Effect of Rhinovirus Infection on the Host Apoptotic · PDF fileEffect of Rhinovirus Infection...

83
Effect of Rhinovirus Infection on the Host Apoptotic Response Kylia Merinda Wall B. Med. Sci Centre for Research in Therapeutic Solutions University of Canberra ACT 2601 A thesis submitted in partial fulfilment of the requirements for the degree of Bachelor of Applied Science (Honours) at the University of Canberra October 2013

Transcript of Effect of Rhinovirus Infection on the Host Apoptotic · PDF fileEffect of Rhinovirus Infection...

Effect of Rhinovirus Infection on the Host

Apoptotic Response

Kylia Merinda Wall

B. Med. Sci

Centre for Research in Therapeutic Solutions

University of Canberra ACT 2601

A thesis submitted in partial fulfilment of the requirements

for the degree of Bachelor of Applied Science (Honours) at

the University of Canberra

October 2013

ii

© Kylia Merinda Wall 2013

iii

Abstract

The human rhinoviruses (HRV) are the most common viral cause of upper respiratory

tract infections and are known to cause asthma exacerbations which may have serious

consequences. One of the host’s defences against viral infection is the induction of

apoptosis, a form of programmed cell death that acts to eliminate the virus with minimal

impact on surrounding cells. It is thought that HRV may delay the induction of

apoptosis in infected cells in order to facilitate its own replication. Previous studies have

shown that some picornaviruses, including the closely related poliovirus, are capable of

regulating induction of the apoptotic pathway.

This study aimed to identify the effect of HRV infection on the apoptotic pathway of

infected cells. Ohio-HeLa cells infected with HRV16 of varying multiplicity of

infections (MOIs) and incubation periods analysed by western blot showed no evidence

of PARP, caspase 3 or caspase 9 cleavage, demonstrating that apoptosis was not

induced. Immunofluorescence assay showed that cytochrome c was not released from

the mitochondria of HRV16 infected cells suggesting that the intrinsic apoptotic

pathway is not induced, even at an early stage.

Investigation of the effect of HRV16 infection on cells treated with the chemical

inducer of apoptosis, Actinomycin D (Act. D), demonstrated that HRV16 may actively

suppress the host apoptotic pathway. A reduced number of apoptotic cells were

observed in cells infected prior to treatment when compared to cells treated with Act. D

alone. It was found that HRV16 infection resulted in the cleavage of the extrinsic

apoptosis intermediate RIPK1 dissimilar to that seen during regular apoptotic induction.

It was found that interestingly, the HRV16 proteases 2A and 3C do not appear to be

responsible for this cleavage.

These results suggest that not only does HRV16 infection avoid apoptotic death, but

that it is capable of actively suppressing the host apoptotic pathways, potentially

through the inhibition of the extrinsic pathway via the indirect alternate cleavage of

RIPK1.

v

Acknowledgements

I would like to express my deep appreciation and gratitude to my supervisor Dr. Reena

Ghildyal. She has provided me with endless support and encouragement throughout and

she has pushed me to develop far beyond my expectations.

I have received generous help, support and encouragement from everyone in CResTS in

particular the RVG team, Dr. Erin Walker, Lora Jensen, Dr. Deborah Heydet and Robert

McCuaig.

I would also like to acknowledge Dr. Scott Bowden of VIDRL Melbourne and Dr. Wai-

Ming Lee of University of Wisconsin for their kind donation of antibodies used

throughout this study.

Lastly I would like to thank my husband Josh for all of his support, patience and

encouragement.

vi

Table of Contents

Abstract ........................................................................................................................................ iii

Acknowledgements ....................................................................................................................... v

List of figures ............................................................................................................................. viii

List of tables ................................................................................................................................. ix

1 Chapter One - Introduction ................................................................................................... 1

1.1 Introduction ................................................................................................................... 1

1.2 Picornaviruses ............................................................................................................... 1

1.2.1 Structure ................................................................................................................ 3

1.2.2 Genome ................................................................................................................. 3

1.2.3 Polyprotein and Protein Processing ...................................................................... 4

1.2.4 Viral Attachment and Entry .................................................................................. 6

1.2.5 Viral Replication ................................................................................................... 8

1.2.6 Viral Assembly and Exit ....................................................................................... 8

1.2.7 Viral Effect on Cellular Processes ...................................................................... 10

1.3 Rhinovirus ................................................................................................................... 12

1.3.1 Classification ....................................................................................................... 12

1.3.2 Epidemiology ...................................................................................................... 13

1.3.3 Clinical Disease................................................................................................... 14

1.4 Apoptosis .................................................................................................................... 15

1.4.1 General Apoptosis ............................................................................................... 15

1.4.2 Viruses and Apoptosis ........................................................................................ 18

1.4.3 Picornaviruses and Apoptosis ............................................................................. 20

1.4.4 Rhinovirus and Apoptosis ................................................................................... 22

1.5 Hypothesis and Aims of Present Study ....................................................................... 24

1.5.1 Hypothesis ........................................................................................................... 24

1.5.2 Aims .................................................................................................................... 24

2 Chapter Two – Materials and Methods ............................................................................... 26

2.1 Materials ..................................................................................................................... 26

2.1.1 Virus and Cell Culture Lines .............................................................................. 26

2.1.1.3 Media Used for Cell Culture and Viral Infection........................................................ 26

2.1.2 Buffers, Solutions and Bacterial Culture Media ................................................. 27

2.1.3 Antibodies ........................................................................................................... 28

2.1.4 Commercial Kits ................................................................................................. 29

2.2 Methods....................................................................................................................... 29

2.2.1 Cell and Virus Cultures ....................................................................................... 29

vii

2.2.2 Analysis ............................................................................................................... 32

3 Chapter Three – Results ...................................................................................................... 34

Rhinovirus Infection Does Not Induce Early or Late Intrinsic Apoptosis .................................. 34

3.1 Introduction ................................................................................................................. 34

3.2 HRV16 Infection Does Not Result in Cleavage of PARP .......................................... 35

3.3 Rhinovirus Serotype 16 Infection Does Not Induce Late Apoptosis .......................... 36

3.4 RV16 Infection Does Not Induce Early Intrinsic Apoptosis ...................................... 40

3.5 Taxol Induces Apoptosis in Ohio-HeLa Cells ............................................................ 43

3.6 Actinomycin D Induces Apoptosis in Ohio-HeLa Cells ............................................. 45

3.7 Summary ..................................................................................................................... 49

4 Chapter Four – Results ........................................................................................................ 50

Effect of HRV16 Infection on Chemically Induced Apoptosis .................................................. 50

4.1 Introduction ................................................................................................................. 50

4.2 Effect of HRV16 Infection on Taxol Induced Apoptosis ........................................... 50

4.3 Effect of HRV16 Infection on Act. D Induced Apoptosis .......................................... 53

4.4 Summary ..................................................................................................................... 57

5 Chapter Five – Results ........................................................................................................ 58

Rhinovirus Infection Leads to the Indirect Cleavage of RIPK1 ................................................. 58

5.1 Introduction ................................................................................................................. 58

5.2 HRV16 Infection and Act. D Treatment Lead to Dissimilar Cleavage of RIPK1 ...... 58

5.3 Cleavage of RIPK1 is Probably Not Carried Out By the 2A or 3C Proteases ............ 60

5.4 Expression of 2A and 3C Proteases Does Not Induce Apoptosis ............................... 61

5.5 Summary ..................................................................................................................... 63

6 Chapter Six – General Discussion ...................................................................................... 64

6.1 Introduction ................................................................................................................. 64

6.2 HRV Infection Does Not Induce Apoptosis................................................................ 64

6.3 HRV16 Inhibits Apoptosis .......................................................................................... 67

6.4 HRV16 Infection Leads to Indirect Cleavage of RIPK1 ............................................ 68

6.5 Conclusion .................................................................................................................. 69

References Cited ......................................................................................................................... 70

viii

List of figures

Figure 1.1 An example of the picornaviral genome……………………………………………...4

Figure 1.2 Post translational processing of the HRV polyprotein………………………………..5

Figure 1.3 An overview of the picornavirus lifecycle…………………………………………..10

Figure 1.4 Schematic diagram of the extrinsic and intrinsic apoptotic pathways as relevant to

picornavirus infection. ………………………………………………………………………….16

Figure 3.1 HRV16 infection does not lead to cleavage of PARP…………………………..…..36

Figure 3.2 Detection of late apoptosis during RV16 infection…………………………...……..39

Figure 3.3 Effect of varying MOI on the detection of late apoptosis during RV16 infection.….40

Figure 3.4 Cytochrome c is not released during RV16 infection…………………………….....42

Figure 3.5 Taxol treatment induces intrinsic apoptosis in Ohio-HeLa cells…………………....44

Figure 3.6 Taxol treatment leads to cytochrome c release from the mitochondria……………..45

Figure 3.7 Act. D treatment induces apoptosis in Ohio-HeLa cells………………………….…47

Figure 3.8 Act. D treatment leads to cytochrome c release from the mitochondria…………….48

Figure 4.1 Taxol treatment during HRV16 infection may reduce expression of VP2………….52

Figure 4.2 Taxol treatment during HRV16 infection may lead to a reduction in the translation of

the viral polyprotein…………………………………………………………………………….53

Figure 4.3 Act. D treatment during HRV16 infection does not reduce expression of VP2…….55

Figure 4.4 HRV16 infection reduces the induction of apoptosis in Act. D treated cells……….56

Figure 5.1 HRV16 and Act. D treatment leads to cleavage of RIPK1………………………….60

Figure 5.2 HRV16 2A and 3C proteases do not cleave RIPK1 or caspase 3…………………...62

ix

List of tables

Table 1.1 Cellular receptors utilised by various picornavirus species……………………...……7

Table 2.1 Cell and virus culture media used during this study………………………………....26

Table 2.2 Buffers and solutions used during this study………………………………………...27

Table 2.3 Western blot primary antibodies used during this study………………………..……28

Table 2.4 Western blot secondary antibodies used during this study…………………………...28

Table 2.5 Immunofluorescence primary antibodies used during this study……………….……28

Table 2.6 Immunofluorescence secondary antibodies used during this study………………….29

Table 2.7 Commercial kits used during this study……………………………………………...29

1

1 Chapter One - Introduction

1.1 Introduction

Human rhinovirus (HRV), a member of the Enterovirus genus of family Picornaviridae,

is the most common viral cause of upper respiratory infections and is known to be

associated with asthma and chronic obstructive pulmonary disease (COPD)

exacerbations which may result in serious complications, even death (Johnston et al.,

1995; Seemungal et al., 2000). Picornaviruses are small, non-enveloped RNA viruses

which feature a single strand of positive sense RNA surrounded by an outer capsid.

They are responsible for a very wide variety of diseases in humans and other vertebrates

including poliomyelitis, liver disease, cardiomyelitis, respiratory illness and foot-and-

mouth disease. One of the host’s defences against viral infections is the induction of

apoptosis via a range of virus associated triggers. Previous studies investigating the

induction of apoptosis during picornavirus infections have shown evidence that the

induction of apoptosis can be either promoted or suppressed under various conditions or

at different time points post infection. It is hypothesised that HRV may delay the

induction of apoptosis in order to facilitate its own replication. Previous studies have

shown that Enterovirus C (poliovirus), another member of the Enterovirus genus of the

Picornaviridae family which is very closely related to HRV, is capable of delaying the

induction of the apoptotic pathway under optimal viral replication conditions whilst also

showing evidence of promoting apoptosis induction when conditions are not ideal.

These results, and the similarities seen between HRV and poliovirus, support the

hypothesis that the HRV is also capable of altering the host apoptotic response for its

own advantage.

1.2 Picornaviruses

The Picornaviridae are a family of viruses belonging to the Picornavirales order of

vertebrate viruses. The picornavirus family is made up of twelve genera including

Aphthoviruses, Avihepatoviruses, Cardioviruses, Enteroviruses, Erboviruses,

Hepatoviruses, Kobuviruses, Parechoviruses, Sapeloviruses, Senecaviruses,

Teschoviruses and Tremoviruses (King et al., 2012). Currently, 29 species of viruses

2

have been identified as belonging within these genera; these are further differentiated

into a large number of viral serotypes (King et al., 2012). There are many medically and

socially important viruses belonging to the Picornaviridae family, including

Enterovirus C (poliovirus (Carstens and Ball, 2009)), Hepatitis A virus (HAV), Foot-

and-mouth disease virus (FMDV), Encephalomyocarditis virus (EMCV), the

Coxsackieviruses (CV) and HRV (Racaniello, 2007; King et al., 2012).

The picornaviral species that have been formally identified to date are responsible for

causing a very broad range of disease and symptoms in vertebrates including humans

and animals such as bovine and swine. The most extensively studied of these,

poliovirus, is a member of the Enterovirus genus of viruses and is responsible for

causing the severely debilitating illness poliomyelitis (Racaniello, 2007; Rhoades et al.,

2011; Minor, 2012). Poliomyelitis is caused by infection of the motor neurons of the

central nervous system and often results in severe disability (Bodian, 1955; Racaniello,

2007; Minor, 2012). Other picornaviruses capable of infecting humans may cause

disease of the liver, as is the case with the Hepatovirus HAV, severe myocarditis, as is

the case with the Enterovirus CV, or in stark contrast, HRV causes a mild respiratory

illness that is most commonly known as the common cold (Racaniello, 2007; Rhoades

et al., 2011). The most notable of the illnesses caused in animals as a result of

picornavirus infection is foot and mouth disease caused by FMDV, which affects

livestock and has had substantial economic impacts worldwide (Racaniello, 2007).

Associated with the highly varied range of illnesses caused by the picornavirus family,

these viruses utilise highly varied modes of infection. Of the human associated viruses,

the respiratory system, digestive system, circulatory and central nervous systems are all

targets, with some viruses capable of targeting multiple systems upon infection

(Racaniello, 2007). For example, poliovirus infection is primarily transmitted via the

faecal-oral route, particularly in developing countries with poor hygiene and sanitation

facilities where it can be transmitted through person to person contact or through

ingestion of contaminated food and water (Bodian, 1955; Pallansch & Roos, 2007;

Minor, 2012). Poliovirus replicates very efficiently within the intestinal tract and is shed

via faeces (Bodian, 1955; Pallansch & Roos, 2007; Minor, 2012). Similarly, HAV is

also transmitted via the faecal-oral route through person to person contact due to poor

hygiene or through ingestion of contaminated food and water (Hollinger & Emerson,

2007). As their name suggests, the HRV’s infect the nasopharyngeal region of the

respiratory system. Human rhinoviruses are acid labile and are therefore unable to

3

penetrate and infect via the digestive system, their primary route of infection is via

direct person to person contact with nasal secretions containing very high levels of virus

particles (Racaniello, 2007).

1.2.1 Structure

Picornavirus virions are simple, spherical structures with a diameter of approximately

30nm (Racaniello, 2007). The picornavirus capsids are made up of four structural viral

proteins, VP1, VP2, VP3 and VP4, with the exception of the Parechovirus genus of

viruses, whose capsids contain the VP1 and VP3 proteins as well as VP0, the precursor

protein of VP2 and VP4 (Stanway & Hyypia, 1999; Racaniello, 2007; Minor, 2012).

These four structural capsid proteins fit together through the formation of protomers to

form an icosahedral shaped sphere of approximately 60 proteins, where VP1, VP2 and

VP3 are present on the surface of the capsid whilst VP4 is present on the inner surface

(Racaniello, 2007; Minor, 2012). The capsids of picornaviruses differ slightly in their

surface shape dependant on the cellular receptor they utilise to allow for optimal

receptor-virion interaction (Racaniello, 2007).

1.2.2 Genome

The genomic RNA of picornaviruses consists of a single strand of uncapped positive

sense RNA which is linked at its 5’ end to a VPg (virion protein, genome linked)

protein, as illustrated in Figure 1.1. (Nomoto et al., 1976; Novak & Kirkegaard, 1999;

Racaniello, 2007; Minor, 2012). VPg proteins, which vary from around 22 to 24 amino

acid residues in length, are essential for the synthesis of both positive and negative

stranded RNA, with the protein found to be linked to both forms (Pettersson et al.,

1978; Rhoades et al., 2011). It is thought that VPg is cleaved upon entry of the genomic

RNA into the cytoplasm by the host cell unlinking enzyme and is not required for viral

translation (Racaniello, 2007).

The single strand of picornavirus RNA contains a long non-coding region at the 5’ end

which is highly structured and contains features such as the internal ribosome entry site

(IRES). The IRES facilitates the binding of cellular ribosomes to the mRNA and allows

for its translation despite the absence of a 5’ cap structure (Racaniello, 2007). The 3’

4

non-coding region of both genomic and mRNA is attached to a poly(A) tail. Between

the 5’ and 3’ non-coding regions is a single continuous open reading frame (ORF),

which encodes a single polyprotein that is proteolytically cleaved by virally encoded

proteases to produce both the structural and non-structural viral proteins (see figures 1.1

& 1.2) (Racaniello, 2007; Grubman et al., 2008).

Figure 1.1 An example of the picornaviral genome. The genome consists of a single strand of positive sense RNA with a poly(A) tail at its' 3' end and a VPg

protein linked to its' 5' end. The RNA contains a single ORF flanked by two highly structured non-coding

regions. The ORF encodes a single polyprotein which undergoes proteolytic cleavage in the host cell to

produce the various functional viral proteins. The leader protein (L) is not present in all species but is

present in FMDV and EMCV. The remainder of the proteins encoded as part of the polyprotein can be

divided into structural and non-structural proteins with the VP1, VP2, VP3 and VP4 proteins making up

the structural capsid proteins and the remainder of the proteins included in the non-structural proteins. Of

the non-structural proteins, 2A and 3C have protease capabilities in most picornaviruses and are

responsible for the cleavage of the polyprotein into the various active viral proteins.

1.2.3 Polyprotein and Protein Processing

The picornavirus polyprotein is cleaved via two internally expressed proteases, 2A and

3C. Cleavage starts concurrently with translation and begins with the autocatalytic

cleavage of 2A. The self-cleavage of 2A forms the first of the primary cleavages and

separates the P1 section of the polyprotein, containing the structural capsid proteins,

from the P2/P3 section containing the remainder of the non-structural viral proteins

(refer to figure 1.2)(Ypma-Wong & Semler, 1987). Following the cleavage of 2A, the

remaining polyprotein cleavages are carried out by the 3C protease either on its own, or

whilst in its precursor state as the 3CD protein (Patick & Potts, 1998; Racaniello, 2007).

Similarly to 2A, the 3C protease cleaves itself from the polyprotein, initially forming

the 3CD precursor protein before finally completing its autocatalytic cleavage to result

in the 3C and 3D proteins. Other cleavages carried out by the 3C protease include the

cleavage of P1 into each of the structural capsid proteins as well as the cleavage of the

remaining non-structural proteins (figure 1.2)(Ypma-Wong & Semler, 1987). There is a

high level of conservation across the picornaviral proteins encoded by each of the viral

family members, with the non-structural proteins showing the highest level of

conservation (Buenz & Howe, 2006).

5

Figure 1.2 Post translational processing of the HRV polyprotein.

Translation of the HRV RNA results in the production of a single polyprotein which undergoes a series of

cleavages to produce the functional viral proteins. Primary cleavage of the polyprotein results in the

production of P1, P2 and P3 proteins with P1 containing the structural capsid proteins. The first of the

primary cleavages is achieved via the autocatalytic cleavage of the 2A protein releasing the P1 section

from the remaining P2/P3 section. The remainder of the cleavages are carried out by the 3C protein

including the autocatalytic cleavage of itself, the cleavage of P1 to produce the capsid proteins and the

cleavage of each of the remaining proteins to form both the active secondary cleaved proteins and the

further cleaved final protein products.

The picornaviral proteases have been shown to play a role in many aspects of viral

infection from the proteolytic processing of the viral proteins, to the shutting off of host

cell processes, including host RNA transcription. Once cleaved from the polyprotein,

the 2A protease of some picornaviruses, including HRV, has been shown to be

responsible for cleavage of the eukaryotic initiation factors eIF4GI and eIF4GII (Gradi

et al., 1998; Gradi et al., 2003). Cleavage of these factors contributes to the shut-off of

host cell translation, freeing the host machinery to focus on viral mRNA translation. In

a study carried out by Carocci et al, it was found that the 2A protein of EMCV is

required for the inhibition of apoptosis in BHK-21 cells, despite the 2A protein of

cardioviruses and aphthoviruses, including EMCV, not being a protease (Buenz &

Howe, 2006; Carocci et al., 2011). Cells infected with wild type EMCV did not show

signs of apoptosis induction, whereas virus containing a 2A deletion appeared to induce

apoptosis via the activation of caspases. These results suggest that the 2A protein of

EMCV is involved in the inhibition or suppression of apoptosis induction (Buenz &

6

Howe, 2006). Cells infected with the altered EMCV were also observed to result in an

accumulation of virus particles within the cytoplasm, indicating that EMCV 2A may

play a role in the release of EMCV from the cell (Buenz & Howe, 2006).

The 3C protease of picornaviruses is responsible for the majority of the processing

cleavages of the viral polyprotein. 3C has also been shown to be responsible for a

number of cellular alterations including alteration of the host cell nucleocytoplasmic

transport pathways (Gustin & Sarnow, 2002). It has been observed that the 3C protease

of a number of picornaviruses is capable of inducing apoptosis when expressed in cell

lines. For example, poliovirus 3C protease induces apoptosis through a caspase

dependant pathway when expressed in HeLa cells (Barco et al., 2000; Buenz & Howe,

2006). Similar results were shown in the human glioblastoma SF268 cells expressing

the 3C protease of the human enterovirus 71 (Li et al., 2002).

1.2.4 Viral Attachment and Entry

The extensive variation of viruses within the picornavirus family requires the utilisation

of a number of different cellular receptors for viral infection (summarised in table 1.1).

Some of the receptors utilised by these viruses include immunoglobulin-like receptors

such as the poliovirus receptor (Pvr) and the major group rhinovirus receptor,

intercellular adhesion molecule 1 (ICAM-1). Others include integrin receptors such as

αvβ3, a receptor used by various CV strains (Racaniello, 2007). This receptor variation

extends to the species level, where for example, HRV serotypes are grouped based on

their cellular receptors, with the major group of rhinoviruses utilising ICAM-1 whilst

the minor group of rhinoviruses use members of the low density lipoprotein receptor

family (Tomassini et al., 1989; Hofer et al., 1994). As well as showing variability in the

selection of receptors required for cellular entry, many viruses within this family also

require the presence of co-receptors for effective infection. A selection of echoviruses,

including echoviruses 3, 6 and 7, not only require the presence of the decay-accelerating

factor (CD55) as their receptor, but also the presence of a β2-microglobulin co-receptor

for entry into the host cell (Racaniello, 2007).

As a result of the variation in cellular receptors, each picornavirus species shows slight

variation in the surface of their capsids. For example, poliovirus and HRV have a

groove within each of the protomers (see section 1.2.6) that make up their capsids. It is

7

within this groove that the virus-receptor interactions take place. In contrast, the

aphthoviruses and cardioviruses do not exhibit such groove structures on their capsid

surfaces (Racaniello, 2007).

Table 1.1 Cellular receptors utilised by various picornavirus species

Virus Receptor

Human enterovirus C Poliovirus receptor (Pvr)

Human rhinovirus – major

group

Intercellular adhesion molecule 1 (ICAM-1)

Human rhinovirus – minor

group

Low density lipoprotein receptor family (LDLR)

Coxsackievirus αvβ3 receptor, CD55, ICAM-1

Foot and mouth disease virus αvβ3 receptor

Human hepatitis A virus Human hepatitis A virus cellular receptor (HAVcr-1)

After attachment has occurred, viral RNA is released from within the capsid and enters

the cytoplasm of the host (illustrated in figure 1.3). Whilst it is not fully understood

exactly how this occurs, two possible methods have been proposed. One proposed

method of picornaviral RNA entry into the cytoplasm is through the formation of a pore

within the plasma membrane of the host cell (Racaniello, 2007). In this method,

interactions between the virus particle and the cellular receptor result in conformational

changes to the viral capsid and lead to the formation of a pore within the plasma

membrane (Racaniello, 2007). This has been identified as being the method through

which poliovirus and the major group HRVs are thought to enter the cell (Perez &

Carrasco, 1993; Schober et al., 1998). Though it is not known if the RNA passes

straight through the pore into the cytoplasm or whether it enters though the host’s

endocytic pathway, it is believed that endocytosis alone is not sufficient for the RNA of

polioviruses to enter the cell (Racaniello, 2007). The other proposed method, through

which EMCV is thought to enter the cell, involves receptor mediated endocytosis,

where the binding of the virus to the receptor simply acts to bring the virus into the

vicinity of the cellular membrane to enable it to enter the endocytic pathway

(Racaniello, 2007). After the initial steps of viral entry, additional variation between the

picornavirus species results from the way in which the viral RNA is released from

within the endosomes into the cytoplasm of the host cell. An example of this can be

8

seen with the differences between the major group and the minor group HRVs (refer to

section 1.3.1). The RNA of the major group HRVs is released from endosomes through

via a disruption to the endosomal membrane caused by an increase in hydrophobicity

within the endosome. This increased hydrophobicity is caused by structural changes to

the viral capsid induced by the cellular receptor ICAM-1 (Racaniello, 2007). In contrast,

the minor group of rhinoviruses are released from within the endosome through pores

created within the endosomal membrane as a result of significant reductions of the

internal pH induced by the formation of the viral-cellular receptor complex (Racaniello,

2007).

1.2.5 Viral Replication

Synthesis of picornaviral RNA takes place within the cytoplasm of the host cell. It

occurs through the creation of a negative sense replicative intermediate strand, that is

then used as a template for the synthesis of positive stranded genomic RNA (illustrated

in figure1.3)(Racaniello, 2007). This synthesis pattern is strongly favoured towards the

synthesis of positive stranded RNA, with approximately 30-70 times more positive

sense RNA synthesised than negative sense (Novak & Kirkegaard, 1991; Racaniello,

2007). Viral RNA synthesis is carried out by a virally coded RNA polymerase, 3Dpol

,

which is cleaved from its precursor protein, the 3CD protease (see figure 1.2). Along

with the 3Dpol

, many other viral proteins have been shown to act as accessory proteins

in the synthesis of viral RNA, including the 2A, 2B and VPg proteins. However the

structural capsid proteins contained within the P1 section of the polyprotein are not

required for RNA synthesis (Racaniello, 2007).

1.2.6 Viral Assembly and Exit

Assembly of the viral capsid occurs following cleavage of the P1 protein from the viral

polyprotein, and the subsequent cleavage of the VP0, VP1 and VP3 structural proteins

by the 3CD protease (refer to figure 1.2) (Racaniello, 2007). Assembly of the capsid

starts with the formation of a protomer containing one copy each of the VP0, VP1 and

VP3 proteins. A pentamer is then formed from five protomers, with the pentamers then

self-assembling with the newly synthesized RNA to form a provirion (Reviewed by

Racaniello, 2007). The final step in viral assembly is the cleavage of the VP0 protein

9

resulting in the VP2 and VP4 proteins, which follows the packaging of newly

synthesised genomic RNA into the provirion. Packaging of viral RNA is highly specific

and ensures that only positive stranded RNA is packaged (Novak & Kirkegaard, 1991).

Once virions are formed, they exit the cell during cellular lyses or degradation resulting

from cytopathic effect or apoptosis. Interestingly in the case of poliovirus, it is thought

the virus utilises microtubules and cellular vesicles to exit the cell in the absence of

cellular lysis (Racaniello, 2007; Taylor et al., 2009).

The picornaviruses have short growth cycles, averaging approximately 8h in cell culture

(Buenz & Howe, 2006). The shedding of HRV particles peaks at 48-72h post infection

as evidenced by viral titres in nasal samples of infected patients (Harris & Gwaltney,

1996; Hendley & Gwaltney, 2004). This peak in viral shedding coincides with an

increase in the release of nasal secretions helping to facilitate the spread of the virus

beyond the host (Hendley & Gwaltney, 2004). Other picornaviruses including

poliovirus, CV, and HAV are shed via faeces, where their major route of transmission is

via the faecal-oral route (Pallansch & Roos, 2007).

10

Figure 1.3 An overview of the picornavirus lifecycle.

1. Viral particles attach to the relevant cell surface receptor, resulting in conformational changes allowing

the virus to enter the cell. 2. The virus enters the cell after attachment via either the formation of a pore

within the plasma membrane or endocytosis. 3. After entry, the viral RNA is released into the cytoplasm.

It is not yet fully understood how the RNA is released from the viral capsid. 4. RNA is translated within

the cytoplasm using host cell machinery to produce the viral polyprotein. 5. The translated polyprotein is

processed via protease cleavage using virally encoded proteases to produce the various functional viral

proteins. 6-7. After entering the cell, the viral RNA is replicated to form both negative and positive sense

strands with there being a far larger proportion of positive sense strands produced than the negative sense

template strands. 8. Following the translation and processing of the structural capsid proteins, they self-

assemble to form the viral capsid. The freshly replicated positive sense viral RNA is then packaged into

the newly formed capsid to form the viral progeny. 9. Viral progeny exit the cell likely helped by the

death and destruction of the host cell.

11

1.2.7 Viral Effect on Cellular Processes

As with most viruses, picornaviruses have developed numerous techniques to control or

disrupt host cellular pathways and machinery in order to enhance their survival and

virulence. Examples have been observed where viruses disrupt major cellular pathways

such as host translation, the secretory pathway and nucleocytoplasmic trafficking.

Various studies have shown that during picornavirus infection, a number of host

proteins associated with translation undergo cleavage. These include the translation

initiation factors eIF4G and eIF4A, the poly(A)-binding protein (PABP), and the

translation initiation factor eIF3 (Belsham et al., 2000; Grubman et al., 2008). Cleavage

of eIF4G and eIF4A leads to the inhibition of cap dependant translation, leaving the

host’s translation machinery free to carry out cap independent translation of the viral

mRNA (Belsham et al., 2000; Grubman et al., 2008). Cleavage of these factors also

contributes to the down regulation of host immune proteins (Grubman et al., 2008).

Proteases of enteroviruses, including poliovirus, cleave PABP, a host protein that plays

a major role in the initiation of cap-dependant translation initiation, potentially

contributing to the shut-off of host protein translation (Kuyumcu-Martinez et al., 2002).

In FMDV infection, it has been shown that the eIF4G proteins are cleaved by the viral

leader protease Lpro

(Buenz & Howe, 2006; Grubman et al., 2008), whereas the 3C

protease has been shown to induce at least a partial cleavage of this factor (Belsham et

al., 2000).

A number of picornaviruses, including poliovirus, coxsackievirus B3 and FMDV, have

been found to cause disruptions to the host cell secretory pathway during infection

(Grubman et al., 2008). FMDV infection has been shown to reduce the number of major

histocompatibility complex (MHC) class I molecules present on the surface of infected

cells, leading to a reduction in the presentation of viral antigens on the cell surface and

consequently a reduction in the induction of the cytotoxic T cell response (Grubman et

al., 2008). Another example of cellular processes being affected include the

identification that picornavirus infection results in the alteration of host

nucleocytoplasmic trafficking. Rhinovirus infection results in the accumulation of a

number of different proteins within the cytoplasm that are normally trafficked across the

nuclear membrane through the classical nuclear import pathway, suggesting this

pathway is inhibited by HRV infection (Gustin & Sarnow, 2002). Another cellular

12

impact of picornaviral infection is the alteration of the calcium homeostasis of the

organelles as demonstrated by Campanella et al (Campanella et al., 2004).

1.3 Rhinovirus

Common colds have been the subject of scientific studies for many years and were

initially thought to be caused by exposure to cold and damp conditions. The idea that

colds were not the result of exposure to cold and damp environments, but rather from

exposure to other infected persons, was first suggested in the 19th

century. This theory

was further developed in the early 20th

century when swabs of nasal secretions of

persons suffering from a cold were effectively able to infect others (Turner & Couch,

2007). Other early studies identified the significant role families played in the spread of

colds. Whilst preliminary studies had identified that there was a virus known to cause

common colds, it was not until the 1960’s that rhinoviruses were formally identified as

the causative virus (Turner & Couch, 2007). By the early 1970’s more than 100

different serotypes of HRV had been described (Turner & Couch, 2007).

1.3.1 Classification

The human rhinoviruses are members of the enterovirus genus of viruses within the

family Picornaviridae. There are over 100 HRV serotypes identified to date which have

been assigned to three genotypes, Rhinovirus A, Rhinovirus B and Rhinovirus C. The

criteria for differentiation of the HRV species from other Enterovirus species is their

acid lability where HRV is inactivated at low pH whilst other enteroviruses are not

(Turner & Couch, 2007). This difference in acid lability reflects the differences seen in

the locations within which infections occur. Rhinoviruses primarily infect the upper

respiratory tract whist other enteroviruses infect via the more acidic digestive tract.

Unlike other enteroviruses, HRV is highly species specific and only grows effectively in

human and a few primate cells. Chimpanzees and gibbons have both been successfully

infected with particular HRV serotypes, however these infections did not cause any

observable illness (Turner & Couch, 2007). This species specificity is caused by the

presence, or absence, of the cellular surface receptors required for HRV entry into the

cell. Three cellular receptors have been identified as being used by various HRV

13

serotypes, the decay-accelerating factor, low-density lipoprotein receptors (LDLR) and

intercellular adhesion molecule 1 (ICAM-1) (Tomassini et al., 1989; Hofer et al., 1994;

Turner & Couch, 2007). The majority of HRV serotypes can be separated into two

groups based on their cellular receptor usage, minor group HRV serotypes and major

group serotypes. The minor group includes the serotypes 1A, 1B, 2, 29, 30, 31, 44, 47,

49 and 62, all of which utilise the LDLR (Hofer et al., 1994; Turner & Couch, 2007).

The remaining HRV serotypes, with the exception of rhinovirus serotype 87 (HRV87),

make up the major group of HRVs and use ICAM-1 as their cellular receptor

(Tomassini et al., 1989; Turner & Couch, 2007). Unlike other serotypes, HRV87

appears to use the decay-accelerating factor as a receptor, similarly to the closely related

Enterovirus 68 (Turner & Couch, 2007).

1.3.2 Epidemiology

As demonstrated by the early investigations into colds, HRV infection is spread through

person to person contact, although infections via inanimate objects such as railings and

door handles and via respiratory aerosols are thought to be possible alternate routes of

transmission. Nasal secretions of HRV infected persons contain a high number of virus

particles, and it is thought that direct person to person contact with these secretions is

the most likely and most effective method of transmission (Hendley et al., 1973; Turner

& Couch, 2007).

The primary site of HRV infection is within the nasopharynx of the upper respiratory

tract. Experiments studying HRV infection routes have shown that the infectious dose

required for infection via nasal drops is significantly lower than that required for

infection via aerosol particles, indicating that the lower respiratory tract is much less

susceptible to HRV infection than the upper respiratory tract (Turner & Couch, 2007).

For a time it was thought that HRV was unable to infect the lower respiratory tract,

however this has since been shown to be incorrect. Despite the nasopharyngeal region

being the primary site of HRV infection, the virus is capable of infecting, and is often

found in, samples collected from the sinuses, the throat and from the middle ear (Turner

& Couch, 2007). Rhinovirus is rarely found in other sites within the body.

Even from the early days of studying the common cold, there has always been the

implication that being exposed to low temperatures or being chilled will induce a cold.

14

Contrary to this long held belief, exposure to cold and damp environments have not

been shown to have any effect on the susceptibility of individuals to colds, and

particularly to HRV infection (Turner & Couch, 2007). Rhinovirus infections occur

more frequently during autumn and spring in temperate climates, with the highest

frequencies occurring during autumn months, however infections do occur all year

round. It is not known why this pattern of infection occurs, however it is likely to be

multifactorial. Rhinovirus infections are far more prevalent in children and their

incidence reduces with age. Family units play a major role in the spread of HRV

infections due to members being in close proximity to each other and having a greater

level of contact with each other than in the general population.

1.3.3 Clinical Disease

Human rhinovirus infections are associated with a wide range of illnesses in addition to

the common cold. An association with asthma and chronic obstructive pulmonary

disease (COPD) exacerbations during HRV infection is well established (Johnston et al.,

1995; Seemungal et al., 2000; Greenberg, 2003). Rhinoviruses are the most common

causative viruses of the common cold, which results in symptoms such as nasal

congestions, sore throat, coughing, sneezing, headaches and general fatigue (Greenberg,

2003). Whilst these symptoms are often not severe and illness is usually resolved within

a few days, HRV infections are responsible for a significant number of absences from

workplaces and schools each year. They are also noted as being one of the most

common reasons for inappropriate use of antibiotics (Greenberg, 2003). These

consequences result in significant impacts both socially and economically. Respiratory

tract infections, including those caused by HRV, often result in more severe illness in

individuals suffering from asthma (Wark et al., 2005). Asthma attacks often occur after

respiratory infections, including those caused by HRV, and it has been shown that these

infections exacerbate asthma symptoms (Johnston et al., 1995; Greenberg, 2003). The

increased severity and exacerbations seen in asthma sufferers during HRV infection,

appear to be a result of inefficiencies in the asthmatic cell’s interferon and apoptotic

responses (Wark et al., 2005). Human rhinovirus infection has also been observed as

being responsible for illnesses such as acute otitis media and serious illness in those

suffering from COPD (Seemungal et al., 2000; Greenberg, 2003).

15

1.4 Apoptosis

1.4.1 General Apoptosis

Apoptosis is a mechanism of controlled cell death, used to eliminate unrequired or

compromised cells without releasing any of the cellular contents, thereby limiting any

potential damage to surrounding cells or triggering an inflammatory response.

Apoptosis can be induced in response to a wide range of triggers, including the removal

of unnecessary cells during foetal development, DNA damage and viral infection

(Barber, 2001). The apoptotic process allows for cells which have been triggered to

partake in apoptosis, to undergo morphological changes which permit them to be

engulfed by neighbouring cells or phagocytes. These morphological changes include

rounding of the cell, cytoplasmic shrinkage, nuclear fragmentation, chromatin

fragmentation and blebbing of the plasma membrane with the cells eventually breaking

down into apoptotic bodies (Barber, 2001; Galluzzi et al., 2008).

The process of apoptosis can be triggered by two distinct pathways, the intrinsic

apoptotic pathway and the extrinsic apoptotic pathway (briefly summarised in figure

1.4). The intrinsic pathway is mediated from within the cell via the mitochondria

(Galluzzi et al., 2008). The mitochondria receive pro- and anti-apoptotic signals passed

on from other organelles within the cell or from within the cytoplasm, resulting in

changes in the permeability of the mitochondrial membranes. In the event of pro-

apoptotic signals being received, the mitochondrial membrane permeability increases,

releasing pro-apoptotic proteins such as cytochrome c (Galluzzi et al., 2008). These

proteins go on to trigger a cascade of signalling events resulting in apoptosis (Galluzzi

et al., 2008). The extrinsic pathway however, is activated via signals external to the cell.

Specific ligands bind to pro-apoptotic receptors on the cell surface such as the

Fas/CD95 receptor and tumour necrosis factor receptors (TNFR-1 & TNFR-2) (Clement

& Stamenkovic, 1994; Micheau & Tschopp, 2003; Galluzzi et al., 2008). This

ligand/receptor binding event causes the formation of a death-inducing signalling

complex (DISC) which in turn triggers a cascade of events including the cleavage of

RIPK1 and caspase 8, ultimately leading to the controlled death of the cell (Micheau &

Tschopp, 2003; Galluzzi et al., 2008). Whilst these pathways are distinctly different, it

is known that they interact and activate each other and the two pathways converge at the

later stages of the cascades. It is also known that in some cases the mitochondria is able

16

to facilitate the activation of death receptors thus resulting in an internal activation of

the extrinsic pathway (Galluzzi et al., 2008).

Figure 1.4 Schematic diagram of the extrinsic and intrinsic apoptotic pathways as relevant to

picornavirus infection.

The extrinsic apoptotic pathway is initiated via the binding of the respective ligand to a pro-apoptotic

receptor on the surface of the plasma membrane. This triggers a signalling cascade that includes the

cleavage of caspase 8 and RIPK1 before the cleavage of the effector caspase 3 which leads to the

induction of the morphological changes characteristic of apoptosis. Induction of the intrinsic apoptotic

pathway is initiated from within the cell. Internal factors act on the mitochondria, increasing the

mitochondrial membrane permeability leading to the release of cytochrome c from within the

mitochondria into the cytoplasm. This leads to a signalling cascade that includes the cleavage of caspase

9. The intrinsic and extrinsic pathways converge as both include the cleavage of caspase 3 which leads to

the morphological changes.

17

Mechanisms behind the induction of apoptosis are complex and there are numerous

triggers and intermediates involved. It has been identified that interferons (IFNs) play a

major role in apoptosis as well as in triggering both the innate and adaptive immune

responses to viral infection (Barber, 2001). IFNs act in the induction of apoptosis

through their regulation of genes such as dsRNA-dependent protein kinase (PKR),

TNF-related apoptosis-inducing ligand (TRAIL) and the interferon regulatory factor

(IRF) family, which in turn utilise apoptosis as part of their antiviral and tumour

suppression functions (Gil & Esteban, 2000; Barber, 2001). Other important molecules

crucial for the apoptotic response include the caspases, the Bcl-2 family of proteins and

the tumour suppressor protein p53. Caspases are a group of enzymes that play a major

role in the apoptotic signalling cascade. They are constantly produced as procaspases by

the cell and are activated via cleavage, either autocatalytically or by upstream enzymes

as part of the apoptotic signalling cascade (Barber, 2001). Once caspases are cleaved

and activated, they in turn cleave specific downstream proteins through the recognition

of specific cleavage sites (Barber, 2001; Richard & Tulasne, 2012). There are

approximately 50 or more caspase substrates identified which when cleaved, act to

cause the characteristic morphological changes associated with apoptotic cells (Barber,

2001; Richard & Tulasne, 2012). For example the Bcl-2 family of proteins are a family

of proteins which act to regulate apoptosis by changing the membrane permeability of

the mitochondria (Galluzzi et al., 2008). The Bcl-2 family is composed of both

proapoptotic and antiapoptotic members that act on each other in response to various

triggers from within the cell, to provide a balance between the induction and inhibition

of apoptosis. This balance helps to prevent the unnecessary premature induction of cell

death (Galluzzi et al., 2008). The cellular expressed tumour suppressor protein p53 is

known to regulate cellular transcription in healthy cells, however it also has the ability

to induce apoptosis when the cell has been compromised, for example during virus

infection or after DNA damage (Barber, 2001). p53 acts to regulate the genes encoding

the Fas and TRAIL apoptotic receptors, as well as the transcription factor NF-κB, to

promote apoptosis (Barber, 2001). Schwarz et al have suggested that the transcription

factor NF-κB is involved in the suppression or delay of apoptosis during EMCV

infection through their studies of knockout mice (Schwarz et al., 1998). It has not been

determined if NF-κB is directly involved in apoptosis suppression or, in the more likely

scenario, that it is responsible for the expression of other antiapoptotic genes.

18

1.4.2 Viruses and Apoptosis

Apoptosis is one of the first cellular responses to viral infection as the cell aims to

eliminate the virus and restrict its replication, further spread and minimise the damage

caused (Barber, 2001; Richard & Tulasne, 2012). It is due to this fact that many viruses

have developed methods of altering the apoptotic pathways to their own advantage.

Viruses have been shown to not only inhibit or delay apoptosis induction in order to

facilitate the production of viral progeny, but have also been shown to have the ability

to induce apoptosis so as to help facilitate virus dissemination. Methods employed by

viruses to regulate apoptosis vary greatly and can affect all stages of the apoptotic

pathway, from the initiation of the signalling cascade right through to the triggering or

prevention of the morphological changes.

Viruses have found many ways of suppressing the host cell apoptotic pathway so as to

allow sufficient time for viral replication. Some viruses, including the human

adenoviruses (ADVs), encode proteins which are homologs of the cellular antiapoptotic

Bcl-2 family proteins that act to block the intrinsic apoptotic pathway (Galluzzi et al.,

2008). The ADV encoded E1B-19K Bcl-2 homolog has also been found to have the

ability to inhibit other apoptotic initiation pathways, including through the prevention of

ligand binding to the TNFR, Fas and TRAIL receptors on the cellular membrane surface

(Galluzzi et al., 2008). The receptor internalisation and degradation complex of ADVs

is capable of inhibiting the extrinsic apoptotic pathway by promoting the internalisation

and degradation of some of the major pro-apoptotic receptors, including TNFR and Fas,

found on the surface of the plasma membrane (Galluzzi et al., 2008).The BHRF1

product of the Epstein-Barr virus (EBV) is an antiapoptotic Bcl-2 protein homolog

which has been found to localise to the outer membrane of the mitochondria where it is

capable of suppressing the intrinsic induction of apoptosis by preventing the

permeabilisation of the mitochondrial membrane (Galluzzi et al., 2008).

The induction of apoptosis during viral infection may be of benefit to either the host or

the virus depending on the timing of induction. Some viruses have been shown to

induce apoptosis in cells as part of viral pathogenesis, for example a number of proteins

encoded by the Human immunodeficiency virus 1 (HIV-1) have been found to promote

an apoptotic response in CD4+ lymphocytes (Galluzzi et al., 2008). There have been a

number of viral proteins that have been identified as direct inducers of apoptosis, as

well as a number act indirectly to induce apoptosis. Additionally, a number of viral

19

proteins are capable of both directly and indirectly inducing the intrinsic pathway of

apoptosis through interactions with the mitochondria. The HIV-1 viral protein R (Vpr)

has been shown to directly interact with the voltage-dependant anion channels of the

outer mitochondrial membrane in order to trigger mitochondrial membrane permeability

and consequentially apoptosis (Galluzzi et al., 2008). The Influenza A virus (IAV)

encoded protein PB1-F2, has the ability to insert into the mitochondrial membranes

where it forms pores within the membranes similar to the proapoptotic Bax member of

the Bcl-2 family (Galluzzi et al., 2008). This pore formation by PB1-F2 is then thought

to increase mitochondrial membrane permeabilisation and ultimately the induction of

apoptosis (Galluzzi et al., 2008). Another example of a viral protein that has been

shown to have the ability to directly induce apoptosis is the hepatitis C virus (HCV)

encoded non-structural protein 4A (NS4A). This protein localises to the mitochondria

where it results in damage to the membranes and causes the release of cytochrome c

into the cytoplasm (Galluzzi et al., 2008). In contrast, the HIV-1 encoded protease

which is required for the processing of mature viral proteins, is able to indirectly induce

intrinsic apoptosis. It does so by promoting the cleavage of caspase 8 which leads to the

cleavage of Bid, a member of the Bcl-2 family, ultimately resulting in the

permeabilisation of the mitochondrial membranes and induction of intrinsic apoptosis

(Galluzzi et al., 2008).

The IFNs are known to play a role in the antiviral response to infection and appear to

increase the sensitivity of infected cells to the induction of apoptosis (Barber, 2001). It

is thought that IFNs are induced by the presence of double-stranded RNA (dsRNA),

which is present during the replication of RNA viruses. Once activated, they act to

increase the sensitivity of cells to the activation of Fas-associated death domain

(FADD) and caspase 8 dependant apoptosis (Barber, 2001). FADD is a molecule

recruited following activation of the proapoptotic death receptors of the extrinsic

pathway (Barber, 2001). The E7 protein of human papillomavirus (HPV) has been

shown to trigger the p53 mediated apoptotic response. However the virus, like many

others, has developed strategies to avoid this induction, including through the

expression of the E6 protein which inhibits the action of p53. Another example of viral

inhibition of the p53 mediated apoptosis is the production of the LANA protein by

human herpes virus 8 (HHV8) viruses which also acts to inhibit the action of the p53

protein (Barber, 2001).

20

1.4.3 Picornaviruses and Apoptosis

In the case of picornaviruses, there is evidence that this family of viruses are capable of

both inducing and inhibiting the apoptotic pathways of the host cells they infect

(reviewed by Buenz & Howe, 2006). Apoptosis triggered by picornaviral infection is

thought to be induced via the intrinsic apoptotic pathway and includes the activation of

the caspases 3 and 9 (Belov et al., 2003). The ability of picornaviruses to control the

balance of apoptosis during infection allows the virus enough time to replicate whilst

suppressing the host apoptotic response. It is thought that the viruses then deliberately

activate the apoptotic pathway to help facilitate the spread of the viral progeny, whilst

avoiding the activation of the host immune response. Despite the fact that some

members of the picornavirus family are capable of altering the apoptotic response, the

exact mechanisms and intermediates involved are yet to be fully understood and this is

an area of research that is becoming of great interest.

The suppression of apoptosis in picornaviral infected cells is thought to benefit the virus

by allowing for maximal replication before cell death occurs. In vitro studies of FMDV

infection have shown no evidence of apoptosis induction, however the mechanisms

behind this apparent suppression of apoptosis are still unknown (Grubman et al., 2008).

Through their studies in knockout mice, Schwarz et al. demonstrated that the

transcription factor NF-κB, may be involved in the suppression or delay of apoptosis

during EMCV infection (Schwarz et al., 1998). Further to this, Carocci et al. found that

the 2A protease also plays a role in the suppression of apoptosis during EMCV infection

(Carocci et al., 2011). Another study investigating apoptosis in EMCV infected cells,

found that apoptosis was not induced in infected HeLa cells, most likely due to actions

of the leader protein L (Romanova et al., 2009). This study also found that EMCV

infection was capable of not only circumventing the induction of host apoptosis, but is

also capable of inhibiting apoptosis induced by chemical apoptotic inducers (Romanova

et al., 2009). Studies investigating apoptosis during poliovirus infection observed that

apoptosis is inhibited during productive poliovirus infection (infection under conditions

optimal for maximum viral replication) of various HeLa sub-line cells, however it was

induced when conditions were not ideal (Tolskaya, et al., 1995; Agol et al., 2000; Belov

et al., 2003). Similarly to that seen in EMCV infection, poliovirus infection is also

capable of inhibiting chemically induced apoptosis (Tolskaya et al., 1995). In a study of

coxsackievirus B3, another member of the enterovirus genus, expression of the 2B

protein led to the inhibition of chemically induced apoptosis in HeLa cells through

21

alterations in the Ca2+

homeostasis of the cellular organelles (Campanella et al., 2004).

Whilst these studies have demonstrated that a number of picornaviruses are capable of

suppressing apoptosis induction, the exact mechanisms involved are yet to be fully

understood. It is possible that the inhibition or suppression of host cell apoptosis by

picornaviruses could be controlled by the virally encoded proteases, 2A and 3C. Their

ability to alter and shut-off other aspects of the host cell machinery, as well as the very

small number of virally encoded proteins, makes these proteases highly plausible targets

when trying to determine the mechanisms responsible for the suppression of apoptosis

during picornavirus infection. However, as demonstrated through the work of

Campanella et al, other viral components cannot be ruled out and it is possible that

multiple processes are involved (Campanella et al., 2004).

Various picornaviral components have been identified as being capable of inducing

apoptosis; however their exact mechanisms of action are yet to be elucidated. Studies

investigating the induction of apoptosis in poliovirus infected cells have shown that

virus infection is capable of both inhibiting and inducing apoptosis (Tolskaya et al.,

1995). It has been found that poliovirus may have the ability to induce apoptosis via

various different triggers which lead to the activation of caspases 3 and 9, following the

efflux of cytochrome c from the mitochondria (Belov et al., 2003). These apoptotic

triggers include the alteration of Bcl-2 family proteins, promotion of mitochondrial

membrane permeability by the viral proteins 2B and 3A and expression of the 2A and

3C proteases (Belov et al., 2003; Buenz & Howe, 2006; Galluzzi et al., 2008). Li et al.

found that the expression of the Enterovirus 71 3C protease in human glioblastoma

SF268 cells results in the induction of apoptosis through the activation of caspases,

however they also found that apoptosis was not induced in cells expressing a

deactivated mutant of the 3C protease (Li et al., 2002). Similar results have been

observed in studies examining poliovirus and HRV infection, where the 2A protease has

been demonstrated to induce a number of cellular alterations associated with the

induction of apoptosis including the fragmentation of DNA (Barco et al., 2000; Buenz

& Howe, 2006) Buenz and Howe 2006). Carthy et al. showed that infection of HeLa

cells with Coxsackievirus B3 (CVB3) resulted in the activation of a range of caspases,

including caspases 3 and 9, as well as the cleavage of the caspase substrate PARP,

indicating the induction of apoptosis (Carthy et al., 2003). In the same study,

significantly more cardiomyocytes were found to be apoptotic in patients infected with

enteroviruses such as coxsackievirus B than in cardiomyocytes from uninfected

22

patients. Another study of CV infection also confirmed that apoptosis is induced during

infections (Gomes et al., 2010). The structural VP3 protein and the non-structural 2C

protein encoded by the avian encephalomyelitis virus (AEV) have been found to

promote the induction of apoptosis via the intrinsic pathway (Galluzzi et al., 2008), with

the VP3 protein shown to localise to the mitochondria where it triggers the activation of

downstream caspases (Galluzzi et al., 2008).

The demonstration that picornaviruses have the ability to both induce and supress

apoptosis at various points post infection suggests that these viruses may have

developed strategies allowing them to alter and regulate the balance of pro-apoptotic

and anti-apoptotic tendencies within the cell in order to support their own replication

and spread.

1.4.4 Rhinovirus and Apoptosis

Similarly to all picornaviruses, the effect of HRV infection on the induction of

apoptosis is not entirely understood. Deszcz et al. have demonstrated that Human

rhinovirus 14 (HRV14) infection triggers apoptosis in both HeLa cells and the human

bronchial epithelial 16HBE14o- cell line, when cells are infected with high

concentrations of virus (Deszcz et al., 2005). They demonstrated that during infection

with HRV14, apoptosis was induced via the intrinsic apoptotic pathway, as

demonstrated by the cleavage of caspase 9 and the release of cytochrome c from within

the mitochondria to the cytoplasm; the universal apoptotic makers, caspase 3 and the

caspase substrate PARP were also found to be cleaved. Similar results were obtained by

Drahos and Racaniello, where they observed that apoptosis was induced in HeLa cells

infected with HRV1a at a multiplicity of infection (MOI) of 10 through the

confirmation that PARP, a known caspase substrate, was cleaved in infected cells

(Drahos & Racaniello, 2009). Taimen et al also found that infection with HRV1b

resulted in apoptosis induction, demonstrated through the cleavage of caspase 3 and

PARP (Taimen et al, 2004). In contrast, Gustin and Sarnow showed that HeLa cells

infected with HRV14 showed no signs of PARP cleavage and only a very small amount

of DNA fragmentation suggesting that apoptosis was inhibited (Gustin & Sarnow,

2002).

23

The effect of HRV infection on the host apoptotic pathways is still not completely

understood. Whilst numerous studies have been performed to investigate apoptosis

during other picornavirus infections, very few have focussed on HRV. The observations

made so far have failed to decisively elucidate the effect of HRV infection on apoptosis,

with a number of contradicting observations described. This remains an area where

further investigations are required to improve our understanding of the processes

involved, and may provide potential future therapeutic targets to be identified.

24

1.5 Hypothesis and Aims of Present Study

1.5.1 Hypothesis

Induction of apoptosis is one of the cells first responses to viral infection. It has been

observed that various viral features are capable of inducing the apoptotic response,

whilst in contrast, it has also been demonstrated that some viruses are capable of

altering the apoptotic pathway in order to improve their chances of survival and spread.

Previous studies investigating the effect of picornaviral infection on the host apoptotic

response, have demonstrated that some members of this family of viruses are capable of

inhibiting apoptosis, while others are capable of inducing apoptosis and some appear to

do both. Poliovirus, a member of the Enterovirus genera of picornaviruses which is very

closely related to HRV, has been shown to both inhibit and induce apoptosis depending

on the availability of optimal viral growth conditions. Few studies have investigated

apoptosis during HRV infection, with those carried out so far demonstrating conflicting

results of both apoptotic induction and inhibition. It is likely that the viral 2A and 3C

proteases may play a role in the regulation of apoptosis, particularly due to their

demonstrated ability alter various host signally pathways.

Based on the results seen in studies investigating the induction of apoptosis during

picornaviral infection, particularly those focusing on poliovirus and the few focussed on

HRV infection, it is hypothesised that HRV infection delays the induction of apoptosis

by the host cell in order to facilitate its own replication.

1.5.2 Aims

The overall objective of this research project is to identify the effect of HRV infection

on the induction of apoptosis. In order to determine the apoptotic response to human

HRV infection, the following aims will be addressed:

Aim 1. To determine if HRV infection inhibits or induces the host’s apoptotic

pathways

The apoptotic response in Ohio-HeLa cells, both uninfected and infected with HRV16,

will be investigated in the presence and absence of known chemical apoptosis inducers

to determine if HRV16 infection alters the host’s apoptotic response. Western blot

25

analysis will be used to detect various apoptotic markers as well as known caspase

substrates to confirm apoptotic activity along with immunofluorescence assays.

Aim 2. To determine if the 3C and 2A proteases have a role in apoptosis

induction/inhibition

In order to determine the role that the HRV proteases 3C and 2A play in the apoptotic

response, Ohio-HeLa cells expressing the viral proteases will be studied to determine if

the expression of the viral proteases alters the induction of apoptosis. The apoptotic

response by the Ohio-HeLa cells will be determined using western blot analysis and

immunofluorescence assays.

It is expected that the results of this study will show that the induction of apoptosis in

Ohio-HeLa cells as a result of HRV16 infection is inhibited or delayed by the virus in

order to improve viral replication. It is also expected that the viral protease, 2A and 3C,

will play a role in this alteration.

26

2 Chapter Two – Materials and Methods

2.1 Materials

2.1.1 Virus and Cell Culture Lines

2.1.1.1 Cell Culture Lines

The Ohio strain of the HeLa human cervical carcinoma cell line was used throughout

this study. Ohio-HeLa cells were maintained in growth media (see table 2.1) at 37°C

with 5% CO2.

COS7 cells were used for transfection of the HRV16 2A and 3C proteases. Cells were

maintained in growth media (see table 2.1) at 37°C with 5% CO2.

2.1.1.2 Virus Lines

Human rhinovirus serotype 16 (HRV16) was used throughout this study.

2.1.1.3 Media Used for Cell Culture and Viral Infection

The following media was used throughout this study during the culture of cell and virus

lines and during viral infections.

Table 2.1 Cell and virus culture media used during this study.

Media Composition

Growth media Dulbecco’s modified Eagle’s Medium (DMEM)

supplemented with 10% foetal calf serum.

Maintenance media Dulbecco’s modified Eagle’s Medium (DMEM)

supplemented with 2% foetal calf serum.

Serum free media Dulbecco’s modified Eagle’s Medium (DMEM)

PBS (1x) 137mM NaCl; 2.7 mM KCl; 8.1mM Na2HPO4; 1.47mM

KH2PO4

0.1% Crystal Violet stain 0.1% crystal violet in ethanol

Trypsin/EDTA Gibco

27

2.1.2 Buffers, Solutions and Bacterial Culture Media

The following buffers and solutions were used throughout this study.

Table 2.2 Buffers and solutions used during this study.

Buffer/Solution Composition

PBS (1x) 137mM NaCl; 2.7 mM KCl; 10mM Na2HPO4; 1.7mM

KH2PO4; pH7.4

RIPA Buffer 150mM NaCl; 1% Triton X 100; 0.5% sodium

deoxycholate; 0.1% SDS; 50mM Tris (pH 8); 1x protease

and phosphatase inhibitors (Roche); H2O

Taxol Sigma-Aldrich 1mg/ml in DMSO

Actinomycin D Sigma-Aldrich 1mg/ml in DMSO

Cyclohexamide 2mg/ml in H2O

z-VAD-fmk 5mM in DMSO

Running Buffer 0.1% SDS; 25mM Tris base; 192mM glycine

Transfer Buffer 25mM Tris base; 192mM glycine

Blocking Solution 3% skim milk powder; PBS

Washing Solution (PBST) 0.1% Tween20; PBS

Ponceau S Stain 2% Ponceau S; 30% trichloroacetic acid; 30%

sulfosalicylic acid

ProLong Gold mounting

media with DAPI

Invitrogen

Kanamycin Sigma 10mg/ml in H2O

Luria-Bertani broth 10g/l bactero-tryptone; 5g/l bacto-yeast extract; 5g/l NaCl

Laemmli buffer 4% SDS; 10% 2-mercaptoethanol; 20% glycerol; 0.004%

bromophenol blue; 0.125M Tris HCl; pH6.8

SDS PAGE stacking gel 5% acrylamide; 0.5M Tris-HCl; 10% SDS; 10% APS;

H2O

SDS PAGE separating gel 12.5% acrylamide; 1.5M Tris-HCl; 10% SDS; 10% APS;

H2O

Western blot stripping

buffer

2% SDS; 62.5 mM Tris-HCl pH 6.8; 114.4 mM β-

mercaptoethanol

28

2.1.3 Antibodies

2.1.3.1 Primary Antibodies – Western blot

The primary antibodies used for western blots throughout this project are listed below.

Table 2.3 Western blot primary antibodies used during this study.

Antibody Dilution Source species Manufacturer

VP2* 1:2000 Mouse Gift from Dr Wai-Ming Lee

Caspase 3 1:500 Mouse Santa Cruz

Caspase 9 1:1000 Mouse Santa Cruz

PARP 1:200 Mouse Santa Cruz

αβ-Tubulin 1:1000 Rabbit Cell Signalling Technology

RIPK1 1:1000 Mouse Thermo Scientific * - gift from Dr Wai-Ming Lee, University of Wisconsin

2.1.3.2 Secondary Antibodies – Western blot

The secondary antibodies used for western blots throughout this study are listed below.

Table 2.4 Western blot secondary antibodies used during this study.

Antibody Dilution Source

species

Manufacturer

Anti-rabbit immunoglobulins conjugated to

horseradish peroxidase (goat-anti-rabbit HRP)

1:5000 Goat Invitrogen

Anti-mouse immunoglobulins conjugated to

horseradish peroxidise (goat-anti-mouse HRP)

1:5000 Goat Invitrogen

2.1.3.3 Primary Antibodies – Immunofluorescence

The primary antibodies used for immunofluorescence are listed below.

Table 2.5 Immunofluorescence primary antibodies used during this study.

Antibody Dilution Source

species

Manufacturer

VP2# 1:2000 Mouse Gift from Dr

Wai-Ming Lee

Cytochrome c 1:200 Mouse Santa Cruz

dsRNA* 1:200 Guinea pig Gift from Dr

Scott Bowden * - gift from Dr Scott Bowden, VIDRL, Melbourne.

# - gift from Dr Wai-Ming Lee, University of Wisconsin

29

2.1.3.4 Secondary Antibodies – Immunofluorescence

The secondary antibodies used for immunofluorescence throughout this study are listed

below.

Table 2.6 Immunofluorescence secondary antibodies used during this study.

Antibody Dilution Source

species

Manufacturer

Goat anti mouse Alexa 568 1:1000 Goat Molecular

Probes

Goat anti mouse Alexa 488 1:1000 Goat Molecular

Probes

Donkey anti goat Alexa 488 1:1000 Donkey Molecular

Probes

Donkey anti mouse Alexa

568

1:1000 Donkey Molecular

Probes

2.1.4 Commercial Kits

The following commercial kits were used during this study to detect the presence of

apoptosis.

Table 2.7 Commercial kits used during this study.

Kit Manufacturer

Plasmid Mini Prep Promega

Western Lighting, ECL Perkin-Elmer

Lipofectamine 2000 Invitrogen

2.2 Methods

2.2.1 Cell and Virus Cultures

2.2.1.1 Cell Culture

Ohio-HeLa cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM)

media supplemented with 10% foetal bovine serum and were incubated at 37°C with

5% CO2. During each passage, at approximately 90-100% confluence the supernatant

was removed from each flask and the adherent cells were washed with 5ml of PBS.

Cells were detached via treatment with 1ml of Trypsin/EDTA for 2-3mins. Cells were

then suspended in 4ml of 10% FBS DMEM. Cells were split 1:5 with 1ml of cell

30

suspension added into each T25ml flask containing 4ml of 10%FBS DMEM and

incubated at 37°C with 5% CO2 until confluent.

2.2.1.2 Virus Culture

Rhinovirus 16 was cultured in Ohio-HeLa cells. Ohio-HeLa cells were cultured and

incubated overnight as described above to give approximately 70% confluence. The

supernatant was removed and cells were washed with 5ml of PBS. 1ml of 2% FBS

DMEM containing enough virus inoculum to give an MOI of 0.1 was added to each

flask of cells which were then incubated at 37°C with 5% CO2 for one hour with gentle

rocking every 15min. After the hour, an additional 4ml of 2% FBS DMEM media was

added to each flask of cells which were then incubated overnight at 37°C with 5% CO2.

The following day cells were checked for evidence of cytopathic effects (CPE) as

confirmation of viral infection before flasks were sealed and stored at -80°C. The flasks

were later thawed at room temperature and the cultured virus was collected and

transferred into 10ml tubes which were then centrifuged at 4000rpm for 15mins at room

temperature to remove cell debris. The supernatant containing the cultured virus was

then removed and distributed into eppendorf tubes and stored at -80°C until required.

2.2.1.3 Virus Titration

All passages of cultured virus were titrated on Ohio-HeLa cells to determine infectious

virus content. Three rows of a 96 well tissue culture plate were seeded with O-HeLa

cells for each virus isolate. Each well was seeded with approximately 5x103 cells per

well diluted in 100µl of 10% FBS DMEM. Ten-fold serial dilutions of the virus culture

were made in 2% FBS DMEM ; 50µl of each virus dilution was added in triplicate to

wells in decreasing concentration with the last three wells having only media added.

The plate was then incubated for 6 days at 37°C with 5% CO2. Following incubation,

the plate was examined under a microscope for signs of CPE. Cells were stained with

0.1% Crystal Violet stain and the CPE positive wells were counted. Virus titration was

then calculated using the Spearman-Karber equation:

Titre = 10 1 + Z(X-0.5)

Where Z equals the log 10 of the dilution factor and X equals the sum of the CPE

positive wells.

31

Three virus cultures were carried out for this study from an original culture provided by

Dr. Reena Ghildyal at passage 2. The virus titrations of each of passage were as follows:

Passage 3 = 6.3 x 106 particles per ml.

Passage 4 = 13.5 x 106 particles per ml.

Passage 5 = 13.5 x 106 particles per ml.

2.2.1.4 Viral Infection

Ohio-HeLa cells were infected with the indicated MOI of HRV16. Cells were seeded in

6-well plates with 2ml of growth media (see section 2.1.3) and incubated overnight at

37°C with 5% CO2 to approximately 70-80% confluence. The media was removed and

cells were washed with 1ml of PBS before the addition of the virus culture diluted in

maintenance media to the indicated MOI to a total of 1ml. Cells were incubated with the

virus for 1hr at 37°C with 5% CO2 with gentle rocking every 15mins. The excess

unincorporated virus was removed and 2ml of fresh maintenance media was added.

Cells were then incubated and treated as indicated.

2.2.1.5 Transfection

2.2.1.5.1 Plasmids

GFP-2A, GFP-2Ainactive, GFP-3C and GFP-3Cinactive clones were generously

provided by Dr. Erin Walker (Walker et al., 2013). Briefly, the sequences for HRV16

2A and 3C were amplified by PCR from the full length HRV16 genome (Lee et al.,

1993) and recombined into the Gateway compatible vector, pEPI-DESTC, to enable the

production of GFP tagged HRV16 2A and 3C proteases (Ghildyal et al., 2005; Ghildyal

et al., 2009). An inactive form of HRV16 2A was generated by mutating the active

Cysteine (Cys 106 (Racaniello, 2007)) to alanine by site-directed mutagenesis and

recombined into the pEPI-DESTC vector as for the wildtype to generate GFP-2Ainac.

Similarly, an inactive form of HRV16 3C was generated by mutating the active

Cysteine (Cys 147 (Racaniello, 2007)) to alanine by site-directed mutagenesis and

recombined into the pEPI-DESTC vector as for the wildtype to generate GFP-3Cinac.

32

The vector (pEPI-GFP) which expresses GFP alone in mammalian cells (Ghildyal et al,

2009) was used as control in all experiments.

2.2.1.5.2 DNA Plasmid Extraction

The GFP-2A, GFP-2Ainactive, GFP-3C and GFP-3Cinactive glycerol stocks were

inoculated in 5ml of Luria-Bertani broth (LB) with 25µl of Kanamycin (final

concentration of 50ug/ml) and incubated at 37°C with constant shaking for 16hrs.

Plasmids were prepared using the Promega Pure Yield miniprep kit as per the

manufacturer’s instructions. The concentration of plasmid DNA was measured using the

Nano-drop spectrophotometer.

2.2.1.5.3 Transfection

COS7 cells were seeded into a 6-well plate in growth medium and grown overnight at

37°C with 5% CO2 until approximately 95% confluent. Cells were then gently washed

with PBS before 250µl of serum free DMEM, 10µl of lipofectamine and 4µg of the

relevant DNA was added and cells were incubated at 37°C with 5% CO2 for 30mins.

2ml of growth media was added to cells and they were incubated overnight at 37°C with

5% CO2 before being lysed and processed for western blot analysis as outlined in

section 2.2.2.1.

2.2.2 Analysis

2.2.2.1 Western Blot Analysis

For each western blot performed, Ohio-HeLa cells were cultured in 6 well plates or

equivalent sized petri dishes. After relevant treatment, cells were lysed with 150µl of

RIPA buffer gently rocked on ice for 30mins. Cell lysates were then transferred into

labelled eppendorf tubes before being stored at -20°C. The cell lysates were mixed with

a 5x Laemmli buffer and heated at 95°C for 5mins. The denatured lysates were

electrophoresed on a 12.5% SDS PAGE gel before being transferred to a nitrocellulose

membrane. Membranes were incubated in 3% skim milk powder in PBS (blocking

33

solution) for 1hr. Following incubation in the blocking solution, the membranes were

incubated with the relevant primary antibody diluted in 1% skim milk powder in PBST

solution overnight at 4°C on a shaker. Membranes were then washed with PBST, three

times for 5mins and once for 10 mins before being incubated for 2hrs in a species

specific secondary antibody conjugated to HRP, diluted in 1% skim milk powder in

PBST. Membranes were washed in PBST followed by enhanced chemiluminiscence

(ECL), exposure and manual development of X-ray films (Kodak film) in order to

visualise the relevant protein bands.

Where required, blots were stripped of probing antibodies using stripping buffer at 50°C

for 10 min, followed by washing in PBST, blocking in 4% skim milk in PBS and

reprobing using different primary antibodies as required.

2.2.2.2 Immunofluorescence Assay

For immunofluorescence (IFA), Ohio-HeLa cells were seeded onto sterile 15mm2

coverslips within a 12well plate and incubated in 1ml of growth media overnight at

37°C with 5% CO2 to approximately 70-80% confluence. Cells were then treated as

outlined for each experiment. After treatment, the media was removed and cells were

washed with 1ml of PBS. Cells were fixed with 500µl of ice cold (-20°C) methanol for

10 mins. Methanol was then washed from cells with two 5 minute washes of 1ml of

PBS. Fixed cells were incubated with 25µl of the relevant primary antibodies for

30mins and then washed with two 5 minute washes of 1ml of PBS. Cells were

incubated with 25µl of the relevant secondary antibody in the dark for 30mins before

again being washed with two 5 min washes with PBS. The coverslips were mounted

onto glass slides with Prolong Gold antifade mounting medium containing DAPI before

curing overnight in the dark. Slides of treated cells were examined and photographed

using Nikon Ti Eclipse confocal laser scanning microscope with Nikon 60x/1.40 oil

immersion lens (Plan Apo VC OFN25 DIC N2; optical section of 0.5µm).

34

3 Chapter Three – Results Rhinovirus Infection Does Not Induce Early or Late Intrinsic Apoptosis

3.1 Introduction

One of the host cell defences against viral infection and spread is the induction of

apoptosis (Barber, 2001; Richard & Tulasne, 2012). Many viruses have developed

strategies to evade this immune mechanism, including a number of picornaviral species,

which have been shown to have the ability to either induce or inhibit the induction of

host apoptosis (Belov et al., 2003; Tolskaya et al., 1995). A few studies investigating

apoptosis during HRV infection have suggested that infection may lead to the induction

of intrinsic, but not extrinsic apoptosis (Taimen et al., 2004; Deszcz et al., 2005; Drahos

& Racaniello, 2009). Interestingly, a study by Gustin and Sarnow showed that infection

of HeLa cells with HRV14 did not result in cleavage of PARP, a marker for late

apoptosis in both intrinsic and extrinsic pathways (Gustin & Sarnow, 2002).

This chapter aimed to determine whether or not apoptosis is induced during infection of

Ohio-HeLa cells with HRV16 through a careful examination of early and late markers

of the intrinsic pathway under various infection conditions.

Following on from the results shown by Deszcz et al, this study paid particular focus on

the induction of the intrinsic apoptotic pathway during HRV16 infection. The intrinsic

apoptotic markers, caspase 9 and cytochrome c, were examined, as well as the universal

apoptotic marker caspase 3. Cytochrome c is released from the mitochondria into the

cytoplasm as one of the initial steps of the intrinsic pathway (refer to figure 1.4).

Caspase 9 is an initiator caspase of the intrinsic apoptotic pathway that once activated,

can go on to activate other apoptotic intermediates, including the caspase 3 effector

caspase. Caspase 3 is an effector caspase that is activated during both the intrinsic and

extrinsic apoptotic pathways (refer to figure 1.4).

As a control, and to determine whether HRV16 induces or is capable of halting

apoptotic cell death, experiments were performed using chemical apoptotic inducers.

Two chemical agents were examined, Taxol and Actinomycin D (Act. D). Paclitaxel

(Taxol) is an anticancer drug derived from the Taxus brevifolia plant (Wani et al., 1971;

35

McGuire et al., 1989). It acts to promote the polymerisation and stabilisation of

microtubules resulting in cell cycle arrest and eventual apoptosis of treated cells (Schiff

et al., 1979; Rowinsky et al., 1988; Woods et al., 1995). Taxol treatment is known to

induce apoptosis in numerous cancer cell lines through the phosphorylation of the anti-

apoptotic gene Bcl-2 (Bhalla et al., 1993; Haldar et al., 1995; Blagosklonny et al.,

1996). Similarly, Act. D is a bacterially derived anti-cancer antibiotic that is known to

induce apoptosis. Act. D acts to inhibit transcription eventually leading to the apoptotic

death of the cell (Vining & Waksman, 1954; Goldberg et al., 1962; Reich et al., 1962).

3.2 HRV16 Infection Does Not Result in Cleavage of PARP

As a first step to elucidating the effect of HRV16 infection on apoptosis, the cleavage of

PARP during infection was examined. Ohio-HeLa cells were seeded and infected with

HRV16 at a MOI of 3 as per section 2.2.1.4. Cells were then treated with 5µg/ml Act. D

and cycloheximide (CHX) for 6hrs, before being lysed with 150µl of RIPA buffer.

Controls were either left untreated, infected for 6h, treated for 6 hrs with Act. D and

cycloheximide or treated for 6 hrs with Act.D and cycloheximide in the presence of the

apoptosis inhibitor z-VAD-fmk. Lysates were processed for western blot analysis and

probed for PARP. As shown in figure 3.1, chemical induction of apoptosis resulted in

cleavage of PARP (lane 2), but HRV16 infection did not (lane 1); infection with

HRV16 was able to inhibit chemical apoptosis as shown by the reduction in intensity of

the cleaved PARP band (lane 4).

The data in figure 3.1 show that HRV16 infection does not induce late apoptosis.

Further investigations in this chapter focussed on determining the stage of the apoptosis

cascade that is inhibited.

36

Figure 3.1 HRV16 infection does not lead to the cleavage of PARP.

Ohio-HeLa cells were infected with HRV16 at MOI 3 and treated with 5µg/ml Act. D and CHX in the

presence or absence of the caspase inhibitor z-VAD-fmk for 6hrs before being lysed for western blot

analysis. Western blot was probed for the presence of PARP with mouse anti PARP primary antibody and

goat anti mouse HRP secondary antibody. Arrows on the left of the blot indicate the approximate protein

size in kDa while arrows on the right of the blot indicate the expected location of PARP and the cleaved

PARP fragment. Results shown are representative of at least 3 independent experiments.

3.3 Rhinovirus Serotype 16 Infection Does Not Induce Late Apoptosis

Further investigation into the effect of HRV16 infection on the induction of host cell

apoptosis was performed using western blot analysis for the cleavage of caspases 3 and

9. Ohio-HeLa cells were seeded and infected as outlined in section 2.2.1.4 with a MOI

of 1. Infected cells were lysed at regular time points post infection (p.i.) (0, 3, 6, 9 and

24hrs p.i.) with 150µl of RIPA buffer (see section 2.1.2) and the cell lysates subjected to

western blot analysis as outlined in section 2.2.2.1. Cells at 24hrs were observed to have

undergone significant CPE with a number of cells having lifted from the well; these

cells were collected and processed along with the attached cells. The blots were initially

probed to detect cleaved caspase 9, as induction of intrinsic apoptosis results in the

activation of caspase 9 via cleavage resulting in a 10kDa cleavage product. The blot

was incubated with the mouse anti caspase 9 antibody followed by HRP-conjugated

secondary antibodies; the antibodies were subsequently stripped from the blot and the

blot re-probed for αβ tubulin as a loading control. As seen in figure 3.2A, there was no

37

detection of the caspase 9 p10 cleavage fragments, nor was there any indication of a

reduction in the procaspase protein. The even loading, as shown by tubulin, supported

the identification that the procaspase 9 protein was not being cleaved, with no obvious

reduction observed over time. Despite the absence of typical apoptotic cleavage of

caspase 9, extra cleavage bands could be seen after 9 and 24hrs in a number of repeat of

this experiment, as demonstrated in figure 3.2A. Due to time constraints this cleavage

was not further investigated, however it may provide a target for future investigation.

The above procedure was repeated, this time probing for the cleavage of caspase 3, an

effector caspase that is cleaved in both the intrinsic and extrinsic apoptotic pathways,

resulting in two cleavage products of 17kDa and 11kDa. Similarly to the caspase 9

results, figure 3.2B shows that procaspase 3 was not cleaved during HRV16 infection,

with no sign of the caspases 3 p11 or p17 cleavage fragments nor any indication of a

reduction in the levels of procaspases 3. Again in some repeats of this experiment,

however not all, extra cleavage of procaspase 3 could be seen at 9 and 24hrs p.i. as seen

in figure 3.2B. Time constraints prevented this cleavage from being further investigated.

These western blots were lastly probed for the HRV structural protein VP2 to confirm

infection had taken place. Figure 3.2C illustrates that VP2 was present from 3hrs post

infection confirming that apoptosis is not induced during HRV16 infection up to 24hrs

p.i. when infected with an MOI of 1. As the viral polyprotein undergoes a series of

cleavages to produce each of the functional viral proteins, each protein may appear in

various lengths dependant on the level of processing that has occurred. This variation is

demonstrated in figure 3.2C where various cleavage products of the viral polyprotein

containing VP2 can be seen.

The data in Figure 3.2 show that HRV16 infection did not induce apoptosis even at 24h

p.i. when extensive cytopathic effect is observed.

To confirm that the lack of caspase cleavage was not due to suboptimal HRV16

infection, Ohio-HeLa cells were infected with HRV16 of varying MOI (0.1, 1 and 10)

for 3, 6 and 24hrs before being lysed for western blot analysis. The cellular lysates were

analysed by western blot analysis as per the methodology used above. As illustrated in

figure 3.3, no p11 cleavage fragment of caspases 3 could be seen, nor is there any

evidence of a reduction in the procaspase 3 protein. As with the results of the previous

experiment, cells at 24hrs were observed to have undergone significant CPE with a

38

number of cells rounding and lifting from the well, these cells were collected and

processed along with the attached cells.

These results suggest that the caspase 9 dependant intrinsic apoptotic pathway is not

induced during HRV16 infection. The identification that caspase 3 is not activated

during infection suggests that neither the intrinsic nor extrinsic pathways are activated,

as caspase 3 is an effector caspase located at the point beyond which both the intrinsic

and extrinsic apoptotic cascades converge (see figure 1.4). These results also suggest

that the absence of apoptotic induction is not dependant on time or MOI.

39

Figure 3.2 Detection of late apoptosis during RV16 infection.

Ohio-HeLa cells were infected with HRV16 at MOI of 1 before being lysed at various time points for

western blot analysis for the detection of markers of late apoptosis. Western blot analysis was carried out

as outlined in section 2.2.2.1. Numbers on the left of each blot indicate the approximate protein size in

kDa. Each blot was incubated with anti-β tubulin to show equal loading in all lanes and is shown below

each blot. A. Blot was incubated with mouse anti caspase 9 p10 primary antibody and goat anti mouse

HRP secondary antibody. Numbers on the right of the blot show the procaspase 9 protein as well where it

would be expected to see the p10 cleaved caspase 9 fragment. B. Western blot was incubated with the

mouse anti caspase 3 primary antibody and the goat anti mouse HRP secondary antibody. Arrows on the

right show the procaspase 3 protein as well as the expected location of the p17 and p11 cleaved caspase 3

fragments. C. Western blot was incubated with mouse anti-VP2 primary antibody and goat anti mouse

HRP secondary antibody to confirm that infection had taken place. Markers on the right side of the blot

demonstrate the presence of the viral VP2 protein in its various stages of cleavage from the polyprotein.

Results shown are representative of at least 3 independent experiments.

40

Figure 3.3 Effect of varying MOI on the detection of late apoptosis during RV16 infection.

Ohio-HeLa cells were infected with RV16 with varying MOI (0.1, 1.0, and 10) before being lysed at

various time points for western blot analysis. Western blot analysis was carried out for the detection of

caspase 3 cleavage, a marker of late apoptosis as outlined in 2.2.2.1. The blot was incubated with mouse

anti caspase 3 p11 primary antibody followed by the goat anti mouse HRP secondary antibody. Arrows

on the left of the blot indicate the approximate protein size in kDa. The arrows on the right of the image

show the procaspase 3 protein as well as the expected location of the p11 and p17 cleaved caspase 3

fragments. Results shown are representative of at least 3 independent experiments.

3.4 RV16 Infection Does Not Induce Early Intrinsic Apoptosis

The apoptotic pathways include a complex cascade of events that ultimately result in the

characteristic apoptotic cell death. This complex cascade provides numerous targets for

viral inhibition of apoptotic death. The finding that the executioner caspase 3 is not

cleaved during HRV16 infection suggests that HRV could possibly interfere with the

pathway further upstream. To identify whether HRV16 may be halting the intrinsic

apoptotic cascade upstream of caspases 3, immunofluorescence assay was performed as

per the methodology outlined in section 2.2.2.2 followed by confocal microscopy. Ohio-

HeLa cells were seeded onto sterile coverslips to approximately 80% confluence. Cells

were then infected with HRV16 with an MOI of 5 and incubated at 37°C with 5% CO2

for 9hrs before being fixed and probed with the mouse anti cytochrome c and guinea pig

anti dsRNA primary antibodies followed by Alexa 568 conjugated goat anti mouse and

Alexa 488 conjugated goat anti guinea pig secondary antibodies. Figure 3.4A

demonstrates the cytochrome c (in red) concentrated within the mitochondria of mock

41

uninfected and untreated cells. Figure 3.4B shows both infected and uninfected cells, all

showing cytochrome c concentrated within the mitochondria. Infected cells are

demonstrated by the presence of dsRNA shown in green. Despite various physical

differences that can be seen between infected and uninfected cells, including

cytoplasmic shrinkage and rounding of the cell, there does not appear to be any change

in the appearance of cytochrome c, with it appearing in concentrated areas rather than

diffuse throughout the cytosol.

These results further confirm that the intrinsic apoptotic pathway is not induced during

HRV16 infection.

42

B.

A.

Figure 3.4 Cytochrome c is not released during RV16 infection.

Ohio-HeLa cells were left uninfected (A) or infected with HRV16 with an MOI of 5 (B) for 9 hrs before

being fixed and incubated with guinea pig anti dsRNA and mouse anti cytochrome c primary antibodies

followed by Alexa 488 conjugated goat anti guinea pig and Alexa 568 conjugated goat anti mouse

secondary antibodies. Coverslips were mounted onto slides using antifade mounting medium containing

DAPI stain A. Uninfected cells show no sign of dsRNA. Arrows indicate the cytochrome c accumulated

within the mitochondria. B. Infected cells are shown by the presence of dsRNA antibody shown in green.

Infected cells show cytochrome c still accumulated within the mitochondria as demonstrated by the

arrows. Results shown are representative of at least 3 independent experiments.

43

3.5 Taxol Induces Apoptosis in Ohio-HeLa Cells

Ohio-HeLa cells were treated with the chemical inducer of apoptosis, Taxol, to

demonstrate that apoptosis could be effectively induced in this cell line. The aim was to

confirm that the results observed during HRV16 infection in sections 3.2 and 3.3 were a

consequence of infection itself rather than the cells’ inherent resistance to apoptosis.

Western blot analysis for the cleavage of procaspase 3 and immunofluorescence assay

for the release of cytochrome c from the mitochondria were used to detect the induction

of apoptosis.

Initially, Ohio-HeLa cells were grown in a 6-well plate in 2ml of growth media

overnight at 37°C with 5% CO2 to approximately 80% confluence as outlined in section

2.2.1.1. Cells were then treated with varying concentrations of Taxol (0.25µg/ml,

0.5µg/ml and 1 µg/ml diluted in maintenance media) or DMSO (Taxol was dissolved in

DMSO before dilution) before being lysed for western blot analysis after 2 or 24 hrs of

treatment as per the method outlined in section 2.2.2.1. The results suggested that the 2

hour incubation was not enough to allow the Taxol to induce apoptosis as there was no

evidence of caspase 3 cleavage (figure 3.5A). With 24h of treatment, cleaved caspase 3

could be seen with all three concentrations of Taxol; 0.5 µg/ml Taxol demonstrated the

strongest level of cleavage at 24hrs treatment (figure 3.5B). Whilst it would be expected

that the level of caspase 3 cleavage would increase with increased Taxol concentration,

the results seen in figure 3.5B do not demonstrate this. It is likely that the lower level of

p11 cleaved fragment seen in the 1 µg/ml cell lysates compared to that seen in the 0.5

µg/ml lysates is a result of the concentration combined with the long incubation leading

to the further degradation of the cleaved fragment or resulting in the cells deteriorating

too far for the cleaved fragment to be recovered.

To further demonstrate that Taxol induced detectable apoptosis in Ohio-HeLa cells,

immunofluorescence assay was carried out for the detection of cytochrome c. Cells

were seeded onto sterile coverslips as outlined in section 2.2.2.2 and grown to

approximately 80% confluence. Cells were either treated with 5µg/ml of Taxol for 3hrs

or left untreated before being washed and fixed as outlined in section 2.2.2.2 and

incubated with the mouse anti cytochome c primary antibody followed by Alexa 488

conjugated goat anti mouse secondary antibody. Coverslips were mounted onto slides

using antifade mounting medium containing DAPI. Slides were then imaged and

analysed using confocal microscopy. Figure 3.6A shows untreated cells demonstrating

44

cytochrome c concentrated within the mitochondria as indicated by the arrows. Figure

3.6B shows a number of cells demonstrating cytochrome c having been released from

the mitochondria into the cytosol, revealing a diffusion of cytochrome c throughout the

cytosol. These cells are most likely to be at the very early stages of apoptosis with none

of the characteristic physical changes associated with apoptosis being observed, which

is expected given the short incubation period of only 3hrs.

The above results show that Ohio-HeLa cells undergo the induction of apoptosis after

treatment with Taxol, a known chemical inducer of apoptosis. The data presented

clearly show that Ohio-HeLa cells respond to Taxol treatment by induction of the

intrinsic apoptotic pathway as shown by release of cytochrome c from mitochondria

into the cytoplasm at 3h post treatment and cleavage of the effector caspase 3 at 24h

post treatment.

Figure 3.5 Taxol treatment induces intrinsic apoptosis in Ohio-HeLa cells.

Ohio-HeLa cells were treated with varying concentrations of Taxol and lysed after 2 or 24hrs. Western

blots were incubated with the mouse anti caspase 3 p11 primary antibody followed by the goat anti mouse

secondary antibody. Arrows on the left of each blot indicate the approximate protein length in kDa. The

arrows on the right of each blot demonstrate the location or expected location of the procaspase 3 protein

or the p11 and p17 cleavage fragments. A. cells were incubated with Taxol for 2hrs. As indicated by the

arrows on the right, no cleavage of the procaspase 3 was observed with no sign of p11 or p17 fragments

nor an obvious reduction in the level of the procaspase protein. B. Cells were treated with Taxol of

various concentrations as indicated. As indicated by the arrows on the right, after 24hrs treatment with 0.5

and 1.0µg/ml of Taxol, both p11 and p17 cleavage fragments can be seen demonstrating that Taxol

treatment induces apoptosis in Ohio-HeLa cells. Results shown are representative of at least 3

independent experiments.

45

Figure 3.6 Taxol treatment leads to cytochrome c release from the mitochondria.

Ohio-HeLa cells were seeded onto sterile coverslips and either left untreated (A) or treated with 5µg/ml

of Taxol (B) for 3 hrs before being fixed and incubated with mouse anti cytochrome c primary and Alexa

488 conjugated goat anti mouse secondary antibodies. Coverslips were mounted onto slides using

antifade mounting medium containing DAPI stain. A. Untreated cells show cytochrome c accumulated

within the mitochondria demonstrated by the arrows. B. Cells treated with Taxol are shown with a

majority of cells having released cytochrome c demonstrated by the absence of concentrated regions of

cytochrome c. The arrow indicates an unaffected cell where cytochrome c has not been released. Results

shown are representative of at least 3 independent experiments.

B.

A.

46

Actinomycin D (Act. D) is an antibiotic used in cancer therapy and is known to induce

apoptosis in treated cells. Similarly to studies with Taxol, western blot analysis for the

cleavage of caspases 3 as well as immunofluorescence assay for the detection of the

release of cytochrome c from the mitochondria was used to demonstrate the induction of

apoptosis in Ohio-HeLa cells treated with Act D.

For western blot analysis, Ohio-HeLa cells were seeded into a 6-well plate in growth

media and incubated overnight at 37°C with 5% CO2 to approximately 90% confluence.

Cells were then either treated with 1 µg/ml Act. D diluted in maintenance media or left

untreated before being lysed after 3 or 5hrs and processed for western blot analysis

(section 2.2.2.1). The western blot was incubated with the mouse anti caspase 3 p11

antibody as per the method outlined in section 2.2.2.1. Figure 3.7 shows some caspase 3

cleavage after 3hrs treatment and an increased level of cleavage in the cells treated for

5hrs. Only partial, low level cleavage was observed at the longest treatment times (5hrs)

in this experiment.

Cells treated with Act. D were also analysed by immunofluorescence for the

mitochondrial release of cytochrome c. Cells were seeded onto sterile coverslips and

incubated overnight at 37°C with 5% CO2 to approximately 80-90% confluent. Cells

were treated with 5µg/ml Act. D for 9hrs before being fixed and incubated with mouse

anti cytochrome c primary antibody followed by Alexa 488 conjugated goat anti mouse

secondary antibody as per the method outlined in section 2.2.2.2. Figure 3.8

demonstrates representative images of both untreated and treated cells. Figure 3.8A

shows untreated cells with the arrows demonstrating cytochrome c accumulated within

the mitochondria. Cells treated with Act. D were seen to undergo apoptosis with figure

3.8B demonstrating cells at various stages of apoptotic death. The arrows indicate

cytochrome c still contained within the mitochondria of cells that are either unaffected

or in the initial stages of apoptosis. The boxed and enlarged cell shows an example of a

cell in the later stages of apoptosis showing cytoplasmic shrinkage and blebbing of the

plasma membrane, changes characteristic of apoptotic death. This cell shows

cytochrome c having been released from the mitochondria and localised diffusely

throughout the cytoplasm rather than concentrated within the mitochondria.

These results confirm that apoptosis can be effectively induced in Ohio-HeLa cells

treated with Act. D demonstrated by the cleavage of caspase 3 and release of

47

cytochrome c from the mitochondria, which was observed after treatment of Ohio-HeLa

cells with 5µg/ml of Act. D.

Figure 3.7 Act. D treatment induces apoptosis in Ohio-HeLa cells.

Ohio-HeLa cells were treated with 1µg/ml Act. D before being lysed at 3 or 5hrs and analysed by western

blot for the identification of cleavage of procaspase 3. The blot was incubated with mouse anti caspase 3

p11 primary antibody followed by goat anti mouse HRP secondary antibody. Arrows on the left indicate

the approximate protein length in kDa. The arrows on the right indicate the location of the procaspase

and cleaved caspase 3 locations. The evidence of both p11 and p17 cleavage fragments in both the 3 and

5hr samples demonstrates that Act. D cleavage induces apoptosis in Ohio-HeLa cells. Results shown are

representative of at least 3 independent experiments.

48

Figure 3.8 Act. D treatment leads to cytochrome c release from the mitochondria.

Ohio-HeLa cells were seeded onto sterile coverslips and either left untreated (A) or treated with 5µg/ml

of Act. D (B) for 9hrs before being fixed and incubated with mouse anti cytochrome c primary and Alexa

488 conjugated goat anti mouse secondary antibodies. Coverslips were mounted onto slides using

antifade mounting medium containing DAPI stain. A. Untreated cells show cytochrome c accumulated

within the mitochondria demonstrated by the arrows. B. Image shows cells at various stages of apoptosis

following treatment with Act. D. Arrows demonstrate cytochrome c not yet released from within the

mitochondria. Inset image shows close up of later stage apoptotic cell with the cytochrome c having been

released from the mitochondria and seen diffuse throughout the cytosol. Results shown are representative

of at least 3 independent experiments.

B.

A.

49

3.6 Summary

The results presented in this chapter indicate that infection of Ohio-HeLa cells with

HRV16 does not induce apoptosis and in particular, does not induce apoptosis through

the intrinsic pathway. It was confirmed that apoptosis could be reliably induced in

Ohio-HeLa cells after treatment with Taxol and Act. D. The lack of apoptosis induction

during HRV16 infection was not limited by the amount of virus used in the experiments

nor the time at which the samples were analysed. This was shown by the use of a high

MOI (10) and analysis through to 24hrs p.i. when significant cell death was observed.

50

4 Chapter Four – Results Effect of HRV16 Infection on Chemically Induced Apoptosis

4.1 Introduction

It has previously been observed that some picornaviral species are capable of avoiding

the apoptotic response of the host cell. Previous studies investigating poliovirus have

found that apoptosis is inhibited during productive poliovirus infection (infection under

conditions optimal for maximum viral replication) within HeLa cells suggesting that

poliovirus is capable of actively inhibiting the induction of apoptosis (Tolskaya et al.,

1995; Agol et al., 2000; Belov et al., 2003). The observation that HRV16 infection did

not induce apoptosis suggests the possibility that HRV16, like poliovirus, may be

capable of actively suppressing the host apoptotic pathways.

This chapter aimed to determine if HRV16 infection is capable of actively suppressing

the induction of chemically induced apoptosis.

Ohio-HeLa cells were infected with HRV16 prior to induction of apoptosis via

treatment with Taxol and Act. D. The efficacy of this chemically induced apoptosis was

then investigated.

4.2 Effect of HRV16 Infection on Taxol Induced Apoptosis

Data presented in Chapter three have shown that HRV16 infection does not induce the

intrinsic apoptotic pathway, raising the possibility that HRV16 actively suppresses

apoptosis. Therefore, the ability of HRV16 infection to inhibit chemically induced

apoptosis was investigated. Ohio-HeLa cells were seeded onto sterile coverslips and

incubated overnight to approximately 80% confluence as outlined in section 2.2.1.1

followed by infection with HRV16 with an MOI of 5 as outlined in section 2.2.1.4.

Following infection, cells were either treated with 5µg/ml of Taxol and incubated for

3hrs before being washed and fixed as per the method in section 2.2.2.2, or control cells

were left untreated. Cells were processed for immunofluorescence assay where cells

were incubated with mouse anti VP2 primary followed by Alexa 568 conjugated goat

51

anti mouse secondary antibodies before being imaged and analysed using confocal

microscopy. Interestingly, it was observed that the level of the viral structural protein

VP2 appeared to be lower in cells both infected with HRV16 and treated with Taxol

(see figure 4.1A) when compared with cells that were infected but not treated with

Taxol (figure 4.1B).

To confirm whether or not VP2 was in fact reduced during Taxol treatment, a similar

experiment was carried out, this time utilising western blot analysis following the same

infection and chemical treatment as that used in the previous experiment. Cells were

lysed and processed for western blot before being incubated with the mouse anti VP2

primary followed by the relevant HRP conjugated secondary antibodies as per section

2.2.2.1, with the results shown in figure 4.2. The results shown in figure 4.2 confirm the

observations in immunofluorescence assay (figure 4.1) that VP2 expression is reduced

in HRV16 infected cells treated with Taxol compared to cells infected only,

demonstrating that cells treated with Taxol had reduced levels of VP2. In particular, the

levels of the larger VP2 containing fragments of the viral polyprotein were lower than

in infected cells that were not treated. As the viral polyprotein undergoes a series of

cleavages to produce each of the functional viral proteins, each protein may appear in

various sizes dependant on the level of processing that has occurred. This variety is

demonstrated in figure 4.2 (as well as that in figure 3.2C) where the cells infected with

HRV16 show VP2 present in protein moieties of various sizes. The reduction in the

level of larger polyprotein fragments in Taxol treated cells suggests that the translation

of new polyproteins may be inhibited by Taxol treatment. This observed reduction was

not due to unequal sample loading as even loading is demonstrated by tubulin.

52

Figure 4.1 Taxol treatment during HRV16 infection may reduce expression of VP2.

Ohio-HeLa cells were seeded onto sterile coverslips and infected with HRV16 with MOI of 5 before

being either treated with 5µg/ml of Taxol or left untreated for 3hrs p.i.. Cells were then fixed and

incubated with mouse anti VP2 primary and Alexa 568 conjugated goat anti mouse secondary antibodies.

Coverslips were mounted onto slides using antifade mounting medium containing DAPI stain. A. Cells

were infected but not treated with Taxol. Infected cells are shown as indicated by the arrows with the

presence of the viral VP2 protein shown in red. B. Cells were infected and treated with Taxol. Infected

cells are shown by the arrows with the viral VP2 protein indicated in red. As compared to infected only

cells seen in A., cells treated with Taxol show lower levels of VP2. Results shown are representative of at

least 3 independent experiments.

B.

A.

53

Figure 4.2 Taxol treatment during HRV16 infection may lead to a reduction in the translation of

the viral polyprotein.

Ohio-HeLa cells were infected with HRV16 with an MOI of 5 before being treated with 5µg/ml Taxol for

3hrs. Western blot was incubated with mouse anti VP2 primary and goat anti mouse HRP secondary

antibodies. The first lane shows lysate from uninfected and untreated cells; second lane shows lysate from

infected cells that were not treated; the third lane shows lysate from cells treated with Taxol and infected;

the fourth lane shows lysate from uninfected cells that were treated with Taxol. Results shown are

representative of at least 3 independent experiments.

4.3 Effect of HRV16 Infection on Act. D Induced Apoptosis

Following on from the results demonstrating that infected cells treated with Taxol

resulted in a reduction in the viral VP2 protein, this study focused on the use of Act. D

to determine if HRV16 had the ability to inhibit or disrupt the apoptotic signalling

cascade.

Initially, the effect of Act. D treatment on the expression of VP2 was investigated to

ensure that Act. D treatment did not interfere with the translation of the viral

polyprotein as was the case with Taxol treatment. Ohio-HeLa cells were seeded onto

sterile coverslips and grown to approximately 80% confluence before being infected

with HRV16 at MOI 5 and treated with 5µg/ml of Act. D for 9hrs. Cells were then fixed

and probed with mouse anti VP2 primary antibody followed by Alexa 568 conjugated

goat anti mouse secondary antibody. Figure 4.3 demonstrates that no difference in the

level of VP2 expression could be seen between infected cells with (figure 4.3A) and

54

without (figure 4.3B) treatment with Act. D. This suggests the Act.D treatment does not

interfere with the translation of the viral polyprotein.

To examine if HRV16 infection had the ability to inhibit or reduce the induction of

apoptosis in cells treated with Act. D, cells were seeded onto sterile coverslips and

grown overnight to approximately 80% confluence (see section 2.2.2.2.). Cells were

then washed and infected with HRV16 with MOI of 5 as outlined in section 2.2.1.4.

Cells were then treated with 5µg/ml of Act. D for 9hrs before being washed and fixed

(see section 2.2.2.2) Coverslips were mounted onto slides using antifade mounting

medium containing a DAPI stain before being photographed by confocal microscopy.

The effect of HRV16 infection on Act. D induced apoptosis was measured by

quantitative analysis. A series of representative widefield images were taken of each

sample and the number of apoptotic and healthy cells were counted. Results in figure

4.4 show pie-charts depicting the percentage of apoptotic and healthy cells counted after

each treatment. The results show a marked reduction in the proportion of apoptotic cells

in cells infected with HRV16 prior to treatment with Act. D in comparison to those cells

treated with Act. D alone. It was observed that 73% of cells treated with Act. D were

apoptotic compared to 45% of cells infected with HRV16 prior to Act. D treatment.

These results suggest that HRV16 infection may have to ability to actively inhibit the

induction of apoptosis of the host cell.

55

Figure 4.3 Act. D treatment during HRV16 infection does not reduce expression of VP2.

Ohio-HeLa cells were seeded onto sterile coverslips and infected with HRV16 with MOI of 5 before

being either treated with 5µg/ml of Act. D or left untreated for 9hrs p.i.. Cells were then fixed and

incubated with mouse anti VP2 primary and Alexa 568 conjugated goat anti mouse secondary antibodies.

Coverslips were mounted onto slides using antifade mounting medium containing DAPI. A. Cells were

infected but not treated with Act. D. Infected cells are shown as indicated by the arrows with the presence

of the viral VP2 protein shown in red. B. Cells were infected and treated with Act. D. Infected cells are

shown by the arrows with the viral VP2 protein indicated in red. As compared to infected only cells seen

in A., cells treated with Act. D show no difference in the levels of VP2. Results shown are representative

of at least 3 independent experiments.

B.

A.

56

Figure 4.4 HRV16 infection reduces the induction of apoptosis in Act. D treated cells.

Ohio-HeLa cells were grown on sterile coverslips and either left untreated (mock), infected with HRV16

with MOI of 5, treated with 5µl/ml of Act. D or a combination of both. Cells were then fixed and the

coverslips were mounted onto slide with antifade mounting medium containing DAPI stain. Cells were

imaged with the Nikon Ti Eclipse confocal microscope as described in section 2.2.2.2. Both apoptotic and

healthy cells were counted from a series of representative images taken of each treatment type as

exemplified by the one healthy and two apoptotic cells shown (B). The percentage of each was then

calculated and represented as pie charts above (A). Results shown are representative of at least 3

independent experiments.

57

4.4 Summary

The results presented in this chapter, together with those presented in Chapter 3, suggest

that HRV16 infection does not result in the induction of apoptosis, probably through its

ability to actively suppress apoptotic pathways. Initially it was found that treatment of

HVR16 infected cells with Taxol may inhibit the translation of the viral polyprotein due

to a reduction seen in the levels of VP2 expression. Since Taxol acts to cause cell cycle

arrest by stabilising the microtubules of the cell, it is likely that this stabilisation of the

microtubules may be the cause of the inhibition of the translation of the viral

polyprotein. Experiments investigating apoptosis in infected cells treated with Act. D

demonstrated a reduction in the number of apoptotic cells when compared to uninfected

cells treated with Act. D, suggesting that HRV16 infection may actively inhibit the

apoptotic pathway.

58

5 Chapter Five – Results Rhinovirus Infection Leads to the Indirect Cleavage of RIPK1

5.1 Introduction

Receptor interacting protein kinase 1 (RIPK1, also known as RIP1) is a 74kDa protein

that interacts with Fas and TNRF1 receptors, crucial to the regulation of programmed

cell death (Clement & Stamenkovic, 1994; Stanger et al., 1995; Ting et al., 1996).

Cleavage activation of RIPK1 has been identified as being crucial to TNF-induced

apoptosis, where it has been demonstrated that RIPK1 cleavage is carried out by

activated caspase 8 (Lin et al., 1999). Cleavage of RIPK1 has also been identified in

other apoptotic pathways including TRAIL and Fas induced apoptosis (Lin et al., 1999).

This makes RIPK1 an effective target in the investigation of extrinsic apoptosis during

HRV16 infection.

The picornaviral proteases are known to play a role in the alteration or inhibition of a

number of host cell signalling pathways including translation initiation and

nucleocytoplasmic trafficking (Gustin & Sarnow, 2002; Grubman et al., 2008). This

ability to interact and alter the host signalling pathways make the viral proteases prime

candidates when investigating the possible mechanisms of HRV16 inhibition of

apoptosis. This chapter aimed to investigate the effect of HRV16 infection on extrinsic

apoptosis, using RIPK1 as a marker, and begins to elucidate the possible mechanisms

through which HRV16 infection may interact with the host cell apoptotic pathways,

with a particular focus on the 2A and 3C proteases.

5.2 HRV16 Infection and Act. D Treatment Lead to Dissimilar Cleavage of RIPK1

After the results in chapter three demonstrated that HRV16 infection did not induce

apoptosis, and in particular did not appear to induce intrinsic apoptosis, the involvement

of the extrinsic apoptotic pathway was investigated in an attempt to elucidate the

possible mechanisms of HRV16 suppression of apoptosis as found in chapter four.

RIPK1 is a key component of the TNFR, TRAIL and Fas receptor mediated apoptotic

59

pathways. The binding of TNFα, Fas and other ligands to their receptors promotes

various signalling cascades, including extrinsic apoptosis within which one of the

crucial steps is the cleavage of RIPK1.

Western blot analysis for the cleavage of RIPK1 was carried out as per the methods

outlined in section 2.2.2.1. Briefly, Ohio-HeLa cells were seeded in a 6-well plate in

growth medium overnight at 37°C with 5% CO2 until approximately 80% confluent.

Cells were then either left untreated, infected with HRV16 with an MOI of 5, treated

with 5µg/ml Act. D or a combination of both. Cells were lysed after 9hrs with RIPA

buffer and processed for western blot analysis. The western blot was incubated with the

mouse anti RIPK1 primary and goat anti mouse HRP secondary antibodies. As shown

in figure 5.1, RIPK1 is cleaved during both HRV16 infection and Act. D treatment. The

cleavage patterns observed differ between the treatments such that cleavage of RIPK1

by Act. D treatment results in a cleavage fragment of approximately 40kDa while

cleavage due to HRV16 infection results in a cleavage fragment of approximately

21kDa. Both cleavage fragments can be seen in samples that were infected with HRV16

and treated with Act.D indicating that mechanisms behind each cleavage are most likely

independent of each other. The cleavage observed during Act. D treatment is consistent

with that seen in studies by Lin et al. (Lin et al., 1999) where they described a 42kDa

cleavage fragment resultant from TNF-induced apoptosis, confirming that the cleavage

observed in this study during Act. D treatment is consistent with the induction of

apoptosis.

The demonstration that HRV16 infection induces RIPK1 cleavage demonstrates the

involvement of the extrinsic apoptotic pathway in the host’s response to HRV16

infection. The observation that the cleavage fragments resulting from HRV16 infection

differ from those observed during Act. D treatment, suggests that this alternate cleavage

may play a role in the inhibition of apoptosis by HRV16.

60

Figure 5.1 HRV16 and Act. D treatment leads to cleavage of RIPK1.

Ohio-HeLa cells were either left untreated, infected with HRV16 with an MOI of 5, treated with 5µg/ml

Act. D or both for 9hrs. Western blot was incubated with mouse anti RIPK1 primary and goat anti mouse

HRP secondary antibodies. The arrows on the left of the blot indicate the approximate protein size in

kDA while the arrows on the right demonstrate the RIPK1 protein and the RIPK1 cleavage fragments

resultant from HRV16 infection and Act. D treatment. Results shown are representative of at least 3

independent experiments.

5.3 Cleavage of RIPK1 is Probably Not Carried Out By the 2A or 3C Proteases

The picornaviral proteases 2A and 3C are known to play a role in the alteration of a

number of host cell pathways including translation initiation and nucleocytoplasmic

trafficking via cleavage of specific host cell proteins (Grubman et al., 2008; Gustin &

Sarnow, 2002). Following on from the identification that HRV16 infection leads to

alternate cleavage of RIPK1 compared to Act. D treatment, this study aimed to

determine if the 2A or 3C proteases play a role in this alternate cleavage.

Active and inactivated 2A and 3C proteases were transfected into COS7 cells as

outlined in section 2.2.1.5. Cells were lysed 20h after transfection, and processed for

western blot analysis (as per section 2.2.2.1) where they were initially probed for the

detection of GFP to confirm the efficacy of the transfection. Figure 5.2A shows that the

GFP-3C, GFP-3C inactive and GFP-2A inactive fusion proteins were effectively

61

transfected into the COS7 cells as demonstrated by the larger protein bands in lanes 1, 2

and 4, with GFP alone being approximately 27kDa. It can be seen that the GFP-2A

fusion protein was not detected; this is most likely the result of the active 2A protease

cleaving itself from GFP. The GFP-2A cells were not discounted from this study as it is

known that the 2A protease is highly active and may therefore have had the opportunity

to act within the host cell despite its presence not being confirmed by western blot.

Following confirmation that the transfections were successful, the blot was incubated

with the mouse anti RIPK1 primary antibody followed by the goat anti mouse HRP

secondary antibody. Figure 5.2B demonstrates that no cleavage of RIPK1 had occurred.

The blot was over developed in order to visualise very low levels of cleaved protein.

This resulted in a large amount of background non-specific bands however after

comparison with the results in figure 5.1, no specific cleavage was seen, particularly not

at the points where cleavage was seen during HRV16 infection and Act. D treatment.

These results suggest that the viral 3C protease is not responsible for the cleavage of

RIPK1 seen during HRV16 infection. It is also suggested that the viral 2A protease is

not likely to be responsible for these cleavages either. It can be confirmed that

inactivated 2A protease does not result in RIPK1 cleavage however this study cannot

rule out that active 2A protease may play a role. Due to time restraints, possible

mechanisms behind the cleavage of RIPK1 in HRV16 infected cells could not be further

investigated.

5.4 Expression of 2A and 3C Proteases Does Not Induce Apoptosis

Previous experiments with various picornaviruses have found that the expression of the

viral proteases alone can induce apoptosis of the host cell. The ability of 2A and 3C

protease to induce apoptosis was examined by western blot following the expression of

the active and inactive forms, in transfected cells. The western blot was incubated with

the mouse anti caspase 3 primary antibody followed by the goat anti mouse HRP

secondary antibody. Figure 5.2C shows that neither the expression of the active nor

inactive proteases induced apoptosis as demonstrated by the absence of cleavage of

caspase 3. As with the previous experiment, it cannot be ruled out that active 2A may

have the ability to individually induce apoptosis, time constraints prevented further

investigation during this study.

62

Figure 5.2 HRV16 2A and 3C proteases do not cleave RIPK1 or caspase 3.

COS7 cells were transfected to express active or inactive 2A or 3C proteases as GFP-fusion proteins.

Western blots of the cell lysates are shown above. Arrows on the left of the blots indicate the approximate

protein size in kDa while arrows on the right indicate the position or predicted position of each of the

proteins being analysed. A. Western blot was incubated with the mouse anti GFP primary antibody

followed by the goat anti mouse HRP secondary antibody to determine the efficacy of the transfection. It

can be seen that the GFP-3C, GFP-3C inactive and GFP-2A inactive were effectively transfected. GFP-

2A appears to have been cleaved, with GFP appearing only alone in the GFP-2A transfected cells. B.

Western blot was incubated with mouse anti RIPK1 primary and goat anti mouse HRP secondary

antibodies. As demonstrated by the arrows on the right, there is no evidence of cleavage of RIPK1 as was

seen during HRV16 or Act. D treatment outside of the background bands seen in all treatments. Blot was

over developed to detect any low level cleavages. Background bands seen are consistent with those seen

in figure 5.1 C. Western blot was incubated with the mouse anti caspase 3 primary and goat anti mouse

HRP secondary antibodies. As demonstrated by the arrows on the right, there is no evidence of cleavage

of caspase 3. Results shown are representative of at least 3 independent experiments.

63

5.5 Summary

This study aimed to elucidate the mechanisms with which HRV16 infection may

interact with the host cell apoptotic pathways. Following on from the findings in the

previous chapter that HRV16 infection did not induce early or late intrinsic apoptosis

and that late stage apoptosis was not induce by either pathway, the possibility of the

involvement of the extrinsic apoptotic pathway was investigated.

A wide range of different initiators of extrinsic apoptosis have been identified, all

resulting in differing signalling pathways that ultimately converge to trigger the effector

caspase 3 and the characteristic physical changes unique to apoptotic cell death. In this

study, the cleavage of RIPK1 during HRV16 infection was investigated. RIPK1 is a

protein that is known to be cleaved during TNFR, TRAIL and Fas induced extrinsic

apoptosis. RIPK1 was found to be cleaved during treatment with Act. D resulting in the

detection of an approximately 40kDa sized cleavage fragment, consistent with results

shown during apoptosis by Lin et al (Lin et al., 1999). HRV16 infection also induced

cleavage of RIPK1, however it resulted in a cleavage fragment of approximately 21kDa.

It was investigated whether the viral 2A or 3C proteases may have been responsible for

this alternate cleavage. Transfection with both active and inactive forms of the 2A and

3C proteases did not result in cleavage of RIPK1 nor cleavage of caspase 3, indicating

that infection by HRV16 leads to the indirect cleavage of RIPK1 with neither of the

HRV16 proteases being responsible. The identification that RIPK1 is cleaved during

HRV16 infection alternately to that demonstrated during apoptosis suggests that the

virus may have the ability to disrupt the induction of the extrinsic apoptotic pathway

and thus may have the ability to inhibit the induction of host apoptosis.

Due to time restraints, the mechanisms behind HRV16 cleavage of RIPK1 could not be

elucidated but provides opportunity for future research.

64

6 Chapter Six – General Discussion

6.1 Introduction

HRV is the most common viral cause of upper respiratory tract infections and causes

significant physical and financial burden throughout the world. Whilst illness caused

directly by HRV is mild and self-limiting, the association of HRV infection with severe

asthma and COPD exacerbations, in combination with its high prevalence throughout

all populations, makes it a virus of significant medical and social interest (Johnston et

al., 1995; Seemungal et al., 2000; Greenberg, 2003).

One of the body’s first responses to the onslaught of viral infection is the cellular

induction of apoptosis, triggered to eliminate viruses in order to limit their effect and

minimise their spread. Just as cells have developed immune responses against viral

pathogens, viruses have developed strategies of their own to circumvent or disrupt these

responses. A number of picornavirus species have been found capable of suppressing

the induction of the host apoptotic response. Of particular interest, poliovirus has been

observed as being capable of suppressing the apoptotic response when viral growth

conditions are ideal in order to help facilitate viral growth (Tolskaya, Romanova et al.,

1995; Agol, Belov et al., 2000; Belov, Romanova et al., 2003). There have been very

few studies investigating the effect of HRV infection on host cell apoptosis. Those that

have been performed have reported somewhat contradictory results.

The ability of viruses to circumvent or suppress the apoptotic pathways of host cells

significantly increases their virulence and can have serious health and societal

consequences. This study aimed to identify and provide further clarification of the effect

of HRV16 infection on the host apoptotic pathways so as to provide opportunities for

the identification of potential novel therapeutic targets.

6.2 HRV Infection Does Not Induce Apoptosis

HRV is an RNA virus that undergoes replication and translation of its genome within

the cytoplasm of the host cell. Whilst the HRV genome, like all picornaviruses, consists

of a single strand of RNA, double stranded RNA (dsRNA) is formed as an intermediate

65

of the viral replication process. The presence of dsRNA is known to induce the

apoptotic response in cells as it is not present under normal cellular conditions

(Iordanov et al., 2005). Cells are capable of recognising viral dsRNA through a range of

cellular dsRNA interacting intermediates. One of the pathways through which

exogenous dsRNA can be detected by the cell is through its interaction with the toll-like

receptor 3 (TLR3) (Alexopoulou et al., 2001). TLR3 is activated by the binding of

dsRNA following its entry into the cell via the endocytic pathway (Akira et al., 2006;

Jiang et al., 2008). Following its activation, TLR3 leads to the activation of NF-κB via

RIPK1 which goes on to induce an apoptotic signalling cascade ultimately leading to

cell death (Alexopoulou et al., 2001; Robbins et al., 2003; Koyama et al., 2008).

Cytosolic sensors of dsRNA include the dsRNA dependant kinase (PKR), melanoma

differentiation-associated gene 5 (MDA-5) and retinoic acid-inducible gene I (RIG-I)

(Akira et al, 2006; Koyama et al, 2008). PKR is activated through the binding of

dsRNA and goes on to induce apoptosis through the subsequent activation of eIF-2,

FADD and NF-κB (Levin & London, 1978; Der et al., 1997; Balachandran et al., 1998;

Srivastava et al., 1998; Williams, 1999; Gil et al., 1999). Similarly, MDA-5 and RIG-I

are activated by binding of dsRNA (Koyama et al, 2008). MDA-5 and RIG-I induced

apoptosis is mediated by the interferon-β promoter stimulator 1 (IPS-1) through

activation of FADD and NF-κB (Kawai et al., 2005; Koyama et al., 2008; Lei et al.,

2009). Thus, several cellular pathways activated by the presence of dsRNA lead to the

apoptotic death of the cell.

Various studies have observed that infection with picornaviruses, including HRV,

induces the recognition of viral dsRNA and subsequent activation of the TLR3, PKR,

MDA-5 and RIG-I mediated pathways (Yeung et al., 1999; Kato et al., 2006; Slater et

al., 2010). These findings are consistent with the knowledge that HRV produces dsRNA

as an intermediate during viral replication and the confirmation of its presence during

HRV16 infection demonstrated in this study (see figure 3.4). With the well-established

recognition of dsRNA by various cellular intermediates, which result in a series of

antiviral responses including the induction of apoptosis, it is expected that HRV, like all

RNA viruses, would induce this response. Previous studies have shown apoptosis to be

both induced and suppressed during picornavirus infection, depending on the viral

species (Tolskaya et al., 1995; Buenz & Howe, 2006). Interestingly, it has been

observed that apoptosis is not induced during EMCV and some poliovirus infections

and that these infections are capable of actively suppressing the induction of chemically

66

induced apoptosis (Tolskaya et al., 1995; Romanova et al., 2009). These findings that a

number of picornaviruses are capable of avoiding host cell apoptosis, despite the

presence of their dsRNA replication intermediates, suggest that they have developed

strategies to actively suppress the apoptotic pathways, however the mechanisms behind

this active suppression are yet to be elucidated.

This study found that HRV16 infection did not lead to the induction of apoptosis in

Ohio-HeLa cells (see chapter 3). These results differ from those observed previously

during HRV 1a, 1b and 14 infections, where it was observed that apoptosis was induced

as demonstrated by the cleavage of caspase 3, the caspase substrate PARP, and release

of cytochrome c from the mitochondria (Taimen et al 2004; Deszcz et al., 2005; Drahos

& Racaniello, 2009). The different results found in chapter 3, in comparison to those

found with HRV1a and HRV1b, may be explained by the fact that these viruses are

minor group viruses whilst HRV16 used in this study belongs to the major group. The

main difference between the major and minor groups is their use of cellular receptors

during entry into the host cell (Tomassini et al., 1989; Hofer et al., 1994). This

difference may play a role in the alternate results seen however further investigations

are required where minor and major group viruses are analysed in parallel for their

apoptosis inducing ability. Studies investigating the major group HRV14 have found

contradicting results, with one study observing cleavage of the caspase substrate PARP

and DNA fragmentation characteristic of apoptosis, whilst the other observed these not

to have occurred (Gustin & Sarnow, 2002; Deszcz et al., 2005). The differences

observed in these studies may be explained by the varying levels of virus used during

infection. Deszcz et al used an MOI of 100 during infection, which may have been

responsible for the induction of apoptosis that was observed due to the saturation of

virus particles present (Deszcz et al., 2005). In contrast, Gustin and Sarnow used an

MOI of 10 during their infections, a level that is more comparable with natural infection

doses (Gustin & Sarnow, 2002). In this study, viral infections were carried out with an

MOI of between 0.1 and 10, comparable to that used by Gustin and Sarnow, as were the

results observed (Gustin & Sarnow, 2002).

This study also demonstrated that not only was apoptosis not observed during HRV16

infection, but that it does not appear as though the intrinsic apoptotic pathway is

induced. This was demonstrated by caspase 9 not being cleaved and cytochrome c not

being released from the mitochondria (see chapter 3). This is in contrast to results

observed by Deszcz et al., who suggested that the intrinsic apoptotic pathway was

67

induced during HRV14 infection (Deszcz et al., 2005). As mentioned above, very high

levels of virus were used during the study carried out by Deszcz et al. particularly in

comparison to those used in this study, which may have had an indirect role in inducing

the intrinsic apoptotic pathway (Deszcz et al., 2005).

6.3 HRV16 Inhibits Apoptosis

The results in chapter 4 suggest that HRV16 infection not only avoids the induction of

host apoptosis, but that it may actively suppress the apoptotic pathways of the host cell.

Ohio-HeLa cells infected with HRV16 prior to treatment with Act. D resulted in a

reduced number of apoptotic cells compared to those treated with Act. D alone. The

observation that both very early and late intrinsic apoptosis were not induced during

infection (chapter 3), raises the possibility that the extrinsic apoptotic pathways may be

associated with HRV infection, and potentially other picornaviruses. The fact that

caspase 3 was not cleaved during HRV16 infection suggests that if extrinsic apoptosis

was induced, it was halted at an early stage of the pathway (refer to figure 1.4). This

study demonstrated that RIPK1, an intermediate of the extrinsic apoptotic pathway, was

cleaved during HRV16 infection differently to that seen during apoptosis induced by

Act. D (chapter 5). RIPK1 is a protein of 74kDa, that interacts with the intracellular

domains of the extrinsic apoptotic receptors Fas and TNFR1, resulting in an apoptotic

response following cleavage by caspase 8 (Stanger et al., 1995; Grimm et al., 1996; Lin

et al., 1999). It has previously been observed that cleavage of RIPK1 occurred during

TRAIL and Fas induced apoptosis resulting in a 42kDa cleavage fragment similar to

that observed in this study in cells treated with Act. D (Lin et al., 1999). The cleavage

fragment observed during HRV16 infection was smaller, at approximately 21kDa

(figure 5.1). This alternate cleavage of RIPK1 may play a role in the potential

suppression of apoptosis by HRV16 infection. The involvement of RIPK1 also suggests

that the extrinsic apoptotic pathway may be implicated in the response of Ohio-HeLa

cells to HRV16 infection.

Interestingly, the cleavage of RIPK1 by Act. D occurs after a short length of treatment,

whist induction by the intrinsic pathway, demonstrated in chapter 3 by the release of

cytochrome c from the mitochondria, occurs only after long treatment times (9hrs

incubation was used in this study). This suggests that Act. D treatment directly induces

68

the extrinsic apoptotic pathway whilst the effect of Act. D on the cellular transcription

may, indirectly, be the cause of intrinsic apoptotic induction.

During this study, it was observed that treatment with Taxol may inhibit the translation

of the HRV polyprotein (chapter 4). Results shown in chapter 4 demonstrate a reduction

in the expression of VP2, with western blot analysis demonstrating a reduction in the

amount of larger VP2 containing polyprotein lengths present in cells treated with Taxol

after infection. These results suggest that processing of the polyprotein was not affected

but that new polyprotein synthesis was inhibited. Taxol acts to stabilise the

microtubules of the cell (Schiff et al., 1979; Rowinsky et al., 1988; Woods et al., 1995).

The results shown in this study suggest that HRV16 infection may utilise the

microtubules of the host cell during translation. It has previously been identified that the

microtubules are utilised by poliovirus for release of the virus from the cell (Taylor et

al., 2009), whilst it has also been observed that HRV utilises the microtubules for

transport of proteins throughout the cell (Grassme et al, 2005). This supports the

possibility that there is an important role microtubules may play in HRV16 polyprotein

translation.

6.4 HRV16 Infection Leads to Indirect Cleavage of RIPK1

Based on previous evidence demonstrating the ability of picornavirus proteases 2A and

3C to cleave or alter a number of cellular components, it was thought likely that these

proteases may be responsible for the alternate cleavage of RIPK1 observed during HRV

infection. Results shown in chapter five demonstrate that cells expressing the HRV16

2A and 3C proteases do not result in cleavage of RIPK1. These results confirmed that

the 3C viral protease is not responsible for the alternate RIPK1 cleavage, however the

potential role of 2A could not be ruled out as the presence of GFP-2A could not be

confirmed by western blot analysis due to the highly active nature of the protease

resulting in it cleaving itself from GFP. Cleavage of RIPK1 by 2A is unlikely however,

as when the translation sequence for the RIPK1 protein was analysed through a

picornaviral protease cleavage site predictor (NetPicoRNA 1.0), no potential 2A

cleavage sites were identified. Two potential 3C cleavage sites were identified at the

89th

and 525th

amino acids, neither of which aligned with the cleavage of RIPK1 seen

during HRV16 infection. Whilst it has been shown here that the HRV proteases are not

responsible for the cleavage of RIPK1, further investigation is required to determine the

69

exact mechanisms behind this cleavage and its potential role in the suppression of

apoptosis during HRV infection.

6.5 Conclusion

Despite a number of studies performed to investigate the induction of apoptosis during

infection with a variety picornaviruses, very little is known about the effect of HRV on

host cell apoptosis. The limited number of studies carried out to date have shown mixed

results, but a majority have suggested that HRV infection induces host cell apoptosis.

The results presented in this study show that host cell apoptosis is not only avoided

during HRV16 infection, but that the virus is capable of actively suppressing it. The

identification that HRV16 infection did not induce the intrinsic apoptotic pathway, but

that it led to the cleavage of RIPK1 not consistent with apoptotic cleavage, suggests that

the virus acts to inhibit the extrinsic apoptotic pathway. Whilst further investigation of

the exact mechanisms behind the cleavage of RIPK1 and suppression of extrinsic

apoptosis are required, these results provide a basis for future studies and an opportunity

for potential new therapeutic targets to be identified.

70

References Cited

Agol, V. I., G. A. Belov, et al. (2000). Competing death programs in poliovirus-infected

cells- commitment switch in the middle of the infectious cycle. Journal of

Virology 74(12): 5534-5541.

Akira, S., S. Uematsu and O. Takeuchi. (2006) Pathogen recognition and innate

immunity. Cell 124: 783-801.

Alexopoulou, L., A. C. Holt, et al. (2001) Recognition of double-stranded RNA and

activation of NF-κB by Toll-like receptor 3. Nature 413: 732-738.

Balachandran, S., C. N. Kim, et al. (1998) Activation of the dsRNA-dependant protein

kinase, PKR, induces apoptosis through FADD-mediated death signalling. The

EMBO Journal 17(23): 6888-6902.

Barber, G. N. (2001). Host defense, viruses and apoptosis. Cell Death and

Differentiation 8: 113-126.

Barco, A., E. Feduchi, et al. (2000). Poliovirus protease 3C kills cells by apoptosis.

Virology 266: 352-360.

Belov, G. A., L. I. Romanova, et al. (2003). The major apoptotic pathway activated and

suppressed by poliovirus. Journal of Virology 77(1): 45-56.

Belsham, G. J., G. M. McInerney and N. Ross-Smith. (2000) Foot-and-mouth disease

virus 3C protease induces cleavage of translation factors eIF4A and eIF4G within

infected cells. J. Virol. 74(1): 272-280.

Bhalla, K., A. M., Ibrado, et al. (1993) Taxol induces internucleosomal DNA

fragmentation associated with programmed cell death in human myeloid leukemia

cells. Leukemia 7(4): 563-568.

Blagosklonny, M. V., T., Schulte, et al. (1996) Taxol-induced Apoptosis and

Phosphorylation of Bcl-2 Protein Involves c-Raf-1 and Represents a Novel c-Raf-

1 Signal Transduction Pathway. Cancer Research 56: 1851-1854.

Bodian, D. (1955) Emerging concept of poliomyelitis infection. Science 122: 105-108.

Buenz, E. J. and C. L. Howe (2006). Picornaviruses and cell death. Trends in

Microbiology 14(1): 28-36.

Campanella, M., A. S. de Jong, et al. (2004). The coxsackievirus 2B protein suppresses

apoptotic host cell responses by manipulating intracellular Ca2 homeostasis. The

Journal of Biological Chemistry 279(18): 18440-18450.

Carocci, M., N. Cordonnier, et al. (2011). Encephalomyocarditis virus 2A protein is

required for viral pathogenesis and inhibition of apoptosis. Journal of Virology

85(20): 10741-10754.

71

Carstens, E. B. and L. A. Ball (2009). Ratification vote on taxonomic proposals to the

International Committee on Taxonomy of Viruses (2008). Archives of Virology

154: 1181-1188.

Carthy, C. M., B. Yanagawa, et al. (2003). Bcl-2 and Bcl-xL overexpression inhibits

cytochrome c release, activation of multiple caspases, and virus release following

coxsackievirus B3 infection. Virology 313: 147-157.

Clement, M. V. and I. Stamenkovic (1994) Fas and tumour necrosis factor receptor-

mediated cell death: similarities and distinctions. Journal of Experimental

Medicine 180: 557-567.

Der, S. D., Y. L. Yang, et al. (1997) A double-stranded RNA-activated protein-kinase

dependant pathway mediating stress-induced apoptosis. Proc. Natl. Acad. Sci.

USA 94: 3279-3283.

Deszcz, L., E. Gaudernak, et al. (2005). Apoptotic events induced by human rhinovirus

infection. Journal of General Virology 86: 1379-1389.

Drahos, J. and V. R. Racaniello (2009). Drahos & racaniello 2009 Cleavage of IPS-1 in

cells infected with human rhinovirus. Journal of Virology 83(22): 11581-11587.

Galluzzi, L., C. Brenner, et al. (2008). Viral control of mitochondrial apoptosis. PLoS

Pathogens 4(5): e1000018.

Ghildyal, R., H. Dagher, et al. (2005) Rhinovirus infects primary human airway

fibroblasts and induces a neutrophil chemokine and a permeability factor. J. Med.

Virol. 75: 608-615.

Ghildyal, R., B. Jordan, et al. (2009) Rhinovirus 3C protease can localise in the nucleus

and alter active and passive nucleocytoplasmic transport. J. Virol. 83: 7349-7352.

Gil, J., J. Alcami and M. Esteban. (1999) Induction of apoptosis by double-stranded

RNA-dependant protein kinase (PKR) involves the α unit of eukaryotic translation

initiation factor 2 and NF-κB. Mol. Cell Biol.19(7): 4653.

Gil, J. and M. Esteban. (2000) Induction of apoptosis by the dsRNA-dependent protein

kinase (PKR): mechanism of action. Apoptosis 5: 107-114.

Goldberg, I. H., M. Rabinowitz and E. Reich. (1962) Basis of actinomycin action, I.

DNA binding and inhibition of RNA-polymerase synthetic reactions by

actinomycin. Proc. Natl. Acad. Sci. 48: 2094-2101.

Gomes, R., R. Guerra-Sa, et al. (2010). Coxsackievirus B5 induced apoptosis of HeLa

cells : Effects on p53 and SUMO. Virology 396: 256-263.

Gradi, A., Y. V. Svitkin, et al. (1998). Proteolysis of human eukaryotic translation

initiation factor eIF4GII but not eIF4GI coincides with the shutoff of host protein

synthesis after poliovirus infection. Proceedings of the National Academy of

Science USA 95: 11089-11094.

Gradi, A., Y. V. Svitkin, et al. (2003). Human rhinovirus 2A proteinase cleavage sites in

eukaryotic initioation factors eIF4Gi and eIF4GII are different. Journal of

Virology 77(8): 5026-5029.

72

Grassme, H., A. Riehle, et al. (2005) Rhinoviruses infect human epithelial cells via

ceramide-enriched membrane platforms. J. Biol. Chem. 280: 26256-26262.

Greenberg, S. B. (2003). Respiratory consequences of rhinovirus infection. Archives of

Internal Medicine 163: 278-284.

Grimm, S., B. Z. Stanger and P. Leder. (1996) RIP and FADD: two “death domain” –

containing proteins can induce apoptosis by convergent, but dissociable,

pathways. Proc. Nat. Acad. Sci. USA 93: 10923-10927.

Grubman, M. J., M. P. Moraes, et al. (2008). Evading the host cell immune response -

how foot and mouth disease virus has become an effective pathogen. FEMS

Immunology and Medical Microbiology 53: 8-17.

Gustin, K. E. and P. Sarnow (2002). Inhibition of nuclear import and alteration of

nuclear pore complex composition by rhinovirus. Journal of Virology 76(17):

8787-8796.

Haldar, S., N., Jena & C. M. Croce. (1995) Inactivation of Bcl-2 by phosphorylation.

Proc. Natl. Acad. Sci. USA 92: 4507-4511.

Harris, J. M., II and J. M. Gwaltney, Jr. (1996). Incubation periods of experimental

rhinovirus infection and illness. Clinical Infectious Diseases 23: 1287-1290.

Hendley, J. O., R. P. Wenzel and J. M. Gwaltney. (1973) Transmission of rhinovirus

colds by self-inoculation. New England Journal of Medicine 288(26): 1361-1364.

Hendley, J. O. and J. M. Gwaltney, Jr. (2004). Viral titers in nasal lining fluid compared

to viral titers in nasal washes during experimental rhinovirus infection. Journal of

Clinical Virology 30: 326-328.

Hofer, F., M. Gruenberger, et al. (1994). Members of the low density lipoprotein

receptor family mediate cell entry of a minor-group common cold virus.

Proceedings of the National Academy of Science USA 91: 1839-1842.

Hollinger, F. B. and S. U. Emerson (2007). Hepatitis A Virus. Fields Virology 5th

Edition. D. M. Knipe and P. M. Howley. Philadelphia PA, Lippincott Williams &

Wilkins. I: 912-947.

Iordanov, M. S., O. P. Ryabinina, et al. (2005) Two mechanisms of caspase 9

processing in double-stranded RNA and virus triggered apoptosis. Apoptosis 10:

153-166.

Jiang, Q., H. Wei and Z. Tian. (2008) Poly I:C enhances cycloheximide-induced

apoptosis of tumor cells through TLR3 pathway. BMC Cancer 8: 12.

Johnston, S. L., P. K. Philip, et al. (1995) Community study of role of viral infections in

exacerbations of asthma in 9-11 year old children. BMJ 310: 1225-1228.

Kato, H., O. Takeuchi, et al. (2006) Differential roles of MDA5 and RIG-1 helicases in

the recognition of RNA viruses. Nature 441(4): 101.

Kawai, T., K. Takahashi, et al. (2005) IPS-1, an adaptor triggering RIG-1 and MDA5-

mediated type I interferon induction. Nature Immunology 6(10): 981.

73

King, A. M. Q., E. Lefkowitz, et al., Eds. (2012). Virus Taxonomy: Ninth Report of the

International Committee on Taxonomy of Viruses. London, Elsevier.

Koyama, S., K. J. Ishii, et al. (2008) Innate immune response to viral infection.

Cytokine

Kuyumcu-Martinez, N. M., M. Joachims, et al. (2002). Efficient cleavage of ribosome

associated poly(A) binding protein by enterovirus 3C protease. Journal of

Virology 76(5): 2062-2074.

Lee, W. M., S. S. Monroe and R. R. Rueckert. (1993) Role of maturation cleavage in

infectivity of picornaviruses: activation of an infectosome. J. Virol. 67:2110-2122.

Lei, Y., C. B. Moore, et al. (2009) MAVS-mediated apoptosis and its inhibition by viral

proteins. PLoS ONE 4(5): e5466.

Levin, D. and I. M. London. (1978) Regulation of protein synthesis: activation by

double-stranded RNA of a protein kinase that phosphorylates eukaryotic initiation

factor-2. Proc. Natl. Acad. Sci. USA 75(3): 1121-1125.

Li, M.-L., T.-A. Hsu, et al. (2002). The 3C protease activity of Enterovirus 71 induces

human neural cell apoptosis. Virology 293: 386-395.

Micheau, O. and J. Tschopp. (2003) Induction of TNF receptor I-mediated apoptosis via

two sequential signalling complexes. Cell 114: 181-190.

Minor, P. D. (2012). The polio-eradication programme and issues of the end game.

Journal of General Virology 93: 457-474.

McGuire, W. P., E. K., Rowinsky, et al. (1989) Taxol: a unique antineoplastic agent

with significant activity in advanced ovarian epithelial neoplasms. Ann. Intern.

Med.111:273-279.

Nomoto, A., Y. F. Lee, et al. (1976). The 5' end of poliovirus mRNA is not capped with

m7G(5')ppp(5')Np. Proceedings of the National Academy of Science USA 73(2):

375-380.

Novak, J. E. and K. Kirkegaard (1991). Improved method for detecting poliovirus

negative strands used to demonstrate specificity of positive strand encapsidation

and the ratio of positive to negative strands in infected cells. Journal of Virology

65(6): 3384-3387.

Pallansch, M. and R. Roos (2007). Enteroviruses: Polioviruses, Coxsackieviruses,

Echoviruses and Newer Enteroviruses. Fields Virology 5th Edition. D. M. Knipe

and P. M. Howley. Philadelphia PA, Lippincott Williams & Wilkins. I: 840-893.

Patick, A. K. and K. E. Potts (1998). Protease inhibitors as antiviral agents. Clinical

Microbiology Reviews 11(4): 614-627.

Perez, L. and L. Carrasco (1993). Entry of Poliovirus into cells does not require a low

pH step. Journal of Virology 67(8): 4543-4548.

Pettersson, R. F., V. Ambros, et al. (1978). Identification of a protein linked to nascent

poliovirus RNA and to the polyuridylic acid of negative strand RNA. Journal of

Virology 27(2): 357-365.

74

Racaniello, V. R. (2007). Picornaviridae: The viruses and their replication. Fields

Virology 5th Edition. D. M. Knipe and P. M. Howley. Philadelphia PA,

Lippincott Williams & Wilkins. I: 796-839.

Reich, E., R. M. Franklin, et al. (1962) Action of actinomycin D on animal cells and

viruses. Proc. Natl. Acad. Sci. 48: 1238-1245.

Rhoades, R. E., J. M. Tabor-Godwin, et al. (2011). Enterovirus infections of the central

nervous system. Virology 411(2): 288-305.

Richard, A. and D. Tulasne (2012). Caspase cleavage of viral proteins, another way for

viruses to make the best of apoptosis. Cell Death and Differentiation 3: e277.

Robbins, M. A., L. Maksumova, et al. (2003) Nuclear factor-κB translocation mediates

double-stranded ribonucleic acid-induced NIT-1 β-cell apoptosis and up-regulates

caspase-12 and tumor necrosis factor-associated ligand (TRAIL). Endocrinology

144(10): 4616-4625.

Romanova, L. I., P. V. Lidsky, et al. (2009). Antiapoptotic activity of the cardiovirus

leader protein, a viral security protein. Journal of Virology 83: 7273-7284.

Rowinsky, E. K., R. C., Donehower, et al. (1988) Microtubule changes and cytotoxicity

in leukemic cell lines treated with taxol. Cancer Research 48: 4093-4100.

Schiff, P. B., Fant, J. & Horwitz, S. B. (1979) Promotion of microtubule assembly in

vitro by taxol. Nature 277: 665-667.

Schober, D., P. Kronenberger, et al. (1998) Major and minor group human rhinoviruses

penetrate from endosomes by different mechanisms. J. Virol. 72(2): 1354-1364.

Schwarz, E. M., C. Badorff, et al. (1998). NF-kB-mediated inhibition of apoptosis is

required for encephalomyocarditis virus virulence. Journal of Virology 72(7):

5654-5660.

Seemungal, T. A. R., R. Harper-Owen, et al., (2000) Detection of rhinovirus in induced

sputum at exacerbation of chronic obstructive pulmonary disease. Eur. Respir. J.

16: 677-683.

Slater, L., N. W. Bartlett, et al. (2010) Co-ordinated role of TLR3, RIG-1 and MDA5 in

the innate response to rhinovirus in bronchial epithelium. PLOS Pathogens 6(11):

e1001178.

Srivastava, S. P., K. U. Kumar and R. J. Kaufman. (1998) Phosphorylation of

eukaryotic translation initiation factor 2 mediates apoptosis in response to

activation of the double-stranded RNA-dependant protein kinase. J. Biol. Chem.

273: 2416-2423.

Stanger, B. Z., P. Leder, et al. (1995). RIP: A novel protein containing a death domain

that interacts with Fas/APO-1 (CD95) in yeast and causes cell death. Cell 81: 513-

523.

Stanway, G. and T. Hyypia (1999). Parechoviruses. Journal of Virology 73(7): 5249-

5254.

Taimen, P., H. Berghall, et al. (2004) NuMA and nuclear lamins are cleaved during

viral infection – inhibition of caspase activity prevents cleavage and rescues HeLa

75

cells from measles virus-induced but not rhinovirus 1B-induced cell death.

Virology 320:85-98.

Taylor, M. P., T. B. Burgon, et al. (2009). Role of microtubules in extracellular release

of poliovirus. Journal of Virology 83(13): 6599-6609

Ting, A. T., F. X. Pimentel-Muinos, et al. (1996). RIP mediates tumour necrosis factor

receptor 1 activation of NF-κB but not Fas/APO-1 initiated apoptosis. The EMBO

Journal 15: 6189-6196.

Tolskaya, E. A., L. I. Romanova, et al. (1995). Apoptosis-inducing and apoptosis-

preventing functions of poliovirus. Journal of Virology 69(2): 1181-1189.

Tomassini, J. E., D. Graham, et al. (1989). cDNA cloning reveals that the major group

rhinovirus receptor on HeLa cells is intracellular adhesion molecule-1.

Proceedings of the National Academy of Science USA 86: 4907-4911.

Turner, R. B. and R. B. Couch (2007). Rhinoviruses. Fields Virology 5th Edition. D. M.

Knipe and P. M. Howley. Philidelphia PA, Lippincott Williams & Wilkins. I.

Vining, L. C. and S. A. Waksman. (1954) Paper chromatographic identification of the

actinomycins. Science 120:389-390.

Walker, E. J., P. Younessi, et al. (2013) Rhinovirus 3C protease facilitates specific

nucleoporin cleavage of nuclear proteins in infected host cells. PLOS 8(8):

e71316.

Wani, M. C., H. L., Taylor, et al. (1971) Plant antitumor agents. VI. Isolation and

structure of taxol, a novel antileukemic and antitumor agent from Taxus

brevifolia J. Am. chem. Soc. 93: 2325–2327.

Wark, P. A. B., S. L. Johnston, et al. (2005). Asthmatic bronchial epithelial cells have a

deficient innate immune response to infection with rhinovirus. Journal of

Experimental Medicine 201(6): 937-947.

Williams, B. R. G. (1999) PKR; A sentinel kinase for cellular stress. Oncogene 18:

6112-6120.

Woods, C. M., J., Zhu, et al. (1995) Taxol-induced mitotic block tirggers rapid onset of

a p53-independent apoptotic pathway. Mol. Med. 1(5): 506-526.

Yeung, M. C., D. L. Chang, et al. (1999) Inhibitory role of the host apoptogenic gene

PKR in the establishment of persistent infection by encephalomyocarditis virus in

U937 cells. PNAS 96(21): 11860-11865.

Ypma-Wong, M. F. and B. L. Semler (1987). In vitro molecular genetics as a tool for

determining the differential cleavage specificities of the poliovirus 3C proteinase.

Nucleic Acids Research 15(5): 2069-2088.