Sei sulla pagina 1di 10

JOURNAL OF VIROLOGY, Jan. 2009, p. 347–356 Vol. 83, No.

1
0022-538X/09/$08.00⫹0 doi:10.1128/JVI.00707-08
Copyright © 2009, American Society for Microbiology. All Rights Reserved.

A Novel C-Type Lectin from the Shrimp Litopenaeus vannamei


Possesses Anti-White Spot Syndrome Virus Activity䌤
Zhi-Ying Zhao,1† Zhi-Xin Yin,1 Xiao-Peng Xu,1 Shao-Ping Weng,1 Xia-Yu Rao,1 Zong-Xian Dai,1
Yong-Wen Luo,1 Gan Yang,1 Zong-Sheng Li,1 Hao-Ji Guan,1 Se-Dong Li,3 Siu-Ming Chan,4
Xiao-Qiang Yu,2* and Jian-Guo He1*
State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen (Zhongshan) University, 135 Xingang Road West,
Guangzhou 510275, People’s Republic of China1; Division of Cell Biology and Biophysics, School of Biological Sciences,
University of Missouri—Kansas City, Kansas City, Missouri 641102; Fisheries Research Institute of Zhanjiang,
Zhanjiang 524039, Guangdong, China3; and Department of Zoology, the University of
Hong Kong, Pokfulam Road, Hong Kong SAR, China4
Received 29 March 2008/Accepted 6 October 2008

C-type lectins play key roles in pathogen recognition, innate immunity, and cell-cell interactions. Here, we
report a new C-type lectin (C-type lectin 1) from the shrimp Litopenaeus vannamei (LvCTL1), which has activity
against the white spot syndrome virus (WSSV). LvCTL1 is a 156-residue polypeptide containing a C-type
carbohydrate recognition domain with an EPN (Glu99-Pro100-Asn101) motif that has a predicted ligand binding
specificity for mannose. Reverse transcription-PCR analysis revealed that LvCTL1 mRNA was specifically
expressed in the hepatopancreas of L. vannamei. Recombinant LvCTL1 (rLvCTL1) had hemagglutinating
activity and ligand binding specificity for mannose and glucose. rLvCTL1 also had a strong affinity for WSSV
and interacted with several envelope proteins of WSSV. Furthermore, we showed that the binding of rLvCTL1
to WSSV could protect shrimps from viral infection and prolong the survival of shrimps against WSSV
infection. Our results suggest that LvCTL1 is a mannose-binding C-type lectin that binds to envelope proteins
of WSSV to exert its antiviral activity. To our knowledge, this is the first report of a shrimp C-type lectin that
has direct anti-WSSV activity.

Lectins are a group of nonimmunogenic proteins possessing addition, MBL has also been shown to link the innate immune
at least one noncatalytic domain that binds reversibly to spe- system to the acquired immune system (2, 10).
cific carbohydrates and usually agglutinates cells (16, 35). They C-type lectins in invertebrates are also involved in innate im-
exist ubiquitously in animals, plants, and bacteria. Due to their mune responses, including the promotion of phagocytosis (18,
ability to bind to specific carbohydrates on the surfaces of 28), nodule formation, encapsulation, melanization (20, 48), and
microorganisms, lectins have been regarded as primary candi- the activation of prophenoloxidase (48, 49). A number of C-type
dates for pattern recognition receptors (PRRs) in animal in- lectins in invertebrates, particularly in insects and shrimps, have
nate immunity. Calcium-dependent (C-type) lectins, which been isolated and characterized (20, 25, 28, 29, 31, 36, 46–49). In
contain a characteristic carbohydrate recognition domain the shrimp Penaeus monodon, two C-type lectins containing a
(CRD) with two or three pairs of disulfide bonds, are believed single CRD have been characterized (28, 29). In Penaeus japoni-
to mediate pathogen recognition and play an important role in cus, an N-acetylglucosamine (GlcNAc)-specific lectin and another
the clearance of pathogens in innate immunity (6, 15, 42, 47). lectin have also been reported (21, 46). A C-type lectin in Litope-
C-type lectins can be further classified into a number of sub- naeus vannamei with two CRDs (LvCTL) is expressed, but only in
groups based on their functions and structures (5, 7). Mam- the hepatopancreas (31). In Fenneropenaeus chinensis, two C-type
malian mannose-binding lectin (MBL), a liver-derived pluri- lectins (Fclectin [FcCLec-1] and Fc-hsL [FcCLec-2]) have been
potent serum C-type lectin, belongs to the collectin subgroup reported (25, 36). FcCLec-1 contains dual CRDs and is expressed
and has been well studied. MBL is a PRR that can activate the in hemocytes, while Fc-hsL contains only one CRD and is ex-
complement system through the lectin pathway (11). It has
pressed specifically in the hepatopancreas. These shrimp C-type
been suggested that MBL plays an important role in host
lectins have ligand binding specificities for carbohydrates such as
defense as a primary immune response to pathogens (9, 38). In
N-acetylglucosamine and lipopolysaccharide, and they can agglu-
tinate erythrocytes and exhibit antimicrobial activity against some
* Corresponding author. Mailing address for Jian-Guo He: State bacteria and fungi. Moreover, many expressed sequence tags
Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen (ESTs) encoding lectins have been identified in cDNA libraries
(Zhongshan) University, Guangzhou 510275, People’s Republic of
from healthy shrimps (P. japonicus, Litopenaeus setiferus, and L.
China. Phone: 86 20 8411 0976. Fax: 86 20 8411 3819. E-mail: lsshjg
@mail.sysu.edu.cn. Mailing address for Xiao-Qiang Yu: Division of vannamei) (12, 14), and partial cDNAs for C-type lectins are also
Cell Biology and Biophysics, School of Biological Sciences, University obtained by subtraction using shrimps (L. vannamei) infected
of Missouri—Kansas City, Kansas City, MO 64110. Phone: (816) 235 with white spot syndrome virus (WSSV) and healthy controls
-6379. Fax: (816) 235-1503. E-mail: Yux@umkc.edu.
(51). However, almost all shrimp C-type lectins exhibit activity
† Present address: Hainan Provincial Fisheries Research Institute,
Haikou 570206, Hainan, China. mainly against bacteria or fungi, and little is known about the

Published ahead of print on 22 October 2008. effects of C-type lectins in response to viruses such as WSSV.

347
348 ZHAO ET AL. J. VIROL.

The Pacific white shrimp, L. vannamei, is a commercially TABLE 1. Primers used for this experiment
important species of cultured penaeid shrimps in China, as well Primer name Sequence (5⬘–3⬘)
as worldwide. With the rapid production and development of
LvCTL1 cDNA cloning
L. vannamei worldwide, shrimp diseases have become wide- GSP1......................................CTGTCCTTACCCCTACGAGCCC
spread and, in recent years, have threatened the shrimp indus- TTGG
try. WSS, which is caused by WSSV, is the most severely GSP2......................................GCTCCTAACCAGAAGTGCTTGCC
damaging disease of shrimps and other crustaceans around the GSP3......................................TGTCGACACTTGACGCAAGACC
GSP4......................................CAGTACTATTCAATATTTCTT
world (13, 24, 26). The outbreak and spread of WSS have
GAGG
resulted in the high mortality of farmed penaeid shrimps and
huge economic losses in many regions of the world, especially RT-PCR
in Southeast Asia (13, 24, 26, 39). The viral pathogen, WSSV, LvCTL1-F .............................TCTGTCGCCAGTGTTCGTTC
is a member of a new genus, Whispovirus, in the Nimaviridae LvCTL1-R.............................CGTATTCCCACTATTAGGCTC
␤-actin-F................................AGATGTGTGACGACGAAGTA
family (41), with double-stranded DNA (about 300 kb) and GCCG
envelope proteins (39, 45). Since the first report of WSSV in ␤-actin-R ...............................TCACGAACGATTTCTCGCTCG
1993, major concerns in the world aquaculture industry have
been raised, and great effort has been made in preventing and Recombinant expression
Re-F.......................................CGG/GATCCAATGACTGTCCTTA
controlling the disease in recent years (24, 26, 39, 45). Even so,
CCCCTACa
shrimp defense mechanisms, particularly those against viruses, Re-R ......................................TCCCC/CCGGGTTAATATTTCTTG
are poorly understood. AGGCAAATGb
The shrimp defense system is believed to rely largely on a
The BamHI restriction site is underlined.
innate immunity. The innate immune systems of shrimps con- b
The XmaI restriction site is underlined.
sist of PRRs that recognize and bind to specific patterns on the
surfaces of pathogens (3, 22). Previously, we employed sup-
pression subtractive hybridization technology to differentially ABI97373), PsCLec from Penaeus semisulcatus (accession no. ABI97372), Lv-
screen cDNA libraries from normal and WSSV-infected L. CLec (accession no. ABI97374), FcCLec-1 from F. chinensis (accession no.
AAX63905), FcCLec-2 (accession no. ABA54612), PmAV from P. monodon
vannamei shrimps by using WSSV-resistant L. vannamei as the
(accession no. AAQ75589), and PmCLec-2 (accession no. AAZ29608). Multiple-
tester and WSSV-susceptible L. vannamei as the driver (51). sequence alignment of the LvCTL1 CRD with CRDs from other shrimp C-type
More than 1,000 qualified ESTs were obtained from the pos- lectins was performed with the ClustalW multiple-alignment software.
itive clones of the cDNA libraries. Many ESTs encoding C- Expression of LvCTL1 in tissues. Total RNA was prepared from the hepato-
type lectins are upregulated in virus-resistant shrimps com- pancreases, hemocytes, gills, eyestalks, muscles, brains, hearts, pyloric ceca,
nerve cords, intestines (midgut), spermaries, and stomachs of healthy WSSV-
pared to their levels of regulation in susceptible shrimps, susceptible L. vannamei shrimps. All of the RNA samples were treated with
particularly in virus-resistant shrimps at 48 h postinfection DNase (Promega) to remove contaminating DNA, and the single-stranded
(51). Subsequently, several of these lectins have been cloned cDNAs were synthesized using Moloney murine leukemia virus reverse trans-
and characterized. criptase (RT; Promega) with an oligo(dT)18 primer by following the manufac-
turer’s instructions. Equal amounts of cDNAs from the shrimp tissues were used
In this study, we report a novel anti-WSSV C-type lectin,
as templates for PCR. The LvCTL1 cDNA fragment was amplified with primers
CTL1, from the shrimp L. vannamei (LvCTL1). LvCTL1 con- LvCTL1-F and LvCTL1-R (Table 1), using the following conditions: initial
tains a single CRD. It exhibits hemagglutinating and sugar denaturation at 94°C for 3 min and then 30 cycles of 94°C for 30 s, 55°C for 30 s,
binding activities as well as a strong affinity for WSSV. LvCTL1 and 72°C for 50 s, followed by a final extension at 72°C for 8 min. The consti-
binds to several envelope proteins from WSSV virions. More tutively expressed ␤-actin gene was amplified with the specific primers ␤-actin-F
and ␤-actin-R (Table 1) and used to normalize PCRs. The PCR products were
importantly, LvCTL1 exhibits high antiviral activity against analyzed on a 1% agarose gel and sequenced to confirm the identity of the
WSSV both in vitro and in vivo. LvCTL1 cDNA.
Expression and purification of recombinant LvCTL1. A cDNA fragment en-
coding a mature LvCTL1 protein (residues 21 to 156) (Fig. 1A) was amplified by
MATERIALS AND METHODS
PCR using Taq polymerase (Promega) with the specific primers Re-F and Re-R
Cloning of full-length LvCTL1 cDNA. A partial cDNA sequence of LvCTL1 (Table 1). BamHI and XmaI restriction sites were added to the 5⬘ ends of Re-F
was obtained from the clone RS4E82, which was isolated from the subtraction and Re-R (after the stop codon), respectively (Table 1). The PCR fragment was
cDNA library described previously (51); the sequence was homologous to a cloned into the pGem-T easy vector (Promega), completely digested with BamHI
C-type lectin from P. monodon (PmAV; GenBank accession no. AAQ75589) and XmaI (NEB, United Kingdom), and then cloned into the BamHI/XmaI sites
(29). Based on the identified EST sequences, gene-specific primers (GSP1 and of the expression vector pQE-30 (Qiagen, Germany). The recombinant plasmid
GSP2) were designed for the rapid amplification of 3⬘ and 5⬘ cDNA ends (pQE-30-LvCTL1) was transformed into competent Escherichia coli M15(pREP4)
(RACE) to amplify the full-length cDNA of LvCTL1. Then, gene-specific prim- (Qiagen, Germany) cells for expression of recombinant proteins. Bacterial cells
ers GSP3 and GSP4 were designed to confirm the full-length cDNA of LvCTL1 expressing recombinant proteins were then harvested and sonicated, and the inclu-
from 3⬘- and 5⬘-end-overlapping fragments (Table 1). RACE was carried out sion bodies were resuspended in 1⫻ phosphate-buffered saline (PBS) containing 8
using a BD SMART RACE cDNA amplification kit (Clontech, CA), using the M urea. Recombinant LvCTL1 protein was purified by affinity chromatography with
adapter-ligated cDNA, which was synthesized from total RNA of the hepato- Ni-nitrilotriacetic acid-agarose (Qiagen, Germany) under denaturing (8 M urea)
pancreas according to the manufacturer’s instructions. DNA was sequenced with conditions according to the manufacturer’s instructions (Qiagen, Germany). Purified
an ABI 3700 DNA sequencer (Perkin-Elmer/Applied Biosystems), and the re- recombinant protein was dialyzed against 1⫻ PBS-glycerol buffer (PBS with 5%
sulting sequences were subjected to cluster and homology analyses. glycerol, pH 7.0) at 4°C overnight. The resulting protein was analyzed by 15%
Sequence analysis. The similarity analysis of the nucleotide and amino acid sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and visu-
sequences of LvCTL1 was carried out using the BLAST programs at the NCBI alized with Coomassie brilliant blue R-250.
(http://www.ncbi.nlm.nih.gov/blast). Translation of the cDNA was performed Production of antibodies. The polyclonal antibody against recombinant
using the Expert Protein Analysis System (http://au.expasy.org/). Signal sequence LvCTL1 was produced by immunizing BALB/c mice according to the conven-
and CRD domain predictions were conducted using SMART software. Shrimp tional method (34). The titer of the antiserum was then determined by an
C-type lectins used for comparisons include PmCLec-1 (GenBank accession no. enzyme-linked immunosorbent assay (ELISA) to determine the optimal working
FIG. 1. L. vannamei LvCTL1 cDNA and amino acid sequences. (A) Full-length cDNA and deduced amino acid sequences. The amino acid sequence
is represented by a single capital letter below the nucleotide sequence (GenBank accession no. DQ858900). The putative signal peptide sequence is
underlined; four conserved cysteine residues that define the C-type lectin domain are shaded, and four additional cysteine residues are shaded and
underlined. The EPN motif for ligand binding specificity is boxed. The polyadenylation signal sequence AATAAA is double underlined. (B) Multiple-
sequence alignment of the CRD of LvCTL1 with CRDs of other shrimp C-type lectins. The CRDs of LvCTL1 and four other shrimp C-type lectins were
aligned using ClustalX. Conserved cysteine residues that define the C-type lectin domain are shaded and indicated by asterisks. The EPN or QPD motif
is also shaded. Shown are LvCLec-1 (accession no. DQ858899, lec5D), PmCLec-1-CRD1 (accession no. ABI97373), PmCLec-1-CRD2 (accession no.
ABI97373), PsCLec-CRD1 (accession no. ABI97372), PsCLec-CRD2 (accession no. ABI97372), LvCTL-CRD1 (accession no. ABI97374), LvCTL-
CRD2 (accession no. ABI97374), FcCLec-1-CRD1 (accession no. AAX63905), FcCLec-1-CRD2 (accession no. AAX63905), FcCLec-2 (accession no.
ABA54612), PmAV (accession no. AAQ75589), and PmCLec-2 (accession no. AAZ29608).

349
350 ZHAO ET AL. J. VIROL.

concentration. Recombinant LvCTL1 protein samples were separated using 15% pended in saline solution and seeded in a 48-well microplate at about 4 ⫻ 105
SDS-PAGE, and the proteins were transferred to a nitrocellulose membrane cells per well. Then, 50 ␮l of the hemocyte suspension in each well of a 48-well
(Roche Biosciences, Germany). The membrane was blocked with blocking buffer microplate was mixed with 50 ␮l of twofold serial dilutions of recombinant
containing 5% nonfat milk powder in PBS with 0.1% Tween 20 (PBS-T) for 2 h LvCTL1 in saline solution and incubated for 1 h at 25°C. Agglutination of
and then incubated with mouse anti-LvCTL1 antibody (or monoclonal antibody hemocytes was observed under a microscope (Nikon, Japan). Hemocytes mixed
against the His tag; Novagen) for 2 h, followed by incubation with peroxidase- with serial dilutions of BSA (Sigma) were processed in parallel as controls.
conjugated secondary antibody (1:10,000 dilution in PBS-T; Promega) for 1.5 h. To test whether calcium is required for the hemagglutination activity of
LvCTL1-specific signals were visualized after the addition of diaminobenzidine LvCTL1, 12.5 ␮l of serial dilutions of EDTA in TBS was mixed with 12.5 ␮l of
colorimetric chromogen (Boster, China). recombinant LvCTL1 (⬃12 ␮g/ml), 25 ␮l 2% trypsin-treated mouse erythrocytes
Expression of LvCTL1 protein in the hemolymph of shrimps after WSSV was added, and the mixture was incubated for 30 min at 25°C. Hemagglutination
infection. To detect LvCTL1 protein in the hemolymph of shrimps after WSSV of erythrocytes was observed under a microscope (Nikon, Japan). Assays were
infection, healthy WSSV-susceptible L. vannamei shrimps were challenged with performed in triplicate.
WSSV as described previously (51), and hemolymph was collected from the Sugar binding specificity of LvCTL1. The sugar binding specificity of recom-
pericardial sinus located at the first abdominal segment of each shrimp at 3, 6, 12, binant LvCTL1 was determined by an inhibitory agglutination assay. Serial di-
24, 36, 48, and 72 h postinjection, using a 1-ml sterile syringe. The hemolymph lutions (12.5 ␮l) of various carbohydrates (Sigma) in TBS-Ca, including D-
was then immediately centrifuged for 10 min at 5,000 ⫻ g at 4°C, and the cell-free mannose, D-fructose, D-galactose, lactose, D-glucose, and sucrose, were mixed
plasma was collected. Plasma from unchallenged shrimps was also prepared by with 12.5 ␮l of recombinant LvCTL1 (⬃12 ␮g/ml) and incubated for 30 min at
the same method. The plasma samples were first diluted with distilled water (1:5, 4°C. Then, 2% trypsin-treated mouse erythrocytes were added, and the mixture
vol/vol) and then subjected to 15% SDS-PAGE, and proteins were transferred to was incubated for 30 min at 25°C. An inhibitory effect was expressed as the
a nitrocellulose membrane (Roche Biosciences, Germany). The membrane was minimum concentration of a carbohydrate required for complete inhibition of
blocked with 5% nonfat milk powder in Tris-buffered saline (TBS) (10 mM the hemagglutinating activity of LvCTL1. This assay was performed in triplicate.
Tris-HCl, pH 7.2, 150 mM NaCl) for 4 h. After being washed three times with Binding of recombinant LvCTL1 to WSSVs. Intact WSSVs were purified from
TBS with 0.1% Tween 20 (TBS-T), the membrane was then incubated with the the gills of infected L. vannamei shrimps according to the method described by
mouse antiserum against LvCTL1 for 2 h. Antibody binding was visualized by a Xie et al. (44). The virus samples were examined using electron microscopy for
colorimetric reaction catalyzed by peroxidase-conjugated goat anti-mouse anti- purity. Purified WSSV was dotted onto a nitrocellulose membrane (Roche Bio-
body (1:10,000 dilution in TBS; Promega). sciences, Germany), and the membrane was blocked with a blocking buffer
ELISA-based assay of LvCTL1 expression in the hemolymph of WSSV-resis- containing 5% milk powder in TBS-T overnight. The membrane was washed
tant and -susceptible shrimps. WSSV-resistant and -susceptible L. vannamei three times in TBS-T and then probed with recombinant LvCTL1, Lypn (a fish
shrimps were obtained by means of a selective-breeding program (51). Two protein also expressed using the pET-32a vector [unpublished]), or pET-32a-
families were selected to perform an ELISA-based assay for LvCTL1 expression. His-tag. (Expression of the empty pET-32a vector [Novagen] was performed in
Family A has a survival rate of up to 70% after WSSV infection, while family B Escherichia coli BL21 cells according to the manufacturer’s instructions, and
has a mortality rate of 100% after WSSV infection. Hemolymph samples were pET-32a-His-tag was purified according to the same method described under
collected in tubes containing 10% saline solution (0.9%, vol/vol, NaCl) from both “Expression and purification of recombinant LvCTL1” above.) Then, Western
WSSV-resistant and -susceptible L. vannamei shrimps at 3, 6, 12, 24, 36, 48, and blotting was performed with monoclonal antibody against the His tag to confirm
72 h postinjection. Hemolymph from unchallenged shrimps was also prepared by the presence of the polypeptides in TBS-T containing 0.1% BSA for 2 h at room
the same method. Hemolymph (200 ␮l) was mixed with commercial extraction temperature. The binding of recombinant proteins to WSSV was detected by
reagent (KeyGen, China) (300 ␮l) on ice for 20 min and then centrifuged at immunoblotting, using mouse monoclonal antibody to the His tag (Novagen) or
12,000 ⫻ g at 4°C for 5 min. The supernatant (diluted plasma) was removed to mouse polyclonal antibody specific for LvCTL1 for 2 h, followed by incubation
new tubes, and phenylmethylsulfonyl fluoride was added to each diluted plasma with goat anti-mouse immunoglobulin G conjugated to peroxidase as a secondary
sample. The protein concentration in the diluted plasma was determined using a antibody (1:10,000 dilution in PBS-T; Promega) for 1.5 h at room temperature.
bicinchoninic acid protein assay kit (Pierce), and the plasma was then stored at Then, a 3,3-diaminobenzidine solution (Boster, China) was added to develop the
⫺80°C for further use. brown product for 5 min, and the reaction was stopped with water.
For ELISA, the 96-well microtiter plates (Costar) were coated overnight at Interaction of recombinant LvCTL1 with WSSV proteins by protein pull-down
4°C with the diluted plasma (100 ␮l per well, ⬃20 ␮g total protein per well). assay. The cDNA fragment encoding mature LvCTL1 (residues 21 to 156) (Fig.
Wells coated with bovine serum albumin (BSA) were used as negative controls. 1A) was cloned into the pMAL-c2X plasmid (NEB, United Kingdom) to express
The plates were washed three times and blocked with blocking buffer containing a maltose-binding protein (MBP)-lectin fusion protein in E. coli BL21 cells. The
0.5% BSA in PBS-T for 2 h at 37°C. The plates were then incubated with mouse MBP or MBP-lectin fusion protein was fixed to amylose resins (NEB, United
anti-LvCTL1 antibody (1:500 dilution with blocking buffer) for 2 h, followed by Kingdom). The purified WSSV particles were lysed with radio immunoprecipi-
incubation with peroxidase-conjugated secondary antibody (goat anti-mouse im- tation buffer supplemented with protease inhibitor (Calbiochem). The viral ex-
munoglobulin G antibody, 1:2,000 dilution in PBS-T; Promega) for 2 h. After the tract was centrifuged at 14,000 rpm for 15 min at 4°C, and the supernatant was
plates were rinsed with PBS-T, the bound peroxidase activity was determined by diluted five times with TBS containing 1% Triton X-100, 10 mM EDTA, and
a reaction with TMB (3,3⬘,5,5⬘-tetramethylbenzidine), and the optical density protease inhibitor. Then, the viral protein extract was added to the MBP or
(OD) value was measured by spectrophotometry, using 450-nm filters. All MBP-lectin fusion protein-fixed amylose resins, and the mixture was incubated
ELISAs were performed in triplicate, with the data given in mean values. for 3 h at 4°C. The amylose resins were then washed with TBS buffer containing
Hemagglutination and hemocyte aggregation assays. The hemagglutinating 1% Triton X-100, and the pull-down proteins were analyzed by SDS-PAGE. The
activity of recombinant LvCTL1 was tested using 2% (vol/vol) erythrocytes from bands pulled down by MBP-lectin but not by MBP were excised from gels, cut
rabbits, mice, chickens, and fish according to the method described by Luo et al. into pieces, and transferred to a 1.5-ml microcentrifuge tube.
(28). Erythrocytes were washed three times with TBS-Ca buffer (50 mM Tris- Protein identification by mass spectrometry. The in-gel enzymatic digestion
HCl, 100 mM NaCl, 10 mM CaCl2, pH 7.0) and then suspended at 2% (vol/vol) and mass spectrometry analysis were performed by the Shanghai Institutes for
in TBS-Ca buffer. Twofold serial dilutions (25 ␮l) of recombinant LvCTL1 in Biological Sciences, Research Center for Proteome Analysis. The digested pro-
TBS-Ca buffer were mixed with 25 ␮l of the erythrocytes in a microtiter U plate. tein was separated and identified with a Finnigan LTQ mass spectrometer (Thermo-
The plate was incubated for 1 h at room temperature, and the hemagglutination Quest, San Jose, CA), coupled with a Surveyor high-performance liquid
was observed under a microscope (Nikon, Japan). Erythrocytes mixed with serial chromatography system (ThermoQuest). First, a Microcore reverse-phase col-
dilutions of BSA (Sigma) in TBS-Ca buffer were processed in parallel with the umn (C18, 0.15 mm by 120 mm; Thermo Hypersil, San Jose, CA) was used to
above-described erythrocytes to serve as controls. separate the protein digests. Solvent A was 0.1% (vol/vol) formic acid, and
To test the aggregation of shrimp hemocytes by recombinant LvCTL1, hemo- solvent B was 0.1% (vol/vol) formic acid in 100% (vol/vol) acetonitrile. The
cytes of L. vannamei shrimps were cultured in modified Leibovitz’s L-15 medium solvent B gradient was held at 2% for 15 min and increased linearly to 98% in 90
(Gibco) in vitro at 24 ⫾ 0.5°C with 5% ⫾ 0.2% CO2, used for the hemocyte min. The peptides were eluted from the C18 microcapillary column at a flow rate
aggregation assay (for hemocyte culture, see “In vitro anti-WSSV activity of of 150 ␮l per min and then electrosprayed directly into the mass spectrometer,
recombinant LvCTL1” below). After 6 days of culture in a 48-well microplate, with the application of a spray voltage of 3.2 kV and with a capillary temperature
the hemocytes were removed by treatment with trypsinase (Promega) at 37°C for of 170°C. The full scan ranges from m/z 400 to 2,000. Protein identification using
5 min, collected, and washed three times with saline solution (0.9% NaCl) by raw data from tandem mass spectrometry was performed with SEQUEST soft-
centrifugation at 200 ⫻ g for 10 min. Hemocytes were then thoroughly resus- ware (University of Washington, licensed to Thermo Finnigan) based on se-
VOL. 83, 2009 C-TYPE LECTIN FROM L. VANNAMEI HAS ANTI-WSSV ACTIVITY 351

quences in the Swiss-Prot database. The species is WSSV. A relative molecular Comparison of LvCTL1 with other shrimp C-type lectins.
mass of 57 Da was added to the average molecular mass of cysteines in tandem- Database searches with the deduced amino acid sequence of
mass spectrometry data searching. Both b ions and y ions were included in the
database search.
LvCTL1 showed that LvCTL1 is most similar to the C-type
In vitro anti-WSSV activity of recombinant LvCTL1. To test whether recom- lectin FcCLec-2 (GenBank accession no. ABA54612) (77%
binant LvCTL1 has anti-WSSV activity, a coculture assay was performed. This identity). The CRD of LvCTL1 was also highly similar to that
assay is based on the percentages of cytopathic effects (CPE) and dead cells of FcCLec-2 (accession no. ABA54612) (79% identity), and it
induced by WSSV infection relative to those of normal cells. Hemocytes from
is also similar to CRDs of PsCLec (accession no. ABI97372)
healthy L. vannamei shrimps were collected as the primary cells and challenged
with WSSV according to the method described by Jiang et al. (17). The L. (34% identity), LvCTL (accession no. ABI97374) (34%),
vannamei hemocytes were cultured in vitro at 24 ⫾ 0.5°C with 5% ⫾ 0.2% CO2 PmCLec-1 (accession no. ABI97373) (32%), and PmAV (ac-
in a 96-well microplate in the modified Leibovitz’s L-15 medium (80% 1⫻ cession no. AAQ75589) (28%). Based on the BLASTP analy-
Leibovitz’s L-15; Gibco) and supplemented with 15% fetal bovine serum (Hy- sis, several shrimp C-type lectins were selected and aligned
Clone), 1.0 g/liter glucose, 0.3 g/liter glutamine, 0.1 mg/liter vitamin C, 12.0 g/liter
NaCl, 100 IU/ml penicillin, and 100 ␮g/ml streptomycin sulfate (pH 7.2). The
with LvCTL1 (Fig. 1B). These shrimp lectins contain at least
cells were observed daily with an inverted phase-contrast microscope (Nikon, one C-type CRD. The alignment showed that the CRD of
Japan), and half of the culture medium was replaced with fresh culture medium LvCTL1 contains all 14 invariant amino acid residues and 16 of
every 3 days. The number of cells was also counted before inoculating them into 18 highly conserved amino acid residues that define a C-type
96-well microplates at about 3 ⫻ 106 cells per well for WSSV challenge exper-
CRD (6), including the conserved cysteine residues that stabi-
iments. The WSSV inoculum was prepared according to the method described in
our previous work (51). WSSV dilutions (50 ␮l for each well) were preincubated lize the C-type lectin domain and the EPN motif for the ligand
with the same volume of serial dilutions of LvCTL1 (the ultimate LvCTL1 binding specificity of mannose (Fig. 1B).
concentrations were 10 ␮g/ml, 20 ␮g/ml, 40 ␮g/ml, and 60 ␮g/ml) or BSA (as a LvCTL1 mRNA is specifically expressed in the hepatopan-
control) in serum-free culture medium for 20 min at 4°C and then added to the creas. Reverse transcription-PCR (RT-PCR) was performed
confluent hemocytes, which had been cultured in a 96-well microplate for 2
weeks. This procedure gave a final WSSV dilution of 10⫺3 virion after the
to determine tissue-specific expression of LvCTL1 mRNA.
addition of the hemocyte suspension. After 2 h of incubation, the viral solutions The LvCTL1 transcript was highly expressed in the hepatopan-
were removed, and completely fresh medium was added. The cells were incu- creas, and a much lower expression of LvCTL1 was detected in
bated at 25°C and monitored for 5 days. CPE and the number of death cells the stomach. However, the LvCTL1 transcript was not de-
induced by WSSV were recorded. Assays were performed in triplicate.
tected in the hemocytes, eyestalks, muscles, brains, gills, intes-
In vivo anti-WSSV activity of recombinant LvCTL1. The anti-WSSV activity of
recombinant LvCTL1 was also tested in healthy L. vannamei shrimps challenged tines (midgut), pyloric ceca, nerve cords, spermaries, or hearts
with WSSV. Healthy Pacific white shrimps with an average weight of 15 g were of healthy L. vannamei shrimps (Fig. 2A and B). These results
kept in separate 500-liter tanks (each tank contained 30 shrimps) of well-aerated suggest that the LvCTL1 transcript is specifically expressed in
seawater (at 26 ⫾ 1°C) and acclimatized to laboratory conditions for 1 week the hepatopancreases of shrimps.
before experiments. Shrimps were fed a commercial diet three times daily, and
approximately 50% of the seawater was changed per day. Twelve tanks of the
Expression and purification of recombinant LvCTL1. Com-
healthy shrimps were divided into four groups (three tanks for each group). petent E. coli M15(pREP4) cells were transformed with the
Shrimps were injected intramuscularly with sterile 1⫻ PBS solution (as a nega- recombinant expression vector pQE30-LvCTL1, and the re-
tive control; group 1), WSSV inoculum (as a positive control; group 2), or WSSV combinant protein, which has a six-His-tag addition to the N
inoculum preincubated with LvCTL1 (60 ␮g/ml; group 3) or pET-32a-His-tag
terminus, was expressed abundantly after IPTG (isopropyl-␤-
(group 4) for 20 min at 4°C (50 ␮l per shrimp). During shrimp culture, shrimp
D-thiogalactopyranoside) induction. The recombinant LvCTL1
mortality was monitored daily for 9 days and calculated, and the dead shrimps
were examined by PCR to confirm infection with WSSV. protein was expressed as inclusion bodies and purified with
Statistical analysis. Student’s t test was used for statistical comparison of the Ni-nitrilotriacetic acid agarose under denaturing conditions.
mean cumulative mortality rates across groups in the antiviral activity experiment After being refolded by gradient dialysis against PBS buffer,
in vivo. P values of ⬍0.05 were considered statistically significant.
almost all of the purified LvCTL1 protein became soluble, and
the purity was estimated to be higher than 95% (Fig. 2C, lane
3). Recombinant LvCTL1 has an apparent mass of ⬃16 kDa as
RESULTS
determined by SDS-PAGE, which matched the predicted mo-
Cloning of the LvCTL1 cDNA from L. vannamei. Based on lecular mass calculated from the deduced amino acid se-
the initial partial cDNA sequence of LvCTL1, gene-specific prim- quence. Recombinant LvCTL1 was specifically recognized by
ers (GSP1 and GSP2) (Table 1) were then designed and used for both monoclonal antibody against the His tag and polyclonal
RACE reactions to obtain both the 5⬘ and 3⬘ ends of the cDNA, antibody against LvCTL1 (Fig. 2C, lanes 4 and 5).
and the full-length cDNA was assembled from multiple overlap- Expression of LvCTL1 protein in shrimp hemolymph is
ping clones. Then, the sequence was verified by reamplifying the induced after WSSV infection. The hemolymph was collected
complete sequence, using gene-specific primers GSP3 and GSP4. from healthy shrimps or shrimps infected with WSSV at dif-
The full-length LvCTL1 cDNA was 638 bp, with an open reading ferent times (3, 6, 12, 24, 36, 48, and 72 h) postinfection, and
frame of 471 bp encoding a protein of 156 amino acids (GenBank LvCTL1 was detected by immunoblotting, using polyclonal
accession no. DQ858900) (Fig. 1A). antibody against recombinant LvCTL1 (Fig. 2D). LvCTL1 pro-
The deduced amino acid sequence of LvCTL1 contains a tein was not detected in the hemolymph of healthy (unchal-
putative signal peptide of 20 residues and a single C-type CRD. lenged) shrimps. After WSSV infection, a 24-kDa protein was
The CRD of LvCTL1 contains an EPN motif (Glu99-Pro100- detected in the hemolymph at 12 h postinfection, which was
Asn101), which has a predicted ligand binding specificity for assumed to be the natural glycoprotein of LvCTL1, just like
mannose in mammalian C-type CRDs (8). This result suggests other shrimp lectin glycoproteins from Fenneropenaeus mer-
that LvCTL1 may bind to mannose. The calculated molecular guiensis, Penaeus monodon, and Macrobrachium rosenbergii
mass of the mature LvCTL1 protein (residues 21 to 156) is (32, 33, 50). The concentration of LvCTL1 in hemolymph
15.86 kDa, with an estimated pI of 5.20. increased steadily up to 48 h postinfection (Fig. 2D, lanes 4, 5,
352 ZHAO ET AL. J. VIROL.

FIG. 2. Expression of LvCTL1 in shrimp tissues and production of recombinant LvCTL1. (A) Expression of LvCTL1 in different tissues as
shown by RT-PCR. Total RNA from different tissues of L. vannamei shrimps was reverse transcribed to cDNA, which was used as a template for
RT-PCR. The tissues include hepatopancreas (Hp), hemocyte (Hm), muscle (Ms), eyestalk (Es), brain (Br), pyloric cecum (Pc), nerve cord (Nc),
gill (Gi), intestine (midgut) (Mg), spermary (Sp), heart (Ht), and stomach (St). Expression of ␤-actin was used as a control. (B) Statistical analysis
of the RT-PCR results. The relative expression level of LvCTL1 is shown as the ratio of lectin to ␤-actin. (C) Production and purification of
recombinant LvCTL1. Bacterial lysates without (lane 1) or with (lane 2) IPTG induction, as well as purified recombinant LvCTL1 (lane 3), were
subjected to SDS-PAGE (15% gel). Proteins were stained with Coomassie brilliant blue R-250. Purified recombinant LvCTL1 was also analyzed
by immunoblotting with mouse monoclonal antibody against the His tag (lane 4) and polyclonal antibody specific for LvCTL1 (lane 5). Lane M,
protein molecular standard. (D) Western blot analysis of LvCTL1 protein in the hemolymph of L. vannamei. Hemolymph from healthy
(unchallenged) shrimps or shrimps injected with WSSV at different time points (0, 3, 6, 12, 24, 36, 48, and 72 h) postinjection was subjected to 15%
SDS-PAGE, and proteins were transferred to a nitrocellulose membrane. LvCTL1 was identified by Western blot analysis, with mouse polyclonal
antiserum against recombinant LvCTL1. Lane 1, unchallenged hemolymph; lanes 2 to 8, hemolymph from shrimps challenged with WSSV at 3 h,
6 h, 12 h, 24 h, 36 h, 48 h, and 72 h postinjection. (E) Comparison of LvCTL1 expression levels in WSSV-resistant and -susceptible shrimp
hemolymph by an ELISA-based assay. Total proteins extracted from hemolymph samples from both the WSSV-resistant and -susceptible L.
vannamei shrimps at 0, 3, 6, 12, 24, 36, 48, and 72 h after WSSV injection were assayed by the ELISA procedure. The 96-well microtiter plates
were coated overnight at 4°C with 20.0 ␮g total protein per well in about 100 ␮l of protein extracts. The bound peroxidase activity was determined
by a reaction with TMB (3,3⬘,5,5⬘-tetramethylbenzidine), and the OD value was measured by spectrophotometry, using 450-nm filters. Negative-
control plates included plates coated with BSA. For each sample, the OD of the control well was subtracted from the OD of the hemolymph sample
well. All ELISAs were performed in triplicate, with the data given in mean values.

6, and 7). However, LvCTL1 in hemolymph decreased dramat- after WSSV injection were gradually increased and obviously
ically at 72 h postinjection (Fig. 2D, lane 8). These results higher than those of the hemolymph samples from the healthy
suggest that LvCTL1 protein in hemolymph was induced after control shrimps. The mean OD values from the hemolymph
WSSV infection. samples of the WSSV-resistant shrimps after WSSV injection
Expression of LvCTL1 in the hemolymph of WSSV-resistant were significantly higher than those of the hemolymph samples
and -susceptible shrimps. To compare the expression levels of from the WSSV-susceptible shrimps (Fig. 2E). These results
LvCTL1 in the hemolymph of WSSV-resistant and -susceptible indicated that the expression levels of LvCTL1 in the hemo-
shrimps, an ELISA-based assay was performed. Cell-free lymph of the WSSV-resistant shrimps were significantly higher
plasma samples were prepared from both the WSSV-resistant than those in the hemolymph of the WSSV-susceptible shrimps
and -susceptible L. vannamei shrimps at 0, 3, 6, 12, 24, 36, 48, after WSSV injection, suggesting that induced expression of
and 72 h after WSSV injection and used for the ELISA. ELISA LvCTL1 is a response against the WSSV infection.
results showed that LvCTL1 in the hemolymph of both the Hemagglutinating activity and carbohydrate binding speci-
WSSV-resistant and -susceptible shrimps was recognized by ficity of recombinant LvCTL1. Refolded recombinant LvCTL1
the mouse antiserum against recombinant LvCTL1. The mean protein was used to perform a hemagglutination assay with
OD values from the hemolymph samples at 12, 24, 36, and 48 h erythrocytes from different animals. Among the animal eryth-
VOL. 83, 2009 C-TYPE LECTIN FROM L. VANNAMEI HAS ANTI-WSSV ACTIVITY 353

TABLE 2. Sugar specificities of LvCTL1 Recombinant LvCTL1 has anti-WSSV activity both in vitro
Sugar name MIC (mM) and in vivo. To test whether recombinant LvCTL1 has direct
anti-WSSV activity, an in vitro assay was first performed with
D-Mannose ..................................................................................... 5
D-Glucose ....................................................................................... 15 a primary culture of L. vannamei hemocytes. Hemocytes from
D-Fructose ...................................................................................... 35 healthy shrimps were collected and cultured in 96-well micro-
D-Galactose .................................................................................... 95 plates. Next, the primary hemocytes were infected with WSSV
Lactose............................................................................................⬎100 that had been preincubated with recombinant LvCTL1 or BSA
Sucrose............................................................................................⬎100
(as a control), and the cells were monitored for 5 days. These
hemocytes were viable after 2 days, but after 3 days, clumps of
infected cells and lysed cells were found in wells that were
inoculated with BSA-treated WSSV (data not shown). There
rocytes tested, LvCTL1 agglutinated mouse and chicken eryth- were no significant changes, however, in the hemocytes inoc-
rocytes more effectively than rabbit erythrocytes with a mini- ulated with recombinant LvCTL1-treated WSSV (data not
mal agglutination concentration of 6.25 ␮g/ml, and it shown). At 5 days postinfection, CPE were apparent in the
agglutinated fish and rabbit erythrocytes at a concentration hemocytes inoculated with BSA-treated WSSV; most cells be-
higher than 12.5 ␮g/ml. Recombinant LvCTL1 also aggluti- came detached from the wells, and cell debris was observed
nated shrimp hemocytes at a concentration higher than 80.0 (Fig.4aB). On the other hand, only small clumps of infected
␮g/ml. However, no agglutination could be observed in the cells and very little cell debris were found in the hemocytes
BSA control under the same conditions. When mouse hemo- inoculated with LvCTL1 (40 ␮g/ml)-treated WSSV (Fig.4aC).
cytes were agglutinated in the presence of EDTA, agglutina- When the concentration of recombinant LvCTL1 was 20 ␮g/ml
tion was inhibited when the EDTA concentration was higher or 60 ␮g/ml, the appearance of infected cell clumps was also
than 2.0 mM (agglutination occurred at EDTA concentrations delayed, and the amounts of cell clumps and CPE induced by
of 0, 0.5, 1.0, and 1.5 mM), indicating that the agglutination of WSSV were decreased (data not shown). The hemocytes with-
animal erythrocytes by recombinant LvCTL1 is calcium depen-
dent.
To test the carbohydrate binding specificity of LvCTL1, an
inhibitory hemagglutination assay was performed. Carbohy-
drates were preincubated with recombinant LvCTL1, and the
mixtures were then added to mouse erythrocytes. Among
the carbohydrates tested, D-mannose and D-glucose inhibited
the hemagglutinating activity of recombinant LvCTL1 most
effectively, with minimal concentrations of 5 and 15 mM, re-
spectively (Table 2). D-Fructose inhibited the hemagglutinating
activity of LvCTL1 at a higher concentration (35 mM), while
D-galactose, lactose, and sucrose had little inhibitory activity
(Table 2). These results suggest that LvCTL1 has ligand bind-
ing specificity for mannose/glucose, which is consistent with the
predicted ligand specificity of mannose.
Recombinant LvCTL1 binds to WSSVs. To test whether FIG. 3. Interaction of recombinant LvCTL1 with WSSV. (a) Bind-
ing of recombinant LvCTL1 to the WSSV virions. Purified WSSV was
LvCTL1 can directly bind to WSSVs, immunostaining was dotted onto a nitrocellulose membrane and probed with recombinant
performed. WSSV virions were spotted onto a nitrocellulose LvCTL1, Lypn (a fish protein that was also expressed with the pET-32a
membrane and then incubated with recombinant LvCTL1, vector), or pET-32a-His-tag. Binding of recombinant proteins to the
Lypn (a His-tagged control protein from fish [unpublished]), or viruses was detected by mouse monoclonal antibody to the His tag or
mouse polyclonal antibody specific for LvCTL1. (A) The membrane
pET-32a-His-tag. Binding of recombinant proteins to WSSVs
was probed with recombinant LvCTL1 and detected with a mouse-
was detected by monoclonal antibody against the His tag or specific antibody for LvCTL1. (B) The membrane was probed with
polyclonal antibody specific for LvCTL1. As shown in Fig. 3a, recombinant LvCTL1 and detected with anti-His tag antibody. (C) The
binding of recombinant LvCTL1 to WSSV was detected by membrane was probed with pET-32a-His-tag and detected with anti-
both anti-His tag and LvCTL1-specific antibodies (Fig. 3aA His tag antibody. (D) The membrane was probed with Lypn and
detected with anti-His tag antibody. (b) Protein pull-down assay for
and B). No Lypn or pET-32a-His-tag bound to WSSVs (Fig. WSSV proteins that interact with the lectin LvCTL1. The cDNA frag-
3aC and D). These results suggest that LvCTL1 may bind to ment encoding mature LvCTL1 (Fig. 1A, residues 21 to 156) was
proteins on the surfaces of WSSVs. cloned into the PMAL-C2X plasmid (NEB, United Kingdom) to ex-
Recombinant LvCTL1 interacts with WSSV envelope pro- press an MBP-lectin fusion protein in E. coli BL21 cells. The MBP or
MBP-lectin fusion protein was fixed to amylose resins. The purified
teins. To further confirm that recombinant LvCTL1 can inter-
WSSV particles were lysed and centrifuged at 14,000 rpm for 15 min at
act with proteins from WSSVs, a pull-down assay was per- 4°C, and the supernatant was diluted five times with TBS containing
formed. WSSV proteins pulled down by MBP-lectin but not by 1% Triton X-100, 10 mM EDTA, and protease inhibitor. Then, the
MBP were analyzed by mass spectrometry and identified as viral protein extract was added to the MBP or MBP-lectin fusion
VP95, VP28, VP26, VP24, VP19, and VP14 (Fig. 3b), with protein-fixed amylose resins, and the mixture was incubated for 3 h at
4°C. The amylose resins were then washed with TBS buffer containing
sequence coverage of 21%, 28%, 62%, 53%, 41%, and 46%, 1% Triton X-100, and the pull-down proteins were analyzed by SDS-
respectively. These results suggest that LvCTL1 may bind to PAGE. Lane 1, proteins pulled down by MBP; lane 2, proteins pulled
these WSSV envelope proteins. down by MBP-lectin; lane M, protein molecular standard.
354 ZHAO ET AL. J. VIROL.

FIG. 4. Anti-WSSV activity of recombinant LvCTL1. (a) In vitro anti-WSSV activity of recombinant LvCTL1 in the primary hemocyte culture
of L. vannamei. Primary hemocytes were inoculated with recombinant LvCTL1-treated WSSV (C) or BSA-treated WSSV (B) or not inoculated
(A). Cells were then incubated for 5 days. Scale bar ⫽ 100 ␮m. (b) In vivo anti-WSSV activity of recombinant LvCTL1 in shrimps. Healthy shrimps
were injected with PBS (negative control), untreated WSSV (positive control), recombinant pET-32a-His-tag-treated WSSV, or LvCTL1-treated
WSSV. The cumulated mortality of shrimps was recorded up to 9 days postinjection. Each column represents the means from triplicates, with
standard deviations. The significance of differences was calculated by the t test (P ⬍ 0.05).

out WSSV inoculation grew well throughout the experimental DISCUSSION


period (Fig.4aA). However, the anti-WSSV activity of LvCTL1
was not significant when the concentration of LvCTL1 was 10 C-type lectins exist in almost all animals. They are capable of
␮g/ml (data not shown). These results suggest that LvCTL1 can binding to specific carbohydrates in a Ca2⫹-dependent manner
protect shrimp hemocytes from WSSV infection. and play an important role in nonself-recognition and the
To determine whether LvCTL1 also has anti-WSSV activity clearance of invading microorganisms (6, 8, 42, 47). In this
in shrimps in vivo, healthy L. vannamei shrimps were infected study, we isolated a cDNA for a novel C-type lectin (named
with untreated WSSV (positive control), recombinant LvCTL1) from the Pacific white shrimp, L. vannamei, by using
LvCTL1- or pET-32a-His-tag-treated WSSV, or PBS (negative ESTs and RACE. LvCTL1 contains a single CRD that is sim-
control), and the mortality of shrimps was monitored daily for ilar to PmAV (black tiger shrimp P. monodon) (29) and Fc-hsL
9 days. Initial mortality was observed after the first or second
day postinjection in all three groups of shrimps infected with
WSSV as well as in the negative-control group (first day). On TABLE 3. Cumulative mortalities of the shrimp L. vannamei after
the injection of PBS, WSSV, or WSSV preincubated with
the fifth day postinfection, the mortality of the shrimps in- recombinant LvCTL1 or pET-32a-His-tag protein
fected with recombinant LvCTL1-treated WSSV (15.6%) was
significantly lower (P ⬍ 0.05) than that of the shrimps infected Initial no. Mortality postinfection (%)a
of
with pET-32a-His-tag-treated WSSV (43.3%) or untreated Treatment
shrimps
WSSV (58.9%), and no mortality was observed in the negative- (n ⫽ 360) Day 3 Day 5 Day 9
control shrimps (Fig. 4b and Table 3). On the ninth day postin- PBS 90 0 0 0
fection, the mortality of shrimps infected with LvCTL1- and WSSV 90 13.3 ⫾ 1.00 58.9 ⫾ 1.53A 100 ⫾ 0.00A
pET-32a-His-tag-treated WSSV reached 47.8% and 100%, re- WSSV ⫹ 90 11.1 ⫾ 0.57 43.3 ⫾ 1.00A 100 ⫾ 0.00A
spectively, and no mortality was observed in the negative-con- PET-32a
WSSV ⫹ 90 6.7 ⫾ 1.00 15.6 ⫾ 0.58B 47.8 ⫾ 1.15B
trol shrimps, whereas the cumulated mortality of the shrimps
LvCTL1
infected with untreated WSSV had already reached 100% on
a
the eighth day postinfection (Table 3 and Fig. 4b). These Results are means from three independent experiments with standard devi-
ations. P values were determined using analysis of variance, and the significance
results suggest that recombinant LvCTL1 can protect shrimps of differences was determined by the t test (P ⬍ 0.05). Values in the same column
from WSSV infection. with different letters (A and B) are significantly different.
VOL. 83, 2009 C-TYPE LECTIN FROM L. VANNAMEI HAS ANTI-WSSV ACTIVITY 355

(FcCLec-2) (Chinese shrimp F. chinensis) (36), but it is differ- C-type lectins, such as MBL, have been shown to be able to
ent from LvCTL, which has two putative CRDs (31). In the bind envelope glycoproteins of human immunodeficiency virus
CRD of LvCTL1, there is an EPN motif (Glu99-Pro100-Asn101) (4) and envelope proteins of the influenza A virus (19), and
that has a predicted ligand binding specificity for mannose (or dendritic cell-specific intercellular adhesion molecule 3-grab-
mannose-related sugars) (Fig. 1A), and recombinant LvCTL1 bing nonintegrins can interact with envelope glycoproteins of
did bind to mannose and glucose with high affinity (Table 2), Ebola virus (1) and bind to Dengue (37) and hepatitis C (27)
indicating that LvCTL1 is mannose specific. LvCTL contains viruses. Thus, it is likely that LvCTL1 may bind to the surface
two CRDs, one with an EPN motif for mannose and the other glycoproteins of WSSV to inhibit the infection.
with a QPD motif for galactose (31). The two L. vannamei So far, approximately 58 structural proteins, including 9 nu-
lectins share about 34% identity in the CRDs. cleocapsid proteins and 32 envelope proteins, have been iden-
LvCTL1 mRNA was specifically expressed in the hepato- tified in the WSSV virions (23), and they may play key roles in
pancreas (Fig. 2A and B), similarly to LvCTL and PmAV (29, the initial infection of WSSV in shrimps (40). These data
31). LvCTL1 protein was not detected in the hemolymph of facilitate us further in studying antiviral activity. It was shown
healthy shrimps, but it was present and its concentration was that even a single WSSV envelope protein, such as VP28, can
increased in the hemolymph of shrimps injected with WSSV prolong the survival of shrimps following WSSV challenge
(Fig. 2D and E), suggesting that expression of LvCTL1 was when it is injected intramuscularly or administered orally, pos-
induced by WSSV infection. Expression of other shrimp lec- sibly by competitive saturation of receptor sites of the host cells
tins, such as PmAV, LvCTL, and FcCLec-1, was also induced relevant for viral adhesion and viral penetration (43). Like-
by WSSV infection (29, 25, 31); however, expression of these wise, antibodies raised against VP28 can also neutralize and
C-type lectins in virus-infected shrimps showed different pat- inhibit WSSV infection (40).
terns. The transcription level of FcCLec-1 reaches a maximum We showed that recombinant LvCTL1 protein, when bound
expression at 3 h postinfection and then decreases from 6 h to to WSSV, significantly increased the survival of shrimps against
48 h postinfection (25). The mRNA level of PmAV is slightly WSSV challenge and also protected shrimp hemocytes from
decreased from 6 h to day 1 postinfection and then starts to WSSV infection (Table 3; Fig. 4). LvCTL1 may serve as a PRR
increase from day 2 and reaches a peak at day 4 postinfection to recognize and bind envelope proteins of WSSV, to block the
(30). Similarly, the transcription level of LvCTL decreases virus from entering cells, or to neutralize the virus. Our future
initially in the first 2 h and then increases to a much higher goal is to investigate how the binding of LvCTL1 to WSSVs
level at 4 h postinfection (31). The LvCTL1 protein appeared inhibits the infection.
in the hemolymph at 12 h postinfection, increased steadily
thereafter, and then remained at a high level until 48 h postin- ACKNOWLEDGMENTS
fection (Fig. 2D and E). A delay in the progression of mortality This work was supported by the National High Technology Re-
in the shrimps infected with recombinant LvCTL1-treated search and Development Program of China (863 Program) under
WSSV was also observed from day 3 postinfection until the grants 2001AA601030 and 2006AA09Z445, the Guangdong Province
end of the challenge (Table 3; Fig. 4b). These results suggest Natural Science Foundation under grant 20023002, and the Science
and Technology Bureau of Guangdong Province.
that LvCTL1 in high concentrations in shrimp hemolymph may
We thank Chuan-Fu Dong for his advice on cell culture and Hua-
bind to WSSV and neutralize the viruses, protecting the Shui Ai for technical assistance.
shrimps from WSSV infection. However, the concentration of
LvCTL1 in the hemolymph decreased dramatically at 72 h REFERENCES
postinfection. It is possible that LvCTL1 recognizes and binds 1. Alvarez, C. P., F. Lasala, J. Carrillo, O. Muñiz, A. L. Corbí, and R. Delgado.
2002. C-type lectins DC-SIGN and L-SIGN mediate cellular entry by Ebola
to the viruses; thus, the free protein in hemolymph decreases. virus in cis and in trans. J. Virol. 76:6841–6844.
Another possibility is that the decrease of LvCTL1 in the 2. Apostolopoulos, V., and I. F. McKenzie. 2001. Role of the mannose receptor
hemolymph might be due to the decrease in shrimp activity at in the immune response. Curr. Mol. Med. 1:469–474.
3. Bayne, C. J. 1990. Phagocytosis and non-self recognition in invertebrates.
the moribund stage, resulting from an increase in the amount BioScience 40:723–731.
of viruses. 4. Curtis, B. M., S. Scharnowske, and A. J. Watson. 1992. Sequence and
Shrimp C-type lectins have been shown to have antibacterial expression of a membrane-associated C-type lectin that exhibits CD4-inde-
pendent binding of human immunodeficiency virus envelope glycoprotein
activity and promote phagocytosis. For example, PmCLec-2, gp120. Proc. Natl. Acad. Sci. USA 89:8356–8360.
FcCLec-1, and FcCLec-2 have been reported to possess anti- 5. Day, A. J. 1994. The C-type carbohydrate recognition domain (CRD) super-
family. Biochem. Soc. Trans. 22:83–88.
bacterial activity (25, 28, 36), and a C-type lectin from P. 6. Drickamer, K. 1993. Ca2⫹-dependent carbohydrate-recognition domains in
japonicus can promote phagocytosis (46). However, only one animal proteins. Curr. Opin. Struct. Biol. 3:393–400.
C-type lectin, PmAV, which contains a QPD motif in the CRD, 7. Drickamer, K. 1988. Two distinct classes of carbohydrate recognition do-
mains in animal lectins. J. Biol. Chem. 263:9557–9560.
has thus far been reported to have antiviral activity by inhib- 8. Drickamer, K. 1992. Engineering galactose-binding activity into a C-type
iting CPE induced by grouper iridovirus in fish cells in vitro mannose-binding protein. Nature 360:183–186.
(29). PmAV was thought to have nonspecific antiviral activity 9. Ezekowitz, R. A. 2003. Role of the mannose-binding lectin in innate immu-
nity. J. Infect. Dis. 187(Suppl. 2):S335–S339.
and may act as an intermediate rather than a recognition factor 10. Fraser, I. P., H. Koziel, and R. A. Ezekowitz. 1998. The serum mannose-
during the antiviral reaction, since no PmAV protein was binding protein and the macrophage mannose receptor are pattern recog-
nition molecules that link innate and adaptive immunity. Semin. Immunol.
found to bind WSSV by an immunohistological assay (29). We 10:363–372.
showed that recombinant LvCTL1 directly bound to the WSSV 11. Fujita, T., M. Matsushita, and Y. Endo. 2004. The lectin-complement path-
virions to exert its antiviral activity (Fig. 3 and 4). LvCTL1 way—its role in innate immunity and evolution. Immunol. Rev. 198:185–202.
12. Gross, P. S., T. C. Bartlett, C. L. Browdy, R. W. Chapman, and G. W. Warr.
interacted with WSSV envelope proteins, including VP95, 2001. Immune gene discovery by expressed sequence tag analysis of hemo-
VP28, VP26, VP24, VP19, and VP14 (Fig. 3b). In mammals, cytes and hepatopancreas in the Pacific White Shrimp, Litopenaeus van-
356 ZHAO ET AL. J. VIROL.

namei, and the Atlantic White Shrimp, L. setiferus. Dev. Comp. Immunol. specific lectin from black tiger prawn (Peneaus monodon). Comp. Biochem.
25:565–577. Physiol. B 97:515–520.
13. He, J. G., M. Deng, Q. X. Long, H. M. Zhou, G. S. Li, P. Yao, L. Lü, Y. G. 33. Rittidach, W. N. Paijit, and P. Utarabhand. 2007. Purification and charac-
Chen, S. P. Weng, S. Y. Miao, S. G. Jiang, Y. Y. Wu, X. Huang, T. Zuo, X. M. terization of a lectin from the banana shrimp Fenneropenaeus merguiensis
Yang, N. Yu, S. T. Xie, W. Zhong, G. F. Li, Q. Z. Ye, J. B. Jiang, S. M. Chan, hemolymph. Biochim. Biophys. Acta 1770:106–114.
and F. Mo. 2000. Theory and strategies for controlling white spot syndrome 34. Sambrook, J., E. F. Fritsch, and T. Maniatis. 1989. Molecular cloning: a
(WSS) of cultured Penaeus monodon in South China. Acta Sci. Nat. Univ. laboratory manual, 2nd ed., p. 165–209. Cold Spring Harbor Laboratory
Sunyatseni 39(Suppl.):147–153. Press, Cold Spring Harbor, NY.
14. He, N. H., Q. W. Qin, and X. Xu. 2005. Differential profile of genes expressed 35. Sharon, N., and H. Lis. 1972. Lectins: cell-agglutinating and sugar-specific
in hemocytes of White Spot Syndrome Virus-resistant shrimp (Penaeus proteins. Science 177:949–959.
japonicus) by combining suppression subtractive hybridization and differen- 36. Sun, Y. D., L. D. Fu, Y. P. Jia, X. J. Du, Q. Wang, Y. H. Wang, X. F. Zhao,
tial hybridization. Antivir. Res. 66:39–45. X. Q. Yu, and J. X. Wang. 2008. A hepatopancreas-specific C-type lectin
15. Hoffmann, J. A., F. C. Kafatos, C. A. Janeway, and R. A. Ezekowitz. 1999. from the Chinese shrimp Fenneropenaeus chinensis exhibits antimicrobial
Phylogenetic perspectives in innate immunity. Science 284:1313–1318. activity. Mol. Immunol. 45:348–361.
16. Janeway, C. A., and R. Medzhitov. 2002. Innate immune recognition. Annu. 37. Tassaneetrithep, B., T. H. Burgess, A. Granelli-Piperno, C. Trumpfheller, J.
Rev. Immunol. 20:197–216. Finke, W. Sun, M. A. Eller, K. Pattanapanyasat, S. Sarasombath, D. L. Birx,
17. Jiang, Y. S., W. B. Zhan, S. B. Wang, and J. Xing. 2006. Development of R. M. Steinman, S. Schlesinger, and M. A. Marovich. 2003. DC-SIGN
primary shrimp hemocyte cultures of Penaeus chinensis to study white spot (CD209) mediates Dengue virus infection of human dendritic cells. J. Exp.
syndrome virus (WSSV) infection. Aquaculture 253:114–119. Med. 197:823–829.
18. Jomori, T., and S. Natori. 1992. Function of the lipopolysaccharide-binding 38. Uemura, K., M. Saka, T. Nakagawa, N. Kawasaki, S. Thiel, J. C. Jensenius,
protein of Periplaneta americana as an opsonin. FEBS Lett. 296:283–286. and T. Kawasaki. 2002. L-MBP is expressed in epithelial cells of mouse small
19. Kase, T., Y. Suzuki, T. Kawai, T. Sakamoto, K. Ohtani, S. Eda, A. Maeda, intestine. J. Immunol. 169:6945–6950.
and Y. Okuno. 1999. Human mannan-binding lectin inhibits the infection of 39. van Hulten, M. C. W., J. Witteveldt, S. Peters, N. Kloosterboer, R. Tarchini,
influenza A virus without complement. Immunology 97:385–392. M. Fiers, H. Sandbrink, R. K. Lankhorst, and J. M. Vlak. 2001. The white
20. Koizumi, N., M. Imamura, T. Kadotani, K. Yaoi, H. Iwahana, and R. Sato. spot syndrome virus DNA genome sequence. Virology 286:7–22.
1999. The lipopolysaccharide-binding protein participating in haemocyte 40. van Hulten, M. C. W., J. Witteveldt, M. Snippe, and J. M. Vlak. 2001. White
nodule formation in the silkworm Bombyx mori is a novel member of the spot syndrome virus envelope protein VP28 is involved in the systemic
C-type lectin superfamily with two different tandem carbohydrate-recogni- infection of shrimp. Virology 285:228–233.
tion domains. FEBS Lett. 443:139–143.
41. Vlak, J. M., J. R. Bonami, T. W. Flegel, G. H. Kou, D. V. Lightner, C. F. Lo,
21. Kondo, M., H. Matsuyama, and T. Yano. 1992. The opsonic effect of lectin
P. C. Loh, and P. W. Walker. 2004. Nimaviridae, p. 187–192. In C. M.
on phagocytosis by hemocytes of Kuruma Prawn, Penaeus japonicus. Fish
Fauquet, M. A. Mayo, J. Maniloff, U. Desselberger, and L. A. Ball (ed.),
Pathol. 27:217–222.
VIIIth report of the International Committee on Taxonomy of Viruses.
22. Lee, S. Y., and K. Söderhäll. 2002. Early events in crustacean innate immu-
Elsevier, Amsterdam, The Netherlands.
nity. Fish Shellfish Immunol. 12(4):21–37.
42. Weis, W. I., M. E. Taylor, and K. Drickamer. 1998. The C-type lectin
23. Li, Z., Q. Lin, J. Chen, J. L. Wu, T. K. Lim, S. S. Loh, X. Tang, and C. L.
superfamily in the immune system. Immunol. Rev. 163:19–34.
Hew. 2007. Shotgun identification of the structural proteome of shrimp white
spot syndrome virus and iTRAQ differentiation of envelope and nucleocap- 43. Witteveldt, J., C. C. Cifuentes, J. M. Vlak, and M. C. W. van Hulten. 2004.
sid subproteomes. Mol. Cell. Proteomics 6:1609–1620. Protection of Penaeus monodon against white spot syndrome virus by oral
24. Lightner, D. V., and R. M. Redman. 1998. Shrimp diseases and current vaccination. J. Virol. 78:2057–2061.
diagnostic methods. Aquaculture 164:201–220. 44. Xie, X. X., H. Y. Li, L. M. Xu, and F. Yang. 2005. A simple and efficient
25. Liu, Y. C., F. H. Li, B. Dong, B. Wang, L. Wei, X. J. Zhang, L. S. Zhang, and method for purification of intact white spot syndrome virus (WSSV) viral
J. H. Xiang. 2007. Molecular cloning, characterization and expression anal- particles. Virus Res. 108:63–67.
ysis of a putative C-type lectin (Fclectin) gene in Chinese shrimp Fennero- 45. Yang, F., J. He, X. Lin, Q. Li, D. Pan, X. Zhang, and X. Xu. 2001. Complete
penaeus chinensis. Mol. Immunol. 44:598–607. genome sequence of the shrimp white spot bacilliform virus. J. Virol. 75:
26. Lo, C. F., C. H. Ho, S. E. Peng, C. H. Chen, H. C. Hsu, Y. L. Chiu, C. F. 11811–11820.
Chang, K. F. Liu, M. S. Su, C. H. Wang, and G. H. Kou. 1996. White spot 46. Yang, H. J., T. Luo, F. Li, S. J. Li, and X. Xu. 2007. Purification and
syndrome baculovirus (WSBV) detected in cultured and captured shrimp, characterization of a calcium-independent lectin (PjLec) from the haemo-
crabs and other arthropods. Dis. Aquat. Organ. 27:215–225. lymph of the shrimp Penaeus japonicus. Fish Shellfish Immunol. 22:88–97.
27. Lozach, P. Y., H. Lortat-Jacob, A. De Lacroix De Lavalette, I. Staropoli, S. 47. Yu, X. Q., and M. R. Kanost. 2004. Immulectin-2, a pattern recognition
Foung, A. Amara, C. Houlès, F. Fieschi, O. Schwartz, J. L. Virelizier, F. receptor that stimulates hemocyte encapsulation and melanization in the
Arenzana-Seisdedos, and R. Altmeyer. 2003. DC-SIGN and L-SIGN are tobacco hornworm, Manduca sexta. Dev. Comp. Immunol. 28:891–900.
high-affinity binding receptors for hepatitis C virus glycoprotein E2. J. Biol. 48. Yu, X. Q., H. Gan, and M. R. Kanost. 1999. Immulectin, an inducible C-type
Chem. 278:20358–20366. lectin from an insect, Manduca sexta, stimulates activation of plasma pro-
28. Luo, T., H. Yang, F. Li, X. Zhang, and X. Xu. 2006. Purification, character- phenol oxidase. Insect Biochem. Mol. Biol. 29:585–597.
ization and cDNA cloning of a novel lipopolysaccharide-binding lectin from 49. Yu, X. Q., and M. R. Kanost. 2000. Immulectin-2, a lipopolysaccharide-
the shrimp Penaeus monodon. Dev. Comp. Immunol. 30:607–617. specific lectin from an insect, Manduca sexta, is induced in response to
29. Luo, T., X. Zhang, Z. Shao, and X. Xu. 2003. PmAV, a novel gene involved gram-negative bacteria. J. Biol. Chem. 275:37373–37381.
in virus resistance of shrimp Penaeus monodon. FEBS Lett. 551:53–57. 50. Zenteno, R., L. Vazquez, C. Sierra, A. Pereyra, and M. C. Slomianny, S.
30. Luo, T., F. Li, K. Lei, and X. Xu. 2007. Genomic organization, promoter Bouquelet, and E. Zenteno. 2000. Chemical characterization of the lectin
characterization and expression profiles of an antiviral gene PmAV from the from the freshwater prawn Macrobrachium rosenbergii (De Man) by MALDI-
shrimp Penaeus monodon. Mol. Immunol. 44:1516–1523. TOF. Comp. Biochem. Physiol. B 127:243–250.
31. Ma, T. H. T., S. H. K. Tiu, J. G. He, and S. M. Chan. 2007. Molecular cloning 51. Zhao, Z. Y., Z. X. Yin, S. P. Weng, H. J. Guan, S. D. Li, K. Xing, S. M. Chan,
of a C-type lectin (LvCTL) from the shrimp Litopenaeus vannamei: early and J. G. He. 2007. Profiling of differentially expressed genes in hepatopan-
gene down-regulation after WSSV infection. Fish Shellfish Immunol. 23: creas of white spot syndrome virus-resistant shrimp (Litopenaeus vannamei)
430–437. by suppression subtractive hybridization. Fish Shellfish Immunol. 22:520–
32. Ratanapo, S., and M. Chulavatnatol. 1990. Monodin, a new sialic acid- 534.

Potrebbero piacerti anche