structure determination and characterization … · structure determination and characterization of...

149
STRUCTURE DETERMINATION AND CHARACTERIZATION OF THE ENVELOPE PROTEINS OF SHRIMP WHITE SPOT SYNDROME VIRUS (WSSV) TANG XUHUA (B. Sc., Xiamen University, China) A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY DEPARTMENT OF BIOLOGICAL SCIENCES NATIONAL UNIVERSITY OF SINGAPORE 2007

Upload: vannga

Post on 13-Mar-2019

230 views

Category:

Documents


0 download

TRANSCRIPT

STRUCTURE DETERMINATION AND

CHARACTERIZATION OF THE ENVELOPE PROTEINS

OF SHRIMP WHITE SPOT SYNDROME VIRUS (WSSV)

TANG XUHUA

(B. Sc., Xiamen University, China)

A THESIS SUBMITTED FOR

THE DEGREE OF DOCTOR OF PHILOSOPHY

DEPARTMENT OF BIOLOGICAL SCIENCES

NATIONAL UNIVERSITY OF SINGAPORE

2007

To My Family

i

Acknowledgements

I would like to express my deepest gratitude to my supervisor, Professor Hew Choy Leong, for his invaluable guidance, advice and mentorship. Thanks for giving me an opportunity to commence my research work in Department of Biological Sciences and providing a motivating, enthusiastic, and critical atmosphere for my research.

I am also deeply indebted to Dr. Jayaraman Sivaraman, for his invaluable and

selfless assistance, from whom I obtained the excellent training in the X-ray crystallography. Thanks for his patience and support in my research project and his constructive comments during my thesis time.

I acknowledge Dr. Wu Jinlu for his collaboration on electron microscopy and virus

purification. I am deeply indebted to Dr. Huang Canhua, Dr. Lin Qingsong, Dr. Li Zhengjun, Dr. Song Wenjun and Mr. Shashikant Joshi for their technical guidance molecular biology and mass spectrometry. Thanks for all the insightful comments and constructive criticisms at different stages of my research.

I would like to extend my thanks to all my colleagues and friends for their full

support and help. Thanks specially go to Ms. Chen Jing and Ms. Zhuang Ying for their sincerity and friendship.

Most importantly, none of my achievements is possible without the love and

patience of my family. My family has been a constant source of love, concern, support and strength for me in all my life. I would like to express my heart-felt gratitude to my dear parents for their selfless love and being my spiritual support all the way. Thanks also go to my elder brother, Don, for all the support and encouragement he gave me. I would also like to thank my husband, zhaohua, for his help, understanding and encouragement throughout my graduate studies. My deepest gratitude is for my late grandmother Mdm. Chen Ruiduan who gave me her unconditional love and support. She taught me the good things that really matter in my life. The happy memory of my grandma will provide a persistent inspiration for my journey in life.

ii

Table of Contents

Acknowledgements ............................................................................................................ i

Table of Contents .............................................................................................................. ii

Summary........................................................................................................................... vi

List of Tables ................................................................................................................... vii

List of Figures................................................................................................................. viii

List of Abbreviations ........................................................................................................ x

List of Abbreviations ........................................................................................................ x

1 Literature Review............................................................................................... 1

1.1 Biology of White Spot Syndrome Virus .............................................................. 2

1.1.1 Overview .............................................................................................................. 2

1.1.2 Morphology of WSSV.......................................................................................... 5

1.1.3 The Genome Structure of WSSV ....................................................................... 11

1.1.4 Taxonomy of WSSV .......................................................................................... 13

1.1.5 Envelope Proteins of WSSV .............................................................................. 16

1.1.6 Problems Faced in Study of Envelope Proteins of WSSV................................. 19

1.2 Structural Genomics ........................................................................................... 20

1.2.1 Structural Genomics as a New Research Initiative ............................................ 20

1.2.2 WSSV as a Model for Structural Genomics Study ............................................ 21

1.3 X-ray Crystallography ........................................................................................ 22

1.3.1 Overview ............................................................................................................ 22

1.3.2 Crystallization..................................................................................................... 24

1.3.3 X-ray Diffraction ................................................................................................ 26

1.3.4 Structure Determination ..................................................................................... 27

1.4 Objectives of the Project .................................................................................... 28

1.5 Significance of the Project.................................................................................. 29

1.6 Scope of the Thesis............................................................................................. 29

2 Materials and Methods .................................................................................... 30

2.1 Bacterial Strains, Vectors, Primers and Bacterial Culture ................................. 31

2.2 DNA Manipulation............................................................................................. 32

iii

2.2.1 Amplification of DNA by Polymerase Chain Reaction (PCR) .......................... 32

2.2.2 DNA Digestion and Ligation.............................................................................. 32

2.2.3 Agarose Gel Electrophoresis and DNA Purification.......................................... 33

2.2.4 Preparation of E.coli Competent Cells ............................................................... 33

2.2.5 Transformation of Bacterial Cells ...................................................................... 34

2.2.6 DNA Sequencing................................................................................................ 35

2.3 Virus Treatment and Analysis ............................................................................ 36

2.3.1 Virus Propagation and Purification .................................................................... 36

2.3.2 Treatment of the Intact Virus with Detergent..................................................... 38

2.3.3 Preparation of WSSV Genomic DNA................................................................ 38

2.3.4 Transmission Electron Microscopy.................................................................... 39

2.3.5 Localization Study by Immunoelectron Microscopy ......................................... 39

2.4 Protein Manipulation .......................................................................................... 40

2.4.1 Protein Expression and Solubility Test .............................................................. 40

2.4.2 Expression of Seleno-Methionine Substituted Protein....................................... 40

2.4.3 GST fusion Protein Purification and Removal of GST tag ................................ 41

2.4.4 Purification of Untagged Protein........................................................................ 42

2.4.5 Antibody Preparation.......................................................................................... 42

2.5 Protein Analytical Techniques ........................................................................... 44

2.5.1 SDS-PAGE Gel Electrophoresis ........................................................................ 44

2.5.2 Western blot Analysis......................................................................................... 45

2.5.3 In-Gel Digestion, MALDI-TOF and Tandem MS Sequencing.......................... 46

2.5.4 N-Terminal Sequencing...................................................................................... 47

2.5.5 Circular Dichroism (CD) Spectrum.................................................................... 48

2.5.6 Dynamic Light Scattering (DLS) ....................................................................... 49

2.6 Crystallization..................................................................................................... 51

2.7 Data Collection................................................................................................... 51

2.8 Structure Determination ..................................................................................... 51

3 Characterization of WSSV Envelope Proteins .............................................. 53

3.1 Characterization of Envelope Protein VP300..................................................... 54

3.1.1 Identification of VP300 by Mass Spectrometry ................................................. 54

iv

3.1.2 Expression and Purification of VP300 ............................................................... 54

3.1.3 Localization Study of VP300 ............................................................................. 56

3.1.3.1 Western Analysis of VP300 ............................................................................... 56

3.1.3.2 TEM Immunogold-Labeling Analysis ............................................................... 59

3.1.4 Neutralization Assay of VP300 .......................................................................... 59

3.1.5 Implications ........................................................................................................ 62

3.2 Localization Study of VP26 in WSSV virion..................................................... 64

3.2.1 Immunogold labeling of VP26 in WSSV particles ............................................ 64

3.2.2 Implications ........................................................................................................ 66

3.3 Candidates for Structure Analysis ...................................................................... 67

3.3.1 Solubility Test of Major Envelope Proteins ....................................................... 67

3.3.2 Purification of VP281 Protein and Crystallization Trials................................... 69

3.3.3 Purification of VP466 Protein and Crystallization Trials................................... 71

4 Crystal Study of Two Major Envelope Proteins............................................ 74

4.1 X-ray Structure of Major Envelope Protein VP28 ............................................. 75

4.1.1 Property of rVP28 Protein .................................................................................. 75

4.1.2 Preparation of SeMet rVP28 Protein.................................................................. 78

4.1.3 Crystallization of rVP28..................................................................................... 78

4.1.4 Structure Determination of rVP28...................................................................... 80

4.1.5 Overall Structure of rVP28................................................................................. 82

4.1.6 Oligomerization of rVP28 .................................................................................. 84

4.2 X-ray Structure of Major Envelope Protein VP26 ............................................. 87

4.2.1 Purification of rVP26 and SeMet rVP26 protein ............................................... 87

4.2.2 Crystallization, Data Collection and Structure Determination of rVP26 ........... 89

4.2.3 Overall Structure of rVP26................................................................................. 92

4.2.4 Oligomerization of rVP26 .................................................................................. 94

4.3 Comparison of VP26 and VP28 ......................................................................... 97

4.3.1 Structure Homolog of VP28 and VP26 .............................................................. 97

4.3.2 Comparison with Other Viral Proteins ............................................................. 104

4.3.3 Implications on Gene Duplication.................................................................... 106

4.3.4 Implications on membrane fusion .................................................................... 109

v

5 General Conclusion and Future Studies ...................................................... 112

Bibliography.................................................................................................................. 118

Appendices..................................................................................................................... 130

List of Publications ....................................................................................................... 134

vi

Summary

White spot syndrome virus (WSSV) is a virulent pathogen known to infect

penaeid shrimp and other crustaceans. It has bacilliform morphology with a tail-like

appendage at one end. The envelope consists of four major proteins. Here we report the

localization and crystal structure of two major envelope proteins VP26 and VP28 from

WSSV at 2.2 and 2.0Å respectively. Their structures are being reported for the first time

for WSSV. Both proteins adopt β-barrels architecture with a protruding region.

Furthermore, we reinvestigated the localization of VP26 and VP28 using immunoelectron

microscopy. Our localization study suggests that VP26 and VP28 are on the outer surface

of the virus and resembled as a spike-like structure on the WSSV envelope, this is the

first convincing observation for VP26. The spike-like structure of VP26 and VP28

observed in our immuno-electron microscopy images matches well with the trimeric

shape of the crystal structure. Based on our present studies combined with previous

findings from other groups, we propose that VP26 and VP28 may anchor on the viral

envelope membrane via their predicted N-terminal transmembrane regions, while leaving

the core β-barrel to protrude outside the envelope to interact with the host receptor or to

fuse with the host cell membrane for effective viral infection.

vii

List of Tables

Table 4.1 Summary of Crystallization Conditions and Cryoprotectants .................... 79

Table 4.2 Data collection and refinement statistics of rVP28..................................... 81

Table 4.3 Summary of Crystallization Conditions and Cryoprotectants .................... 90

Table 4.4 Data collection and refinement statistics of rVP26..................................... 91

viii

List of Figures

Figure 1.1 Clinical sign of WSSV in shrimp and crayfish............................................. 4

Figure 1.2 EM of negatively stained intact WSSV virions ............................................ 7

Figure 1.3 Electron micrograph of WSSV particles treated with 1% Triton X-100 ...... 8

Figure 1.4 EM of negatively stained, naked WSSV nucleocapsids ............................... 9

Figure 1.5 Proposed WSSV structure........................................................................... 10

Figure 1.6 Circular representation of the WSSV genome............................................ 12

Figure 1.7 Bootstrap analysis (100 replicates) of an uprooted phylogenetic tree of the DNA polymerase from WSSV.................................................................... 15

Figure 1.8 Synopsis of structure determination by X-ray crystallography .................. 23

Figure 2.1 Banding of purified WSSV Virus in the sucrose gradient.......................... 37

Figure 3.1 1D SDS-PAGE of the WSSV proteins ....................................................... 55

Figure 3.2 Expression of the purified (His)6-VP300 protein........................................ 57

Figure 3.3 Western blot analysis of WSSV with anti-VP300 IgG............................... 58

Figure 3.4 Localization of WSSV VP300 proteins by immunoelectron microscopy .. 60

Figure 3.5 Neutralization Analysis of WSSV infections ............................................. 61

Figure 3.6 localization of WSSV VP26 proteins by immunoelectron microscopy...... 65

Figure 3.7 Solubility study on the envelope proteins of WSSV .................................. 68

Figure 3.8 Expression of the purified VP281 protein .................................................. 70

Figure 3.9 Expression of the purified VP466 protein .................................................. 72

Figure 4.1 DAS transmembrane prediction result for full length VP28 and VP26...... 76

Figure 4.2 Expression of the rVP28 protein in E.coli BL21 DE3 star ......................... 77

Figure 4.3 Ribbon diagram of rVP28 monomer........................................................... 83

Figure 4.4 Ribbon diagram of rVP28 trimer. ............................................................... 85

ix

Figure 4.5 Western blot analysis of WSSV by anti-VP28 ........................................... 86

Figure 4.6 Expression of the rVP26 protein in E.coli BL21 DE3 star ......................... 88

Figure 4.7 Ribbon Diagram of rVP26 monomer.......................................................... 93

Figure 4.8 Ribbon Diagram of rVP26 trimer ............................................................... 95

Figure 4.9 Western blot analysis of WSSV by anti-VP26 ........................................... 96

Figure 4.10 Stereo Cα superposition of rVP28 and rVP26............................................ 98

Figure 4.11 Structure-based sequence alignment of rVP28 and rVP26......................... 99

Figure 4.12 Simulated annealing Fo-Fc omit map showing the conserved region of rVP28 ........................................................................................................ 100

Figure 4.13 Simulated annealing Fo-Fc omit map showing the conserved region of rVP26 ........................................................................................................ 101

Figure 4.14 The molecular surface representation for VP26 and VP28 along with the proposed viral envelope. ........................................................................... 103

Figure 4.15 Sequence Alignment for VP24, VP26 and VP28 ..................................... 107

Figure 4.16 Fusion Proteins of Class I and Class II ..................................................... 110

x

List of Abbreviations

CCP4 collaborative computational project No.4

CD circular dichroism

CNS crystallographic and NMR system

cryo-EM cryo-electron microscopy

DMSO dimethyl sulfoxide

DNase deoxyribonuclease

DLS dynamic light scattering

dsDNA double-stranded DNA

DTT dithiothreitol

dUTPase deoxyuridine-triphosphatase

EDTA ethylenediamine tetraacetic acid

FOM figure of merit

HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

HRP horseradish peroxidase

HSQC heteronuclear single-quantum coherence

HIC hydrophobic interaction chromatography

HIV human immunodeficiency virus

Hrs homologous regions

Ig immunoglobulins

IPTG isopropyl-β-D-thiogalactopyranoside

kbp kilo base pair

kDa kilo Dalton

MAD multiple wavelength anomalous dispersion

MALDI matrix assisted laser desorption/ionization

Mbp mega-base pair

MIR multiple isomorphous replacements

MOPS 3-(N-Morpholino)-propanesulfonic acid

MR molecular replacement

MS mass spectrometry

xi

mg milligram

NCS non-crystallographic symmetry

NMR nuclear magnetic resonance

ORF open reading frame

PAGE polyacrylamide gel electrophoresis

PCR polymerase chain reaction

PEG polyethylene glycol

PMSF phenyl-methyl-sulfonyl fluoride

Q-TOF quadrupole-TOF

RMS root mean square

RMSD root-mean-square deviation

SDS sodium dodecyl sulfate

TFA trifluoroacetic acid

TM transmembrane

TOF time-of-flight

Tris tris (hydroxymethyl) aminomethane

UV ultraviolet

WSSV white spot syndrome virus

1

CHAPTER ONE

1 Literature Review

2

1.1 Biology of White Spot Syndrome Virus

1.1.1 Overview

Viruses can only be reproduced by invading and taking over host cells due to their

lack of cellular machinery for reproduction. The term “virus”, which comes from the

Latin word virus, means poison things and usually refers to those particles that infect

eukaryotes including both multi-cellular and many single-cell organisms and prokaryotes

(phages) and archae as well. Typically, these particles carry a small amount of nuclei

acids (either DNA or RNA) surrounded by some forms of protective coat consisting of

proteins, or proteins and lipids (Knipe and Howley 2001).

The discovery that viruses are very abundant in natural waters, surpassing

bacteria by an order of magnitude, has inspired a resurgence of interest in viruses in the

aquatic environment (Wommack and Colwell 2000). The viral pathogen is a major threat,

especially for the aquaculture industry. Intensive cultivation, inadequate sanitation and

worldwide trade of livestock have greatly increased the chance of virus outbreak that

consequently causing catastrophic economic losses. Therefore, effective control and

prevention of viral disease is of upmost important to ensure the long-term operation of

aquaculture.

In the shrimp aquaculture industry, out of more than 20 strains of viruses have

been reported, among which White Spot Syndrome Virus (WSSV) is the most serious

3

pathogen (Chou et al. 1995; Lo et al. 1996; Chen et al. 1997). WSSV first appeared in the

1990s in Taiwan and has since been spread quickly to Southeast Asia, the Indian

continent and the Central- and Latin America (Xie and Yang 2005). The virulence of

WSSV is very high, resulting in a mortality rate of up to 90 to 100% within 3 to 7 days,

which can devastate the regional shrimp culture industry. A major outbreak of WSSV

infection in 1993 resulted in a 70% reduction in production of shrimps in China (Chen et

al. 1997; Cen 1998). The worldwide economic loss caused by WSSV outbreak is

catastrophic and in the range of multi-billion U.S. dollars.

The symptom of the disease caused by this virus include a red color on the entire

body and obvious white spots on the carapace, appendages and the inside surface of body.

That is why scientists named this virus as “white spot syndrome virus”. Most organs and

tissues of the shrimp, except for hepatopancreatocytes and epithelial cells of the midgut,

can be infected by WSSV. Moreover, WSSV has a broad range of hosts, including salt

and brackish water penaeid, crab, spiny lobster, freshwater shrimps and crayfishs (Figure

1.1) (Chou et al. 1995; Wang et al. 1995; Lo et al. 1996; Chen et al. 1997; Huang and

Song 1999; Chen et al. 2000; Huang et al. 2001). As such, WSSV is becoming an

epizootic disease that causes great threat not only to the shrimp aquaculture industry but

also to the marine ecology in general.

4

Figure 1.1 Clinical sign of WSSV in shrimp and crayfish

(A) WSSV infected shrimp (P. monodon) with an apparent presence of white spots on the cuticle. (B) Crayfish (Cherax quadricarinatus) one of WSSV infected hosts, was used in our laboratory for virus propagation and purification.

A

B

5

Prevention and inhibition of infection by this virus, however, is very difficult

largely due to the ability of WSSV to survive for a long period of time in the environment

(around 2 years in a shrimp pond) and also due to a poor understanding of this virus at the

molecular level (Yang et al. 2001). At present, except for the use of nonspecific

immunostimulants such as bacterial glycans (with minimal protection) and low-density

culture conditions, there is no effective treatment protocol for this disease. Therefore

novel control strategies against this virus are highly desirable (Westenberg et al. 2005).

In the following sections, some basic information of the biology on WSSV including its

morphology, genome structure and the taxonomy will be introduced in detail. These

would provide a in-depth and comprehensive understanding of the uniqueness of this

virus and the significance of scientific research on the related disease.

1.1.2 Morphology of WSSV

As mentioned before, WSSV is the major viral pathogen of shrimp with a very

high virulence. In the past decade, WSSV has attracted intensive investigation resulting

in many exciting research progress. The morphology of WSSV has been studied by

electron microscopy technology (Chou et al. 1995; Wang et al. 1995; Wongteerasupaya

et al. 1995; Durand et al. 1997; Lu et al. 1997; Leu et al. 2005). A previous electron

microscopy study showed the that intact WSSV virion is a non-occluded, enveloped

particle with an olive-to-bacilliform shape. The average size of the intact viral particle is

6

approximately 110-130 nm in diameter and 260-350 nm in length. The most prominent

feature of WSSV is the presence of a long tail-like envelope extension at one extremity

that highly resemble to a bacterial flagellum (Huang et al. 2001). The negatively stained

intact WSSV virions observed under EM are shown in Figure 1.2.

The WSSV virion is very sensitive to detergents and the envelope proteins wll be

removed from the virion when treated with 1% Triton X-100 to expose the nucleocapsid

(Figure 1.3). As shown in Figure 1.4, the naked viral nucleocapsid is about 80×350 nm

(Huang et al. 2001). Interestingly, the nucleocapsid is composed of 15 or 16 vertical

segments that are perpendicular to the long axis and about 18 to 20 nm thick. Huang and

coworkers (Huang et al. 2001) have proposed that each segment is composed of double

rows of 14 globular subunits of 8 nm in diameter (Figure 1.5). A “ring” structure could

also be seen in some of the degraded viral nucleocapsid. However, the existence of these

discrete globular subunits is debatable. In a recent paper, Leu et al (2005) have identified

that VP664, the largest viral structural protein ever found, is the major component of the

nucleocapsid. Gold labeling Immunoelectron microscopy showed that the gold particles

indicating the location of VP664 were regularly distributed around the periphery of the

nucleocapsid with a periodicity that matched the characteristic stacked ring subunits

which appear as striations. From this observation, they hypothesized that the stacked,

patterned rings of the nucleocapsid are comprised of the VP664 viral protein.

7

Figure 1.2 EM of negatively stained intact WSSV virions

A) Purified WSSV is shown with the Scale bar equals to 416 nm. B) An electron micrograph of a single WSSV virion. Note the long, tail-like structure that represents its long envelope extension. Scale bar equals to 104 nm. (Huang et al. 2001)

8

Figure 1.3 Electron micrograph of WSSV particles treated with 1% Triton X-100

This treatment leads to the distinct separation of the double layered structure of the viral envelop as shown in A (arrow →). The nucleocapsid wrapped inside the double layer envelope shows a "spindle" shape at one extremity (arrow →), while a ‘papilla’ shape structure in the other extremity (arrow →) as shown in B. In A, scale bar equals to 250 nm and in B, scale bar equals to 138 nm. (Huang et al. 2001)

9

Figure 1.4 EM of negatively stained, naked WSSV nucleocapsids

The average size of the nucleocapsid in A is 80 nm in diameter and 350 nm in length with the scale bar equals to 50 nm. Note the 15 conspicuous, vertical helix located horizontally along the long axis of the naked rod-shape nucleocapsid core in B. Each helix has two parallel striations which contain 14 global capsomers or subunit. Scale bar in B equals to 54 nm. (Huang et al. 2001)

10

Figure 1.5 Proposed WSSV structure

(Top) the overall proposed structure of WSSV virion. (Bottom) The average size of the WSSV nucleocapsid encapsidated within the double-layered envelope is 80×350 nm. There are 15 distinct and spiral helices along its long axis. Each spiral helix has two striations composed of seven pairs of globular capsomers, each of which is 8 nm in diameter. The spacing between each spiral helix is 7 nm. A ‘ring-like’ structure is associated at one terminus of the nucleocapsid core. (Huang et al. 2001)

11

1.1.3 The Genome Structure of WSSV

The complete genome sequence of three different isolates of WSSV has been

published (van Hulten et al. 2001a; Yang et al. 2001; Chen et al. 2002a) (Genebank

Accession Nos. AF332093, AF369029, AF440570 for viruses isolated from China,

Thailand and Taiwan, respectively). WSSV is the first marine invertebrate virus that has

its genome completely sequenced and it is also at present the largest animal virus whose

complete genome has been sequenced.

The complete WSSV genome is a double-stranded circular DNA of 305,107 bp,

very close to the previous estimate of 290 kb (Yang et al. 1997). Since the origin of

replication was unknown, the start of the largest BamHI fragment was chosen to be base

1. Three percent of the WSSV genome is made up of nine homologous regions (hrs),

while the remaining 97% of the sequences are unique (see description below). The

genome has a total G+C content of 41% that are uniformly distributed (Yang et al. 2001).

A total of 531 putative open reading frames (ORFs) were identified by sequence

analysis, among which 181 ORFs encoding more than 50 amino acids are likely for

functional proteins. This corresponds to an average gene density of one gene per 1.7kb.

The relative positions of the ORFs and homologous regions (hrs) in the genome are

shown in Figure 1.6. There is a potential polyadenylation site (AATAAA) downstream of

the ORFs for 80% of the 181 putative ORFs. The putative proteins encoded by 18 ORFs

show 40 to 68% identity to known proteins from other viruses or organisms and contain

12

Figure 1.6 Circular representation of the WSSV genome

Arrows, positions (outer ring) of 181 ORFs (red and blue indicate the different directions of transcription); green rectangles, 9 homologous regions (HRs). B, sites of BamHI restriction enzymes (inner ring; their positions are indicated in parentheses) (Yang et al. 1997)

WSSV

13

an identifiable functional domain. These proteins include enzymes involved in nucleic

acid metabolism and DNA replication, a collagen-like protein, and three viral structural

proteins. Proteins predicted from thirty ORFs also show partial homology (20 to 39%

identity) to known proteins or contain one or two sequence motifs (versus a real

functional domain). However, the remaining 133 ORFs encode novel proteins with no

homology to any known proteins or motifs (Yang et al. 2001).

The most unique feature of WSSV is the presence of an collagen gene, a gene

encodes an extracellular matrix protein of animal cells that has never been found in any

other viruses (Yang et al. 2001). The genome information provides us the molecular basis

of viral replication and infection.

The analysis of the genome showed that WSSV differs from all known viruses,

suggesting that it represents a novel class of viruses and perhaps implying a significant

evolutionary distance between marine and terrestrial viruses. The next section will

introduce studies on the taxonomy of WSSV during the past decades.

1.1.4 Taxonomy of WSSV

WSSV was classified in the Baculoviridae family and the subfamily of the non-

occluded baculoviruses with its rod-shaped and enveloped morphology that are similar to

insect baculovirus (Francki et al. 1991). However, more in-depth studies on the general

14

and molecular characteristics of WSSV (Inouye et al. 1994; Momoyama et al. 1994;

Wongteerasupaya et al. 1995; Lo et al. 1997) and analysis of the complete genome

sequence (van Hulten et al. 2001a; Yang et al. 2001; Chen et al. 2002a) suggest that

WSSV is not related to any virus of the Baculoviridae family or any other family of

viruses. Looking at the phylogenetic tree of the genes for WSSV DNA polymerase

(Figure 1.7), ribonucleotide reductase large (rr1) and small (rr2) subunits, protein kinase,

thymidine-thymidylate kinase and endonuclease, no obvious relationship was revealed

between WSSV and other established family of large dsDNA virus (Chen et al. 2002b).

Based on the uniqueness of WSSV, many scientists have suggested that it should

be excluded from the Baculovirus family and be classified as a new virus family.

Recently, the International Committee on Taxonomy of Viruses approved a proposal to

elect WSSV as the type species of the genus Whispovirus, family Nimaviridae

(www.ncbi.nlm.nih.gov/ICTVdb/Ictv/index.htm). The family name reflects the most

notable physical feature of the virus, the tail-like projection extending from one end of

the WSSV virion. The uniqueness of WSSV has attracted extensive research into its

evolution and infection mechanism. In the protein level, most researches are focused on

the envelope proteins that play important roles in the infection and maturation

mechanisms of the virus.

15

Figure 1.7 Bootstrap analysis (100 replicates) of an uprooted phylogenetic tree of the

DNA polymerase from WSSV.

The numbers at the branches indicate frequency of the clusters and frequencies over 70% are indicated by thick lines.

(http://www.danforthcenter.org/iltab/ictvnet/images/paris/Nimaviridae.pdf)

16

1.1.5 Envelope Proteins of WSSV

In general, viral proteins are divided into three temporal classes: early proteins

synthesized prior to the replication of the DNA, the intermediate and late proteins

synthesized after the onset of DNA replication (Jensen et al. 1996). The late proteins that

synthesized from 5 to 6 hours after infection are assumed to be virion-associated proteins.

In characterizing any virus, its virion-associated proteins are particularly important for

understanding the mechanisms of viral infection and morphogenesis.

A virion is a complex assembly of macromolecules exquisitely suited for the

protection and delivery of viral genomes (Tsai et al. 2006). As a double-layer enveloped

virus, virion-associated proteins of WSSV include proteins of both the envelope and

nucleocapsid. Thirty nine virion-associated proteins have been identified due to the gel

based mass spectrometry techniques (Huang et al. 2002a; Huang et al. 2002b; Li et al.

2004; Tsai et al. 2004). Shotgun proteomic studies have identified fifty five virion-

associated proteins in our laboratory recently (Li et al, unpublished data).

Among these virion-associated proteins, the envelope proteins are of extremely

importance because they are believed to be the first molecules to interact with the host

and hence play critical roles in targeting the host cell and triggering host defenses.

Determining the localization of structural proteins in the virion is important to elucidate

their roles in both virus assembly and infection. Up to date, four major envelope proteins

(VP19, VP24, VP26 and VP28) and several minor envelope proteins (VP31, VP39,

17

VP124, VP187, VP281, VP292, VP466) were detected by Western blotting technique

and localization study under electron microscope (Huang et al. 2002a; Huang et al. 2002b;

Zhang et al. 2002a; Zhang et al. 2002b; Zhang et al. 2004; Li et al. 2005b; Li et al. 2006;

Xie et al. 2006; Zhu et al. 2006).

There are different classes of membranes proteins: Integral proteins interact with

hydrophobic parts of the bilayer’s phospholipids, and they are not easy to remove. Most

span the bilayer, with their hydrophilic domains extending past both of its surfaces.

Peripheral proteins are positioned at the membrane surface, not in the bilayer. Weak

interactions, including hydrogen bonds, allow them to associate with integral proteins and

with polar heads of the membrane lipids. For major envelope proteins of WSSV such as

VP26 and VP28, they both have a hydrophobic transmembrane domain that serves to

anchor them to the surface of the lipid bilayer.

Using SDS-PAGE profiles of envelope fractions from WSSV, VP19, VP24, VP26

and VP28 have been shown to constitute the major part of the entire envelope.

Interestingly, there are striking similarities between VP24, VP26 and VP28 at the amino

acid and the nucleotide sequence level. VP26 and VP28 share a sequence similarity of

41%, whereas VP24 shares significant sequence similarities of 41% and 46% with VP26

and VP28 respectively. These three proteins have the same size (~ 206 aa) and all are

predicted to have a transmembrane region of approximately 30 amino acid in length at

the N-terminus. This strongly suggests that the genes of these three envelope protein may

have evloved by gene duplication. Another major envelope protein, VP19, has no

18

sequence homology with other WSSV or other known viral proteins. The transmembrane

region of VP19 is predicted to be situated at the middle of the protein sequence.

As far as functional study is concerned, VP28, VP68, VP281, and VP466 were

believed to play an important role in the systemic WSSV infection in shrimp by using an

in vivo neutralization assay (van Hulten et al. 2001b; Li et al. 2005a; Xu et al. 2005).

VP28 is often used for detecting the WSSV in crustaceans because of its abundance in

the proteins profile (Yoganandhan et al. 2004). VP26 is reported to be capable of binding

to actin or actin-associated proteins (Xie and Yang 2005). Another envelope protein,

VP281, which contains a cell attachment RGD motif, is also supposed to play an

important role in mediating infection by WSSV (Huang et al. 2002a; Liang et al. 2005).

19

1.1.6 Problems Faced in Study of Envelope Proteins of WSSV

Envelope proteins have been investigated extensively because of their importance

in viral infection and this project will also focus on the characterization of these envelope

proteins. There are, however, many well-known technical difficulties that hinder in-depth

studies to elucidate the function of the envelope proteins from WSSV. The lack of

established shrimp cell lines for in vitro reproduction of WSSV is one of the major

obstacles. The function of these envelope proteins are also hard to predict because of

their lack of homology with other known proteins. Therefore, we will utilize structural

genomics approaches to elucidate the structure of the WSSV proteins to understand their

function towards the drug design.

Among the 55 structural proteins identified by mass spectrometry, 11 and 3 of

them have been assigned as the envelope and the nucleocapsid proteins respectively. The

localizations of the other 40 structural proteins are still unknown. At present, only VP28

has been confirmed to be involved in the attachment and penetration of the WSSV into

shrimp cells (Yi et al. 2004). The roles of other envelope proteins in the life cycle of the

virus remain to be defined. Moreover, up to now, tertiary structures are not available for

any WSSV proteins. This project will therefore focus on the characterization and

elucidation of the three dimensional structure of the envelope proteins of WSSV.

20

1.2 Structural Genomics

1.2.1 Structural Genomics as a New Research Initiative

New research initiatives in biology are being driven by complete sequencing of

the genomes from various organisms (Green 2001; Lander et al. 2001; Aparicio et al.

2002; Yu et al. 2002), which gave us the protein repertoires of diverse organisms from all

kingdoms (Marsden et al. 2006). The genome-sequence projects have accumulated lots of

sequence data providing us nearly complete lists of macromolecules present in an

organism. However, the knowledge of the components relatively little about the function

and organization of biological systems (Bravo and Aloy 2006). The challenge to

understand these gene products has led to the development of functional genomics

approaches, which collectively aim to improve the biological understanding of the

genetic sequence. Structural genomics is one of such approaches, with an unique promise

to reveal the molecular function of protein domains (Ashburner et al. 2000). A good

understanding of molecular functions & interactions can come from high-resolution

three-dimensional (3D) structures, as they provide key atomic details on the binding

surfaces. Structural genomics projects, together with computational structure prediction

methods (Bradley et al. 2005), are likely to provide 3D structures for most of the proteins

within an organism.

Worldwide structural genomics initiatives, advanced by the development of

improved technologies in X-ray crystallography and NMR, are expanding our knowledge

on structural families with increasing number of novel protein folds. Methods for

21

detecting remote sequence similarities have also been improved in sensitivity and enabled

us to map domains from these structural families onto genome sequences directly to

understand how these families are distributed throughout the genomes. This in turn reveal

how these proteins might influence the functional repertoires and biological complexities

of the organisms (Marsden et al. 2006).

1.2.2 WSSV as a Model for Structural Genomics Study

WSSV provides an important and interesting model for structural genomics study.

It has a relatively small genome with only about 181 open reading frames, much smaller

than herpes virus and other viruses with large genomes. Many of the WSSV putative

ORFs do not have match with any known proteins and functions. Structural genomics

therefore offers an opportunity to elucidate the function of these proteins through

structure determination and prediction. Structure elucidation of WSSV will provide

insights on the mechanisms of host-pathogen interaction, the maturation and assembly of

the virus particles and many other important biological processes. More importantly, it

will provide a wealth of information for structure based drug design.

22

1.3 X-ray Crystallography

1.3.1 Overview

It is necessary to know the precise arrangement of atoms in biological

macromolecules in order to understand how they function at the molecular level. In

addition, this information is required for the rational design of drugs that target a specific

activity of the molecule or prevent the molecule from binding to other molecules with

which it normally interacts (Yaffe 2005). X-ray crystallography is one of the most

powerful techniques that are used to obtain atomic information about molecular structure.

In brief, structure determination of a protein by X-ray crystallography requires the

growth of high-quality crystals from the purified protein, measurement of the directions

and intensities of X-ray beams diffracted from the crystals, and the use of a computer to

calculate the electron density map after the phase problem is being solved. Finally, the

protein backbone is fitted into the electron density map and refined to give the structure

coordinates (Rhodes 2000).

In Figure 1.8, the synopsis of structure determination by X-ray crystallography is

clearly shown. The macromolecule of interest is crystallized, and a single crystal that

contains thousands of copies of the macromolecule is placed in an X-ray beam. The

crystal is slowly rotated in the X-ray beam and diffraction patterns similar to the one

shown are collected for every 0.5–1 degree of rotation until almost all points in the

23

Figure 1.8 Synopsis of structure determination by X-ray crystallography

The macromolecule of interest is crystallized, and a single crystal containing thousands of copies of the macromolecule is placed into an X-ray beam. The crystal is slowly rotated in the X-ray beam and diffraction images similar to the one shown are collected every 0.5–1 degree of rotation until all points in the crystal have been sampled. The intensity of the diffracted X-ray, together with information about their phases, is collected and converted by a Fourier transforming to an electron density map of the protein. A model of the protein structure is then built into the electron density map (Yaffe, 2005).

24

reciprocal space have been sampled. Structure determination of a molecule requires

knowing both the amplitude and the phase of the photon wave being diffracted from the

crystalline sample. Although a detector can measure the intensity of a wave, which is

proportional to its amplitude, there is no way to measure its phase. The phase angel is the

difference of the angles between the combined X-ray scattered by all atoms of this

molecule and the original X-ray. Thus, it is common to hear that one has "lost the phase"

when measuring a diffraction pattern. Reconstructing the phase of the diffracted wave is

the crux of the phase problem. The intensity of the diffracted X-ray, together with

information about their phases, is converted by Fourier transformation into an electron

density map of the protein. A model of the protein structure is then built into the electron

density map (Yaffe 2005).

1.3.2 Crystallization

X-ray crystallography is totally dependent on highly ordered crystals. Obtaining

such crystals is the rate-limiting step in structure determination and the pressure is to

produce crystals. As a result, the science of crystallization is gathering a new momentum

and is becoming a rapidly developing field (Chayen 2004).

Crystallization is a phase transition phenomenon. Crystals grow from an aqueous

protein solution when the solution is brought into supersaturation (Ataka 1993; Ducruix

and Giege 1999; McPherson 1999). Supersaturation is achieved by varying the

25

concentration of precipitant, protein and additives, pH, temperature and other parameters.

The task to produce suitable crystals can be tackled using two different approaches. The

first relies on empirical techniques that is based mainly on trial and error, and what is

perceived to be the “art” of crystallization. The second approach is aimed at gaining an

understanding of the fundamental principles that govern crystallization; this knowledge

may be applied to design experimental methodology for producing high-quality crystals

of medical and industrial interest (Chayen 2004).

The past few years have seen some of the greatest achievements in the field of

protein crystallization e.g. automation and miniaturization of crystallization trials (Kuhn

et al. 2002; Walter et al. 2005). The high-throughput approach has reduced the time

needed to set up a series of experiments. The subsequent phases of different pattern

capture and analysis of the data is also progressing at high speed (Luft et al. 2003).

However, the high-throughput has not led to high output. Production of suitable crystals

still remains as a rate-limiting step. It is still not understood why some proteins crystallize

easily while others stubbornly refuse to produce suitable crystals. Obviously there is an

urgent need to solve the problem by a more systematic & scientific approach (Chayen

2004).

The success rate to obtain high-quality crystals is improving rapidly and it will

improve further as we progress to the more sophisticated techniques, which will play a

major role in crystallization and structural biology.

26

1.3.3 X-ray Diffraction

X-ray is an electromagnetic radiation with wavelengths in the range from 0.1 to

1000 Å. X-ray rays are generated when electrons (or other charged particles) undergo

severe acceleration (or deceleration). There are two types of X-ray sources used for

macromolecular crystallography, those from rotating anode and the synchrotron radiation.

The rotating anode type X-ray generator is the laboratory scale sources. An anode

of appropriate target metal (copper, molybdenum etc.), a cathode and a tungsten filament

are kept inside an evacuated tube. When a high voltage difference is maintained between

the anode and cathode, electrons, produced by the filament by thermionic emission, hit

the anode and X-rays are produced. Depending on the target and transition, the radiation

is named as Cu Kα, Cu Kβ etc. The anode is rotated by a motor for efficient distribution

and dissipation of heat and a high intensity beam is produced. The X-rays produced at the

impact point are distributed over a wide angular range and cover a wide range of energies

up to a maximum energy determined by the voltage at which the tube is operated. In a

synchrotron, electrons travel in a storage ring at a high speed and are emitted as radiation

and it is possible to change the wavelength of synchrotron radiation. Here bunches of

very high energy electrons (several GeV) move in a closed loop. At each bending point

an intense and very narrowly defined pencil beam of radiation is emitted. The beam

contains radiation of a wide range of energies - UV through to X-rays. Synchrotrons are

very large central facilities that have their own operating procedures and safety systems

that visitors and users must follow.

27

1.3.4 Structure Determination

To determine the structure of a protein means that position of all atoms of the

protein in the unitcell represented by the electron density map are known. There is a

direct relationship between the electron density map of the unitcell and the structure

factors of the diffraction pattern as related by the Fourier transformation. However, in

order to calculate the electron density map of the unitcell, one needs the experimental

structure amplitudes and their phase angles.

As the phase angle is an immeasurable quantity, the information can only be

derived by one of the following methods. In the molecular replacement (MR) method, an

available similar structure is used as a model to probe the experimental data to identify

the structure of interest. Direct methods are based on the positivity and atomicity of

electron density that leads to phase relationships between structure factors. The phase

angle is obtained by mathematical means. In the multiple isomorphous replacement (MIR)

method, the data set for a native crystal and data sets for two derivative crystals, obtained

by soaking the native crystal in two different heavy metal containing solutions, are

collected. In the multiwavelength anomalous dispersion method (MAD), a crystal

containing an anomalously diffracting atom is used for data collection at three different

wavelengths using a synchrotron. Positions of these anomalous atoms are first

determined and the electron density map is made. SAD (single-wavelength anomalous

diffraction) method is becoming increasingly possible to collect data at just a single

28

wavelength, typically at the absorption peak, and use density-modification protocols to

break the phase ambiguity and provide interpretable maps.

The electron density map is calculated after the phase problem is being solved.

The protein model is refined until the difference between the experimental and calculated

(using the model) structure factors (R-factor) reaches at an acceptable minimum. The

most used refinement program is “CNS” (Crystallographic and NMR System). CNS

adopts the algorithm of energy minimization for the refinement

1.4 Objectives of the Project

The characterization of viral structural proteins especially the envelope proteins is

of significant importance to study the virus taxonomy, infection assembly. Due to the

lack of a shrimp cell line for in vitro reproduction of this virus and low homology with

other proteins of known function, we have decided to take a structural proteomics

approach to study the envelope proteins of WSSV. The objectives of this project are:

1) To identify more envelope proteins of WSSV. Among the 55 structural proteins

identified by mass spectrometry, only 11 have been assigned as envelope proteins and 3

as nucleocapsid proteins. There are 40 structural proteins unassigned. We will use

western blotting and immuno-gold-labeling electron microscopy to study the localization

of these envelope proteins. Neutralization assay that can determine whether the envelope

proteins are involved in the virus entry during infection will also be performed.

29

2) To elucidate the three dimensional structure of the viral envelope proteins. The

study attempts to determine the structures of these envelope proteins to understand their

function towards the drug design.

1.5 Significance of the Project

Once the 3D structures of major envelope proteins are solved, their structural

features should provide us an unique opportunity to postulate the function of these

envelope proteins. Also, the structure of these envelope proteins and their interactions

explain the unique morphological features of WSSV. The structural elucidation of WSSV

envelope proteins will provide us much useful information for further research on host-

virus interaction, the maturation and assembly of the virus particles and many other

important questions. It will also help us to better understand the infection mechanism and

life cycle of WSSV. Finally, it will provide a wealth of information for structure based

drug design that can be used to develop specific inhibitors to control or even prevent the

outbreak of WSSV.

1.6 Scope of the Thesis

X-ray crystallography techniques will be used to determine the structures of the

envelope proteins. The major envelope proteins like VP19, VP24, VP26 and VP28 are the

primary candidates for crystallography and functional study.

30

CHAPTER TWO

2 Materials and Methods

31

2.1 Bacterial Strains, Vectors, Primers and Bacterial Culture

Bacterial Strain DH5α (Invitrogen) was used in this study as host for cloning

experiments and BL21 Star (DE3) strain (Invitrogen) for protein expression. Vector

pGEX6p-1 (Amersham Bioscience) was used for GST fusion protein expression and

pET32a(+) (Novagen) for His-tag protein expression. Escherichia coli starins were

grown at 37 °C in LB (Luria broth) liquid media (10g Tryptone, 5g yeast extract, 10g

NaCl, pH7.5) or 1.5 % agar medium (Sambrook and Russell 2001). For long-term storage,

all bacterial strains were stored in LB with 50% glycerol at -80 °C freezer.

The sequences of some primers used in this study are shown as following:

N-terminus truncated VP28 (residues 31-204)

CGC GGA TCC AAC ACT GTG ACC AGG ACC ATC GAA

CCG GAA TTC TTA CTC GGT CTC AGT GCC AGA

N-terminus truncated VP26 (residues 35-204)

CGC GGA TCC ATG AAC ACA CGT GTT GGA AGA

CCG GAA TTC TTA CTT CTT CTT GAT TTC GTC CTT

Full-length VP300

CGC GGA TCC ATG GGA GAT AAG CAA AAG GTG GAA

CGC GAA TTC TTA GGA GCA TGT GCA TGT GAT CCT

Full-length VP281

CGC GGA TCC ATG GCG GTA AAC TTG GAT AAT

CGC GAA TTC TTA TGT CCA ACA ATT TAA AAA GAA

32

2.2 DNA Manipulation

DNA manipulations were carried out following standard molecular protocols

cited from Molecular Cloning (Sambrook and Russell 2001) with some modifications.

2.2.1 Amplification of DNA by Polymerase Chain Reaction (PCR)

DNA fragments and related genes were amplified by the basic polymerase chain

reaction (PCR). High fidelity Pfu DNA polymerase (Promega) was used to amplify the

genes from WSSV genomic DNA. The composition of PCR reaction mixture was shown

as following: 5 µl 10 × PCR buffer (without MgCl2), 5 µl MgCl2 (25 mM), 1 µl 5’ Primer,

1 µl 3’ Primer, 1 µ dNTPs (25 mM) l, 1 µl Template DNA (20-100 ng/ µl), 1 µl Tag DNA

polymerase (1 unit/ µl), and distilled water to a final volume of 50 µl.

The PCR was run on an iCycler Thermal Cycler (Bio-rad) with the following

program. 1 cycle of 95 °C for 1 ~ 2 minutes, 30 cycles of each at 95 °C for 30 seconds,

50 °C for 30 seconds and 72 °C for 1 minute per 1kb DNA, and 1 cycle of 72 °C for 15

minutes.

2.2.2 DNA Digestion and Ligation

DNA digestion and ligation were conducted according to the instruction manual

by the supplier. The DNA digestion mixture comprised of 10 µl DNA, 1 µl restriction

enzymes, 2 µl 10×BSA, 2 µl reaction buffer and 4 µl sterile H2O. The mixture was

33

incubated at 37 °C for 2 hours. For single-digestion vectors, dephosphorylation is

required for the subsequent ligation. In this case, the vector mixture was treated

additionally with alkaline phophatase (Promega) at 37 °C for 1 hour, followed by

inactivation of the alkaline phophatase at 75 °C for 10 minutes. The digested DNA

products were further purified using agarose gel electrophoresis and QIAquick Gel

Extraction Kit (QIAGEN). Bacteriophage T4 DNA ligase was used to ligate the digested

DNA fragments to the digested vectors. The ligation was performed at room temperature

for 3 ~ 4 hours or at 4 °C for overnight.

2.2.3 Agarose Gel Electrophoresis and DNA Purification

DNA fragments were separated by agarose gel electrophoresis in TAE buffer

(0.04M Tris-acetate, 0.001 M EDTA, pH 8.0) along with a standard DNA ladder

(Fermentas). Normally, the concentration of agarose gel is 1% and ethidium bromide (EB)

was directly added to the agarose gel to a final concentration of 0.5 µg/ml. The separated

DNA fragments were recovered from the agarose gel by using the QIAquick Gel

Extraction Kit (QIAGEN) according to the manufacturer’s instructions.

2.2.4 Preparation of E.coli Competent Cells

Highly efficient competent cells of E. coli DH5α and BL21 (DE3) strains were

prepared by the rubidium chloride method. Freshly growing E. coli colonies from LB

34

agar plates were inoculated into 100 ml of Psi broth medium (5g yeast extract, 20g

tryptone, 5g magnesium sulfate, pH7.6) in a 1-liter conical flask and then allowed to

grow at 37 °C with vigorous shaking (200 rpm). After the cells grew to an OD550 of

0.45~0.5, the culture was chilled on ice for 15 min and then the cells were collected by

centrifugation at 5,000 rpm (Beckman J2-21 ) for 5 min at 4 °C. The cell pellet was

resuspended in 40 ml of ice-cold TfbI buffer (30 mM potassium acetate, 100 mM

rubidium chloride, 10 mM CaCl2, 50 mM MgCl2, 15% v/v glycerol, pH 5.8) and

incubated on ice for 15 min. After centrifugation, the cell pellet was resuspended by 4 ml

TfbII buffer (10 mM MOPS, 10 mM rubidium chloride, 75 mM CaCl2, 15% v/v glycerol,

pH 6.5). The competent cells can be either used immediately or quickly frozen as 0.25 ~

0.5 ml aliquots in liquid nitrogen prior to storage in a -80 °C freezer.

(http://130.15.90.245/e__coli_competent_cells.htm)

2.2.5 Transformation of Bacterial Cells

In most operations, DNA was transferred into E. coli cells by heat-shock

following the standard protocol (Sambrook and Russell 2001). Frozen competent cells

(50 µl) were thawed on ice. The transforming DNA (up to 25 ng per 50 µl of competent

cells), including plasmid or ligation product, was added to the competent cell suspension

in a volume not exceeding 5 % of that of the competent cells. The competent cells were

mixed with the transforming DNA by swirling gently and then incubated on ice for 30

minutes. The mixture of cells and DNA were heat-shocked at 42 °C for 1 minute and

immediately chilled on ice for 2 min. Eight hundred microliters of fresh LB medium were

35

added to the transformed cells. The transformed cells were incubated at 37 °C for 45 min

with shaking to allow the bacteria to recover and to express the antibiotic resistance

marker encoded by the plasmid vector. The cells were collected by centrifugation and

spread onto LB agar plates containing the appropriate antibiotic. Colonies usually

appeared after overnight incubation at 37 °C.

2.2.6 DNA Sequencing

The sequencing PCR run mixture was 10 µl and contained: 2 µl BigDyeTM Ready

Mix (v 3.0), 0.5 µl primer, 2 µl template DNA (purified plasmids or PCR products,

100~200 ng µl-1) and 5.5 µl deionized water. The sequencing cycle was normally

performed using the following program: 1 cycle at 96 °C for 0.5 ~ 1 min and then 25

cycles each at 96 °C for 15 seconds, annealing at 50 °C for 15 seconds and extension at

60 °C for 4 min. The sequence product was kept at 4 °C until ready to purify. After the

cycle sequencing was finished, the contents in the PCR tube were spun down and then

transferred into a clear 1.5-ml tube. 80 µl of acetate-ethanol mixture (3 µl of 3M sodium

acetate, 62.5 µl 95% ethanol and 14.5 µl water) was added to the tube. The contents in

the tube were mixed by vortexing and the tube was allowed to stand at room temperature

for 15 minutes to precipitate the extension products. The pellet was collected by

centrifugation at 14,000 rpm (Eppendorf 5417C) for 20 min and then washed twice with

70% ethanol before centrifugation at 14,000 rpm (Eppendorf 5417C) for 10 min. After

the supernatant was carefully aspirated from the tube, the pellet was dried and stored at –

20 °C. This pellet is ready to be submitted for running electrophoresis.

36

2.3 Virus Treatment and Analysis

2.3.1 Virus Propagation and Purification

The WSSV isolate used in this study was originated from WSSV infected

Penaeus chinensis (Ningbo, China) and was propagated in an alternate host, red claw

crayfish, Cherax quadricarinatus. Preparation of WSSV virions was carried out based on

the method described previously (Huang et al. 2001) with some modifications. The

haemolymph collected from infected crayfish were diluted in PBS buffer and stored in -

80 oC as the stock. One tube of the stock haemolymph was thawed and after

centrifugation at 1000 × g for 10 minutes, the supernatant was filtered (0.45 µm filter)

and diluted 10 times in PBS buffer as the virus inoculum. Then we injected 0.3-0.4ml of

the virus inoculant was injected intramuscularly into the healthy red claw crayfish in the

lateral area of the fourth abdominal segment. After four to five days later, hemolymph

freshly extracted from moribund crayfish was first centrifuged at 2,000 × g to get rid of

the tissue debris. The supernatant was layered on the top of a 30 – 60% (w/v) stepwise

sucrose gradient and centrifuged at 20,000 rpm using a SW28 rotor in a Beckman

ultracentrifuge (XL-90; Beckman Coulter) for 2 hours at 4°C. Virus bands (Figure 2.1)

were collected and then diluted two or three times with TN buffer (20mM Tris-HCl and

400mM NaCl, pH7.4) before centrifugation again at 20,000 rpm using a SW28 rotor for 1

hour. The pellets were washed with TN buffer followed by a centrifuge at 12,000 × g for

30 minutes at 4 °C. The wash step was repeated to remove all the sucrose. The resulting

pellets were resuspended in TN buffer and the intact virion samples were stored at -80 oC.

37

Figure 2.1 Banding of purified WSSV Virus in the sucrose gradient

The stepwise sucrose gradient used was 30%, 40%, 50%, 60% (w/v) from the top. The

virus band showed in the interface between 40% and 50% gradient after

ultracentrifugation at 20,000rpm for 1 hour.

WSSV

38

2.3.2 Treatment of the Intact Virus with Detergent

The virus envelope was separated from the virus particles by treatment with Triton

X-100. Generally, the purified WSSV was mixed with an equal volume of 0.2% Triton

X-100 and incubated for 1 h at room temperature. The nucleocapsids were purified by

centrifugation at 20,000 × g for 20 min at 4 °C. The envelope fraction was collected in

the supernatant. The nucleocapsid fraction was subjected to a second round of Triton X-

100 extraction to ensure the envelopes were removed completely. The degree of purity of

the intact virions isolated and the nucleocapsid fractions were evaluated by negative-

staining transmission electron microscopy.

2.3.3 Preparation of WSSV Genomic DNA

WSSV genomic DNA from gradient-purified virions was extracted by protease K

and cetyltrimethylammonium bromide (CTAB) treatments, followed by phenol-

chloroform extraction and ethanol precipitation (Lo et al. 1996). Briefly, protease K and

N-laurysarcosine were added to the purified viral particles suspended in TNE buffer to a

final concentration of 0.5 mg/ml and 0.5%, respectively. After 1 hour incubation at 65 °C,

the solution was then treated with 1% CTAB for 15 minutes at 65 °C. This was followed

by successive extractions with an equal volume of phenol once, an equal volume of

phenol:chloroform:isoamyl alcohol (25:24:1) for 3 ~ 4 times, and then an equal volume

of chloroform:isoamyl alcohol once. Viral DNA was recovered by ethanol precipitation,

dried and dissolved in a small volume of 0.1 × TE buffer (50 mM Tris, 1 mM EDTA,

39

pH 8.0) containing RNAse A (10ug/ml) before quantified by spectrophotometry with the

Shimadzu spectrophotometer (Model UV-300).

2.3.4 Transmission Electron Microscopy

For transmission electron microscopy (TEM) examination, virus samples were

deposited onto formvar-coated, carbon-stabilized copper grids (200-mesh, Electron

Microscopy Sciences) before they were negatively stained with 2% (w/v) sodium

phosphotungastate (PTA, pH 7.0). The grids were dried and examined under electron

microscope (JEOL JEM 2010F or PHILIPS CM 10).

2.3.5 Localization Study by Immunoelectron Microscopy

WSSV intact virion and nuclocapsid suspension were mounted on carbon-coated

nickel grids (300 meshs, Electron Microscopy Sciences) and kept for 5 minutes before

blocking in 5% BSA (in 20 mM phosphate buffer) for 30 minutes at room temperature.

The first antibody (diluted with 5% BSA buffer, concentration of 40 mg/ml) was added

and kept at 4°C overnight. After washing at least three times for 10 minutes, the gold

labeled second antibody (goat-anti-mouse/rabbit IgG, 1:40 dilution, Electron Microscopy

Sciences) was added and incubated for 2 ~3 hour at room temperature. The grids were

washed by 5% BSA buffer twice and followed by water once. Negative stain by 2% PTA

was performed for 2 ~3 minutes and the grids were dried before examining under TEM.

40

2.4 Protein Manipulation

2.4.1 Protein Expression and Solubility Test

The constructs encoding WSSV envelope proteins were introduced into E. coli.

The bacterial cells were grown to a concentration of approximately OD600 = 0.5 ~ 0.6 at

37 °C. IPTG was added to the cultures in a final concentration of 0.1 mM to induce the

expression of fusion proteins. The bacterial cultures were allowed to grow for additional

5 ~6 hours at 25 °C or overnight at 18°C. All the subsequent operations were performed

at 4 °C. The cells from 500 ml of culture were harvested by centrifugation and

resuspended in 25 ml lysis buffer (20 mM Tris, 500 mM NaCl; 10 mM EDTA, 2mM

DTT, 100 µM PMSF, 5% Glycerol, pH 8.0). After treatment with chicken egg lysozyme

(final concentration of 1 mg/ml) for 30 minutes, the cell suspension was sonicated to near

clarity and centrifuged at 20,000 rpm for 1 hour using a J6-HC centrifuge (Beckman

Coulter). The supernatant and pellets sample were loaded into SDS-PAGE to determine

the solubility of the fusion proteins expressed.

2.4.2 Expression of Seleno-Methionine Substituted Protein

The selenomethionine substituted protein was expressed in defined LeMaster

medium using the methionine auxotrophic strain (DL41). The LeMaster medium amino

acid mixture for 1L medium were prepared as following: 0.5 g Alanin, 0.58 g

Arginine.HCL, 0.4 g Aspartic acid, 0.03 g Cystine, 0.67 g Glutamic acid, 0.33 g

41

Glutamine, 0.54 g Glycine, 0.06 g Histidine, 0.23 g Isoleucine, 0.23 g Leucine, 0.42 g

Lysine.HCL, 0.13 g Phenylalanine, 0.1 g Proline, 2.08 g Serine, 0.23 g Valine, 0.5 g

Adenine, 0.67 g Adenine, 0.67 g Guanosine, 0.17 g Thymine, 0.5 g Uracil, 1.5 g Sodium

acetate, 1.5 g Succinic acid, 0.75 g Ammonium chloride, 1.08 g Sodium hydroxide, 10.5

g Dibasic K2HPO4. This autoclaved mixture should be stored dry at -20°C.

Dissolve 24.1 g of the amino acid mixture to 1 Liter of deionized water and

autoclave for 20 minutes. Dissolve 10 g D-glucose, 0.25 g MgSO4.7H2O and 4.18 mg

FeSO4.7H2O in 90ml deionized water. 8.3 µl of concentrated H2SO4 and 10 ml Kao and

Michayluk vitamen solution (Sigma) were added and filter sterilize. All these are added

into the above autoclaved medium. Add 25 mg L-selenomethionine (Sigma) and 1 ml 0.1

g/ml amipicillin immdediately prior to inoculation. Inoculate the methionine auxotrophic

strain (DL41) containing the plasmid of interest in defined LeMaster medium and grow at

37 °C until the reading of OD600 is above 0.6. IPTG was then added into the culture

medium for the protein over expression.

2.4.3 GST fusion Protein Purification and Removal of GST tag

All the subsequent operations were performed at 4 °C. The soluble fusion proteins

were present in the supernatant of cell lysate from above steps. The supernatant was

filtered through 0.45 µm filter (Millpore) and loaded onto a Glutathione Sepharose 4B

column (Amersham Biosciences), followed by washing with at least 10 column volume

of lysis buffer. The protein was eluted with 20 mM reduced glutathione freshly prepared

42

in lysis buffer. The concentration of eluted GST fusion protein was determined with

Coomassie Protein Assay kit (Pierce) according to the manufacturer’s instructions.

ProScission™ protease (Amersham Biosciences) was added to the eluted protein solution

and kept at 4°C overnight for reaction. After digestion, the solution was loaded onto a

HiPrep Desalting column (Amersham Biosciences) with the desalting buffer (20 mM Tris,

100 mM NaCl, 2 mM DTT, pH8.0) to get rid of the reduced glutathione. After desalting,

the protein solution was passed through the Glutathione Sepharose 4B column to remove

the GST cleaved away from the fusion protein.

2.4.4 Purification of Untagged Protein

After removal of GST, the solution of untagged protein was further purified by

Superdex 75 size exclusion chromatography (Amersham Biosciences) using gel filtration

buffer. The purified protein fractions were collected and concentrated to a final

concentration of 8 mg/ml with a VIVASPIN 20 (Vivascience) concentrator. The purity of

the protein was examined by SDS-PAGE as well as by native gel electrophoresis.

2.4.5 Antibody Preparation

To obtain the polyclonal anti-rabbit antibody, New Zealand wild type female

rabbits were used in this study. A small amount (about 5 ml) of preimmune blood was

drawn from each rabbit prior to any injections as negative controls. 0.5 ml of purified

43

protein (0.1 mg) was mixed and emulsified with an equal volume of complete Freund’s

Adjuvant. The freshly prepared emulsion was immediately injected into one limb of the

rabbit. The first boost was done four weeks later on another limb. The dosage for each

booster was 50 µg, which was emulsified in 0.5 ml of Incomplete Freund’s Adjuvant

(IFA) emulsion and injected into another limb. After the first booster, the rabbit was

injected every week and the limbs were used in rotation during the subsequent boost

injections.

To obtain the polyclonal anti-mouse antibody, the purified protein was used to

immunize 3- to 4- week-old Swiss Albino mice once every 2 weeks by intradermal

injection over an 8-week period. Antigen (20 µg) was mixed with an equal volume of

Freund’s complete adjuvant (Sigma) for the first injection. Subsequent injections were

conducted using 20 µg of protein mixed with an equal volume of Freund’s incomplete

adjuvant (Sigma).

The blood collected from rabbit or mouse was allowed to clot at room

temperature for overnight. The antiserum was collected from the clot by centrifugation at

1,500 × g for 10 minutes at room temperature. Titers of the antiserum was determined by

ELISA (Harlow and Lane 1988). Protein A Sepharose CL-4B was used to isolate anti-

mouse antibody according to the manufacturer’s instructions (Amersham Biosciences).

44

2.5 Protein Analytical Techniques

2.5.1 SDS-PAGE Gel Electrophoresis

Proteins were separated by SDS-PAGE gel electrophoresis according to their

molecular weight. The electrophoresis apparatus used in SDS-PAGE gel electrophoresis

was the Mini-Protean III Electrophoresis Cell (BioRad). The apparatus was assembled

according to the instruction manual provided by the manufacturer. For the routine

analysis of proteins, 12% PAGE gel was used. The polyacrylamide gels (PAGE) for the

separating gels were prepared as following:

Gel concentration 7% 10% 12% 15% 17.5%

buffer (1.5M Tris, pH8.8) 1.9 ml 1.9ml 1.9ml 1.9ml 1.9ml

30% Acrylamide/bis-acrylamide (29:1) 3.5 ml 5 ml 6 ml 7.5 ml 8.75 ml

10% (w/v) SDS 150 µl 150 µl 150 µl 150 µl 150 µl

10% (W/V) APS 150 µl 150 µl 150 µl 120 µl 100 µl

TEMED 9 µl 9 µl 9 µl 8 µl 6 µl

H2O 9.21ml 7.71ml 6.71ml 5.24ml 4.01ml

Total volume 15 ml 15 ml 15 ml 15 ml 15 ml

Stacking gel was 2.5% and prepared by mixing 2 ml stacking gel buffer (0.5M

Tris, pH6.8), 1.3ml 30% acrylamide/bis-acrylamide, 80 µl 10% SDS, 80 µl 10% APS, 8

µl TEMED and 4.5 ml H2O.

45

Before loading, proteins sample was mixed with 2 × loading buffer (100mM Tris

pH6.8, 200mM DTT, 4% SDS, 0.2% bromophenol blue, 20% Glycerol) and boiled for 3

~ 5 minutes to fully denature the protein and reduce disulfide bonds. The gel was run at a

constant voltage of 100 V until the bromophenol blue reached the bottom of the gel. After

SDS-PAGE, the gel was stained in Coomassie staining solution (45% methanol, 10%

acetic acid and 0.25% coomassie brilliant blue R-250) for 2 ~4 hours and then destained

with destain solution containing 5% methanol and 7.5% acetic acid. The destained gel

was washed in water to remove the organic solvents before drying.

2.5.2 Western blot Analysis

For Western blot analysis, proteins separated by SDS-PAGE gel electrophoresis

were transferred electrophoretically onto a nitrocellular membrane with Mini Trans-Blot

Cell (Bio-rad). The transfer buffer used in this study was Towbin buffer (3.03g/l Tris,

14.4 g/l Glycine, 20% V/V Methanol). Transfer was performed either at 75 mA for 2~3

hours or at 30 mA for overnight at 4°C. After the transfer was completed, the membrane

was removed from the sandwich and then incubated with 5% non-fat milk in TBST

buffer (20 mM Tris, 150 mM NaCl, 0.05% V/V Tween-20, pH 7.4) to block the

remaining protein-binding sites in the membrane. The blocking reaction was carried out

for 1 hours at room temperature on an orbital shaker. After blocking reaction was

completed, the membrane was washed at least three times with TBST buffer for ten

minutes each on a rocking platform. Then, the membrane was incubated with the TBST-

diluted primary antibody (usually use 100ug/ml as final concentration) for 1 hour at room

46

temperature while continuously rocking the sample. To remove the unbound primary

antibody, the membrane must be washed at least three times as aforementioned. After the

washing was completed, the membrane was incubated with the TBST-diluted second

antibody (usually in a 1:5000 dilution). The unbound second antibodies were washed

away by three extensive TBST washes. The fluorescence produced by the HRP

(horseradish peroxidase) -conjugated second antibody was detected with the SuperSignal

West Pico System (Pierce).

2.5.3 In-Gel Digestion, MALDI-TOF and Tandem MS Sequencing

Protein bands were carefully excised from the gel, cut into small pieces, and then

destained with several washes of 50 mM ammonium bicarbonate in 50% aqueous

acetonitrile. The gel pieces were dehydrated with 100% acetonitrile and dried in Savant

Speed-vac. The dried gel pieces were rehydrated with a solution containing 10 mM DTT

and 100 mM ammonium bicarbonate and incubated at 57°C for 60 min to reduce the

disulfide bonds. The reduced sulfhydryl group was alkylated by 55 mM iodoacetamide in

100 mM ammonium bicarbonate at room temperature for 60 minutes. After alkylation,

the gel pieces were first washed with 100 mM ammonium bicarbonate solution, then

dehydrated with 100% acetonitrile at least 6 times, and then dried in Savant Speed-vac

again. Sequencing grade modified trypsin (12.5 mg l-1

in 50 mM ammonium bicarbonate,

pH 8.0) was added to the dried gel pieces and incubated overnight at 37°C. The resulting

trypsinized peptides were extracted with 20 mM ammonium bicarbonate solution, 5%

formic acid in 50% aqueous acetonitrile, and 100% acetonitrile, respectively. The

47

extracted peptides were combined, lyophilized, and then resuspended in 0.1%

trifluoroacetic acid (TFA). An aliquot of the resuspended peptides was spotted onto the

stainless steel MALDI sample plate and overlaid with equal volume of matrix solution

(20 g l-1 α-cyano-4-hydroxycinnamic acid in 0.1% TFA; 50% aqueous acetonitrile). The

sample (matrix mixture) was allowed to air dry. An Applied Biosystems Voyager-DE

STR MALDI mass spectrometer was used to acquire the MALDI-time of flight (TOF)

spectra. The instrument was operated in the positive reflector delayed extraction mode.

Peptide mass spectra were searched against theoretically spectra derived from proteins in

the non-redundant databases of National Center for Biotechnology Information (NCBI) to

match peptide mass fingerprints. Partial amino acid sequence of a single peptide was

obtained by quadrupole-time of flight (Q-TOF) hybrid tandem mass (MS/MS)

spectrometer. For Q-TOF MS/MS analysis, the trypsinized peptide samples were

concentrated and desalted by the Zip-Tip method. Based on the mass spectra acquired by

MALDI-TOF, dominant peptides were selected for MS/MS analysis. After peptides were

selected, the Q-TOF mass spectrometer was automatically switched from MS to MS/MS

mode and the Q-TOF MS/MS spectra were collected. The amino acid sequences were

determined by searching the Q-TOF MS/MS spectra against the NCBI non-redundant

protein databases (Westermeier 2002).

.

2.5.4 N-Terminal Sequencing

Purified protein sample (1 ~ 2 mg/ml) was loaded into the ABI Procise 494

Protein Sequencer for N-terminal sequencing analysis. Principally, in this reaction

48

phenylisothiocyanate (PITC) reacts with the amino acid residue at the amino terminus

under basic conditions (provided by n-methylpiperidine/methanol/water) to form a

phenylthiocarbamyl derivative (PTC-protein). Trifluoroacetic acid was then used to

cleave off the first amino acid as its anilinothialinone derivative (ATZ-amino acid) and

leave the new amino terminus for the next degradation cycle. The ATZ amino acid is then

removed by extraction with N-butyl chloride and converted to a phenylthiohydantoin

derivative (PTH-amino acid) with 25% TFA/water. The PTH-amino acid is transferred to

a reverse-phase C-18 column for detection at 270 nm. This chromatogram provided

standard retention times of the amino acids for comparison with each Edman degradation

cycle chromatogram. The HPLC chromatograms were collected using a computer data

analysis system. To determine the amino acid present at a particular residue number, the

chromatogram from the residues of interest was compared with the chromatogram from

the previous residue by overlaying one on top of the other. From this, the amino acid for

the particular residue can be determined. This process was repeated sequentially to

provide the N-terminal sequence of the protein.

(http://www.protein.iastate.edu/nsequence494.html)

2.5.5 Circular Dichroism (CD) Spectrum

Circular Dichroism (CD) spectroscopy is a type of absorption spectroscopy that

can provide information on the secondary & tertiary structures of many types of

biological macromolecules. Circular Dichroism is the difference between the absorption

of left and right handed circularly-polarised light and is scanned as a function of

49

wavelength. Biological macromolecules, such as proteins, carbohydrates and nucleic

acids, are composed of many optically active or chiral units that exhibit large CD signals.

CD spectroscopy has therefore been used extensively in the study of proteins, where

asymmetric carbon atoms in their amino-acid 'backbone' give rise to a CD spectrum. The

shape of the spectrum depends on the protein's secondary structure content and allows the

proportions of helix, beta structure, turns and random coil to be determined.

Purified protein was diluted in phosphate buffer to final concentration of 100

µg/ml. 200 µl protein sample was loaded and use water as blank control.

(http://www.cryst.bbk.ac.uk/BBS/whatis/cd_website.html)

2.5.6 Dynamic Light Scattering (DLS)

The dynamic light scattering (DLS) is a well established technique for measuring

particle size over the size range from a few nanometers to a few microns. The technique

uses the idea that small particles in a suspension move in a random pattern. When a beam

of light passes through a colloidal dispersion, the particles or droplets scatter some of the

50

light in all directions. When the particles are very small compared with the wavelength of

the light, the intensity of the scattered light is uniform in all directions; for larger particles

(above approximately 250 nm diameter), the intensity is angle dependent. If the light is

coherent and monochromatic, as from a laser for example, it is possible to observe time-

dependent fluctuations in the scattered intensity using a suitable detector such as a

photomultiplier capable of operating in photon counting mode.

(http://www.brookhaven.co.uk/dynamic-light-scattering.html). Observation of larger

particles compared to smaller particles will show that the larger particles tumble more

slowly than the smaller ones if the temperature is the same. According to Einstein's

developments in his Kinetic Molecular Theory (applied to heat), molecules that are much

smaller than the particles can impart a change to the direction of the particle and its

velocity. Thus water molecules (0.00033 microns) can move polystyrene particles as

large as a couple of microns. The combination of these effects is observed as vibrations

and an overall movement of the particle. Concentrated and purified protein prior to

crystallization was tested for homogeneity using the Dynamic Light Scattering (DynaPro,

Protein Solutions). A few minutes are required for the sample and cell to equilibrate with

the actively controlled temperature environment. The cuvette must be washed with sterile

water until the counts are below 10. Then 50 µl concentrated sample was loaded into the

cuvette and any bubbles should be avoided. At least 15 times of counting should be

collected. (http://www.microtrac.com/dynamicscattering.cfm)

51

2.6 Crystallization

Crystallization screens were performed using the hanging-drop method with

Hampton 24-well plastic plates at 20 °C. The initial screens were carried out using

Hampton crystallization screen kits (crystal screen 1&2, index). A drop composed of a

mixture of 1 µl protein solution and 1µl reservoir solution is placed in vapor equilibration

with a liquid reservoir of regent. The initial conditions were optimized to get diffraction-

quality crystals. The crystals were soaked into the cryoprotectant buffer before flash-

frozen in liquid nitrogen for data collection.

2.7 Data Collection

X-ray diffraction data were collected at 100°K using synchrotron radiation. A

three wavelength experimental data set for the rVP28 crystals was collected at beamline

X29, NSLS, USA while that of rVP26 crystals was collected at beamline X26C, NSLS,

USA. Data for rVP26 and rVP28 were processed and scaled using the HKL suite

(Otwinowski and Minor 1997).

2.8 Structure Determination

The structure of rVP28 and rVP26 were determined using the MAD method. The

position of the seleno-methionine were determined and phases computed with SOLVE

(Terwilliger and Berendzen 1999). Density modification was effected by using

52

RESOLVE (Terwilliger 2000). The resulting electron density map was not good enough

and the model was built manually by using the program O (Jones et al. 1991). The initial

model was refined by rigid body, individual B-factor and energy minimization refinement

using CNS (Brunger et al. 1998). For rVP28, the restrained non-crystallographic

symmetry (NCS) was applied to increase the observation to parameter ratio throughout

the CNS refinement. Also, the test set (Rfree) was introduced into the refinement to

monitor and validate the model refinement. At each round of refinement, the geometry of

the structure was checked with PROCHECK using the Ramachandran plot (Laskowski et

al. 1993). Water molecules are added in the final stage of refinement. Structural

superposition was performed with the program O (Jones et al. 1991).

53

CHAPTER THREE

3 Characterization of WSSV Envelope Proteins

54

3.1 Characterization of Envelope Protein VP300

3.1.1 Identification of VP300 by Mass Spectrometry

Purified WSSV virions were separated by SDS-PAGE (Figure 3.1). More than 30

bands were visualized using Coomassie brilliant blue R-250 staining. Many major bands

have been identified previously (such as VP19, VP24, VP26 and VP28 as envelope

proteins and VP15 as nucleocapsid) however many minor bands are still undefined. The

band highlighted by red arrow was excised and digested by trypsin and analyzed by

MALDI-TOF mass spectrometry. Search the WSSV ORF database with the masses of a

list of trypsin-treated peptides has identified a protein as the product of the vp300 gene.

The ORF is located between nucleotide position 132994-133893 on the genome (Yang et

al. 2001). This 900 bp ORF presumably encoded a 300 aa protein (hence termed the

vp300 gene, GenBank accession number AF403003, and the resulting protein termed

VP300). One more interesting finding from the mass spectrometry result is that VP292 is

also identified by MALDI and Q-TOF from the same band. The mass spectrometry result

is consistent with previous proteomic research (Tsai et al. 2004; Zhang et al. 2004). It

suggests that VP300 might be a WSSV virion-associated protein.

3.1.2 Expression and Purification of VP300

The VP300 gene was amplified by PCR from the WSSV genome and inserted

into the vector pET-32(a) to express the (His)6-VP300. The recombinant protein was

55

Figure 3.1 1D SDS-PAGE of the WSSV proteins Viral proteins were purified and separated via one-dimensional SDS-PAGE. Left lane is the protein marker and the right lane is WSSV proteins. Four major structural proteins (VP19, VP24, VP26 and VP28) are indicated by black arrow. The band highlighted by red arrow was excised and digested by trypsin and then analyzed by MALDI-TOF mass spectrometry. The search of WSSV ORF database with a list of trypsin-treated peptide masses identified one of the proteins as the product of the vp300 gene. .

25

15

37

50

75

200150100

kDa

VP300

VP28 VP26 VP24

VP19

56

expressed in the BL21 (DE3) cell and purified by Ni-NTA under denaturing condition.

Figure 3.2 shows that SDS-PAGE gel of the purified VP300 protein. The purified protein

was injected into mouse to raise the mouse anti-(His)6-VP300 antibody as mentioned in

Chapter Two.

3.1.3 Localization Study of VP300

3.1.3.1 Western Analysis of VP300

To test specificity of the anti-VP300 antibody, western blot analysis was

performed. From Figure 3.3A, the purified mouse anti-(His)6-VP300 IgGs was used and

it can detect the VP300 protein among a mixture of all viral proteins as well as the

positive control of purified (His)6-VP300 protein. As a negative control, the antibody did

not react with purified VP281, another envelope protein of WSSV. It showed that the

purified mouse anti-(His)6-VP300 IgGs reacted specifically with VP300. Also it proved

that VP300 was of the WSSV origin rather than a contaminating protein linked

superficially to the WSSV virion. To further identify the location of VP300 in the virion,

the virus sample was fractionated into envelope and nucleocapsid fractions by the

treatment with 2% Triton X-100. As in Figure 3.3B, the western blot result shows that

VP300 was clearly detected in the WSSV whole virion and the purified viral envelope

fraction. However, the anti-VP300 antibodies did not react with the proteins of WSSV

nucleocapsid. This indicates that VP300 belongs to viral envelope, instead of

nucleocapsid.

57

Figure 3.2 Expression of the purified (His)6-VP300 protein

Shown are Coomassie Brilliant Blue –stained 12% SDS-PAGE gels. Lane 1, protein marker; Lane 2, recombinant pET-32(a) plasmid containing vp300 gene, non-induced; Lane 3, recombinant pET-32(a) plasmid containing vp300 gene, induced; Lane 4, purified (His)6-VP300 protein by Ni-NTA resin.

14.4

21.5

31.0

45.0

66.0

200.0

kDa

1 2 3 4

(His)6-VP300

58

Figure 3.3 Western blot analysis of WSSV with anti-VP300 IgG

Figure 3.3A is Coomassie Brilliant Blue-stained SDS-PAGE gels and the corresponding western blot analysis with anti-VP300 IgG. Lane 1, purified (His)6-VP300 protein as positive control; Lane 2, purified VP281 protein as negative control; Lane 3, protein marker; Lane 4, WSSV virion. Figure 3.3B is the western blot analysis of the different fractions of WSSV with anti-VP300 IgG. Lane 1, WSSV whole virion; Lane 2, WSSV viral envelope; Lane 3, WSSV nucleocapsid.

VP300

Western blot with anti-VP300 antibody Coomassie Brilliant Blue-stained gel

1 2 3 4 1 2 3 4

A

B

1 2 3

59

3.1.3.2 TEM Immunogold-Labeling Analysis

TEM immunogold-labeling method was further used to confirm the location of

VP300. The results shown in Figure 3.4 demonstrated that high-density gold particles

were located specifically at the viral envelope, while no apparent signals could be

observed in the nucleocapsid. The mouse anti-(His)6 antibody was also used as control to

replace the anti-(His)6-VP300 IgGs but no gold labeling signals were observed in the

viral envelope. From the Western blotting assay and TEM observations, it is concluded

that VP300 is a WSSV envelope protein.

3.1.4 Neutralization Assay of VP300

Virus envelope proteins play important roles in viral infection, such as recognition

and attachment to receptors on the surface of host cell. So being an envelope protein, is

VP300 involved in viral entry during infection? In an attempt to identify the envelope

protein’s involvement in WSSV infection on shrimp, the neutralization assay of infection

by WSSV virion with antibodies against VP300 was performed. First, the suitable dosage

of WSSV for shrimp infection was determined by performing an in vivo virus titration.

As expected, the mortality varied among groups of shrimp injected with different amount

of viruses. Delay in shrimp mortality was observed with the serial dilution of virus. When

the WSSV virion was diluted in PBS buffer with a ratio of 1:106, a mortality of 100%

was observed in a period of 11 days (Figure 3.5B). Based on the in vivo virus titration,

the WSSV virion concentration of 106 dilutions was selected for later experiments

60

Figure 3.4 Localization of WSSV VP300 proteins by immunoelectron microscopy

A, gold particles localize in virus envelope (arrow indicated); B, No gold particles was found in the nucleocapsid. The primary antibody is the purified mouse anti-(His)6-VP300 IgG, and the second antibody is goat anti-mouse IgG conjugated with 15 nm of gold.

A

B

61

Neutralization

0

0.2

0.4

0.6

0.8

1

1.2

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16

day

mor

talit

y 0.9%NaClWSSV+Anti-GSTWSSV(1:1000000)WSSV + Anti-VP300

0.00%

20.00%

40.00%

60.00%

80.00%

100.00%

120.00%

1 2 3 4 5 6 7 8 9 10 11

1 / 1001 /10001 / 100001 /1000001 /1000000

Figure 3.5 Neutralization Analysis of WSSV infections

A, Neutralization of WSSV infection in crayfish by antibody against VP300. B, Dosage of WSSV for shrimp infection. Days post-infection were shown on the abscissa and accumulated mortality on the ordinate.

B

A

62

because it is expected to give the optimal response to the neutralization in terms of

mortality. After we selected the suitable dosage of WSSV, we performed the

neutralization assay by adding specific antibodies. The antibodies against VP300 and

WSSV virion, as well as the anti-GST antibodies as control, were used in the in vivo

neutralization assays. After incubation of WSSV with each antibody, the mixtures were

injected into shrimp. The results are shown in Figure 3.5A. It showed that the shrimp

mortalities were very low, almost zero for the negative control (0.9% NaCl), whereas the

shrimp from the positive control (WSSV only) displayed 100% mortality in 15 days post-

infection. However, when the mixture of WSSV and anti-VP300 IgGs was injected, the

shrimp mortality fell significantly to almost zero, indicating that the infection of WSSV

could be delayed or neutralized by anti-VP300 IgGs.

3.1.5 Implications

Proteomic study by mass spectrometry had identified the existence of VP300

protein in the WSSV virion. From the SDS gel, the observed molecular mass of VP300 is

around 38 KDa, which is greater than the theoretical molecular mass (34.4 KDa) of the

vp300 gene product. This difference indicates that some posttranslational modifications

might have occurred. Computer assisted analysis revealed that there are three possible

phoshporylation sites and one potential N-linked glycosylation site in VP300.

Furthermore, VP292, an envelope protein that previously confirmed by Huang et al

(2002), was also detected by mass spectrometry from the same SDS PAGE band. When

we compared the protein sequence of VP300 and VP292, there is no significant

63

homology. Since the functional characterization of VP292 is still in progress, we cannot

confirm any functional relationships between these two proteins.

Besides the mass spectrometry results, the observations from Western blot and

immuno-gold labeling TEM further confirmed that VP300 is a WSSV envelope protein

but not a nucleocapsid protein. Similar to many known membrane proteins, VP300 has a

transmembrane region predicted by sequence analysis using the DAS server (Stockholm

University). It suggests that this hydrophobic region was responsible for insertion into

membrane lipid bilayer and VP300 may be a membrane anchor protein.

To date, several envelope proteins have been identified (VP19, VP31, VP24,

VP26, VP28, VP124, VP281, VP292, VP466). Previous neutralization study of six

WSSV envelope proteins indicates that some of the envelope proteins had function on

viral infection while some do not. In this study, neutralization assay showed that the anti-

VP300 IgGs functions to delay or neutralize the infection of shrimp by WSSV. The

antibody against VP300 could bind to envelope spikes on the virion and prevent the

attachment of the virus to the cell surface or uncoating of the virus (Burton et al. 2000;

Wu et al. 2005). VP300 might be a key protein in viral invasion and could be a good

candidate for screening of receptors on the cell surface. Further study on VP300 would

help to understand the mechanism of the virus infection. Up until now, no efficient

method has been found to prevent the infection of shrimp by WSSV. Antibody against

VP300 could be used directly as a passive immune strategy to control the WSSV

infection.

64

3.2 Localization Study of VP26 in WSSV virion

3.2.1 Immunogold labeling of VP26 in WSSV particles

(This part of experiments was done by Dr. Jinlu Wu)

Compared with the confirmed envelope protein of VP28, the location of VP26 in

the virion is still unclear. VP26 was first reported as a nucleocapsid protein (van Hulten

et al. 2001b) and later reported as an envelope protein (Zhang et al. 2002b). However, the

most recent publications claimed that VP26 is a linker (Xie and Yang 2005) or a

tegument protein (Tsai et al. 2006). Clarification of the location of VP26 should improve

our understanding on function of the protein.

To study the location of VP26 in the virion, the virus was treated with Tween 20

to get partial and complete separation of viral envelope from the nucleocapsid so that we

can distinguish the location of gold particles. Figure3.6 shows that the detergent

treatment separated the envelope from the nucleocapsid. Interestingly, some completely

separated envelope formed vesicles of different sizes and shapes. The treatment also

resulted in visible surface projections and spike-like structures, which were not observed

on the envelope of an intact virion. Figure 3.6A-3.6D show immunogold-labeled VP26,

and Figure 3.6E show immunogold-labeled VP28. All gold particles were found to bind

at the outer surface of the envelope, while none bind to the inner surface of envelope,

nucleocapsid or the space between them. Some gold particles were clearly localized on

the spike-like structures when using anti-VP26 for immunogold labeling (insert in Figure

3.6D). The negative control (Figure 3.6F) shows very low level of non-specific binding

65

A CB

i

iiii

ii

i

i

D E

i

F

ii

i

A CB

i

iiii

ii

i

i

D E

i

F

ii

i

Figure 3.6 localization of WSSV VP26 proteins by immunoelectron microscopy

VP26 and VP28 are both envelope proteins. Purified WSSV virion was treated with 0.1% Tween 20 and then was immunogold labeled with VP26 and VP28 antibodies. A-D show VP26 antibody specially binds to outer surface and the gold particles are localized on the outer surface of virions (A-i, C-i), semi-separated envelope (B-i) and fully separated envelope (envelope vacuoles, A-ii, C-ii, D-i). E shows immunogold labeled VP28 is also localized on the outer surface of envelope. Surface projects (solid arrows) and spike-like (empty arrow) structures clearly appeared on some of the envelope (C-i and E-i) and vesicles (D-i), respectively. The insert in D shows a large view of spike-like structures. F shows a very low binding background when a pre-immune rabbit antibody or gold-conjugated secondary antibody was used. The F also shows a possible process of vesicle formation (bar=100 nm). The EM experiment was preformed by Dr. Jinlu Wu.

66

and a possible mechanism for the formation of vesicles. On the other hand, if the virion

was over treated with Tween 20, VP26 can only be scarcely labeled. We also tried to treat

the virion with 0.1% Triton X-100, but such treatment always resulted in complete loss of

viral envelope, only nucleocapsid remained after the subsequent immunogold labeling

was finished.

3.2.2 Implications

Due to the dispute over the location of VP26, we have carried out an examination

on VP26 by immunoelectron microscopy in this study. In this study we chose to use

Triton X-100 and Tween 20 to treat the virion to expose antibody binding sites and to

separate the envelope from the nucleocapsid. Our result showed that 0.1% Tween 20

could effectively separate the envelope from the nucleocapsid, but the treatment with

0.1% Triton X-100 was too harsh and resulted in complete loss of envelope during

subsequent steps of immunogold labeling. More interestingly, surface projections and

spike-like structures could be observed in the outer surface of the envelope after

treatment with Tween 20. It suggests that the envelope structure became loosely packed,

thus more antibody binding sites for both VP26 and VP28 were exposed resulting in

higher labeling efficiency. Even though, no gold particles were observed in the space

between the inner surfaces of envelope or the nucleocapsid, all the gold particles were

localized on the outer surface of the semi or fully separated envelope. Taken together, we

proposed that VP26 itself is an envelope protein and is anchored at the viral lipid bilayer

by its N-terminal transmembrane region.

67

3.3 Candidates for Structure Analysis

3.3.1 Solubility Test of Major Envelope Proteins

Although many structural genomic programs have used different criteria or

priorities for the selection of target proteins (Brenner 2001), the common theme is to

restrict the selection to candidates to proteins of unknown structure, expected interest and

accessibility (yields, solubility and ease of labeling etc). In the present proposal, we have

focused on the envelope proteins what are important for viral infection, assembly and

morphogenesis. These candidates were expressed in a bacterial system. The yield and

solubility of these proteins were examined and protocols will be developed for protein

purifications and characterization. To express and purify all these envelope proteins, the

encoding DNA fragment was cloned into different kinds of vectors, including pET15b,

pET32(a), pGEX-4T-2 and pGEX-6P-1 and then transformed into the BL21 (DE3) cell

for expression as mentioned in Chapter Two. All envelope proteins, except for VP281,

are predicted to contain a transmembrane domain. So the first step is to check the

solubility of these full length proteins. Initial trials were performed by expression using

0.1 mM IPTG at 30 °C for 4 hours. The expression condition was optimized by changing

the final concentration of IPTG and culture temperature. As shown in Figure 3.7, when

expressed as GST fusion proteins, VP281 and VP466 were partially soluble when the

cells with the recombinant plasmid were cultured at the optimal condition. But for VP19,

VP24, VP28 and VP68, they were all expressed as insoluble proteins. VP281 and VP466

were thus first selected as candidates for X-ray crystallography study because of their

higher solubilities.

68

VP281 VP466

1 2 1 2

VP281 VP466

1 2 1 2

VP28 VP26 VP19 VP68

1 2 1 21 21 2

VP28 VP26 VP19 VP68

1 2 1 21 21 2

Figure 3.7 Solubility study on the envelope proteins of WSSV

Six envelope proteins are tested for their solubility in their optimal conditions. Lane 1, supernatant; Lane 2, pellet. VP281 and VP466 are partially expressed in soluble fraction while VP28, VP26, VP19 and VP68 are expressed in the inclusion body.

69

3.3.2 Purification of VP281 Protein and Crystallization Trials

The solubility test showed that VP281 and VP466 were expressed as soluble

proteins in a bacterial system. Thus we performed the large scale expression and

purification of these two proteins. The vp281 gene was cloned into pGEX-6P-1 using the

restriction enzyme sites, BamHI and EcoRI. The VP281-GST fusion protein was first

purified using glutathione sepharose and then the GST tag was removed by the cleavage

using ProScission protease as mentioned in Chapter Two. The expressed protein is 286 aa

in size with predicted MW of 31970.80 Da and pI of 4.47. There are 4 cysteine residues

in the VP281 sequence indicating that disulfide bonds might form inside VP281 to

stabilize its fold or/and to form linkage between two VP281 molecules.

The SDS-PAGE gel of purified VP281 is shown in Figure 3.8. Further

purification of VP281 was performed by ion exchange, hydrophobic interaction

chromatography (HIC) and size exclusion chromatography. MALDI-TOF MS and Q-

TOF MS as well as N-terminal sequencing were carried out to confirm the identity of the

purified protein. MALDI-TOF MS result showed that the amino acid sequence coverage

is about 42% (120 of 281). And the Q-TOF MS result showed that the MW of VP281 is

31973.80 Da, almost the same as the theoretical MW of 31970.80 Da. The N-terminal

sequence of 12 amino acids also confirmed identity of the purified VP281 protein. A CD

spectrum was also taken to study the secondary structure of this protein and from the

result we interpreted that VP281 consisted mainly of α-helical secondary structure.

70

GST-VP281

VP281

GST

1 2 3 4 5 6 7 8

14.4

21.5

31.0

45.0

66.0

200.0

kDa

GST-VP281

VP281

GST

1 2 3 4 5 6 7 8

14.4

21.5

31.0

45.0

66.0

200.0

kDa

14.4

21.5

31.0

45.0

66.0

200.0

kDa

Figure 3.8 Expression of the purified VP281 protein

Shown is Coomassie Brilliant Blue –stained 12% SDS-PAGE gel. Lane 1, protein marker; Lane 2, pGEX-6P-1 (vector only as control)-pLysS, non-induced; Lane 3, pGEX-6P-1-pLysS, induced; Lane 4, recombinant pGEX-6P-1 plasmid containing vp281 gene, non-induced; Lane 5, recombinant pGEX-6P-1 plasmid containing vp281 gene, induced; Lane 6, purified GST-VP281 protein; Lane 7, GST-VP281 fusion protein cleavage with ProScission™ protease; Lane 8, purified VP281 protein.

71

Homogeneity of the purified protein is the key to grow a well-diffracting crystal.

Single band purity on native PAGE is neither a sufficient nor a necessary prerequisite to

obtaining crystals but has always been regarded as a good starting point for crystal

growth. The native gel and SDS-gel together will give some hints about the extent of

protein aggregation. The purified VP281 protein was concentrated to 10 mg/ml and ran in

native gel. The gel filtration profile showed that the protein was eluted as a dimeric but

when running in the native gel we could clearly see several bands, indicating that there

was significant aggregation happened in VP281 protein. The most frequent cause of non-

specific aggregation is suboptimal protein solvent conditions. We have tried to change

the buffer condition, such as the concentrations of NaCl, glycerol and the reducing agent,

but to no avoid. However, there was no apparent difference of all in the variations in the

native gel. It is impossible to obtain any diffraction-quality crystals from this poor quality

protein. Further purification steps or truncation treatments are required for this protein for

crystallization.

3.3.3 Purification of VP466 Protein and Crystallization Trials

The vp466 gene was cloned into pGEX-4T-1 between the restriction enzyme sites,

BamHI and XhoI. VP466 was first expressed as a GST fusion protein using Glutathione

sepharose 4B beads and then the GST tag was removed by thrombin protease cleavage.

The SDS-PAGE gel of purified VP466 is shown in Figure 3.9. The corresponding bands

were excised from the gel, digested by trypsin and then portion of tryptic peptide

fragments was studied by MALDI-TOF MS. The fitted amino acids residues covered

72

14.4

21.5

31.0

45.0

66.0

200.0

kDa

GST-VP466

VP466

GST

1 2 3 4 5 6 7 8

14.4

21.5

31.0

45.0

66.0

200.0

kDa

14.4

21.5

31.0

45.0

66.0

200.0

kDa

GST-VP466

VP466

GST

1 2 3 4 5 6 7 8

Figure 3.9 Expression of the purified VP466 protein

Shown is Coomassie Brilliant Blue –stained 12% SDS-PAGE gel. Lane 1, protein marker; Lane 2, pGEX-4T-1 (vector only as control)-pLysS, non-induced; Lane 3, pGEX-4T-1-pLysS, induced; Lane 4, recombinant pGEX-4T-1 plasmid containing vp466 gene, non-induced; Lane 5, recombinant pGEX-4T-1 plasmid containing vp466 gene, induced; Lane 6, purified GST-VP466 protein; Lane 7, GST-VP466 fusion protein cleavage with thrombin protease; Lane 8, purified VP466 protein.

73

42% of the whole sequence (198 of 466). And the Q-TOF MS result showed that the MW

of VP466 was 52053.20 Da, slightly larger than the predicted M.W. of 52052.30 Da, but

the difference was within the allowable error range. Furthermore, the N-terminal

sequence of 14 amino acids also confirmed the identity of the purified VP466 protein. A

CD spectrum showed that VP466 has a typical α-helical secondary structure.

The single band of purified VP466 protein in native gel indicated that the protein

was homogenous. However we were unable to get any crystals from the crystallization

screening. More crystallization trials on purified VP466 will be performed under different

conditions. Different crystal growth techniques like sitting drop vapor diffusion or under

oil will also be attempted.

74

CHAPTER FOUR

4 Crystal Study of Two Major Envelope Proteins

75

4.1 X-ray Structure of Major Envelope Protein VP28

4.1.1 Property of rVP28 Protein

VP28 and VP26 are two most abundant envelope proteins that consist 60% of the

envelope. Previous experiments have shown VP28 play important role in WSSV

infection and VP26 was able to interact with actin. As their importance and abundance in

viral envelope, we selected these two proteins for the structure determination. As we

mentioned before that the expression and purification trials on full-length VP28 and

VP26 resulted in a very low level of expression and the production of an insoluble

protein. Extensive detergent screen to solubilize and crystallize the protein was

unsuccessful. Sequence analysis using DAS server (Stockholm University) indicated the

presence of a transmembrane region at the N-terminal region of VP28 and VP26 (Figure

4.1). We hypothesized that removal of the transmembrane region would increase the

solubility and the yield of crystals. Thus we designed a construct encoding the major part

(Asn31-Glu204) of VP28 without the N-terminal transmembrane domain (Met01-His30).

Theoretical pI and MW of rVP28, the truncated protein, is 4.63 and 19258.47 Da

respectively. The SDS-PAGE electrophoresis is shown in Figure 4.2. The homogeneity of

purified rVP28 at different concentration was tested by native gel and dynamic light

scattering experiment respectively. The gel filtration profile as well as the dynamic light

scattering data showed that rVP28 is a monomer in solution. rVP28 at a concentration of

8-10 mg/ml was homogenous and suitable for crystallization trials.

76

Figure 4.1 DAS transmembrane prediction result for full length VP28 and VP26

These curves are obtained by pairwise comparison of the proteins in the test set in

"each against the rest" fashion. There are two cutoffs indicated on the plots: a "strict" one at 2.2 DAS score, and a "loose" one at 1.7. The hit at 2.2 is informative in terms of the number of matching segments, while a hit at 1.7 gives the actual location of the transmembrane segment.

VP28

VP26

77

GST-rVP28

GST

rVP28

1 2 3 4 5 6

15

20

25

37

50

75100

Figure 4.2 Expression of the rVP28 protein in E.coli BL21 DE3 star

Shown is Coomassie Brilliant Blue –stained 12% SDS-PAGE gel Lane 1, protein marker; Lane 2, recombinant pGEX-6P-1 plasmid containing vp28 gene, non-induced; Lane 3, recombinant pGEX-6P-1 plasmid containing vp28 gene, induced; Lane 4, purified GST-rVP28 protein; Lane 5, GST-rVP28 fusion protein cleavage with ProScission™ protease; Lane 6, purified rVP28 protein.

KDa

78

4.1.2 Preparation of SeMet rVP28 Protein

As VP28 lacks homology with any other known structure of viral proteins, the

MAD method was used to solve the structure of rVP28. The seleno-methionine rVP28

was expressed in defined LeMaster medium using the methionine auxotrophic strain

(DL41). The purification procedure was the same as that for the native protein. The

SeMet rVP28 was purified by Superdex-75 gel filtration chromatography using a gel

filtration buffer containing 20 mM Tris-Cl (pH 7.4), 150 mM NaCl, 10 mM DTT and 5%

Glycerol. The purified SeMet rVP28 was concentrated to around 10 mg/ml for

crystallization. To confirm the substitution status, the MW of seleno-methionine rVP28

and the native rVP28 were determined by MALDI TOF MS. The MW difference of

native and seleno-methionine rVP28 was around 271 Dalton, while the MW difference of

selenium and sulfur is 46.895 Dalton, indicating that all of the six methionine residues of

rVP28 were fully substituted by seleno-methionine.

4.1.3 Crystallization of rVP28

The initial crystals were obtained using under oil method. The condition was

later optimized using the hanging drop method to obtain diffraction-quality crystals.

Glycerol was used as the cryoprotectant for flash-frozen in liquid nitrogen. The details of

the conditions were listed in Table 4.1.

79

Table 4.1 Summary of Crystallization Conditions and Cryoprotectants

for rVP28 Crystals

Initial crystallization condition

20% PEG8K

0.2 M Ca acetate

0.1 M Na HEPES (pH 7.5)

Optimized Crystallization condition

25% PEG8K

0.2 M Ca acetate

0.1 M Na HEPES (pH 7.5)

1.5% w/v 1,2,3-heptanetriol

Cyroprotectant

25% PEG8K

0.2 M Ca acetate

0.1 M Na HEPES (pH 7.5)

20% glycerol

80

4.1.4 Structure Determination of rVP28

The structure of the recombinant SeMet VP28 was solved using the MAD method.

Three data sets in different wavelength were collected from the synchrotron beam line

X29 at the Brookhaven National Laboratories. Data were processed and scaled using the

HKL suite. BNP was used to locate the Se sites for VP28.

The phases were further developed using RESOLVE which improved the overall

FOM (figure of merit) to 0.75. The initial models were built automatically by RESOLVE

containing ~50% of the total residues. The rest of the model was manually built using

program O and refined using the program CNS interspersed with several rounds of

manual refitting. All the measured reflections were used in the refinement. The

asymmetric unit of VP28 consists of twelve molecules. Throughout the refinement the

Non Crystallographic Symmetry (NCS) restraints were used for VP28. The final R-

factors for VP28 is 0.24 (Rfree=0.28) at 2.0Å resolutions. The model was refined with

good stereochemical parameters. Statistics of the Ramachandran plot from an analysis by

PROCHECK gave an overall 89.7% of non-glycine residues in the most favored region

and only Asp166 in the generously allowed regions. This residue belongs to a less well-

ordered segment connecting two β strands. The details of data collection and refinement

are listed in Table 4.2.

81

Table 4.2 Data collection and refinement statistics of rVP28

Experiment

Cell parameters and angles a=105.33, b=106.71, c=200.37, α=β=γ=90°

Space group P212121

Data collection Peak Inflection Remote

Resolution range (Å) 50-2.0 50-2.1 50-2.1

Wavelength (Å) 0.9796 0.9799 0.9600

Observed Reflections>1 1818612 1608124 774774

Unique Reflections 152547 132247 131772

Overall (I/σI) 15.3 15.9 15.5

Redundancya 11.9(9.7) 12.2(11.2) 5.9(5.4)

Completenessa (%) 100(99.9) 100(100) 99.7(99.5)

Rsyma,b (%) 0.08(0.209) 0.072(0.179) 0.073(0.181)

Refinement Resolution range (Å) 45.0-2.0

Number of Reflections 294116

R factorc (%) 24.82

Rfreed (%) 28.12

RMSD bond lengths (Å) 0.008

RMSD bond angles ( °) 1.24

Average B-factors (Å2) 60.00

Water molecules 1422

Ramachandran Plot

Most favored region (%) 89.7

Additional allowed regions (%) 9.7

Generously allowed regions (%) 0.6

Disallowed regions (%) 0.0

a Last shell values givn in parentheses. b.Rsym = |Ii -<I>|/|Ii| where Ii is the intensity of the ith measurement, and <I> is the mean intensity for that reflection. b Rwork = | Fobs - Fcalc|/|Fobs| where Fcalc and Fobs are the calculated and observed structure factor amplitudes, respectively. c Rfree is calculated using the same equation as that for R work but 3% of reflections where chosen randomly and omitted from the refinement.

82

4.1.5 Overall Structure of rVP28

The full-length rVP28 exhibits single β-barrel architecture and an α-helix is

protruding from the β-barrel. The N-terminal extended (protruding) α-helix is 15 residues

(Thr32-Asn47) of approximately 23 Å in length from the surface of the β-barrel. A total

of nine β-strands (β1↓β2↑β3↓β4↓ß5↑β6↓β7↑β8↓β9↑), mostly anti-parallel, form the

barrel. The N-terminal protruding α-helix is linked by 2 residues to the core β-barrel. The

overall dimension of the β-barrel is approximately 35 Å (height) x 15 Å (diameter)

(Figure 4.3).

The pore of the β-barrel is highly hydrophobic in nature with 25 hydrophobic side

chains lining the inner surface, contributed by side chains from seven Phe, eight Ile, one

Leu, four Met and five Val residues. Taken together the size and the hydrophobic nature

of the pore, the latter may help to recognize small, specific, hydrophobic ligands/ions

which could potentially establish hydrophobic or van der Waals interactions with the side

chains of the pore residues. The exact role of this pore for the function of VP28 is yet to

be established.

83

Figure 4.3 Ribbon diagram of rVP28 monomer. The N and C termini are labled. Figure was prepared using the programs MOLSCRIPT (Kraulis 1991) and Raster3D (Merrit 1997).

N

C

84

4.1.6 Oligomerization of rVP28

In the crystal, an asymmetric unit consists of twelve copies of rVP28 molecules,

each with 170 residues from Thr32-Thr201 assembled into four trimers (Figure 4.4).

However, Gel filtration, SDS-PAGE and dynamic light scattering results showed that

rVP28 is a monomer in solution. Purified WSSV was treated with RIPA buffer and

analyzed by non-reducing SDS-PAGE and Western blotting. The bands detected by anti-

VP28 were excised from the corresponding gel and analyzed by MALDI TOF TOF MS.

There were three bands with different M.W. that matched to masses of a list of trypsin-

treated peptide of VP28. The bands correspond to a trimer (~75KD), dimer (~50 KD) and

monomer (~25KD, the most abundant species) (shown in Figure 4.5). This suggests that

VP28 naturally forms trimers in the viral envelope but the trimer species may be very

fragile during virus purification and lysis treatment, hence it becomes a monomer in

solution. However, during crystal formation the monomers tend to associate as trimers,

the most natural oligomerization state of VP28.

In the crystal of rVP28, a channel is formed by the N-terminal α-helix and the

wall of the β-barrel from each monomer of the trimeric rVP28. The length and weight of

the channel are approximately 58 X 12 Å. No salt bridges are apparent between any of

these residues. There are 10 hydrogen bonding contacts (<3.2 Å) and several hydrophobic

interactions between each monomers of the trimeric rVP28 to keep the integrity of the

channel. The surface area of a monomer is around 9024 Å2, whereas around 1170 Å2

(around 13%) is buried at the monomer interface of the trimer.

85

Figure 4.4 Ribbon diagram of rVP28 trimer. A) One trimer of the asymmetric unit. Each monomer of the trimer is shown in different color. B) Top view of the trimer. Figures were prepared using the programs MOLSCRIPT (Kraulis 1991) and Raster3D (Merrit 1997). The N and C termini are labled

N

C

86

Figure 4.5 Western blot analysis of WSSV by anti-VP28

The bands that were detected by anti-VP28 IgGs were excised for MALDI TOF TOF MS. The bands indicated by arrows match to VP28 of WSSV.

.

87

4.2 X-ray Structure of Major Envelope Protein VP26

4.2.1 Purification of rVP26 and SeMet rVP26 protein

The expression and purification trials on full-length VP26 had resulted in a very

low expression and the production of an insoluble protein as we have mentioned before.

We hypothesize that the low solubility was due to the N-terminal strong hydrophobic

region which was predicted as a transmembrane region by sequence analysis. The

transmembrane truncated form of VP26 (Asn34-Lys204) were constructed, with the

theoretical pI and M.W. of 9.57 and 19038.8 Da respectively. The analysis of the

expressed protein by SDS-PAGE is shown in Figure 4.6.

Similar to the native protein, SeMet substituted rVP26 was purified by Superdex-

75 gel filtration chromatography using a gel filtration buffer containing 20 mM Tris-Cl

(pH 7.4), 200 mM NaCl, 10 mM DT and 5% Glycerol. It is a monomer in solution as

confirmed by the gel filtration profile and the native gel. MALDI TOF mass spectrometry

was used for the measurement of the molecular weight of purified SeMet rVP26 as well

as native rVP26, to make sure that all of the nine methionine residues of rVP26 were

fully substituted by seleno-methionine. The purified SeMet rVP26 protein was

concentrated. The DLS result showed that the purified rVP26 protein was homogenous

up to a concentration of 10 mg/ml. However aggregation will start to appear in solutions

with higher concentration. Thus a sample of concentration of 10 mg/ml was used later for

crystallization trials.

88

15

20

25

37

50

75

kDa

100

GST-rVP26

GST

rVP26

1 2 3 4 5 6

15

20

25

37

50

75

kDa

100

15

20

25

37

50

75

kDa

100

GST-rVP26

GST

rVP26

1 2 3 4 5 6

Figure 4.6 Expression of the rVP26 protein in E.coli BL21 DE3 star

Shown is Coomassie Brilliant Blue –stained 12% SDS-PAGE gel Lane 1, protein marker; Lane 2, recombinant pGEX-6P-1 plasmid containing vp26 gene, non-induced; Lane 3, recombinant pGEX-6P-1 plasmid containing vp26 gene, induced; Lane 4, purified GST-rVP26 protein; Lane 5, GST-rVP26 fusion protein cleavage with ProScission™ protease; Lane 6, purified rVP26 protein.

89

4.2.2 Crystallization, Data Collection and Structure Determination of rVP26

Crystals of rVP26 were grown using the hanging drop vapor diffusion method at

room temperature. Extensive additives screening was performed to optimize the

crystallization condition to obtain best diffraction-quality crystals. Glycerol was used as

the cryoprotectant for flash-frozen the crystals in liquid nitrogen. The details of the

crystallization conditions and cryoprotectants are listed in Table 4.3.

The structure of the recombinant SeMet rVP26 protein was solved using a MAD

data set collected from the synchrotron beam line X26 at the Brookhaven National

Laboratories. Data were processed and scaled using the HKL suite. SOLVE was used to

locate the Se sited and the phases were further developed using RESOLVE which

improved the overall FOM (figure of merit) to 0.67. The initial models were built

automatically by RESOLVE containing ~50% of the total residues. The rest of the model

was manually built using program O and refined with all the measured reflections using

the program CNS interspersed with several rounds of manual refitting. The structure was

refined at 2.1 Å resolution, to a final R-factor of 0.21 (Rfree=0.27). The model has been

refined with good stereochemical parameters. Statistics for the Ramachandran plot from

an analysis using PROCHECK (Laskowski et al., 1993) gave an overall 85.3% of non-

glycine residues in the most favored region, with only Ala 58 in the generously allowed

regions. This residue belongs to a poorly ordered peptide segment connecting two β

strands. No residue was observed in the disallowed region. The details of data collection

and refinement are listed in Table 4.4.

90

Table 4.3 Summary of Crystallization Conditions and Cryoprotectants

for rVP26 Crystals

Initial crystallization condition

0.1 M citric acid (pH 3.5)

3.0 M sodium chloride

Optimized Crystallization condition

0.1 M citric acid (pH 3.5)

3.0 M sodium chloride

1% w/v polyethylene glycol 3350

Cyroprotectant

0.1 M citric acid (pH 3.5)

3.0 M sodium chloride

30% glycerol

91

Table 4.4 Data collection and refinement statistics of rVP26

Experiment

Cell parameters and angels a=b=73.915, c=199.313; α=β =90°, γ=120°

Space group R32

Data collection Peak Inflection Remote

Resolution range (Å) 50-2.2 50-2.6 50-2.7

Wavelength (Å) 0.9782 0.9787 0.9600

Observed Reflections >1 187787 188815 120101

Unique Reflections 10798 10841 11519

Overall (I/σI) 23 19.1 12.3

Redundancya 17.4(13.9) 17.4(13.5) 10.4(10.1)

Completenessa (%) 99.9(99.4) 99.7(97.4) 99.8(98.8)

Rsyma,b (%) 0.090(0.327) 0.088(0.382) 0.082(0.259)

Refinement

Resolution range (Å) 45.0-2.2

Number of Reflections 19998

R factorc (%) 21.68

Rfreed (%) 27.85

RMSD bond lengths (Å) 0.007

RMSD bond angles ( °) 1.22

Average B-factors (Å2) 73.74

Water molecules 153

Ramachandran Plot

Most favored region (%) 85.3

Additional allowed regions (%) 14.0

Generously allowed regions (%) 0.7

Disallowed regions (%) 0.0

a Last shell values givn in parentheses. b.Rsym = |Ii -<I>|/|Ii| where Ii is the intensity of the ith measurement, and <I> is the mean intensity for that reflection. b Rwork = | Fobs - Fcalc|/|Fobs| where Fcalc and Fobs are the calculated and observed structure factor amplitudes, respectively. c Rfree is calculated using the same equation as that for R work but 3% of reflections where chosen randomly and omitted from the refinement.

92

4.2.3 Overall Structure of rVP26

Figure 4.7 shows the crystal structure of rVP26. The asymmetric unit contains one

VP26 molecule comprising 161 residues from Ser41 to Ile201 and a total of 153 water

molecules. Residues 34-40 and 202-204 from part of the N-terminal/C-terminal region

and had no interpretable density and were not modeled. The full-length rVP26 consists of

a nine stranded β-barrel with mostly anti-parallel β-strands in the orientation of

β1↓β2↑β3↓β4↓ß5↑β6↓β7↑β8↓β9↑. In addition, a two strand β-sheet (β1↓β9↑) protrudes

out of the β-barrel parallel to the barrel axis. Besides, there is a two turn α-helix hanging

outside of the β-barrel structure. In the protruding β-sheet, one strand is the N-terminal

part of the molecule, whereas the second strand is the C-terminal strand of VP26. The N-

and C-terminal of the molecule comes together to form this β-sheet. The N-terminal β-

strand is the longest in the molecule, having around 50Å in length; this β-strand is the

part of the β-barrel as well as the protruding β-sheet. The pore of the β-barrel is highly

hydrophobic in nature with side chains from three Phe, seven Ile, two Leu, five Met and

four Val, a total of 21 hydrophobic side chains lining the inner pore surface. The

approximate dimensions of the β-barrel are 38.5 Å (height) and 15.5 Å (diameter). We

suggest that this pore, similar to the β-barrel pore of rVP28, may allow the binding of

small, specific ligand/ions. The exact role of this pore for the function of rVP26 is yet to

be established.

93

Figure 4.7 Ribbon Diagram of rVP26 monomer

The β strand is colored green, while α helix and loop are colored as red and blue respectively. The N and C termini are labled. Figure was prepared using the programs MOLSCRIPT (Kraulis 1991) and Raster3D (Merrit 1997).

N

C

94

4.2.4 Oligomerization of rVP26

The oligomerization of rVP26 was investigated by gel filtration chromatography,

SDS-PAGE and native gel. All the experiments showed an apparent molecular mass of

20 KDa that corresponded to a monomer in solution. Only one molecule of rVP26 is

observed in an asymmetric unit. Interestingly, rVP26 crystallized in R32 space group

which has a three fold axis. The crystal packing showed a trimeric arrangement within

the crystal lattices from the symmetry related molecules (Figure 4.8).

Non-reduced SDS-PAGE and western blot analysis on the purified WSSV in

Figure 4.9 showed the presence of trimer (~66KD), dimer (~44 KD) and monomer

(~22KD, the most abundant species) of VP26. The arrow indicates the result

corresponding to MALDI TOF TOF MS. Similar to the rVP28 trimer, we speculate that

formation of the trimer may be the desirable aggregated form of this protein in the viral

envelope. The monomers of the trimer may be held together by very weak interactions

that are but not sufficiently strong to retain its oligomerization state in solution. The

immuno-electron images of VP26 that are shown in Chapter three indicate that the

observed spike-like structure should be a trimer.

Similar to the rVP28 trimer, a channel is formed by the protruding N terminal

region and the wall of the β-barrel from each monomer of the rVP26 trimer. There are

several water molecules present in the channel. The height and diameter are

approximately 58 × 12 Å. The channel may allow the passage of ions or small molecules,

but the exact role of these channels remains to be established.

95

Figure 4.8 Ribbon Diagram of rVP26 trimer

A) The rVP26 trimer (Crystallographic symmetry related molecules). Each monomer is shown in different color. B) Top view of the VP26 trimer. Figures were prepared using the programs MOLSCRIPT (Kraulis 1991) and Raster3D (Merrit 1997). The N and C termini are labled.

C

N

96

Figure 4.9 Western blot analysis of WSSV by anti-VP26

The bands were detected by anti-VP26 IgGs and the corresponding gel was excised for MALDI TOF TOF MS. The bands indicated by arrow match only to the VP26 of WSSV.

97

4.3 Comparison of VP26 and VP28

4.3.1 Structure Homolog of VP28 and VP26

A search of structural similarities for both full length rVP26 and rVP28 using the

DALI (Holm and Sander 1995) database failed to recognize significant matches to any

known structure. Interestingly, rVP26 and rVP28 display strong structural as well as

sequence homology among themselves. The structural superposition of rVP26 on rVP28

gave an rmsd of 2.6Å for 144 Cα atoms, as shown in Figure 4.10, with a sequence

similarity of ~36%. The structure based sequence alignment revealed that most of the

structurally invariant residues are located at β1, β3, α1 (equivalent of α2 in rVP28), β4

and β5. The rVP26 and rVP28 monomers possess a pore of similar size that is very

hydrophobic in nature. The hydrophobic side chain residues lining inside the pore are

quite conserved in these two proteins (Figure 4.11, Figure 4.12 and Figure 4.13). The

most significant difference between them is an extended β-strand in rVP26 and an α-

helix in rVP28 as their protruding parts. In the case of rVP26, the N-terminal part and the

C-termial strand come together to form a two stranded ß-sheet (β1↓β9↑) protruding out

of the β-barrel and parallel to the barrel axis. Whereas in rVP28, a long α-helix is

hanging outside the barrel at the N-terminus.

98

Figure 4.10 Stereo Cα superposition of rVP28 and rVP26

Cα of rVP28 in blue and rVP26 in green is shown. The superposition was generated using the program O (Jones et al. 1991), using the consered residues as the starting point. This figure was prepared using the program MOLSCRIPT (Kraulis 1991).

99

Figure 4.11 Structure-based sequence alignment of rVP28 and rVP26

The alignment was performed using the program O (Jones et al. 1991). The secondary structural elements and the sequence numbering for VP26 and VP28 were shown at the top and the bottom respectively. The conserved residues are highlighted in red as well as with blue line box. This figure was created by the program ESPript (Gouet et al. 1999).

100

Figure 4.12 Simulated annealing Fo-Fc omit map showing the conserved

region of rVP28

All the residues shown in the figure as well as all the atoms within 3Å of these residues were omitted prior to refinement and map calculation. The map was contoured at a level of 2.5σfor VP28. This figure was prepared using the program Bobscript (Esnouf 1997).

101

Figure 4.13 Simulated annealing Fo-Fc omit map showing the conserved

region of rVP26

All the residues shown in the figure as well as all the atoms within 3Å of these residues were omitted prior to refinement and map calculation. The map was contoured at a level of 2σfor rVP26. This figure was prepared using the program Bobscript (Esnouf 1997).

102

Furthermore, an interesting feature of these two proteins is their extreme opposite

charges on the surface, although the inner pore region of the β-barrel maintains a strong

hydrophobic environment with highly conserved residues lining the pore. The rVP26

trimer (crystallographic symmetry related molecules) depicts patches of positively

charged residues on the surfaces, whereas the rVP28 trimer depicts patches of negative

charged regions (Figure 4.14). It has been reported that VP28 will interact with VP26 by

far western blot analysis and pulldown assay (Xie et al. 2006; Xie and Yang 2006).

However, it is unclear that what these kinds of charge distribution contribute to the

function of the proteins such as the interaction between these envelope proteins or with

receptors on host cells.

Here we propose the trimeric VP26 and VP28 models along with the viral

envelope. The trimer arrangement of VP26 and VP28 matches well with the shape of the

spike-like structure in EM images after the treatment of Tween 20 (sees Figure 3.6 A-D,

as well as inserts Figure 3.6 D). It has been proven that viral structural proteins can exert

multiple functions by adopting different conformations (Hartlieb & Weissenhorn, 2006)

such as the Ebola virus matix protein VP40 was monomers when targeted to cellular

membranes but change its conformation to form hexamer via specific interactions of the

N-terminus (Nguyen et al., 2005). We propose that VP28/VP26 of WSSV could also

adopt a different conformation for different function, in which the trimer of VP28/VP26

may play an important role in membrane fusion during viral infection. However, the

questions that what is the factor to trigger the trimerization of these two proteins and

what is the role of monomeric protein are to be answered by further investigations.

103

Figure 4.14 The molecular surface representation for VP26 and VP28 that anchor the proposed viral envelope by the transmembrane (TM) domain.

VP26 trimer (crystallographic symmetry related molecules) depicting patches of positively charge surfaces. Whereas, VP28 trimer, part of one asymmetric unit depicting the negatively charged regions. These figures were prepared using the program GRASP (Nicholls et al. 1991). The immuno-electron microscopic image of WSSV stained against VP26 and VP28 shows the spike-like structure appearance for VP26 and VP28 after Tween20 treatment. Shape of the spike-like structure matches well with trimer arrangement of VP26 and VP28 (see Figure 3.6 A-D, as well as insert Figure 3.6 D).

TM

TM

104

4.3.2 Comparison with Other Viral Proteins

The β structure is one of the most common structures for viral structural protein

and has been found in various viral proteins in RNA viruses from plant, insect and animal

origins ((Rossmann and Johnson 1989), in double-stranded DNA viruses such as

adenovirus (Roberts et al. 1986), PRD1 (Benson et al. 1999) and SV40 (Stehle et al.

1996), and in dsRNA viruses such as blue tongue virus (Grimes et al. 1998) and

orthoreovirus (Reinisch et al. 2000). The β structures of VP26 and VP28 reveal

interesting structural similarities between these two proteins as well as with several

apparently unrelated virus types, although neither VP28 nor VP26 shares significant

sequence homology with known structural proteins from any other viruses.

However, a search of structural similarities for both full length rVP26 and rVP28

using the DALI (Holm and Sander 1995) database failed to recognize any significant

matches to any known protein structure. The DALI search shows that the highest

structural homology is with the gamma COP appendage domain of an intracellular

trafficking pathway protein (pdb code 1 pzd; rmsd is 3.3 Å for 87 Cα atoms of rVP28 and

89 Cα atoms of rVP26). The only viral protein structures that show partial structural

similarity were VP7 protein of Bluetongue virus (pdb code 1bvp; rmsd is 3.6 Å for 82 Cα

atoms of rVP26), A21 fragment of human coxsackievirus (pdb code 1z7z; rmsd is 3.6 Å

only for 72 Cα atoms of rVP28) and the capsid protein of carnation mottle virus (pdb

code 1opo; rmsd is 3.6 Å for only 64 Cα atoms of rVP28 and 3.7 Å for only 63 Cα atoms

of rVP26). Structurally the most common feature among these proteins is the presence of

105

a β-barrel structure, which is exposed on the outer surface. The topologies of these

proteins are, however, differenced from each other.

Unlike the predominant eight-stranded jelly roll topology, VP26 and VP28 has a

nine stranded ß-barrel that is folded into a novel topology and has not been seen in any

other virus up to date. The novelty of the structure of the major envelope proteins of

WSSV from any other virus to date therefore places WSSV outside of any of the

structurally linked groupings that have been identified so far. This unique feature of

WSSV is consistent with the phylogenetic tree of the WSSV DNA polymerase genes,

showing that WSSV is not related to any of the larger dsDNA virus family (Chen et al.

2002b). This further supports the proposition that WSSV should be a representative of a

new family (Nimavirida) under a new viral genus (Whispovirus).

106

4.3.3 Implications on Gene Duplication

VP24 is another important envelope protein of WSSV and interestingly it is also

highly homologous to VP26 and VP28 with 41% and 46% sequence similarity

respectively (Figure 4.15). Similar to VP26 and VP28, a highly hydrophobic region in the

N-terminus of VP24 might represent a transmembrane region. From the homolog of

VP26 and VP28 and the conservation of the sequence, we can hypothesize that VP24

adopts a nine-stranded β-barrel fold with a hydrophobic pore similar to that observed for

VP26 and VP28. However, the expression and purification trials on full-length VP24 or

the truncated form invariably resulted in the production of an insoluble protein.

Sequence comparison of VP24, VP26 and VP28 revealed that these three proteins

are highly related and most likely arise from by gene duplication. The structure similarity

of VP26 and VP28 further confirms this view. As the β sheet angle reflects packing of

the hydrophobic pore, it should also be conserved upon gene duplication. Conversely,

horizontal gene transfer would be less likely to yield a virus in which all three proteins

would possess the same β sheet angle. Thus we believe that the structures of VP26 and

VP28 bear evidence that support their evolution by gene duplication within the viral

genome. A similar example can be seen in the outer coat proteins P3-V1, P3-V2 and P5

of Bacteriophage PRD1. All these proteins have similar β barrel structure of same β

sheet angles and are believed to be evolved by gene duplication (Merckel et al. 2005).

107

Figure 4.15 Sequence Alignment for VP24, VP26 and VP28

The sequence alignment of VP24, VP26 and VP28 were analyzed by DNAman. The identity residues are highlighted in black. The residues with similar properties are highlighted in blue.

40VP28 40VP26 37VP24

Consensus

MM.

DE.

LF.

SGM

FNH

TLM

LTW

SNG

VLV

VDY

SVAa

AAAi

III

LILa

AAA

IIG

TLL

AST

VIL

IAI

AIL

VIV

FAV

ILI

VIS

IVI

FIV

RMV

YVT

HIN

NMI

TIE

VVL

TFN

KNK

TTK

IRL

EVD

TGK

HRK

80VP28 76VP26 77VP24

Consensus

T.D

D.K

NSD

IVA

EVY

TAP

NNV

MYE

DDS

EQE

NMI

LMI

RRN

IVL

PPT

VII

TQN

ARG

ERV

VAA

GKR

SVG

GMN

YSH

FIF

KRN

MGF

T.V

DEN

VRG

SST

FYL

DNQ

S.T

DTR

TPN

LLYg

GGGk

KKK

IVV

116VP28 112VP26 117VP24

Consensus

KAY

IMV

RKA

.

.G

NNQg

GGG

KLTs

SSSd

DDD

AKS

QDE

MML

KKV

EDK

EVK

DSK

AAGd

DDD

LLI

VVI

IIL

TST

PTS

VVL

ETL

GAG

RPD

.

.G

.

.D

.

.H

ART

LTL

EDN

VPV

TAN

VGK

GTA

QGE

NAS

LEK

155VP28 151VP26 157VP24

Consensus

TNE

FSL

ENE

GML

TTY

FLA

KKR

VIV

WLYn

NNNn

NNNt

TTT

SGK

RVR

KDD

ILI

NLT

IIV

TND

GDS

MIV

QTS

MVL

VRSp

PPP

KTG

IVL

NIN

PAA

.

.T

SGG

KNR

AIE

FKF

VGS

GNA

STN

SMK

NSF

TNV

193VP28 191VP26 195VP24

Consensus

STL

SYYf

FFF

TSK

PSP

VKT

SDV

IIL

DKK

ESK

DSN

.S.

.S.

EKR

VII

GTN

TLT

FIL

VDV

CVF

GCG

TSA

TKTf

FFF

GED

ADE

PGD

IAI

AAD

AFD

TET

AAN

GTR

GMH

NNY

LIL

FGL

DFS

MTM

YSR

204VP28 203VP26 207VP24

Consensus

VKF

HNS

VVP

TIG

YDN

SID

GKL

TDF

EEK

TIV

EKG

.KE

108

It is surprised that WSSV encodes three most abundant envelope proteins of

similar structure. One possible explanation is that these membrane anchor proteins can

aggregate on the surface of membrane with β-barrels in which there are no "edge" with

unfulfilled hydrogen bonding. The presence of similar hydrophobic barrel indicated that

these three proteins most probably are capable of forming homo- and heteromultimers. It

is recently reported that VP28 interacts with VP26 and VP24 to form a complex (Xie et al.

2006; Xie and Yang 2006). Intermolecular interactions between thee similar structures

may play a very important role in virus morphogenesis.

Another way to explain the highly similar structure to assume that these proteins

have been evolved with different functions in the WSSV virion such as VP26 was

previously reported to be capable of binding to actin or actin-associated proteins (Xie and

Yang 2005). Or they maybe related with infection to different host cell as the virus has a

broad range of hosts including salt and brackish water penaeid, crab, spiny lobster,

freshwater shrimps and crayfishes. However, whether these three proteins are just a

redundant copy of each other for backup or tave been evolved with functional divergence

is still unclear and needs further investigations.

109

4.3.4 Implications on membrane fusion

Viral structural proteins can exert multiple functions by adopting different

conformations (Hartlieb & Weissenhorn, 2006). For example, the Ebola virus matix

protein VP40 was shown to form monomers when targeted to cellular membranes but

change its conformation to form hexamer via specific interactions of the N-terminal

domain (Nguyen et al., 2005). We propose that VP28/VP26 of WSSV could also adopt a

different conformation for different function, in which the trimer of VP28/VP26 may

play an important role in membrane fusion during viral infection.

Enveloped viruses gain entry into the cytoplasm by the fusion of their lipid

envelope with the host cell membrane and the virus–induced membrane fusion is

triggered by conformational changes of viral envelope proteins (Da Poian et al. 2005). To

date, two classes of virus membrane-fusion proteins have been defined based on their key

structural features (Kielian 2006; Kielian and Rey 2006). Viruses that are currently

assigned to Class I or Class II are listed in Figure 4.16A. The Class I membrane-fusion

protein is mediated by the refolding of the fusion protein to a highly stable rod-like

structure with a central trimeric α-helical coiled coil. The Class II membrane-fusion

protein undergoes an oligomeric rearrangement during fusion, converting from the

metastable prefusion dimer to a considerably more stable homotrimer conformation. β

structure is the predominant secondary structure of Class II fusion protein. The details of

the structure features of the proteins in these two classes are list in Figure 4.16B. The

structures of some examples of these fusion proteins are shown in Figure 4.16C.

110

Figure 4.16 Fusion Proteins of Class I and Class II

A) Class I or Class II virus fusion proteins B) Comparison of Class I & Class II virus membrane fusion proteins. C) Structures of some fusion proteins. (Kielian and Rey 2006)

A

C

B

111

Though, VP26 and VP28 are not highly homologous to any of the viral envelope

fusion proteins, it retains the ß-barrel as well as trimer architecture similar to other viral

envelope fusion proteins such as E1 structural protein of Semliki Forest Virus (SFV)

(Gibbons et al. 2004). Previous study has shown that VP28 plays an important role in the

systemic WSSV infection in shrimp (Li et al. 2005a) and VP28 can bind to shrimp cells

as an attachment protein only in a low pH condition and can help the virus enter to the

cytoplasm (Yi et al. 2004). It has been well established that some of the enveloped

viruses enter the cells by endocytic pathway and this fusion depends on the acidification

of the endosomal compartment. More recently it has been shown that the interaction

between the host and WSSV is mediated through the host protein PmRab7 and VP28

(Sritunyalucksana et al. 2006). Rab proteins act as molecular switches to control

trafficking of endocytic vesicles within cells, as well as their subsequent fusion to

endosomes. Considering this we suggest that the fusion of WSSV viral envelope and the

host membrane through VP26 and VP28 is triggered by the exposure to low pH in the

endocytic pathway or the combination of receptor binding and low pH.

This completely different fusion mechanism also indicates that WSSV is a very

unique virus that is not closely related to any other virus, underscoring the distinct

taxonomic position of WSSV among invertebrate viruses. The many questions on the

fusion of WSSV will be very important topics to be answered in the future.

112

CHAPTER FIVE

5 General Conclusion and Future Studies

113

The characterization of viral structural proteins especially the envelope proteins is

of significant importance to study the taxonomy of the virus, its mechanism of infection

and also the assembly process of the mature virus. Due to the lack of a suitable shrimp

cell line and a low homology with other proteins of known function, we took the

structural approach to study WSSV envelope proteins.

A novel envelope protein, VP300, was identified by mass spectrometry. A

localization study using Western blotting and immuno-gold-labeling electron microscopy

are performed to confirm that VP300 localizes in WSSV envelope, not in the

nucleocapsids. The neutralization assay showed that anti-VP300 IgGs functions to delay

or neutralize the infection of WSSV to shrimp. Perhaps the antibody of VP300 could bind

to envelope spikes on the virion and prevent virus uncoating or the attachment of the

virus to the cell surface or virus uncoating. Further study on VP300 would be helpful to

realize the mechanism of the viral infection. Up to now, no efficient method has been

found to prevent the infection of shrimp by WSSV. Antibodies against VP300 could be

used directly as a passive immune strategy to control the WSSV infection and also might

be a choice for the virus control in the shrimp industry.

In this study, localization of VP26 with immuno-electron microscopy has also

been investigated. Compared with the highly confirmed envelope protein of VP28, the

location of VP26 in the virion is still unclear. VP26 was first reported as a nucleocapsid

protein (van Hulten et al. 2000), and later reported as an envelope protein (Zhang et al.

2002b). However, most recent publications claimed that VP26 is a linker (Xie and Yang

114

2005), or a tegument protein (Tsai et al. 2006). Clarification of the location of VP26

should improve our understanding on function of the protein. In this study we have first

used Tween 20 treatment to separate viral envelope from the nucleocapsid and then

localized VP26 by immunogold labeling. Both the VP26 and VP28 were clearly observed

on the outer surface of the virus envelope by electron microscopy and that is the very first

time we observed the spike-like structure in WSSV envelope.

As the continuation of our effort to understand the structure and function of

WSSV proteins, we determinate the crystal structure of two major envelop proteins VP26

and VP28 from WSSV, the first two structural envelop proteins to be reported from

WSSV for the first time. We have analyzed the three dimensional structures of VP26 and

VP28 and postulated that VP26 and VP28 might fuse with the envelop membrane by

their N-terminal transmembrane domains. A search for full length VP26 and VP28

structural similarities using the DALI (Holm and Sander 1995) database failed to

recognize any significant matches. Interestingly, VP26 and VP28 display strong

structural as well as significant sequence homology. Both proteins adopt the β-barrel

architecture with a protruding N-terminal region. The mushroom-like shape of the

trimeric VP26 and VP28 matches well with the spike structure we have observed in the

localization study. They are highly likely function to interact with host receptor proteins

or fuse with host cell membrane, representing a different model from the fusion proteins

of class I and class II.

115

This is the first report that structures of WSSV major envelope proteins. It greatly

inspires us to further determine the structure of other envelope proteins or nucleocapsid

proteins as they have important role in mechanism of viral morphogenesis and the

infection mechanism.

There are four major envelope proteins (VP19, VP24, VP26 and VP28) proteins

that consist almost 80-90% of the entire envelope. We have speculated that VP24 may

adopt a β-barrel structure with a hydrophobic pore similar to that of VP26 and VP28

because of their very high sequence homology. However, VP19, which almost constitutes

30% of the envelope, has no sequence similarities to proteins from WSSV or other

known viruses. Structural elucidation of VP19 will be extremely important for studying

the evolution of WSSV and also for the infection mechanism of this unique virus. A brief

attempt at the expression and purification trials of full length VP19 resulted in an

insoluble protein. Considering a predicted transmembrane region at the middle of

sequence, we will try to express the two sides of the fragment that may represent two

domains that are located outside and inside of the membrane respectively.

VP281, with a cell attachment RGD motif, is supposed to play an important role

in mediating WSSV infectivity (Huang et al. 2002a; Liang et al. 2005). As another

candidate for a structural genomics study, this protein has been successfully expressed

and purified as a soluble protein in the bacterial system as we mentioned in Chapter three.

However, we are still unable to grow any crystal of this protein from the crystallization

trials. We suspect that the failure of crystal growth is because of a probable floppy

116

domain in the protein that might cause aggregation. So in the future work, the following

approachs can be used to remove this floppy region. The first one is to construct different

truncated forms of VP281, such as to remove the N-terminus or C-terminus. The second

approach is to remove the flexible region by performing limited proteolysis. An

experiment using intact proteins and small amounts of exogenously added proteases can

be performed to isolate a fragment that will be more amenable to crystallization. The

ratio of protein to protease and the reaction time can be varied for limited proteolysis.

Once identified, the protease resistant fragment can be purified for crystallization.

More recently it has been shown that the interaction between the host and WSSV

is mediated through the host protein PmRab7 and VP28. Rab proteins are known to act as

molecular switches to control trafficking of endocytic vesicles within cells, as well as

their subsequent fusion to endosomes. It has been reported that once the interaction of

these two proteins was blocked by their specific antibodies the efficiency of the WSSV

infection were greatly reduced. Thus we can try to solve the complex structure of VP28

and PmRab7 to figure out the key residues which are involved in their interaction. Based

on these residues, peptides or small molecules can be designed to inhibit their interaction.

Besides the structure determination, an infection assay can be done to figure out

the key factors affecting the viral infection. Such as this assay can be done at various

different pH to prove the proposal that acidic pH is essential for viral infection of WSSV.

These experiments can also lead to the question of why pH will affect viral infection and

whether this is somehow related to the charge distribution on VP26 and VP28. Similar to

117

the neutralization study by antibodies, an experiment can be set up to use mutant VP26 or

VP28 to map out the host receptor binding region of these proteins. Purified wild type

protein should be able to neutralize WSSV infection as it should compete with the virus

to the host cell receptor. By mutating residues on the surface of the protein, the mutant

may lose the ability to inhibit WSSV infection. Those mutated residues can be identified

to be essential for interaction with the host receptor. However, the neutralization assay is

not that easy to be carried out because that it is very hard for us to select out the WSSV-

free shrimps or crayfishes for the assay. Majority of the shrimps or crayfishes we brought

for experiments are tested to be the carrier of the WSSV and unable to be used in the

neutralization assay. Therefore, we will continue to screen out the shrimp (crayfish/crab)

cell lines for in vitro reproduction of WSSV what is suitable for the infection assay.

In a conclusion, after the 3D structures of these envelope and nucleocapsid

proteins are solved, we can obtain more comprehensive information for further research

on host-virus interaction, the maturation and assembly of the virus particles and answer

many other important questions. Besides that, infection assays can be done to figure out

the key factors that affect viral infectivity of WSSV and select the inhibitors to block the

interaction to the host cell receptor that can greatly reduce the infection. Finally, our

future study should also extend towards drug design and vaccine development against

WSSV.

118

6 Bibliography

Abbott, A. (2002). "The society of proteins." Nature 417(6892): 894-6.

Aloy, P. and R. B. Russell (2002). "The third dimension for protein interactions and

complexes." Trends Biochem Sci 27(12): 633-8.

Aparicio, S., J. Chapman, E. Stupka, N. Putnam, J. M. Chia, P. Dehal, A.

Christoffels, S. Rash, S. Hoon, et al. (2002). "Whole-genome shotgun assembly and

analysis of the genome of Fugu rubripes." Science 297(5585): 1301-10.

Ashburner, M., C. A. Ball, J. A. Blake, D. Botstein, H. Butler, J. M. Cherry, A. P.

Davis, K. Dolinski, S. S. Dwight, et al. (2000). "Gene ontology: tool for the unification

of biology. The Gene Ontology Consortium." Nat Genet 25(1): 25-9.

Ataka, M. (1993). "Protein crystal growth: ab approach based on phase diagram

determination." Phase Transitions 45: 205-219.

Ban, N., P. Nissen, J. Hansen, P. B. Moore and T. A. Steitz (2000). "The complete

atomic structure of the large ribosomal subunit at 2.4 A resolution." Science 289(5481):

905-20.

Ben-Shem, A., F. Frolow and N. Nelson (2003). "Crystal structure of plant photosystem

I." Nature 426(6967): 630-5.

Benson, S. D., J. K. Bamford, D. H. Bamford and R. M. Burnett (1999). "Viral

evolution revealed by bacteriophage PRD1 and human adenovirus coat protein

structures." Cell 98(6): 825-33.

119

Bradley, P., K. M. Misura and D. Baker (2005). "Toward high-resolution de novo

structure prediction for small proteins." Science 309(5742): 1868-71.

Bravo, J. and P. Aloy (2006). "Target selection for complex structural genomics." Curr

Opin Struct Biol.

Brenner, S. E. (2001). "A tour of structural genomics." Nat Rev Genet 2(10): 801-9.

Brunger, A. T., P. D. Adams, G. M. Clore, W. L. DeLano, P. Gros, R. W. Grosse-

Kunstleve, J. S. Jiang, J. Kuszewski, M. Nilges, et al. (1998). "Crystallography &

NMR system: A new software suite for macromolecular structure determination." Acta

Crystallogr D Biol Crystallogr 54(Pt 5): 905-21.

Burton, D. R., R. A. Williamson and P. W. Parren (2000). "Antibody and virus:

binding and neutralization." Virology 270(1): 1-3.

Cen, F. (1998). "The existing condition and development strategy of shrimp culture

industry in China " In Y.Q.Su(ed.), The health culture of shrimps. China Ocean Press,

Beijing, People's Republic of China 32-38.

Chance, M. R., A. R. Bresnick, S. K. Burley, J. S. Jiang, C. D. Lima, A. Sali, S. C.

Almo, J. B. Bonanno, J. A. Buglino, et al. (2002). "Structural genomics: a pipeline for

providing structures for the biologist." Protein Sci 11(4): 723-38.

Chayen, N. E. (2004). "Turning protein crystallisation from an art into a science." Curr

Opin Struct Biol 14(5): 577-83.

Chen, L. L., J. H. Leu, C. J. Huang, C. M. Chou, S. M. Chen, C. H. Wang, C. F. Lo

and G. H. Kou (2002a). "Identification of a nucleocapsid protein (VP35) gene of shrimp

white spot syndrome virus and characterization of the motif important for targeting VP35

to the nuclei of transfected insect cells." Virology 293(1): 44-53.

120

Chen, L. L., C. F. Lo, Y. L. Chiu, C. F. Chang and G. H. Kou (2000). "Natural and

experimental infection of white spot syndrome virus (WSSV) in benthic larvae of mud

crab Scylla serrata." Dis Aquat Organ 40(2): 157-61.

Chen, L. L., H. C. Wang, C. J. Huang, S. E. Peng, Y. G. Chen, S. J. Lin, W. Y. Chen,

C. F. Dai, H. T. Yu, et al. (2002b). "Transcriptional analysis of the DNA polymerase

gene of shrimp white spot syndrome virus." Virology 301(1): 136-47.

Chen, X. F., C. Chen, D. H. Wu, H. Huai and X. C. Chi (1997). "A new baculovirus of

cultured shrimp." Sci. China Ser 40: 630-635.

Chou, H. Y., C. Y. Huang, C. H. Wang, H. C. Chiang and C. F. Lo (1995).

"Pathogenicity of a baculovirus infection causing white spot syndrome in cultured

penaeid shrimp in Taiwan." Dis. Aquat. Org. 23: 165-173.

Cramer, P., D. A. Bushnell and R. D. Kornberg (2001). "Structural basis of

transcription: RNA polymerase II at 2.8 angstrom resolution." Science 292(5523): 1863-

76.

Da Poian, A. T., F. A. Carneiro and F. Stauffer (2005). "Viral membrane fusion: is

glycoprotein G of rhabdoviruses a representative of a new class of viral fusion proteins?"

Braz J Med Biol Res 38(6): 813-23.

Ducruix, A. and R. Giege (1999). "Crystallisation of Nucleic Acids and Proteins. A

practical Approach, edn 2. Oxford: Oxford University Press."

Durand, S., D. V. Lightner, R. M. Redman and J. R. Bonami (1997). "Ultrastructure

and morphogenesis of white spot syndrome baculovirus (WSSV)." Dis Aquat Organ 29:

205-211.

121

Egea, P. F., S. O. Shan, J. Napetschnig, D. F. Savage, P. Walter and R. M. Stroud

(2004). "Substrate twinning activates the signal recognition particle and its receptor."

Nature 427(6971): 215-21.

Francki, R. I. B., C. M. Fanquet, D. L. Knudson and F. Brown (1991). "Classification

and nomenclature of viruses. Fifth report of the International Committee on Taxonomy of

Viruses." Arch Virol Suppl. 2: 1-450.

Gouet, P., E. Courcelle, D. I. Stuart and F. Metoz (1999). "ESPript: analysis of

multiple sequence alignments in PostScript." Bioinformatics 15(4): 305-8.

Green, E. D. (2001). "Strategies for the systematic sequencing of complex genomes."

Nat Rev Genet 2(8): 573-83.

Grimes, J. M., J. N. Burroughs, P. Gouet, J. M. Diprose, R. Malby, S. Zientara, P. P.

Mertens and D. I. Stuart (1998). "The atomic structure of the bluetongue virus core."

Nature 395(6701): 470-8.

Harlow, E. and D. Lane (1988). "Antibodies: A laboratory Manual." Cold Spring

Harbor, NY: Cold Spring Harbor Laboratory.: 553-612.

Holm, L. and C. Sander (1995). "Dali: a network tool for protein structure comparison."

Trends Biochem Sci 20(11): 478-80.

Huang, C., L. Zhang, J. Zhang, L. Xiao, Q. Wu, D. Chen and J. K. Li (2001).

"Purification and characterization of White Spot syndrome virus (WSSV) produced in an

alternate host: crayfish, Cambarus clarkii." Virus Res 76(2): 115-25.

Huang, C., X. Zhang, Q. Lin, X. Xu and C. L. Hew (2002a). "Characterization of a

novel envelope protein (VP281) of shrimp white spot syndrome virus by mass

spectrometry." J Gen Virol 83(Pt 10): 2385-92.

122

Huang, C., X. Zhang, Q. Lin, X. Xu, Z. Hu and C. L. Hew (2002b). "Proteomic

analysis of shrimp white spot syndrome viral proteins and characterization of a novel

envelope protein VP466." Mol Cell Proteomics 1(3): 223-31.

Huang, C. C. and Y. L. Song (1999). "Maternal transmission of immunity to white spot

syndrome associated virus (WSSV) in shrimp (Penaeus monodon)." Dev Comp Immunol

23(7-8): 545-52.

Inouye, K., S. Miwa, N. Oseko, H. Nakano and T. Kimura (1994). "Mass mortalities

of cultured kuruma shrimp, Penaeus japonicus, in Japan in 1993: electron microscopic

evidence of the causative virus." Fish Pathol. 29: 149-158.

Jensen, O. N., T. Houthaeve, A. Shevchenko, S. Cudmore, T. Ashford, M. Mann, G.

Griffiths and J. Krijnse Locker (1996). "Identification of the major membrane and core

proteins of vaccinia virus by two-dimensional electrophoresis." J Virol 70: 7485-7497.

Jones, T. A., J. Y. Zou, S. W. Cowan and M. Kjeldgaard (1991). "Improved methods

for building protein models in electron-density maps and the location of errors in these

models." Acta Crystallogr A 47: 110-119.

Kielian, M. (2006). "Class II virus membrane fusion proteins." Virology 344(1): 38-47.

Kielian, M. and F. A. Rey (2006). "Virus membrane-fusion proteins: more than one way

to make a hairpin." Nat Rev Microbiol 4(1): 67-76.

Knipe, D. M. and P. M. Howley (2001). "Fundamental Virology 4th Edition."

Lippincott Williams and Wilkins Press.

Koppers-Lalic, D., E. A. Reits, M. E. Ressing, A. D. Lipinska, R. Abele, J. Koch, M.

Marcondes Rezende, P. Admiraal, D. van Leeuwen, et al. (2005). "Varicelloviruses

123

avoid T cell recognition by UL49.5-mediated inactivation of the transporter associated

with antigen processing." Proc Natl Acad Sci U S A 102(14): 5144-9.

Kraulis, P. J. (1991). "MOLSCRIPT: a program to produce both detailed and schematic

plots of protein structures." J. Appl. Cryst. 24: 946-950.

Kuhn, P., K. Wilson, M. G. Patch and R. C. Stevens (2002). "The genesis of high-

throughput structure-based drug discovery using protein crystallography." Curr Opin

Chem Biol 6(5): 704-10.

Lander, E. S., L. M. Linton, B. Birren, C. Nusbaum, M. C. Zody, J. Baldwin, K.

Devon, K. Dewar, M. Doyle, et al. (2001). "Initial sequencing and analysis of the human

genome." Nature 409(6822): 860-921.

Laskowski, R. A., D. S. Moss and J. M. Thornton (1993). "Main-chain bond lengths

and bond angles in protein structures." J. Mol. Biol. 231: 1049-1067.

Leu, J. H., J. M. Tsai, H. C. Wang, A. H. Wang, C. H. Wang, G. H. Kou and C. F.

Lo (2005). "The unique stacked rings in the nucleocapsid of the white spot syndrome

virus virion are formed by the major structural protein VP664, the largest viral structural

protein ever found." J Virol 79(1): 140-9.

Li, H., Y. Zhu, X. Xie and F. Yang (2006). "Identification of a novel envelope protein

(VP187) gene from shrimp white spot syndrome virus." Virus Res 115(1): 76-84.

Li, H. X., X. L. Meng, J. P. Xu, W. Lu and J. Wang (2005a). "Protection of crayfish,

Cambarus clarkii, from white spot syndrome virus by polyclonal antibodies against a

viral envelope fusion protein." J Fish Dis 28(5): 285-91.

124

Li, L., X. Xie and F. Yang (2005b). "Identification and characterization of a prawn

white spot syndrome virus gene that encodes an envelope protein VP31." Virology

340(1): 125-32.

Li, Q., Y. Chen and F. Yang (2004). "Identification of a collagen-like protein gene from

white spot syndrome virus." Arch Virol 149(2): 215-23.

Liang, X., B. Chow, C. Raggo and L. A. Babiuk (1996). "Bovine herpesvirus 1 UL49.5

homolog gene encodes a novel viral envelope protein that forms a disulfide-linked

complex with a second virion structural protein." J Virol 70(3): 1448-54.

Liang, Y., J. Huang, X. L. Song, P. J. Zhang and H. S. Xu (2005). "Four viral proteins

of white spot syndrome virus (WSSV) that attach to shrimp cell membranes." Dis Aquat

Organ 66(1): 81-5.

Liu, Z., H. Yan, K. Wang, T. Kuang, J. Zhang, L. Gui, X. An and W. Chang (2004).

"Crystal structure of spinach major light-harvesting complex at 2.72 A resolution."

Nature 428(6980): 287-92.

Lo, C. F., C. H. Ho, C. H. Chen, K. F. Liu, Y. L. Chiu, P. Y. Yeh, S. E. Peng, H. E.

Hsu, H. C. Liu, et al. (1997). "Detection and tissue tropism of white spot syndrome

baculovirus (WSBV) in captured brooders of Penaeus monodon with a special emphasis

on reproductive organs." Dis Aquat Organ 30: 53-72.

Lo, C. F., C. H. Ho, S. E. Peng, C. H. Chen, H. E. Hsu, Y. L. Chiu, C. F. Chang, K. F.

Liu, M. S. Su, et al. (1996). "White spot syndrome associated virus (WSBV) detected in

cultured and captured shrimp, crabs and other arthropods." Dis. Aquat. Org. 27: 215-225.

Lu, C. P., S. Zhu, F. S. Guo and S. Y. Wu (1997). "Electron microscopic observation

on a non-occluded baculo-like virus in shrimps." Arch Virol 142(10): 2073-8.

125

Luft, J. R., R. J. Collins, N. A. Fehrman, A. M. Lauricella, C. K. Veatch and G. T.

DeTitta (2003). "A deliberate approach to screening for initial crystallization conditions

of biological macromolecules." J Struct Biol 142(1): 170-9.

Marsden, R. L., J. A. Ranea, A. Sillero, O. Redfern, C. Yeats, M. Maibaum, D. Lee,

S. Addou, G. A. Reeves, et al. (2006). "Exploiting protein structure data to explore the

evolution of protein function and biological complexity." Philos Trans R Soc Lond B

Biol Sci 361(1467): 425-40.

McPherson, A. (1999). "Crystallization of biological Macromolecules. Cold Spring

Harbor: Cold Spring Harbor Laboratory press."

Merckel, M. C., J. T. Huiskonen, D. H. Bamford, A. Goldman and R. Tuma (2005).

"The structure of the bacteriophage PRD1 spike sheds light on the evolution of viral

capsid architecture." Mol Cell 18(2): 161-70.

Merrit, E. A., and Bacon, D. J. (1997). Methods Enzymol. 277: 505-524.

Momoyama, K., M. Hiraoka, H. Nakano, H. Koube, K. Inouye and N. Oseko (1994).

"Mass mortalities of cultured kuruma shrimp, Penaeus japonicus, in Japan in 1993:

histopathological study." Fish Pathol. 29: 141-148.

Otwinowski, Z. and W. Minor (1997). "Processing of X-ray diffraction data collected in

oscillation mode." Methods Enzymol. 276: 307-326.

Reinisch, K. M., M. L. Nibert and S. C. Harrison (2000). "Structure of the reovirus

core at 3.6 A resolution." Nature 404(6781): 960-7.

Rhodes, G. (2000). "crystallography made crystal clear, second edition. Academic

Press."

126

Roberts, M. M., J. L. White, M. G. Grutter and R. M. Burnett (1986). "Three-

dimensional structure of the adenovirus major coat protein hexon." Science 232(4754):

1148-51.

Rossmann, M. G. and J. E. Johnson (1989). "Icosahedral RNA virus structure." Annu

Rev Biochem 58: 533-73.

Sambrook, J. and D. W. Russell (2001). "Molecular Cloning: A Laboratory Manual

(3rd Edition)." Cold Spring Harbor Laboratory.

Spahn, C. M., R. Beckmann, N. Eswar, P. A. Penczek, A. Sali, G. Blobel and J.

Frank (2001). "Structure of the 80S ribosome from Saccharomyces cerevisiae--tRNA-

ribosome and subunit-subunit interactions." Cell 107(3): 373-86.

Stehle, T., S. J. Gamblin, Y. Yan and S. C. Harrison (1996). "The structure of simian

virus 40 refined at 3.1 A resolution." Structure 4(2): 165-82.

Terwilliger, T. C. (2000). "Maximum-likelihood density modification." Acta Crystallogr

D Biol Crystallogr 56(Pt 8): 965-72.

Terwilliger, T. C. and J. Berendzen (1999). "Automated MAD and MIR structure

solution." Acta Crystallogr D Biol Crystallogr 55(Pt 4): 849-61.

Tsai, J. M., H. C. Wang, J. H. Leu, H. H. Hsiao, A. H. Wang, G. H. Kou and C. F.

Lo (2004). "Genomic and proteomic analysis of thirty-nine structural proteins of shrimp

white spot syndrome virus." J Virol 78(20): 11360-70.

Tsai, J. M., H. C. Wang, J. H. Leu, A. H. Wang, Y. Zhuang, P. J. Walker, G. H. Kou

and C. F. Lo (2006). "Identification of the nucleocapsid, tegument, and envelope

proteins of the shrimp white spot syndrome virus virion." J Virol 80(6): 3021-9.

127

van Hulten, M. C., M. Westenberg, S. D. Goodall and J. M. Vlak (2000).

"Identification of two major virion protein genes of white spot syndrome virus of

shrimp." Virology 266(2): 227-36.

van Hulten, M. C., J. Witteveldt, S. Peters, N. Kloosterboer, R. Tarchini, M. Fiers,

H. Sandbrink, R. K. Lankhorst and J. M. Vlak (2001a). "The white spot syndrome

virus DNA genome sequence." Virology 286(1): 7-22.

van Hulten, M. C., J. Witteveldt, M. Snippe and J. M. Vlak (2001b). "White spot

syndrome virus envelope protein VP28 is involved in the systemic infection of shrimp."

Virology 285(2): 228-33.

Walter, T. S., J. M. Diprose, C. J. Mayo, C. Siebold, M. G. Pickford, L. Carter, G. C.

Sutton, N. S. Berrow, J. Brown, et al. (2005). "A procedure for setting up high-

throughput nanolitre crystallization experiments. Crystallization workflow for initial

screening, automated storage, imaging and optimization." Acta Crystallogr D Biol

Crystallogr 61(Pt 6): 651-7.

Wang, C. H., C. F. Lo, J. H. Leu, C. M. Chou, P. Y. Yeh, M. C. Tung, C. F. Chang,

M. S. Su and G. H. Kou (1995). "Purification and genomic analysis of baculovirus

associated with white spot syndrome (WSBV) of Penaeus monodon." Dis Aquat Organ

23: 239-242.

Westenberg, M., B. Heinhuis, D. Zuidema and J. M. Vlak (2005). "siRNA injection

induces sequence-independent protection in Penaeus monodon against white spot

syndrome virus." Virus Res 114(1-2): 133-9.

Westermeier, R. N., Tom (2002). Proteomics in Practice. A Laboratory Manual of

Proteome Analysis, Wiley-VCH, Weinheim.

128

Wommack, K. E. and R. R. Colwell (2000). "Virioplankton: viruses in aquatic

ecosystems." Microbiol Mol Biol Rev 64(1): 69-114.

Wongteerasupaya, C., J. E. Vickers, S. Sriurairatana, G. L. Nash, A. Akarajamorn,

V. Boonsaeng, S. Panyim, A. Tassanakajon, B. Withyachumnarnkul, et al. (1995).

"A non-occluded, systemic baculovirus that occurs in cells of ectodermal and

mesodermal origin and causes high mortality in black tiger prawn Penaeus monodon "

Dis Aquat Organ 21: 69-77.

Wu, W., L. Wang and X. Zhang (2005). "Identification of white spot syndrome virus

(WSSV) envelope proteins involved in shrimp infection." Virology 332(2): 578-83.

Xie, X., L. Xu and F. Yang (2006). "Proteomic analysis of the major envelope and

nucleocapsid proteins of white spot syndrome virus (WSSV)." J Virol.

Xie, X. and F. Yang (2005). "Interaction of white spot syndrome virus VP26 protein

with actin." Virology 336(1): 93-9.

Xie, X. and F. Yang (2006). "White spot syndrome virus VP24 interacts with VP28 and

is involved in virus infection." J Gen Virol 87(Pt 7): 1903-8.

Xu, Y. X., X. F. Wu, Y. F. Zhu, Z. R. Xu and W. D. Shen (2005). "[The expression of

VP19 gene from prawn white spot syndrome virus in silkworm, Bombyx mori using host

range-expanded HyNPV]." Sheng Wu Gong Cheng Xue Bao 21(5): 837-9.

Yaffe, M. B. (2005). "X-ray crystallography and structural biology." Crit Care Med

33(12 Suppl): S435-40.

Yang, F., J. He, X. Lin, Q. Li, D. Pan, X. Zhang and X. Xu (2001). "Complete genome

sequence of the shrimp white spot bacilliform virus." J Virol 75(23): 11811-20.

129

Yang, F., W. Wang, R. Z. Chen and X. Xu (1997). "A simple and efficient method for

purification of prawn baculovirus DNA." J Virol Methods 67(1): 1-4.

Yi, G., Z. Wang, Y. Qi, L. Yao, J. Qian and L. Hu (2004). "Vp28 of shrimp white spot

syndrome virus is involved in the attachment and penetration into shrimp cells." J

Biochem Mol Biol 37(6): 726-34.

Yoganandhan, K., S. Syed Musthaq, R. B. Narayanan and A. S. Sahul Hameed

(2004). "Production of polyclonal antiserum against recombinant VP28 protein and its

application for the detection of white spot syndrome virus in crustaceans." J Fish Dis

27(9): 517-22.

Yu, J., S. Hu, J. Wang, G. K. Wong, S. Li, B. Liu, Y. Deng, L. Dai, Y. Zhou, et al.

(2002). "A draft sequence of the rice genome (Oryza sativa L. ssp. indica)." Science

296(5565): 79-92.

Zhang, X., C. Huang, X. Tang, Y. Zhuang and C. L. Hew (2004). "Identification of

structural proteins from shrimp white spot syndrome virus (WSSV) by 2DE-MS."

Proteins 55(2): 229-35.

Zhang, X., C. Huang, X. Xu and C. L. Hew (2002a). "Identification and localization of

a prawn white spot syndrome virus gene that encodes an envelope protein." J Gen Virol

83(Pt 5): 1069-74.

Zhang, X., C. Huang, X. Xu and C. L. Hew (2002b). "Transcription and identification

of an envelope protein gene (p22) from shrimp white spot syndrome virus." J Gen Virol

83(Pt 2): 471-7.

Zhu, Y. B., H. Y. Li and F. Yang (2006). "Identification of an envelope protein (VP39)

gene from shrimp white spot syndrome virus." Arch Virol 151(1): 71-82.

130

7 Appendices

A. Media and solutions

LB broth (per liter) To 950 mL of MQ H2O, add: Bacto-tryptone 10 g Bacto-yeast extract 5 g NaCl 10 g Dissolve solutes. Adjust pH to 7.0 with 1 M NaOH. Adjust volume to 1L with

MQ H2O. Sterilize by autoclaving. 2 × YT medium (per liter) To 950 mL of MQ H2O, add: Bacto-tryptone 16 g Bacto-yeast extract 10 g NaCl 5 g Dissolve solutes. Adjust pH to 7.0 with 1 M NaOH. Adjust volume to 1L with

MQ H2O. Sterilize by autoclaving. SOC medium (per liter) To 950 mL of MQ H2O, add: Bacto-tryptone 20 g Bacto-yeast extract 5 g NaCl 0.58 g Dissolve solutes. Add 10 mL of 250 mM KCl. Adjust pH to 7.0 with 1 M NaOH.

Adjust volume to 980 mL with MQ water. Sterilize by autoclaving. Allow to cool to 60°C or less, and then add 20 mL of sterile 1M glucose. Add 10 mL of sterile 1 M MgCl2 just prior to use.

Tfb I (per liter) To 950 mL of MQ H2O, add: CH3COOK 2.94 g RbCl 12.1 g

131

CaCl2·2H2O 14.7 g MnCl2·4H2O 9.9 g Glycerol 150 mL Adjust pH to 5.8 with dilute acetic acid, add MQ H2O to 1 L and autoclave. Tfb II (per liter) To 950 mL of MQ H2O, add: MOPS 2.1 g CaCl2 11.1 g RbCl 1.21 g Glycerol 150 mL Adjust pH to 6.5 with 1 M NaOH, add MQ H2O to 1 L and autoclave. 10 × TNE buffer (per liter) To 950 mL of MQ H2O, add: Tris 6.05 g NaCl 5.85 g EDTA 0.292 g Adjust pH to 7.4 with 1 M HCl and add MQ H2O to 1 L. 10 × TE buffer (per liter) To 950 mL of MQ H2O, add: Tris 6.05 g EDTA 0.292 g Adjust pH to 7.4 with 1 M HCl and add MQ H2O to 1 L. 10 × TN buffer (per liter) To 950 mL of MQ H2O, add: Tris 6.05 g NaCl 9 g Adjust pH to 7.4 with 1 M HCl and add MQ H2O to 1 L.

132

B. Restriction enzymes All restriction enzymes were bought from New England Biolabs (NEB). BamH I NEB, #R0136L EcoR I NEB, #R0101L Hind III NEB, #R0104L Xho I NEB, #R0146L C. Other enzymes Protease K Sigma, #P2308 T4 DNA polymerase NEB, #M0203S T4 DNA ligase NEB, #M0202L T4 polynucleotide kinase Promega, #M4103 DNA Taq polymerase Promega, #M1665 RNase-free DNase I QIAGEN, #79254 Sequencing grade modified trypsin Promega, #V511A Trypsin-EDTA GIBCO, #25200-072 D. Antibiotics Ampicillin Sigma, #A-6140 E. Kits High pure PCR product purification kit Roche, #11732668001 Rapid DNA ligation kit Roche, #11635379001 QIAprep spin miniprep kit QIAGEN, #27106 QIAquick gel extraction kit QIAGEN, #28706 RNeasy mini kit QIAGEN, #74106 OneStep RT-PCR kit QIAGEN, #210212 BigDye® terminator v3.1 cycle sequencing kit Applied Biosystems, #4337455 Hi-Di formamide Applied Biosystems, #4311320 RC DC protein assay kit Bio-Rad, #500-0119 F. Crystallization screen kits Index Hampton Research, HR2-144 PEG/Ion Screen Hampton Research, HR2-126 Crystal Screen Kit Hampton Research, HR2-110 Crystal Screen 2 Hampton Research, HR2-112

133

Natrix Hampton Research, HR2-116 MembFac kit Hampton Research, HR2-114 Additive Screen Hampton Research, HR2-428 G. Equipments High Speed Centrifuge Beckman, J2-21 Ultracentrifuge Beckman, XL-90 MicroPulser® Electroporator Bio-Rad Thermal Cycler Bio-Rad ABI PRISM™ 3100 Genetic Analyzer Applied Biosystems Voyager-DE™ STR BioSpectrometry Workstation Applied Biosystems 4700 Proteomics Analyzer with TOF/TOF™ optics Applied Biosystems Ultimate™ LC system Dionex LC Packings

134

8 List of Publications Publications: Xiaobo Zhang, Canhua Huang, Xuhua Tang, Ying Zhuang, Choy Leong Hew

Identification of structural proteins from shrimp white spot syndrome virus (WSSV) by 2DE-MS. Proteins: Structure, Function, and Bioinformatics, Volume 55, Issue 2, Pages 229-235 (1 May 2004)

Xuhua Tang, Jinlu Wu, Jayaraman Sivaraman, Choy Leong Hew

Crystal Structure of two major envelope proteins from shrimp white spot syndrome virus (WSSV). Journal of Virology.(accepted, under revision)

Xuhua Tang, Jayaraman Sivaraman, Choy Leong Hew

Expression, purification and crystallization of two major envelope proteins from white spot syndrome virus at 2.0 and 2.2Å resolution (submitted)

Abstract Publications: Xuhua Tang, Jayaraman Sivaraman, choy-leong Hew

X-ray crystal structural studies of VP26 of WSSV. FASEB Journal, 20(4), Part 1 (2006): A489.

Xuhua Tang, Jayaraman Sivaraman, choy-leong Hew Structural studies of envelop proteins of white spot syndrome virus. FASEB Journal, 19(4), Part 1 Suppl. S (2005): A312-A313.

Poster Presentations on Conferences:

1. X-ray Crystal Structural Studies of VP26 of WSSV Xuhua Tang, Jayaraman Sivaraman, choy-leong Hew

Experimental Biology 2006, April 1-5, 2006, San Francisco, U.S.A **Graduate Travel Award sponsored by ASBMB (American Society for Biochemistry and

Molecular Biology) 2. Structural studies of envelope proteins of white spot syndrome virus

Xuhua Tang, Jayaraman Sivaraman, choy-leong Hew Experimental Biology 2005, April 2-6, 2005, San Diego, U.S.A **Graduate Travel Award sponsored by ASBMB (American Society for Biochemistry and

Molecular Biology)

135

3. Crystal Structure of envelope protein VP26 of white spot syndrome virus

Xuhua Tang, Jayaraman Sivaraman, choy-leong Hew Biology in Asia International Conference, Dec 7-10, 2004, Nanyang Technological

University, Singapore **Graduate Conference Fellowship sponsored by Department of Biological Sciences, NUS

4. Identification and Structure Determination of envelope proteins of WSSV Xuhua Tang, Canhua Huang, choy-leong Hew

The 10th SCBA (International Symposium, July 18 -23, 2005, Beijing, China

5. Structural studies of envelope proteins of whit spot syndrome virus Xuhua Tang, Jayaraman Sivaraman, choy-leong Hew Third International Conference on Structural Biology & Functional Genomics, Dec 2004, Singapore

6. Functional and structural Genomics Studies of Marine Viral pathogens.

Xuhua Tang, Wenjun Song, Yang Liu, Jing Chen, Ying Zhuang and Choy Leong Hew International Conference on Marine Science and Technology, May 13-14, 2004, Taiwan

7. Structural Genomics Studies of Shrimp White Spot Syndrome Virus. Choy Leong Hew, Yang Liu, Xuhua Tang, Ying Zhuang, Canhua Huang, Zhengjun Li, Jianxing Song,

World Aquaculture Society, Hawaii, U.S.A. March 2004

8. Structural Genomics Studies of Shrimp White Spot Syndrome Virus. Yang Liu, Xuhua Tang, Ying Zhuang, Canhua Huang, Zhengjun Li, Jianxing Song, Choy-Leong Hew

Asian Pacific Aquaculture, Bangkok, September 2003

9. Structural Determination of the envelope proteins from the shrimp white spot syndrome virus.

Xuhua Tang and Choy Leong Hew The 4th Sino-Singapore Conference in Biotechnology, 11-13 Nov 2003, Singapore

10. Structural Determination and characterization of the envelope proteins from the shrimp white spot syndrome virus.

Xuhua Tang and Choy Leong Hew 8th Biological Science Graduate Congress, Department of Biological Sciences, NUS,

3-6 Dec, 2003

136