165
i Nuclear Shuttling Proteins of Paramyxoviruses: Identification of Previously Unknown Function of Respiratory Syncytial Virus Matrix Protein in Viral Proliferation and Potential Localisation Signals in Human Metapneumovirus Nucleocapsid Protein A thesis submitted for the Degree of Master in Applied Science (Research) October, 2012 Wahyu Nawang Wulan Biomedical Sciences Faculty of Applied Science University of Canberra Canberra, Australia

Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

Embed Size (px)

Citation preview

Page 1: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

i

Nuclear Shuttling Proteins of

Paramyxoviruses: Identification of

Previously Unknown Function of

Respiratory Syncytial Virus Matrix

Protein in Viral Proliferation and Potential

Localisation Signals in Human

Metapneumovirus Nucleocapsid Protein

A thesis submitted for the

Degree of Master in Applied Science (Research)

October, 2012

Wahyu Nawang Wulan

Biomedical Sciences

Faculty of Applied Science

University of Canberra

Canberra, Australia

Page 2: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

iii

Abstract

Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) are the major

causes of viral lower respiratory tract infections (LRTIs). There are currently no licensed

vaccines or antivirals for RSV and hMPV; and so research is directed towards the

development of live-attenuated vaccines. One pathway to attenuation is to disrupt the

nucleocytoplasmic transport of structural proteins of a virus. The nuclear localisation of a

structural protein is a strategy used by many viruses to inhibit host antiviral responses.

RSV matrix (M) protein shuttles between the cytoplasm and the nucleus, with nuclear

localisation occurring early in infection (around 18 – 24 hours post infection) and

cytoplasmic localisation later. The disruption of RSV M nuclear import decreases the titre of

infectious particles while the disruption of nuclear export ceases the production of infectious

particles. The nucleocytoplasmic transport of RSV M is regulated by CK2 phosphorylation

although the detailed mechanism is not well understood. RSV M(205) (T), which is adjacent

to the nuclear export signal, has been predicted as a putative regulatory site. The

nucleocytoplasmic transport of hMPV nucleocapsid (N) protein, however, has never been

described. The hMPV N has been shown to localise in the nucleus late in the replication cycle

(day 5 post infection), but further research is required to fully understand the mechanism.

Published literature (RSV M) and unpublished results (hMPV N) from the Ghildyal group led

to a hypothesis that disruption of the nucleocytoplasmic transport of a viral structural protein

can lead to attenuation. In this study, two hypotheses were tested: (1) mutation in RSV M205

reduces the production of infectious progeny and the induction of pro-inflammatory

responses; and (2) the hMPV N has inherent nucleocytoplasmic transport ability. The

hypotheses were tested in two aims. Firstly, to investigate the role of the RSV M(205) (T) in

virus growth and induction of pro-inflammatory response in Vero E6 and A549 cells; and

secondly, to investigate the nucleocytoplasmic transport ability of hMPV N in transfected

Cos-7 cells.

To understand the role of RSV M(205) (T), cells were infected with recombinant RSV strain

A2 having threonine (T) substitution to alanine (A) at M205 (rRSV A2 M(T205A)). Viral

titres and expression of IL-8 and RANTES were determined using plaque assay and ELISA,

Page 3: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

iv

following single cycle and multiple cycle replication assays. To define hMPV N

nucleocytoplasmic transport, full-length and truncated hMPV N constructs fused to green

fluorescent protein were expressed in transfected Cos-7 cells.

rRSV A2 M(T205A) was found to be capable of infecting Vero E6 and A549 cells, but was

unable to transmit the infection between cells. The inability to spread leads to a reduced

induction of IL-8 and RANTES expression, which was significantly suppressed in the

interferon (IFN)-α/β-producing A549 cells, compared to the wild type rRSV A2 M205 (T).

hMPV N was found to have inherent ability of nucleocytoplasmic transport, with a potential

nuclear export signal (NES) identified at amino acid 1 – 15 and a region containing a nuclear

localisation signal (NLS) identified at amino acid 192 – 250.

This study shows for the first time that the RSV M(205) (T) is an important site for the

success of transmission of infection and RSV M has a role in the suppression of IFN-α/β.

This study was also the first to show that hMPV N is capable of nucleocytoplasmic transport,

with potential nuclear transport motifs have been identified. By describing the

nucleocytoplasmic transport function of the RSV M and hMPV N proteins, this study

contributes to attempts to develop live-attenuated vaccines for RSV and hMPV.

Page 4: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

v

Table of Contents

Abstract………………………………………………………………………………………………………..iii

Table of Contents………………………………………………………………………………..…………..v

List of Tables and Figures………………………………………………………………….……………..ix

Form B – Certificate of Authorship of Thesis…………………………………………………...……xi

Acknowledgement………………………………………………………………….………………………xiii

List of Abbreviation………………………………………………...……………………………..………xv

1 Introduction .........................................................................................................................1

1.1 Viral respiratory tract infections ....................................................................................3 1.2 Respiratory syncytial virus ............................................................................................3

1.2.1 Epidemiology................................................................................................... 4 1.2.2 Clinical disease ................................................................................................ 4

1.2.3 Taxonomy ........................................................................................................ 5 1.2.4 The structure of an RSV particle (the virion) .................................................... 6

1.2.5 The life cycle of RSV....................................................................................... 8 1.3 The nucleocytoplasmic transport of viral proteins .........................................................9

1.3.1 Overview ......................................................................................................... 9 1.3.2 The components of nucleocytoplasmic transport ............................................ 10

1.3.3 The mechanism of nuclear import .................................................................. 13 1.3.4 The mechanism of nuclear export ................................................................... 14

1.3.5 Regulation of nucleocytoplasmic transport by phosphorylation ...................... 14 1.3.6 Regulated nucleocytoplasmic transport of viral proteins ................................. 17

1.4 The nucleocytoplasmic transport of the matrix (M) protein of RSV ............................. 19 1.4.1 Overview of the M protein of RSV ................................................................ 19

1.4.2 The nucleocytoplasmic transport of the M protein in infected cells................. 20 1.5 Regulation of cellular innate immunity against RSV infection ..................................... 22

1.5.1 Overview ....................................................................................................... 22 1.5.2 Innate immune responses against RSV infection ............................................ 23

1.5.3 Activation of NF-κB transcription factor ........................................................ 24 1.5.4 Activation of IRF3 transcription factor ........................................................... 25

1.6 RSV vaccine ............................................................................................................... 26 1.7 Human metapneumovirus (hMPV) .............................................................................. 28

1.7.1 Epidemiology................................................................................................. 28 1.7.2 hMPV infection ............................................................................................. 29

1.7.3 Taxonomy ...................................................................................................... 30 1.7.4 The structure of an hMPV particle (the virion) ............................................... 30

1.7.5 The life cycle of hMPV .................................................................................. 32 1.7.6 The role of N protein in virus replication ........................................................ 32

1.7.7 The nucleocytoplasmic transport of nucleocapsid proteins ............................. 34 1.8 Hypotheses and aims ................................................................................................... 36

2 Materials and Methods ....................................................................................................... 39 2.1 Materials ..................................................................................................................... 41

2.1.1 Cell lines ........................................................................................................ 41 2.1.2 Viruses........................................................................................................... 41

2.1.3 Plasmids and bacterial cultures....................................................................... 41 2.1.4 Primers .......................................................................................................... 42

2.1.5 Enzymes/buffers for molecular biology assays ............................................... 43 2.1.6 Antibodies ..................................................................................................... 43

Page 5: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

vi

2.1.7 Kits ................................................................................................................ 43 2.1.8 Buffers, media, and solutions ......................................................................... 44

2.2 Methods ...................................................................................................................... 46 2.2.1 Cell culture .................................................................................................... 46

2.2.2 Virus propagation .......................................................................................... 48 2.2.3 Replication kinetics assay .............................................................................. 51

2.2.4 Enzyme-linked immunofluorescence assay (ELISA) ...................................... 52 2.2.5 hMPV N constructs ........................................................................................ 53

2.2.6 Cloning of hMPV N gene using GatewayTM

Technology ............................... 55 2.2.7 Confirmation of the hMPV N gene inserted into vectors using PCR and DNA

sequencing method .............................................................................................................. 57 2.2.8 Measurement of nucleic acid concentration .................................................... 59

2.2.9 Agarose gel electrophoresis ............................................................................ 59 2.2.10 Plasmid storage in glycerol solution ............................................................... 59

2.2.11 hMPV N protein expression ........................................................................... 60 2.2.12 Bacterial culture ............................................................................................. 61

3 Replication Efficiency of RSV in Cell Culture ................................................................... 63 3.1 Introduction ................................................................................................................ 65

3.2 Methods ...................................................................................................................... 66 3.2.1 Cells and virus ............................................................................................... 66

3.2.2 Single cycle growth curve .............................................................................. 67 3.2.3 Multiple cycle growth curve ........................................................................... 67

3.2.4 Data analysis .................................................................................................. 67 3.2.5 Confirmation of the stability of rRSV M constructs ........................................ 68

3.3 Results ........................................................................................................................ 68 3.3.1 Single cycle replication kinetics ..................................................................... 68

3.3.2 Multiple cycle replication kinetics .................................................................. 72 3.3.3 Confirmation of sequence of rRSV A2 and rRSV A2 M(T205A) ................... 74

3.4 Discussion................................................................................................................... 76 3.5 Summary .................................................................................................................... 78

4 Pro-Inflammatory Response in RSV Infection .................................................................... 79 4.1 Introduction ................................................................................................................ 81

4.2 Methods ...................................................................................................................... 82 4.2.1 Cells and virus ............................................................................................... 82

4.2.2 Single cycle replication assay ......................................................................... 82 4.2.3 Multiple cycle replication assay ..................................................................... 83

4.2.4 Enzyme-linked immunosorbent assay (ELISA) .............................................. 83 4.2.5 Data analysis .................................................................................................. 84

4.3 Results ........................................................................................................................ 84 4.3.1 Single cycle replication assay ......................................................................... 84

4.3.2 Multiple cycle replication assay ..................................................................... 88 4.4 Discussion................................................................................................................... 92

4.5 Summary .................................................................................................................... 94 5 The Subcellular Localisation of hMPV N Protein in Transfected Cells ............................... 95

5.1 Introduction ................................................................................................................ 97 5.2 Methods ...................................................................................................................... 99

5.2.1 Generation of constructs and recombination cloning ...................................... 99 5.2.2 Transient transfection into Cos-7 cells.......................................................... 101

5.2.3 Confocal laser scanning microscopy (CLSM) .............................................. 103

Page 6: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

vii

5.3 Results ...................................................................................................................... 104 5.3.1 The generation of hMPV N constructs ......................................................... 104

5.4 The GatewayTM

recombination cloning (RC) technology system ............................... 107 5.4.1 BP recombination reaction ........................................................................... 107

5.4.2 LR recombination reaction ........................................................................... 109 5.5 Expression of hMPV N constructs in transfection cells .............................................. 111

5.5.1 Expression of full-length hMPV N (1-394) observed in live Cos-7 cells ....... 111 5.5.2 Expression of hMPV N deleted constructs observed in Cos-7 cells............... 113

5.6 Discussion................................................................................................................. 115 6 General Discussion .......................................................................................................... 119

6.1 Respiratory syncytial virus ........................................................................................ 121 6.2 Human metapneumovirus.......................................................................................... 124

6.3 Conclusion ................................................................................................................ 125 7 Reference List .................................................................................................................. 127

8 Appendix ......................................................................................................................... 147

Page 7: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

ix

List of Tables and Figures

Figure 1-1 Syncytia. ............................................................................................................. 5

Figure 1-2 The structure of respiratory syncytial virus .......................................................... 7 Figure 1-3 The life cycle of respiratory syncytial virus.. ....................................................... 9

Figure 1-4 Overview of a nuclear pore complex (NPC).. .................................................... 10 Table 2-1 Cell lines used in this study ................................................................................. 41

Table 2-2 Viruses used in this study .................................................................................... 41 Table 2-3 Plasmid/bacteria used in this study ...................................................................... 41

Table 2-4 Primers used in this study .................................................................................... 42 Table 2-5 Enzymes used in this study .................................................................................. 43

Table 2-6 Antibodies used in this study ............................................................................... 43 Table 2-7 Kits used in this study ......................................................................................... 43

Table 2-8 Culture media used in this study .......................................................................... 44 Table 2-9 List of commercial chemicals used in this study .................................................. 44

Table 2-10 List of in-house prepared buffers and solution ................................................... 45 Table 2-11 Components of PCR for Mycoplasma sp. detection ........................................... 47

Table 2-12 The reaction mix I to generate cDNA from hMPV RNA.................................... 53 Table 2-13 The reaction mix II to generate cDNA from hMPV RNA .................................. 54

Table 2-14 The PCR components to generate hMPV N constructs....................................... 54 Table 2-15 List of primer pairs used to generate hMPV N construct .................................... 54

Table 2-16 The components of BP recombination reaction .................................................. 55 Table 2-17 The components of LR recombination reaction .................................................. 56

Table 2-18 PCR components to confirm the presence of hMPV N insert in cloning vectors. 58 Table 2-19 Components of DNA sequencing reaction, with PCR product as template ......... 58

Table 2-20 Components of DNA sequencing reaction, with ds plasmid as template ............. 58 Figure 3-1 The growth curve of single cycle replication kinetics of rRSV A2 and rRSV A2

M(T205A) in Vero E6 and A549 cells. ........................................................................ 70 Figure 3-2 The growth curve of multiple-step replication kinetics of rRSV A2 and rRSV A2

M(T205A) in Vero E6 and A549 cells.. ....................................................................... 73 Figure 3-3 Chromatogram of DNA sequencing of the matrix gene of rRSV A2 and rRSV A2

M(T205A) showing the difference between the threonine and alanine codons at amino

acid 205. ...................................................................................................................... 75

Figure 4-1 Expression of interleukin 8 (IL-8) in cells infected with rRSV A2 or rRSV A2

M(T205A) in single cycle replication assay.. ............................................................... 85

Figure 4-2 Expression of regulated upon activation, normal T-cell expressed, and secreted

(RANTES) in cells infected with respiratory syncytial virus (RSV) in single cycle

replication assay. ......................................................................................................... 87 Figure 4-3 Expression of interleukin 8 (IL-8) in cells infected with respiratory syncytial virus

(RSV) in multiple cycle replication assay.. .................................................................. 89 Figure 4-4 Expression of regulated upon activation, normal T-cell expressed, and secreted

(RANTES) in cells infected with respiratory syncytial virus (RSV) in multiple cycle

replication assay.. ........................................................................................................ 91

Figure 5-1 Schematic representation of hMPV N constructs generated in this study.......... 105 Figure 5-2 Human metapneumovirus N (hMPV N) gene constructs and vectors used to

investigate the subcellular localisation of the constructs.. ........................................... 106 Figure 5-3 Screening for E. coli colonies containing the pDONR

®207 – hMPV N constructs

using PCR. ................................................................................................................ 108

Page 8: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

x

Figure 5-4 Screening for E. coli colonies containing the pEPI-DESTC – hMPV N constructs

using PCR. ................................................................................................................ 110

Figure 5-5 Subcellular localisation of GFP-tagged full-length hMPV N (1-394) protein

observed in Cos-7 cells.. ............................................................................................ 112

Figure 5-6 Subcellular localisation of GFP-tagged hMPV N constructs in live Cos-7 cell. 114 Appendix: Confirmation of hMPV N constructs ligated in the entry and expression clones by

DNA sequencing ....................................................................................................... 149

Page 9: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

xi

Form B

Certificate of Authorship of Thesis

Except where clearly acknowledged in footnotes, quotations and the bibliography, I certify

that I am the sole author of the thesis submitted today entitled –

Nucleocytoplasmic Transport of Respiratory Syncytial Virus Matrix Protein and Human

Metapneumovirus Nucleocapsid Protein in Infection

I further certify that to the best of my knowledge the thesis contains no material previously

published or written by another person except where due reference is made in the text of the

thesis.

The material in the thesis has not been the basis of an award of any other degree or diploma

except where due reference is made in the text of the thesis.

The thesis complies with University requirements for a thesis as set out in Gold Book

Part 7: Examination of Higher Degree by Research Theses Policy, Schedule Two (S2).

Refer to http://www.canberra.edu.au/research-students/goldbook

31 October 2012

Signature of Candidate

Wahyu Nawang Wulan Signature of chair of the supervisory panel

Date: ……………………………..

Page 10: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

xiii

Acknowledgment

I would like to thank my supervisors, Dr Reena Ghildyal and Dr Michelle Gahan for great

supervision, guidance, patience, and encouragement. Their vast knowledge tells that they are

world class scientists, and it is truly a great honour for me to be under their supervision.

Other than my supervisors, I would like to thank all the people who have helped me

accomplishing my study, in particular Dr Erin Walker and Dijana Townsend for huge

research assistance. A huge thank you to Parisa, Raj, Robert, James, Hamid, and Kylia from

the Ghildyal lab, for providing technical assistance and making the lab a great place to work.

I would also like to thank the BMS students for sharing the experience of research works and

the friendship, particularly the members of room 3D29: Guifang Shang, Khloud, Bilquis,

Sandy, Matt, Jade, Jessica, Rebecca, Fan, Sherry, and Andrew. Also, a big thank you to Ke

Jun, Marwa, Rafat, and other BMS staffs/ post-docs; the 3D level is a supportive place to

work because of them.

My master’s education could not have been carried out without the Australian Development

Scholarship, for which I am greatly indebted. I am particularly grateful to Rozana Muir, the

AUSAid Liaison Officer, for her invaluable assistance during my study.

Thank you to my family, relatives, and friends, for the support and encouragement. I am

particularly indebted to the lengthy support and encouragement given by my mother and my

aunt Djarwani, who have inspired me to pursue my dreams and kept on cheering me through

the ups and downs.

Finally, I would like to thank God for giving me the strength in accomplishing this study.

Page 11: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

xv

List of Abbreviation

A Alanine

BDV Borna disease virus

BRAP2 BRCA1 binding protein 2

BRCA1 breast cancer antigen 1

CARD caspase recruitment domain

Cdk cyclin-dependent kinase

cDNA complementary deoxyribonucleic acid

CDV canine distemper virus

CK2 casein kinase 2

CLSM confocal laser scanning microscopy

cp cold passaged

CPE cytopathic effect

Crm chromosomal region maintenance

CTD carboxy terminal domain

D aspartic acid

d.p.i day post infection

DEN dengue virus

dsRNA double-stranded ribonucleic acid

EAV equine arteritis virus

ELISA enzyme-linked immunosorbent assay

F phenylalanine; fusion protein

FG phenylalanine glycine

FI formaline-inactivated

G glycine; attachment glycoprotein

GAP GTPase-activating protein

GDP guanosine diphosphate

GE gene end

GEF guanine–nucleotide exchange factor

GFP green fluorescent protein

GS gene start

GTP guanosine triphosphate

h.p.i hour post infection

HCMV human cytomegalovirus

hMPV human metapneumovirus

I isoleucine

IFN interferon

IKK inhibitor of κB kinase

IL interleukin

ILI influenza-like illness

Page 12: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

xvi

Imp importin

IPS1 interferon-β promoter stimulator 1

IRAK IL-1 receptor-associated kinases

IRF interferon regulatory factor

ISG interferon-stimulated gene

ISGF IFN-stimulated growth factor

IκB inhibitor of κB

JAK Janus kinase

K lycine

kDa kilo dalton

L leucine

LRTIs lower respiratory tract infections

M methionine; matrix

MAVS mitochondrial antiviral signalling protein

MOI multiplicity of infection

MV measles virus

N nucleocapsid; asparagine

NES nuclear export signal

NF-κB nuclear factor kappa B

NLS nuclear localisation signal

NODSH+

nucleolar detention signal regulated by H+

NoLS nucleolar localisation signal

NPC nuclear pore complex

NS non-structural protein

NTD amino terminal domain

NTF nuclear transport factor

Nups nucleoporins

ORF open reading frame

PAMP pathogen-associated molecular pattern

PKC protein kinase C

ppp triphosphate

PRR pathogen recognition receptor

Q glutamine

R arginine

RABV rabies virus

RANTES regulated upon activation, normal T-cell expressed, and secreted

RC recombinational cloning

RCC regulator of chromosome condensation

RIG-1 retinoic-acid inducible gene

RPV rinderpest virus

rRNA ribosomal RNA

rRSV recombinant respiratory syncytial virus

Page 13: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

xvii

RSV respiratory syncytial virus

RTIs respiratory tract infections

S serine

SEM standard error of the means

SH small hydrophobic

ssRNA single-stranded ribonucleic acid

T threonine

TAB TAK1-binding protein

TAK tumour growth factor-β-activated kinase

TLR Toll-like receptor

TNF tumour necrosis factor

TRAF tumour necrosis factor receptor-associated factor

ts temperature sensitive

Tyk tyrosine kinase

URTIs upper respiratory tract infections

UV ultraviolet

V valine

VISA virus-induced signalling adaptor

VSV vesicular stomatitis virus

ZFD zinc finger domain

Page 14: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

1

CHAPTER 1

INTRODUCTION

1 Introduction

Page 15: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

2

Page 16: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

3

1.1 Viral respiratory tract infections

Viral respiratory tract infections (RTIs) are the most common infections in humans

(Lieberman et al., 2002), and morbidity and mortality contribute to a significant loss of

productivity that puts a strong economic burden on society. RTIs are highly infectious due to

the ease of the transmission of the respiratory viruses and therefore there is a need to prevent

the feasibility of RTIs distribution in the population. The disease is mostly self-limiting with

symptoms that include ‘runny nose’ (rhinorrhea), nasal congestion, throat inflammation

(pharyngitis), and cough, which are most commonly known as influenza-like illness (ILI)

(Kesson, 2007). However, disease severity varies according to the infecting virus, the age of

the infected person, as well as the host inflammatory responses. In general, upper respiratory

tract infections (URTIs), which general symptoms include runny/stuffy nose and sneezing,

are usually benign, transitory, and self-limiting; while lower respiratory tract infections

(LRTIs), which mostly progress to bronchitis/bronchiolitis and pneumonia, can be fatal

(Dasaraju and Liu, 1996). The viruses that are associated with serious LRTIs are the closely-

related negative single-stranded ribonucleic acid ((-)ssRNA) viruses of the Paramyxoviridae

family: respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) (Simoes,

1999, Wat, 2004, Kesson, 2007), to which there are no licensed vaccines available for the

general public.

1.2 Respiratory syncytial virus

Respiratory syncytial virus (RSV) was first isolated in 1956 from a laboratory chimpanzee

showing symptoms of ILI (Collins and Crowe Jr., 2007). It is the primary cause of severe

LRTIs among infants and very young children (less than 2 years old), among the elderly,

particularly those who have underlying cardiopulmonary diseases, as well as among

immunocompromised people of any age (Englund et al., 1988, Falsey and Walsh, 2005,

C.D.C, 2011). RSV infection in immunocompetent adults is seldom asymptomatic, generally

only develops into mild ILIs, and is not treated as threatening illness; yet poses a potential

risk of transmission to the susceptible age groups because immunity against subsequent

infection is short-lived (< 2 months) and reinfections are common (Anderson and Heilman,

1995, Hashem and Hall, 2003, Falsey et al., 2006). The virus is transmitted by large droplets

and fomites (Hall, 1982). The development of an RSV vaccine has been hindered by

immaturity of the immune system and immunosuppressive effects of maternally-derived

Page 17: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

4

antibodies in the young, as well as difficulties in conducting research due to poor virus

growth in vitro and physical instabilities of virus particles (Dudas and Karron, 1998).

1.2.1 Epidemiology

RSV infection is highly associated with bronchiolitis and pneumonia in infants/ very young

children and the elderly (Ruuskanen et al., 2011). The global burden of RSV disease is

estimated at 64 million cases and 160,000 deaths annually (W.H.O, 2011). In regions where

RSV infection is well documented, such as in the United States of America, the mortality rate

of RSV infection is recorded as the highest in people aged less than 1 year and more than 65

years old, reaching 5.4 and 29.6 cases per 100,000 person-years, respectively (Thompson et

al., 2003). Epidemics occur throughout the year worldwide and are characterised by a strong

pattern of seasonality, particularly in temperate regions. The epidemics mostly occur during

the winter/spring months, with a peak of incidence in January – March in the northern

hemisphere and July – September in the southern hemisphere (Al-Toum et al., 2006, Checon

et al., 2002, Roche et al., 2003, Tatochenko et al., 2010). In the tropical regions, the epidemic

period is less clear-cut, but most cases occur during the rainy season (Weber et al., 1998).

1.2.2 Clinical disease

RSV-associated illness starts as an URTI, as a result of the replication of the virus in the

nasopharyngeal epithelial cells; followed by the migration of the virus to the lower part of the

respiratory tract, where the virus targets the ciliated cells of the small bronchioles and the

type-1 pneumocytes to develop LRTI (Brooks et al., 2007, Collins and Graham, 2008). The

spread of virus particles from cell to cell is facilitated by the beating of the epithelial cilia

(Stokes-Peebles-Jr. and Graham, 2005). The incubation period between exposure and onset of

illness is 3 – 5 days; however, once symptoms have developed, progress to death may occur

rapidly (Brooks et al., 2007). Virus spread induces the fusion between adjacent plasma

membranes, leading to the development of the characteristic cytopathic effect (CPE) of

multinucleated giant cells known as syncytia (sing.: a syncytium), which can contain 20 – 30

nuclei (Figure 1-1).

Page 18: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

5

Figure 1-1 Syncytia (circled, and shown by arrow head) induced by respiratory syncytial

virus (RSV) infection in Vero E6 cells, 48 hours after infection.

RSV infection leads to the production of various inflammatory mediators that signal and

recruit immune cells to the infection sites, such as RANTES (eosinophil chemoattractant) and

interleukin 8 (neutrophil chemoattractant). However, overexpression of the mediators that

leads to the over recruitment of the immune cells may exaggerate the inflammatory responses

that subsequently promote airway damage and enhanced disease pathogenesis, which

manifests primarily as bronchiolitis and/or pneumonia (Domachowske and Rosenberg, 1999,

Openshaw and Tregoning, 2005, Oshansky et al., 2009). Bronchiolitis is a condition where

sloughed necrotic infected cells and intense inflammatory mediators infiltrate and obstruct

the bronchial lumen, while pneumonia refers to inflammation in the alveoli or when the lungs

are filled with fluid (Brooks et al., 2007, Williams and Crowe Jr., 2007). RSV-associated

bronchiolitis and pneumonia often contribute to respiratory failure and fatalities (Dasaraju

and Liu, 1996).

1.2.3 Taxonomy

RSV is classified in the order Mononegavirales, family Paramyxoviridae, subfamily

Pneumovirinae, and genus Pneumovirus (Collins, 1991, Lamb and Parks, 2006, Collins and

Graham, 2008). Other viruses of significant importance for human health in the family

Paramyxoviridae include measles virus (genus Morbillivirus), mumps virus (genus

Page 19: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

6

Rubulavirus), human metapneumovirus (genus Metapneumovirus), and the deadly emerging

virus, Nipah virus (genus Henipaviruses). RSV is differentiated into antigenic subgroups A

and B based on the neutralisation with monoclonal antibody against the G protein (Collins,

2011).

1.2.4 The structure of an RSV particle (the virion)

RSV particles are pleomorphic, having various shapes that include round, kidney, and

filamentous, with the latter the most observed (Norrby et al., 1970, Bachi and Howe, 1973).

The size of RSV is approximately 80-350 nm in diameter as a spherical unit and

approximately 60-100 nm in diameter and up to 10 µm in length as a filamentous unit

(Collins, 1991, Ogra, 2004). The main architecture of RSV is a nucleocapsid core that is

enclosed by a lipid bilayer envelope structure, with a matrix layer that bridges between the

two structures, as seen in Figure 1-2 (Collins, 1991, Lamb and Parks, 2006). The viral

genome is a negative sense single-stranded ribonucleic acid ((-)ssRNA) sized 15.2 kilobase

(kb) and contains 10 genes that are arranged in the order of 3’-NS1-NS2-N-P-M-SH-G-F-

M2-L-5’, which encode for 11 proteins, in sequential order, the non-structural 1 and 2 (NS1,

NS2), the nucleocapsid (N), the phosphoprotein (P), the matrix (M), the small hydrophobic

(SH), the attachment glycoprotein (G), the fusion (F), the transcription elongation factor

(M2-1) and a replication-regulator factor (M2-2), and the large polymerase subunit (L)

(Collins and Murphy, 2005).

The internal virion is built from the helical nucleocapsid complex that consists of the

(-)ssRNA genome, N protein, the L and P polymerase complex (García-Barreno et al., 1996,

Collins, 2011). Viral envelope is derived from host plasma membrane and contains the SH

protein and the two RSV major neutralising antigens: the attachment G protein and the fusion

F protein. The G and F proteins mediate the binding of virion to cellular receptors to establish

efficient infection (Collins, 2011). The homotrimeric-arranged F protein specifically

functions to fuse between viral and host membranes to initiate infection and between the

plasma membranes of infected cells to develop the characteristic syncytial CPE (Morton et

al., 2003). RSV F is capable of attachment function in the absence of the G protein (Lamb

and Parks, 2006, Collins, 2011). The G protein is less neutralising and more diverged than the

F protein (Collins, 1991). However, together with the SH protein, the G protein is not

required for growth in cell culture (Karron et al., 1997a, Teng and Collins, 1998). The RSV

Page 20: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

7

M protein underlies the viral envelope and is associated with the nucleocapsid complex; its

positioning is the result of a hydrophobic-electrostatic interaction with the plasma membrane

and the binding to the cytoplasmic tail of the G glycoprotein and to the M2-1 protein of the

nucleocapsid core (Lenard, 1996, Gaudier et al., 2002, Ghildyal et al., 2002, Latiff et al.,

2004, Ghildyal et al., 2005b, Li et al., 2008).

The proteins that comprise the virion are called the structural proteins, while the non-

structural proteins, which comprise M2-1, M2-2, NS1, and NS2, are either underrepresented

or absent in the virion but synthesised in infected cells to function in regulation of viral

proliferation (Collins, 1991, Lamb and Parks, 2006, Collins, 2011). For example, the NS1

and NS2 proteins facilitate successful replication by counteracting the antiviral action of the

host innate immune response, the M2-1 protein is the transcription elongation factor, and the

M2-2 protein is the regulatory switch between transcription and replication of viral RNA

(Hardy and Wertz, 1998, Bermingham and Collins, 1999, Spann et al., 2004, Spann et al.,

2005).

Figure 1-2 The structure of respiratory syncytial virus. The general structure of an RSV

virion is a nucleocapsid core that is sheathed by an envelope structure, which is the host-

derived lipid bilayer plasma membrane. The nucleocapsid core consists of the negative-sense

(-) single-strand (ss) RNA genome bound by the nucleocapsid (N) protein and is associated

with the phosphoprotein (P) and large (L) polymerase subunit. The nucleocapsid core

interacts with the envelope structure by the mediation of the matrix (M) protein that layers

the inner surface of the envelope. The surface glycoproteins consisting of the fusion (F),

attachment glycoprotein (G), and the small hydrophobic (SH) proteins are inserted into the

envelope

Page 21: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

8

1.2.5 The life cycle of RSV

The main stages in the life cycle of RSV are adsorption/entry, RNA synthesis, and

assembly/budding (Figure 1-3). Adsorption/entry involves the binding between the viral and

cellular receptors, followed by fusion of the F protein with the plasma membrane, and the

release of the nucleocapsid core into the cytoplasm, which is known as uncoating (Collins,

1991, Lamb and Parks, 2006, Collins and Crowe Jr., 2007). RNA synthesis consists of

transcription of monocistronic mRNAs from (-)ssRNA genome and genome replication. The

initiation and termination of transcription is signalled by specific internal sequences, namely

gene start (GS) and gene end (GE), that flank each gene; and transcription is attenuated at

succeeding genes such that the relative abundance of each mRNA decreases in the 3’ – 5’

direction of the (-)ssRNA genome (Lamb and Parks, 2006). Genome replication involves the

production of the complementary (+)ssRNA antigenome that acts as an intermediate template

for the synthesis of new (-)ssRNA (Collins, 1991, Lamb and Parks, 2006). Virion assembly is

initiated by the formation of the nucleocapsid core in the cytoplasm and the arrangement of

the envelope glycoproteins in the plasma membrane (Collins, 1991, Lamb and Parks, 2006).

New infectious particles are formed by the joining together between the nucleocapsid and

envelope structures by the M protein, followed by maturation via budding through plasma

membrane, during which the cellular membrane is acquired as virion envelope (Norrby et al.,

1970, Bachi and Howe, 1973, Roberts et al., 1995, Garoff et al., 1998).

The single cycle growth of RSV in cell culture takes place between 20 – 40 hours post

infection (h.p.i) and the viral spread from cell to cell induces syncytia formation that has been

shown to peak between 72 and 96 h.p.i (Bachi and Howe, 1973, Lamb and Parks, 2006).

However, it is arguable whether the production of infectious particles is the highest at this

point since syncytia formation can be induced by RSV F protein independent of other viral

components (Morton et al., 2003).

Page 22: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

9

1.3 The nucleocytoplasmic transport of viral proteins

1.3.1 Overview

The nucleocytoplasmic transport of molecules inside a eukaryotic cell plays an important role

in the integration of DNA replication/RNA biogenesis and protein synthesis, since the first

occurs in the nucleus and the latter in the cytoplasm. The two compartments are separated by

the nuclear envelope, which is a double membrane system perforated by nuclear pores

(Alberts et al., 2008). Molecules are transported through the pore, which itself is a complex

protein structure built from subunits called nucleoporins (Nups). The nuclear pore complex

(NPC) consists of three domains: the cytoplasmic filaments, the central core, and the nuclear

basket (Sorokin et al., 2007) (Figure 1-4). The spatial segregation provides the cells with a

Figure 1-3 The life cycle of respiratory syncytial virus. All stages of replication occur in the

cytoplasm of infected cells. (1) The RSV virion attaches to the cellular receptor via G

glycoprotein, followed by (2) the fusion of viral membrane with the cellular plasma membrane

mediated by F glycoprotein. The consequence of which is the release of the helical nucleocapsid

containing the (-)ssRNA genome into the cytoplasm. The (-)ssRNA is transcribed into mRNAs

(3) and replicated into progeny genome (4). Viral proteins are translated (5) from the mRNAs.

The nucleocapsid complex is built in the cytoplasm (6) from free N subunits, the (-)ssRNA

genome, the P(hospho) protein, L polymerase subunits, and the M2-1 protein. Meanwhile, the

glycoproteins are transported via the exocytic pathway to the plasma membrane where they are

arranged at the assembly sites (7). The matrix (M) protein interacts with the cytoplasmic tail of G

glycoprotein of the envelope structure and with the M2-1 protein of the nucleocapsid complex in

the cytoplasm; then brings the two structures together to form new infectious particles, which

mature by budding out of the plasma membrane (8).

Page 23: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

10

means of regulation of gene expression. In terms of virus infection, nucleocytoplasmic

transport of viral genomes or proteins is crucial for the successful replication of many

viruses, one of the reasons is as a strategy to evade the host immune responses (Fulcher and

Jans, 2011). The transport of viral proteins between the cytoplasm and the nucleus is most

commonly facilitated by a mechanism known as Ran-dependent nucleocytoplasmic transport

of proteins. Nucleocytoplasmic transport is bidirectional, consisting of nuclear import and

nuclear export, and is regulated at the level of individual cargo molecule, nuclear transport

receptors, and nucleoporins; with the efficiency of nuclear transport is determined by the

affinity between nuclear transport factors and nuclear transport signals (Nigg, 1997,

Henderson and Eleftheriou, 2000, Lam et al., 2001, Sorokin et al., 2007, Terry et al., 2007,

Chumakov and Prasolov, 2010).

1.3.2 The components of nucleocytoplasmic transport

The main components in nuclear transport are transport factors, nuclear transport signals of a

cargo molecule, phenylalanine glycine nucleoporins (FG-Nups) of the NPC, and GTPase Ran

with its main regulators (RanGAP1 and RanGEF) (Gorlich and Mattaj, 1996, Nigg, 1997,

Terry et al., 2007, Sorokin et al., 2007, Alberts et al., 2008, Chumakov and Prasolov, 2010).

Figure 1-4 Overview of a nuclear pore complex (NPC). The cytoplasmic filaments, outer

and inner ring, the NPC core, nuclear basket and distal ring are shown with their constituent

nucleoporins (Nups).

Page 24: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

11

1.3.2.1 Transport factors

Transport factors are proteins from the large evolutionarily-conserved karyopherin β family,

which carry the cargo molecule across the NPC and can be differentiated into importins and

exportins (Sorokin et al., 2007). Importins transport the cargo molecule from the cytoplasm to

the nucleus, while exportins travel in the opposite direction. Importin α (Imp-α) and importin

β (Imp-β) are members of the importin family that are known to play a major role in nuclear

import; while the most widely characterized exportin is the Crm1 (chromosomal region

maintenance 1), which is a homologue of Imp-β (Alberts et al., 2008, Chumakov and

Prasolov, 2010). The nucleocytoplasmic transport of the M protein of RSV has been shown to

be facilitated by Imp-β1 and Crm1 (Ghildyal et al., 2005a).

1.3.2.2 Nuclear transport signals

The nuclear localisation signal (NLS) specifies translocation across the NPC; it provides a

high affinity interaction between a cargo and an importin (Imp) and is typically present as a

repeat (monopartite) of positively charged basic residues (lysine (K) or arginine (R)), or two

clusters of the repeat (bipartite) separated by a spacer (Corbett and Silver, 1997). The most

well-studied monopartite NLS is of the SV40 T antigen, PKKKRKV132

, while the

nucleoplasmin’s KRPAATKKAGQAKKKK, is the best-studied bipartite NLS (Gorlich and

Mattaj, 1996, Nigg, 1997, Sorokin et al., 2007). Bold characters indicate the key residues of

an NLS motif. The interaction of the positively-charged basic NLS with the acidic residues in

the inner surface of the importin results in the strong affinity of the cargo-importin complex

(Jamali et al., 2011). This binding affinity governs the formation of a functional nuclear

import complex that eventually determines the nuclear import rate (Timney et al., 2006).

It has been suggested that proteins having NLS rich in arginines can be transported directly

by Imp-β; while proteins with NLS rich in lysines require the mediation of Imp-α for binding

to Imp-β (Palmeri and Malim, 1999). The differentiation between direct binding to Imp-β1

and binding with mediation of Imp-α is specific, since the NLS of a protein that directly binds

to Imp-β1 tends to bind to an area of Imp-β1 that recognises Imp-α; therefore there is a

competition to use the Imp-β1 binding region between Imp-α and cargo, making the presence

of Imp-α inhibits Imp-β1-cargo binding (Henderson and Percipalle, 1997).

Page 25: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

12

The nucleolar localisation signal (NoLS) ensures accumulation of a cargo protein in the

nucleolus. There is not much known about the consensus sequence of a NoLS; however it is

believed that the motif is also rich in basic residues like NLS and tends to be located at both

ends of a protein (Scott et al., 2011). Commonly, cytoplasmic proteins that localise in the

nucleolus also possess NLS to facilitate transport across the NPC (Scott et al., 2010). A

protein can also localise in the nucleolus as a result of retention in the nucleolus following a

high affinity binding to the nucleolar components such as ribosomal DNA or RNA (rDNA or

rRNA), where this high binding affinity is also facilitated by a specific sequence known a s

nucleolar detention signal regulated by H+ (NoDS

H+) (Carmo-Fonseca et al., 2000, Mekhail et

al., 2007). In this case, it was proposed that the transport signal in these proteins is most

likely NLS, but then the proteins have the capability of binding to nucleolar components, and

this binding facilitates the nucleolar localisation (Jans and Hübner, 1996).

The nuclear export signal (NES) is a strong cytoplasmic localisation signal that is recognised

by Crm1 and consists of a short hydrophobic sequence most commonly conforms to the motif

LXXXLXXLXL, where L can be the hydrophobic residues of leucine (L), methionine (M),

phenylalanine (F), valine (V), or isoleucine (I), and X is any amino acid, as exemplified by

the HIV Rev NES LQLPPLERLTL83

(Gorlich and Mattaj, 1996, Nigg, 1997, Hutten and

Kehlenbach, 2007, Sorokin et al., 2007, Jamali et al., 2011). The transport signal is usually

positioned in an exposed region or in a flexible region that unfolds easily (la-Cour et al.,

2004).

1.3.2.3 FG-Nups

FG-Nups fill the central core of a NPC and impose a selective transport following a specific

interaction with transport factors (Grunwald et al., 2011). FG-Nups that are critical for

nucleocytoplasmic transport are characterised by the GLFG, FxFG, or FG repeats motif,

separated from each other by spacers rich in glutamine (Q), serine (S), threonine (T), or

asparagine (N). The F and L residues of this motif produce weak hydrophobic interactions

with karyopherins, facilitating translocation of transport complex across a nuclear pore by

acting as docking sites for the translocation of transport factor (Sorokin et al., 2007, Jamali et

al., 2011).

Page 26: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

13

1.3.2.4 Ran and its regulators

The formation and dissociation of the transport complex are controlled by GTPase Ran

through hydrolysis of GTP into GDP. The hydrolysing activity of Ran is induced by

RanGTPase activating protein 1 (RanGAP1) that is compartmentalized in the cytoplasm

(Nigg, 1997, Sorokin et al., 2007, Chumakov and Prasolov, 2010, Jamali et al., 2011). Ran-

bound GDP is converted into a RanGTP by the nuclearly-compartmentalised guanine–

nucleotide exchange factor (RanGEF), which in mammals is known as the regulator of

chromosome condensation (RCC1) (Gorlich and Mattaj, 1996, Nigg, 1997, Sorokin et al.,

2007). Another important factor is the nuclear transport factor 2 (NTF2), which does not play

a direct role in translocating a cargo protein but is involved in governing the directionality of

nucleocytoplasmic transport/karyopherin recycling by maintaining a high concentration of

nuclear Ran-GTP and cytoplasmic Ran-GDP (Chumakov and Prasolov, 2010, Jamali et al.,

2011, Grunwald et al., 2011).

1.3.3 The mechanism of nuclear import

In terms of Imp-β1-facilitated nuclear import, the transport of proteins from the cytoplasm to

the nucleus occurs through three major steps: firstly, the association of cargo-Imp-β1 and

subsequent docking to the NPC, secondly, the translocation of nuclear import complex

through the NPC, and thirdly, the dissociation of nuclear import complex. In the cytoplasm,

the Imp-β1 binds to a cargo protein at its NLS to form an import complex, which

subsequently docks at FxFG repeats of the Nup358 that is positioned in the cytoplasmic

filaments of the NPC (Sorokin et al., 2007, Chumakov and Prasolov, 2010). From Nup358,

the complex translocates to the nuclear side by multiple associations-dissociations between

the import complex and FxFG repeats, particularly those contained in Nup62 of the central

core of the NPC (Gorlich and Mattaj, 1996, Nigg, 1997, Sorokin et al., 2007). Recent

findings suggest that translocation is energy-independent, taking place only through repeated

low-affinity hydrophobic interactions between Imp-β1 and the phenylalanine residues of

FxFG repeats (Sorokin et al., 2007, Terry et al., 2007, Chumakov and Prasolov, 2010, Jamali

et al., 2011). Reaching the nuclear side of the NPC, the Imp-β1 docks the import complex to

Nup153 of the nuclear filaments of NPC (Sorokin et al., 2007). RanGTP, which is highly

concentrated in the nucleus, breaks this connection by binding to Imp-β1, thus releasing

cargo protein into the nucleoplasm. RanGTP has a very strong affinity for karyopherins, and

Page 27: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

14

its association to Imp-β1 disassembles the import complex: possibly due to the competition

with the binding domain in Imp-β1 for NLS or because RanGTP interaction with Imp-β1

drives conformational change that detaches cargo (Henderson and Percipalle, 1997, Lam et

al., 2001, Pemberton and Paschal, 2005, Chumakov and Prasolov, 2010, Jamali et al., 2011).

The RanGTP–Imp-β1 complex is recycled back into the cytoplasm, where RanGTP is

hydrolysed into RanGDP by RanGAP.

1.3.4 The mechanism of nuclear export

In contrast to nuclear import, translocation of export complex from nucleus to cytoplasm is

an energy-dependent process. A functional nuclear export complex commonly comprises

Crm1, a cargo protein, RanGTP, and RanBP3, which is a cofactor that enhances the binding

of Crm1 to RanGTP as well as increases the affinity of Crm1 to NES (Hutten and

Kehlenbach, 2007, Sorokin et al., 2007, Chumakov and Prasolov, 2010). The complex

translocates through the NPC in a mechanism that has not been well understood but is

believed to involve the FG-Nups (Macara, 2001). The translocation ends at the terminal

docking sites, which, although still in debate, are thought to involve the cytoplasmic Nup358

(Hutten and Kehlenbach, 2007, Sorokin et al., 2007, Chumakov and Prasolov, 2010).

In the terminal docking sites, the export complex dissociates through the hydrolysis of

RanGTP, which causes a conformational change that breaks the strong bond of RanGTP –

Crm1. The hydrolysis is facilitated by RanGAP1, which itself is targeted to the Nup358 by

the Sumo E3 ligase (Jamali et al., 2011). RanGTP – karyopherin association is very stable

and requires RanBP1 to first penetrate the bond (Chumakov and Prasolov, 2010, Jamali et al.,

2011). The Crm1 affinity to NES weakens as RanGTP is hydrolysed into RanGDP, thus

allowing the release of cargo protein into the cytoplasm. Meanwhile, RanGDP is transported

back to the nucleus by NTF2, where it is converted into RanGTP by RanGEF (Chumakov

and Prasolov, 2010, Jamali et al., 2011).

1.3.5 Regulation of nucleocytoplasmic transport by phosphorylation

Phosphorylation is a common means of regulating the subcellular localisation of various

proteins in eukaryotic cells. It is a reversible posttranslational modification of the proteins

that consists of phosphorylation and dephosphorylation. Phosphorylation is the transfer of a

phosphate group (PO32-

) from the kinase-facilitated hydrolysis of adenosine tri-phosphate

Page 28: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

15

(ATP) into adenosine di-phosphate (ADP), mainly to the amino acid residues serine (S),

threonine (T), or tyrosine (Y) (Alberts et al., 2008, Nelson and Cox, 2008).

Dephosphorylation is phosphatase-catalysed removal of the phosphate group from a

phosphorylated amino acid residue (Alberts et al., 2008). To phosphorylate or

dephosphorylate, kinases or phosphatases, specifically recognise common structural motifs.

Kinases that phosphorylate serine and threonine cluster in one subfamily, the most prominent

of which are casein kinase 2 (CK2) and cyclin-dependent kinase (cdk) cdc2 that recognise the

motifs –X-(S/T)-X-X-(E/D)-X- and –X-(S/T)-P-X-(K/R)-, respectively (Alberts et al., 2008,

Nelson and Cox, 2008). Whereas the tyrosine kinase subfamily clusters in another

evolutionarily-related group, the most prominent enzyme of which is Src (Alberts et al.,

2008).

Phosphorylation, particularly at an individual amino acid residue that plays a critical role for

the tertiary or quaternary structure of polypeptides, can dramatically change the overall

structure of a protein, hence altering its interaction with other proteins (Jans and Hübner,

1996, Harreman et al., 2004). As a result, phosphorylation is an important mechanism that

regulates the nucleocytoplasmic transport of viral proteins, as described in the following

models.

1.3.5.1 Phosphorylation promotes the interaction between NLS and importins

This model best describes the upregulating impact of phosphorylation in the vicinity of NLS,

as evidenced by the CKII-(cdk) cdc2-NLS (CcN) motif of simian virus 40 (SV40) T-antigen

(T-Ag), a tumour-inducing protein, and the CKII-CKII-NLS2 (C2N) motif of human

cytomegalovirus (HCMV) processivity factor of DNA polymerase, ppUL44 (Jans, 1995,

Alvisi et al., 2005).

The proposed mechanism of how phosphorylation in the vicinity of NLS regulates the

nucleocytoplasmic transport of viral proteins is based on the electrostatic interaction between

the positively charged basic residues of NLS and the acidic amino acid residues in the inner

side of Imp-α. Phosphorylation at a distance of approximately 5 – 25 amino acid residues

from NLS creates additional negative charges that are necessary for the interaction between

NLS and Imp-α, whereas phosphorylation inside the NLS will impair the interaction due to

electrostatic repulsion (Rihs et al., 1991, Hübner et al., 1997, Alvisi et al., 2008). The

enhanced interaction between NLS and importin eventually increases the rate of nuclear

Page 29: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

16

import. CKII phosphorylation at S112

of SV40 T-Ag and at S413

of HCMV ppUL44 has been

shown to increase the binding of NLS to Imp-α/β complex and subsequently enhance the

nuclear accumulation of both proteins (Rihs et al., 1991, Hübner et al., 1997, Alvisi et al.,

2005).

1.3.5.2 Cytoplasmic or nuclear retention

In this model, the negative charge introduced by phosphorylation strengthens the interaction

affinity between a protein and a cytoplasmic retention factor. It has been studied at length in

the (cdk) cdc2 phosphorylation at T124

of SV40-Tag and PKC phosphorylation at T427

of

HCMV ppUL44, in which phosphorylation enhances the binding of the proteins to a

cytoplasmic retention factor named breast cancer antigen 1 (BRCA1) binding protein 2

(BRAP2) (Fulcher et al., 2010). As a result, proteins are retained in the cytoplasm and

nuclear import is inhibited.

Phosphorylation provides negative electric charge to regulate nuclear import. Substituting

kinases-recognisable residues (S/T) with negatively-charged residue, such as aspartic acid

(D), maintains the impact of phosphorylation in a protein’s function; while substitution with

neutrally-charged residue, such as glycine (G) or alanine (A), eliminates the phosphorylation

impact(s). S112

G substitution in the SV40 T-Ag and S413

G/A substitution in HCMV ppUL44

disables CK2 phosphorylation at these sites and reduces the rate of nuclear accumulation of

both proteins (Rihs et al., 1991, Alvisi et al., 2005). In terms of nuclear import inhibition by

phosphorylation, T124

A and T427

A of SV40 T-Ag and HCMV ppUL44 disables CK2 and

PKC phosphorylation, leading to reduced affinity of the proteins to BRAP2 and regular

nucleocytoplasmic transport rate (Fulcher et al., 2010). Conversely, T124

D substitution of

SV40 T-Ag and T427

D substitution of HCMV ppUL44, which were aimed to generate

phosphorylation mimicry, increases the affinity of both proteins to bind to BRAP2,

comparable with the impact of CK2 and PKC phosphorylation at T124

of SV40 T-Ag and at

T427

of HCMV ppUL44, respectively (Fulcher et al., 2010). Thus, kinase-facilitated

phosphorylation can enhance or inhibit the nucleocytoplasmic transport of a protein,

depending on the phosphorylation site, the altered structure of proteins, and the biological

pathways affected.

Page 30: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

17

1.3.6 Regulated nucleocytoplasmic transport of viral proteins

1.3.6.1 Vesicular stomatitis virus (VSV) matrix (M) protein

VSV belongs to the genus Vesiculovirus of the family Rhabdoviridae, and is a (-)ssRNA

virus (Lyles and Rupprecht, 2007). It replicates in the cytoplasm and has its M protein

localised in the nucleus at a particular stage of infection through NLS-facilitated active

transport (Glodowski et al., 2002). The nuclear transport of VSV M protein is crucial for the

establishment of efficient infection, in that it allows VSV M to inhibit the nucleocytoplasmic

transport of molecules to and from the host nucleus, including the export of host mRNA (Her

et al., 1997, von Kobbe et al., 2000, Petersen et al., 2000). von Kobbe et al. (2000) showed

that VSV M protein strongly associates with Nup98 of the NPC, which may competitively

eliminate the ability of Ran-dependent nucleocytoplasmic transport components to use the

Nups in translocating molecules through the nuclear pore. VSV M accesses the Nup98

through an active nuclear import mechanism that is facilitated by a bipartite NLS at amino

acid 47 – 229; while VSV M association to Nup98, which leads to the inhibition of host gene

expression, is facilitated by methionine (M) at position 51 (Petersen et al., 2000, Glodowski

et al., 2002). This association prevents the nuclear export of many cellular molecules, leading

to the sequestering of host mRNA inside the nucleus and shut off of host gene expression,

including the suppression of IFN-β expression (Ahmed et al., 2003). IFN-β is a cytokine

produced to respond to viral infection and generates an ‘antiviral state’ in target cells to

restrict viral replication (Fontana et al., 2008). The failure to induce an ‘antiviral state’, as the

result of inhibition of IFN-β production, could eliminate the barrier for viral dissemination

throughout the organism. Therefore, the impact of the nucleocytoplasmic transport of VSV M

protein disrupts the host defence capacity to fight virus infection.

1.3.6.2 Dengue virus serotype 2 (DEN-2) NS5 protein

Dengue virus belongs to the genus Flavivirus of the family Flaviviridae, is a (+)ssRNA virus,

and, based on serology, is classified into dengue virus serotype 1, 2, 3, and 4 (DEN-1,-2,-3,-

4) (Henchal and Putnak, 1990). The NS5 protein is the viral RNA-dependent RNA

polymerase that performs its role in the cytoplasm in association with the viral helicase NS3.

In the much studied DEN-2, NS5 has been shown to possess two NLSs and one NES, namely

an Imp-α/β-recognised NLS (aNLS) within amino acids 369 – 389, an Imp-β1-recognised

NLS (bNLS) within amino acids 320 – 368, and a Crm1-recognised NES within amino acids

Page 31: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

18

327 – 343, which enable it to shuttle between the nucleus and the cytoplasm (Pryor et al.,

2007, Rawlinson et al., 2009). The nuclear import is primarily facilitated by the aNLS;

whereas the bNLS overlaps with the NS3 binding site that poses a competitive binding to

Imp-β1 (Johansson et al., 2001, Pryor et al., 2007). The nucleocytoplasmic transport is

regulated by a cellular-kinase phosphorylation that affects at least four distinct serine residues

(Kapoor et al., 1995, Reed et al., 1998). Phosphorylation has been proposed to introduce

conformational changes to the cytoplasmic NS5 in order to release NS3 and unmask NLSs,

thus allowing the binding of importins; while the nuclear export may involve

dephosphorylation within the nucleus such that NES is exposed for Crm1 binding (Alvisi et

al., 2008).

The nuclear localisation of NS5 is associated with increased virus production and reduced IL-

8 synthesis, particularly during 48 – 96 hours post infection (Pryor et al., 2007, Rawlinson et

al., 2009). IL-8 is a chemotactic cytokine (chemokine) that recruits potent phagocytes, such

as neutrophils, to inflammatory sites (Mukaida et al., 1998). IL-8 function in trafficking

neutrophil migration to the infection sites for pathogen clearance; but if overexpressed, can

cause damage to the body tissues due to the cytotoxic effects of neutrophils’ granular proteins

to uninfected cells (Kindt et al., 2007), which in doing so, contributes to the development of

severe disease.

1.3.6.3 Rabies virus P protein

The neurotropic rabies virus (RABV) of the family Rhabdoviridae has a 12-kb genomic

ss(-)RNA genome that encodes nucleoprotein (N), phosphoprotein (P), matrix protein (M),

glycoprotein (G), and polymerase (L) (Lyles and Rupprecht, 2007). RABV P mRNA, in

addition to be translated into a full-length P protein (297 amino acids), is also translated into

four shorter proteins namely P2, P3, P4, and P5, due to a leaky-scanning mechanism at

internal M codons 20, 53, 69, and 83, respectively (Chenik et al., 1995). The RABV P can

localise in both nuclear and cytoplasmic compartments due to having a Crm1-dependent

amino (N)-terminal domain (NTD) NES (amino acids 49 – 58), carboxy (C)-terminal domain

(CTD) NLS (amino acids 172 – 297, active residues 211

KKYK214

and R260

), and a Crm1-

dependent protein kinase C (PKC)-activated CTD NES (amino acid 221 – 238, active

residues 227

FEQLKM232

) (Pasdeloup et al., 2005, Moseley et al., 2007). The cytoplasmic

compartmentalisation of these proteins interferes with signal transduction in the innate

Page 32: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

19

immune response against RABV infection, by tampering with the activation of type I

interferon (IFN-α/β).

In RABV infection, the recognition of pathogen-associated molecular pattern (PAMP) viral

5’-triphosphate (ppp) ssRNA by the pattern-recognition receptor (PRR) retinoic-acid

inducible gene 1 (RIG-1) leads to a cascade of intracellular signalling that activates the type I

IFN (Faul et al., 2010). RABV P restrains the type I IFN activation in two ways. Firstly, an

internal domain of RABV P, (amino acids 176-186), masks the S386

phosphorylation of

interferon regulatory factor 3 (IRF3), which dimerisation and nuclear translocation are

required to trigger the transcription factor complex for early expression of IFN-β (Hiscott,

2007, Katze et al., 2008, Randall and Goodbourn, 2008, Takeuchi and Akira, 2008,

Mogensen, 2009). Secondly, the CTD of RABV P (amino acids 288 – 297) develops a strong

affinity to the DNA-binding domain of phosphorylated STAT1 (pSTAT1), an activator of the

transcription of interferon-stimulated genes (ISGs); thus, preventing the transcription of

products that facilitate the antiviral action of type I IFN (Vidy et al., 2005, Brzózka et al.,

2006, Vidy et al., 2007, Schnell et al., 2010). The cytoplasmic localisation of RABV P has

been shown to be regulated by the interplay between NES and NLS, and the recently

discovered PKC phosphorylation at amino acid residue S210

that activates the CTD NES

whilst silencing the NLS (Moseley et al., 2007).

1.4 The nucleocytoplasmic transport of the matrix (M) protein of RSV

1.4.1 Overview of the M protein of RSV

The RSV M protein (256 amino acids, 28.7 kDa) underlies the viral envelope, is associated

with viral nucleocapsid complex, consists mostly of basic amino acid residues, has an

increased hydrophobicity from NTD (residues 1 – 126) towards CTD (residues 140 – 255),

and has a strong tendency to self-assembly (Latiff et al., 2004, Kraus et al., 2005, Money et

al., 2009). RSV M is capable of binding to various structural viral components. RSV M

association to the G protein is influenced by the amino acid 2 (S) and 6 (D) in the NTD of

RSV G; while the binding to M2-1 protein requires the NTD of RSV M (amino acid residues

1 – 110) (Ghildyal et al., 2005b, Li et al., 2008). RSV M is also capable of independent

binding to the nuclear components via a zinc-finger domain (ZFD) contained in the residues

110 – 150; in which basic residues (K or R) at position 121, 130, 156, 157, and 170 have

been shown to be important for RNA-binding capacity (Rodriguez et al., 2004, Ghildyal et

Page 33: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

20

al., 2005a). The interaction with various parts of the virus, added to the strong predisposition

to self-oligomerise (Kraus et al., 2005, Lamb and Parks, 2006), has made RSV M the driving

force/central organiser of the formation of new infectious particles. Further, this crucial

function has been linked to the trafficking of RSV M between the nucleus and the cytoplasm.

1.4.2 The nucleocytoplasmic transport of the M protein in infected cells

RSV M is an intracellular transport protein that is transported into the nucleus directly by

Imp-β1 and brought out of the nucleus by Crm1 (Ghildyal et al., 2005a, Ghildyal et al.,

2009). Imp-β1 recognises the bipartite NLS 154

TSKKVIIPTYLRSISVRNK172

; while Crm1

recognises a CTD NES 194

IIPYSGLLLVITV206

(Ghildyal et al., 2009). In addition, the

nuclear concentration of RSV M is also promoted by ZFD binding to nuclear component(s)

(Ghildyal et al., 2005a). The nuclear localisation of RSV M can be observed in the early

hours of infection while the cytoplasmic localisation takes place in the late stage of infection,

with the demarcating time point between the two processes is approximately 18 hours post

infection (Ghildyal et al., 2002). The nuclear export is up-regulated by a late stage CK2

phosphorylation (Alvisi et al., 2008).

1.4.2.1 Functions of the M protein in the nucleus

The nucleocytoplasmic transport of RSV M has been thought to associate with non-structural

roles of the protein since the whole replicative events, which require the structural functions

of RSV M, are cytoplasmic. One possibility is that RSV M might interfere with host genetic

machinery since its nuclear localisation has been shown to coincide with a decrease in host

transcriptional activity (Ghildyal et al., 2003). Although the precise mechanism is unclear, an

inhibition of host transcriptional activity could be facilitated by the capability of RSV M to

bind the nuclear component(s). Inhibition of host transcriptional activity might be directed to

enhanced infectious processes, as has been observed in the nuclear localisation of VSV M,

where inhibition of cellular transcription dampens the activation of the antiviral IFN-β

(Ahmed and Lyles, 1998, Ahmed et al., 2003). This suggests that RSV M nuclear localisation

could promote molecular events that lead to the generation of abundant progenies, as shown

by substitution of the positively-charged basic residues 156

KK157

of NLS into the neutrally-

charged 156

TT157

that inhibited RSV M nuclear localisation in the early stage of infection and

led to the reduced production of infectious particles (Ghildyal et al., 2009). RSV M nuclear

localisation might also facilitate efficient progeny production by delaying the

Page 34: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

21

assembly/budding of new infectious particles until enough viral components have been

synthesised in the cytoplasm (Harrison et al., 2010). Further studies are required to elucidate

the role(s) of RSV M nuclear localisation.

1.4.2.2 Functions of the M protein in the cytoplasm and plasma membrane

RSV M cytoplasmic localisation is associated with its structural function as the driving

force/central organiser of the formation of viral progenies in the assembly/budding process.

Successful assembly/budding is important for the production of infectious particles. Virions

that cannot bud out of the infected cells are trapped inside and so the infection will not

spread. The involvement of matrix protein in the assembly/budding step can be described as

follow. Firstly, RSV M transports the nucleocapsid core from the cytoplasm to the assembly

/budding site, condenses it for incorporation to new particles and switches off viral

transcription in favour of infectious particles release; secondly, RSV M joins the

nucleocapsid complex with the glycoproteins complex arranged in the budding sites, while

developing its own hydrophobic-electrostatic interaction with the plasma membrane; and

lastly, RSV M builds infectious particles via self-aggregation followed by pinching-off of

new particles through the plasma membrane (Garoff et al., 1998, Chazal and Gerlier, 2003,

Jayakar et al., 2004, Takimoto and Portner, 2004). The matrix protein has also been shown to

mediate the cellular factors that regulate infectious particle release (Harty et al., 1999).

Considering the importance, a threshold level of functional matrix protein must be available

for the assembly/budding process as evidenced by matrix-deficient mutant strains of a

(-)ssRNA virus (rabies virus, Rhabdovirus, Rhabdoviridae, Mononegavirales) that produced

extremely low numbers of progenies to the point of near-complete defects of particle

formation (Mebatsion et al., 1999). As such, RSV M needs to be involved throughout the

assembly/budding process that takes place in the cytoplasm and plasma membrane in the late

stage of infection.

1.4.2.3 The importance of timely regulated nucleocytoplasmic transport of

RSV M

Given the transient nuclear or cytoplasmic localisation of RSV M relates to important

regulations of viral proliferation, RSV M must localise precisely and punctually in the

designated compartment in order to establish successful infection. Disruption of these

localisations has been shown to impair the production of infectious particles. Mutant RSV

Page 35: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

22

with substitution in key basic residues of NLS (rRSV A2 M(156

KK157

156TT

157)) lost nuclear

accumulation in the early stage of infection (prior to 18 hours post infection) and underwent

reduced titre of viral proliferation in A549 cells, while mutant RSV with substitution in key

hydrophobic residues of NES (rRSV A2 M(200

LLLV203

200AAAA

203)) did not release

detectable infectious particles (Ghildyal et al., 2009). The study also found that RSV M with

200LLLV

203

200AAAA

203 did not localise in the assembly/budding sites at 24 hours post

infection in a transfection system using Vero cells. The study suggests that early stage

nuclear localisation promotes viral proliferation, although the precise mechanism is still

unknown, while positioning of RSV M in assembly/budding sites in late stage of infection is

important for production of infectious particles.

Phosphorylation is a main regulatory mechanism in the nucleocytoplasmic transport of a

protein (Jans and Hübner, 1996) The nuclear export of RSV M has been shown to be up-

regulated by CK2 phosphorylation in the late stage of infection (Alvisi et al., 2008). Potential

CK2 phosphorylation motifs have been identified along RSV M, although the precise

phosphorylation site is still under investigation (Ghildyal et al., 2006). A potential site, the

amino acid residue T205

that is positioned adjacent to the key hydrophobic residues of NES,

has been disproven as the designated site based on transfection experiments using the mutant

rRSV A2 M(T205A) (R. Ghildyal, personal communication). Further analysis revealed that

RSV mutant strain rRSV A2 M(T205A) had grown to a lower titre in cell lines expressing

IFN-α/β gene (HEp2 and A549 cell lines) compared to cell line deficient in expressing the

antiviral IFN-α/β response (Vero E6). The evidence has proposed a possibility that RSV M

might have an active role in affecting the regulation of the antiviral IFN-α/β expression

during the course of infection and, considering that RSV M is a transport protein, that the

regulation might associate with the nucleocytoplasmic transport of RSV M.

1.5 Regulation of cellular innate immunity against RSV infection

1.5.1 Overview

There are two strategies of mammalian immunity against viral infection, the innate and

adaptive immune responses. The innate immunity pre-exists prior to infection and is activated

within a short timeframe following infection, while the adaptive immunity is activated later

due to the complexity of its biological processes, but provides immunological memory that

results in more specific and faster protection on subsequent infection (Kindt et al., 2007).

Page 36: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

23

The innate immunity is important for the first line of protection against viruses since it aims

to restrict viral spread and/or prevent disease development by inhibiting viral replication in

the early stage of infection. The immediate protection is resulted from the development of

‘antiviral state’ in infected cells that is subsequently relayed to the surrounding uninfected

cells by a signalling molecule known as cytokines, in a system known as the inflammatory

response (Arnold et al., 1994). Widely-recognised cytokines include interleukin (IL), tumour

necrosis factor (TNF), interferon (IFN), and chemotactic cytokine (chemokine) (Kindt et al.,

2007). An innate immune response generally conforms to the following cascade: recognition

of pathogen-associated molecular patterns (PAMPs) by pathogen recognition receptors

(PRRs), followed by the induction of signal transduction pathways involving transcription

factors, kinases, and adaptor molecules, which in turn regulate the expression of genes coding

for antiviral responses such as pro-inflammatory cytokines (Katze et al., 2008, Mogensen,

2009).

1.5.2 Innate immune responses against RSV infection

The hallmark of innate immunity against RSV infection in the epithelial cells of the

respiratory tract is the time- and dose-dependent secretions of two chemokines, interleukin-8

(IL-8) and regulated upon activation, normal T-cell expressed, and secreted (RANTES)

(Garofalo et al., 1996, Saito et al., 1997, Harrison et al., 1999, Domachowske et al., 2001).

Chemokines are chemotactic factors of leukocytes, with IL-8 is the chemoattractant of

neutrophils and RANTES is the chemoattractant of eosinophil (Olszewska-Pazdrak et al.,

1998, Garofalo and Haeberle, 2000). The phagocytic role and cytotoxic granule products of

leukocytes contribute to viral clearance by killing infected cells; however, their hyper-

activation, which is caused by excessive concentration of chemokines at inflammatory sites,

can affect uninfected cells and lead to tissue damage and subsequent exacerbation of illness,

which in the case of RSV includes mucus overproduction and airway epithelial cells necrosis

(bronchitis/bronchiolitis) (Sekido et al., 1993, Mukaida et al., 1998, Harrison et al., 1999,

Miller et al., 2003, Kindt et al., 2007). IL-8 and RANTES accumulation has been observed to

be higher in infants with severe RSV bronchiolitis than in those without bronchiolitis (Noah

et al., 2002, Smyth et al., 2002).

As the secretion level of IL-8 and RANTES dictates the subsequent inflammatory response

and, potentially, the eventual clinical symptoms; it is possible to use their expression as an

Page 37: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

24

early predictor of the pathogenesis of RSV-associated disease and as a model of the immune

response and disease development against infection with an uncharacterised RSV strain. The

synthesis of IL-8 and RANTES is preceded by the activation of transcriptional factor NF-κB,

and is augmented by the presence of IFN-α/β (Mukaida et al., 1994, Garofalo et al., 1996,

Thomas et al., 1998, Genin et al., 2000, Tian et al., 2002, Spann et al., 2005). RANTES

expression is also induced by the activation of IRF3, which firstly leads to the production of

IFN-β (Bao et al., 2008).

NF-κB is natively cytoplasmic due to sequestration by the inhibitor of κB (IκB) that masks its

NLS and DNA binding domain (Bonizzi and Karin, 2004). IκB degradation by the inhibitor

of κB kinase (IKK) unmasks the NLS; therefore allowing Imp-α5 to translocate IκB into the

nucleus (Bose and Banerjee, 2003). The NF-κB binding site in RANTES gene is located in

the 5’-untranslated region, specifically at nucleotide -32 of the start codon of the first exon;

while the NF-κB binding site in IL-8 gene is situated at nucleotide -80 – (-)70 of the start

codon of exon 1 (Nelson et al., 1993, Mukaida et al., 1994).

1.5.3 Activation of NF-κB transcription factor

RSV is recognised in two ways: in the cytoplasm by the interferon (IFN)-inducible double-

stranded RNA (dsRNA)-activated serine-threonine protein kinase (PKR), and at the cellular

membrane, where RSV F protein is recognised by Toll-like receptor (TLR) 4 (Kurt-Jones et

al., 2000, Zamanian-Daryoush et al., 2000, Haynes et al., 2001, Bose and Banerjee, 2003,

Gern et al., 2003). PKR activation is replication-dependent; while TLR4 activation is

replication-independent (Fiedler et al., 1996, Olszewska-Pazdrak et al., 1998, Haeberle et al.,

2002, Miller et al., 2003). Chemokine expression is biphasic, where the weak early peak of

expression is the result of induction following viral entry, which is recognition of RSV F by

TLR4, and that the more prominent late production comes from the development of immune

response against RSV replication; which corresponds to the PKR activation (Miller et al.,

2003, Spann et al., 2005).

In PKR-activated pathway, dsRNA activates PKR by inducing dimerisation and

autophosphorylation, which in turn phosphorylates the IKK complex (Zamanian-Daryoush et

al., 2000). Activated IKK complex catalyses I-KB phosphorylation and degradation by 26S

proteasome to promote dissociation from the NF-κB subunits in order to expose the NLS and

DNA binding domain in NF-κB (Bose and Banerjee, 2003, Bonizzi and Karin, 2004). In

Page 38: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

25

TLR4-activated pathway, the extracellular part of TLR4, leucine-rich repeat region (LRR),

initially binds to RSV F and the cytoplasmic part, Toll/interleukin-1 receptor homology (TIR)

domain, binds to an adaptor, which is the myeloid differentiation primary response gene 88

(MyD88) (Mogensen, 2009). MyD88 recruits IL-1 receptor-associated kinases (IRAKs) 4 and

1 by binding its death domain to IRAK4’s; following which the IRAK pair

autophosphorylates and IRAK1 recruits and activates TNF receptor-associated factor 6

(TRAF6) (Mogensen, 2009). Phosphorylated (p)IRAK1 dissociates the heterodimer IRAK-

MyD88 complex by forming a new complex consisting of IRAK1, TRAF6, TGF-β-activated

kinase 1 (TAK1), TAK1-binding protein 2 (TAB2) and TAK1-binding protein 3 (TAB3);

then the newly-formed complex translocates to the cytoplasm and TAK1 activates the IKK

complex (Takeda and Akira, 2004).

1.5.4 Activation of IRF3 transcription factor

IRF3 is activated following recognition of RSV 5’-triphosphate (ppp) ssRNA by retinoic-acid

inducible gene I (RIG-I) (Faul et al., 2010). The RIG-I DEx(D/H) box RNA helicase domain

binds to the RNA, while its N-terminal caspase recruitment domain (CARD) binds to the

CARD of an adaptor, IFN-β promoter stimulator 1 (IPS-1; also known as virus-induced

signalling adaptor (VISA) or mitochondrial antiviral signalling protein (MAVS) or Cardif)

(Randall and Goodbourn, 2008, Takeuchi and Akira, 2008, Mogensen, 2009). The IPS-I

mediates an association with TNF-receptor-associated factor 3 (TRAF3), which then recruits

TANK-binding kinase I (TBK-I) and inducible IκB kinase (IKKϵ) (Hiscott, 2007). The

kinases activate IRF3 at C-terminal region, driving its dimerization and nuclear translocation

to join a transcription factor complex consisting of NF-κB, p300/CBP, and ATF-2/c-Jun

heterodimer to promote the transcription of the early-expressed IFN-β (Hiscott, 2007, Katze

et al., 2008, Randall and Goodbourn, 2008, Takeuchi and Akira, 2008, Mogensen, 2009).

The release of early IFN-β signals for the production of IFN-α/β in neighbouring cells by

associating with their receptors, the interferon α receptor 1 and 2 (IFNAR1/2), to activate

receptor-associated JAK1 and Tyk2 tyrosine-kinases (Platanias, 2005, Randall and

Goodbourn, 2008). Tyk2 phosphorylates Y466

of IFNAR1 in order to create docking site(s)

for the incoming STAT2, as well as phosphorylates Y690

of STAT2 in preparation for

association with STAT1. Meanwhile, JAK1 phosphorylates Y701

of STAT1 (Platanias, 2005,

Randall and Goodbourn, 2008). The phosphorylated (p)STAT1 and pSTAT2 form a stable

Page 39: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

26

heterodimer, in which the Imp-α5-dependent NLS in STAT1 is exposed and NES in STAT2

is inactivated (Platanias, 2005, Vidy et al., 2005, Randall and Goodbourn, 2008). The

pSTAT1/pSTAT2 complex detaches from the IFNAR1/IFNAR2 oligomer and binds to the

DNA binding protein p48 (IFN regulatory factor 9 (IRF9)) to form the IFN-stimulated

growth factor 3 (ISGF3) (Platanias, 2005, Randall and Goodbourn, 2008). The ISGF3 then

translocates to the nucleus and binds to IFN-stimulated response elements (ISREs) that is

located in the promoters of IFN-stimulated genes (ISGs); therefore activating the

transcription of pro-inflammatory mediators (Platanias, 2005, Randall and Goodbourn, 2008).

1.6 RSV vaccine

A licensed RSV vaccine is currently not available due to a major difficulty. The population

having the highest risk of developing severe LRTIs is infants younger than 3 months old, who

may not respond adequately to the vaccination due to suppression of immune response by

maternal antibody or immaturity of the immune system (Dudas and Karron, 1998). In

addition, it is important to vaccinate other age groups to control the transmission of RSV as

these act as the reservoir of infection, such that the introduction of infection to the main target

group can be limited (Haydon et al., 2002).

Other problems that slow the progression of RSV vaccine as described by Collins and

Murphy (2002) include the following. Firstly, the presence of maternally-derived antibody

presents hurdles in evaluating the safety and efficacy of the vaccine, since it is impossible to

find infants who are RSV-naïve. Secondly, RSV does not replicate efficiently in vitro and is

structurally frail, such that cautious handling is required to maintain its infectivity.

In addition to difficulties in handling RSV, significant obstacle to developing RSV vaccines

has been the experience with formalin-inactivated (FI) RSV, which elicits partial immunity,

as reviewed in Dudas and Karron (1998). FI-inactivated RSV induced exaggerated clinical

symptoms in infants who were RSV-naïve before vaccination since formalin inactivation

selectively altered the epitopes in the F and G surface glycoproteins. This alteration resulted

in inadequate levels of neutralising antibodies in serum and induced the production of IL-4,

IL-5, and IL-10, followed by the influx of lymphocytes and eosinophil, and the release of

additional mediators; the combination of which caused inflammation and airway constriction

that leads to bronchiolitis.

Page 40: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

27

Further, an ideal RSV vaccine would be in the form of live-attenuated virus that is

administered via the intranasal route. Live-attenuated vaccine is expected to induce an

immune response that closely resembles the response to natural infection, without causing

severe disease. Intranasal administration can induce both local mucosal and systemic

immunity; therefore is expected to overcome the suppressive effects of maternal serum

antibodies (Collins and Murphy, 2005).

Technology used to achieve attenuation must serve the purpose that the best vaccine would

be highly immunogenic, such that it can be given to people from all age groups, but must not

cause disease following natural infection. Attenuated strains are generally less immunogenic

than clinical isolates, because of, in part, reduced replication and antigen production in vivo

(Collins and Murphy, 2005). Therefore, it is important to ensure that the attenuation

technology can fulfil the requirements of suitable RSV vaccine.

Conventional methods to attenuate RSV have been based on strain RSV A/Australia/A2/1961

via combination of extensive in vitro passaging in multiple temperatures and chemical

mutagenesis to result in cold passaged (cp) and temperature sensitive (ts) mutants (Dudas and

Karron, 1998). The mutants are expected to be restricted at human body temperature (37o C),

but replicate efficiently at suboptimal temperature. However, the best candidate has not been

discovered yet, with in vivo characterisation of these mutants revealing either under or over

attenuation (Karron et al., 1997b).

The progression in molecular biology techniques, known as reverse genetics, allows for a

method to generate recombinant RSV strain A2 (rRSV A2) containing attenuating mutations.

This technique allows for prediction of attenuation and fine-tunes the degree of attenuation at

the stage of designing the recombinant virus. Attenuation can be achieved by point mutation,

gene deletion, targeting a specific protein and replacing charged amino acids with a non-

charged amino acid, or changing the gene order (Collins and Murphy, 2002). Recombinant

RSV is generated from cDNA by expression in Vero cells, which are approved for vaccine

production according to the principle that the attenuating mutation should not reduce virus

yield in vitro since RSV does not replicate to a high titre and reduction of replication

efficiency in vitro could interfere with vaccine manufacture (Collins and Murphy, 2005).

Page 41: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

28

1.7 Human metapneumovirus (hMPV)

The other human virus that is taxonomically closest to RSV is human metapneumovirus

(hMPV). Although hMPV was only recently identified in 2001 from a 20-year collection of

respiratory secretions obtained from children who suffered from respiratory tract infection,

seroprevalence analysis indicated that it has been circulating in human population at least

since 1958 (van-den-Hoogen et al., 2001). hMPV infects all age groups and commonly

causes ILI, although the prevalence and disease pathogenesis is heightened in the population

of infants/very young children (less than 2 years old), the elderly, and the

immunocompromised (van-den-Hoogen et al., 2001, Boivin et al., 2002, van-den-Hoogen et

al., 2004b, Williams et al., 2004, Falsey et al., 2006, Sarasini et al., 2006, Liao et al., 2012).

hMPV particles are thought to be spread by large respiratory secretions and fomites, are

stable at room temperature for 6 hours, and can resist 10 freeze-thaw cycles (Broor et al.,

2008, Papenburg and Boivin, 2010, Tollefson et al., 2010). The development of a hMPV

vaccine is complicated by the fact that the population most at risk of developing severe

clinical manifestations is the least able to develop a sufficient immune response.

1.7.1 Epidemiology

Globally, hMPV circulates all year round, but in the temperate regions it has a seasonal peak

that tends to be approximately 2 months later than that of RSV, occurring mostly during late

winter to early spring (Falsey et al., 2003, Esper et al., 2004, Gray et al., 2006, Kahn, 2006,

Sloots et al., 2006, Feuillet et al., 2012). RSV and hMPV co-infection, which is enabled by

the overlapping temporal distribution, has been attributed as the risk factor in the

development of severe RSV-associated bronchiolitis (Greensill et al., 2003, Semple et al.,

2005). As a single aetiology, hMPV is the second most common cause of bronchiolitis-

associated hospitalisation in infants and young children after RSV (Kahn, 2006, Gaunt et al.,

2009). Global annual epidemic of hMPV may be caused by many different strains in separate

regions; which differs from influenza virus, where the yearly epidemic is caused by 2 – 3

strains that circulate simultaneously worldwide (Esper et al., 2004, Williams et al., 2004,

Kahn, 2006). Re-infection is common, very possibly reflecting short-term immunity due to

the high genetic variability of the hMPV and the rapid evolution of the circulating genotypes

that overcome pre-existing immunity in the population (Williams et al., 2004).

Page 42: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

29

1.7.2 hMPV infection

The main replication site of hMPV is the ciliated epithelial cells of the respiratory tract,

which span from the nasal cavity to bronchioles; however hMPV also replicates in the type 1

pneumocytes (Kuiken et al., 2004). Viral replication eliminates ciliation, causes the

sloughing-off of necrotic epithelial cells, recruits neutrophils, and induces the rare formation

of syncytia (Kuiken et al., 2004). The clinical symptoms of infection range from mild upper

respiratory tract illnesses, which encompass fever, cough, rhinorrhoea (runny nose), and

rhinitis (nasal inflammation), to the more severe lower respiratory tract illnesses including

bronchitis and pneumonia (van-den-Hoogen et al., 2001, Esper et al., 2004, Williams et al.,

2004). Additional symptoms include wheezing, myalgia (muscle pain), diarrhoea, and

vomiting (van-den-Hoogen et al., 2001, Crowe-Jr. and Williams, 2011). Overall, the

characteristics of hMPV infection are quite indistinguishable from RSV infection; although

hMPV infections are likely to be milder among immunocompetent adults, less associated

with pneumonia, inclined to be associated with hospitalisation in an older age group (median

age 6 – 12 months (hMPV) vs 6 months (RSV)), and more associated with exacerbation of

asthma (Boivin et al., 2002, Falsey et al., 2003, Williams et al., 2004, Papenburg and Boivin,

2010, Li et al., 2012). The severe hMPV-associated illness in infants/children less than 2-year

old and in the elderly, very likely is the consequence of deficient immune responses (van-

den-Hoogen et al., 2004b, Liao et al., 2012). In addition, the degree of disease severity has

been associated with nasopharyngeal viral load, with a higher viral titre is linked to the

development of LRTIs and results in hospitalisation (Bosis et al., 2008).

In cell culture, hMPV replicates more slowly than RSV, undergoes a delayed CPE

development, is trypsin-dependent, and tends to grow to a lower peak titre (van-den-Hoogen

et al., 2001, Tollefson et al., 2010, Crowe-Jr. and Williams, 2011). These characteristics are

often claimed as the cause of the recent discovery of hMPV despite its long-term circulation

in the population. CPEs are generally observed at days 10 –14 post infection, and include the

formation of syncytia, followed by rapid internal disruption of the cells and subsequent

detachment of the cells from the surface of culture plate (van-den-Hoogen et al., 2001, Boivin

et al., 2002, van-den-Hoogen et al., 2004b, Williams et al., 2004). hMPV can be propagated

in epithelial cells, such as Vero, Vero E6, A549, and LLC-MK2 cell lines; the most

permissive cell lines being the trypsin-resistant LLC-MK2 of Macaca mulatta kidney and

Page 43: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

30

BEAS-2B of human bronchus (van-den-Hoogen et al., 2001, Tollefson et al., 2010). Syncytia

formation is cell-type dependent (Deffrasnes et al., 2005).

1.7.3 Taxonomy

Based on the relatedness of nucleotide sequences, hMPV is classified into the family

Paramyxoviridae, subfamily Pneumovirinae, and genus Metapneumovirus (van-den-Hoogen

et al., 2001, van-den-Hoogen et al., 2002). Based on the variability in structural genes,

including N, P, L, F, and G, hMPV is consistently classified into two genetic lineages: the

genotypes A and B, with subgenotypes A1, A2, B1, and B2 inside each genotype (Peret et al.,

2002, van-den-Hoogen et al., 2004a). hMPV is the taxonomically closest human pathogen to

RSV; the other member of the Metapneumovirus genus is an animal virus, the avian

metapneumovirus (aMPV) previously known as turkey rhinotracheitis virus (TRTV) or avian

pneumovirus (APV).

1.7.4 The structure of an hMPV particle (the virion)

The morphology of hMPV resembles RSV, i.e pleomorphic, spherical, or filamentous, in

which a nucleocapsid structure is contained within a space that is sheathed by an envelope

containing projections of glycoproteins (Peret et al., 2002).

The hMPV genome is a nonsegmented (-)ssRNA of 13.3 kb, is equipped with noncoding 3’-

leader and 5’-trailer sequences, contains 8 genes namely 3’-N-P-M-F-M2-SH-G-L-5’ that

code for 9 proteins, which in order of the gene sequence are the nucleoprotein (N),

phosphoprotein (P), matrix (M) protein, fusion (F) protein, transcription elongation factor

(M2-1), RNA synthesis regulatory factor (M2-2), small hydrophobic surface protein (SH),

major attachment glycoprotein (G), and large polymerase subunit (L) (van-den-Hoogen et al.,

2002). Each gene is flanked by a well conserved GS and a less conserved GE sequences

(Bastien et al., 2003).

In hMPV-infected cells, the N, P, and L proteins constitute a structure central for genome

replication known as ribonucleoprotein (RNP) complex, which appears as the inclusion

bodies in the cytoplasm, with the N and P protein association being the core of the inclusion

(Collins and Crowe Jr., 2007, Derdowski et al., 2008). Unlike in RSV, hMPV M2-1 is not

included in the RNP complex, does not bind to N, is not required for in vitro growth–

although is required for the optimum in vivo replication, and does not function as the

Page 44: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

31

antitermination of transcriptional read-through (Herfst et al., 2004, Buchholz et al., 2005,

Derdowski et al., 2008).

Consistent with the general structure of Paramyxoviruses, the replication complex is sheathed

by a lipid bilayer envelope that is derived from the plasma membrane of the host cell, and

contains the F, G, and SH glycoproteins (Lamb and Parks, 2006). The homotrimeric-arranged

F protein is the hMPV major neutralisation antigen, is first synthesised as an F0 precursor that

is subsequently cleaved into the F1 and F2 subunits, functions in the viral attachment and

fusion, determines cell tropism, and induces syncytia formation (Crowe-Jr. and Williams,

2011). hMPV F cleavage site is unique, consisting of a R-Q-X-R motif compared to the

common R/K-X-R/K-R furin cleavage site found in the Paramyxovirus F, and is possibly not

cleaved by furin protease such that trypsin supplementation is required for in vitro growth

(van-den-Hoogen et al., 2002, Bastien et al., 2003, Collins and Crowe Jr., 2007). The hMPV

G and SH are not required for in vitro growth; but, the hMPV G is involved in the activation

of NF-κB and IRF3 in vitro (Biacchesi et al., 2004, Bao et al., 2008).

The hMPV M protein and its biology has so far been deduced from the biology of RSV M,

such as being located between the RNP complex and the envelope glycoproteins, binding to

the cytoplasmic tail of the envelope glycoproteins, and having a significant role in virion

assembly by binding to the nucleocapsid complex and ceasing RNA synthesis (Collins and

Crowe Jr., 2007, Crowe-Jr. and Williams, 2011). However, hMPV M has been hypothesised

to induce the secretion of proinflammatory mediators in vitro by promoting the maturation of

dendritic cells (Bagnaud-Baule et al., 2011).

There is not much known about hMPV M2 gene products, but they are predicted to control

RNA synthesis based on the roles of M2-1 and M2-2 proteins of RSV, where RSV M2-1

promotes the read-through of intergenic junction during transcription and M2-2 acts as the

controlling switch between RNA replication and transcription (Hardy and Wertz, 1998,

Bermingham and Collins, 1999). hMPV M2-2 has been shown to upregulate RNA

transcription, and together with M2-1, is not important for in vitro growth although is

required for optimal in vivo replication (Buchholz et al., 2005).

Page 45: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

32

1.7.5 The life cycle of hMPV

The replicative cycle of hMPV has not been studied in detail, but is very likely to be similar

to that of RSV (Crowe-Jr. and Williams, 2011). Replication processes take place in the

cytoplasm and encompass the adsorption/entry, RNA synthesis, and assembly/budding

processes. Notable differences are that the kinetics of hMPV infection are slower compared

to other Paramyxoviruses: the peak of hMPV intracellular protein expression is 48 – 72 hours

post infection while Paramyxoviruses commonly complete a growth cycle in 14 – 30 hours

(Lamb and Parks, 2006). Compared to RSV, the CPE is less prominent and the yield of

infectious particles is approximately 5 – 10 fold lower (Collins and Crowe Jr., 2007).

1.7.6 The role of N protein in virus replication

The N protein of nonsegmented (-)ssRNA viruses associates directly with the genomic/(-) or

antigenomic/(+) strands of RNA in infected cells as well as within virus particles

(Ball, 2007). The known functions of N-RNA interaction include: physical protection for the

RNA, regulation of template fidelity, formation of viral replication complex, and RNA

packaging in preparation of virion assembly/budding and maturation.

1.7.6.1 RNA protection

The N protein protects RNA against ribonuclease degradation and high concentration of salts,

as well as maintains the rigidity of RNA structure following cycles of

folding/dissociation/refolding during replication cycle (Lynch and Kolakofsky, 1978, Mir and

Panganiban, 2006, Raymond et al., 2010). The N subunits self-associate, are aligned in a

continuous tunnel, and cover the full length of viral RNA, such that the closed conformation

prevents accesses to RNA except for a very short time when the conformation is altered to

allow copying by RNA polymerase (Green et al., 2011).

1.7.6.2 Regulation of template fidelity

The N protein functions in regulating the fidelity of viral genome by maintaining the ratio of

association between N subunits and ribonucleotides, which serves as a screening mechanism

to eliminate pseudo genomes containing non-templated insertions from the genome pool

(Barr and Fearns, 2010). Little is known about the N:ribonucleotide ratio in hMPV; known

stoichiometry include one N molecule binds to nine ribonucleotides (1:9) in Rhabdoviridae,

Page 46: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

33

1:12 in Bunyaviridae, and 1:6 in Paramyxoviridae (Calain and Roux, 1993, Mohl and Barr,

2009). In Paramyxoviruses, pseudo genomes that arise from the copying errors of RNA

polymerase and do not follow the N:ribonucleotides stoichiometry, decrease RNA synthesis

by approximately 50 – 100 fold (Kolakofsky et al., 1998).

1.7.6.3 Formation of replication complex/ ribonucleoprotein (RNP) complex

The N protein is the driving force for the formation of RNP complex due to the propensity to

self-associate and because N possesses binding sites for other RNP components, including

RNA, P, and L (Becker et al., 1998, Coronel et al., 2001, DiCarlo et al., 2007, Raymond et

al., 2010, Green et al., 2011). N has an inherent ability to interact with P and this interaction

is particularly important because N-RNA association is not specific, while N-P conformation

confers specificity for encapsidation such that N-P associated RNA is the form that can be

recognised by RNA polymerase (Baric et al., 1988, Howard and Wertz, 1989, Myers et al.,

1999, Yoo et al., 2003, Nayak et al., 2009). In hMPV, the N-P interaction is also required to

prevent N condensation due to self-oligomerisation (Derdowski et al., 2008). This interaction

has been shown to result in the very early formation of cytoplasmic inclusion bodies in

hMPV and RSV, once the first molecules of N and P proteins have been translated (García-

Barreno et al., 1996, Derdowski et al., 2008).

1.7.6.4 RNA packaging in assembly, budding, and particle release

The packaging signal in N activate the packaging signal in RNA by masking the polymerase

recognition site that overlaps with RNA packaging signal during the late stage of infection,

i.e once enough N protein has accumulated (Fosmire et al., 1992, Rein et al., 1994).

Following genome packaging, N binds to M in order to stabilise the replication complex and

facilitates interaction with plasma membrane for the release of new particles (Stricker et al.,

1994, Coronel et al., 2001, Dancho et al., 2009). M-N interaction is specific, that is, M only

binds to N that encapsidates full-length RNA; thus ensuring the released progeny virions are

infectious and have a fully-functioning genome (Narayanan et al., 2000).

hMPV N is coded by a 1185-bp gene and contains 395 amino acids with a molecular mass of

43.5 kDa (Bastien et al., 2003, Petraityte-Burneikiene et al., 2011). hMPV N has important

function for the success of viral proliferation as shown by transcriptional silencing assay

Page 47: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

34

using siRNA, where the absence of hMPV N decreases viral load in vitro and in vivo (Darniot

et al., 2012).

1.7.7 The nucleocytoplasmic transport of nucleocapsid proteins

Viruses with (-)ssRNA genome naturally replicate in the cytoplasm; therefore there is a need

for the N protein to localise in the cytoplasm as it forms the central part of viral replication

complex. However, the N protein of several cytoplasmic-replicating viruses has been shown

to localise in the nucleus or nucleolus of infected cells during certain stages of infection. The

N nuclear/nucleolar localisation usually takes place early in the infectious cycle as soon as it

is translated, possibly to play non-structural functions, and then returns in the cytoplasm in

the late stage of infection to participate in assembly (Tijms et al., 2002, Yoo et al., 2003).

1.7.7.1 Examples of viruses in which N protein undergoes nucleocytoplasmic

transport

The N protein of equine arteritis virus (EAV), porcine reproductive and respiratory

syndromes virus (PRRSV), severe acute respiratory syndrome coronavirus (SARS Co-V),

infectious bronchitis virus (IBV), Borna disease virus (BDV), and some Paramyxoviruses:

canine distemper virus (CDV), rinderpest virus (RPV), and measles virus (MV) has been

shown to localise in the nucleus or nucleolus during infection.

The EAV N nuclear import has not been clearly understood, but the nuclear export is

facilitated by Crm1 (Tijms et al., 2002). The PRRSV N possesses NLS, NoLS, and NES

motifs that facilitate independent nucleocytoplasmic transport and accumulation in the

nucleoli of infected cells, where it binds to the 18S and 28S rRNAs and associates to

fibrillarin, an RNA-associated nucleolar protein that plays a role in assembly of ribosomes

and regulation of cell cycle (Rowland et al., 1999, Yoo et al., 2003). The nuclear/nucleolar

localisation does not affect the capability to produce infectious particles, but attenuates the

amount of the released particles and in vivo pathogenesis (Lee et al., 2006). The IBV N also

interacts with, and even reorganises fibrillarin distribution, where the interaction and/or

redistribution are associated with delay in cell growth (Hiscox et al., 2001, Chen et al., 2002).

There are three putative NLSs and a Crm1-independent NES that have been identified in

SARS Co-V N (You et al., 2005). The NES is the predominant signal and SARS Co-V N

cytoplasmic localisation has been shown to be facilitated by 14-3-3 (tyrosine 3-

Page 48: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

35

monooxygenase/tryptophan 5-monooxygenase activation protein) that recognised

phosphorylated serine residues within RSXS*XP motif (Macara, 2001, Surjit et al., 2005).

The genome of BDV is replicated in the nucleus and the N protein is equipped with NLS and

Crm1-associated NES, such that N can facilitate the nuclear import of RNP complex for

replication and nuclear export to the cytoplasm for virion assembly (Kobayashi et al., 2001).

The N proteins of CDV and RPV possess conserved NLS and NES motifs, where the NES,

although a leucine-rich motif, is Crm-1 independent (Sato et al., 2006). MV is a virus whose

inclusion bodies were produced in the nucleus of infected cells. More importantly, the

nuclear translocation of MV N protein also affects the host immune response, which is done

by inhibiting the nuclear translocation of STATs without interfering the nuclear transport

factor of STATs, Imp-α5; therefore causing disruption of the IFN-α/β and IFN-γ signalling

pathways (Takayama et al., 2012).

1.7.7.2 The possible functions of nuclear and/or nucleolar localisation of N

The nuclear/nucleolar translocation of the N protein of viruses that replicate in the cytoplasm

is mostly associated with optimisation of replication. For example IBV N redistributes

fibrillarin and delays cytokinesis to divert biosynthetic resources from the dividing nucleus to

the cytoplasm, where viral replication takes place (Chen et al., 2002).

Other possibilities include ‘accidental’ translocation schemes, which include the molecular

mimicry of nuclear or nucleolar localisation signal (NLS/NoLS), the association with

ribosomal proteins, and passive diffusion. In molecular mimicry, the K/R-rich RNA binding

domains may resemble NLS/NoLS motifs, allowing the N protein to be transported into the

nucleus by a transport factor (Rowland and Yoo, 2003). Association with ribosomal proteins

in cytoplasm might result in N being coincidentally translocated into the nucleolus, where the

ribosomal proteins associate with rRNA to form ribosomal subunits (Tijms et al., 2002). The

NPC allows for passive diffusion for proteins that size less than 50 kDa or 5 nm (Wurm et al.,

2001). The accidental localisation might serve a purpose by maintaining the balance of

N:ribonucleotides stoichiometry in the cytoplasm in order to produce functional genomes,

since N can be overproduced and transient nuclear/nucleolar localisation can be a means to

keep the appropriate concentration of N molecules in the cytoplasm (Rowland and Yoo,

2003).

Page 49: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

36

1.8 Hypotheses and aims

The need for a safe and immunogenic live-attenuated RSV vaccine has led to the use of

reverse genetics to facilitate efficient vaccine production. Reverse genetics allows for

achieving attenuation from altering the genomic composition of RSV, which is done by

introducing mutation in the viral genome, followed by rescuing a recombinant virus in Vero

cells transfected with cDNAs of the mutated genome. The mutation targets in the genome are

regions, or sites, that control important functions for RSV replication.

The nucleocytoplasmic transport of structural proteins facilitates the replication processes in

many viruses; making the nucleocytoplasmic transport of viral proteins a viable target for

attenuation. In RSV M, timely and appropriate nucleocytoplasmic transport of the M protein

has been shown to be important for determining the success of replication. Disruption of the

nuclear import leads to decreased particle production and disruption of the nuclear export

ceases the infection process. As a result, the RSV M nucleocytoplasmic transport can be

targeted for attenuation. One mechanism is to alter the components of nucleocytoplasmic

transport in the structure of RSV M, such as the NLS, NES, or regulatory phosphorylation

sites.

The success of RSV M nucleocytoplasmic transport is determined by the functionality of the

NLS, NES, and the regulatory sites. A potential CK2 phosphorylation site was identified

adjacent to the NES, M205(T). Since RSV M nucleocytoplasmic transport is regulated by a

phosphorylation mechanism (Alvisi et al., 2008), the role of RSV M205 in virus replication

can be assessed by mutating the positively charged threonine (T) residue into the neutrally

charged alanine (A)

The same approach can also be applied to attenuate other viruses where nucleocytoplasmic

transport of a structural protein serves a function in replication. Therefore, this study attempts

to assess the potential of attenuating hMPV, a virus closely related to RSV, by interfering

with the nucleocytoplasmic transport of the N protein. Previous work in the laboratory has

shown that hMPV N protein can localise in the nucleus in the very late stage of infection (day

5 post infection) (Ghildyal, R & Gahan, M; unpublished); therefore, it is important to gather

evidence in order to facilitate further study.

Page 50: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

37

The hypotheses of this study are:

1. Mutation in a CK2 phosphorylation motif adjacent to the NES reduces the production

of infectious progeny and dampens the activation of pro-inflammatory responses.

2. The hMPV N protein has inherent nuclear transport capability.

The aims of this study are:

1. To determine the impact of mutation at a CK2 phosphorylation motif adjacent to the

NES on virus replication and on the activation of pro-inflammatory responses in cell

culture.

2. To examine the nuclear localisation of hMPV N in transfected cells.

Page 51: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

38

Page 52: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

39

CHAPTER 2

MATERIALS AND METHODS

2 Materials and Methods

Page 53: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

40

Page 54: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

41

2.1 Materials

2.1.1 Cell lines

Table 2-1 Cell lines used in this study

2.1.2 Viruses

Table 2-2 Viruses used in this study Virus Strain Description Reference

Respiratory syncytial

virus

rRSV A2 Recombinant RSV strain A2 (Collins et al.,

1995)

rRSV A2 M(T205A) Recombinant RSV strain A2 with TA

substitution at M 205*

(Ghildyal et al.,

2009)

Human

metapneumovirus

hMPV CAN97-83 Clinical isolate from a patient with

respiratory diseases in Quebec, Canada

GenBank accession number AY297749

(Peret et al., 2002)

*NES-associated-CK2-phosphorylation-motif of the M protein

2.1.3 Plasmids and bacterial cultures

Table 2-3 Plasmid/bacteria used in this study

Plasmid/bacteria Source Description pDONR®207 [Invitrogen, Carlsbad, USA] Entry vector

pEPI-DESTC [R. Ghildyal, University of Canberra, Canberra, Australia]

(Ghildyal et al., 2005a)

Expression vector

E. coli ElectroMAXTM

DH10BTM

[Invitrogen, Carlsbad, USA] Competent cells

Cell line Source ATCC®

Number

Description

Vero E6 Epithelial cells from the kidney of

Cercopithecus aethiops (African green

monkey)

CRL-1586TM Lack functional type I interferon

response

A549 Carcinoma epithelial cells from human lungs CCL-185TM Active type I interferon

signalling pathway

Cos-7 Cells from the kidney of Cercopithecus

aethiops (African green monkey) that are transformed with SV40 T-antigen

CRL-1651TM Transfection host cell line

Page 55: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

42

2.1.4 Primers

Table 2-4 Primers used in this study Use Primer name Template Sequence (5’ 3’) Reference Supplier

Sequencing pEPIseqFWD pEPI-DESTC GCC CCG TGC TGC TGC CCG (Ghildyal et

al., 2005a)

[GeneWorks,

Hindmarsh, Australia]

pEPIseqREV pEPI-DESTC GTG AAA AGA AAG TAT ACC

DONRFWD pDONR®207. TCG CGT TAA CGC TAG CAT GGA TCT C

DONRRev pDONR®207 GTA ACA TCA GAG CTT TTG AGA CAC

HMPV83Nf_inner hMPV.N TGA TGC ACT CAA AAG ATA CCC This study

HMPV83Nr_inner hMPV.N GTT TGA CCA GCA CCA TAA GC

PCR/cloning attB1RSVM1 RSV M *GGG GAC AAG TTT GTA CAA AAA AGC

AGG CTT GGA AAC ATA CGT GAA CAA GC

(Ghildyal et

al., 2005a)

attB2RSVM256 RSV M *GGG GAC CAC TTT GTA CAA GAA AGC

TGG GTT AAT CTT CCA TGG GTT TGA TTG C

HMPV83NattB1 hMPV N *GGG GAC AAG TTT GTA CAA AAA AGC

AGG CTC ATC TCT TCA AGG GAT TCA CCT

GAG

This study

HMPV83NattB2 hMPV N *GGG GAC CAC TTT GTA CAA GAA AGC

TGG GTC TTA CTC ATA ATC ATT TTG ACT

GTC GTC AC

hMPVN_f_attB1 hMPV N *GGG GAC AAG TTT GTA CAA AAA AGC AGG CTT GAT GCA CTC AAA AGA TAC CC

hMPVN_r_attB2 hMPV N *GGG GAC CAC TTT GTA CAA GAA AGC TGG GTG TTT GAC CAG CAC CAT AAG C

hMPVN_f-

rev_attB2

hMPV N *GGG GAC CAC TTT GTA CAA GAA AGC

TGG GTG GGT ATC TTT TGA GTG CAT CA

Mycoplasma

test

Myco F1 Mycoplasma sp. ACA CCA TGG GAG CTG GTA AT

Myco R1 Mycoplasma sp GTT CAT CGA CTT TCA GAC CCA AGG CAT

Myco F2 Mycoplasma sp GTT CTT TGA AAA CTG AAT

Myco R2 Mycoplasma sp GCA TCC ACC AAA AAC TCT

*bold characters = attB sequences

Page 56: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

43

2.1.5 Enzymes/buffers for molecular biology assays

Table 2-5 Enzymes used in this study

Enzymes Manufacturer Mango TaqTM DNA polymerase [Bioline, Alexandria, Australia]

Gateway® BP ClonaseTM II Enzyme Mix [Invitrogen, Carlsbad, USA]

Gateway® LR ClonaseTM II Enzyme Mix [Invitrogen, Carlsbad, USA]

2.1.6 Antibodies

Table 2-6 Antibodies used in this study

Antibodies Target Manufacturer Use Mouse anti-RSV fusion protein

monoclonal antibody

RSV F [Millipore, Billerica,

USA]

RSV titration

Ms X Hu Metapneumovirus hMPV nuclear

protein

[Millipore, Billerica,

USA]

hMPV titration

Goat anti-mouse IgG-alkaline

phosphatase

Mouse IgG [Sigma-Aldrich, St.

Louis, USA]

RSV and hMPV

titration

2.1.7 Kits

Table 2-7 Kits used in this study

Kits Manufacturer Vector® Black Alkaline Phosphatase Substrate Kit II [Vector Laboratories Inc., Burlingame, USA]

cDNA Synthesis Kit [Bioline, Alexandria, Australia]

Wizard® SV Gel and PCR Clean-Up System [Promega, Madison, USA]

Human RANTES ELISA Development Kit [Peprotech, Rocky Hill, USA]

Human IL-8 ELISA Development Kit [Peprotech, Rocky Hill, USA]

QIAprep® Spin Miniprep Kit [Qiagen, Hilden, Germany]

QIAGEN Plasmid Midi Kit [Qiagen, Hilden, Germany]

Page 57: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

44

2.1.8 Buffers, media, and solutions

Table 2-8 Culture media used in this study

Media Composition

Cell culture Methyl cellulose 2% methyl cellulose [Sigma-Aldrich, St. Louis, USA]; 15% HBSS [Sigma-

Aldrich, St. Louis, USA]; 2% FBS [Bovogen, Melbourne, Australia]; 80.9%

Dulbeccos’s Modified Eagle Medium (DMEM) [Sigma-Aldrich, St. Louis,

USA]; 0.1% 1000X antibiotic mix: penicillin+streptomycin+neomycin (PSN)

Vero E6 growth medium (RSV) DMEM [Sigma-Aldrich, St. Louis, USA] + 5% FBS [Bovogen, Melbourne, Australia]

Vero E6 growth medium (hMPV) OPTI-MEM® Reduced Serum Medium (1X) [GibcoTM, Invitrogen Corp.,

Carlsbad, USA] + 5% FBS [Bovogen, Melbourne, Australia]

A549 growth medium F12K Nutrient Mixture Kaighn’s Modification (1X) [Invitrogen, Carlsbad,

USA] + 10% FBS [Bovogen, Melbourne, Australia]

Cos-7 growth medium DMEM [Sigma-Aldrich, St. Louis, USA] + 5% FBS [Bovogen, Melbourne,

Australia] + 1X PSN

Bacterial culture Luria Bertani (LB) 10 g/l tryptone [Sigma-Aldrich, St. Louis, USA]; 5 g/l yeast extract [Oxoid,

Basingstoke, England]; 10 g/l NaCl [Univar APS, Seven Hills, Australia]

LB agar LB with 2% bacterial agar [Oxoid, Basingstoke, England]

SOB -Mg medium 20 g/l tryptone [Sigma-Aldrich, St. Louis, USA]; 5 g/l yeast extract [Oxoid,

Basingstoke, England]; 10 mM NaCl [Univar APS, Seven Hills, Australia];

2.5 mM KCl [Univar APS, Seven Hills, Australia]

LB + gentamycin LB + 7 μg/ml Gentamycin [Sigma-Aldrich, St. Louis, USA]

LB + kanamycin LB + 100 μg/ml Kanamycin [Sigma-Aldrich, St. Louis, USA]

LB agar + gentamycin LB agar + 7 μg/ml Gentamycin [Sigma-Aldrich, St. Louis, USA]

LB agar + kanamycin LB agar + 100 μg/ml Kanamycin [Sigma-Aldrich, St. Louis, USA]

Table 2-9 List of commercial chemicals used in this study

Buffers/solutions Manufacturer Foetal bovine serum (FBS) [Bovogen, Melbourne, Australia]

Hank’s Balanced Salt Solution (HBSS) [Sigma-Aldrich, St. Louis, USA]

Trizol® reagent [Invitrogen, Carlsbad, USA]

(5X) DNA loading dye, blue [Bioline, Alexandria, Australia]

Agarose [Amresco, Solon, USA]

GelRedTM fluorescent nucleic acid gel stain, 10,000X in H2O [Biotium, Hayward, USA]

LipofectamineTM 2000 Transfection Reagent [Invitrogen, Carlsbad, USA]

2, 2’-Azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)

(ABTS liquid substrate)

[Sigma-Aldrich, St. Louis, USA]

RNAse-free water [Qiagen, Hilden, Germany]

HyperLadderTM I DNA ladder [Bioline, Alexandria, USA]

HyperPage Protein ladder [Bioline, Alexandria, USA]

Gentamycin [Sigma-Aldrich, St. Louis, USA]

Kanamycin [Sigma-Aldrich, St. Louis, USA]

Bovine serum albumin (BSA) [Sigma-Aldrich, St. Louis, USA]

Dimethyl sulfoxide [Sigma-Aldrich, St. Louis, USA]

Glycerol [Unilab APS, Seven Hills, Australia]

Page 58: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

45

Table 2-10 List of in-house prepared buffers and solution

Buffers/solutions Composition 10X phosphate-buffered saline (PBS) 1.36 M NaCl [Univar APS, Seven Hills, Australia]; 101.44

mM Na2HPO4 [ChemSupply, Gilman, Australia]; 26.83 mM

KCl [Univar APS, Seven Hills, Australia]; 16.90 mM

KH2PO4; pH 7.5

Trypsin/EDTA 2.5% Trypsin [Sigma-Aldrich, St. Louis, USA]; 26.5 mM

EDTA [Unilab APS, Seven Hills, Australia]

Trypan blue 0.16% trypan blue [Sigma-Aldrich, St. Louis, USA]; 0.85%

NaCl [Univar APS, Seven Hills, Australia]; distilled H2O

1000X PSN 30.072 g/l Penicillin G [ICN-MP Biomedicals, Solon, USA];

50 g/l Streptomycin sulphate [Sigma-Aldrich, St. Louis, USA];

50 g/l Neomycin sulphate [Sigma-Aldrich, St. Louis, USA]

50X Tris-acetic acid ethylenediamine tetraacetic acid (TAE)

2 M Tris [Amresco, Solon, USA], 1 M acetic acid [Fronine, Taren Point, Australia], 0.05 M EDTA [Unilab APS, Seven

Hills, Australia]

Developing solution for Vector® Black

Alkaline Phosphatase Substrate Kit II

0.1 M Tris-HCl [Amresco, Solon, USA]; 0.1 M NaCl [Univar

APS, Seven Hills, Australia]; 0.05 M MgCl2 [Unilab APS,

Seven Hills, Australia]; pH 9.5

Block solution (immunostaining plaque assay) 5% skim milk powder [Diploma, Mt. Waverley, Australia];

PBS

Wash solution (immunostaining plaque assay) 0.5% Tween-20 [Amresco, Solon, USA]; PBS

1X TE buffer 10 mM Tris-HCl [Amresco, Solon, USA] pH 7.5; 1 mM

EDTA [Unilab APS, Seven Hills, Australia] pH 8.0

Block solution (ELISA) 1% BSA [Sigma-Aldrich, St. Louis, USA]; PBS

Wash solution (ELISA) 0.05% Tween-20 [Amresco, Solon, USA]; PBS

Diluent (ELISA) 0.05% Tween-20 [Amresco, Solon, USA]; 0.1% bovine serum

albumin (BSA) [Sigma-Aldrich, St. Louis, USA]; PBS

30% LB-Glycerol 30% glycerol [Unilab APS, Seven Hills, Australia]; LB

Page 59: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

46

2.2 Methods

2.2.1 Cell culture

2.2.1.1 Routine culturing

Vero E6 cells were grown in DMEM and OPTI-MEM® Reduced Serum Medium (1X), both

supplemented with 5% (v/v) heat-inactivated FBS. A549 cells were grown in F-12K Nutrient

Mixture, Kaighn’s Modification (1X), supplemented with 10% (v/v) heat-inactivated FBS.

Cos-7 cells were grown in DMEM supplemented with 10% (v/v) heat-inactivated FBS and

1X PSN. The cells were maintained in a humidified 37o C incubator supplied with 5% CO2

and subcultured 2 – 3 times per week. Trypsin/EDTA solution was used to detach cells from

the culture flask. All media, buffers, and solutions were pre-warmed to 37o C before use.

2.2.1.2 Freezing and thawing cells

5 X 106 – 1 X 10

7 cells/ml were resuspended in freezing medium (growth medium

supplemented with 10% (v/v) dimethyl sulfoxide) and 1 ml aliquots frozen slowly overnight

in a Cryo 1o C freezing container [Nalgene

TM, Rochester, USA]. Frozen cell stocks were

prepared for each cell line and stored at -80o C or in liquid nitrogen for later use.

When cells reached a high passage number (> 35), a vial of frozen cells was retrieved from

storage and thawed quickly in a 37o C waterbath. The cell suspension was added to 9 ml of

growth medium, resuspended, and cells pelleted by centrifugation at 400x g (Techcomp

CT15RT) [Techcomp Ltd., Kwai Chung, Hong Kong], at room temperature, for 5 minutes.

The pellet was then resuspended in 5 ml of growth medium and cells cultured in a 25 cm2

flask [Corning Inc., Lowell, USA].

2.2.1.3 Mycoplasma test

Cells were tested using polymerase chain reaction (PCR) for contamination by Mycoplasma

sp. Firstly, cells were cultured without antibiotics. For cells cultured in a 25 cm2 flask

[Corning Inc., Lowell, USA], the monolayer was washed once with PBS, detached with

trypsin/EDTA solution, and resuspended in 2 ml of growth medium. 250 µl of the cell

suspension was pelleted at 10,000x g and 4o C for 10 minutes. The cell pellet was washed

twice using 1 ml PBS followed by centrifugation at 10,000x g and 4o C for 10 minutes, and

resuspended in 240 µl of nuclease-free water. 20 µg/µl of Proteinase K was added to the cell

Page 60: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

47

suspension, followed by mixing and 1 hour incubation at 50o C. Cells were then lysed by

heating at 100o C for 10 minutes, and then pelleted by centrifugation at 13,000x g for 5

minutes at room temperature. Supernatant was collected and used directly as the PCR

template or stored at -20o C until further use. The PCR mixture, detailed in table 2-11, was

prepared on ice.

Table 2-11 Components of PCR for Mycoplasma sp. detection

Component Final concentration (5X) Mango Taq Colored Reaction Buffer 1 X

50 mM MgCl2 1.5 mM

2 mM dNTPs 0.2 mM

5 pmoles/μl MycoF1 primer 0.25 pmoles

5 pmoles/μl MycoR1 primer 0.25 pmoles

5 U/μl MangoTaq DNA polymerase 0.075 U

DNA template 1 µl

Nuclease-free water to 20 µl

PCR cycling conditions were 94o C for 3 minutes, followed by 30 cycles of 94

o C for 30

seconds, 54o C for 2 minutes, and 72

o C for 1 minute, and terminated by a final extension step

of 72o C for 5 minutes in a Mastercycler

® gradient thermal cycler model 5533 [Eppendorf,

Hamburg, Germany].

The positive control was Mycoplasma sp. contaminated cell culture and the negative control

was nuclease-free water. PCR results were viewed using agarose gel electrophoresis, as

described in section 2.2.8.2, by running 15 µl of PCR product mixed with 3 µl of (5X) DNA

loading dye. If the PCR results were negative, 1 µl of the PCR product was used as the

template for a nested PCR using the pair of MycoF2 and MycoR2 primers (Table 2-4) and

similar mixture and cycling conditions as described in this section.

2.2.1.4 Trypan blue exclusion assay

The test is based on the principle that live cells possess intact cell membranes that exclude the

trypan blue dye, while dead cells do not. A viable cell should present a clear cytoplasm while

a nonviable cell should appear completely blue. Trypan blue was prepared by mixing 200 µl

of 0.2% (w/v) trypan blue in distilled water with 50 µl of 4.25% (w/v) NaCl in distilled

water. Diluted trypan blue and cell culture in growth medium were mixed in a 1:1 ratio and

applied to a haemocytometer. Cells were observed using an inverted light microscope

Page 61: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

48

[Olympus, Mt. Waverley, Australia], and viable cell number determined using the following

equation:

Viable cell count (cells/ml) =

2.2.1.5 Heat inactivation of FBS

500 ml of frozen FBS was slowly thawed overnight at 4o C. The FBS and a temperature

control consisting of an FBS bottle filled with 500 ml of water and inserted with a

thermometer were slowly heated to 37o C in a waterbath, and then transferred to a 56

o C

waterbath. The heat inactivation was carried out for 30 minutes at 56o C, starting from once

the thermometer inserted into the control reached 56o C.

2.2.1.6 Preparation of 2% methyl cellulose

Methyl cellulose powder (10 g) was slowly added into 75 ml of boiled HBSS, with the

solution mixed through stirring. The solution was autoclaved at 121o C for 37 minutes, and

then cooled to 37o C on a stirrer. 40 ml of HBSS, 10 ml of FBS, 0.5 ml of 1,000X PSN, and

409.5 ml of serum-free DMEM was added to the methylcellulose-HBSS mix. The solution

was then shaken vigorously, stirred again for 30 minutes at room temperature, transferred to

4o C for 1 hour, while being continuously stirred, until the solution reached a consistent

thickness. The 2% methyl cellulose was stored at 4o C and pre-heated to 37

o C before each

use.

2.2.2 Virus propagation

2.2.2.1 Cell preparation

Vero E6 cells were prepared as 80%-confluent monolayer in a 150 cm2 flask [Corning Inc.,

Lowell, USA], which was approximately 8 X 106 cells. For RSV propagation, cells were

grown in DMEM + 5% FBS; while for hMPV propagation, cells were grown in OPTI-MEM®

+ 5% FBS.

Page 62: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

49

2.2.2.2 Virus inoculation

2.2.2.2.1 RSV

The culture medium was removed and Vero E6 monolayer washed with HBSS. Cells were

infected with recombinant RSV in 8 ml DMEM, at a multiplicity of infection (MOI) of 0.1 –

0.5, depending on the titre of available virus stock. Virus was adsorbed for 1 hour in a

humidified 37o C incubator supplied with 5% CO2; following which 6 ml of DMEM + 5%

FBS was added. A flask that was mock infected served as the control for the infection.

Infected cells were incubated at 37o C, 5% CO2 until approximately 50% syncytia had

developed evenly across the flask, accompanied by minimal cell death.

2.2.2.2.2 hMPV

Vero E6 monolayer was first rinsed with HBSS, followed by a second rinse with

OPTI-MEM® + 5 μg/ml trypsin. Cells were infected with hMPV in MOI of 0.5 in 6 ml of

OPTI-MEM® + 5 μg/ml trypsin and adsorbed for 2 hours in a 37

o C incubator supplied with

5% CO2. After adsorbtion, 6 ml of OPTI-MEM® + 5% FBS was added and infected cells

were incubated at 37o C, 5% CO2. hMPV was harvested when 25 – 50% CPE was observed

evenly across the flask and cell death was minimal.

2.2.2.3 Harvest

RSV and hMPV were harvested using a similar technique. Firstly, infection medium was

removed while leaving 2.5 – 3 ml in the flask and infected cells detached using a scraper

[Corning Inc., Lowell, USA]. Infectious particles contained within cells were released using

five times 75% amplitude sonication [Sonics®, Newton, USA]. Each sonication lasted five

seconds, with a two-minute incubation on ice between each sonication. The virus was

separated from cell debris by centrifugation at 1200x g and 40 C for 7 minutes. The virus-

containing supernatant was collected in cryovials [GREINER Cryo.sTM

, Kaysville, USA] and

stored at -800 C.

2.2.2.4 Virus titration

RSV and hMPV was titrated to determine viral infectivity as previously described (Tripp,

2006, Tripp, 2010). Slight modifications were made to these procedures and these are

detailed below.

Page 63: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

50

Briefly, 1 X 105 Vero E6 cells were seeded in each well of a 24-well plate in 0.5 ml of growth

medium, which was DMEM + 5% FBS for RSV and OPTI-MEM® + 5% FBS for hMPV, and

grown overnight (less than 24 hours) to obtain 80% confluent monolayer on the day of virus

inoculation.

10-fold serial dilutions of virus in serum-free DMEM (RSV) or OPTI-MEM® + 5 μg/ml

trypsin (hMPV) were used for quadruplicate inoculation. The growth medium was removed

from the well, cells briefly washed with HBSS, and 200 μl of diluted virus was added into

each well. Serum-free DMEM or OPTI-MEM® + 5 μg/ml trypsin alone was used as a control

(mock) infection for RSV and hMPV, respectively. Virus was adsorbed to the cells for 1 hour

at 37o C, 5% CO2, following which the infection was overlaid with 1 ml of 2% methyl

cellulose (RSV) or 200 μl of OPTI-MEM® + 5% FBS (hMPV) and the plate incubated at 37

o

C, 5% CO2 until day 6 post infection.

The culture medium was removed using suction and the cells washed with PBS and fixed

using ice-cold acetone-methanol (6:4) for 10 minutes. Following fixation, monolayers were

washed with PBS and air-dried. Fixed cells were blocked with PBS + 5% skim milk powder

for 30 minutes at room temperature for RSV titration, and for 1 hour at 37o C for hMPV.

Block solution was then replaced by 200 µl of anti-RSV F protein monoclonal antibody clone

131-2A (1:1000 dilution in block solution) for RSV titration or Ms X Hu Metapneumovirus

(1:1000 dilution in block solution) for hMPV titration, followed by 2 hours incubation at 37o

C. Cells were rinsed and washed twice with PBS + 0.5% Tween-20, 5 minutes each, on a

rocker [Labnet, Woodbridge, USA]. 200 µl of the secondary antibody, goat anti-mouse IgG

conjugated to alkaline phosphatase (diluted 1:1000 in block solution), was added and cells

incubated for 1 hour at 37o C, then washed as described previously. Vector

® Black Alkaline

Phosphatase Substrate Kit II was prepared according to the manufacturer’s protocol shortly

before use and 200 µl added to each well. Staining was developed for 20 minutes and stained

cells washed with water to stop the reaction. The plate was then air-dried and stained

immunoplaques counted using an inverted light microscope [Olympus, Mt. Waverley,

Australia]. Virus titre (plaques/ml) was determined using the formula

dilution factor 5,

where N is the number of immunoplaques in each well.

Page 64: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

51

2.2.3 Replication kinetics assay

The replication kinetics assay was done in two cell lines, Vero E6 and A549. The cells were

prepared as 80%-confluent monolayer in 500 µl of growth medium, which was

approximately 1 X 105 cells per well in a 24-well plate. A control (mock infection) consisting

of serum-free media alone was included. RSV was diluted to an MOI of 0.03 and 3.0

PFU/cell in 200 µl of serum-free DMEM for Vero E6 cells and F12K Nutrient Mixture

Kaighn’s Modification for A549 cells. The growth medium was removed from the well, cell

monolayers were briefly washed with HBSS, and the diluted RSV added in quadruplicate.

Virus was adsorbed to cells for 1 hour in a 37o C incubator supplemented with 5% CO2. The

infecting medium was replaced with 600 µl of 1:1 mix of serum-free medium and cell growth

medium, and then plates were incubated at 37o C, 5% CO2 until the specified endpoints. The

endpoints of single cycle growth infection (MOI 3.0) were 0 hour (H), 18 H, 24 H, 30 H, 36

H, and 48 H; while the end points of multi-cycle infection (MOI 0.03) were day (D) 0, D1,

D2, D3, D4, and D5. At the specified end point, 450 μl of the 600 μl infection medium was

collected as the supernatant of the samples and the remaining 150 μl was used to generate cell

suspension by scraping off monolayer. Both supernatant and cell suspension were stored at -

80o C until further use. The supernatant was used to measure the secretion level of cellular

IL-8 and RANTES via ELISA, as described in section 2.2.4. The cell suspension was used

for measuring viral titre at each end point of infection via immunostaining plaque assay

described in section 2.2.2.4.

Viral titres were calculated in pfu/ml. A linear graph was built based on the time point of

termination of infection (x-axis) and log10 of viral titres (y-axis), virus titres were represented

by the means of quadruplicate titres from independent assays with a standard error. The

nonparametric Mann-Whitney U test (2-tailed) was used to compare the mean titres between

viruses. All calculation and graph building were done using GraphPad (Prism) v.5 software

(GraphPad, La Jolla, USA).

Page 65: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

52

2.2.4 Enzyme-linked immunofluorescence assay (ELISA)

2.2.4.1 Plate-based optical density measurement

The concentrations of IL-8 and RANTES in RSV-infected cells were measured using the

ELISA development kit according to the manufacturer’s protocol. The ELISA plate [Thermo

Scientific, Roskilde, Denmark] was first prepared by overnight coating with 100 µl of capture

antibody (0.5 µg/ml) in PBS at room temperature. Thereafter, the liquid was aspirated and

plate washed 4 times, each with 300 µl of wash buffer (PBS + 0.05% Tween-20). 300 µl of

block buffer (PBS + 1% BSA) was applied to the wells and incubated for 1 hour at room

temperature, following which the plate was washed as described.

The kit-provided standard was two-fold serially diluted in diluent (0.1% BSA, 0.05% Tween-

20, PBS). The RANTES standard curve ranged from 2 ng/ml to 0 ng/ml and IL-8 from 1

ng/ml to 0 ng/ml. Samples (see section 2.2.3) were diluted 1:2 in diluent. 100 µl of diluted

samples and standards were added to the plate in triplicate. The reaction was incubated for 2

hours at room temperature and then washed. Detection antibody was diluted 0.25 µg/ml in

diluent, 100 µl added per well, incubated for another 2 hours at room temperature, and then

the plate was washed. 100 µl of avidin-HRP conjugate in diluent (1:2000) was applied into

wells, the plate incubated for 30 minutes at room temperature, and washed. As the final step,

100 µl of ABTS liquid substrate solution was added and colour development monitored with

a Benchmark-Plus ELISA plate reader [Bio-Rad Laboratories Inc., Hercules, USA] at 405

nm, with wavelength correction set at 650 nm.

2.2.4.2 Enumeration of IL-8 and RANTES concentration (pg/ml)

The standard curve was built based on the optical density values of the standard dilutions,

which were plotted on an xy graph, with the optical density values as the dependent variable

(y) and the concentration of the standard dilutions as the independent variable (x). A

regression analysis of scatterplots in the standard curve was done using Microsoft Excel

software [Microsoft Corporation]. The scatterplot exhibited a quadratic regression and a

polynomial regression curve and its function in was determined. The

IL-8 and RANTES concentrations in supernatant of infected cells were calculated using the

equation, based on the optical density values of the diluted samples. The kinetics of

chemokine expression was then evaluated based on the mean values of the concentration of

Page 66: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

53

the chemokine in the samples, with variability of the values expressed as standard error, and a

graph was built based on the means value of the titres with the standard error of the means.

The nonparametric Mann-Whitney U test (2-tailed) was used to compare the mean values of

chemokine expression induced by the two viruses. All calculation and graph building were

done using GraphPad (Prism) v.5 software (GraphPad, La Jolla, USA).

2.2.5 hMPV N constructs

2.2.5.1 RNA extraction

hMPV was first propagated in Vero E6 cells that were cultured in a 25 cm2 flask [Corning

Inc., Lowell, USA] and the N gene isolated from RNA, which was extracted using TRIzol®

reagent as per manufacturer’s instruction. The growth medium was replaced with 2.5 ml of

TRIzol® reagent, and flask incubated for 5 minutes at room temperature. The suspension was

transferred into a 10 ml conical tube [Cellstar®, Greiner Bio One, Frickenhausen, Germany],

mixed vigorously with 0.5 ml chloroform, and incubated for 3 minutes at room temperature.

Suspension was centrifuged at 12,000x g for 15 minutes at 4o C. The aqueous phase was

transferred into a new 10 ml conical tube, and RNA precipitated with 1.25 ml of isopropyl

alcohol for 10 minutes at room temperature, followed by centrifugation at 12,000x g and 4o C

for 10 minutes. The RNA pellet was then washed by vigorous mixing with 2.5 ml of 75%

ethanol, followed by centrifugation at 7,500x g and 4o C for 5 minutes. As the final step, the

RNA was air dried, resuspended in nuclease-free water, and stored at -80o C until further use.

2.2.5.2 Reverse transcription and polymerase chain reaction (RT-PCR)

The cDNA of hMPV RNA was generated using the cDNA synthesis kit. RNA concentration

was first measured using NanoDropTM

1000 UV/Vis spectrophotometer [NanodropTM

,

Wilmington, USA]. Firstly, reaction mix I (table 2-12) was first prepared on ice.

Table 2-12 The reaction mix I to generate cDNA from hMPV RNA

Component Final concentration RNA 1 µg

270 ng/μl Oligo(dT)10 270 ng

10 mM dNTP Mix 2 mM

DEPC-treated water To a final volume of 10 µl

The reaction was incubated for 10 minutes at 65o C, immediately chilled on ice for 2 minutes,

and added to reaction mix II (table 2-13).

Page 67: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

54

Table 2-13 The reaction mix II to generate cDNA from hMPV RNA

Component Final concentration (5X) RT Buffer 2X

10 U/μl RNase Inhibitor 10 U

200 U/µl Reverse Transcriptase 5 U

DEPC-treated water To a final volume of 10 µl

The final reaction was incubated for 45 minutes at 45o C, and reverse transcription terminated

by 15 minutes incubation at 70o C. cDNA was stored at -80

o C until further use.

The hMPV N gene was amplified via PCR using the cDNA of hMPV genome as template. In

addition to amplification, the PCR also equipped the N gene with attB sequences that

function as site-specific recombination required for generating the entry clone that is

described in section 2.2.6.1. The PCR is also the means to generate the truncated ORF

constructs of hMPV N gene, which was done by adjusting the specific primer pair to the

desired ORF. The PCR components are detailed in table 2-14 and the primer pairs that were

used to generate the hMPV N constructs are listed in table 2-15.

Table 2-14 The PCR components to generate hMPV N constructs

Component Final concentration (5X) Mango Taq Colored Reaction Buffer 1X

50 mM MgCl2 1.5 mM

2 mM dNTPs 0.2 mM

5 pmoles/μl forward primer 0.25 pmoles

5 pmoles/μl reverse primer 0.25 pmoles

5 U/μl MangoTaq DNA polymerase 0.025 U

cDNA 5 μl

Nuclease-free water To a final volume of 40 µl

Table 2-15 List of primer pairs used to generate hMPV N construct

ORF Primer pair hMPV N HMPV83NattB1 + HMPV83NattB2

hMPV N – fragment A HMPV83NattB1+hMPVN_r_attB2

hMPV N – fragment B HMPV83NattB1+hMPVN_f-rev_attB2

hMPV N – fragment C hMPVN_f_attB1+HMPV83NattB2

PCR cycling conditions were 95o C for 7 minutes, followed by 35 cycles of 96

o C for 30

seconds, 58o C for 45 seconds, and 72

o C for 1 minute. PCR results were viewed via agarose

gel electrophoresis as described in section 2.2.8.2, from 5 µl of PCR product that was mixed

with 1 µl of (5X) DNA loading dye.

2.2.5.3 DNA clean up

PCR product was purified from agarose after separation via electrophoresis. DNA

purification was carried out to eliminate the excess salts and primers that might interfere with

Page 68: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

55

the recombination reaction. Briefly the total volume of PCR product was mixed with DNA

loading dye in 5:1 ratio and electrophoresed in 1% agarose gel, as described in section 2.2.9.

The desired DNA fragment was visualised using UV light from a transilluminator and cut

from the gel using a clean scalpel. The DNA was then purified using Wizard® SV Gel and

PCR Clean-Up System [Promega, Madison, USA]. Excised gel was first dissolved with

Membrane Binding Solution in a ratio of 1 µl solution for 1 mg gel via incubation at 65o C

for 10 minutes. The solution was then transferred to an SV Minicolumn and incubated for 1

minute at room temperature. The column was centrifuged at 19,000x g for 1 minute and the

membrane washed twice with Membrane Wash Solution. The first wash used 700 µl of

solution and the second wash used 500 µl of solution. The column was centrifuged at 19,000x

g for 1 minute in each wash. An additional centrifugation was carried out to remove the

residual wash solution, then the column transferred to a 1.5 ml microfuge tube and the DNA

eluted with 30 µl of nuclease-free water. The purified DNA was stored at -80o C until further

use.

2.2.6 Cloning of hMPV N gene using GatewayTM

Technology

The hMPV N protein was expressed in mammalian cells as an expression clone, by being

inserted into an expression vector. The expression clone was prepared using the Gateway®

recombination cloning. This technology makes use of the specific att recombination sites

present in E. coli bacterial chromosome (attB) and bacteriophage lambda DNA (attP) that

ensures the correct directionality of the recombinant. The Gateway® technology consists of

two steps: the construction of an entry clone and the construction of an expression clone. The

entry clone was generated by inserting the PCR product of hMPV N gene into the entry

vector pDONR®207 via BP recombination. The expression clone was generated by transport

the entry clone into the destination vector, pEPI-DESTC, via LR recombination.

2.2.6.1 Entry clone construction

Table 2-16 The components of BP recombination reaction

Component Final concentration attB-tagged ORF (section 2.2.5.3) 16 – 40 ng

attP-containing pDONR®207 150 ng

Gateway ® BP Clonase® Enzyme Mix 2 µl

TE buffer pH 8.0 To 10 µl

The reaction was incubated at 25o C for 18 hours and terminated with addition of 2 µl of

Proteinase K in 10-minutes incubation at 37o C. 1 – 2 µl of the reaction was transformed into

Page 69: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

56

ElectroMAXTM

DH10BTM

Cells (as described in section 2.2.10.2). 100 µl of the transformed

E. coli suspension was plated on LB agar supplemented with 7 µg/ml gentamycin and

incubated overnight at 37o C. Positive colonies were picked up and cultured in 5 ml LB

supplemented with 7 µg/ml of gentamycin, at 37o C, 220 rpm, for plasmid isolation (section

2.2.6.3).

2.2.6.2 Expression clone construction

Table 2-17 The components of LR recombination reaction

Component Final concentration attP containing pDONR®207.hMPV N construct (section 2.2.6.1) 50 – 150 ng

attB containing pEPI-DESTC 150 ng

LR Clonase® 2 µl

TE buffer pH 8.0 To 10 µl

The reaction was incubated 25o C for 18 hours and terminated with addition of 2 µl of

Proteinase K in 10 minutes incubation at 37o C. 1 – 2 µl of the reaction was transformed into

ElectroMAXTM

DH10BTM

Cells (as described in section 2.2.10.2). 100 µl of the transformed

E. coli suspension was plated on LB agar supplemented with 100 µg/ml kanamycin and

incubated overnight at 37o C. Positive colonies were picked up and cultured in 5 ml LB

supplemented with 100 µg/ml of kanamycin at 37o C, 220 rpm, for plasmid isolation (section

2.2.6.3).

2.2.6.3 Plasmid isolation

The desired plasmid, obtained from BP or LR recombination reaction, was isolated using the

the QIAprep® Spin Miniprep or Midiprep Kits [Qiagen].

2.2.6.3.1 Plasmid isolation using Miniprep Kit (Qiagen)

Bacterial culture grown overnight in 5 ml of LB supplemented with appropriate antibiotics

(section 2.2.6.1 and 2.2.6.2) was first pelleted through centrifugation at 4,100x g and 4o C for

20 minutes, and resuspended in 250 µl of buffer PI. 250 µl of buffer P2 was added and mixed

thoroughly by 4 – 6 times inversion. 350 µl of buffer N3 was added and mixed in the same

way. The solution was then centrifuged at 16,500x g for 10 minutes and supernatant loaded

on to a QIAprep® spin column. The column was briefly centrifuged at 16,500x g, washed

with 0.5 ml of buffer PB in a brief centrifugation at 16,500x g, followed by a secondary wash

with 0.75 ml of buffer PE. An additional centrifugation was carried out to remove the

residual wash buffers. Finally, the column was transferred to a 1.5 ml microfuge tube and the

Page 70: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

57

plasmid eluted with 30 µl of RNase-free water. The plasmid was stored at -20o C until further

use.

2.2.6.3.2 Plasmid isolation using Midiprep Kit (Qiagen)

A starter culture was first prepared by growing the desired bacterial colonies (section 2.2.6.1

and 2.2.6.2) in 5 ml of LB supplemented with appropriate antibiotics at 37o C, 300 rpm, for 8

hours. A bacterial culture was made by re-culturing 50 μl of the starter culture in 25 ml of LB

supplemented with appropriate antibiotic solution at 37o C, 300 rpm, overnight. The bacteria

were harvested by centrifugation at 6000x g and 4o C for 15 minutes, then resuspended in 4

ml of buffer P1. 4 ml of buffer P2 was added and mixed by 4 – 6 times tube inversion, and

incubated at room temperature for 5 minutes. 4 ml of buffer P3 was then added and mixed by

4 – 6 times tube inversion, followed by 15 minutes incubation on ice. The tube was

centrifuged at 21,800x g and 4o C for 30 minutes; then supernatant transferred into a new

15-ml conical tube and re-centrifuged at 21,800x g and 4o C for 15 minutes. Subsequently,

the supernatant was applied to a QIAGEN-tip 100. The tip was washed twice with 10 ml of

buffer QC and plasmid eluted in 5 ml of buffer QF. The plasmid was precipitated using 3.5

ml isopropanol accompanied by centrifugation at 18,000x g and 4o C for 18 minutes. The

DNA pellet was washed with 2 ml of 70% room-temperature ethanol, followed by

centrifugation at 18,000x g for 10 minutes. As the final step, the pellet was air dried for 10

minutes and plasmid redissolved in 500 μl of RNase-free water. The plasmid was stored at -

80o C until further use.

2.2.7 Confirmation of the hMPV N gene inserted into vectors using PCR and

DNA sequencing method

2.2.7.1 PCR

The second method to verify entry and expression clones was via PCR using the same primer

pair used to generate the ORF of interest. The components of the PCR are listed in table 2-18.

Page 71: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

58

Table 2-18 PCR components to confirm the presence of hMPV N insert in cloning vectors.

PCR cycling conditions were 95o C for 7 minutes, followed by 35 cycles of 96

o C for 30

seconds, 58o C for 45 seconds, and 72

o C for 1 minute. PCR results were viewed via agarose

gel electrophoresis as described in section 2.2.7.6, from 5 µl of PCR product that was mixed

with 1 µl of (5X) DNA loading dye.

2.2.7.2 DNA sequencing

The sequence of ORF generated via PCR was confirmed via DNA sequencing. The

sequencing reaction is described table 2-19.

Table 2-19 Components of DNA sequencing reaction, with PCR product as template

Component Final concentration PCR product 30 – 75 ng

5 pmoles/μl primer* 0.5 pmoles

Nuclease-free water To a final volume of 12 μl

*the primers used were listed in table 2-15.

The results of each Gateway® recombination reaction were also verified via DNA

sequencing, using sequencing primers listed in table 2-4. The sequencing reaction is

described in table 2-20.

Table 2-20 Components of DNA sequencing reaction, with ds plasmid as template

Component Final concentration ds plasmid 600 ng

Primer 0.5 pmoles

Nuclease-free water To a final volume of 12 μl

DNA sequencing was outsourced to the Australian Genome Research Facility

(www.agrf.org.au), and the reaction was prepared according to the company’s requirement.

The chromatograms were analysed using BioEdit 7.0.4 (Hall, 2005, Ibis Biosciences)

software.

Component Final concentration (5X) Mango Taq Colored Reaction Buffer 1X

50 mM MgCl2 1.5 mM

2 mM dNTPs 0.2 mM

5 pmoles/µl forward primer 0.25 pmoles

5 pmoles/µl reverse primer 0.25 pmoles

5 U/μl MangoTaq DNA polymerase 0.025 U

DNA template (entry or expression clones) 5 μl

Nuclease-free water To a final volume of 40 µl

Page 72: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

59

2.2.8 Measurement of nucleic acid concentration

DNA/RNA quantitation was carried out using the NanoDropTM

1000 UV/Vis

spectrophotometer [NanodropTM

, Wilmington, USA]. Briefly, 2 μl of DNA/RNA sample was

loaded on the pedestal and DNA/RNA concentration was measured by absorbance at 260 nm

(A260), A280, and A230. The ratios of A260/A280 and A260/A230 were analysed by the

NanoDropTM

v3.7.1 software. The spectrophotometer was blanked with the solution in which

the nucleic acid was resuspended.

2.2.9 Agarose gel electrophoresis

Agarose gel (1%) was prepared from 0.5 g agarose powder in 50 ml 1X TAE, which was

dissolved by heating the mix in a microwave for 1 minute, and adding 50 µl GelRedTM

nucleic acid gel stain (1:10) into the mix. The DNA was mixed with (5X) DNA loading dye

(1 part DNA loading dye + 5 parts DNA) and the mix loaded into gel wells. The

electrophoresis was carried out at 80 volts for 80 minutes and the result viewed using GelDoc

[Biorad] UV transilluminator and analysed with Quantity One® v4.6.5 software [Bio-Rad

Laboratories Inc., Hercules, USA].

For confirmation of hMPV N constructs (Figures 5-3 and 5-4), agarose gel images were

digitally modified using Adobe Photoshop CS5 software [Adobe Systems Incorporated] by

rearranging the lanes containing confirmation using PCR and uncut plasmid, for the purpose

of presentation clarity.

2.2.10 Plasmid storage in glycerol solution

Plasmids (entry and expression clones) were stored in transformed ElectroMAXTM

DH10BTM

competent E. coli. The bacteria were grown overnight on LB agar plates supplemented with

appropriate antibiotics at 37o C. The colonies that grew all over the agar surface were made

into a bacterial suspension by adding 1 ml of LB + 30%-glycerol and scraping off the

colonies using a glass spreader. The suspension was pipetted into cryovials [GREINER

Cryo.sTM

, Kaysville, USA] and stored at -80o C.

Page 73: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

60

2.2.11 hMPV N protein expression

2.2.11.1 Cell transfection

Cos-7 cells (3 X 105 cells in 2 ml growth medium) were seeded overnight in advance, on

glass coverslips (diameter = 24 mm) [ProSciTech, Thuringuwa, Australia] that were placed in

a 6-well plate to obtain 80%-confluent monolayer at the time of transfection.

LipofectamineTM

2000 Transfection Reagent was diluted 1:25 in serum-free DMEM. Green

fluorescent protein (GFP) – tagged ORFs in pEPI-DESTC was diluted in serum-free DMEM

to a concentration of 2 µg per well. Diluted LipofectamineTM

2000 Transfection Reagent and

diluted plasmid was mixed in 1:1 (v:v) ratio and incubated 20 minutes at room temperature.

The monolayer was washed with PBS, then 200 µl of LipofectamineTM

2000 Transfection

Reagent/plasmid solution added in a dropwise fashion, and incubated at 37o C, 5% CO2 for

20 minutes. 2 ml growth medium was added and transfected cells were maintained at 37o C,

5% CO2, until a specified time point, when the expression of green fluorescent protein (GFP)-

tagged protein was viewed and recorded using confocal laser scanning microscopy (CLSM)

as described in section 2.2.11.2.

2.2.11.2 Confocal laser scanning microscopy (CLSM)

For live cells imaging, 24 or 48 hours after transfection the coverslip was placed on a ring-

shaped coverslip holder and cells covered with 37o C PBS. A drop of immersion oil was

placed on top of the objective of Nikon Eclipse T [Nikon Instruments Inc., Melville, USA]

to layer between the lens and coverslip.

The NIS-Elements 64 bit 3.21.00 software [Nikon Instruments Inc., Melville, USA] captured

and recorded the cell images. First, the wavelength of imaging light was adjusted to the

fluorescent dye used to label cells in the ‘channel’ menu of the scan setting window; the most

generally used was 488 nm, which recognises the light emitted by the GFP. To select the cells

whose images were to be recorded, the menu from NIS-Elements 64 bit 3.21.00 software was

shifted to ‘widefield’, and the sight field scrutinised while the lens focus was being adjusted.

To capture and record the images, the menu was shifted to ‘confocal’, the desired cells

localised in the ‘scan area’ function of the navigation window, and lens focus manually

adjusted during ‘live’ viewing. The frozen images were captured using 4X averaging of the

‘XY’ navigation and saved in JPEG 2000 Image format (.jp2). The pixel density of

Page 74: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

61

fluorescence in nucleus (Fn) and in cytoplasm (Fc) were obtained using the ImageJ 1.45M

software (Ferreira & Rasband, 2011) and the fluorescence ratio determined using the formula

Fn/c =

, where Fn is the pixel density fluorescence in nucleus, Fc is the pixel density of

fluorescence in the cytoplasm, and Fb is the pixel density of background fluorescence.

The values of nuclear and cytoplasmic fluorescence ratio (Fn/Fc) of subcellular localisation

of hMPV N constructs were determined according to section 2.2.11.3. The mean value of

Fn/Fc ratio for each construct was determined from total cells observed (generally > 10), and

presented as a bar, with variability expressed as the standard error of the mean (SEM). The

statistical difference of Fn/Fc ratio between each of hMPV N construct was assessed using

the nonparametric Mann-Whitney U test (2-tailed). All calculation and graph building were

done using GraphPad (Prism) v.5 software (GraphPad, La Jolla, USA).

2.2.12 Bacterial culture

2.2.12.1 Preparation of competent E. coli

The ElectroMAXTM

DH10BTM

Cells [Invitrogen] are specifically designed to facilitate

recombinational cloning using GatewayTM

technology. From the initial frozen culture, a loop

of ElectroMAXTM

DH10BTM

Cells [Invitrogen] was streaked on to an antibiotic-free LB agar

plate at 37o C. A single colony was selected and grown overnight in 5 ml of SOB –Mg

medium at 37o C in a shaker incubator [Bioline]. Four ml of the culture was then re-cultured

in 200 ml of SOB –Mg medium for 2 – 3 hours at 37o C until OD600 ranged between 0.8 and

1.0. The suspension was pelleted at 2500x g centrifugation at 4o C for 10 minutes, the pellet

resuspended in 100 ml of 10% pre-chilled glycerol, and re-pelleted by centrifugation for 10

minutes at 2500x g and 4o C. The supernatant was carefully removed and the pellet

resuspended in 200 ml of 10% pre-chilled glycerol and centrifuged 2500x g at 4o C for 10

minutes. As the final step, bacterial pellet was resuspended in approximately 2 ml of the

remaining liquid, aliquoted in 50 µl volume, and stored at -80o C until further use.

2.2.12.2 Cell transformation

On ice, 1 – 10 µl plasmid that was generated from BP or LR reaction was mixed with 50 µl

of ElectroMAXTM

DH10BTM

competent E. coli cell suspension and inserted into a pre-chilled

1 ml cuvette [Bio-Rad Laboratories Inc., Hercules, USA]. The plasmid was transformed into

bacterial cells via electroporation using GenePulser® II electroporator [Bio-Rad Laboratories

Page 75: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

62

Inc., Hercules, USA] with the following conditions: 25 µF capacitance, 200 Ω resistance, 2.0

kV voltage, then 450 μl of LB medium was added. Transformed bacteria were incubated at

37o C and 220 rpm in a shaker incubator [Bioline, Alexandria, Australia] for an hour. 100 –

150 µl of cell suspension was cultured overnight on an LB agar plate supplemented with

appropriate antibiotic at 37o C. Surviving colonies were either inoculated in 5 ml LB for

further assay or re-cultured on LB agar plate for storage.

2.2.12.3 Long term storage of E. coli

Transformed or non-transformed E. coli were preserved in 30% glycerol solution as

described in section 2.2.10.

Page 76: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

63

CHAPTER 3

IN VITRO REPLICATION EFFICIENCY

OF rRSV A2 M(T205A)

3 Replication Efficiency of RSV in Cell Culture

Page 77: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

64

Page 78: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

65

3.1 Introduction

The replication efficiency, or fitness, measured in cell culture can correlate with the clinical

outcome of infections and host-to-host transmission in whole organism (Dietzschold et al.,

1985, Dietzel et al., 2011). Replication efficiency includes two components: the capabilities

of the virus to, firstly, establish infection and, secondly, to transmit the infection. Viral

infectivity can be studied through a replication kinetics assay wherein a high ratio of virus

particles to cells (multiplicity of infection (MOI)) is used and observing infectious particle

production in a short period following infection (typically hours), which is essentially a single

cycle growth curve. Viral transmissibility can be studied by conducting replication kinetics

assay using a low MOI and observing viral growth over a longer period following infection

(typically days); therefore studying viral growth over multiple replication cycles.

Studying viral growth in cell cultures (in vitro) allows determination of viral fitness over a

period of time in a system where host antiviral mechanisms are well characterised, compared

to the more complex antiviral network in a whole organism (in vivo). The replication

efficiency of RSV was studied in two cell lines: Vero E6 and A549 cells. Vero E6 is a cell

line that originates from the kidney epithelium of the African green monkey (Cercopithecus

aethiops) and lacks the structural genes of the antiviral IFN-α/β (Emeny and Morgan, 1979),

therefore is highly susceptible to infection and in this study acted as the control system for the

IFN-α/β antiviral response. A549 is a cell line that originates from the human respiratory

epithelial cells and possesses the functional structural genes of IFN-α/β; and therefore are a

relevant model for studying RSV infection, in a system that resembles the natural site of

infection (Stoltz and Klingström, 2010). IFN-α/β is a strong innate immune response against

viral infection and in RSV infection contributes to virus clearance as well as directs the

biology of the subsequent pro-inflammatory responses, which often determine the disease

outcome (Noah et al., 2002, Smyth et al., 2002).

The wild type and mutant viruses are both well-characterised recombinants and have identical

genomes, except for threonine into alanine substitution (TA) at RSV M205. The wild type

virus was generated using reverse genetics technology based on the genome of RSV strain A2

and confers the CK2 phosphorylatable phenotype of RSV M 205 (T). The mutation of M205

(TA) in the mutant virus was designed on the basis of previous work that showed that CK2

phosphorylation regulates nuclear export of RSV M (Ghildyal et al., 2006). The recombinant

Page 79: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

66

RSV were produced by our collaborators in University of South Florida (Dr Michael Teng

and Ms Kim Tran) who performed full genome sequencing for rRSV A2 and rRSV A2

M(T205A) prior to us importing the virus into Australia. It was expected that the difference

in viral titres would represent the impact of this single amino acid substitution on the

replication efficiency. Previously, the impact of M(T205) substitution has been studied using

expression in transfected cells and in viral infection, but the results have not yet been

published (R. Ghildyal, personal communication).

In this study, both rRSV A2 and rRSV A2 M(T205A) were passaged multiple times in Vero

E6 cells prior to replication kinetics assay. Multiple passaging of a recombinant mutant virus

confers a possibility that selection, which favours the wild type genome, may cause the

mutant phenotype to revert into the wild type. In this study, the multiple passaging of rRSV

A2 M(T205A) could by chance mutate the M(A205) into the wild type M(T205), and in

doing so, the result obtained for the mutant may not be the true phenotype. Therefore, the

genotypes of the rRSV M constructs were confirmed using DNA sequencing after the

multiple passaging.

This study aimed to investigate the impact of mutation of a putative CK2 kinase

phosphorylation site within the M protein on the replication efficiency of RSV in an

environment that exerts the antiviral activity of IFN-α/β, as is found in the human respiratory

tract. The investigation was done by comparing the replication fitness of a wild type

(rRSV A2) and a mutant (rRSV A2 M(T205A)) virus in a single cycle replication assay. The

impact of RSV M (T205A) mutation on viral transmissibility was also studied in a multiple

cycle replication assay. Investigation of the spread of the virus in the monolayer of cell

culture, which is a closed population with a controlled environment, could give insights into

important determinants of transmissibility in a more complex system, i.e a whole organism.

3.2 Methods

3.2.1 Cells and virus

Vero E6 and A549 cells were maintained in culturing medium as described in section 2.2.1.

The wild type (rRSV A2) and mutant (rRSV A2 M(T205A)) virus were grown in Vero E6,

followed by immunostaining plaque assay to determine the titre of infectious virus particle

(pfu/ml) as described in section 2.2.2.

Page 80: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

67

3.2.2 Single cycle growth curve

Quadruplicate monolayers of Vero E6 and A549 cells were infected with recombinant wild

type (rRSV A2) or mutant (rRSV A2 M(T205A)) RSV at an MOI of 3, in 200 µl of serum-

free medium. Two hours after inoculation, the medium was replaced with 600 µl of infecting

medium, and infection developed until specified end points, which were 18, 24, 30, 36, and

48 hours post infection as described in section 2.2.3. At each specified time point, 450 µl of

culture supernatant was collected for measuring chemokine expression (Chapter 4) and the

remaining 150 µl was used to collect cell lysates by scraping the well bottom. The cells were

lysed by three freeze-thaw cycles, followed by a brief centrifugation, to release the virus

particles. The count of infectious viral particles was estimated from the clarified lysates by

immunostaining plaque assay (2.2.2.4).

3.2.3 Multiple cycle growth curve

Quadruplicate monolayers of Vero E6 and A549 cells were infected with recombinant wild

type (rRSV A2) or mutant (rRSV A2 M(T205A)) RSV at an MOI of 0.03, in 200 µl of

serum-free medium. Two hours after inoculation, the medium was replaced with 600 µl of

infecting medium, and infection developed until specified end points, which were 0, 1, 2, 3,

4, and 5 days post infection as described in section 2.2.3. At each specified time point, 450 µl

of culture supernatant was collected for measuring chemokine expression (Chapter 4) and the

remaining 150 µl was used to collect cell lysates by scraping the well bottom. The cells were

lysed by three freeze-thaw cycles, followed by a brief centrifugation, to release the virus

particles. The count of infectious viral particles was estimated from the clarified lysates by

immunostaining plaque assay (2.2.2.4).

3.2.4 Data analysis

The RSV titres at each end point were plotted on a log scale (log pfu/ml) against time point of

collection. The mean ± standard error of the mean (SEM) from four replicates of each sample

were used. Statistical differences between the titres of rRSV A2 and rRSV A2 M(T205A) at

each time point were determined by the nonparametric Mann-Whitney U test (GraphPad

(Prism) v.5 software (GraphPad, La Jolla, USA), and a difference was considered significant

if P<0.05.

Page 81: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

68

3.2.5 Confirmation of the stability of rRSV M constructs

rRSV M constructs were obtained by isolating total RNA from infected Vero E6 cells,

followed by generating cDNA using oligo(dT)10, and capturing the M gene using the primer

pair attB1RSVM1 and attB2RSVM256 in PCR. The amplicons obtained in PCR were then

agarose gel-purified and sequenced in the forward and reverse direction using the primer

attB1RSVM1 (forward) and attB2RSVM256 (reverse). The raw sequence data was examined

using BioEdit 7.0.4 (Hall, 2005; Ibis Biosciences).

3.3 Results

3.3.1 Single cycle replication kinetics

Figure 3-1 represents the single cycle growth curve of the rRSV A2 and rRSV A2 M(T205A)

in Vero E6 (A) and A549 cells (B).

Figure 3-1 A shows that the rRSV A2 had exponential growth during 18 – 30 hours post

infection and a lag phase or levelling-off afterwards. However, the rRSV A2 M(T205A) did

not follow the pattern since there were no exponential or lag phases observed. Instead, the

growth curve fluctuated from an initial high titre (18 h.p.i), level off (24 h.p.i), a decrease

from 24 to 36 h.p.i, and then another increase (48 h.p.i). The different shape of the growth

curve was supported by the significantly different titres between the two rRSV at 18, 30, and

36 hours post infection. At 18 h.p.i, the rRSV A2 had a lower titre (mean titre = 2.7 X 103

pfu/ml) than rRSV A2 M(T205A) (mean titre 1.1 X 105 pfu/ml). However, at 30 and 36 h.p.i,

the rRSV A2 had a higher titre (mean titres = 3.5 X 105 pfu/ml at 30 h.p.i, 8.6 X 10

5 pfu/ml at

36 h.p.i) than rRSV A2 M(T205A) (mean titres = 6.2 X 104 pfu/ml at 30 h.p.i, 5.2 X 10

4

pfu/ml at 36 h.p.i). At 24 h.p.i, the rRSV A2 had a higher titre than rRSV A2 M(T205A)

although this was not statistically significant. At 48 h.p.i, there was no statistically significant

difference between rRSV A2 and rRSV A2 M(T205A); however, the titre of rRSV A2 (7.7 X

105 pfu/ml) was lower than rRSV A2 M(T205A) (1.9 X 10

6 pfu/ml).

Figure 3-1 B details the replication of the recombinant RSV in A549 cells. The rRSV A2 had

an exponential phase from 18 – 30 h.p.i and then lagged toward 48 h.p.i., where virus

production slowly decreased. The rRSV A2 M(T205A) proliferated exponentially only up to

24 h.p.i, then lagged in a decreasing fashion for the next 24 hours.

Page 82: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

69

The rRSV A2 M(T205A) yielded lower amounts of infectious particles at each time point

observed, compared to rRSV A2 (Figure 3-1 B). Its exponential peak was represented by a

lower titre of infectious particle compared to the amount produced by rRSV A2 at the same

time point (24 h.p.i., mean titres = 6.0 X 104 pfu/ml (rRSV A2) and 1.6 X 10

4 pfu/ml (rRSV

A2 M(T205A)). At the time rRSV A2 reached the exponential peak (30 h.p.i), the rRSV A2

M(T205A) has started decreasing, and it was reflected by the significantly different amounts

Page 83: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

70

Figure 3-1 The growth curve of single cycle replication kinetics of rRSV A2 and rRSV A2

M(T205A) in (A) Vero E6 and (B) A549 cells. Quadruplicate monolayers of cells were infected at

a multiplicity of infection (MOI) of 3. Whole cell lysates were collected at the indicated time

points and titrated using immunostaining plaque assay described in section 2.2.2.4. Shown is the

means of log10 viral titres (pfu/ml) ± standard error, n ≥ 4, with error bars indicating standard error

of the means. Continuous black line (– ) represents rRSV A2. Continuous pink line represents

rRSV A2 M(T205A). Dotted line indicates limit of detection by immunostaining plaque

assay (50 pfu/ml). Asterisk (*) denotes that the viral titres are significantly different between the

two viruses (P < 0.005) based on the nonparametric Mann-Whitney U-test (GraphPad (Prism), La

Jolla, USA).

Page 84: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

71

of infectious particles produced at that time point. The mean titre of rRSV A2 was 2.3 X 105

pfu/ml, while the mean titre of rRSV A2 M(T205A) was 6.3 X 103 pfu/ml.

The rRSV A2 M(T205A) was capable of producing a relatively comparable amount of virus

particles to rRSV A2 at 48 h.p.i in IFN-α/β-free environment (Figure 3-1 A), but it did not do

so in the presence of IFN-α/β (Figure 3-1 B). The rRSV A2 produced a significantly higher

viral titre (mean titre = 9.4 X 104 pfu/ml) than rRSV A2 M(T205A) (mean titre = 2.1 X 10

3

pfu/ml) at 48 h.p.i.

Comparison between the rRSV A2 growth in a non-IFN-α/β-expressing environment (Vero

E6 cells, Figure 3-1 A) and in an IFN-α/β-expressing environment (A549 cells, Figure 3-1 B)

shows that the exponential phase lasted during 18 – 30 h.p.i and the lag phase took place 30 –

48 h.p.i in both cells. The rRSV A2 grown in A549 cells peaked at a significantly lower titre

(mean = 2.3 X 105 pfu/ml) than rRSV A2 grown in Vero E6 cells (mean = 3.5 X 10

5 pfu/ml).

While rRSV A2 grown without the presence of IFN-α/β continued to produce more particles

in the lag phase, rRSV A2 grown in IFN-α/β expressing environment was affected by the

IFN-α/β such that the production of infectious particles declined during the lag phase. This

was shown by the significantly different amount of infectious particles produced at 48 h.p.i

(mean titre: rRSV A2 A549 = 9.4 X 104 pfu/ml; rRSV A2 Vero E6 = 7.7 X 10

5 pfu/ml).

rRSV A2 M(T205A) replicated at significantly lower titres in A549 cells (Figure 3-1 B) than

in Vero E6 cells (Figure 3-1 A), at 18, 30, and 48 h.p.i. At 18 h.p.i., the mean titres were 1.1

X 105 pfu/ml (Vero E6) and 3.6 X 10

2 pfu/ml (A549). At 30 h.p.i., the mean titres were 6.2 X

104 pfu/ml (Vero E6) and 6.3 X 10

3 pfu/ml (A549). At 48 h.p.i, the mean titres were 1.9 X

106 pfu/ml (Vero E6) and 2.1 X 10

3 pfu/ml (A549). This result shows that overall, the

replication of rRSV A2 M(T205A) was significantly inhibited by the antiviral action of IFN-

α/β.

Page 85: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

72

3.3.2 Multiple cycle replication kinetics

Figure 3-2 represents the multiple cycle growth curve of the rRSV A2 and rRSV A2

M(T205A) in Vero E6 (A) and A549 cells (B).

In the IFN-α/β-free environment, both the recombinant RSV grew by following the

exponentiallag phases of virus replication curve (Figure 3-2 A). The exponential peak took

place at day 2 post infection, and the lag phase followed until the endpoint of observation

(day 5 post infection). The lag phase of rRSV A2 had a trend toward a slight increase in the

number of infectious particle production; while the lag phase of rRSV A2 M(T205A) showed

a decrease in the number of infectious particle production. The rRSV A2 M(T205A)

proliferated into significantly lower number of progeny than the rRSV A2, as evidenced at

the days 2, 4, and 5 post infection, where the mean titres of rRSV A2 were 9.7 X 105 pfu/ml

(D2), 1 X 106 pfu/ml (D4), and 3.7 X 10

6 pfu/ml (D5) while the mean titres of rRSV A2

M(T205A) were 4.9 X 104 pfu/ml (D2), 4 X 10

4 pfu/ml (D4), and 5.7 X 10

3 pfu/ml (D5).

In an environment that exerted the antiviral IFN-α/β pressure (A549 cells, Figure 3-2 B),

rRSV A2 grew exponentially, peaking at day 2 post infection, and then had a lag phase with a

slight increase in particle production. Although showing a similar pattern of growth kinetics,

the amount of infectious particles produced in A549 cells (Figure 3-2 B) were significantly

lower than the amount produced in Vero E6 cells (Figure 3-2 A) at each time point of

observation.

The titres of rRSV A2 M(T205A) in A549 cells were below the detection limit of the

immunostaining plaque assay (50 pfu/ml, dotted line) at each time point (Figure 3-2 B).

Page 86: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

73

Figure 3-2 The growth curve of multiple-step replication kinetics of rRSV A2 and rRSV A2

M(T205A) in (A) Vero E6 and (B) A549 cells. Quadruplicate monolayers of cells were

infected at a multiplicity of infection (MOI) of 0.03. Whole cell lysates were collected at the

indicated time points and titrated using immunostaining plaque assay described in section

2.2.2.4. Shown is the means of log10 viral titres (pfu/ml) ± standard error, n ≥ 4, with error

bars indicating standard error of the means. Continuous black line (– ) represents rRSV A2.

Continuous pink line represents rRSV A2 M(T205A). Dotted line indicates limit of

detection by immunostaining plaque assay (50 pfu/ml). Asterisk (*) denotes that the viral titres

are significantly different between the two viruses (P < 0.005) based on the nonparametric

Mann-Whitney U-test (GraphPad (Prism), La Jolla, USA).

Page 87: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

74

3.3.3 Confirmation of sequence of rRSV A2 and rRSV A2 M(T205A)

The codon at amino acid 205 was shown to be ACA, which codes for threonine (T) for the

wild type rRSV A2, and GCT, which codes for alanine (A) for the mutant rRSV A2

M(T205A) (Figure 3-3). This shows that the rRSV M constructs were stable throughout all

passages and experimental procedures.

Page 88: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

75

Figure 3-3 Chromatogram of DNA sequencing of the matrix gene of rRSV A2 and rRSV A2

M(T205A) showing the difference between the threonine and alanine codons at amino acid 205.

Page 89: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

76

3.4 Discussion

Reverse genetics technology, which was used to generate recombinant RSVs, ensures that the

mutant and wild type viruses being compared have identical genomes except at the sites

where mutation was performed. Therefore, the effects of the compensatory mutations in other

genes, which may appear due to natural selection in naturally-circulating strains, are not

present, allowing a direct study of the effect of a single mutation on the replication efficiency

of a virus.

RSV M205 (T) was initially selected for mutation due to its proximity to the NES

(194

IIPYSGLLLVITV206

) and its potential as a CK2 phosphorylation site. The nuclear export

of RSV M has been proposed to be regulated by a phosphorylation mechanism (Alvisi et al.,

2008); therefore RSV M205 (T) was suggested to have a possible role in the regulation of

nuclear export. In this study, the rRSV A2 M(T205A) was able to produce a relatively similar

titre to the rRSV A2 in a single cycle of replication (Vero E6 cells, Figure 3-1 A). This

suggests that amino acid residue 205 (T) is not a site that is critical for the success of RSV M

nuclear export to facilitate assembly of ribonucleoproteins and glycoproteins to produce new

infectious particles. Furthermore, although not directly tested, T205 may not be the

regulatory phosphorylation site that promotes the nuclear export of RSV M, as suggested by

the fact that the assembly of progeny virus did not appear to be affected (Vero E6 cells,

Figure 3-1 A). Regulated, timely-mannered localisation of the M protein is essential for

optimal RSV replication and assembly (Ghildyal et al., 2009), and the results obtained in this

study, from the single cycle replication assay, suggest that M is present in the cytoplasm to

facilitate particle assembly after 24 h.p.i.

Many factors regulate the capability of a virus to spread, including the inherent properties of

the virus, such as the structural proteins. In this study, the impact of M(T205A) substitution

on the capability of RSV to spread the infection from cell to cell was investigated by

conducting multiple-step replication kinetics assay with a low virus to cell ratio (MOI =

0.03). Multiple cycle replication assay is more sensitive than single cycle replication assay to

identify different properties between two virus because it can magnify the impact of small

differences (Wang and Bushman, 2006). When the initial virus-to-cell ratio is low (multiple

cycle replication assay, MOI 0.03), viral particles need more than one replication cycle to

Page 90: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

77

maximally infect all cells, and this requires the capability of the virus to reinitiate infection in

uninfected cells.

In this study, the multiple cycle replication assay showed that rRSV A2 M(T205A) has a

defect in spreading the infection from infected to uninfected cells. In Vero E6 cells, the

multiple cycle replication assay resulted in a lower amount of infectious progeny in the rRSV

A2 M(T205A) compared to the rRSV A2 (Figure 3-2 A), unlike in the single cycle

replication assay where the rRSV A2 M(T205A) proliferated more than the rRSV A2 (Figure

3-1 A). This suggests that without the confounding effect of the antiviral IFN-α/β, rRSV A2

M(T205A) managed to produce progeny but suffered from a restricted ability to spread the

infection. This finding corroborates the observation that the processes in the replication that

associate with particle spread was likely to be affected by M205 (TA) substitution, or

M205 (T) plays an important role in the budding or release of RSV. In addition, it is

important to note the fact that rRSV A2 M(T205A) produced a higher titre than rRSV A2, in

single cycle replication assay at 18 h.p.i, may be attributed to a high starting titre in Vero E6

cells possibly the result of unremoved inoculum.

The lower titres of rRSV A2 in A549 cells (Figure 3-1 B and 3-2 B) compared to Vero E6

cells (Figure 3-1 A and 3-2 A) suggests that the products of the antiviral IFN-α/β gene restrict

the replication efficiency of RSV. This finding corroborates a previous study describing that

the absence of IFN-α/β in Vero E6 cells allows for a higher level of viral replication

compared to other cell lines such as A549 and HUVEC (Kraus et al., 2004). In the case of

rRSV A2 M(T205A), the IFN-α/β activity severely restricts its spread to the extent that viral

titre could not be detected by the immunostaining plaque assay (Figure 3-2 B). Thus, RSV M

could have an important role for IFN-α/β suppression to facilitate the spread of infection and

that M205 (T) might be functional in this role. Overall, the M(T205A) substitution confers a

defect in biological processes of RSV replication that prevent the virus to re-initiate infection

and to suppress the antiviral activity of IFN-α/β. The actual mechanism of how the single-site

M(T205A) substitution reduces RSV capability to inhibit the antiviral activity of IFN-α/β

cannot be assessed in detail based on the result obtained in this study. Likewise, whether

RSV M functions independently or in cooperation with other cellular or viral components

still requires further study.

The results obtained in this study clearly represent the characterisation of the mutant rRSV

A2 M(T205A) in comparison to the wild type rRSV A2. This was confirmed by performing

Page 91: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

78

DNA sequencing of the M constructs of rRSV, which showed that the amino acid residue

threonine (T) in M205 of rRSV A2 was coded by the codon ‘ACA’ and the amino acid

residue alanine (A) in M205 of rRSV A2 M(T205A) was coded by the codon ‘GCT’.

Therefore, the two rRSV M constructs were stable and the viral titres, and immune responses

(detailed in Chapter 4), observed during this study truly represent the phenotypes of the

mutant rRSV A2 M(T205A) and the wild type rRSV A2 in in vitro growth.

3.5 Summary

There are some points that can be inferred based on the findings presented in this chapter.

Firstly, the M protein has a substantial role in ensuring the success of viral egress in RSV

replication. Secondly, the M protein might have a function in counteracting the antiviral

activity of IFN-α/β at the level of replication in infected cells and in the spread of infection.

Thirdly, the M205 (T) is potentially a site that is critical for M to facilitate the egress of

progeny, as well as to suppress the antiviral activity of IFN-α/β. In summary, this study

shows that the M protein is crucial for the replication efficiency of RSV.

Page 92: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

79

CHAPTER 4

IN VITRO EXPRESSION OF PRO-

INFLAMMATORY MEDIATORS IN

RESPONSE TO INFECTION BY rRSV

A2 M(T205A)

4 Pro-Inflammatory Response in RSV Infection

Page 93: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

80

Page 94: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

81

4.1 Introduction

Inflammatory mediators, such as chemokines (chemotactic cytokines), are of particular

relevance in RSV infection because the inflammatory responses triggered by RSV infection

contribute largely to RSV-induced bronchiolitis and pneumonia (Garofalo et al., 1996, Dudas

and Karron, 1998, Casola et al., 2001a, Noah et al., 2002). Chemokines function in

trafficking leukocyte migration to the infection sites for pathogen clearance; but if

overexpressed, can cause damage to the body tissues due to cytotoxicity (Kindt et al., 2007).

IL-8 and RANTES are two chemokines that are activated prominently in response to RSV

infection, with IL-8 the attractant for neutrophils and RANTES the attractant of eosinophils

(Collins and Graham, 2008). Overexpression of IL-8 and RANTES during RSV infection has

been shown to associate with the development of severe bronchiolitis in infants (Mukaida et

al., 1998, Genin et al., 2000).

The expression of IL-8 and RANTES in infected cells is regulated by the activation of

transcriptional regulatory proteins. The main regulators for RANTES expression following

RSV infection are NF-κB and IRF3 (Casola et al., 2001b). Viral infection activates IRF3 by

inducing phosphorylation at serine/threonine residues in the CTD (S385, 386, 396, 398, 402,

405, and T404), which causes IRF3 nuclear translocation to promote the transcription of

interferon-stimulated genes (ISGs), including RANTES, and IFN-α/β that in turn can induce

RANTES expression via JAK-STAT signalling (Bose and Banerjee, 2003). IRF3 can also

induce RANTES activation directly, bypassing the JAK-STAT signalling pathway (Lin et al.,

1999). The induction of RANTES gene transcription by virus infection requires cooperation

between the IRF3 and NF- κB. Disruption of either pathway abolishes the ability of the other

to activate RANTES expression; however, only IRF3 is targeted by RSV, whereas NF- κB

plays more of a cofactor role (Lin et al., 1999, Genin et al., 2000, Casola et al., 2001b).

The transcriptional activators of IL-8 expression following RSV infection consist of NF- κB

and NF-IL-6, with IRF-1 acts as a cofactor (Mukaida et al., 1994, Garofalo et al., 1996,

Casola et al., 2000). The combinational activation of NF-κB and NF-IL-6 binding sites in the

5’-flanking region of the gene is required for the transcription of IL-8 gene following

sensitisation by RSV infection; neither site alone is sufficient for gene activation (Mukaida et

al., 1994, Mastronarde et al., 1996).

Page 95: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

82

The secretion of IL-8 and RANTES has been shown to dictate the subsequent inflammatory

response and the eventual clinical symptoms in RSV disease (Olszewska-Pazdrak et al.,

1998, Miller et al., 2003). Therefore, it is of great value to study their expression in cell

culture, particularly with the aim of assessing their activation as the potential early predictor

of the severity of RSV-associated illness.

This chapter aimed to assess the impact of M(T205A) substitution on the production of pro-

inflammatory mediators RANTES and IL-8 in RSV infection, with and without the influence

of the antiviral action of IFN-α/β. The pro-inflammatory response directed to rRSV A2

M(T205A) was studied in Vero E6 cells that have a genetic defect in the IFN-β locus and

thus cannot express type-1 IFN (IFN-α/β) (Emeny and Morgan, 1979), and in A549 cells that

express the strong antiviral IFN-α/β signalling pathway. In doing so, the experimental results

from Vero E6 cells can serve as the control for expression of the IFN-α/β–mediated pro-

inflammatory response. In addition, Vero E6 cells are useful to study NF-κB-mediated

immune response without the confounding effects of IFN-α/β activation.

4.2 Methods

4.2.1 Cells and virus

Vero E6 and A549 cells were maintained in culturing medium as described in section 2.2.1.

The wild type (rRSV A2) and mutant (rRSV A2 M(T205A)) virus were passaged in Vero E6

cells, followed by immunostaining plaque assay to determine the titre of infectious virus

particle (pfu/ml) as described in section 2.2.2.

4.2.2 Single cycle replication assay

Quadruplicate monolayers of Vero E6 and A549 cells were infected with rRSV A2 or rRSV

A2 M(T205A) at an MOI of 3, in 200 µl of serum-free medium. Mock infection was also

performed using just serum-free medium. Two hours after inoculation, the medium was

replaced with 600 µl of infecting medium, and infection developed until specified end points,

which were 18, 24, 30, 36, and 48 hours post infection as described in section 2.2.3. At each

specified time point, 450 µl of culture supernatant was collected.

Page 96: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

83

4.2.3 Multiple cycle replication assay

Quadruplicate monolayers of Vero E6 and A549 cells were infected with rRSV A2 or rRSV

A2 M(T205A) at an MOI of 0.03, in 200 µl of serum-free medium. Mock infection was also

performed using just serum-free medium. Two hours after inoculation, the medium was

replaced with 600 µl of infecting medium, and infection developed until specified end points,

which were 0, 1, 2, 3, 4, and 5 days post infection as described in section 2.2.3. At each

specified time point, 450 µl of culture supernatant was collected.

4.2.4 Enzyme-linked immunosorbent assay (ELISA)

Chemokine measurement using ELISA was performed as detailed in section 2.2.4.1. Briefly,

ELISA plate was coated with 100 µl of capture antibody (0.5 µg/ml in PBS) at room

temperature overnight. Plate was then washed four times with PBS + 0.05% Tween-20,

blocked with 300 µl PBS + 1% BSA for an hour at room temperature, and washed again as

described. IL-8 and RANTES standards and the culture supernatants were two-fold-serially-

diluted in diluent (PBS + 0.1% BSA+ 0.05% Tween-20). 100 µl diluted standard and samples

were added into the plate in triplicate and incubated for two hours at room temperature. The

plate was washed, 100 µl of 0.25 µg/ml detection antibody in diluent added, and incubated at

room temperature for 2 hours. The plate was washed again, then 100 µl of avidin-HRP

conjugate in diluent (1:200) was applied and plate incubated for 30 minutes at room

temperature, then washed. 100 µl of ABTS liquid substrate solution was added and optical

density (OD) values collected based on absorbance of light with wavelength set at 405 nm in

ELISA plate reader.

The concentration of chemokine (pg/ml) was calculated as explained in section 2.2.4.2.

Briefly, the optical density values of the standard (y-axis) were plotted against time point (x-

axis). A regression analysis of the scatterplots was done to determine the function of the

polynomial regression curve. The chemokine concentration was determined by entering the

OD values into the function. The values of chemokine concentration were then normalised

against that of the mock, and presented as the mean value from triplicate reactions with

variability expressed as the standard error of the mean.

Page 97: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

84

4.2.5 Data analysis

Data was analysed using the nonparametric Mann-Whitney U-test (GraphPad (Prism) v5, La

Jolla, USA), as explained in section 2.2.13.2. Results were considered statistically significant

if P<0.05.

4.3 Results

4.3.1 Single cycle replication assay

The chemokine expression in single cycle replication assay was investigated to understand

the initial pro-inflammatory response directed towards infection by rRSV A2 M(T205A) in

only one cycle of viral replication.

4.3.1.1 Expression of IL-8

Following infection by either rRSV A2 or rRSV A2 M(T205A), IL-8 was produced at only a

very low concentration in Vero E6 cells (Figure 4-1 A), but was expressed higher in A549

cells (Figure 4-1 B). In A549 cells, but not Vero E6 cells, rRSV A2 M(T205A) induced a

significantly higher concentration of IL-8 than rRSV A2 at 30, 36, and 48 h.p.i (Figure 4-1

B). It was shown that IL-8 had a trend toward decreased expression after 30 h.p.i in rRSV

A2-infected A549 cells; however, the trend was toward increasing expression in rRSV A2

M(T205A)-infected A549 cells (Figure 4-1 B).

Page 98: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

85

Figure 4-1 Expression of interleukin 8 (IL-8) in cells infected with rRSV A2 or rRSV A2

M(T205A) in single cycle replication assay. (A) Expression of IL-8 in Vero E6 cells. (B)

Expression of IL-8 in A549 cells. Cells were infected with RSV at a multiplicity of infection = 3

and supernatant collected at 18, 24, 30, 36, and 48 hours post infection to measure the chemokine

concentration secreted overtime using enzyme-linked immunoassay (ELISA). The concentration

of cell-secreted chemokines is plotted as a function of time after infection. Shown are the mean

values of three separate optical density readings that were then converted into concentration

(pg/ml), with standard error of the means. Black line (– ) represents rRSV A2. Pink line

represents rRSV A2 M(T205A). Asterisk (*) denotes that the chemokine concentrations are

significantly different between the two viruses (P<0.05) based on the nonparametric Mann-

Whitney U-test (GraphPad (Prism), La Jolla, USA).

Page 99: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

86

4.3.1.2 Expression of RANTES

In contrast to IL-8, RANTES was produced in both the Vero E6 and A549 cells that were

infected by either rRSV A2 or rRSV A2 M(T205A) (Figure 4-2).

The production of RANTES in Vero E6 cells infected with rRSV A2 was significantly higher

than in cells infected with rRSV A2 M(T205A) at 18 and 48 h.p.i (Figure 4-2 A). In rRSV A2

M(T205A)-infected Vero E6 cells, RANTES expression was low, with the mean

concentration below 250 pg/ml, except at 24 h.p.i (344 pg/ml); as compared to the expression

in rRSV A2-infected Vero E6 cells, which was over 250 pg/ml in all the time points of

observation.

In A549 cells, however, there was no significant difference in RANTES expression in cells

infected by either rRSV A2 or rRSV A2 M(T205A) (Figure 4-2 B). RANTES appeared to be

induced by rRSV A2 in a biphasic fashion, where two peaks of production were observed at

30 and 48 h.p.i. The biphasic production of RANTES was not observed in rRSV A2

M(T205A)-infected cells.

Page 100: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

87

Figure 4-2 Expression of regulated upon activation, normal T-cell expressed, and secreted

(RANTES) in cells infected with respiratory syncytial virus (RSV) in single cycle replication

assay. (A) Expression of RANTES in Vero E6 cells. (B) Expression of RANTES in A549 cells.

Cells were infected with RSV at a multiplicity of infection = 3 and supernatant collected at 18,

24, 30, 36, and 48 hours post infection to measure the chemokine concentration secreted

overtime using enzyme-linked immunoassay (ELISA). The concentration of cell-secreted

chemokines is plotted as a function of time after infection. Shown are the mean values of three

separate optical density readings that were then converted into concentration (pg/ml), with

standard error of the means. Black line (– ) represents rRSV A2. Pink line represents rRSV

A2 M(T205A). Asterisk (*) denotes that the chemokine concentrations are significantly

different between the two viruses (P<0.05) based on the nonparametric Mann-Whitney U-test

(GraphPad (Prism), La Jolla, USA).

Page 101: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

88

4.3.2 Multiple cycle replication assay

The chemokine expression in multiple cycle replication assay was investigated to understand

the pro-inflammatory response directed towards infection by rRSV A2 M(T205A) in the

condition of more than one cycle of viral replication.

4.3.2.1 Expression of IL-8

There are three notable observations from IL-8 production in multiple cycle replication assay.

Firstly, IL-8 production in multiple cycle replication assay, in agreement with the single cycle

replication assay (Figure 4-1), was observed only in rRSV-infected A549, and not in Vero E6

cells (Figure 4-3). In A549 cells, a significantly higher titre of IL-8 was observed in cells

infected with rRSV A2 compared to rRSV A2 M(T205A) at days 0 – 4 post infection (Figure

4-3 B). This pattern is in contrast to IL-8 production in rRSV-infected A549 cells in single

cycle replication assay, where rRSV A2 M(T205A)-infected A549 cells secreted a

significantly higher titre of IL-8 than rRSV A2-infected A549 cells (Figure 4-1 B).

Secondly, to compare between IL-8 expression in rRSV A2-infected A549 cells in single

cycle replication assay (Figure 4-1 B) and multiple cycle replication assay (Figure 4-3 B)

during a similar period of observation (48 h.p.i (single cycle) or 2 d.p.i (multiple cycle)), the

values of IL-8 concentration in single cycle replication assay were less variable than in

multiple cycle replication assay. In single cycle replication assay, the lowest concentration of

secreted IL-8 was 34.72 pg/ml, the highest concentration was 1031.07 pg/ml, and the mean

value was 703.13 pg/ml. In multiple cycle replication assay, the lowest concentration was 0

pg/ml, the highest concentration was 1044.75 pg/ml, while the mean value was 417.97 pg/ml.

Thirdly, the rRSV A2 M(T205A)–infected A549 cells produced a lower IL-8 concentration in

multiple cycle replication assay than in single cycle replication assay in a period of 2 d.p.i (48

h.p.i) (Figure 4-1 B and 4-3 B). In single cycle replication assay, the mean value was 915.9

pg/ml; while in multiple cycle replication assay, the mean value was 27.16 pg/ml.

Page 102: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

89

Figure 4-3 Expression of interleukin 8 (IL-8) in cells infected with respiratory syncytial virus

(RSV) in multiple cycle replication assay. (A) Expression of IL-8 in Vero E6 cells. (B)

Expression of IL-8 in A549 cells. Cells were infected with RSV at a multiplicity of infection =

0.03 and supernatant collected at day 0, 1, 2, 3, 4, and 5 post infection to measure the

concentration of chemokines expressed overtime using enzyme-linked immunoassay (ELISA).

The concentration of cell-secreted chemokines is plotted as a function of time after infection.

Shown are the mean values of three separate optical density readings that were then converted

into concentration (pg/ml) with standard error of the means. Black line (– ) represents rRSV A2.

Pink line represents rRSV A2 M(T205A). Asterisk (*) denotes that the chemokine

concentrations are significantly different between the two viruses (P<0.05) based on the

nonparametric Mann-Whitney U-test (GraphPad (Prism), La Jolla, USA).

Page 103: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

90

4.3.2.2 Expression of RANTES

RANTES was produced in Vero E6 and A549 cells in multiple cycle replication assay

(Figure 4-4), but only rRSV A2-infected cells expressed RANTES at significantly higher

concentration than rRSV A2 M(T205A)-infected cells. In addition, Vero E6 cells expressed

an overall higher concentration of RANTES than A549 cells. The mean titre of RANTES

expression in rRSV A2-infected Vero E6 cells was 952.5 pg/ml and in A549 cells was 532.5

pg/ml. In rRSV A2 M(T205A)-infected cells, the mean titre was 184.8 pg/ml (Vero E6) and

49.15 pg/ml (A549).

In rRSV A2-infected Vero E6 and A549 cells, expression of RANTES peaked at day 4 post

infection, where the mean titre in Vero E6 cells was 2210.41 pg/ml and the mean titre in

A549 cells was 1275 pg/ml. The rRSV A2 M(T205A)-infected cells, however, produced peak

RANTES expression at day 1 post infection (24 h.p.i), albeit in much lower titre. The mean

titre in Vero E6 cells was 66.80 pg/ml and the mean titre in A549 cells was 196.64 pg/ml.

Based on the findings, two differences were observed between RANTES expression in

multiple cycle and single cycle replication assays. Firstly, rRSV A2-infected Vero E6 and

A549 cells always produced a higher titre of RANTES in multiple cycle replication assay; in

contrast to in single cycle replication assay, where rRSV A2 M(T205A)-infected A549 cells

managed to secrete a relatively similar titre of RANTES during a period of 48 h.p.i.

Secondly, in multiple cycle replication assay overall RANTES expression in rRSV A2-

infected cells was higher in Vero E6 (mean titre = 952.50 pg/ml) than in A549 cells (mean

titre = 448.57 pg/ml), while in single cycle replication assay the overall titre was lower in

Vero E6 (878.70 pg/ml) than in A549 cells (1177.09 pg/ml).

Page 104: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

91

Figure 4-4 Expression of regulated upon activation, normal T-cell expressed, and secreted

(RANTES) in cells infected with respiratory syncytial virus (RSV) in multiple cycle

replication assay. (A) RANTES expression in Vero E6 cells. (B) RANTES expression in

A549 cells. Cells were infected with RSV at a multiplicity of infection = 0.03 and

supernatant collected at day 0, 1, 2, 3, 4, and 5 post infection to measure the concentration

of chemokines expressed overtime using enzyme-linked immunoassay (ELISA). The

concentration of cell-secreted chemokines is plotted as a function of time after infection.

Shown are the mean values of three separate optical density readings that were then

converted into concentration (pg/ml) with standard error of the means. Black line (– )

represents wild type rRSV A2. Pink line represents CK2 phosphorylation mutant

rRSV A2 M(T205A). Asterisk (*) denotes that the chemokine concedntrations are

significantly different between the two viruses (P<0.05) based on the nonparametric Mann-

Whitney U-test (GraphPad (Prism), La Jolla, USA).

Page 105: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

92

4.4 Discussion

IL-8 was expressed in rRSV-infected A549 cells, but not in Vero E6 cells, in both single

cycle and multiple cycle replication assays. There are two possible mechanisms to explain

this observation. Firstly, IL-8 production in RSV-infected respiratory epithelial cells (A549

cells) has been shown to be controlled by the combinational activation of NF-κB and NF-IL-6

transcriptional factors (Mastronarde et al., 1996). Vero E6 cells have a functional NF-κB

transcriptional factor, but no studies have confirmed the presence of NF-IL-6; therefore, this

potentially partial activation might lead to the lack of IL-8 expression in Vero E6 cells.

Secondly, the IFN-α/β expression in A549 cells might provide a positive feedback, as a

soluble mediator that can signal the ‘antiviral state’, resulting in the amplification of IL-8

expression, particularly during the secondary activation (Bose and Banerjee, 2003).

The concentration of IL-8 in the rRSV A2 M(T205A)-infected A549 cells was significantly

higher than that of the rRSV A2-infected A549 cells in single cycle replication assay (Figure

4-1 B), but it was the opposite way in multiple cycle replication assay (Figure 4-3 B). Based

on this finding, there are two points that can be inferred. Firstly, M(T205A) substitution

might potentially dampen the ability of rRSV in suppressing the IL-8 production, allowing

the secretion of a high IL-8 concentration during the initial recognition. However, continuing

IL-8 production requires particle proliferation, particularly to activate the NF-κB activation.

(Fiedler et al., 1996, Garofalo et al., 1996). Therefore, the rRSV A2 M(T205A) infection in

multiple cycle replication assay resulted in muted IL-8 production because the rRSV A2

M(T205A) lacked the ability to spread (Figure 3-2 B). Secondly, IL-8 expression has been

shown to be dependent on a certain threshold of the initiating inoculum titre (Arnold et al.,

1994), probably representing the number of infected cells. As a result, the rRSV A2

M(T205A) induced a higher concentration of IL-8 in single cycle replication assay where

cells were initially infected with 100 times greater virus to cell ratio (MOI of 3), but not in

multiple cycle replication assay, where cells infected with an MOI of 0.03.

M (T205A) substitution was shown to also affect RANTES, with expression only occurring

in an infection system that exerts the antiviral IFN-α/β (Figure 4-2 B). The expression of

RANTES is regulated by IRF3, which is the transcriptional factor of IFN-α/β, independently

or with the assistance of NF-κB (Lin et al., 1999, Casola et al., 2001b, Chew et al., 2009).

Both the transcriptional factors are present in Vero E6 cells, but the IFN-α/β-facilitated

Page 106: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

93

JAK/STAT signalling, that is present only in A549 cells, amplifies chemokine production by

introducing an ‘antiviral state’ in the population of cells (Bose and Banerjee, 2003). In doing

so, the redirection might represent the effect of IFN-α/β in elevating RANTES production.

By taking the fact that rRSV A2 M(T205A) induced the production of a higher titre of

RANTES in single cycle replication (MOI = 3) (Figure 4-2 B) than in multiple cycle

replication (MOI = 0.03) (Figure 4-4 B) into consideration, it can be inferred that viral

replication is required for RANTES expression. Indeed, it has been shown that RANTES

secretion in RSV-infected cells requires replicating particles in infected cells (Miller et al.,

2003). One importance of the viral replication for RANTES expression is by inducing the

production of the IFN-α/β, which functions to amplify RANTES production. The

transcriptional factor of IFN-α/β, IRF3, is activated only by the recognition of dsRNA, which

in the case of (-) ssRNA virus like RSV, is produced as the genomic intermediate during viral

replication (Stetson and Medzhitov, 2006). Therefore, the disrupted viral replication can

eliminate the optimal IFN-α/β-facilitated signalling pathway. As such, the disrupted

proliferation ability of rRSV A2 M(T205A) likely contributed to the low induction of

RANTES production despite the presence of IFN-α/β.

The suggestion that M(T205A) substitution potentially dampen the ability of rRSV in

suppressing the IL-8 production to result in a higher IL-8 concentration during the initial

recognition, might work for the RANTES activation as well. RANTES expression has been

shown to be elevated in 24 h.p.i (Figures 4-2 A, 4-4 A, and 4-4 B), but depleted afterwards,

consistent with the requirement of viral replication for RANTES expression. As such, it can

be inferred that M(T205A) substitution might result in the suppression of pro-inflammatory

mediators, but the phenotype was not expressed since the rRSV A2 M(T205A) lacks the

ability to spread.

The results presented in this chapter suggest that RSV M has a significant role in the

regulation of the expression of pro-inflammatory mediators in the innate immune response.

This role is suggested to be associated with the proliferating ability of RSV, since replication

processes have been shown to be important for the production of RANTES and IL-8 in RSV

infection (Fiedler et al., 1996, Miller et al., 2003). RSV proteins that have been shown to alter

the expression of IL-8 and RANTES, as well as their transcriptional factors IRF3 and NF-

κB, are NS1 and NS2 (Spann et al., 2005). Therefore, it is interesting to propose a possible

cooperation between RSV M, NS1, and NS2 in regulating the expression of IL-8 and

Page 107: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

94

RANTES. Further study is needed to elucidate the exact role of the M protein in inducing

pro-inflammatory mediators in RSV infection.

4.5 Summary

The findings presented in this chapter shows that the M(T205A) substitution led to a lower

chemokine production in infected cells, very likely associated with the reduced ability of the

mutant to undergo proliferation. One mechanism underlying this observation is that the

proliferation defect leads to the weakened induction of IFN-α/β that has a role in amplifying

chemokine expression by introducing an ’antiviral state’ among the population of cells.

Page 108: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

95

CHAPTER 5

THE SUBCELLULAR LOCALISATION

OF hMPV N PROTEIN IN

TRANSFECTED CELLS

5 The Subcellular Localisation of hMPV N Protein in Transfected Cells

Page 109: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

96

Page 110: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

97

5.1 Introduction

The nucleocapsid (N) protein of several (-)ssRNA viruses has been shown to undergo

independent nucleocytoplasmic transport in order to perform non-structural functions in the

nucleus. For example, the nuclear localisation of infectious bronchitis virus (IBV) N is

associated with the reorganisation of fibrillarin distribution in an attempt to delay cell growth

(Hiscox et al., 2001, Chen et al., 2002), while the nuclear localisation of measles virus (MV)

N protein blocks the nuclear translocation of STATs (Takayama et al., 2012). Preliminary

investigation showed that the hMPV N localises to the nucleus in the late stage of infection

(day 5 post infection) (Ghildyal R & Gahan M, personal communication). Putative NES and

NLS motifs were identified in the ORF of hMPV Can97-83 N gene (GenBank accession

number AY145278). The putative NES motif 1MSLQGIHLSDLSYKH

15, was identified by

submitting the hMPV Can97-83 N amino acid sequence to EXPASY

(http://prosite.expasy.org/), a resource portal that provides access to databases and software

tools in motif recognition. This motif conforms to the common NES motif LXXXLXXLXL,

which is rich in hydrophobic residues (la-Cour et al., 2004). A putative bipartite NLS-like

motif 185

RRANRVLSDALKRYPR200

was manually identified (R. Ghildyal), and resembles

the RSV M NLS 154

TSKKVIIPTYLRSISVRNK172

(Ghildyal et al., 2006). As a first step

towards determining the functionality of these motifs, and as a prelude to elucidating the

possible nuclear functions of hMPV N, the subcellular localisation of hMPV N was studied in

transfected mammalian cells.

The functionality of the NES and NLS was studied in the context of full-length hMPV N and

as independent transport motifs. Identifying the functionality of a structural motif as a part of

a protein, and as an individual motif, is important because of the following reasons. Firstly,

motifs that are exposed on the surface are more likely to be recognised by nuclear transport

factors; secondly, the functionality of a motif might be assisted by unique flanking

sequence(s) (Silver, 1991). The subcellular localisation of full-length hMPV N and the

transport motif was studied by expressing full-length and deletion constructs generated based

on hMPV N gene in a transfection host.

To study the subcellular localisation, hMPV N constructs were expressed as green fluorescent

protein (GFP)-fusion proteins, in which GFP is positioned in the N-terminal end of each

construct. GFP is a green-light emitting marker for gene expression that has stable

Page 111: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

98

fluorescence, even after formaldehyde treatment, does not have a toxic effect, and does not

interfere with cell growth and function (Chalfie et al., 1994). The localisation of hMPV N

constructs in transfected cells can be traced via GFP fluorescence upon excitation by the blue

laser (395 – 490 nm).

Cloning in an expression vector is one approach to generate a GFP-tagged ORF without

interfering with the function of the ORF itself. The GatewayTM

recombination cloning (RC)

technology was used to clone the hMPV N constructs into an expression vector. The

GatewayTM

RC technology is highly accurate in generating expression clones, being

facilitated by two specific recombination steps and stringent negative and positive selection,

to specifically maintain the directionality of the ORF (Hartley et al., 2000). The first

recombination step is to produce an entry clone and the second recombination is to generate

the expression clone. Both recombination are facilitated by specific recombination sites,

which is a pair of attB sites in hMPV N constructs and the complementary pair of attP sites in

the vector, to allow for conservative (no nucleotides are added or lost), directional

(orientations of segments in new clones are fixed according to the orientations of the att

sites), and concerted (all strand exchanges occur in protein-DNA complexes, no free ends are

available for side reactions) cloning (Hartley et al., 2000, Hopkins et al., 2012). The selection

for each clone is facilitated by the negative selection marker, ccdB gene, and the antibiotic

resistance gene in each vector. ccdB gene encodes a cell-killing protein, which affects

bacterial gyrase, an enzyme that facilitates DNA unwinding during replication (Reyrat et al.,

1998). Importantly, the 5’-end recombination site of the GatewayTM

RC, attB1, contains the

translation signals for E. coli and mammalian cells, making the technology versatile for

protein expression in both prokaryotic and mammalian cells (Hartley et al., 2000).

Transfection is a method of introducing foreign genetic material into a cell with the aim of

analysing gene expression. The use of mammalian cells give the advantage of assisting

peptide folding, providing posttranslational modifications compared to prokaryotic cells

(E. coli), and allowing the expression of protein in the proper intracellular compartments

(Kingston, 2003, Brondyk, 2009, Hopkins et al., 2012). Additionally, transfection has the

advantage of protein overexpression, allowing the researcher to observe the protein separately

from other components of the cells. Also, cells used in transfection have generally been well

characterised such that no cellular parameters interfere with the investigation. Transient

transfection has the advantage of not requiring integration of the gene of interest into host

Page 112: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

99

chromosomal DNA; therefore can allow quick observation of the subcellular localisation of

hMPV N constructs (24 – 48 hours post transfection) (Liew et al., 2007).

This study aimed to investigate the nucleocytoplasmic transport of hMPV N in transiently

transfected Cos-7 cells, by using the GatewayTM

RC technology to generate an expression

clone containing GFP-tagged hMPV N constructs.

5.2 Methods

5.2.1 Generation of constructs and recombination cloning

Presented is a brief overview of generating hMPV N constructs and recombinational cloning

using Gateway technology. Total RNA was first extracted from hMPV-infected Vero E6 cells

(5.2.1.1) and reverse transcribed into cDNA (5.2.1.2); then the hMPV N constructs were

generated via PCR using the cDNA as template and specific attB-tagged primer pairs (table

2-15) (5.2.1.3). The PCR products, were purified (5.2.1.4), and each construct was then

inserted into the donor vector (pDONR®207) by recombination between attB1/attP1 and

attB2/attP2 sites, catalysed by BP ClonaseTM

(5.2.1.5). The hMPV N construct was inserted

into the expression clone by recombination between the entry clone and the expression vector

pEPI-DESTC (recombination between attL1/attR1 and attL2/attR2 sites), catalysed by LR

ClonaseTM

(5.2.1.5). The products of the BP and LR recombination reactions were

transformed into electrocompetent E. coli to generate the entry and expression clone

(5.2.1.6).

5.2.1.1 Extraction of total RNA from hMPV-infected cells

80%-confluent Vero E6 monolayer was infected with hMPV at an MOI of 1 and incubated at

37o C for 24 hours (section 2.2.2.2). The infection medium was replaced with 2.5 ml of

TRIzol® reagent and flask incubated for 5 minutes at room temperature to lyse the cells. Cell

suspension was transferred into a 10 ml conical tube, mixed with 0.5 ml chloroform,

incubated for 3 minutes at room temperature, and centrifuged at 12,000x g for 15 minutes at

4o C. The aqueous phase (clear) was transferred into a new 10 ml conical tube. From this

aqueous phase, RNA was precipitated with 1.25 ml of isopropyl alcohol for 10 minutes at

room temperature, and pelleted by centrifugation at 12,000x g and 4o C for 10 minutes. RNA

was washed with 2.5 ml of 75% ethanol, and re-pelleted by centrifugation at 7,500x g and 4o

Page 113: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

100

C for 5 minutes. The RNA was air dried and resuspended in nuclease-free water. The RNA

extraction is described in detail in section 2.2.6.1.

5.2.1.2 Reverse transcription

1 µg hMPV RNA was mixed with 270 ng of Oligo(dT)10, 2mM dNTP, and DEPC-treated

water in a final volume of 10 µl, then incubated at 65o C for 10 minutes. The reaction was

added into a mixture of 2X RT buffer, 10 U RNase inhibitor, 5 U reverse transcriptase, and

DEPC-treated water (to a final volume of 10 µl). The final reaction was incubated at 45o C

for 45 minutes. The reverse transcription was terminated by incubation at 70o C for 15

minutes.

5.2.1.3 Polymerase chain reaction (PCR)

PCR was done to generate the hMPV N constructs, by using cDNA as template and a specific

primer pair (Table 2-15) for each construct. 5 µl of cDNA was mixed with 1X Mango Taq

reaction buffer, 1.5 mM MgCl2, 0.2 mM dNTP, 0.25 pmoles of each forward and reverse

primer, 0.025 U Mango Taq DNA polymerase, and nuclease-free water to a final volume of

40 µl. The reaction was subjected to cycling conditions consisting of 95o C incubation for 7

minutes, followed by 35 repeats of 96o C for 30 seconds, 58

o C for 45 seconds, and 72

o C for

1 minute.

5.2.1.4 Gel purification of PCR products

The detailed description of gel purification of PCR products is explained in section 2.2.5.3.

Briefly, whole PCR product was mixed with DNA loading dye (5:1) and electrophoresed in

1% agarose gel (section 2.2.9). The gel was visualised using UV light and the desired DNA

band excised and dissolved in Membrane Binding Solution (1 µl solution for 1 mg gel) via

incubation at 65o C for 10 minutes. The dissolved gel was transferred to an SV Minicolumn

and incubated for 1 minute at room temperature. The column was centrifuged at 19,000x g

for 1 minute and the membrane washed twice with Membrane Wash Solution: the first wash

used 700 µl and the second wash used 500 µl, with 19,000x g and 1 minute centrifugation for

each wash. An additional centrifugation was carried out to remove the residual wash solution,

then the purified DNA eluted with 30 µl of nuclease-free water via 19,000x g for 1 minute

centrifugation.

Page 114: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

101

5.2.1.5 DNA ligation

To generate the entry clones, purified PCR products of a hMPV N construct, pDONR®207,

BP ClonaseTM

, and TE buffer (pH 8.0) were incubated at 25o C overnight, then transformed

into ElectroMAXTM

DH10BTM

competent E. coli (2.2.12.2) and plasmid DNA isolated using

the Miniprep or Midiprep kit (Qiagen) (section 2.2.6.3).

To generate the expression clones, entry clones (pDONR®207.hMPV N construct), pEPI-

DESTC, LR ClonaseTM

, and TE buffer (pH 8.0) were incubated at 25o C overnight, then

transformed into ElectroMAXTM

DH10BTM

competent E. coli (2.2.12.2) and plasmid DNA

isolated using the Miniprep or Midiprep kit (Qiagen) (section 2.2.6.3).

Selection for the growth of entry clone- or expression clone-containing bacteria was done by

plating bacterial suspension on antibiotic-supplemented LB agar, with gentamycin for

selection of entry clone and kanamycin for selection of expression clone.

5.2.1.6 Transformation

1 – 10 µl of BP or LR reaction was mixed with 50 µl of ElectroMAXTM

DH10BTM

competent

E. coli cell suspension, inserted into a pre-chilled 1 ml cuvette, and transformed into the

bacterial cells via electroporation using 25 µF capacitance, 200 Ω resistance, and 2.0 kV

voltage. 450 μl of LB medium was added, then the transformed bacteria incubated at 37o C

and 220 rpm for 1 hour. 100 – 150 µl of the bacterial suspension was plated on an antibiotic-

supplemented LB agar plate, and cultured overnight at 37o C.

5.2.2 Transient transfection into Cos-7 cells

5.2.2.1 Plasmid preparation

The expression clones (pEPI.DESTC - hMPV N constructs) were firstly selected in LB agar

by the lethal marker ccdB gene product and antibiotic selection, gentamycin for entry clone

and kanamycin for expression clone, then plasmid DNA isolated as described in section

2.2.6.3. Plasmids were isolated using Miniprep Kit (Qiagen), or Midiprep Kit (Qiagen) if a

larger amount of DNA was required.

For Miniprep plasmid isolation, selected colonies from antibiotic-supplemented LB agar were

re-cultured in 5 ml of antibiotic-supplemented LB medium (37o C, 220 rpm, 24 h). Bacterial

Page 115: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

102

cells were pelleted through centrifugation at 4,100x g and 4o C for 20 minutes, and

resuspended in 250 µl of buffer PI. 250 µl of buffer P2 was added, followed by 350 µl of

buffer N3. The solution was centrifuged at 16,500x g for 10 minutes and supernatant loaded

onto a QIAprep® spin column. The column was centrifuged at 16,500x g, washed with 0.5 ml

of buffer PB via centrifugation at 16,500x g, and washed again with 0.75 ml of buffer PE and

centrifugation at 16,500x g. An additional centrifugation was carried out to remove the

residual wash buffers, then the plasmid eluted with 30 µl of RNase-free water.

For Midiprep plasmid isolation, a starter culture was prepared by growing the selected

bacterial colonies in 5 ml of antibiotic-supplemented LB medium at 37o C, 300 rpm, for 8

hours. 50 μl of the starter culture was cultured in 25 ml of antibiotic-supplemented LB at 37o

C, 300 rpm, overnight. Bacterial cells were pelleted by centrifugation at 6000x g and 4o C for

15 minutes, then resuspended in 4 ml of buffer P1. 4 ml of buffer P2 was added, the solution

incubated at room temperature for 5 minutes, then 4 ml of buffer P3 added, and solution

incubated for 15 minutes on ice. The tube was centrifuged at 21,800x g and 4o C for 30

minutes; then supernatant transferred into a new 15-ml conical tube and re-centrifuged at

21,800x g and 4o C for 15 minutes. Subsequently, the supernatant was applied to a QIAGEN-

tip 100, which was washed twice with 10 ml of buffer QC. Plasmid was then eluted in 5 ml of

buffer QF, precipitated using 3.5 ml isopropanol, and pelleted by centrifugation at 18,000x g

and 4o C for 18 minutes. The DNA was washed with 2 ml of 70% room-temperature ethanol

via centrifugation at 18,000x g for 10 minutes. The DNA was air dried for 10 minutes and

redissolved in 500 μl of RNase-free water.

The concentration of plasmid was determined using NanoDropTM

1000 UV/Vis

spectrophotometer (section 2.2.8). Briefly, 2 μl of DNA sample was loaded on the pedestal

and DNA concentration was measured by absorbance at 260 nm (A260), A280, and A230. The

ratios of absorbance at A260/A280 and A260/A230 determine the purity of DNA. The DNA

concentration was presented in ng/µl.

5.2.2.2 Transfection

The transient transfection of Cos-7 cells was performed using cationic-lipid (liposome)

platform of Lipofectamine 2000 (Invitrogen), as described in detail in section 2.2.11.1.

Briefly, 3 X 105 of Cos-7 cells were seeded on 24-mm glass coverslips placed in a 6-well

plate, then incubated at 37o C, with 5% CO2 supplementation, for a maximum of 24 hours. 2

Page 116: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

103

µg of expression clones containing GFP alone or GFP–tagged hMPV N construct was

prepared in serum-free DMEM and mixed in 1:1 (µg:ml) ratio with LipofectamineTM

2000

Transfection Reagent diluted in serum-free DMEM (1:25), and incubated for 20 minutes at

room temperature. The transfection mix was dropped onto the monolayer, incubated at 37o C,

5% CO2 for 20 minutes, then 2 ml of DMEM + 10% FBS was added. The transfected cells

were maintained at 37o C, 5% CO2 and cells observed at 24 and 48 hours post tranfection.

5.2.3 Confocal laser scanning microscopy (CLSM)

5.2.3.1 Image analysis

The detail steps of cell imaging using CLSM are presented in section 2.2.11.2. The coverslip

was placed on a ring-shaped coverslip holder, covered with 37o C PBS. A drop of immersion

oil was placed on top of the objective of Nikon Eclipse T to layer between the lens and

coverslip, and cells were directly observed.

Cell images were captured and recorded in a JPEG2000 format using the NIS-Elements 64 bit

3.21.00 software. Digital images were analysed with ImageJ 1.45M software (Ferreira &

Rasband, 2011). The pixel density of fluorescence in the nucleus (Fn) and in the cytoplasm

(Fc) were measured and the ratio of nuclear and cytoplasmic fluorescence was calculated

based on the formula Fn/c =

, where Fn is the pixel density of nuclear fluorescence, Fc

is the pixel density of cytoplasmic fluorescence, and Fb is the pixel density of background

fluorescence.

5.2.3.2 Statistical analysis

The mean value of Fn/Fc ratio for each construct was determined from total cells observed

(≥ 10), and presented in a bar chart, with variability expressed as the standard error of the

mean (SEM). The value of 1.0 (in live cells) and 1.4 (in fixed cells) was used to draw a line,

where Fn/Fc greater than 1.0 or 1.4 denotes nuclear localisation and Fn/Fc less than 1.0 or 1.4

denotes cytoplasmic localisation. The statistical difference of Fn/Fc ratio between each

hMPV N construct was assessed using the nonparametric Mann-Whitney U test (2-tailed)

(GraphPad (Prism) v.5 software (GraphPad, La Jolla, USA)).

Page 117: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

104

5.3 Results

5.3.1 The generation of hMPV N constructs

Four constructs were generated by RT-PCR from total RNA isolated from hMPV-infected

Vero E6 cells as template and the specific attB-tagged primer pairs (Figure 5-1). The four

constructs were the full-length hMPV N, fragment A (hMPV N(1-250)) that contains both the

NES and NLS motifs, fragment B (hMPV N(1-199)) that contains the NES motif and part of

the NLS motif, and fragment C (hMPV N(192-394)) that lacks the NES and contains part of

the NLS motif (Figure 5-1). The constructs were confirmed as correct by PCR (Figure 5-2)

and sequencing (Appendix).

Page 118: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

105

Figure 5-1 Schematic representation of hMPV N constructs generated in this study. The

constructs were designed to accommodate independent investigation of nuclear transport. PCR

was conducted as described in section 2.2.5, using cDNA of total RNA from hMPV-infected

Vero E6 cells as template and specific pair of primers for each construct. NES stands for nuclear

export signal. NLS stands for nuclear localisation signal. Sequence of the putative NES and

NLS is shown with residues in red denoting essential residues within the motif. Right arrow

indicates forward direction of primer. Left arrow indicates reverse direction of primer. Full

length hMPV N (residue 1 – 394) contains both NES and NLS. Fragment (fr) A hMPV N

(residue 1 – 250) contains both NES and NLS, but without the carboxy end of the protein.

Fragment (fr) B hMPV N (residue 1 – 199) contains intact NES, but partial NLS. Fragment (fr)

C (residue 192 – 394) contains partial NLS. attB1 and attB2 sequences attached to the forward

and reverse primers specify the attB recombination sites that are complementary to the attP

recombination sites in the entry vector to facilitate the recombinational cloning.

Page 119: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

106

Figure 5-2 Human metapneumovirus N (hMPV N) gene constructs and vectors used

to investigate the subcellular localisation of the constructs. A. Lane 1 - HyperLadderTM

DNA molecular weight marker, lane 2 – full-length hMPV N, lane 3 – pDONR® 207

(empty entry vector), lane 4 – pEPI.DESTC (empty expression vector). hMPV N gene

was obtained from reverse transcription polymerase chain reaction (RT-PCR) of total

RNA isolated from hMPV-infected Vero E6 cells and then gel-purified as described in

section 2.2.5. pDONR®207 and pEPI-DESTC were isolated from re-culturing the

glycerol stock as described in section 2.2.6.3. B. Lane 1 - HyperLadderTM

DNA

molecular weight marker, lane 2 – fragment A (hMPV N(1-250)), lane 3 – fragment B

(hMPV N(1-199)), lane 4 – fragment C (hMPV N(192-394)). The truncated constructs

of hMPV N gene were generated using PCR with cDNA as template, together with

appropriate primer pairs (table 2-15, figure 5-1), and then gel purified as described in

section 2.2.5. 1 μl of DNA was loaded on to 1% agarose gel stained with 0.01%

GelRedTM

and electrophoresed at 80 volts for 90 minutes. Numbers on the left indicate

molecular weight in base pairs (bp). The length of each PCR product is indicated on

the right.

Page 120: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

107

5.4 The GatewayTM

recombination cloning (RC) technology system

5.4.1 BP recombination reaction

The success of BP recombination was assessed to identify the correct transfer of hMPV N

constructs into pDONR®207

using PCR and DNA sequencing methods (section 2.2.7).

The confirmation of the hMPV N constructs recombination into the entry vector

pDONR®207 using PCR is presented in Figure 5-3. The size of DNA (hMPV N constructs)

identified by PCR was as follows: full length hMPV N (Figure 5-3 A, Lane 1) 1185 bp,

fragment A (Figure 5-3 B, Lane 2) 770 bp, fragment B (Figure 5-3 C, lane 2) 596 bp, and

fragment C (Figure 5-3 D, Lane 2) 609 bp. The size of PCR products is as expected and

confirms the correctness of the insert.

Uncut plasmid containing each of the hMPV N construct was also run on an agarose gel and

the size was as follows: pDONR®207.full length hMPV N (Figure 5-3 A, Lane 2) 4553 bp,

pDONR®207.fragment A (Figure 5-3 B, Lane 1) 4110 bp, pDONR

®207.fragment B (Figure

5-3 C, lane 1) 3964 bp, and pDONR®207.fragment C (Figure 5-3 D, Lane 1) 3977 bp. The

size of entry clones shown in the agarose gel may not in accordance with their predicted size

because a plasmid can move in an agarose gel in the form of coiled DNA that does not allow

for the preview of the actual base-pair (bp) measurement.

The presence of the hMPV N constructs inserted into the pDONR®207 was also confirmed

using DNA sequencing according to a method described in section 2.2.7.2, and as shown in

Appendix. In the hMPV N constructs, there are some nucleotide changes compared to the N

gene of Can97-83. The similar changes occurred in all constructs generated in this study, but

do not change the amino acid sequence. Therefore, the presence of each of hMPV N

construct in the entry clone was confirmed.

Page 121: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

108

Fig

ure

5-3

Scr

eenin

g f

or

E.

coli

co

lonie

s co

nta

inin

g t

he

pD

ON

R®207 –

hM

PV

N c

onst

ruct

s u

sing

PC

R a

s d

escr

ibed

in s

ect

ion

2.2

.7.

Pla

smid

was

extr

acte

d f

rom

ind

ivid

ual

colo

nie

s an

d u

sed a

s te

mp

late

fo

r P

CR

usi

ng t

he

pair

s o

f pri

mer

use

d t

o g

ener

ate

each

const

ruct

. 1 µ

l o

f p

lasm

id D

NA

or

5 µ

l o

f P

CR

pro

duct

was

lo

aded

on t

o t

he

well

s o

f 1%

agar

ose

gel st

ained

wit

h 0

.01%

GelR

edT

M a

nd e

lect

rophore

sis

per

form

ed u

sing 8

0V

ele

ctri

c vo

ltag

e. I

mag

e w

as t

aken

wit

h U

V i

llu

min

atio

n.

L –

Hyp

erL

adder

TM

DN

A m

ole

cula

r w

eig

ht

mar

ker

. (A

) L

ane

1 –

co

nfi

rmat

ion o

f hM

PV

N i

n p

DO

NR

®2

07

.hM

PV

N v

ia P

CR

(si

ze 1

18

5 b

p),

Lane

2 –

uncu

t pD

ON

R®20

7.h

MP

V N

. (B

) L

ane

1 –

uncu

t pD

ON

R207.f

rA,

Lan

e 2 –

co

nfi

rmat

ion o

f fr

agm

ent

A (

hM

PV

N (

1-2

50))

in

pD

ON

R®207.f

rA v

ia P

CR

(si

ze 7

70 b

p).

(C

) L

ane

1 –

uncu

t pD

ON

R®20

7.f

rB,

Lan

e 2 –

co

nfi

rmat

ion o

f fr

agm

ent

B (

hM

PV

N (

1-

199))

in p

DO

NR

®207.f

rB v

ia P

CR

(si

ze 5

96 b

p).

(D

) L

ane

1 –

uncu

t pD

ON

R®207.f

rC,

Lane

2 –

co

nfi

rmat

ion o

f fr

agm

ent

C

(hM

PV

N (

200

-394))

in p

DO

NR

®207.f

rC v

ia P

CR

(si

ze 6

09 b

p).

Page 122: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

109

5.4.2 LR recombination reaction

The success of LR recombination was assessed via PCR and DNA sequencing to identify the

correct transfer of hMPV N constructs into pEPI-DESTC using PCR and DNA sequencing

methods (section 2.2.7).

The confirmation of hMPV N constructs recombination into the expression vector pEPI-

DESTC using PCR is presented in Figures 5-4. The size of DNA (hMPV N constructs)

identified by PCR was as follows: full length hMPV N (Figure 5-4 A, Lane 2) 1185 bp,

fragment A (Figure 5-4 B, Lane 2) 770 bp, fragment B (Figure 5-4 C, lane 2) 596 bp, and

fragment C (Figure 5-4 D, Lane 2) 609 bp. The size of PCR products is as expected and

confirms the correctness of the insert.

Uncut plasmid containing each of the hMPV N construct was also run on an agarose gel and

the size was as follows: pEPI-DESTC.full length hMPV N (Figure 5-4 A, Lane 1) 7945 bp,

pEPI-DESTC.fragment A (Figure 5-4 B, Lane 1) 7530 bp, pEPI-DESTC.fragment B (Figure

5-4 C, lane 1) 7415 bp, and pEPI-DESTC.fragment C (Figure 5-4 D, Lane 1) 7369 bp. The

size of entry clones shown in the agarose gel may not in accordance with their predicted size

because a plasmid can move in an agarose gel in the form of coiled DNA that does not allow

for the preview of the actual base-pair (bp) measurement.

The presence of the hMPV N constructs inserted into the pEPI-DESTC was also confirmed

using DNA sequencing according to a method described in section 2.2.7.2, and as shown in

Appendix. In the hMPV N constructs, there are some nucleotide changes compared to the N

gene of Can97-83. The similar changes occurred in all constructs generated in this study, but

do not change the amino acid sequence. Therefore, the presence of each of hMPV N

construct in the expression clone was confirmed.

Page 123: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

110

Fig

ure

5-4

Scr

eenin

g f

or

E.

coli

co

lonie

s co

nta

inin

g t

he

pE

PI-

DE

ST

C –

hM

PV

N c

onst

ruct

s usi

ng P

CR

as

des

crib

ed i

n

sect

ion 2

.2.7

. P

lasm

id w

as e

xtr

acte

d f

rom

ind

ivid

ual co

lonie

s and u

sed a

s te

mp

late

fo

r P

CR

usi

ng t

he p

air

s o

f pri

mer

use

d

to g

ener

ate

each

co

nst

ruct

. 1 µ

l o

f p

lasm

id D

NA

or

5 µ

l o

f P

CR

pro

duct

was

lo

aded

on t

o t

he

well

s o

f 1%

agar

ose

gel

stai

ned

wit

h 0

.01%

Gel

Red

TM

and e

lect

rophore

sis

per

form

ed u

sing 8

0V

ele

ctri

c vo

ltag

e. I

mag

e w

as t

aken

wit

h U

V

illu

min

atio

n.

L –

Hyper

Lad

der

TM

DN

A m

ole

cula

r w

eig

ht

mar

ker

. (A

) L

ane

1 –

uncu

t pE

PI-

DE

ST

C.h

MP

V N

, L

ane

2 –

confi

rmat

ion o

f hM

PV

N i

n p

EP

I-D

ES

TC

.hM

PV

N v

ia P

CR

(si

ze

1185

bp).

(B

) L

ane

1 –

uncu

t pE

PI-

DE

ST

C.f

rA,

Lan

e 2

– c

onfi

rmat

ion o

f fr

agm

ent

A (

hM

PV

N (

1-2

50))

in p

EP

I-D

ES

TC

.frA

via

PC

R (

size

770 b

p).

(C

) L

ane

1 –

uncu

t pE

PI-

DE

ST

C.f

rB,

Lan

e 2 –

co

nfi

rmat

ion o

f fr

agm

ent

B (

hM

PV

N (

1-1

99))

in p

EP

I-D

ES

TC

.frB

via

PC

R (

size

596 b

p).

(D

)

Lane

1 –

uncu

t pE

PI-

DE

ST

C.f

rC,

Lane

2 –

co

nfi

rmat

ion o

f fr

agm

ent

C (

hM

PV

N (

192

-394))

in p

EP

I-D

ES

TC

.frC

via

PC

R (

size

609 b

p).

Page 124: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

111

5.5 Expression of hMPV N constructs in transfection cells

5.5.1 Expression of full-length hMPV N (1-394) observed in live Cos-7 cells

Subcellular localisation of GFP-tagged full-length hMPV N was studied using CLSM, with

GFP alone used as control. GFP was shown to diffuse easily across the nuclear envelope and

localise equally in the cytoplasm and the nucleus of transfected cells (Figure 5-5), showing

the transfection was efficient.

GFP-tagged full-length hMPV N was present in both the nucleus and cytoplasm of

transfected Cos-7 cells at 24 hours post transfection (Figure 5-5 A, right image), as

determined by green fluorescence in live cells that is similar to the distribution of GFP alone

(Figure 5-5 A, left image). There was no change in the localisation of either GFP alone, or

GFP-tagged full-length hMPV N, at 48 hours post transfection (Figure 5-5 A, lower images),

with both equally present in the nucleus and cytoplasm.

The fluorescence intensity within the nucleus (Fn) and cytoplasm (Fc) of Cos-7 cells

transfected with pEPI.DESTC-GFP and pEPI.DESTC-hMPV N was measured to generate the

nuclear to cytoplasmic ratio (Fn/Fc) (Figure 5-5 B), as detailed in section 2.2.11.3. GFP

accumulated equivalently in the cytoplasm and in the nucleus at both 24 (Fn/Fc = 1.21) and

48 hours post infection (Fn/Fc = 1.03), as did GFP-tagged hMPV N (Fn/Fc 24H = 1.07,

Fn/Fc 48H = 1.04). The values of Fn/Fc ratio support the cytoplasmic vs nuclear

accumulation observed from fluorescence in cell images (Figure 5-5 A). Based on this

finding, it can be inferred that the hMPV N protein localised in both the cytoplasm and in the

nucleus of transfected cells in the period of 48 hours after transfection, and the equal

distribution of hMPV N in the cytoplasm and in the nucleus was stable over the time of the

experiment.

Page 125: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

112

Figure 5-5 Subcellular localisation of GFP-tagged full-length hMPV N (1-394) protein observed in

Cos-7 cells. Cos-7 cells were transfected with an expression clone containing full-length hMPV N

gene, as described in section 2.2.11.1 and images were taken from live cells 24 and 48 hours post

transfection using confocal laser scanning microscopy (CLSM) as described in section 2.2.11.2. A

Subcellular localisation of GFP and GFP-tagged hMPV N observed in live Cos-7 cells 24 and 48

hours post infection. B Quantitative analysis of CLSM imaging is presented as the ratio between

nuclear and cytoplasmic concentration (Fn/Fc) of hMPV N, which were determined from pixel

density in the image and the means of the ratio ± SEM presented.

Page 126: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

113

5.5.2 Expression of hMPV N deleted constructs observed in Cos-7 cells

Cos-7 cells transfected with GFP alone or GFP fused to full-length hMPV N or its truncated

constructs, were observed using CLSM at 24 hours after transfection (Figure 5-6 A). Full

length hMPV N (1-394) localised in a similar manner as GFP in transfected cells, which is

equally localised in both the cytoplasm and in the nucleus, 24 hours after transfection. This

similar localisation was supported by the mean value of Fn/Fc, which are 1.110 (GFP) and

1.109 (hMPV N) (Figure 5-6 B), and the statistical analysis (nonparametric Mann-Whitney

U-test) that showed no statistical difference (P = 5.194; Figure 5-6 C).

Fragment A (hMPV N (1-250)), which contains both NLS and NES, was distributed in both

the nucleus and cytoplasm (Figure 5-6 A). This observation was also supported by Fn/Fc

ratio average of 1.018 (Figure 5-6 B), which resulted in no statistically different localisation

compared to full-length hMPV N (P = 0.2076; Figure 5-7 C). In other words, fragment A was

distributed similarly as hMPV N.

Fragment B (hMPV N (1-199)), which contains intact NES and partial NLS, presented with

more cytoplasmic localisation compared to the other hMPV N constructs and GFP (Figure 5-

6 A). The determination of Fn/Fc ratio also supports the cell image analysis that fragment B

localised predominantly in the cytoplasm 24 hours after transfection, with the mean of Fn/Fc

values 0.7008 (Figure 5-6 B). The cytoplasmic localisation of fragment B was significantly

different to the nucleocytoplasmic distribution of full length hMPV N (P < 0.0004; Figure 5-6

C).

The localisation of fragment C (hMPV N (192-394)) containing C-terminal domain (CTD)

NLS resembled that of the full length hMPV N and fragment A (hMPV N (1-250)), which

was equally distribution in both the cytoplasm and the nucleus (Figure 5-6 A). This

observation was supported by a mean value of Fn/Fc = 0.1057 (Figure 5-6 B). However, a

statistical difference to full length hMPV N was observed (P = 0.0249).

Page 127: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

114

Fig

ure

5-6

Subce

llu

lar

loca

lisa

tio

n o

f G

FP

-tag

ged

hM

PV

N c

onst

ruct

s in

liv

e C

os-

7 c

ell

s. (

A)

Co

s-7 c

ell

s

wer

e tr

ansf

ecte

d w

ith e

xpre

ssio

n c

lone

conta

inin

g t

runca

ted a

nd f

ull

-length

hM

PV

N g

ene,

as

des

crib

ed i

n

sect

ion 2

.2.1

1.1

and i

mag

es f

rom

liv

e ce

lls

wer

e ta

ken

24 h

ours

po

st t

ransf

ecti

on a

s des

crib

ed i

n s

ecti

on

2.2

.11.2

. (B

) Q

uan

tita

tive

analy

sis

of

CL

SM

im

ag

ing i

s pre

sente

d a

s th

e ra

tio

bet

wee

n n

ucle

ar a

nd

cy

top

lasm

ic

conce

ntr

atio

n (

Fn/F

c) o

f G

FP

-tag

ged

hM

PV

N c

onst

ruct

s, w

hic

h w

ere

det

erm

ined

fro

m p

ixel densi

ty i

n t

he

imag

e and t

he

mea

n o

f th

e ra

tio

± S

EM

pre

sente

d. (C

) T

he

Fn/F

c ra

tio

valu

es o

f ea

ch h

MP

V N

co

nst

ruct

wer

e

com

par

ed a

gain

st G

FP

and f

ull

-length

hM

PV

N u

sing n

onp

aram

etri

c M

ann-W

hit

ney U

Tes

t (G

raphP

ad P

rism

,

La

Joll

a, U

SA

), (

*)

den

ote

s P

<0.5

; (*

*)

den

ote

s P

<0.0

05, (*

**)

den

ote

s P

<0.0

00

1.

Page 128: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

115

5.6 Discussion

The N protein of RNA viruses that replicate in the cytoplasm has been shown to localise into

the nucleus during the course of infection, with the facilitation of NLS and NES. The

nuclear/nucleolar localisation of N protein has been well-characterised in the Coronaviridae

(SARS, borna disease virus, transmissible gastroenteritis virus, mouse hepatitis virus),

Arteriviridae (equine arteritis virus, porcine reproductive and respiratory viruses),

Bornaviridae (borna disease virus), and Paramyxoviridae (measles virus) families (Rowland

et al., 1999, Hiscox et al., 2001, Kobayashi et al., 2001, Wurm et al., 2001, Tijms et al., 2002,

Calvo et al., 2005, Surjit et al., 2005, You et al., 2005, Lee et al., 2006, Sato et al., 2006,

Takayama et al., 2012). The precise role of nuclear/nucleolar translocation of these N

proteins in virus replication has not been defined, but the transient localisation tends to

associate with processes that give advantage for virus replication. For example, the

Coronavirus N associates with nucleolar proteins including fibrillarin and nucleolin to hold

the cell cycle in interphase, during which the maximum translation of viral mRNA can occur

(Wurm et al., 2001, Chen et al., 2002). N association with fibrillarin has also been

documented in arteriviruses and inhibition of nuclear localisation of the N protein results in

attenuated virus replication (Yoo et al., 2003, Lee et al., 2006). In Paramyxoviridae (measles

virus), the nuclear localisation of N has been shown to block host interferon responses by

inhibiting the nuclear translocation of STATs (Takayama et al., 2012). These studies suggest

that disruption of the nucleocytoplasmic transport of N protein offers a way for attenuation.

Little is known about the nuclear localisation of hMPV N protein, but this study has

identified that there are putative NLS (185

RRANRVLSDALKRYPR200

) and NES

(1MSLQGIHLSDLSYKH

15) motifs in the N protein. To determine whether the motifs are

functional and can facilitate the nucleocytoplasmic localisation of hMPV N, GFP-tagged

hMPV N constructs were synthesised and the nucleocytoplasmic localisation observed in

transfected cells. Fusing a coding sequence to a fluorescent gene marker, followed by protein

expression in transfected cells, is a standard approach to characterise previously unknown

nucleocytoplasmic properties of a protein (Tijms et al., 2002, Rowland and Yoo, 2003, Surjit

et al., 2005, You et al., 2005, Sato et al., 2006).

The results presented in this chapter indicates that GFP-tagged full length hMPV N

containing NES and NLS, GFP-tagged fragment A containing NES and a potential nuclear

Page 129: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

116

localisation motif of NTD, and GFP-tagged fragment C containing the a potential nuclear

localisation motif of CTD localised in both the nucleus and cytoplasm at 24 hours after

transfection. In contrast, the GFP-tagged fragment B, containing intact NES and a potential

nuclear localisation motif of NTD, localised to the cytoplasm. Two indications can be

inferred based on this result. Firstly, the nuclear localisation motif of fragment B is functional

in facilitating nuclear import, considering that the nucleus presented a green fluorescence,

albeit dimmer than the cytoplasm (Figure 5-6 A). However, the rate of nuclear export

facilitated by the intact NES might be higher than the rate of nuclear import facilitated by the

disrupted nuclear localisation motif for hMPV N (1-199), such that it was more cytoplasmic

than nuclear. Secondly, hMPV N contains a functional NLS, based on the fact that full-length

hMPV N and fragment A constructs, having both the full-length putative NES (1-15) and

potential nuclear localisation motif of NTD (185-200) localised evenly in the cytoplasm and

the nucleus. This suggests that R200

has a determining role in facilitating the nuclear

localisation of hMPV N, and that the nuclear localisation activity might be contained in

residues 185 – 250. The highly cytoplasmic concentration of fragment B might be caused by

a dysfunctional nuclear localisation motif, most likely due to the lack of key residue R200

.

The size of the GFP-tagged constructs is an important factor that needs to be considered in

assessing the subcellular distribution of a construct in transfected cells. Proteins less than 50

kDa have been shown to passively diffuse across the NPC, causing even distribution in the

cytoplasm and in the nucleus; while proteins larger than 50 kDa require an active nuclear

transport system to facilitate the nucleocytoplasmic transport; causing cytoplasmic

localisation for inherently cytoplasmic proteins and nuclear concentration for the inherently

nuclear proteins (Terry et al., 2007). GFP has a molecular weight of 27 kDa; therefore it is

able to passively diffuse across the NPC, which results in equal distribution between the

cytoplasm and the nucleus (Chalfie et al., 1994, Macara, 2001). hMPV N is 43.5 kDa, which

may be small enough for a protein to passively diffuse but is most likely actively transported

across the NPC (Petraityte-Burneikiene et al., 2011). However, GFP-tagged hMPV N is 70.5

kDa, which is too big for passive diffusion and requires active transport to pass through the

NPC. Therefore, the equal distribution of hMPV N in the cytoplasm and in the nucleus is

most likely the result of active transport, which represents the ability of hMPV N to shuttle

between the nucleus and the cytoplasm.

Page 130: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

117

GFP-tagged fragment A is approximately 57 kDa, while GFP-tagged fragment B and C are

approximately 51 kDa each. Therefore, the localisation of these hMPV N constructs is also

likely the result of nucleocytoplasmic transport.

This study suggests that there is an active NES in hMPV N coding sequence and a region

containing NLS motifs; and therefore confirmed that the hMPV N possesses inherent ability

for nucleocytoplasmic transport. The N protein is a structural protein that has various core

functions in viral replication, most notably in genome protection and packaging, ensuring that

only functional genome is inherited by infectious progeny (Narayanan et al., 2000). The

nucleocytoplasmic transport of hMPV N probably has a determining function in virus

replication, as already described in porcine reproductive and respiratory syndrome virus (Lee

et al., 2006). In that case, attempts to disrupt the movement can be useful for attenuation,

which is one known method to develop vaccines or antivirals. As such, by describing that

hMPV N possesses inherent nucleocytoplasmic transport ability; this study has opened a new

research avenue for future investigation on the development of vaccines/antivirals against

hMPV.

Page 131: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

118

Page 132: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

119

CHAPTER 6

GENERAL DISCUSSION

6 General Discussion

Page 133: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

120

Page 134: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

121

RSV and hMPV are the main cause of bronchiolitis and pneumonia in infants/ very young

children and the elderly. Being dispersed by aerosols, RSV and hMPV are easily transmitted.

This poses the risk of spreading the virus to a large proportion of the population in a short

time. Attempts to lessen the burden of RSV and hMPV are therefore required.

Options include the development of prophylaxis to ameliorate the severity of clinical

symptoms, antivirals to control infection, and vaccines to provide the human body with the

tools to fight viral infection such that the infection does not develop and virus spread can be

controlled. Of the three mechanisms, a vaccine is considered the best option for limiting the

risks of lower respiratory tract infections posed by RSV and hMPV because vaccination can

be applied to all members of the human population. This is important considering that,

although the main target of RSV and hMPV are the very young and elderly, people from

other age groups can become the reservoir of the virus once infected, spreading the virus to

the population in the high-risk age group.

To develop an effective vaccine, it is important to understand the molecular biology of viral

replication because disrupting the biological processes of viral replication can facilitate virus

attenuation for the purpose of vaccine development. Viruses that replicate in the cytoplasm

have been shown to have structural proteins transiently translocating into the nucleus to play

a non-structural function that optimise viral replication; therefore, can be used as a target of

attenuation.

To attain attenuated virus by disrupting the nucleocytoplasmic transport of the structural

proteins involves four major steps. Firstly, the nucleocytoplasmic shuttling of a structural

protein in infected and transfected cells needs to be identified. Secondly, the nuclear transport

motifs need to be defined by expressing the protein in cell culture. Thirdly, to gather evidence

on whether the nucleocytoplasmic transport of the structural protein has a significant function

in virus replication, the key residues of the nuclear transport motifs need to be mutated and

the impact of the mutation on virus production analysed in vitro and in vivo. The final stage is

to apply the knowledge for the production of live-attenuated virus vaccine or antivirals.

6.1 Respiratory syncytial virus

Attempts to develop a vaccine for RSV have been hindered by problems such as the

difficulties in culturing the virus in cell lines (Collins and Crowe Jr., 2007), to allow further

Page 135: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

122

study, and the partial immunity developed against the formalin-inactivated virus that turned

out to cause deadly clinical outcomes in infants (Dudas and Karron, 1998). After years of

study, live-attenuated virus has been shown to be the best option for an RSV vaccine because

of the capability to induce immune responses similar to the natural infection (Collins, 2011).

However, attenuating RSV for vaccine purpose is a challenge itself, particularly in terms of

balancing the immunogenicity and the safety (not causing severe disease). Methods to

attenuate RSV include multiple passaging to obtain mutants that are temperature sensitive (ts)

and host specific. However, multiple passaging is a costly and time-consuming method to

generate attenuated RSV. The phenotypes of the mutants are not predictable, and subsequent

in vitro and in vivo characterisation might result in phenotypes that are not suitable for

vaccine production (Karron et al., 1997b). To overcome the obstacles in the development of

RSV vaccine, attempts of attenuation are directed towards disrupting biological processes in

virus replication (Collins et al., 1995), with the nucleocytoplasmic transport of RSV M being

one potential target of attenuation.

The nucleocytoplasmic transport mechanism of RSV M is well characterised. The RSV M

translocates to the nucleus early in infection and localises in the cytoplasm later (Ghildyal et

al., 2002, Ghildyal et al., 2003, Ghildyal et al., 2005b). The nuclear localisation of RSV M is

associated with binding to the host DNA and the inhibition of host transcription; therefore it

is thought that RSV M nuclear localisation aims to hold host transcriptional activity to allow

the viral replication machinery to function optimally in the cytoplasm (Ghildyal et al., 2003,

Latiff et al., 2004). The cytoplasmic localisation of RSV M in the late stage of infection aims

to facilitate the formation of infectious progeny during assembly and budding processes (Li et

al., 2008). Disrupting nuclear localisation has been shown to decrease the production of

infectious progeny, while disrupting cytoplasmic localisation (nuclear export) has been

shown to cease the infection as no infectious progeny are produced due to the failure of

assembly/budding (Ghildyal et al., 2009). The RSV M nucleocytoplasmic transport has been

shown to be regulated by CK2 phosphorylation, which is thought to promote nuclear export

(Alvisi et al., 2008). RSV M205 (T) was proposed as the possible candidate of CK2

phosphorylation site. Therefore, the site was mutated into alanine (A) to identify whether this

key residue has a role in viral replication.

This study characterises the ability of a recombinant mutant having RSV M205 (A) to

replicate and to induce pro-inflammatory response in vitro. The impact of RSV M205 for

Page 136: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

123

virus replication was studied in single cycle replication and multiple cycle replication assays.

The single cycle replication assay aimed to observe the capability of RSV to infect, while the

multiple cycle replication assay aimed to observe the capability of RSV to transmit the

infection.

In contrast to the starting hypothesis regarding the role of M205 (T) as a putative CK2

phosphorylation site that promotes nuclear export, this study, indirectly, identified that

M(205) T is not the regulatory CK2 phosphorylation site and that the RSV M nuclear export

was not affected by the substitution. This was evidenced by the ability of rRSV A2

M(T205A) to produce a relatively similar titre of infectious particle at the end of single cycle

replication assay (Figure 3-1 A). However, this site does appear to be required to facilitate

viral egress to reinitiate infection in uninfected cells, as shown by the results of the multiple

cycle replication assay (Figure 3-2 A). In doing so, results obtained in this study present a

new finding that the M205 (T) is an important site for the ability of RSV to transmit the

infection in vitro. The M(T205A) substitution restricted viral spread (Figure 3-2 A), leading

to reduced amount of infectious progeny; and therefore, attenuating the virus.

The ability of rRSV A2 M(T205A) to replicate further affected the pro-inflammatory

responses that were produced upon RSV infection. The expression of pro-inflammatory

mediators, IL-8 and RANTES, requires the production of infectious particles (Fiedler et al.,

1996, Saito et al., 1997). As the M(T205A) substitution limited the growth of RSV, the

induction of IL-8 and RANTES production (Chapter 4) was further suppressed.

In addition, this study also suggested that the M protein possesses an important role for

suppressing the antiviral IFN-α/β activity that can restrict viral spread, and that the M205 (T)

might be a key residue in regulating this role. The failure to suppress the antiviral activity of

rRSV A2 M(T205A) was shown by the reduced titres in A549 cells, compared to the rRSV

A2 (Figure 3-1 B). The titre of rRSV A2 M(T205A), however, was relatively similar to the

rRSV A2 in the IFN-α/β-defective Vero E6 cells (Figure 3-1 A); suggesting that M may have

a role in regulation of IFN response in RSV infected cells. This is the first evidence that

suggests the role of RSV M in the induction of the IFN-α/β activation.

The attenuation of RSV presented in this study provides an early characterisation of an

attenuated RSV strain. It is important to repeat the characterisation in a whole organism (in

vivo) to ensure whether the attenuated phenotype of rRSV A2 M(T205A) displayed in cell

Page 137: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

124

culture (in vitro) is stably expressed in vivo, considering that cell culture is an environment

with limited immune capabilities, while an organism has a complex immune network.

Attenuation by generating recombinant virus poses a good potency of a safe and useful

vaccine or antiviral. For example, many forms of recombinant RSV has been generated via

reverse genetics, including rRSV A2 M(T205A) that was studied in this thesis. Questions

about the length of protection it may give cannot be assessed based on the experimental

results presented in this thesis. However, to infer from another experience using recombinant

mutant RSV as a vaccine candidate, a recombinant mutant RSV has been shown to provide

protection up to 147 days against challenge of natural RSV infection (Widjojoatmodjo et al.,

2010). Based on this experience, the potential of rRSV A2 M(T205A) to provide protection

against natural RSV infection is possible to last more than two months (approximately 4.5

months). Given that RSV infections mostly peaks during late spring-winter (4 – months), a

single dose of vaccination could be expected to protect against RSV during the RSV season.

Even if the protection is induced for only one season, RSV vaccination should lead to

significant reduction in morbidity, mortality and socio-economic costs given that more than

10,000 elderly patients die each year from RSV disease.

6.2 Human metapneumovirus

Obstacles of developing an hMPV vaccine include, like RSV, difficulties in growing the

virus in cell culture, to enable further study of the biology of the virus (van-den-Hoogen et

al., 2001, Kuiken et al., 2004). This results in limited availability of knowledge regarding the

development of host immune responses and viral factors of infection that can be targeted as a

basis of vaccine development. As hMPV is from the same family as RSV, i.e

Paramyxoviridae, knowledge of the development of immune response and clinical symptoms

for hMPV can be inferred from knowledge assessed in RSV, including the development of a

live-attenuated vaccine.

In the case of hMPV, the N protein has been shown to localise in the nucleus in the very late

stage of infection (day 5 post infection) (Ghildyal R and Gahan, M; unpublished). The

mechanism of nuclear localisation has never been described; therefore, this study attempts to

gather evidence of the inherent ability of the hMPV N for nucleocytoplasmic transport. As

the first step in characterising the nucleocytoplasmic transport ability, the presence of nuclear

Page 138: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

125

transport motifs in hMPV N was identified using the expression of full-length and truncated

hMPV N constructs in transfected cells.

This study identified the presence of components for active nucleocytoplasmic transport in

hMPV N protein. The NES motif identified in EXPASY (hMPV N 1 – 15:

MSLQGIHLSDLSYKH) was indicated to be functional by the cytoplasmic localisation of

fragment B that contains this motif and lacks the complete sequence of the potential nuclear

localisation motif (Figure 5-6). A region containing the nuclear localisation motif was

manually identified in amino acid 185 – 200: RRANRVLSDALKRYPR. This study

confirmed that an active NLS is contained within this sequence; with the key residues being

the amino acid residue R200

and other important residues might reside in amino acids 185 –

250. This finding was based on the ability of fragment C (hMPV N(192-394)) and fragment

A (hMPV N (1-250)) to be equally distributed in both the nucleus and cytoplasm, while

fragment B (hMPV N (1 – 199)) was more cytoplasmic (Figure 5-6).

The ability of active nucleocytoplasmic transport in hMPV N was confirmed by the size of

GFP-tagged hMPV N constructs, either full-length or truncated, which were larger than the

size threshold of proteins that can diffuse passively across the nuclear pore complex (50 kDa)

(Terry et al., 2007). In addition, the biology of cells used as the transfection host has been

well characterised; therefore, ensuring that the expression of a transfected gene truly

represents the phenotype of a protein without interference from other components (Kingston,

2003). Therefore, it can be concluded that the hMPV N possesses inherent nucleocytoplasmic

transport ability.

Nevertheless, the precise NLS motif, as well as the mechanism of localisation, cannot be

determined based on the result obtained in this study. This prompts further study to define the

precise NLS motif in transfected cells and investigate the subnuclear localisation of hMPV N

in infected cells, as a basis to determine its nuclear functions and suitability as an attenuation

target.

6.3 Conclusion

Study of the nucleocytoplasmic transport of viral structural proteins contributes to the

knowledge of attenuation targets for the purpose of developing a safe live-attenuated vaccine,

which is needed to control the spread of RSV and hMPV worldwide. The nuclear transport of

Page 139: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

126

RSV M protein has been well characterised and recombinant mutants were generated by

targeting this mechanism. This study characterised a recombinant RSV with M(T205A)

substitution in vitro. The results showed that RSV M205 (T) is a site required to transmit

infection, and that RSV M has a role in the suppression of IFN-α/β. On the other hand, the

subcellular localisation of hMPV N has never been assessed and this study identified for the

first time that the protein has inherent nucleocytoplasmic transport ability, therefore

providing a possible means of attenuation. To sum up, the disruption of the

nucleocytoplasmic transport of a structural protein provides a new method of attenuation, and

enables biological processes of a virus to be studied and characterised.

Page 140: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

127

REFERENCES

7 Reference List

Page 141: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

128

Page 142: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

129

AHMED, M. & LYLES, D. S. 1998. Effect of Vesicular Stomatitis Virus Matrix Protein on

Transcription Directed by Host RNA Polymerases I, II, and III. Journal of Virology,

72, 8413-8419.

AHMED, M., MCKENZIE, M. O., PUCKETT, S., HOJNACKI, M., POLIQUIN, L. &

LYLES, D. S. 2003. Ability of the Matrix Protein of Vesicular Stomatitis Virus To

Suppress Beta Interferon Gene Expression Is Genetically Correlated with the

Inhibition of Host RNA and Protein Synthesis. Journal of Virology, 77, 4646-4657.

AL-TOUM, R., BDOUR, S. & AYYASH, H. 2006. Epidemiology and Clinical

Characteristics of Respiratory Syncytial Virus Infections in Jordan. Journal of

Tropical Pediatrics, 52, 282-287.

ALBERTS, B., JOHNSON, A., LEWIS, J., RAFF, M., ROBERTS, K. & WALTERS, P.

2008. Molecular Biology of the Cell, New York, Garland Science.

ALVISI, G., JANS, D. A., GUO, J., PINNA, L. A. & RIPALTI, A. 2005. A Protein Kinase

CK2 Site Flanking the Nuclear Targeting Signal Enhances Nuclear Transport of

Human Cytomegalovirus ppUL44. Traffic, 6, 1002 - 1013.

ALVISI, G., RAWLINSON, S. M., GHILDYAL, R., RIPALTI, A. & JANS, D. A. 2008.

Regulated nucleocytoplasmic trafficking of viral gene product: A therapeutic target?

Biochimica and Biophysica Acta, 1784, 213-227.

ANDERSON, L. J. & HEILMAN, C. A. 1995. Protective and Disease-Enhancing Immune

Responses to Respiratory Syncytial Virus. The Journal of Infectious Diseases, 171, 1

- 7.

ARNOLD, R., HUMBERT, B., WERCHAU, H., GALLATI, H. & KONIG, W. 1994.

Interleukin-8, Interleukin-6, and Soluble Tumour Necrosis Factor Receptor Type I

Release from a Human Pulmonary Epithelial Cell Line (A549) Exposed to

Respiratory Syncytial Virus. Immunology, 82, 126 - 133.

BACHI, T. & HOWE, C. 1973. Morphogenesis and Ultrastructure of Respiratory Syncytial

Virus. Journal of Virology, 12, 1173-1180.

BAGNAUD-BAULE, A., REYNARD, O., PERRET, M., BERLAND, J.-L., MAACHE, M.,

PEYREFITTE, C., VERNET, G., VOLCHKOV, V. & PARANHOS-BACCALA, P.

2011. The Human Metapneumovirus Matrix Protein Stimulates the Inflammatory

Immune Response In Vitro. PLOS ONE, 6, e17818.

BALL, L. A. 2007. Virus Replication Strategies. In: KNIPE, D. M. & HOWLEY, P. M.

(eds.) Virus Replication Strategies. 5th ed. Riverwoods: Wolters Kluwer

Health/Lippincott Williams & Wilkins.

BAO, X., LIU, T., SHAN, Y., LI, K., GAROFALO, R. & CASOLA, A. 2008. Human

Metapneumovirus G Protein Inhibits Innate Immune Responses. PLOS Pathogens, 4,

e1000077.

BARIC, R. S., NELSON, G. W., FLEMING, J. O., DEANS, R. J., KECK, J. G., CASTEEL,

N. & STOHLMAN, S. A. 1988. Interactions Between Coronavirus Nucleocapsid

Protein and Viral RNAs: Implications for Viral Transcription. Journal of Virology,

62, 4280 - 4287.

BARR, J. N. & FEARNS, R. 2010. How RNA Viruses Maintain Their Genome Integrity.

Journal of General Virology, 91, 1373 - 1387.

BASTIEN, N., NORMAND, S., TAYLOR, T., WARD, D., PERET, T. C. T., BOIVIN, G.,

ANDERSON, L. J. & LI, Y. 2003. Sequence Analysis of the N, P, M, and F Genes of

Canadian Human Metapneumovirus Strains. Virus Research, 93, 51 - 62.

BECKER, S., RINNE, C., HOFSÄSS, U., KLENK, H.-D. & MÜHLBERGER, E. 1998.

Interactions of Marburg Virus Nucleocapsid Proteins. Virology, 249, 406 - 417.

Page 143: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

130

BERMINGHAM, A. & COLLINS, P. L. 1999. The M2-2 Protein of Human Respiratory

Syncytial Virus Is a Regulatory Factor Involved in the Balance between RNA

Replication and Transcription. Proceedings of the National Academy of Sciences

(PNAS), 96, 11259 - 11264.

BIACCHESI, S., SKIADOPOULOS, M. H., YANG, L., LAMIRANDE, E. W., TRAN, K.

C., MURPHY, B. R., COLLINS, P. L. & BUCHHOLZ, U. J. 2004. Recombinant

Human Metapneumovirus Lacking the Small Hydrophobic SH and/or Attachment G

Glycoprotein: Deletion of G Yields a Promising Vaccine Candidate. Journal of

Virology, 78, 12877 - 12887.

BOIVIN, G., ABED, Y., PELLETIER, G., RUEL, L., MOISAN, D., COTÉ, S., PERET, T.

C. T., ERDMAN, D. D. & ANDERSON, L. J. 2002. Virological Features and Clinical

Manifestations Associated with Human Metapneumovirus: A New Paramyxovirus

Responsible for Acute Respiratory-Tract Infections in All Age Groups. The Journal of

Infectious Diseases, 186, 1330 - 1334.

BONIZZI, G. & KARIN, M. 2004. The Two NF-κB Activation Pathways and Their Role in

Innate and Adaptive Immunity. Trends in Immunology, 25, 280 - 288.

BOSE, S. & BANERJEE, A. K. 2003. Innate Immune Response Against Nonsegmented

Negative Strand RNA Viruses. Journal of Interferon & Cytokine Research, 23, 401 -

412.

BOSIS, S., ESPOSITO, S., OSTERHAUS, A. D. M. E., TREMOLATI, E., BEGLIATTI, E.,

TAGLIABUE, C., CORTI, F., PRINCIPI, N. & NIESTERS, H. G. M. 2008.

Association between High Nasopharyngeal Viral Load and Disease Severity in

Children with Human Metapneumovirus Infection. Journal of Clinical Virology, 42,

286 - 290.

BRONDYK, W. H. 2009. Selecting an Appropriate Method for Expressing a Recombinant

Protein. Methods in Enzymology. Elsevier, Inc.

BROOKS, G. F., CARROLL, K. C., BUTEL, J. S. & MORSE, S. A. 2007. Jawetz, Melnick

& Adelberg's Medical Microbiology, New York, The McGraw-Hill Companies, Inc.

BROOR, S., BHARAJ, P. & CHAHAR, H. S. 2008. Human Metapneumovirus: A New

Respiratory Pathogen. Journal of Biosciences, 33, 483 - 493.

BRZÓZKA, K., FINKE, S. & CONZELMANN, K.-K. 2006. Inhibition of Interferon

Signaling by Rabies Virus Phosphoprotein P: Activation-Dependent Binding of

STAT1 and STAT2. Journal of Virology, 80, 2675-2683.

BUCHHOLZ, U. J., BIACCHESI, S., PHAM, Q. N., TRAN, K. C., YANG, L., LUONGO,

C. L., SKIADOPOULOS, M. H., MURPHY, B. R. & COLLINS, P. L. 2005. Deletion

of M2 Gene Open Reading Frames 1 and 2 of Human Metapneumovirus: Effects on

RNA Synthesis, Attenuation, and Immunogenicity. Journal of Virology, 79, 6588 -

6597.

CALAIN, P. & ROUX, L. 1993. The Rule of Six, a Basic Feature for Efficient Replication of

Sendai Virus Defective Interfering RNA. Journal of Virology, 67, 4822 - 4830.

CALVO, E., ESCORS, D., LÓPEZ, J. A., GONZÁLEZ, J. M., ÁLVAREZ, A., ARZA, E. &

ENJUANES, L. 2005. Phosphorylation and Subcellular Localization of Transmissible

Gastroenteritis Virus Nucleocapsid Protein in Infected Cells. Journal of General

Virology, 86, 2255-2267.

CARMO-FONSECA, M., MENDES-SOARES, L. & CAMPOS, I. 2000. To Be or Not To Be

in the Nucleolus. Nature Cell Biology, 2, E107 - E112.

CASOLA, A., BURGER, N., LIU, T., JAMALUDDIN, M., BRASIER, A. R. &

GAROFALO, R. P. 2001a. Oxidant Tone Regulates RANTES Gene Expression in

Page 144: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

131

Airway Epithelial Cells Infected with Respiratory Syncytial Virus. The Journal of

Biological Chemistry, 276, 19715-19722.

CASOLA, A., GAROFALO, R., HAEBERLE, H., ELLIOTT, T. F., LIN, R.,

JAMALUDDIN, M. & BRASIER, A. R. 2001b. Multiple Cis Regulatory Elements

Control RANTES Promoter Activity in Alveolar Epithelial Cells Infected with

Respiratory Syncytial Virus. Journal of Virology, 75, 6428 - 6439.

CASOLA, A., GAROFALO, R. P., JAMALUDDIN, M., VLAHOPOULOS, S. & BRASIER,

A. R. 2000. Requirement of A Novel Upstream Response Element in Respiratory

Syncytial Virus-Induced IL-8 Gene Expression. The Journal of Immunology, 164,

5944-5951.

CENTERS FOR DISEASE CONTROL AND PREVENTION (C.D.C). 2011. Respiratory

Syncytial Virus (RSV) Season, U.S., 2010-2011 [Online]. Atlanta. Available:

http://www.cdc.gov/Features/dsRSV/ [Accessed 04 December 2011].

CHALFIE, M., TU, Y., EUSKIRCHEN, G., WARD, W. W. & PRASHER, D. S. 1994.

Green Fluorescent Protein as a Marker for Gene Expression. Science, 263, 802-805.

CHAZAL, N. & GERLIER, D. 2003. Virus Entry, Assembly, Budding, and Membrane Rafts.

Microbiology and Molecular Biology Reviews, 67, 226 - 237.

CHECON, R. E., SIQUEIRA, M. M., LUGON, A. K., PORTES, S. & DIETZE, R. 2002.

Short Report: Seasonal Pattern of Respiratory Syncytial Virus in A Region with A

Tropical Climate in Southeastern Brazil. The American Journal of Tropical Medicine

and Hygiene, 67, 490-491.

CHEN, H., WURM, T., BRITTON, P., BROOKS, G. & HISCOX, J. A. 2002. Interaction of

the Coronavirus Nucleoprotein with Nucleolar Antigens and the Host Cell. Journal of

Virology, 76, 5233 - 5250.

CHENIK, M., CHEBLI, K. & BLONDEL, D. 1995. Translation Initiation at Alternate In-

Frame AUG Codons in the Rabies Virus Phosphoprotein mRNA Is Mediated by a

Ribosomal Leaky Scanning Mechanism. Journal of Virology, 69, 707-712.

CHEW, T., NOYCE, R., COLLINS, S. E., HANCOCK, M. H. & MOSSMAN, K. L. 2009.

Characterization of the Interferon Regulatory Factor 3-Mediated Antiviral Response

in A Cell Line Deficient for IFN Production. Molecular Immunology, 46, 393-399.

CHUMAKOV, S. P. & PRASOLOV, V. S. 2010. Organization and Regulation of

Nucleocytoplasmic Transport. Molecular Biology, 44, 186-201.

COLLINS, P. L. 1991. The Molecular Biology of Human Respiratory Syncytial Virus (RSV)

of the Genus Pneumovirus. In: KINGSBURY, D. W. (ed.) The Paramyxoviruses.

New York: Plenum Press.

COLLINS, P. L. 2011. Human Respiratory Syncytial Virus. In: SAMAL, S. K. (ed.) The

Biology of Paramyxoviruses. Norfolk: Caister Academic Press.

COLLINS, P. L. & GRAHAM, B. S. 2008. Viral and Host Factors in Human Respiratory

Syncytial Virus Pathogenesis. Journal of Virology, 82, 2040-2055.

COLLINS, P. L., HILL, M. G., CAMARGO, E., GROSFIELD, H., CHANOCK, R. M. &

MURPHY, B. R. 1995. Production of Infectious Human Respiratory Syncytial Virus

from Cloned cDNA Confirms An Essential Role for the Transcription Elongation

Factor from the 5'-Proximal Open Reading Frame of the M2 mRNA in Gene

Expression and Provides a Capability for Vaccine Development Proceedings of the

National Academy of Sciences 92, 11563-11567.

COLLINS, P. L. & CROWE JR., J. E. 2007. Respiratory Syncytial Virus and

Metapneumovirus. In: KNIPE, D. M. & HOWLEY, P. M. (eds.) Fields Virology.

Lippincott Williams and Wilkins.

Page 145: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

132

COLLINS, P. L. & MURPHY, B. R. 2002. Respiratory Syncytial Virus: Reverse Genetics

and Vaccine Strategies. Virology, 296, 204 - 211.

COLLINS, P. L. & MURPHY, B. R. 2005. New Generation Live Vaccines Against Human

Respiratory Syncytial Virus Designed by Reverse Genetics. Proceedings of the

American Thoracic Society, 2, 166-173.

CORBETT, A. H. & SILVER, P. A. 1997. Nucleocytoplasmic Transport of Macromolecules.

Microbiology and Molecular Biology Reviews, 61, 193-211.

CORONEL, E. C., AKIMOTO, T., MURTI, K. G., VARICH, N. & PORTNER, A. 2001.

Nucleocapsid Incorporation into Parainfluenza Virus Is Regulated by Specific

Interaction with Matrix Protein. Journal of Virology, 75, 1117 - 1123.

CROWE JR., J. E. & WILLIAMS, J. V. 2011. Metapneumoviruses. In: SAMAL, S. K. (ed.)

The Biology of Paramyxoviruses. Norfolk: Caister Academic Press.

DANCHO, B., MCKENZIE, M. O., CONNOR, J. H. & LYLES, D. S. 2009. Vesicular

Stomatitis Virus Matrix Protein Mutations that Affect Association with Host

Membranes and Viral Nucleocapsids. The Journal of Biological Chemistry, 284, 4500

- 4509.

DARNIOT, M., SCHILDGEN, V., SCHILDGEN, O., SPROAT, B., KLEINES, M., DITT,

V., PITOISET, C., POTHIER, P. & MANOHA, C. 2012. RNA Interference In Vitro

and In Vivo Using siRNA Targeting the Nucleocapsid N mRNA of Human

Metapneumovirus. Antiviral Research, 93, 364 - 373.

DASARAJU, P. V. & LIU, C. 1996. Infections of the Respiratory System. In: BARON, S.

(ed.) Medical Microbiology. 4 ed. Galveston (TX): University of Texas Medical

Branch at Galveston.

DEFFRASNES, C., CÔTÉ, S. & BOIVIN, G. 2005. Analysis of Replication Kinetics of the

Human Metapneumovirus in Different Cell Lines by Real-Time PCR. Journal of

Clinical Microbiology, 43, 488 - 490.

DERDOWSKI, A., PETERS, T. R., GLOVER, N., QIAN, R., UTLEY, T. J., BURNETT, A.,

WILLIAMS, J. V., SPEARMAN, P. & CROWE JR., J. E. 2008. Human

Metapneumovirus Nucleoprotein and Phosphoprotein Interact and Provide the

Minimal Requirements for Inclusion Body Formation. Journal of General Virology,

89, 2698 - 2708.

DICARLO, A., MOLLER, P., LANDER, A., KOLESNIKOVA, L. & BECKER, S. 2007.

Nucleocapsid Formation and RNA Synthesis of Marburg Virus Is Dependent on Two

Coiled Coil Motifs in the Nucleoprotein. Virology Journal, 4, 105 - 162.

DIETZEL, E., ANDERSON, D. E., CASTAN, A., MESSLING, V. V. & MAISNER, A.

2011. Canine Distemper Virus Matrix Protein Influences Particle Infectivity, Particle

Composition, and Envelope Distribution in Polarized Epithelial Cells and Modulates

Virulence. Journal of Virology, 85, 7162-7168.

DIETZSCHOLD, B., WIKTOR, T. J., TROJANOWSKI, J. Q., MACFARLAN, R. I.,

WUNNER, W. H., TORRES-ANJEL, M. J. & KOPROWSKI, H. 1985. Differences

in Cell-to-Cell Spread of Pathogenic and Apathogenic Rabies Virus In Vivo and In

Vitro. Journal of Virology, 56, 12-18.

DOMACHOWSKE, J. B., BONVILLE, C. A. & ROSENBERG, H. F. 2001. Gene

Expression in Epithelial Cells in Response to Pneumovirus Infection. Respiratory

Research, 2, 225 - 233.

DOMACHOWSKE, J. B. & ROSENBERG, H. F. 1999. Respiratory Syncytial Virus

Infection: Immune Response, Immunopathogenesis, and Treatment. Clinical

Microbiology Reviews, 12, 298-309.

Page 146: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

133

DUDAS, R. A. & KARRON, R. A. 1998. Respiratory Syncytial Virus Vaccines. Clinical

Microbiology Reviews, 11, 430 - 439.

EMENY, J. M. & MORGAN, M. J. 1979. Regulation of the Interferon System: Evidence that

Vero Cells Have a Genetic Defect in Interferon Production Journal of General

Virology, 43, 247-252.

ENGLUND, J. A., SULLIVAN, C. J., JORDAN, C., DEHNER, L. P., VERCELOTTI, G. M.

& JR., H. H. B. 1988. Respiratory Syncytial Virus Infection in Immunocompromised

Adults. Annals of Internal Medicine, 109, 203-208.

ESPER, F., MARTINELLO, R. A., BOUCHER, D., WEIBEL, C., FERGUSON, D.,

LANDRY, M. L. & KAHN, J. S. 2004. A 1-Year Experience with Human

Metapneumovirus in Children Aged <5 Years. The Journal of Infectious Diseases,

189, 1388 - 1396.

FALSEY, A. R., CRIDDLE, M. C. & WALSH, E. E. 2006. Detection of Respiratory

Syncytial Virus and Human Metapneumovirus by Reverse Transcription Polymerase

Chain Reaction in Adults with and without Respiratory Illness. Journal of Clinical

Virology, 35, 46 - 50.

FALSEY, A. R., ERDMAN, D., ANDERSON, L. J. & WALSH, E. E. 2003. Human

Metapneumovirus Infections in Young and Elderly Adults. The Journal of Infectious

Diseases, 187, 785 - 790.

FALSEY, A. R. & WALSH, E. E. 2005. Respiratory Syncytial Virus Infection in Elderly

Adults. Drugs Aging, 22, 577-587.

FAUL, E. J., WANJALLA, C. N., SUTHAR, M. S., GALE JR., M., WIRBLICH, C. &

SCHNELL, M. J. 2010. Rabies Virus Infection Induces Type I Interferon in an IPS-1

Dependent Manner while Dendritic Cell Activation Relies On IFNAR Signaling.

PLoS Pathogens, 6, e1001016.

FEUILLET, F., LINA, B., ROSA-CALATRAVA, M. & BOIVIN, G. 2012. Ten Years of

Human Metapneumovirus Research. Journal of Clinical Virology, 53, 97 - 105.

FIEDLER, M. A., WERNKE-DOLLRIES, K. & STARK, J. M. 1996. Inhibition of Viral

Replication Reverses Respiratory Syncytial Virus-Induced NF-κB Activation and

Interleukin-8 Gene Expression in A549 Cells. Journal of Virology, 70, 9079 - 9082.

FONTANA, J. M., BANKAMP, B. & ROTA, P. A. 2008. Inhibition of Interferon Induction

and Signaling by Paramyxoviruses. Immunological Reviews, 225, 46-67.

FOSMIRE, J. A., HWANG, K. & MAKINO, S. 1992. Identification and Characterization of

a Coronavirus Packaging Signal. Journal of Virology, 66, 3522 - 3530.

FULCHER, A. J. & JANS, D. A. 2011. Regulation of Nucleocytoplasmic Trafficking of

Viral Proteins: An Integral Role in Pathogenesis? . Biochimica et Biophysica Acta

(BBA) - Molecular Cell Research, 1813, 2176-2190.

FULCHER, A. J., ROTH, D. M., FATIMA, S., ALVISI, G. & JANS, D. A. 2010. The

BRCA-1 binding protein BRAP2 is a novel, negative regulator of nuclear import of

viral proteins, dependent on phosphorylation flanking the nuclear localization signal.

The FASEB Journal, 24, 1454 - 1466.

GARCÍA-BARRENO, B., DELGADO, T. & MELERO, J. A. 1996. Identification of Protein

Regions Involved in the Interaction of Human Respiratory Syncytial Virus

Phosphoprotein and Nucleoprotein: Significance for Nucleocapsid Assembly and

Formation of Cytoplasmic Inclusion. Journal of Virology, 70, 801 - 808.

GAROFALO, R., SABRY, M., JAMALUDDIN, M., YU, R. K., CASOLA, A., OGRA, P. L.

& BRASIER, A. R. 1996. Transcriptional activation of the Interleukin-8 Gene by

Respiratory Syncytial Virus Infection in Alveolar Epithelial Cells: Nuclear

Page 147: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

134

Translocation of the RelA Transcription Factor as a Mechanism Producing Airway

Mucosal Inflammation. Journal of Virology, 70, 8773 - 8781.

GAROFALO, R. P. & HAEBERLE, H. 2000. Epithelial Regulation of Innate Immunity to

Respiratory Syncytial Virus. American Journal of Respiratory Cell and Molecular

Biology, 23, 581 - 585.

GAROFF, H., HEWSON, R. & OPSTELTEN, D.-J. E. 1998. Virus Maturation by Budding.

Microbiology and Molecular Biology Reviews, 62, 1171-1190.

GAUDIER, M., GAUDIN, Y. & KNOSSOW, M. 2002. Crystal Structure of Vesicular

Stomatitis Virus Matrix Protein. The EMBO Journal, 21, 2886 - 2892.

GAUNT, E., MCWILLIAM-LEITCH, E. C., TEMPLETON, K. & SIMMONDS, P. 2009.

Incidence, Molecular Epidemiology and Clinical Presentations of Human

Metapneumovirus; Assessment of Its Importance as A Diagnostic Screening Target.

Journal of Clinical Virology, 46, 318 - 324.

GENIN, P., ALGARTE, M., ROOF, P., LIN, R. & HISCOTT, J. 2000. Regulation of

RANTES Chemokine Gene Expression Requires Cooperativity Between NF-κB and

IFN-Regulatory Factor Transcription Factors. The Journal of Immunology, 164, 5352

- 5361.

GERN, J. E., FRENCH, D. A., GRINDLE, K. A., BROCKMAN-SCHNEIDER, R. A.,

KONNO, S. & BUSSE, W. W. 2003. Double-Stranded RNA Induces the Synthesis of

Specific Chemokines by Bronchial Epithelial Cells. American Journal of Respiratory

and Cellular and Molecular Biology, 28, 731 - 737.

GHILDYAL, R., BAULCH-BROWN, C., MILLS, J. & MEANGER, J. 2003. The Matrix

Protein of Human Respiratory Syncytial Virus Localises to the Nucleus of Infected

Cells and Inhibits Transcription. Archives of Virology, 148, 1419-1429.

GHILDYAL, R., HO, A., DIAS, M., SOEGIYONO, L., BARDIN, P. G., TRAN, K. C.,

TENG, M. N. & JANS, D. A. 2009. The Respiratory Syncytial Virus Matrix Protein

Possesses a Crm1-Mediated Nuclear Export Mechanism. Journal of Virology, 83,

5353-5362.

GHILDYAL, R., HO, A. & JANS, D. A. 2006. Central Role of the Respiratory Syncytial

Virus Matrix Protein in Infection. FEMS Microbiology Reviews, 30, 692 - 705.

GHILDYAL, R., HO, A., WAGSTAFF, K. M., DIAS, M. M., BARTON, C. L., JANS, P.,

BARDIN, P. & JANS, D. A. 2005a. Nuclear Import of the Respiratory Syncytial

Virus is Mediated by Importin β1 Independent of Importin α. Biochemistry, 44,

12887-12895.

GHILDYAL, R., LI, D., PEROULIS, I., SHIELDS, B., BARDIN, P. G., TENG, M. N.,

COLLINS, P. L., MEANGER, J. & MILLS, J. 2005b. Interaction Between the

Respiratory Syncytial Virus G Glycoprotein Cytoplasmic Domain and the Matrix

Protein. Journal of General Virology, 86, 1879-1884.

GHILDYAL, R., MILLS, J., MURRAY, M., VARDAXIS, N. & MEANGER, J. 2002.

Respiratory Syncytial Virus Matrix Protein Associates with Nucleocapsids in Infected

Cells. Journal of General Virology, 83, 753-757.

GLODOWSKI, D. R., PETERSEN, J. M. & DAHLBERG, J. E. 2002. Complex Nuclear

Localization Signal in the Matrix Protein of Vesicular Stomatitis Virus. The Journal

of Biological Chemistry, 277, 46864-46870.

GORLICH, D. & MATTAJ, I. W. 1996. Nucleocytoplasmic transport. Science, 271, 1513-

1518.

GRAY, G. C., CAPUANO, A. W., SETTERQUIST, S. F., ERDMAN, D. D., NOBBS, N. D.,

ABED, Y., DOERN, G. V., STARKS, S. E. & BOIVIN, G. 2006. Multi-Year Study

Page 148: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

135

of Human Metapneumovirus Infection at A Large US Midwestern Medical Referral

Center. Journal of Clinical Virology, 37, 269 - 276.

GREEN, T. J., ROWSE, M., TSAO, J., KANG, J., GE, P., ZHOU, Z. H. & LUO, M. 2011.

Access to RNA Encapsidated in the Nucleocapsid of Vesicular Stomatitis Virus.

Journal of Virology, 85, 2714 - 2722.

GREENSILL, J., MCNAMARA, P. S., DOVE, W., FLANAGAN, B., SMYTH, R. L. &

HART, C. A. 2003. Human Metapneumovirus in Severe Respiratory Syncytial Virus

Bronchiolitis. Emerging Infectious Diseases, 9, 372-375.

GRÜNWALD, D., SINGER, R. H. & ROUT, M. 2011. Nuclear Export Dynamics of RNA-

Protein Complexes. Nature, 475, 333-341.

HAEBERLE, H. A., TAKIZAWA, R., CASOLA, A., BRASIER, A. R., DIETERICH, H.-J.,

ROOIJEN, N. V., GATALICA, Z. & GAROFALO, R. P. 2002. Respiratory Syncytial

Virus-Induced Activation of Nuclear Factor-κB in the Lung Involves Alveolar

Macrophages and Toll-Like Receptor 4-Dependent Pathways. The Journal of

Infectious Diseases, 186, 1199 - 1206.

HALL, C. B. 1982. Respiratory Syncytial Virus: Its Transmission in the Hospital

Environment. The Yale Journal of Biology and Medicine, 55, 219-223.

HARDY, R. W. & WERTZ, G. W. 1998. The Product of the Respiratory Syncytial Virus M2

Gene ORF1 Enhances Readthrough of Intergenic Junctions during Viral

Transcription. Journal of Virology, 72, 520 - 526.

HARREMAN, M. T., KLINE, T. M., MILFORD, H. G., HARBEN, M. B., HODEL, A. E. &

CORBETT, A. H. 2004. Regulation of Nuclear Import by Phosphorylation Adjacent

to Nuclear Localization Signals. The Journal of Biological Chemistry, 279, 20613 -

20621.

HARRISON, A. M., BONVILLE, C. A., ROSENBERG, H. F. & DOMACHOWSKE, J. B.

1999. Respiratory Syncytial Virus-Induced Chemokine Expression in Lower Airways.

American Journal of Respiratory and Critical Care Medicine, 159, 1918 - 1924.

HARRISON, M. S., SAKAGUCHI, T. & SCHMITT, A. P. 2010. Paramyxovirus Assembly

and Budding: Building Particles that Transmit Infections. Int J Biochem Cell Biol, 42,

1416 - 1429.

HARTLEY, J. L., TEMPLE, G. F. & BRASCH, M. A. 2000. DNA Cloning Using In Vitro

Site-Specific Recombination. Genome Research, 10, 1788 - 1795.

HARTY, R. N., PARAGAS, J., SUDOL, M. & PALESE, P. 1999. A Proline-Rich Motif

within the Matrix Protein of Vesicular Stomatitis Virus and Rabies Virus Interacts

with WW Domains of Cellular Proteins: Implications for Viral Budding. Journal of

Virology, 73, 2921 - 2929.

HASHEM, M. & HALL, C. B. 2003. Respiratory Syncytial Virus in Healthy Adults: The

Cost of A Cold. Journal of Clinical Virology, 27, 14 - 21.

HAYDON, D. T., CLEAVELAND, S., TAYLOR, L. H. & LAURENSON, M. K. 2002.

Identifying Reservoirs of Infection: A Conceptual and Practical Challenge. Emerging

Infectious Diseases, 8, 1468-1473.

HAYNES, L. M., MOORE, D. D., KURT-JONES, E. A., FINBERG, R. W., ANDERSON,

L. J. & TRIPP, R. A. 2001. Involvement of Toll-Like Receptor 4 in Innate Immunity

to Respiratory Syncytial Virus. Journal of Virology, 75, 10730 - 10737.

HENCHAL, E. A. & PUTNAK, J. R. 1990. The Dengue Viruses. Clinical Microbiology

Reviews, 3, 376-396.

HENDERSON, B. R. & ELEFTHERIOU, A. 2000. A Comparison of the Activity, Sequence

Specificity, and CRM1-Dependence of Different Nuclear Export Signals.

Experimental Cell Research, 256, 213 - 224.

Page 149: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

136

HENDERSON, B. R. & PERCIPALLE, P. 1997. Interactions between HIV Rev and Nuclear

Import and Export Factors: The Rev Nuclear Localisation Signal Mediates Specific

Binding to Human Importin-B. Journal of Molecular Biology, 274, 693 - 707.

HER, L.-S., LUND, E. & DAHLBERG, J. E. 1997. Inhibition of Ran Guanosine

Triphosphatase-Dependent Nuclear Transport by the Matrix Protein of Vesicular

Stomatitis Virus. Science, 276, 1845-1847.

HERFST, S., GRAAF, M. D., SCHICKLI, J. H., TANG, R. S., KAUR, J., YANG, C.-F.,

SPAETE, R. R., HALLER, A. A., HOOGEN, B. G. V. D., OSTERHAUS, A. D. M.

E. & FOUCHIER, R. A. M. 2004. Recovery of Human Metapneumovirus Genetic

Lineages A and B from Cloned cDNA. Journal of Virology, 78, 8264 - 8270.

HISCOTT, J. 2007. Triggering the Innate Antiviral Response through IRF-3 Activation.

Journal of Biological Chemistry, 282, 15325-15329.

HISCOX, J. A., WURM, T., WILSON, L., BRITTON, P., CAVANAGH, D. & BROOKS, G.

2001. The Coronavirus Infectious Bronchitis Virus Nucleoprotein Localizes to the

Nucleolus. Journal of Virology, 75, 506 - 512.

HOPKINS, R. F., WALL, V. E. & ESPOSITO, D. 2012. Optimizing Transient Recombinant

Protein Expression in Mammalian Cells. In: HARTLEY, J. L. (ed.) Methods in

Molecular Biology. SpringerLink.

HOWARD, M. & WERTZ, G. 1989. Vesicular Stomatitis Virus RNA Replication: a Role for

the NS Protein. Journal of General Virology, 70, 2683 - 2694.

HÜBNER, S., XIAO, C.-Y. & JANS, D. A. 1997. The Protein Kinase CK2 Site (Ser111/112)

Enhances Recognition of the Simian Virus 40 Large T-antigen Nuclear Localization

Sequence by Importin. The Journal of Biological Chemistry, 272, 17191 - 17195.

HUTTEN, S. & KEHLENBACH, R. H. 2007. CRM1-Mediated Nuclear Export: To The Pore

and Beyond. TRENDS in Cell Biology, 17, 193-201.

JAMALI, T., JAMALI, Y., MEHRBOD, M. & MOFRAD, M. R. K. 2011. Nuclear Pore

Complex: Biochemistry and Biophysics of Nucleocytoplasmic Transport in Health

and Disease. International Review of Cell and Molecular Biology, 287, 233 - 286.

JANS, D. A. 1995. The Regulation of Protein Transport to the Nucleus by Phosphorylation.

Biochemical Journal, 311, 705-716.

JANS, D. A. & HÜBNER, S. 1996. Regulation of Protein Transport to the Nucleus: Central

Role of Phosphorylation. Physiological Reviews, 76, 651 - 685.

JAYAKAR, H. R., JEETENDRA, E. & WHITT, M. A. 2004. Rhabdovirus Assembly and

Budding. Virus Research, 106, 117 - 132.

JOHANSSON, M., BROOKS, A. J., JANS, D. A. & VASUDEVAN, S. G. 2001. A Small

Region of the Dengue Virus Encoded RNA-Dependent RNA Polymerase, NS5,

Confers Interaction with Both the Nuclear Transport Receptor Importin-B and the

Viral Helicase, NS3. Journal of General Virology, 82, 735-745.

KAHN, J. S. 2006. Epidemiology of Human Metapneumovirus. Clinical Microbiology

Reviews, 19, 546 - 557.

KAPOOR, M., ZHANG, L., RAMACHANDRA, M., KUSUKAWA, J., EBNER, K. E. &

PADMANABHAN, R. 1995. Association between NS3 and NS5 Proteins of Dengue

Virus Type 2 in the Putative RNA Replicase Is Linked to Differential Phosphorylation

of NS5. The Journal of Biological Chemistry, 270, 19100-19106.

KARRON, R. A., BUONAGURIO, D. A., GEORGIU, A. F., WHITEHEAD, S. S.,

ADAMUS, J. E., CLEMENTS-MANN, M. L., HARRIS, D. O., RANDOLPH, V. B.,

UDEM, S. A., MURPHY, B. R. & SIDHU, M. S. 1997a. Respiratory Syncytial Virus

(RSV) SH and G Proteins Are Not Essential for Viral Replication In Vitro: Clinical

Evaluation and Molecular Characterization of A Cold-Passaged, Attenuated RSV

Page 150: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

137

Subgroup B Mutant. Proceedings of the National Academy of Sciences, 94, 13961-

13966.

KARRON, R. A., WRIGHT, P. F., CROWE JR, J. E., CLEMENTS-MANN, M. L.,

THOMPSON, J., MAKHENE, M., CASEY, R. & MURPHY, B. R. 1997b.

Evaluation of Two Live, Cold-Passaged, Temperature-Sensitive Respiratory Syncytial

Virus Vaccines in Chimpanzees and in Human Adults, Infants, and Children. The

Journal of Infectious Diseases, 176, 1428-1436.

KATZE, M. G., FORNEK, J. L., PALERMO, R. E., WALTERS, K.-A. & KORTH, M. J.

2008. Innate Immune Modulation by RNA Viruses: Emerging Insights from

Functional Genomics. Nature Reviews Immunology, 8, 644-653.

KESSON, A. M. 2007. Respiratory Virus Infections. Paediatric Respiratory Reviews, 8, 240

- 248.

KINDT, T. J., GOLDSBY, R. A. & OSBORNE, B. A. 2007. Kuby Immunology, New York,

W.H. Freeman and Company.

KINGSTON, R. E. 2003. Introduction of DNA into Mammalian Cells. Current Protocols in

Molecular Biology. John Wiley & Sons, Inc.

KOBAYASHI, T., KAMITANI, W., ZHANG, G., WATANABE, M., TOMONAGA, K. &

IKUTA, K. 2001. Borna Disease Virus Nucleoprotein Requires both Nuclear

Localization and Export Activities for Viral Nucleocytoplasmic Shuttling. Journal of

Virology, 75, 3404 - 3412.

KOLAKOFSKY, D., PELET, T., GARCIN, D., HAUSMANN, S., CURRAN, J. & ROUX,

L. 1998. Paramyxovirus RNA Synthesis and the Requirement for Hexamer Genome

Length: the Rule of Six Revisited Journal of Virology, 72, 891 - 899.

KRAUS, A. A., RAFTERY, M. J., GIESE, T., ULRICH, R., ZAWATZKY, R.,

HIPPENSTIEL, S., SUTTORP, N., KRÜGER, D. H. & SCHONRICH, G. 2004.

Differential Antiviral Response of Endothelial Cells after Infection with Pathogenic

and Nonpathogenic Hantaviruses. Journal of Virology, 78, 6143-6150.

KRAUS, I., BOGNER, E., LILIE, H., EICKMANN, M. & GARTEN, W. 2005.

Oligomerization and Assembly of the Matrix Protein of Borna Disease Virus. FEBS

Letters, 579, 2686 - 2692.

KUIKEN, T., VAN DEN HOOGEN, B. G., VAN RIEL, D. A. J., LAMAN, J. D., VAN

AMERONGEN, G., SPRONG, L., FOUCHIER, R. A. M. & OSTERHAUS, A. D. M.

E. 2004. Experimental Human Metapneumovirus Infection in Cynomolgus macaques

(Macaca fascicularis) Results in Virus Replication in Ciliated Epithelial Cells and

Pneumocytes with Associated Lesions throughout the Respiratory Tract. American

Journal of Pathology, 164, 1893 - 1900.

KURT-JONES, E. A., POPOVA, L., KWINN, L., HAYNES, L. M., JONES, L. P., TRIPP,

R. A., WALSH, E. E., FREEMAN, M. W., GOLENBOCK, D. T., ANDERSON, L. J.

& FINBERG, R. W. 2000. Pattern Recognition Receptors TLR4 and CD14 Mediate

Response to Respiratory Syncytial Virus. Nature Immunology, 1, 398 - 401.

LA COUR, T., KIEMER, L., MOLGAARD, A., GUPTA, R., SKRIVER, K. & BRUNAK, S.

2004. Analysis and Prediction of Leucine-Rich Nuclear Export Signals. Protein

Engineering, Design & Selection, 17, 527-536.

LAM, M. H. C., HU, W., XIAO, C.-Y., GILLESPIE, M. T. & JANS, D. A. 2001. Molecular

Dissection of the Importin β1-Recognized Nuclear Targeting Signal of Parathyroid

Hormone-Related Protein. Biochemical and Biophysical Research Communications,

282, 629 - 634.

Page 151: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

138

LAMB, R. A. & PARKS, G. D. 2006. Paramyxoviridae: The Viruses and Their Replication.

In: KNIPE, D. M. & HOWLEY, P. M. (eds.) Fields Virology 5th ed.: Lippincot

Williams & Wilkins.

LATIFF, K., MEANGER, J., MILLS, J. & GHILDYAL, R. 2004. Sequence and Structure

Relatedness of Matrix Protein of Human Respiratory Syncytial Virus with Matrix

Proteins of Other Negative-Sense RNA Viruses Clinical Microbiology and Infection,

10, 945-948.

LEE, C., HODGINS, D., CALVERT, J. G., WELCH, S.-K. W., JOLIE, R. & YOO, D. 2006.

Mutations within the Nuclear Localization Signal of the Porcine Reproductive and

Respiratory Syndrome Virus Nucleocapsid Protein Attenuate Virus Replication.

Virology, 346, 238 - 250.

LENARD, J. 1996. Negative Strand Virus M and Retrovirus MA Proteins: All in a Family?

Virology, 216, 289-298.

LI, D., JANS, D. A., BARDIN, P. G., MEANGER, J., MILLS, J. & GHILDYAL, R. 2008.

Association of Respiratory Syncytial Virus M Protein with Viral Nucleocapsids Is

Mediated by the M2-1 Protein. Journal of Virology, 82, 8863-8870.

LI, J., WANG, Z., GONZALEZ, R., XIAO, Y., ZHOU, H., ZHANG, J., PARANHOS-

BACCALA, G., VERNET, G., JIN, Q., WANG, J. & HUNG, T. 2012. Prevalence of

Human Metapneumovirus in Adults with Respiratory Tract Infection in Beijing,

China. Journal of Infection, 64, 96 - 103.

LIAO, R. S., APPELGATE, D. M. & PELZ, R. K. 2012. An Outbreak of Severe Respiratory

Tract Infection due to Human Metapneumovirus in a Long-Term Care Facility for the

Elderly in Oregon. Journal of Clinical Virology, 53, 171 - 173.

LIEBERMAN, D., KORSONSKY, I., BEN-YAKOOV, M., LAZAROVICH, Z.,

FRIEDMAN, M. G., DVOSKIN, B., LEINONEN, M., OHANA, B. & BOLDUR, I.

2002. A Comparative Study of the Etiology of Adult Upper and Lower Respiratory

Tract Infections in the Community. Diagnostic Microbiology and Infectious Disease,

42, 21 - 28.

LIEW, C.-G., DRAPER, J. S., WALSH, J., MOORE, H. & ANDREWS, P. W. 2007.

Transient and Stable Transgene Expression in Human Embryonic Stem Cells. Stem

Cells, 25, 1521-1528.

LIN, R., HEYLBROECK, C., GENIN, P., PITHA, P. M. & HISCOTT, J. 1999. Essential

Role of Interferon Regulatory Factor 3 in Direct Activation of RANTES Chemokine

Expression. Molecular and Cellular Biology, 19, 959-966.

LYLES, D. S. & RUPPRECHT, C. E. 2007. Rhabdoviridae. In: KNIPE, D. M. & HOWLEY,

P. M. (eds.) Fields Virology. Lippincotts Williams & Wilkins.

LYNCH, S. & KOLAKOFSKY, D. 1978. Ends of the RNA within Sendai Virus Defective

Interfering Nucleocapsids Are Not Free. Journal of Virology, 28, 584 - 589.

MACARA, I. G. 2001. Transport into and out of the Nucleus. Microbiology and Molecular

Biology Reviews, 65, 570 - 594.

MASTRONARDE, J. G., HE, B., MONICK, M. M., MUKAIDA, N., MATSUSHIMA, K. &

HUNNINGHAKE, G. W. 1996. Induction of Interleukin(IL)-8 Gene Expression by

Respiratory Syncytial Virus Involves Activation of Nuclear Factor (NF)-κB and NF-

IL-6. The Journal of Infectious Diseases, 174, 262 - 267.

MEBATSION, T., WEILAND, F. & CONZELMANN, K.-K. 1999. Matrix Protein of Rabies

Virus Is Responsible for the Assembly and Budding of Bullet-Shaped Particles and

Interacts with the Transmembrane Spike Glycoprotein G. Journal of Virology, 73, 242

- 250.

Page 152: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

139

MEKHAIL, K., RIVERO-LOPEZ, L., AL-MASRI, A., BRANDON, C., KHACHO, M. &

LEE, S. 2007. Identification of a Common Subnuclear Localization Signal. Molecular

Biology of the Cell, 18, 3966-3977.

MILLER, A. L., BOWLIN, T. L. & LUCACS, N. W. 2003. Respiratory Syncytial Virus-

Induced Chemokine Production: Linking Viral Replication to Chemokine Production

In Vitro and In Vivo. The Journal of Infectious Diseases, 1819, 1419 - 1430.

MIR, M. A. & PANGANIBAN, A. T. 2006. The Bunyavirus Nucleocapsid Protein Is an

RNA Chaperone: Possible Roles in Viral RNA Panhandle Formation and Genome

Replication. RNA, 12, 272 - 282.

MOGENSEN, T. 2009. Pathogen Recognition and Inflammatory Signaling in Innate Immune

Defenses. Clinical Microbiology Reviews, 22, 240-273.

MOHL, B.-P. & BARR, J. N. 2009. Investigating the Specificity and Stoichiometry of RNA

Binding by the Nucleocapsid Protein of Bunyamwera Virus. RNA, 15, 391 - 399.

MONEY, V. A., MCPHEE, H. K., MOSELY, J. A., SANDERSON, J. M. & YEO, R. P.

2009. Surface Features of a Mononegavirales Matrix Protein Indicate Sites of

Membrane Interaction Proceedings of the National Academy of Sciences, 106, 4441-

4446.

MORTON, C. J., CAMERON, R., LAWRENCE, L. J., LIN, B., LOWE, M., LUTTICK, A.,

MASON, A., MCKIMM-BRESCHKIN, J., PARKER, M. W., RYAN, J., SMOUT,

M., SULLIVAN, J., TUCKER, S. P. & YOUNG, P. R. 2003. Structural

Characterization of Respiratory Syncytial Virus Fusion Inhibitor Escape Mutants:

Homology Model of the F Protein and a Syncytium Formation Assay. Virology, 311,

275 - 288.

MOSELEY, G. W., FILMER, R. P., DEJESUS, M. A. & JANS, D. A. 2007.

Nucleocytoplasmic Distribution of Rabies Virus P-Protein Is Regulated by

Phosphorylation Adjacent to C-Terminal Nuclear Import and Export Signals.

Biochemistry, 46, 12053-12061.

MUKAIDA, N., HARADA, A. & MATSUSHIMA, K. 1998. Interleukin-8 (IL-8) and

Monocyte Chemotactic and Activating Factor (MCAF/MCP-1), Chemokines

Essentially Involved in Inflammatory and Immune Responses. Cytokine & Growth

Factor Reviews, 9, 9 - 23.

MUKAIDA, N., OKAMOTO, S., ISHIKAWA, Y. & MATSUSHIMA, K. 1994. Molecular

Mechanism of Interleukin-8 Gene Expression. Journal of Leukocyte Biology, 56, 554

- 558.

MYERS, T. M., SMALLWOOD, S. & MOYER, S. A. 1999. Identification of Nucleocapsid

Protein Residues Required for Sendai Virus Nucleocapsid Formation and Genome

Replication Journal of General Virology, 80, 1383 - 1391.

NARAYANAN, K., MAEDA, A., MAEDA, J. & MAKINO, S. 2000. Characterization of the

Coronavirus M Protein and Nucleocapsid Interaction in Infected Cells. Journal of

Virology, 74, 8127 - 8134.

NAYAK, D., PANDA, D., DAS, S. C., LUO, M. & PATTNAIK, A. K. 2009. Single-Amino-

Acid Alterations in a Highly Conserved Central Region of Vesicular Stomatitis Virus

N Protein Differentially Affect the Viral Nucleocapsid Template Functions. Journal

of Virology, 83, 5525 - 5534.

NELSON, D. L. & COX, M. M. 2008. Lehninger Principles of Biochemistry, New York, W

H Freeman and Company.

NELSON, P. J., KIM, H. T., MANNING, W. C., GORALSKI, T. J. & KRENSKY, A. M.

1993. Genomic Organizations and Transcriptional Regulation of the RANTES

Chemokine Gene. The Journal of Immunology, 151, 2601 - 2612.

Page 153: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

140

NIGG, E. A. 1997. Nucleocytoplasmic Transport: Signals, Mechanisms, and Regulation.

Nature, 386, 779-786.

NOAH, T. L., IVINS, S. S., MURPHY, P., KAZACHKOVA, I., MOATS-STAATS, B. &

HENDERSON, F. W. 2002. Chemokines and Inflammation in the Nasal Passages of

Infants with Respiratory Syncytial Virus Bronchiolitis. Clinical Immunology, 104, 86

- 95.

NORRBY, E., MARUSYK, H. & ORVELL, C. 1970. Morphogenesis of Respiratory

Syncytial Virus in a Green Monkey Kidney Cell Line (Vero). Journal of Virology, 6,

237-242.

OGRA, P. L. 2004. Respiratory Syncytial Virus: The Virus, The Disease, and The Immune

Response. Paediatric Respiratory Reviews, 5, S119-S126.

OLSZEWSKA-PAZDRAK, B., CASOLA, A., SAITO, T., ALAM, R., CROWE, S. E., MEI,

F., OGRA, P. L. & GAROFALO, R. P. 1998. Cell-Specific Expression of RANTES,

MCP-1, and MIP-1A by Lower Airway Epithelial Cells and Eosinophils Infected with

Respiratory Syncytial Virus. Journal of Virology, 72, 4756 - 4764.

OPENSHAW, P. J. M. & TREGONING, J. S. 2005. Immune Responses and Disease

Enhancement During Respiratory Syncytial Virus Infection. Clinical Microbiology

Reviews, 18, 541-555.

OSHANSKY, C. M., ZHANG, W., MOORE, E. & TRIPP, R. A. 2009. The Host Response

and Molecular Pathogenesis Associated with Respiratory Syncytial Virus Infection.

Future Microbiol., 4, 279 - 297.

PALMERI, D. & MALIM, M. H. 1999. Importin β Can Mediate the Nuclear Import of an

Arginine-Rich Nuclear Localization Signal in the Absence of Importin α. Molecular

and Cellular Biology, 19, 1218-1225.

PAPENBURG, J. & BOIVIN, G. 2010. The Distinguishing Features of Human

Metapneumovirus and Respiratory Syncytial Virus. Reviews in Medical Virology, 20,

245 - 260.

PASDELOUP, D., POISSON, N., RAUX, H., GAUDIN, Y., RUIGROK, R. W. H. &

BLONDEL, D. 2005. Nucleocytoplasmic Shuttling of the Rabies Virus P Protein

Requires a Nuclear Localization Signal and a CRM1-Dependent Nuclear Export

Signal. Virology, 334, 284-293.

PEMBERTON, L. F. & PASCHAL, B. M. 2005. Mechanisms of Receptor-Mediated Nuclear

Import and Nuclear Export. Traffic, 6, 187-198.

PERET, T. C. T., BOIVIN, G., LI, Y., COUILLARD, M., HUMPHREY, C., OSTERHAUS,

A. D. M. E., ERDMAN, D. D. & ANDERSON, L. J. 2002. Characterization of

Human Metapneumoviruses Isolated from Patients in North America. The Journal of

Infectious Diseases, 185, 1660 - 1663.

PETERSEN, J. M., HER, L.-S., VARVEL, V., LUND, E. & DAHLBERG, J. E. 2000. The

Matrix Protein of Vesicular Stomatitis Virus Inhibits Nucleocytoplasmic Transport

When It Is in the Nucleus and Associated with Nuclear Pore Complexes. Molecular

and Cellular Biology, 20, 8590-8601.

PETRAITYTE-BURNEIKIENE, R., NALIVAIKO, K., LASICKIENE, R., FIRANTIENE,

R., EMUZYTE, R., SASNAUSKAS, K. & ZVIRBLIENE, A. 2011. Generation of

Recombinant Metapneumovirus Nucleocapsid Protein As Nucleocapsid-Like Particles

and Development of Virus-Specific Monoclonal Antibodies. Virus Research, 161, 131

- 139.

PLATANIAS, L. C. 2005. Mechanisms of Type-I and Type-II-Interferon-Mediated

Signalling. Nature Reviews Immunology, 5, 375-387.

Page 154: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

141

PRYOR, M. J., RAWLINSON, S. M., BUTCHER, R. E., BARTON, C. L., WATERHOUSE,

T. A., VASUDEVAN, S. G., BARDIN, P. G., WRIGHT, P. J., JANS, D. A. &

DAVIDSON, A. D. 2007. Nuclear Localization of Dengue Virus Nonstructural

Protein 5 through Its Importin α/β-Recognized Nuclear Localization Sequences is

Integral to Viral Infection. Traffic, 8, 795-807.

RANDALL, R. E. & GOODBOURN, S. 2008. Interferons and Viruses: An Interplay between

Induction, Signalling, Antiviral Response, and Virus Countermeasures. Journal of

General Virology, 89, 1-47.

RAWLINSON, S. M., PRYOR, M. J., WRIGHT, P. J. & JANS, D. A. 2009. CRM1-

Mediated Nuclear Export of Dengue Virus RNA Polymerase NS5 Modulates

Interleukin-8 Induction and Virus Production The Journal of Biological Chemistry,

284, 15589-15597.

RAYMOND, D. D., PIPER, M. E., GERRARD, S. R. & SMITH, J. L. 2010. Structure of the

Rift Valley Fever Virus Nucleocapsid Protein Reveals Another Architecture for RNA

Encapsidation. Proccedings of the National Academy of Sciences (PNAS), 107, 11769

- 11774.

REED, K. E., GORBALENYA, A. E. & RICE, C. M. 1998. The NS5A/NS5 Proteins of

Viruses from Three Genera of the Family Flaviviridae Are Phosphorylated by

Associated Serine/Threonine Kinases. Journal of Virology, 72, 6199-6206.

REIN, A., HARVIN, D. P., MIRRO, J., ERNST, S. M. & GORELICK, R. J. 1994. Evidence

that a Central Domain of Nucleocapsid Protein Is Required for RNA Packaging in

Murine Leukemia Virus. Journal of Virology, 68, 6124 - 6129.

REYRAT, J.-M., PELICIC, V., GICQUEL, B. & RAPPUOLI, R. 1998. Counterselectable

Markers: Untapped Tools for Bacterial Genetics and Pathogenesis. Infection and

Immunity, 66, 4011-4017.

RIHS, H.-P., JANS, D. A., FAN, H. & PETERS, R. 1991. The Rate of Nuclear Cytoplasmic

Protein Transport Is Determined by the Casein Kinase II Site Flanking the Nuclear

Localization Sequence of the SV40 T-antigen. The EMBO Journal, 10, 633 - 639.

ROBERTS, S. R., COMPANS, R. W. & WERTZ, G. W. 1995. Respiratory Syncytial Virus

Matures at the Apical Surfaces of Polarized Epithelial Cells. Journal of Virology, 69,

2667-2673.

ROCHE, P., LAMBERT, S. & SPENCER, J. 2003. Surveillance of Viral Pathogens in

Australia: Respiratory Syncytial Virus. Communicable Diseases Intelligence, 27, 117-

122.

RODRIGUEZ, L., CUESTA, I., ASENJO, A. & VILLANUEVA, N. 2004. Human

Respiratory Syncytial Virus Matrix Protein Is an RNA-Binding Protein: Binding

Properties, Location, and Identity of the RNA Contact Residues. Journal of General

Virology, 85, 709-719.

ROWLAND, R. R., KERVIN, R., KUCKLEBURG, C., SPERLICH, A. & BENFIELD, D.

A. 1999. The Localization of Porcine Reproductive and Respiratory Syndrome Virus

Nucleocapsid Protein to the Nucleolus of Infected Cells and Identification of a

Potential Nucleolar Localization Signal Sequence. Virus Research, 64, 1 - 12.

ROWLAND, R. R. & YOO, D. 2003. Nucleolar-Cytoplasmic Shuttling of PRRSV

Nucleocapsid Protein: A Simple Case of Molecular Mimicry or the Complex

Regulation by Nuclear Import, Nucleolar Localization, and Nuclear Export Signal

Sequences. Virus Research, 95, 23 - 33.

RUUSKANEN, O., LAHTI, E., JENNINGS, L. C. & MURDOCH, D. R. 2011. Viral

Pneumonia. The Lancet, 377, 1264 - 1275.

Page 155: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

142

SAITO, T., DESKIN, R. W., CASOLA, A., HAEBERLE, H., OLSZEWSKA, B., ERNST, P.

B., ALAM, R., OGRA, P. L. & GAROFALO, R. 1997. Respiratory Syncytial Virus

Induces Selective Production of the Chemokine RANTES by Upper Airway Epithelial

Cells. The Journal of Infectious Diseases, 175, 497 - 504.

SARASINI, A., PERCIVALLE, E., ROVIDA, F., CAMPANINI, G., GENINI, E.,

TORSELLINI, M., PAOLUCCI, S., BALDANTI, F., MARCHI, A., GRAZIA-

REVELLO, M. & GERNA, G. 2006. Detection and Pathogenicity of Human

Metapneumovirus Respiratory Infection in Pediatric Italian Patients during a Winter-

Spring Season. Journal of Clinical Virology, 35, 59 - 68.

SATO, H., MASUDA, M., MIURA, R., YONEDA, M. & KAI, C. 2006. Morbillivirus

Nucleoprotein Possesses A Novel Nuclear Localization Signal and a CRM1-

Independent Nuclear Export Signal. Virology, 352, 121 - 130.

SCHNELL, M. J., MCGETTIGAN, J. P., WIRBLICH, C. & PAPANERI, A. 2010. The Cell

Biology of Rabies Virus: Using Stealth to Reach the Brain. Nature Reviews

Microbiology, 8, 51-61.

SCOTT, M. S., BOISVERT, F.-M., MCDOWALL, M. D., LAMOND, A. I. & BARTON, G.

J. 2010. Characterization and Prediction of Protein Nucleolar Localization Sequences.

Nucleic Acid Research, 38, 7388-7399.

SCOTT, M. S., TROSHIN, P. V. & BARTON, G. J. 2011. NoD: A Nucleolar Localization

Sequence Detector for Eukaryotic and Viral Proteins. BMC Bioinformatics, 12, 317.

SEKIDO, N., MUKAIDA, N., HARADA, A., NAKANISHI, I., WATANABE, Y. &

MATSUSHIMA, K. 1993. Prevention of Lung Reperfusion Injury in Rabbits by a

Monoclonal Antibody against Interleukin-8. Nature, 365, 655 - 657.

SEMPLE, M. G., COWELL, A., DOVE, W., GREENSILL, J., MCNAMARA, P. S.,

HALFHIDE, C., SHEARS, P., SMYTH, R. L. & HART, C. A. 2005. Dual Infection

of Infants by Human Metapneumovirus and Human Respiratory Syncytial Virus Is

Strongly Associated with Severe Bronchiolitis. The Journal of Infectious Diseases,

191, 382-386.

SILVER, P. A. 1991. How Proteins Enter the Nucleus. Cell, 64, 489-497.

SIMOES, E. A. F. 1999. Respiratory Syncytial Virus Infection. The Lancet, 354, 847 - 852.

SLOOTS, T. P., MACKAY, I. M., BIALASIEWICZ, S., JACOB, K. C., MCQUEEN, E.,

HARNETT, G. B., SIEBERT, D. J., MASTERS, I. B., YOUNG, P. R. & NISSEN, M.

D. 2006. Human Metapneumovirus, Australia, 2001-2004. Emerging Infectious

Diseases, 12, 1263 - 1266.

SMYTH, R. L., MOBBS, K. J., O'SHEA, U., ASHBY, D. & HART, C. A. 2002. Respiratory

Syncytial Virus Bronchiolitis: Disease Severity, Interleukin-8, and Virus Genotype.

Pediatric Pulmonology, 33, 339 - 346.

SOROKIN, A. V., KIM, E. R. & OVCHINNIKOV, L. P. 2007. Nucleocytoplasmic Transport

of Proteins. Biochemistry (Moscow), 72, 1439-1457.

SPANN, K. M., TRAN, K.-C., CHI, B., RABIN, R. L. & COLLINS, P. L. 2004. Suppression

of the Induction of Alpha, Beta, and Gamma Interferons by the NS1 and NS2 Proteins

of Human Respiratory Syncytial Virus in Human Epithelial Cells and Macrophages.

Journal of Virology, 78, 4363-4369.

SPANN, K. M., TRAN, K. C. & COLLINS, P. L. 2005. Effects of Nonstructural Proteins

NS1 and NS2 of Human Respiratory Syncytial Virus on Interferon Regulatory Factor

3, NF-κB, and Proinflammatory Cytokines. Journal of Virology, 79, 5354 - 5362.

STETSON, D. B. & MEDZHITOV, R. 2006. Antiviral Defense: Interferons and Beyond. The

Journal of Experimental Medicine, 203, 1837-1841.

Page 156: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

143

STOKES PEEBLES JR., R. & GRAHAM, B. S. 2005. Pathogenesis of Respiratory Syncytial

Virus in the Murine Model. Proceedings of the American Thoracic Society, 2, 110-

115.

STOLTZ, M. & KLINGSTRÖM, J. 2010. Alpha/Beta Interferon (IFN-α/β)-Independent

Induction of IFN- l1 (Interleukin-29) in Response to Hantaan Virus Infection. Journal

of Virology, 84, 9140 - 9148.

STRICKER, R., MOTTET, G. & ROUX, L. 1994. The Sendai Virus Matrix Protein Appears

To Be Recruited in the Cytoplasm by the Viral Nucleocapsid to Function in Viral

Assembly and Budding. Journal of General Virology, 75, 1031 - 1042.

SURJIT, M., KUMAR, R., MISHRA, R. N., REDDY, M. K., CHOW, V. T. K. & LAL, S. K.

2005. The Severe Acute Respiratory Syndrome Coronavirus Nucleocapsid Protein Is

Phosphorylated and Localizes in the Cytoplasm by 14-3-3-Mediated Translocation.

Journal of Virology, 79, 11476 - 11486.

TAKAYAMA, I., SATO, H., WATANABE, A., OMI-FURUTANI, M., SUGAI, A. &

KANKI, K. 2012. The Nucleocapsid Protein of Measles Virus Blocks Host Interferon

Response. Virology, 424, 45 - 55.

TAKEDA, K. & AKIRA, S. 2004. TLR Signaling Pathways. Seminars in Immunology, 16, 3

- 9.

TAKEUCHI, O. & AKIRA, S. 2008. MDA5/RIG-I and Virus Recognition. Current Opinion

in Immunology, 20, 17-22.

TAKIMOTO, T. & PORTNER, A. 2004. Molecular Mechanism of Paramyxovirus Budding.

Virus Research, 106, 133 - 145.

TATOCHENKO, V., UCHAIKIN, V., GORELOV, A., GUDKOV, K., CAMPBELL, A.,

SCHULZ, G., PRAHL, R. & NOTARIO, G. 2010. Epidemiology of Respiratory

Syncytial Virus in Children Less or Equal Than 2 Years of Age Hospitalized with

Lower Respiratory Tract Infections in the Russian Federation: A Prospective,

Multicenter Study. Clinical Epidemiology, 2, 221-227.

TENG, M. N. & COLLINS, P. L. 1998. Identification of the Respiratory Syncytial Virus

Proteins Required for Formation and passage of Helper-Dependent Infectious

Particles. Journal of Virology, 72, 5707-5716.

TERRY, L. J., SHOWS, E. B. & WENTE, S. R. 2007. Crossing the Nuclear Envelope:

Hierarchical Regulation of Nucleocytoplasmic Transport. Science, 318, 1412-1416.

THOMAS, L. H., FRIEDLAND, J. S., SHARLAND, M. & BECKER, S. 1998. Respiratory

Syncytial Virus-Induced RANTES Production from Human Bronchial Epithelial Cells

Is Dependent on Nuclear Factor-κB Nuclear Binding and Is Inhibited by Adenovirus-

Mediated Expression of Inhibitor of κB-α. Journal of Immunology, 161, 1007 - 1016.

THOMPSON, W. W., SHAY, D. K., WEINTRAUB, E., BRAMMER, L., COX, N.,

ANDERSON, L. J. & FUKUDA, K. 2003. Mortality Associated with Influenza and

Respiratory Syncytial Virus in the United States. Journal of the American Medical

Association, 289, 179-186.

TIAN, B., ZHANG, Y., LUXON, B. A., GAROFALO, R. P., CASOLA, A., SINHA, M. &

BRASIER, A. R. 2002. Identification of NF-κB-Dependent Gene Networks in

Respiratory Syncytial Virus Infected Cells. Journal of Virology, 76, 6800 - 6814.

TIJMS, M. A., VAN DER MEER, Y. & SNIJDER, E. J. 2002. Nuclear Localization of Non-

Structural Protein 1 and Nucleocapsid Protein of Equine Arteritis Virus. Journal of

General Virology, 83, 795 - 800.

TIMNEY, B. L., NOVATT-TETTENBAUM, J., AGATE, D. S., WILLIAMS, R., ZHANG,

W., CHAIT, B. T. & ROUT, M. P. 2006. Simple Kinetics Relationships and

Page 157: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

144

Nonspecific Competition Govern Nuclear Import Rates In Vivo. The Journal of Cell

Biology, 175, 579-593.

TOLLEFSON, S. J., COX, R. G. & WILLIAMS, J. V. 2010. Studies of Culture Conditions

and Environmental Stability of Human Metapneumovirus. Virus Research, 151, 54 -

59.

TRIPP, R. 2006. Immunostaining for Human Metapneumovirus (HMPV) [Online]. Athens:

The University of Georgia College of Veterinary Medicine. Available:

http://www.vet.uga.edu/ID/tripp/Assets/docs/RTLP-HMPV-3.pdf [Accessed 31

January 2012].

TRIPP, R. 2010. Immunostaining Plaque Assay for Respiratory Syncytial Virus: From Stock

Virus [Online]. Athens: The University of Georgia College of Veterinary Medicine.

Available: http://www.vet.uga.edu/ID/tripp/protocols.php [Accessed 5 May 2011].

VAN DEN HOOGEN, B. G., BESTEBROER, T. M., OSTERHAUS, A. D. M. E. &

FOUCHIER, R. A. M. 2002. Analysis of the Genomic Sequence of a Human

Metapneumovirus. Virology, 295, 119 - 132.

VAN DEN HOOGEN, B. G., HERFST, S., SPRONG, L., CANE, P. A., FORLEO-NETO,

E., SWART, R. L. D., OSTERHAUS, A. D. M. E. & FOUCHIER, R. A. M. 2004a.

Antigenic and Genetic Variability of Human Metapneumoviruses. Emerging

Infectious Diseases, 10, 658 - 666.

VAN DEN HOOGEN, B. G., JONG, J. C. D., GROEN, J., KUIKEN, T., GROOT, R. D.,

FOUCHIER, R. A. M. & OSTERHAUS, A. D. M. E. 2001. A Newly Discovered

Human Pneumovirus Isolated from Young Children with Respiratory Tract Disase.

Nature Medicine, 7, 719-724.

VAN DEN HOOGEN, B. G., OSTERHAUS, A. D. M. E. & FOUCHIER, R. A. M. 2004b.

Clinical Impact and Diagnosis of Human Metapneumovirus Infection. The Pediatric

Infectious Disease Journal, 23, S25 - S32.

VIDY, A., BOUGRINI, J. E., CHELBI-ALIX, M. K. & BLONDEL, D. 2007. The

Nucleocytoplasmic Rabies Virus P Protein Counteracts Interferon Signaling by

Inhibiting both Nuclear Accumulation and DNA Binding of STAT1. Journal of

Virology, 81, 4255-4263.

VIDY, A., CHELBI-ALIX, M. & BLONDEL, D. 2005. Rabies Virus P Protein Interacts with

STAT1 and Inhibits Interferon Signal Transduction Pathways. Journal of Virology,

79, 14411-14420.

VON KOBBE, C., DEURSEN, J. M. A. V., RODRIGUES, J. P., SITTERLIN, D., BACHI,

A., WU, X., WILM, M., CARMO-FONSECA, M. & IZAURRALDE, E. 2000.

Vesicular Stomatitis Virus Matrix Protein Inhibits Host Cell Gene Expression by

Targeting the Nucleoporin Nup 98. Molecular Cell, 6, 1243-1252.

WANG, G. P. & BUSHMAN, F. D. 2006. A Statistical Method for Comparing Viral Growth

Curves. Journal of Virological Methods, 135, 118-123.

WAT, D. 2004. The Common Cold: A Review of the Literature. European Journal of

Internal Medicine, 15, 79 - 88.

WEBER, M. W., MULHOLLAND, E. K. & GREENWOOD, B. M. 1998. Respiratory

Syncytial Virus Infection in Tropical and Developing Countries. Tropical Medicine

and International Health, 3, 268-280.

WIDJOJOATMODJO, M. N., BOES, J., VAN BERS, M., VAN REMMERDEN, Y.,

ROHOLL, P. J. M. & LUYTJES, W. 2010. A Highly Attenuated Recombinant

Human Respiratory Syncytial Virus Lacking the G Protein Induces Long-Lasting

Protection in Cotton Rats. Virology Journal, 7: 114

Page 158: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

145

WILLIAMS, J. V., HARRIS, P. A., TOLLEFSON, S. J., HALBURNT-RUSH, L. L.,

PINGSTERHAUS, J. M., EDWARDS, K. M., WRIGHT, P. F. & CROWE-JR., J. E.

2004. Human Metapneumovirus and Lower Respiratory Tract Disease in Otherwise

Healthy Infants and Children. The New England Journal of Medicine, 350, 443 - 450.

WILLIAMS, J. V. & CROWE JR., J. E. 2007. Paramyxoviruses: Measles Virus and

Respiratory Syncytial Virus. In: ENGLEBERG, N. C., DIRITA, V. & DERMODY,

T. S. (eds.) Schaechter's Mechanisms of Microbial Disease. Baltimore: Lippincott

Williams & Wilkins.

WORLD HEALTH ORGANIZATION (W.H.O). 2011. Initiative for Vaccine Research

(IVR): Acute Respiratory Infections [Online]. Available:

http://www.who.int/vaccine_research/diseases/ari/en/index2.html.

WURM, T., CHEN, H., HODGSON, T., BRITTON, P., BROOKS, G. & HISCOX, J. A.

2001. Localization to the Nucleolus Is a Common Feature of Coronavirus

Nucleoproteins, and the Protein May Disrupt Host Cell Division. Journal of Virology,

75, 9345 - 9356.

YOO, D., WOOTTON, S. K., LI, G., SONG, C. & ROWLAND, R. R. 2003. Colocalization

and Interaction of the Porcine Arterivirus Nucleocapsid Protein with the Small

Nucleolar RNA-Associated Fibrillarin. Journal of Virology, 77, 12173 - 12183.

YOU, J., DOVE, B. K., ENJUANES, L., DEDIEGO, M. L., ALVAREZ, E., HOWELL, G.,

HEINEN, P., ZAMBON, M. & HISCOX, J. A. 2005. Subcellular Localisation of the

Severe Acute Respiratory Syndrome Coronavirus Nucleocapsid Protein. Journal of

General Virology, 86, 3303 - 3310.

ZAMANIAN-DARYOUSH, M., MOGENSEN, T. H., DIDONATO, J. A. & WILLIAMS, B.

R. G. 2000. NF-κB Activation by Double-Stranded-RNA-Activated Protein Kinase

(PKR) Is Mediated through NF-κB-Inducing Kinase and IκB Kinase. Molecular and

Cellular Biology, 20, 1278 - 1290.

Page 159: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

146

Page 160: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

147

APPENDIX

8 Appendix

Page 161: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

148

Page 162: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

1

Appendix: Confirmation of hMPV N constructs ligated in the entry and expression clones by DNA sequencing

10 20 30 40 50 60 70 80 90 100

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 ATGTCTCTTCAAGGGATTCACCTGAGTGATCTATCATACAAGCATGCTATATTAAAAGAGTCTCAGTATACAATAAAGAGAGATGTAGGCACAACAACAG

pDONR_hMPVN TCA.................................................................C...............................

pDONR_hMPVN-frA TCA.................................................................C...............................

pDONR_hMPVN-frB TCA.................................................................C...............................

pDONR_hMPVN-frC ----------------------------------------------------------------------------------------------------

pEPI_hMPVN TCA.................................................................C...............................

pEPI_hMPVN-frA TCA.................................................................C...............................

pEPI_hMPVN-frB TCA.................................................................C...............................

pEPI_hMPVN-frC ----------------------------------------------------------------------------------------------------

110 120 130 140 150 160 170 180 190 200

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 CAGTGACACCCTCATCATTGCAACAAGAAATAACACTATTGTGTGGAGAAATTCTATATGCTAAGCATGCTGATTACAAATATGCTGCAGAAATAGGAAT

pDONR_hMPVN .....................................G..............................A...............................

pDONR_hMPVN-frA .....................................G..............................A...............................

pDONR_hMPVN-frB .....................................G..............................A...............................

pDONR_hMPVN-frC ----------------------------------------------------------------------------------------------------

pEPI_hMPVN .....................................G..............................A...............................

pEPI_hMPVN-frA .....................................G..............................A...............................

pEPI_hMPVN-frB .....................................G..............................A...............................

pEPI_hMPVN-frC ----------------------------------------------------------------------------------------------------

210 220 230 240 250 260 270 280 290 300

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 ACAATATATTAGCACAGCTCTAGGATCAGAGAGAGTACAGCAGATTCTAAGAAACTCAGGCAGTGAAGTCCAAGTGGTTTTAACCAGAACGTACTCCTTG

pDONR_hMPVN ........................G................................................AC.....................T...

pDONR_hMPVN-frA ........................G................................................AC.....................T...

pDONR_hMPVN-frB ........................G................................................AC.....................T...

pDONR_hMPVN-frC ----------------------------------------------------------------------------------------------------

pEPI_hMPVN ........................G................................................AC.....................T...

pEPI_hMPVN-frA ........................G................................................AC.....................T...

pEPI_hMPVN-frB ........................G................................................AC.....................T...

pEPI_hMPVN-frC ----------------------------------------------------------------------------------------------------

49

Page 163: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

150

310 320 330 340 350 360 370 380 390 400

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 GGGAAAGTTAAAAACAACAAAGGAGAAGATTTACAGATGTTAGACATACACGGAGTAGAGAAAAGCTGGGTGGAAGAGATAGACAAAGAAGCAAGAAAAA

pDONR_hMPVN .................T.........................................A........................................

pDONR_hMPVN-frA .................T.........................................A........................................

pDONR_hMPVN-frB .................T.........................................A........................................

pDONR_hMPVN-frC ----------------------------------------------------------------------------------------------------

pEPI_hMPVN .................T.........................................A........................................

pEPI_hMPVN-frA .................T.........................................A........................................

pEPI_hMPVN-frB .................T.........................................A........................................

pEPI_hMPVN-frC ----------------------------------------------------------------------------------------------------

410 420 430 440 450 460 470 480 490 500

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 CAATGGCAACTTTGCTTAAAGAATCATCAGGCAATATTCCACAAAATCAGAGGCCTTCAGCACCAGACACACCTATAATCTTATTATGTGTAGGTGCCTT

pDONR_hMPVN .............A......................................................................................

pDONR_hMPVN-frA .............A................................................................................C.....

pDONR_hMPVN-frB .............A......................................................................................

pDONR_hMPVN-frC ----------------------------------------------------------------------------------------------------

pEPI_hMPVN .............A......................................................................................

pEPI_hMPVN-frA .............A................................................................................C.....

pEPI_hMPVN-frB .............A......................................................................................

pEPI_hMPVN-frC ----------------------------------------------------------------------------------------------------

510 520 530 540 550 560 570 580 590 600

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 AATATTTACCAAACTAGCATCAACTATAGAAGTGGGATTAGAGACCACAGTCAGAAGAGCTAACCGTGTACTAAGTGATGCACTCAAAAGATACCCTAGG

pDONR_hMPVN ...................................................................................................A

pDONR_hMPVN-frA ...................................................................................................A

pDONR_hMPVN-frB ................................................................................................----

pDONR_hMPVN-frC ----------------------------------------------------------------------------------------------------

pEPI_hMPVN ...................................................................................................A

pEPI_hMPVN-frA ...................................................................................................A

pEPI_hMPVN-frB ................................................................................................----

pEPI_hMPVN-frC ----------------------------------------------------------------------------.......................A

Page 164: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

151

610 620 630 640 650 660 670 680 690 700

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 ATGGACATACCAAAAATCGCTAGATCTTTCTATGATTTATTTGAACAAAAAGTGTATTACAGAAGTTTGTTCATTGAGTATGGCAAAGCATTAGGCTCAT

pDONR_hMPVN .................T........C........C................................................................

pDONR_hMPVN-frA .................T........C........C................................................................

pDONR_hMPVN-frB ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frC .................T........C........C................................................................

pEPI_hMPVN .................T........C........C................................................................

pEPI_hMPVN-frA .................T........C........C................................................................

pEPI_hMPVN-frB ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frC .................T........C........C................................................................

710 720 730 740 750 760 770 780 790 800

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 CCTCTACAGGCAGCAAAGCAGAAAGTTTATTCGTTAATATATTCATGCAAGCTTACGGTGCTGGTCAAACAATGCTGAGGTGGGGAGTCATTGCCAGGTC

pDONR_hMPVN .......................................................T.............................G.....C........

pDONR_hMPVN-frA .......................................................T...............-----------------------------

pDONR_hMPVN-frB ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frC .......................................................T.............................G.....C........

pEPI_hMPVN .......................................................T.............................G.....C........

pEPI_hMPVN-frA .......................................................T...............-----------------------------

pEPI_hMPVN-frB ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frC .......................................................T.............................G.....C........

810 820 830 840 850 860 870 880 890 900

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 ATCTAACAATATAATGTTAGGACATGTATCTGTCCAAGCTGAGTTAAAACAAGTCACAGAAGTCTATGACCTGGTGCGAGAAATGGGCCCTGAATCTGGG

pDONR_hMPVN .................................G.....C.....G.....G....................A........G..................

pDONR_hMPVN-frA ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frB ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frC .................................G.....C.....G.....G....................A........G..................

pEPI_hMPVN .................................G.....C.....G.....G....................A........G..................

pEPI_hMPVN-frA ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frB ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frC .................................G.....C.....G.....G....................A........G..................

Page 165: Wahyu Nawang Wulan - canberra.edu.au · iv following single cycle and multiple cycle replication assays. To define hMPV N nucleocytoplasmic transport, full-length and truncated hMPV

152

910 920 930 940 950 960 970 980 990 1000

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 CTCCTACATTTAAGGCAAAGCCCAAAAGCTGGACTGTTATCACTAGCCAATTGTCCCAACTTTGCAAGTGTTGTTCTCGGCAATGCCTCAGGCTTAGGCA

pDONR_hMPVN ....................T....................CT......................................G..................

pDONR_hMPVN-frA ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frB ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frC ....................T....................CT......................................G..................

pEPI_hMPVN ....................T....................CT......................................G..................

pEPI_hMPVN-frA ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frB ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frC ....................T....................CT......................................G..................

1010 1020 1030 1040 1050 1060 1070 1080 1090 1100

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 TAATAGGTATGTATCGCGGGAGAGTGCCAAACACAGAACTATTTTCAGCAGCAGAAAGCTATGCCAAGAGTTTGAAAGAAAGCAATAAAATTAACTTTTC

pDONR_hMPVN ................A...................................C.............................T........C........

pDONR_hMPVN-frA ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frB ----------------------------------------------------------------------------------------------------

pDONR_hMPVN-frC ................A...................................C.............................T........C........

pEPI_hMPVN ................A...................................C.............................T........C........

pEPI_hMPVN-frA ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frB ----------------------------------------------------------------------------------------------------

pEPI_hMPVN-frC ................A...................................C.............................T........C........

1110 1120 1130 1140 1150 1160 1170 1180

....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|....|

N_Can97-83 TTCATTAGGACTCACAGATGAAGAAAAAGAGGCTGCAGAACACTTTCTAAATGTGAGTGACGACAGTCAAAATGATTATGAGTAA

pDONR_hMPVN .............................................C.......................................

pDONR_hMPVN-frA -------------------------------------------------------------------------------------

pDONR_hMPVN-frB -------------------------------------------------------------------------------------

pDONR_hMPVN-frC .............................................C.......................................

pEPI_hMPVN .............................................C.......................................

pEPI_hMPVN-frA -------------------------------------------------------------------------------------

pEPI_hMPVN-frB -------------------------------------------------------------------------------------

pEPI_hMPVN-frC .............................................C.......................................

Legends: Full-length and truncated hMPV N constructs were cloned into entry (pDONR207) and expression (pEPI-DESTC) vectors and sequenced as described in section 2.2.7.2. Sequences resulted in this study were compared to the reference N gene of hMPV Can97-83 (GenBank accession number AY145278). Dots (…) = residues similar to reference

Dash (---) = regions that cannot be sequenced using primers that were used in this study