated with beta-integrin [29, 30]. Integrin is another type receptor of laminin. It has been suggested that the large protein 67LR facilitates interactions between laminin and integrins [36]. Furthermore, VP31 can also bind with Lamr and integrin. Binding of VP31 both with shrimp Lamr and integrin suggested that PmLamr may also serve as a co-receptor in shrimp cells to promote interactions between VP31 and integrin, and have a role in pathogenesis of WSSV infection.

Many believe that interactions between cellular receptors and viral attachment proteins are important in determining virus tissue tropism and host range. The WSSV is a virus with wide host range and often causes systemic infection in hosts. Homologues to Lamr were detected in all taxonomic domains of life [13]. In addition, Lamr was widely distributed in shrimp tissues [32]. Therefore, Lamr thus may contribute to the virus’ wide host range and tissue tropism. Elucidating relationships between WSSV components and Lamr should provide further insights regarding pathogenesis of WSSV infections and contribute to development of antiviral strategies for WSSV.

Acknowledgments

This investigation was supported financially by the Ministry of Science and Technology, R.O.C (MOST 104-2311-B-845-001). The authors thank Dr. Han-Ching Wang (Institute of Biotechnology, National Cheng Kung University) for providing the WSSV inoculum and purified WSSV virion.

Author Contributions

Conceived and designed the experiments: WJL. Performed the experiments: WJL YCL. Analyzed the data: WJL YCL GHK CFL. Contributed reagents/materials/analysis tools: WJL GHK CFL. Wrote the paper: WJL.

References

  1. 1. Vlak JM, Bonami JR, Flegel TW, Kou G-H, Lightner DV, Lo C-F, et al. Nimaviridae. In: Fauquet CM, Mayo MA, Maniloff J, Desselberger U, Ball LA, editors. VIIIth report of the international committee on taxonomy of viruses. Elsevier; 2005. p. 187–92.
  2. 2. Lo C-F, Ho C-H, Peng S-E, Chen C-H, Hsu H-C, Chiu Y-L, et al. White spot syndrome baculovirus (WSBV) detected in cultured and captured shrimp, crabs and other arthropods. Dis Aquat Organ. 1996;27:215–25.
  3. 3. Lo C-F, Peng S-E, Chang Y-S, Kou G-H. White spot syndrome–What we have learned about the virus and the disease. In: Walker PJ, Lester RG, Bondad-Reantaso MG, editors. Diseases in Asian Aquaculture V. Proceedings of the 5th Symposium on Diseases in Asian Aquaculture. Fish Health Section, Asian Fisheries Society. Manila; 2005 p. 421–33.
  4. 4. Escobedo-Bonilla CM, Alday-Sanz V, Wille M, Sorgeloos P, Pensaert MB, Nauwynck HJ. A review on the morphology, molecular characterization, morphogenesis and pathogenesis of white spot syndrome virus. J Fish Dis. 2008;31:1–18. pmid:18086030
  5. 5. Sánchez-Paz A. White spot syndrome virus: an overview on an emergent concern. Vet Res. 2010;41:43. pmid:20181325
  6. 6. Li DF, Zhang MC, Yang HJ, Zhu YB, Xu X. Beta-integrin mediates WSSV infection. Virology. 2007;368:122–32. pmid:17655902
  7. 7. Sritunyalucksana K, Wannapapho W, Lo C-F, Flegel TW. PmRab7 is a VP28-binding protein involved in white spot syndrome virus infection in shrimp. J Virol. 2006;80:10734–42. pmid:17041224
  8. 8. Chen L-L, Lu L-C, Wu W-J, Lo C-F, Huang W-P. White spot syndrome virus envelope protein VP53A interacts with Penaeus monodon chitin-binding protein (PmCBP). Dis Aquat Organ. 2007;74:171–8. pmid:17465302
  9. 9. Liang Y, Cheng JJ, Yang B, Huang J. The role of F1 ATP synthase beta subunit in WSSV infection in the shrimp, Litopenaeus vannamei. Virol J. 2010;7:144. pmid:20591132
  10. 10. Huang H-T, Leu J-H, Huang P-Y, Chen L-L. A putative cell surface receptor for white spot syndrome virus is a member of a transporter superfamily. PLoS One. 2012;7:e33216. pmid:22427993
  11. 11. Huang H-T, Chan H-L, Shih T-Y, Chen L-L. A study of the role of glucose transporter 1 (Glut1) in white spot syndrome virus (WSSV) infection. Fish Shellfish Immunol. 2015;46:305–14. pmid:26142142
  12. 12. Chen J, He WR, Shen L, Dong H, Yu J, Wang X, et al. The laminin receptor is a cellular attachment receptor for classical Swine Fever virus. J Virol. 2015;89:4894–906. pmid:25694590
  13. 13. DiGiacomo V, Meruelo D. Looking into laminin receptor: critical discussion regarding the non-integrin 37/67-kDa laminin receptor/RPSA protein. Biol Rev. 2015. [Epub ahead of print 2015 Jan 28]
  14. 14. Castronovo V. Laminin receptors and laminin-binding proteins during tumor invasion and metastasis. Invasion Metastasis. 1993;13:1–30. pmid:8407208
  15. 15. Senapin S, Phongdara A. Binding of shrimp cellular proteins to Taura syndrome viral capsid proteins VP1, VP2 and VP3. Virus Res. 2006;122:69–77. pmid:16889862
  16. 16. Busayarat N, Senapin S, Tonganunt M, Phiwsaiya K, Meemetta W, Unajak S, et al. Shrimp laminin receptor binds with capsid proteins of two additional shrimp RNA viruses YHV and IMNV. Fish Shellfish Immunol. 2011;31:66–72. pmid:21414409
  17. 17. Charoensapsri W, Sangsuriya P, Lertwimol T, Gangnonngiw W, Phiwsaiya K, Senapin S. Laminin receptor protein is implicated in hemocyte homeostasis for the whiteleg shrimp Penaeus (Litopenaeus) vannamei. Dev Comp Immunol. 2015;51:39–47. pmid:25720979
  18. 18. Liu W-J, Shiung H-J, Lo C-F, Lai Y-J, Lee T-L, Huang W-T, et al. Characterization and interactome study of white spot syndrome virus envelope protein VP11. PLoS One. 2014;9:e85779. pmid:24465701
  19. 19. Krogh A, Larsson B, von Heijne G, Sonnhammer EL. Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes. J Mol Biol. 2001;305:567–80. pmid:11152613
  20. 20. Tusnády GE, Simon I. Principles governing amino acid composition of integral membrane proteins: application to topology prediction. J Mol Biol. 1998;283:489–506. pmid:9769220
  21. 21. Tusnády GE, Simon I. The HMMTOP transmembrane topology prediction server. Bioinformatics. 2001;17:849–50. pmid:11590105
  22. 22. Tsai J-M, Wang H-C, Leu J-H, Wang AH-J, Zhuang Y, Walker PJ, et al. Identification of the nucleocapsid, tegument, and envelope proteins of the shrimp white spot syndrome virus virion. J Virol. 2006;80:3021–9. pmid:16501111
  23. 23. Kang S-T, Leu J-H, Wang H-C, Chen L-L, Kou G-H, Lo C-F. Polycistronic mRNAs and internal ribosome entry site elements (IRES) are widely used by white spot syndrome virus (WSSV) structural protein genes. Virology. 2009;387:353–63. pmid:19285701
  24. 24. Xie X, Yang F. Interaction of white spot syndrome virus VP26 protein with actin. Virology. 2005;336:93–9. pmid:15866074
  25. 25. Leu J-H, Kuo Y-C, Kou G-H, Lo C-F. Molecular cloning and characterization of an inhibitor of apoptosis protein (IAP) from the tiger shrimp, Penaeus monodon. Dev Comp Immunol. 2008;32:121–33. pmid:17628672
  26. 26. Liu W-J, Chang Y-S, Wang AH-J, Kou G-H, Lo C-F. White spot syndrome virus annexes a shrimp STAT to enhance the expression of the immediate-early gene ie1. J Virol. 2007;81:1461–71. pmid:17079306
  27. 27. Li L, Xie X, Yang F. Identification and characterization of a prawn white spot syndrome virus gene that encodes an envelope protein VP31. Virology. 2005;340:125–32. pmid:16023692
  28. 28. Li LJ, Yuan JF, Cai CA, Gu WG, Shi ZL. Multiple envelope proteins are involved in white spot syndrome virus (WSSV) infection in crayfish. Arch Virol. 2006;151:1309–17. pmid:16489508
  29. 29. Li L, Lin Z, Xu L, Yang F. The RGD motif in VP31 of white spot syndrome virus is involved in cell adhesion. Arch Virol. 2011;156:1317–21. pmid:21448738
  30. 30. Zhang JY, Liu QH, Huang J. Multiple proteins of white spot syndrome virus involved in recognition of beta-integrin. J Biosci. 2014;39:381–8. pmid:24845502
  31. 31. Huang P-Y, Leu J-H, Chen L-L. A newly identified protein complex that mediates white spot syndrome virus infection via chitin-binding protein. J Gen Virol. 2014;95(Pt 8):1799–808. pmid:24836670
  32. 32. Senapin S, Phiwsaiya K, Anantasomboon G, Sriphaijit T, Browdy CL, Flegel TW. Knocking down a Taura syndrome virus (TSV) binding protein Lamr is lethal for the white leg shrimp Penaeus vannamei. Fish Shellfish Immunol. 2010;29:422–9. pmid:20451618
  33. 33. Kielian M, Chanel-Vos C, Liao M. Alphavirus entry and membrane fusion. Viruses. 2010;2:796–825. pmid:21546978
  34. 34. Nelson J, McFerran NV, Pivato G, Chambers E, Doherty C, Steele D, et al. The 67 kDa laminin receptor: structure, function and role in disease. Biosci Rep. 2008;28:33–48. pmid:18269348
  35. 35. Akache B, Grimm D, Pandey K, Yant SR, Xu H, Kay MA. The 37/67-kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8, 2, 3, and 9. J Virol. 2006;80:9831–6. pmid:16973587
  36. 36. Ardini E, Tagliabue E, Magnifico A, Buto S, Castronovo V, Colnaghi MI, et al. Co-regulation and physical association of the 67-kDa monomeric laminin receptor and the α6β4 integrin. J Biol Chem. 1997;272:2342–5. pmid:8999943