• 沒有找到結果。

Grouper (Epinephelus coioides) CXCR4 Is Expressed in Response to Pathogens Infection and Early Stage of Development

N/A
N/A
Protected

Academic year: 2021

Share "Grouper (Epinephelus coioides) CXCR4 Is Expressed in Response to Pathogens Infection and Early Stage of Development"

Copied!
40
0
0

加載中.... (立即查看全文)

全文

(1)

1

Grouper (Epinephelus coioides) CXCR4 is expressed in response to pathogens infection 1

and early stage of development 2

3

Ching-Yu Lina, Young-Mao Chena,b,c, Hao-Hsuan Hsua,c, Chia-Tai Shiua,c, Hsiao-Che 4

Kuoa,b,c, Tzong-Yueh Chena,b,c* 5

6

aLaboratory of Molecular Genetics, Institute of Biotechnology, College of Bioscience and 7

Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan 8

bResearch Center of Ocean Environment and Technology, National Cheng Kung University, 9

Tainan 70101, Taiwan 10

cAgriculture Biotechnology Research Center, National Cheng Kung University, Tainan 11 70101, Taiwan 12 13 *Corresponding author: 14 Dr. Tzong-Yueh Chen 15

Laboratory of Molecular Genetics, Institute of Biotechnology, College of Bioscience and 16

Biotechnology, National Cheng Kung University, Tainan 70101, Taiwan 17

Telephone: 886-6-2757575 ext. 65622 ext.610 18

Fax: 886-6-2766505 19

E-mail: ibcty@mail.ncku.edu.tw 20

Source:Developmental and Comparative Immunology, Vol. 36, No. 1, pp. 112-120 Year of Publication:2011

ISSN:0145-305X Publisher:Elsevier

DOI:10.1016/j.dci.2011.06.009 © 2011 Elsevier Ltd. All rights reserved

(2)

2 Abstract

21

Chemokine (C-X-C motif) receptor 4 (CXCR4) from orange-spotted grouper (Epinephelus 22

coioides) was identified and characterized in this study. gCXCR4 shared common features 23

in protein sequence and predicted structure of CXCR4 family. This suggested that gCXCR4 24

is a member of G protein-coupled receptors with seven transmembrane domains. The 25

expression patterns revealed that gCXCR4 may play a key role in early development of 26

grouper. Furthermore, overexpression of gCXCR4-GFP for 48 h had significant effects on 27

the GF-1 cell viability. gCXCR4 protein was mainly expressed in the marginal zone of head 28

kidney and on the surface of intestinal villi. gCXCR4 expression can be detected in all the 29

examined tissues and significantly up-regulated in eye and brain, which are the main targets 30

for nervous necrosis virus (NNV) infection and replication. gCXCR4 gene expression can 31

be induced in the spleen and eye by lipopolysaccharide and NNV, respectively. Our data 32

suggested that gCXCR4 may not only play a role in the early immune response to microbial 33

infection but also restrain to the immune system and central nervous system. 34

Keywords: Epinephelus coioides; CXCR4; G-protein-coupled receptors; chemokine 35

(3)

3 1. Introduction

37

The orange-spotted grouper, Epinephelus coioides, is a commercially important fish that is 38

widely farmed in tropical waters of many countries. Considerable economic losses have 39

been sustained in grouper aquaculture due to the infection of grouper by piscine nodavirus, 40

i.e. nervous necrosis virus (NNV). The virus causes viral nervous necrosis (VNN) on 41

grouper hatchery larvae and juvenilies, resulting in a high mortality rate (80-100%) (Kuo et 42

al., 2011; Munday et al., 2002). 43

Chemokines are a group of small molecular weight (6–14 kDa) cytokines which play 44

important roles not only in against microbial infection by guiding leukocyte migration but 45

also in embryonic growth and development (Kim et al., 1999; Olson et al., 2002). 46

Chemokines can be classified into four different kinds, CXC, CC, C and CX3C (Murphy et 47

al., 2000), according to its cysteine motif in the N-terminal region. Chemokine receptors are 48

a group of G protein-coupled receptors with seven transmembrane domains. Upon 49

stimulation by chemokines, chemokine receptors trigger a series of intracellular signal 50

transductions via interaction with the G proteins. To this day, different chemokine receptors 51

have been found on various cells such as monocytes, T lymphocytes, B lymphocytes, 52

natural killer cells, macrophages, endothelial cells and neuron cells in mammals (Horuk et 53

al., 2009). To date, CXCR4 has been identified in several species including human 54

(Federsppiel et al., 1993), mouse (Heesen et al., 1996) and dog (Tsuchida et al., 2007), but 55

(4)

4

less is known in fish (Daniels et al., 1999; Chong et al., 2001; Alabyev et al., 2000; Jia and 56

Zhang, 2009), and no functional characterization of CXCR4 in orange-spotted grouper has 57

been reported. 58

Chemokines and their receptors serve an important role in viral infections and among 59

the chemokine receptors, CXCR4 is also a co-receptor for human immunodeficiency virus 60

(HIV) entry into target cells (Feng et al., 1996; Berson et al., 1996). Nonetheless, CXCR4 is 61

not only involved in the pathogenesis of viral infections but also plays a critical role in 62

organogenesis and embryonic development-related vascularization, lymphopoiesis and 63

myelopoiesis (Tachibana et al., 1998). CXCR4 deficiency produces a lethal phenotype and 64

abnormal development of central nervous system, such as abnormal migration of granule 65

cells and an altered location of Purkinje cells in mice malformed cerebellum (Ma et al., 66

1998). In fish, CXCR4 has been found in the early stage of zebrafish embryo, it can be 67

detected in the lateral mesoderm and posterior midbrain (Chong et al., 2001), moreover the 68

migration of lateral-line-primordium is impeded in CXCR4 homologue-mutated zebrafish 69

and is completely absent in SDF-1a defective zebrafish (Valentin et al., 2007). In addition, 70

CXCR4 plays a crucial role for tissue polarity (Haas and Gilmour, 2006). CXCR4 71

homologue deficiency leads to the random migration of cells and the loss of coordinated 72

motility within the posterior lateral line primordium in zebrafish (Haas and Gilmour, 2006). 73

These observations indicate that CXCR4 is multifunctional and plays crucial roles in 74

(5)

5 embryonic growth development and hematopoiesis. 75

However, no functional characterization of grouper CXCR4 has been reported. 76

Previously, a partial portion of grouper CXCR4 cDNA was identified by subtractive cDNA 77

hybridization from healthy and NNV-infected groupers (Chen et al., 2010). In the present 78

study, orange-spotted grouper CXCR4 (gCXCR4) was cloned and the expression profile 79

that response to lipopolysaccharide (LPS) and NNV infection was investigated. In addition, 80

we showed that cell proliferation was impeded after gCXCR4 overexpression for 48 h. 81

(6)

6 2. Materials and methods

83

2.1. Fish and challenge experiments 84

Fish weighing approximately 300g (150 days post-hatching) and different ages (1-40 85

days-old) of orange-spotted grouper (E. coioides) were collected from an indoor fish farm in 86

Linyuan and maintained in 10L containers at 27 ± 1 °C. For challenge experiments, 15 87

300g-in-weight-fish were divided into five groups (n=3 per group) and challenged with 100 88

μl phosphate buffered saline (PBS) contained approximately 20 μg of purified Escherichia 89

coli 0127:B8 LPS (Sigma-Aldrich, St. Louis, MO) per fish via intraperitoneal injection. 90

Fish with 100 μl PBS injection was used as a control group. The fish were sacrificed and the 91

spleens were collected at 0, 6, 24, 48 and 72 h post-injection. In the experiments of virus 92

challenging, juvenile groupers (about 0.8 g in weight, 40–45 days post-hatching) were 93

collected from Linyuan fish farms in southern Taiwan. Twelve juvenile groupers were 94

divided into two groups, NNV infection group and control group. Each six fish were 95

immersed into 500 ml of rearing water which contained 50 ml of a viral solution (106 96

TCID50/0.1 ml) or saline for 2 h. The fish were transferred to a virus-free aquarium, which 97

had been exposure to ultraviolet (UV) light for 24 h, and cultured at 28 °C. Real time PCR 98

was then used to confirm the fish was infected by NNV after 72 h of challenging. 99

2.2. Total RNA extraction and cDNA synthesis 100

Eye or whole fish samples (n=3 per group) were used for total RNA extraction by 101

(7)

7

homogenized using a MagNALysis homogenizer (Roche, Basel, Switzerland) following the 102

manufacture’s recommendations of TRIzol™ (Invitrogen) method.. cDNA was synthesized 103

with 2 μg RNA, 0.1 μM oligo dT primer, 12.5 μM dNTP (Bioman Scientific Co. Ltd., 104

Taipei, Taiwan) and 50 units Molony Murine Leukemia Virus (MMLV) reverse transcriptase 105

(Promega, Madison, WI) at 42 oC for 1 h. 106

RNA and cDNA were quantified using an Ultrospec 3300 Pro spectrophotometer 107

(Amersham Biosciences, Piscataway, NJ, USA); nucleic acids were diluted using sheared 108

salmon sperm DNA (5 ng mL-1) as a carrier. 109

2.3 RNA gel electrophoresis 110

To confirm the integrity of RNA samples, the extracted RNA was evaluated by RNA 111

electrophoresis. In brief, 2 μl RNA sample was mixed with 18 μl 1× reaction buffer (1× 112

MOPS, 20 % formaldehyde and 50 % formamide), 2 µl of 10× formaldehyde gel loading 113

buffer (50 % glycerol, 10 mM EDTA, pH8, 0.25% bromphenol blue and 0.25 % xylene 114

cyanol) and was visualized by using ethidium bromide staining 115

2.4. Rapid amplification of cDNA ends (RACE) 116

A 950 bp cDNA fragment obtained from our previous study by subtractive cDNA 117

hybridization from healthy and NNV infected groupers (Chen et al., 2010). The sequence 118

showed 75% similarity to CXCR4 from Psetta maxima by blasting 119

(http://www.ebi.ac.uk/blastall/).Full length cDNA was obtained by 5’/3’ RACE which was 120

(8)

8

performed by using 5’/3’ RACE Kit. Gene-specific primers (Table 1) for 5’ and 3’ RACE 121

were designed based on partial sequence of gCXCR4 For 5’ RACE, mRNA was transcribed 122

by MMLV reverse transcriptase (Sigma-Aldrich) with primer gCXCR4-5SP1and 123

gCXCR4-5SP2 and gCXCR4-5SP3 were used for PCR and nested PCR. 3’ RACE was 124

performed by using primers gCXCR4-3SP1 and gCXCR4-3SP2 for PCR and nested PCR, 125

respectively. PCR condition was one cycle of 3 min at 95 oC, followed by 35 cycles each at 126

95 oC for 30 s, 58 oC for 30 s, 72 oC for 30s and a final extension at 72 oC for 10 min. The 127

primers gCXCR-F and gCXCR-R were used to amplify the gCXCR4 cDNA fragment. 128

2.5 Bioinformatic analyses of gCXCR4 129

The transmembrane domains, extracellular domains and cytoplasmic domains of CXCR4 130

were identified by the TMHMM Server 2.0 program 131

(http://www.cbs.dtu.dk/services/TMHMM/). The protein sequences of different CXCR4 132

species were obtained from GenBank and were aligned using Vector NTI 10 software. A 133

phylogenetic tree of CXCR4 was constructed by the neighbor-joining method using 134

MEGA4.0. The reliability of the tree was established by bootstrap analysis, based on 1,000 135

bootstrap replications. 136

2.6. RT-PCR 137

The tissue distribution of gCXCR4 gene expressions was investigated by RT-PCR. Total 138

RNA was extracted from different tissues of grouper such as eye, fin, gill, muscle, head 139

(9)

9

kidney, heart, spleen, intestine and brain. PCR condition was one cycle of 3 min at 95 oC, 140

followed by 35 cycles each at 95 oC for 30 s, 58 oC for 30 s, 72 oC for 30 s and a final 141

extension at 72 oC for 10 min. To detect the expression of gCXCR4 in different 142

developmental stages of grouper larvae, total RNA were prepared from pooled larvae that 143

contained 20 fish fry in each group at 1, 2, 4, 6, 8, 10, 14, 18 and 20-days post hatch (dph), 144

and 3 fish larvae were pooled at 24, 26, 28, 30, 32, 34, 38 and 40 dph. 145

2.6. Real Time-quantitative PCR 146

Real-time quantitative PCR was performed by StepOne™ real-time PCR system (Applied 147

Biosystems, Foster City, CA, USA). 1ul of cDNA (from 100 ng RNA) was mixed with 12.5 148

μl 2×SYBR® Green Master Mix (Applied Biosystems) and 1 μl of each 10 μM specific 149

primer (Table 1). The thermal profile for real-time PCR was 95 °C for 10 min followed by 150

40 cycles of 95 °C for 30 s, 60 °C for 30 s and a final stage at 95 °C for 15 s, 60 °C for 1 151

min, 95°C for 15 s. The results of real-time PCR were analyzed with StepOne Software 152

v2.1. 153

2.7 Statistical analysis 154

The CT for gCXCR4 and β-actin were determined for each sample. β-actin was used as 155

internal control. △CT (Differences between gCXCR4 and β-actin) was calculated to 156

normalize the differences in the efficiency of reverse transcription reactions. The △CT for 157

each sample was subtracted from the △CT of the calibrator, and the difference was 158

(10)

10

designated as the △△CT value. The relative expression level of gCXCR4 could be 159

calculated by 2-△△CT. All real-time PCR data were subjected to analysis of t-test and are 160

presented as the mean ± S.E. of the relative mRNA expression. P-values of < 0.05 were 161

considered significantly different. 162

2.8 Plasmid construction 163

To prepare anti-gCXCR4 antiserum, extracellular domain I and III of gCXCR4 were 164

constructed into pET29b expression vector (Novagen, USA) by PCR using primers. Primers 165

gCSCR4-EXI-F (BamHI) and gCSCR4-EXI-R (SalI) were used for extracellular domain I 166

and gCXCR4-EXIII-F (SalI) and gCXCR4-EXIII-R (XhoI) were used for amplifying 167

extracellular domain III (181-215 a.a.). This recombinant plasmid was named 168

pET29b-gCXCR4-EXI-EXIII which can express a fusion protein of gCXCR4 extracellular 169

domains I and III along with a 6×His tag. 170

The gCXCR4 overexpression vector, pcDNA3.1-gCXCR4-GFP, was constructed by 171

PCR amplifying gCXCR4 using primers gCXCR-F and gCXCR4-GFP-R (Table 1) and the 172

PCR products were then cloned into the pcDNA3.1-CT-GFP-TOPO expression vector 173

(Invitrogen). The inserted DNA fragments of each clone were confirmed by sequencing 174

(Mission Biotech Co., Ltd., Taipei, Taiwan). 175

2.9 Recombinant protein and anti-gCXCR4 antiserum preparation 176

The gCXCR4-EXI-EXIII-His recombinant protein was expressed by transforming 177

(11)

11

pET-29b-gCXCR4-EXI-EXIII intoBL21(DE3) cells (Novagen)and induced by adding 178

isopropyl-beta-D-thiogalactopyranoside (IPTG; MDBIO, Frederick, MD) to a final 179

concentration of 0.1 mM. Protein purification was performed using a HisTrap HP 1 ml 180

column (Amersham Biosciences, Piscataway, NJ). Antisera against gCXCR4 was obtained 181

by immunizing (injection of 1 mg/ml protein which was mixed with Freund's complete 182

adjuvant [Sigma-Aldrich] on days 1, 14 and 28) a New Zealand White rabbit (Taiwan 183

Livestock Research Institute, Tainan, Taiwan) with recombinant gCXCR4-extracellular 184

domain I-extracellular domain III fusion protein. The antiserum collected at day 0 (before 185

treatment) was used as control. The blood samples were incubated at 37℃for 1 h and left 186

overnight at 4 ℃ . The supernatant (containing rabbit anti-gCXCR4 antiserum) was 187

collected after centrifuging at 900 ×g for 10 min at 4 °C. The rabbit anti-gCXCR4 188

antiserum was stored at –20 °C. 189

2.10 Immunofluorescence staining 190

The head kidney and intestines were obtained from healthy groupers and treated with 30 % 191

sucrose at 4 ℃ overnight. The different tissue blocks were covered with an optimal cutting 192

temperature compound (Tissue-Tek®; Sakura Finetek, Tokyo, Japan), and the samples were 193

slowly placed into liquid nitrogen. The frozen tissue block was transferred into a cryotome 194

cryostat and 5 μm-thick sections were cut. Each slide was fixed with 3% paraformaldehyde 195

(Kanto Chemical, Tokyo, Japan) and incubated at room temperature for 30 min. The 196

(12)

12

samples were then washed with 1× PBST (0.1% Tween 20, 1× PBS) and blocked with 5 % 197

skim milk. Rabbit anti-gCXCR4 antisera (1:200 dilution) were added and subsequence 198

Alexa Fluor® 594 goat anti-rabbit IgG (1:200 dilution) (H+L) (Invitrogen) secondary 199

antibody was added. The nuclei were stained with Hoechst 33342 (Invitrogen) at room 200

temperature for 20 min, then washed extensively with 1×PBS and mounted on a coverslip 201

with mounting medium. 202

2.11 Cell proliferation assay 203

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrasodium bromide (MTT) assay was performed 204

using Cell Proliferation Kit I ( Roche ) to analyze the effects of gCXCR4-GFP 205

overexpression. 5×104 Epinephelus coicoides fin cells (GF-1, BCRC 960094) were seeded 206

in 24-well plate and grown in a humidified incubator operating at 28°C in an antibiotic-free 207

L15 medium (Life Technologies, Carlsbad, CA, USA) supplemented with 5% v/v 208

heat-inactivated fetal bovine serum (FBS) (Chen et al., 2008). After 24hr, when cells 209

attached completely, pcDNA3.1-CT-GFP-CXCR4 or pcDNA3.1-CT-GFP vector was 210

transfected using Lipofectamine 2000 (Invitrogen). Then 20μl of MTT labeling reagent was 211

added to each well and cultured for 4 hours in incubator. To dissolve formazan crystals, 212

200μl solubilization solution was added to each well. Covered with tinfoil and agitate cells 213

on orbital shaker for 10 min. Read absorbance at 570 nm. 214

(13)

13 3. Results

215

3.1. Characterization of full-length gCXCR4 216

Orange-spotted grouper CXCR4 (gCXCR4) contained an open reading frame (ORF) of 217

1,104 nucleotides encoding a 367 a.a. protein with a predicted molecular weight of 40.37 218

kDa (Fig. 1). The structure of the protein was predicted to have seven-transmembrane 219

domains, four extracellular domains and four cytoplasmic domains. The DRY motif was 220

found at the second intracellular loop. A conserved cysteine residue on each of the four 221

extracellular domains located at positions Cys33, Cys118, Cys198 and Cys292. The C-terminal 222

is a region rich in serine and threonine residues. 223

The results of different species CXCR4 alignment (Fig.2) showed that gCXCR4 not 224

only similar to fish (zebrafish [58%], carp [54%], rainbow trout [53%] and turbot [52%]) 225

but also to mammalian (human [51%) and mouse [50%)) and the conserved regions 226

appeared in seven putative transmembrane domains (Fig. 2). The sequence of extracellular 227

domains showed very diverse in different species. gCXCR4 has 22.1%, 8.5% , 28.8%, 228

32.2%, 33.6% and 29.3% similarity to human, mice, common carp, zebrafish, rainbow trout 229

and turbot, respectively (Table 2). 230

3.2. Phylogenetic analysis of gCXCR4 231

There are six groups of CXCRs on the phylogenetic tree. Interestingly, within the group of 232

CXCR4, mammalian and fish origins of CXCR4s was clear separated except for gCXCR4 233

(14)

14

and Petromyzon marinus CXCR4 (Fig. 3). This indicated that gCXCR4 might be a common 234

ancestor to other CXCR4 proteins. 235

3.3. in vitro and in vivo expression of gCXCR4 236

To clarify the role of gCXCR4 in grouper, the expression of gCXCR4 in different 237

growth stages of grouper was measured by real-time PCR. gCXCR4 can be detected in all 238

examined fish samples (from 1dph to 40 dph). The expression levels of gCXCR4 was up 239

regulated and fluctuated in the period 1-4 dph and 6-8 dph , the gCXCR4 expressions are < 240

50. Two higher expression peaks (>100, p < 0.05) were observed at 18 dph and 38 dph (Fig. 241

4). 242

To evaluate the effect of gCXCR4 overexpression on cell proliferation, GF-1 cells 243

were transfected by pcDNA3.1-gCXCR4-GFP or pcDNA3.1-GFP overexpression vector. 244

The results showed that overexpression of gCXCR4-GFP for 12 h, 24 h, and 36 h had no 245

significant effects on cell viability (Fig. 5) but significantly repressed after 48 h (p < 0.05) 246

(Fig. 5) (n=5 per group). 247

3.4. Expression patterns of gCXCR4 on head kidney and intestine 248

gCXCR4 was mainly expressed in the head kidney (Fig. 6A) and on the surface of intestinal 249

villi of intestine (Fig. 6B). 250

3.5 The expressions of gCXCR4 in different organs of grouper 251

gCXCR4 was highly expressed in eye, gill, brain and important immune organs such as 252

(15)

15

spleen and head kidney (Fig. 7). Higher levels of expression were detected in eye, gill, 253

spleen, brain and head kidney tissues. Lower levels of expression were detected in fin, 254

muscle and heart tissues. Barely any gCXCR4 transcript was detected in the intestine (Fig. 255

7B). 256

3.6 The expressions of gCXCR4 after LPS or NNV challenge 257

The expression level of gCXCR4 in spleen was significantly increased after 6 h 258

post-injection of LPS (p < 0.05) and decreased at 24 h and 48 h post-injection of LPS (p < 259

0.05). At 72 h post-injection, gCXCR4 had returned to the base level as the control (Fig. 260

8A). 261

Forty-eight h after NNV infection, the juvenile groupers exhibited abnormal behaviors 262

such as loss of equilibrium and spiral swimming pattern and NNV can be detected at 72 h. 263

gCXCR4 expression was also up-regulated at the time point which was 72 h post-NNV 264

infection in eyes (p < 0.05) (Fig. 8B). 265

(16)

16 4. Discussion

267

The chemokine system has an important role in the host immune response against microbial 268

pathogens and provides a link between innate and adaptive immunity (Murphy et al., 2000). 269

The similar structure of gCXCR4 to other species (Alabyev et al., 2000; Tsuchida et al., 270

2007; Jia and Zhang, 2009) contains seven transmembrane regions, four extracellular 271

regions and four intracellular regions, and a conserved DRY motif (Fig. 2). The predicted 272

function of the gCXCR4 DRY motif was supported by the results of amino acid sequence 273

alignments of gCXCR4 and CXCR4 of other species, which function had been 274

demonstrated as important to G protein coupling (Doranz et al., 1999). The transmembrane 275

regions as well as the cysteine residue positions in the extracellular regions appear to be 276

highly conserved in CXCR4 evolution (Federsppiel et al., 1993; Heesen et al., 1996; 277

Alabyev et al., 2000; Tsuchida et al., 2007; Jia and Zhang, 2009). The posttranslational 278

modification, i.e. the tyrosine residues of the N terminus are sulfated in Golgi, of human 279

CXCR4 plays a crucial role on the infective ability of HIV (Farzan et al., 2002). However, 280

these tyrosine residues were not conserved in gCXCR4 (Figs.1 and 2), suggesting that the 281

posttranslational modification of gCXCR4 N-terminus is different. In addition, many serine 282

and threonine residues were identified in the C-terminus of gCXCR4 and might have the 283

modifications, i.e. phosphorylated as a prerequisite of signal transfer (Berson et al., 1996), 284

like other protein in CXCR4 family. 285

(17)

17

CXCR4 is expressed mainly in immune organs and central nervous system: thymus 286

and spleen of mouse (Heesen et al., 1996), chicken bursa (Liang et al., 2001), primate 287

(Macaca mulatta) brain (Federsppiel et al., 1993) and cattle locus coeruleus, cerebellum and 288

pons (Rimland et al., 1991). In grouper, gCXCR4 was highly expressed in NNV major target 289

organs, such as eyes and brain, and major lymphoid organs, such as gill, spleen and head 290

kidney. This also been reported in other fish species (Daniels et al., 1999; Jia and Zhang, 291

2009) in which CXCR4 expressed in central nervous system and immune system. 292

Interestingly, the expression of gCXCR4 in eye other than the immune related organs or 293

central nervous system has never been reported which raised the other possible function of 294

gCXCR4. Accordingly the grouper major lymphoid organs such as spleen, head kidney, gill 295

and mucosa-associated tissues appeared to be regions of gCXCR4 overproduction (Press 296

and Evensen., 1999). Furthermore, CXC chemokine system originates from the central 297

nervous system and may participate in central nervous system development (Huising et al., 298

2003). 299

SDF1/CXCR4 signaling plays a critical role in embryonic development and is essential 300

for development of cardiovascular, central nervous system, bone marrow colonization and 301

hematopoiesis in mice (Ma et al., 1998; Tachibana et al., 1998). In fish, CXCR4 has been 302

found in the early stage of zebrafish embryo and related to tissue polarity (Chong et al., 303

2001; Haas and Gilmour, 2006). The gene expression of gCXCR4 was highly expressed in 304

(18)

18

the period day14-day20 and day34-day40 larva that is coincided with dorsal spine formation 305

and pigmentation (Katsutoshi and Hiroshi, 2009). The results implied that chemokine 306

system exist in early developmental stage and play a key role in grouper development. 307

Immunohistofluorescence staining suggested that the protein gCXCR4 is expressed in 308

lymphoid organs (Fig. 6A) and mainly on the surface of intestinal villi. This may be due to 309

eyes, gills and surface of intestinal villi are continuously exposed to an environment which 310

may have potentially pathogenic microbes. LPS is an endotoxin constituent of the outer 311

membrane of Gram-negative bacteria which can induce immune responses and 312

inflammation (Raetz et al., 2008). In fish, it has been demonstrated that LPS can stimulate 313

the proliferation of neutrophils, monocytes, B lymphocytes and macrophages in a response 314

against LPS-induced inflammation (Swain et al., 2008). The results shown that LPS can 315

up-regulate the expression of gCXCR4 in the spleen and this also been showed in head 316

kidney and spleen of turbot after challenging with Vibrio harveyi (Jia et al., 2009). 317

gCXCR4 mRNA was up-regulated at 3 days post-infection and was significantly 318

increased in the eyes (Fig. 8B), suggesting that gCXCR4 is not only involved in the 319

response to bacteria invasion, but also have response to NNV infection. Although the eye 320

has been known to express chemokine receptors, such as CXCR1 and CXCR2 in mammals 321

(Goczalik et al., 2008), our detection of abundant gCXCR4 in the organs and significantly 322

up-regulated in NNV-infected fish was unexpected. Interestingly, grouper eye is one of the 323

(19)

19

main organs for NNV replication (Munday et al., 2002) and the immune response of NNV 324

infection involving macrophage-like cells and lymphocytes migrate to the eyes (Grotmol et 325

al., 1997; Nilsen et al., 2001; Munday et al., 2002). Taking those results together, we 326

hypothesized that the gCXCR4 expression is related to NNV infection and which may cause 327

by the immune related cells migration. However, too much gCXCR4 in cell could result in 328

significant growth obstruction which might due to the other functions of CXCR4 (Bleul et 329

al., 1996; Ganju et al., 1998). 330

In summary, our data indicated that the expression of grouper CXCR4 is regulated by 331

LPS or NNV challenge, and is expressed during embryogenesis, speculating its importance 332

in both immune and early developmental stage. The characterization of gCXCR4 between a 333

teleost fish and mammalians has provided valuable information for future functional 334

analysis of the gene. 335

(20)

20 Acknowledgements

337

We thank Dr. Brian D. Hoyle for editing the manuscript. This research was supported 338

by the National Science Council (NSC97-2313-B-006-001-MY3), and the Landmark 339

Project (B0127) of National Cheng Kung University, the plan of University Advancement, 340

Ministry of Education, Taiwan. 341

(21)

21 References

342

Alabyev, B.Y., Najakshin, A.M., Mechetina, L.V., Taranin, A.V., 2000. Cloning of a CXCR4 343

homolog in chondrostean fish and characterization of the CXCR4-specific structural 344

features. Dev Comp Immunol 24, 765-770. 345

Baldwin, J.M., 1994. Structure and function of receptors coupled to G proteins. Curr Opin 346

Cell Biol 6, 180-190. 347

Berson, J.F., Long, D., Doranz, B.J., Rucker, J., Jirik, F.R., Doms, R.W., 1996. A 348

seven-transmembrane domain receptor involved in fusion and entry of T- cell-tropic 349

human immunodeficiency virus type 1 strains. J Virol 70, 6288-6295. 350

Bleul, C.C., Farzan, M., Choe, H., Parolin, C., Clark-Lewis, I., Sodroski, J., Springer, T. A., 351

1996. The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks 352

HIV-1 entry. Nature 6594, 829-833. 353

Chen, Y.M., Su, Y.L., Shie, P.S., Hwang, S.L., H.L., Chen, T.Y., 2008. Grouper Mx confers 354

resistance to nodavirus and interacts with coat protein. Dev Comp Immunol 32, 355

825-836. 356

Chen, Y.M., Kuo, C.E., Wang, T.Y., Shie, P.S., Wang, W.C., Huang, S.L., Tsai, T.J., Chen, 357

P.P., Chen, J.C., Chen, T.Y., 2010. Cloning of an orange-spotted grouper Epinephelus 358

coioides heat shock protein 90AB (HSP90AB) and characterization of its expression 359

in response to nodavirus. Fish Shellfish Immunol 28, 895-904. 360

(22)

22

Chong, S.W., Emelyanov, A., Gong, Z., Korzh, V., 2001. Expression pattern of two 361

zebrafish genes, cxcr4a and cxcr4b. Mech Dev 109, 347-354. 362

Daniels, G.D., Zou, J., Charlemagne, J., Partula, S., Cunningham, C., Secombes, C.J., 1999. 363

Cloning of two chemokine receptor homologs (CXC-R4 and CC-R7) in rainbow trout 364

Oncorhynchus mykiss. J Leukoc Biol 65, 684-690. 365

Doranz, B.J., Orsini, M.J., Turner, J.D., Hoffman, T.L., Berson, J.F., Hoxie, J.A., Peiper, 366

S.C., Brass, L.F., Doms, R.W., 1999. Identification of CXCR4 domains that support 367

coreceptor and chemokine receptor functions. J Virol 73, 2752-2761. 368

Farzan, M., Babcock, G.J., Vasilieva, N., Wright, P.L., Kiprilov, E., Mirzabekov, T., Choe, 369

H., 2002. The role of post-translational modifications of the CXCR4 amino terminus 370

in stromal-derived factor 1 alpha association and HIV-1 entry. J Biol Chem 277, 371

29484-29489. 372

Federsppiel, B., Melhado, I.G., Duncan, A.M.V., Delaney, A., Schappert, K., Clark-Lewis, I., 373

Jirik, F.R., 1993. Molecular cloning of the cDNA and chromosomal localization of the 374

gene for a putative seven-transmembrane segment (7-TMS) receptor isolated from 375

human spleen. Genomics 16, 707-712. 376

Feng, Y., Broder, C.C., Kennedy, P.E., Berger, E.A., 1996. HIV-1 entry cofactor: functional 377

cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 272, 378

872-877. 379

(23)

23

Ganju, R.K., Brubaker, S.A., Meyer, J., Dutt, P., Yang, Y., Qin, S., Newman, W., Groopman, 380

J.E., 1998. The alpha-chemokine, stromal cell-derived factor-1alpha, binds to the 381

transmembrane G-protein-coupled CXCR-4 receptor and activates multiple signal 382

transduction pathways. J Biol Chem 273, 23169-23175 383

Goczalik, I., Ulbricht, E., Hollborn, M., Raap, M., Uhlmann, S., Weick, M., Pannicke, T., 384

Wiedemann, P., Bringmann, A., Reichenbach, A., Francke, M., 2008. Expression of 385

CXCL8, CXCR1, and CXCR2 in neurons and glial cells of the human and rabbit 386

retina. Invest Ophthalmol Vis Sci 49, 4578–4789. 387

Grotmol, S., Totland, G.K., Torud, K., Hjeltnes, B.K., 1997. Vacuolating encephalopathy 388

and retinopathy associated with a nodavirus-like agent: a probable cause of mass 389

mortality of cultured larval and juvenile Atlantic halibut Hippoglossus hippoglossus. 390

Dis Aquat Organ 29, 85-97. 391

Haas, P., Gilmour, D., 2006. Chemokine signaling mediates self-organizing tissue migration 392

in the zebrafish lateral line. Dev Cell 10, 673-680. 393

Heesen, M., Berman, M.A., Benson, J.D., Gerard, C., Dorf, M.E., 1996. Cloning of the 394

mouse fusin gene, homologue to a human HIV-1 co-factor. J Immunol 157, 395

5455-5460. 396

Hesselgesser, J., Taub, D., Baskar, P., Greenberg, M., Hoxie, J., Kolson, D. L., Horuk, R., 397

1998. Neuronal apoptosis induced by HIV-1 gp120 and the chemokine SDF-1 alpha is 398

(24)

24

mediated by the chemokine receptor CXCR4. Curr Biol 10, 595-598. 399

Horuk, R., 2009. Chemokine receptor antagonists: overcoming developmental hurdles. Nat 400

Rev Drug Discov 8, 23-33. 401

Huising, M.O., Stet, R.J., Kruiswijk, C.P., Savelkoul, H.F., Lidy Verburg-van Kemenade, 402

B.M., 2003. Molecular evolution of CXC chemokines: extant CXC chemokines 403

originate from the CNS. Trends Immunol 24, 307-313. 404

Jia, A., Zhang, X.H., 2009. Molecular cloning, characterization, and cxpression analysis of 405

the CXCR4 gene from turbot: Scophthalmus maximus. J Biomed Biotechnol 406

doi:10.1155/2009/767893. 407

Katsutoshi, K., Kohno, H., 2009. Morphological development of larval and juvenile 408

blacktip grouper, Epinephelus fasciatus. Fish Sci 75, 1239–1251. 409

Kim, C.H., Broxmeyer, H.E., 1999. Chemokines: signal lamps for trafficking of T and B 410

cells for development and effector function. J Leukoc Biol 65, 6-15. 411

Kuo, H.C., Wamg, T.Y., Chen, P.P., Chen, Y.M., Chuang, H.C., Chen T.Y., 2011. Real-time 412

quantitative PCR asaay for monitoring of nervous necrosis virus infection in grouper 413

aquaculture. J Clin Microbiol 49, 1090-1096. 414

Liang, T.S., Hartt, J.K., Lu, S., Martins-Green, M., Gao, J.L., Murphy, P.M., 2001. Cloning, 415

mRNA distribution, and functional expression of an avian counterpart of the 416

chemokine receptor/HIV coreceptor CXCR4. J Leukoc Biol 69, 297-305. 417

(25)

25

Ma, Q., Jones, D., Borghesani, P.R., Segal, R. A., Nagasawa, T., Kishimoto, T., Bronson, 418

R.T., Springer, T.A., 1998. Impaired B-lymphopoiesis, myelopoiesis, and derailed 419

cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad 420

Sci U S A 95, 9448-9453. 421

Munday, B.L., Kwang, J., Moody, N., 2002. Betanodavirus infections of teleost fish: a 422

review. J Fish Dis 25, 127-142. 423

Murphy, P.M., Baggiolini, M., Charo, I.F., Hebert, C.A., Horuk, R., Matsushima, K., Miller, 424

L.H., Oppenheim, J.J., Power, C.A., 2000. International union of pharmacology. XXII. 425

nomenclature for chemokine receptors. Pharmacol Rev 52, 145-176. 426

Nilsen, R., Ranheim, T., Hansen, M.K., Taksdal, T., Totland, G.K., 2001. Pathology in 427

persistent nodavirus infected juvenile Atlantic halibut Hippoglossus hippoglossus. In: 428

Tenth International Conference of the European Association of Fish Pathologists 429

Abstract P-117, 9-14. 430

Olson, T.S., Ley, K., 2002. Chemokines and chemokine receptors in leukocyte trafficking. 431

Am J Physiol Regul Integr Comp Physiol 283, R7-28. 432

Press, C. M.,and Evensen, Ø., 1999. The morphology of the immune system in teleost fishes. 433

Fish Shellfish Immunol 9, 309-318. 434

Raetz, C.R., Whitfield, C., 2002. Lipopolysaccharide endotoxins. Annu Rev Biochem. 71, 435

635-700. 436

(26)

26

Rimland, J., Xin, W., Sweetnam, P., Saijoh, K., Nestler, E.J., Duman, R.S., 1991. Sequence 437

and expression of a neuropeptide Y receptor cDNA. Mol Pharmacol 40, 869-875. 438

Swain, P., Nayak, S.K., Nanda, P.K., Dash, S., 2008. Biological effects of bacterial 439

lipopolysaccharide (endotoxin) in fish: a review. Fish Shellfish Immunol 25, 191-201. 440

Tachibana, K., Hirota, S., Iizasa, H., Yoshida, H., Kawabata, K., Kataoka, Y., Kitamura, Y., 441

Matsushima, K., Yoshida, N., Nishikawa, S., Kishimoto, T., Nagasawa, T., 1998. The 442

chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal 443

tract. Nature 393, 591-594. 444

Tsuchida, S., Kagi, A., Takahashi, T., 2007. Characterization of cDNA and genomic 445

sequences encoding a canine chemokine receptor, CXCR4 and its ligand CXCL12. 446

Vet Immunol Immunopathol 116, 219-225. 447

Valentin, G., Haas, P., Gilmour, D., 2007. The chemokine SDF1a coordinates tissue 448

migration through the spatially restricted activation of Cxcr7 and Cxcr4b. Curr Biol 449

17, 1026-1031. 450

(27)

27 Legends of figures

452

Fig. 1. Nucleotide sequence and deduced amino acid sequence of the open reading 453

frame of Epinephelus coioides cDNA. The bold letters represent the start codon (ATG) and 454

the stop codon (TAG). The regions of seven-transmembrane domains are at amino acids 455

50 –72, 85–104, 119–141, 161–180, 216–238, 259–281 and 301–323. The Genbank 456

accession number of gCXCR4 is HQ185191. 457

Fig. 2. Protein alignment and analysis of gCXCR4 with homologues from other species. 458

The other species used for comparison were Homo sapiens (GenBank accession number 459

CAA12166), Mus muluscus (GenBank accession number AAH31665), Cyprinus carpio, 460

Oncorhynchus mykiss (GenBank accession number CAA04493) and Psetta maxima 461

(GenBank accession number ABP48751). The black letters denote the consensus sequence 462

of CXCR4 of the different species. The bars represent transmembrane region of gCXCR4 463

determined using the TMHMMM program in the ExPASy Proteomics Server database. The 464

DRY motif is boxed. The asterisk represents conserved cysteine residue on each of the four 465

extracellular domains that located at positions 33aa, 118aa, 198aa and 292aa. 466

Fig. 3. Phylogenetic analysis of CXCR4 protein family members. The amino acids of the 467

different CXCR4 species obtained from the NCBI GenBank were aligned using ClustalW. 468

The Neighbor-Joining tree was created by MEGA4.0 software with a bootstrap value of 469

1,000. Accession numbers of chemokine receptors amino acid sequences obtained from 470

(28)

28

GenBank were: Homo sapiens CXCR1 NP_000625; Mus musculus CXCR1 NP_839972; 471

Cyprinus carpio CXCR1 BAA31458; Takifugu rubripes CXCR1 NP_001072110; Homo 472

sapiens CXCR2 NP_001161770; Mus musculus CXCR2 NP_034039; Bos taurus CXCR2 473

ABC59060; Homo sapiens CXCR3 EAX05283; Danio rerio CXCR3a NP_001082899; 474

Ctenopharyngodon idella CXCR3 AAW69766; Petromyzon marinus CXCR4 AAO21209; 475

Epinephelus coioides CXCR4 HQ185191; Homo sapiens CXCR4 CAA12166; Mus 476

musculus CXCR4 AAH98322; Bos taurus CXCR4 NP_776726; Sus scrofa CXCR4 477

AAZ32767; Cyprinus carpio CXCR4 BAA32797; Oncorhynchus mykiss CXCR4 478

CAA04493; Salmo salar CXCR4 BT060355; Danio rerio CXCR4 AAF1756; Ictalurus 479

punctatus CXCR4 ACS45337; Acipenser ruthenus CXCR4 CAB60252; Xenopus laevis 480

CXCR4 AAI10722; Oryzias latipes CXCR4 ABC41565; Psetta maxima CXCR4 481

ABP48751; Homo sapiens CXCR5 AAI10353; Mus musculus CXCR5 AAH64059; 482

Ctenopharyngodon idella CXCR5 ACZ06880; Mus musculus CXCR6 NP_109637; Homo 483

sapiens CXCR6 NP_006555; Bos taurus CXCR6 NP_001014859; Homo sapiens CXCR7 484

NP_064707; Mus musculus CXCR7 NP_031748 and Xenopus laevis CXCR7 485

NP_001082236. 486

Fig. 4. Gene expression profile of gCXCR4 was examined in different development 487

stages of Epinephelus coioides. The total RNA was isolated from different stages and gene 488

expression of gCXCR4 was determined by real-time PCR. 489

(29)

29

Fig. 5. Effects of overexpression of gCXCR4 on GF-1 cell proliferation. The cells 490

proliferation was quantified by measuring MTT absorbance at 570 nm. Vertical bars 491

indicate the mean ± S.E (N=3). **p < 0.01. The Blank was the spontaneous proliferation of 492

GF-1 cells without treating any plasmid; the GFP was the group transfected with the same 493

backbone of the plasmid to the gCXCR4-GFP group without inserting the gCXCR4. 494

Fig. 6. Expression of gCXCR4 in (A) head kidney and (B) intestine of healthy grouper 495

using immunohistofluorescence staining. (a) and (d): Nucleus was detected using Hoechst 496

33342 (blue). (b) Control experiments were carried out with control rabbit antiserum as the 497

primary antiserum, and visualized with Alexa Fluor® 594 goat anti-rabbit IgG (H+L) (red). 498

(e) The expression of gCXCR4 was detected using rabbit anti-gCXCR4 antiserum and 499

visualized with Alexa Fluor® 594 goat anti-rabbit IgG (H+L). (c) Merged image from 500

figures (a) and (b). (f) Merged image from figures (d) and (e). Bars = 1 mm (A) and 50 μm 501

(B). 502

Fig. 7. Gene expression of gCXCR4 in different tissues. (A) RT-PCR and (B) real-time 503

PCR analysis of gCXCR4 gene expression in different tissues including eye, fin, gill, muscle, 504

head kidney, heart, spleen ,intestine and brain of healthy adult grouper. β-actin 505

amplification was used as an internal control. Vertical bars indicate the mean ± S.E (N=3). 506

*p < 0.05. 507

Fig. 8. Expression level of gCXCR4 mRNA in grouper after challenge with LPS (A) 508

(30)

30

and NNV (B). (A) Relative expression level of gCXCR4 mRNA in spleen of grouper after 509

challenge with LPS or PBS. (B) Analysis of expression of gCXCR4 gene in control or NNV 510

infected juvenile grouper or eye of juvenile grouper groups by real-time RT-PCR. gCXCR4 511

mRNA levels (relative to β-actin mRNA) between different time were compared by the 512

t-test. Vertical bars indicate the mean ± S.E (N=3). *p < 0.05. 513

(31)

31

Figure 1

(32)

32

Figure 2

(33)

33

Figure 3

(34)

34

Figure 4

(35)

35

Figure 5

(36)

36

Figure 6

(37)

37

Figure 7

(38)

38

Figure 8

(39)

39

Table1. Primers used in this study

Name Sequence gCXCR4-5SP1 CAGTAACACACAAGGAGGACCAG gCXCR4-5SP2 GCTGATGAATGCCAGGATGAGCACACT gCXCR4-5SP3 AGCAGCAGAGAGATGTAGCCG gCXCR4-3SP1 CGCTACCTGGCTGTAGTCCGA gCXCR4-3SP2 CTTCGTGTGCTGGCTGCCGTATGG gCXCR-F ATGTCGTACTATGAGCATATCGTCTTCG gCXCR-R CTAGCTGGAATGTAAACTGGAGGACTC gCXCR4-GFP-R GGCTGGAATGTAAACTGGAGGACTC gCXCR4-EXI-F ATCGTAGGATCCGATGTCGTACTATGAGCAT gCXCR4-EXI-R ACGATGTCGACAGGTAAGAAGACCTGCTGGAG gCXCR4-EXIII-F ATCGTAGTCGACCCGGACTTGATCTATGCCC gCXCR4-EXIII-R ACGATCTCGAGGTGGAAGACTGCAACCCAG Q-gCXCR4-F CACACTGCTGCCTGAACCCACTGCT Q-gCXCR4-R CTAGCTGGAATGTAAACTGGAGGACTC β-actin-F TGCCTCTGGTCGTACCACTGGTATTGTC β-actin-R GGCAGCAGTGCCCATCTCCTGCTCGA

(40)

40

Table 2. Protein sequences similarity* of CXCR4 from different species E. coioides H. sapiens M. musculus C. carpio D. rerio O. mykiss P. maxima Extracellular domains E. coioides 100 22.1 8.5 28.8 32.2 33.6 29.3 H. sapiens 100 15.9 30.3 36.3 31.9 30.3 M. musculus 100 13.7 16.2 14.6 14.9 C. carpio 100 75.5 43.4 33.1 D. rerio 100 51.4 37.3 O. mykiss 100 46.6 P. maxima 100 Transmembrane domains E. coioides 100 57.7 32.9 59.0 59.3 59.1 62.3 H. sapiens 100 35.4 71.6 70.7 75.0 75.2 M. musculus 100 37.9 36.0 37.3 37.9 C. carpio 100 92.3 80.1 84.0 D. rerio 100 77.8 82.7 O. mykiss 100 84.8 P. maxima 100 Intracellular domains E. coioides 100 43.6 15.8 44.0 45.4 41.4 38.8 H. sapiens 100 24.8 64.4 62.7 58.4 59.4 M. musculus 100 20.2 19.8 22.2 21.4 C. carpio 100 89.8 73.5 74 D. rerio 100 71.4 73.5 O. mykiss 100 76 P. maxima 100

數據

Table 2. Protein sequences similarity* of CXCR4 from different species

參考文獻

相關文件

Late Qing Master Taixu, recognized as the leader of Buddhist reform movement, and several Buddhist intellectuals collaborated to remodel and revive Buddhist

Therefore, the “Buddhism for this World” is a movement, in certain aspects, of “returning to the India’s primitive Buddhism and early Mahāyāna Buddhism.” The proposing of

“The Story of Sadāprarudita in the Aṣṭasāhasrikā Prajñāpāramitā Sūtra and its Contribution to the Study of Early Mahāyāna Buddhism.” In Peter Skilling and Justin McDaniel

In response to the changing needs of society, the rapid development of science and technology, the views of stakeholders collected through various surveys and

In response to the changing needs of society, the rapid development of technology, views of stakeholders collected through various engagement activities and events

Xianggang zaji (miscellaneous notes on Hong Kong) was written by an English and translated into Chinese by a local Chinese literati.. Doubts can therefore be cast as to whether

An information literate person is able to recognise that information processing skills and freedom of information access are pivotal to sustaining the development of a

Office of Early Childhood Development, Virginia Department of Social Services (2013). Milestones of child development: A guide to young children’s learning and development from