213
The Effective Contribution of Viral Respiratory Infection to Wheezing Illness in Hospitalised Young Children Chisha Teza Sikazwe BSc (Biomedical Sciences and Molecular biology) Master of Infectious Diseases This thesis is presented for the degree of Doctor of Philosophy in Microbiology of The University of Western Australia School of Biomedical Sciences 2018

Chisha Teza Sikazwe - University of Western Australia · I, Chisha Teza Sikazwe, certify that: This thesis has been substantially accomplished during enrolment in the degree. This

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

  • The Effective Contribution of Viral Respiratory Infection to Wheezing Illness

    in Hospitalised Young Children

    Chisha Teza Sikazwe

    BSc (Biomedical Sciences and Molecular biology)

    Master of Infectious Diseases

    This thesis is presented for the degree of Doctor of Philosophy in Microbiology of

    The University of Western Australia

    School of Biomedical Sciences

    2018

  • ii

  • iii

    Thesis declaration

    I, Chisha Teza Sikazwe, certify that:

    This thesis has been substantially accomplished during enrolment in the degree.

    This thesis does not contain material which has been accepted for the award of any

    other degree or diploma in my name, in any university or other tertiary institution.

    No part of this work will, in the future, be used in a submission in my name, for an y

    other degree or diploma in any university or other tertiary institution without the

    prior approval of The University of Western Australia and where applicable, any

    partner institution responsible for the joint-award of this degree.

    This thesis does not contain any material previously published or written by another

    person, except where due reference has been made in the text.

    The work(s) are not in any way a violation or infringement of any copyright,

    trademark, patent, or other rights whatsoever of any person.

    Technical assistance was kindly provided by Tom Chung for Cytokine multiplex bead

    experiment that is described in Chapter seven of this thesis.

    This thesis contains only sole-authored work, some of which has been published

    and/or prepared for publication under sole authorship.

    XChisha Teza Sikazwe

    Date: 25.06.2018

  • iv

    Abstract

    Respiratory tract infections are a leading cause of morbidity and mortality worldwide. Our

    emerging understanding of the importance of acute lower respiratory viral infe ction in

    early childhood predisposing to chronic inflammatory respiratory disease, coupled with the

    complex interplay between virus and host underscores the need for investigations to

    understand the clinical manifestation and the interplay between the magnitude of

    infection and the corresponding host response. Rhinoviruses, specifically RV -C have been

    identified as an important contributor to wheezing illness in paediatric medicine. Though,

    little is known about its contribution to infection. Thus, this the sis aims to improve

    understanding of the underlying pathophysiological mechanisms in the context of RV -C

    wheezing illness through the development of a reliable method of quantifying RV -C load

    and characterisation of the host response following infection.

    RV-C was the most common virus detected in preschool aged children hospitalised with an

    acute asthma exacerbation (Chapter 6). This was also apparent in preschool aged non-

    asthmatic children hospitalised with a wheezing illness (chapter 7). Interestingly, i n young

    infants under the age of two years hospitalised (Chapter 5) with a wheezing illness, RSV

    rather than RV-C was found to be the most common virus detected. Viral load studies

    revealed that both RSV and RV-C replicate to significantly higher levels in hospitalised

    patients than in non-respiratory disease controls. In asthmatics and non-asthmatic

    patients, RV-C induced respiratory wheeze appears to be driven by a Th2 response that is

    independent of viral load. The magnitude of the host immune response is more apparent

    in children with asthma compared to those without asthma. In addition, RV-C infection in

    pre-school aged children with wheeze appears to induce a selective increase in neutrophil

    chemokines and a significant increase in neutrophil numbers with a simultaneous increase

    in illness severity.

  • v

    The findings in this thesis extend the existing knowledge on RV-C mediated wheezing

    illness and demonstrate that magnitude of replication does not significantly contribute to

    disease outcome. Conversely, it appears that the host immune response for which in part

    is driven by a pro-inflammatory, neutrophilic inflammation may play a substantial role in

    severity of disease. Host directed therapy targeting neutrophil pathways may prove to be

    beneficial in young children infected with RV-C.

  • vi

    Table of contents

    Thesis declaration ......................................................................................................... iii

    Abstract........................................................................................................................ iv

    List of Figures................................................................................................................. x

    List of Tables ................................................................................................................ xii

    Acknowledgments....................................................................................................... xiii

    AUTHORSHIP DECLARATION: SOLE AUTHOR PUBLICATIONS .................................... xv

    Peer reviewed papers and Conference presentations .................................................... xvi

    List of Abbreviations................................................................................................... xvii

    1 Literature review .................................................................................................... 2

    1.1 Introduction ............................................................................................ 3

    1.1.1 Respiratory syncytial virus ........................................................................ 6

    1.1.2 Rhinoviruses ............................................................................................ 6

    1.1.3 Human Metapneumovirus ........................................................................ 8

    1.1.4 Influenza virus ......................................................................................... 8

    1.1.5 Parainfluenza virus ................................................................................... 9

    1.1.6 Adenovirus ............................................................................................ 10

    1.1.7 Human Coronaviruses ............................................................................ 10

    1.2 Laboratory Diagnosis of Respiratory Infections ................................................... 11

    1.2.1 Upper and Lower airway samples............................................................ 12

    1.2.2 Virus isolation by cell culture .................................................................. 13

    1.2.3 Direct detection of respiratory viruses by immunofluorescence ................ 14

    1.2.4 Nucleic Acid Tests .................................................................................. 15

    1.3 Viral kinetics of acute respiratory tract infections................................................ 19

    1.4 Viral respiratory infection and asthma................................................................ 20

    1.5 Innate immune response to viral respiratory infection ........................................ 23

  • vii

    1.6 Aims of Project ................................................................................................. 26

    2 Materials and Methods ......................................................................................... 27

    2.1 Sample collection .............................................................................................. 27

    2.2 Nucleic acid extraction and viral detection.......................................................... 28

    2.3 Design of primers and probes ............................................................................ 29

    2.4 Production and quantification of transcribed RNA standards ............................... 31

    2.5 Quantitative Real time PCR (Viral load)............................................................... 32

    2.6 Digital Droplet PCR ............................................................................................ 33

    2.7 PCR reagents .................................................................................................... 34

    2.8 Gel Electrophoresis ........................................................................................... 34

    2.9 Thermocyclers .................................................................................................. 34

    2.10 Computational analysis ..................................................................................... 35

    2.11 Multiplex immunoassay..................................................................................... 35

    2.12 Quality Control ................................................................................................. 36

    2.13 Statistical Analysis ............................................................................................. 36

    2.14 Ethics Approval ................................................................................................. 36

    3 The design and development of quantitative detection assays for the common

    causative viral pathogens of acute lower respiratory tract infection ................................ 38

    3.1 Introduction ..................................................................................................... 39

    3.2 Samples............................................................................................................ 41

    3.3 Results ............................................................................................................. 41

    3.4 Discussion......................................................................................................... 55

    4 The development of a reliable PCR assay to measure RV-C load in clinical samples ... 61

    4.1 Introduction ..................................................................................................... 62

    4.2 Samples............................................................................................................ 64

    4.3 Results ............................................................................................................. 66

    4.4 Discussion......................................................................................................... 75

    5. The Quantitative Detection of Respiratory Syncytial Virus in Hospitalized Young South

    African Children ........................................................................................................... 78

    5.1 Introduction ..................................................................................................... 79

    5.2 Samples............................................................................................................ 81

    5.3 Results ............................................................................................................. 82

    5.3.1 Baseline characteristics .......................................................................... 82

    5.3.2 RSV infection and clinical outcome .......................................................... 84

  • viii

    5.3.3 RSV disease and HIV infection ................................................................. 87

    5.3.4 RSV load .................................................................................................... 88

    5.4 Discussion......................................................................................................... 91

    6 Determinants of acute asthma exacerbation severity following RV-C infection ......... 95

    6.1 Introduction ..................................................................................................... 96

    6.2 Samples............................................................................................................ 97

    6.3 Results ............................................................................................................. 99

    6.3.1 Study participants characteristics ............................................................ 99

    6.3.2 Virus Detection ...................................................................................... 99

    6.3.3 RV-C load..............................................................................................101

    6.3.4 Surrogate markers of inflammation........................................................104

    6.3.5 Performance of RV-C load, neutrophils and eosinophils in predicting the

    severity AAE ........................................................................................................108

    6.4 Discussion........................................................................................................109

    7. Cytokine profiles in nasal secretions of patients hospitalised with Rhinovirus Species C

    associated respiratory wheeze .....................................................................................115

    7.1 Introduction ....................................................................................................116

    7.2 Samples...........................................................................................................117

    7.3 Results ............................................................................................................119

    7.3.1 Virus Detections....................................................................................119

    7.3.2 Nasal cytokine profiles of wheezing patients following RV-C infection ......124

    7.3.3 Relationships between cytokines, RV-C load and clinical outcomes ..........132

    7.4 Discussion........................................................................................................135

    8. General Discussion and Conclusions......................................................................142

    8.1 Introduction .........................................................................................143

    8.2 Epidemiology of respiratory viruses in young children under the age of five

    years 145

  • ix

    8.3 Reliable methods of accurately determining viral load in RV-C infected

    patients 148

    8.4 Viral Determinants of severity of RV-C induced wheezing illness ..............150

    8.6 Conclusion .......................................................................................................154

    Bibliography ...............................................................................................................156

    Appendices .................................................................................................................176

    Appendix 1 ..........................................................................................................176

    Appendix 2 .................................................................................................................181

    Published work completed during PhD .........................................................................181

  • x

    List of Figures

    Figure 4.3-1 A BioEdit sequence alignment of primer and probe regions that were targeted

    by assays one to four. Sequences of forward primer region (left box), probe region (center

    box) and reverse primer region (right box). Identical bases at the same position are

    represented by dots whereas capitalized bases indicate mismatches between sequences.

    ................................................................................................................................... 66

    Figure 4.3-2 the algorithm for the determination of RV-C viral load in clinical samples ..... 72

    Figure 4.3-3: Box plots of RV-C load in samples from young children presenting to the

    Emergency Department with acute wheeze. .................................................................. 74

    Figure 5.3-1: Viruses detected in NPAs collected from ALRI cases and NRD controls. RV

    (RV-A, RV-B, RV-C), HCoV (OC43, 229E, HKU-1, NL63), HPIV (PIV I-IV), IFV (A/H1N1, A/H3N2,

    B and C) ....................................................................................................................... 84

    Figure 5.3-2: The distribution of RSV positive and RSV-negative ALRI cases by age. RSV

    disease was more prevalent in children within their first year of life. Peak hospitalization

    rate was observed in the 0-2 month age group. ............................................................ 86

    Figure 5.3-3: Distribution of respiratory viruses detected in HIV infected ALRI patients .... 87

    Figure 5.3-4: A box plot comparing RSV load between ALRI cases and NRD controls......... 88

    Figure 5.3-5 A box plot comparing RSV load by subtype and clinical diagnosis.................. 89

    Figure 5.3-6: A box plot comparing RSV loads in ALRI cases with a viral co-infection [n=12;

    RSV with either hAdV (75%, n=9), hCoV (17%, n=2) or RV (8%, n=1)] versus sole RSV

    infection (n=15). .......................................................................................................... 90

    Figure 6.3-1 A bar graph comparing the frequency (%) of viruses detected in cases and

    controls. Rhinovirus-C (RV-C), Rhinovirus-A (RV-A), parainfluenza virus (PIV), respiratory

    syncytial virus (RSV), human adenovirus (hAdV), Influenza viruses (IFV), human corona

    virus (hCoV) Rhinovirus-B (RV-B) and human metapneumovirus (hMPV) ........................100

    Fig 6.3-2: Box plot summarising RV-C load in children hospitalised with an acute asthma

    exacerbation (cases) and otherwise healthy individuals with a non-respiratory disease

    (controls). Median RV-C load of AAE cases was 2.6 log10 copies/mL higher than that of the

    non-respiratory disease control group . ........................................................................102

    Figure 6.3-3: A boxplot of RV-C loads for cases(n=21) stratified by disease severity and

    controls (n=2). Groups were compared using the Mann-Whitney U test,. median viral load

    between the two severity groups did not differ significantly. .........................................103

    Figure 6.3-4 Surrogate markers of asthma exacerbation in acute samples from patients

    infected with RV-C stratified by illness severity. A) Absolute neutrophil peripheral blood

    counts B) absolute eosinophils peripheral blood counts C) Total serum IgE levels counts.

    Mann- Whitney U or Kruksall-Wallis test were used for testing on the apporpriate group of

    subjects. All data are expressed as box and whisker plots. *:p=0.05 (severe vs non-severe)

    ** p

  • xi

    Figure 7.3-2: RV detection rates stratified by species. RV-C was the predominant species

    detected in children hospitalised with respiratory wheeze ............................................120

    Figure 7.3-3- A comparison of virus detection rates between patients with classified with

    asthma compared to those not classified with asthma. .................................................121

    Figure 7.3-4: Area under the curve analysis of RV-C load to predict hospitalisation. RV-C

    load was poor predictor of hospitalisation with an AUC of 0.5 (95%CI, 0.24-0.74) ...........122

    Figure 7.3-5 Levels of IL-4 (a) and IL-13 (b) in nasal secretions of non-respiratory disease

    controls, RV-C infected patients with asthma and without asthma. IL-4 and IL-13 were both

    significantly elevated in the asthmatic group but IL-13 levels did not differ significantly in

    the non asthmatic group compared to controls.* p

  • xii

    List of Tables Table 1-1: Seasonal and clinical profiles of the commonly detected viruses associated with

    acute respiratory tract infection....................................................................................... i

    Table 4.3-1 A comparison of RNA transcript concentration and Cq values for the different

    RV-C assays.................................................................................................................. 68

    Table 4.3-2 the performance of the individual PCR assays for the detection of matched RV -

    C RNA transcript........................................................................................................... 69

    Table 4.3-3 Intra and Inter assay variability of the four RV-C qRT-PCR assays (Assay 1-4) .. 70

    Table 4.3-4 Variation in calculated copy number yield (%) of transcripts 1-4 compared to

    the number of probe mismatches ................................................................................. 71

    Table 5.3-1 Demographic and clinical details of study participants .................................. 83

    Table 6.3-1 Baseline characteristics of children with acute asthma exacerbation (AAE) and

    controls ....................................................................................................................... 99

    Table 6.3-2: A statistical summary of the risk of being diagnosed with acute asthma

    exacerbation follwoing respiratory virus detection........................................................101

    Table 7.3-1 Summary of clinical and demographic data of hospitalised children with RV -C

    respiratory wheeze .....................................................................................................123

    Table 7.3-2 nasal cytokine levels of healthy non-respiratory disease controls, RV-C

    infected patients with asthma and without asthma. ....................................................130

    Table 7.3-3: An illustration of the relationship between RV-C load and inflammatory

    mediator production in the nasal secretions of children with asthma .............................133

    Table 7.3-4: An illustration of the relationship between RV-C load and inflammatory

    mediator production in the nasal secretions of children without asthma ........................133

    Table 7.3-5: Association between cytokine production and hospitalisation of children

    hospitalised following RV-C infection............................................................................134

    Table 0-1 The performance of the individual PCR assays for the detection of matched RV -C

    RNA transcript ............................................................................................................176

    Table 0-2 A comparison of RNA transcript concentration and Cq values for the different RV-

    C assays ......................................................................................................................177

    Table 0-3 Intra and Inter assay variability of the four RV-C qRT-PCR assays (Assay 1-4) ....178

    Table 0-4 The RV-C load determinations for patients enrolled in the PREVIEW study. Clinical

    samples were tested in triplicate and mean viral load calculated. ..................................179

  • xiii

    Acknowledgments

    First and foremost, I want to thank Prof. David Smith, A/Prof. Allison Imrie, Dr. Glenys

    Chidlow and Dr. Gerald Harnett. It has been an honour to be your Ph.D. student. You all

    have taught me, both consciously and unconsciously, how good research is conducted. I

    appreciate all your contributions of time, ideas, and funding to make my Ph.D. experience

    productive and stimulating. The joy and enthusiasm you all had for this research was

    contagious and motivational for me, even during tough times. I am also thankful for the

    excellent example you have provided as successful leaders in your respective fields. Special

    thanks to Robyn Thompson for her countless efforts to ensure I had access to everything

    (conferences, meetings and training courses) and everyone I needed to accomplish my

    objectives.

    I would like to thank all the members of the PathWest Molecular Diagnostics Laboratory

    for accommodating me into your busy laboratory. You have been a source of friendships as

    well as good advice. Many thanks to our collaborators at the Telethon Kids Institute,

    especially Professor Peter Le Souef, Dr. Ingrid Laing and Dr Kim Khoo. I would also like to

    thank Dr. Abha Chopra at the Institute of Immunology and Infectious diseases for providing

    access to their digital PCR instrument.

    Last but not the least, I want to thank my partner Emma, my family and my closest friend’s

    you guys have been amazing during this time. Mum you have been my biggest supporter

    from the get go. Dad thank you for your wise words, my brothers Kapembwa and

    Kamando, you are the best brothers and friends one could ever ask for. Jarrad, thank you

    for your friendship during this phase it was one where we learnt a lot from each other and

    I will forever cherish the times spent at coffee. My dearest Emma, thank you for being so

    patient and supportive during this time.

  • xiv

    I gratefully acknowledge the funding sources that made my Ph.D. work possible. I was

    funded by University of Western Australia Postgraduate award and University of Western

    Australia Top-up Scholarship. Material, consumables and software were made available by

    PathWest Laboratory Medicine WA. This research was supported by an Australian

    Government Research Training Program (RTP) Scholarship.

  • xv

    AUTHORSHIP DECLARATION: SOLE AUTHOR PUBLICATIONS

    This thesis contains the following sole-authored work that has been published and/or prepared for publication.

    Deta ils of the work: Comparison of Droplet Digital RT-PCR to qPCR for the quantitative detection of Respiratory

    Syncytia l Virus Chisha T. Sikazwe, Angela Fonceca, Avram Levy, Glenys R. Chidlow, Al lison Imrie, Mark Everard,

    David W. Smith. RSV16: 10th International Respiratory syncytial vi rus symposium, Sept 2016, Patagonia,

    Argentina. Location in thesis: Chapter Three

    Deta ils of the work: Reliable quantification of rhinovirus species C using real-time PCR. Sikazwe CT, Chidlow GR,

    Imrie A, Smith DW. J Vi rol Methods. 2016 May 20;235:65-72. doi :10.1016/j.jvi romet.2016.05.014. [Epub ahead

    of print] PMID: 27216896. Location in thesis: Chapter Four

    Deta ils of the work: The Quantitative Detection of Respiratory Syncytial Virus in Hospitalized Young South

    African Children Chisha T. Sikazwe, Al icia A. Annamalay, Glenys R. Chidlow, Salome Abbott, Siew -Kim Khoo,

    Joelene Bizzintino, Robin Green, Allison Imrie, Peter LeSouëf, David W. Smith. Submitted to Influenza and other

    Respiratory vi ruses. Location in thesis: Chapter Five

    Deta ils of the work: Sikazwe CT, Chidlow GR, Imrie A, Smith DW. Determinants of acute asthma exacerbation

    severity following RV-C infection. Location in thesis: Chapter Six

    Deta ils of the work: Sikazwe CT, Chidlow GR, Imrie A, Smith DW. Cytokine profiles in nasal secretions of

    patients hospitalised with Rhinovirus Species C associated respiratory wheeze, Cytokine. (In preparation).

    Location in thesis: Chapter Seven

    XChisha Teza Sikazwe

    Date:

    XAllison Imrie

    Co-ordinating Supervisor

    Date:

    25/06/2018

    25/06/2018

  • xvi

    Peer reviewed papers and Conference presentations

    1 Rachael Lappan; Kara Imbrogno; Chisha Sikazwe; Denise Anderson; Danny Mok;

    Harvey Coates; Shyan Vijayasekaran; Paul Bumbak; Christopher Blyth; Sarra Jamieson;

    Christopher Peacock A microbiome case-control study on recurrent acute otitis media

    identified potentially protective bacterial genera. Microbiome, (In submission)

    2 Bjerregaard, A., Laing, I.A., Backer, V., Fally, M., Khoo, S.-K., Chidlow, G., Sikazwe, C.,

    Smith, D.W., Le Souëf, P. and Porsbjerg, C. (2016) Clinical characteristics of eosinophilic

    asthma exacerbations. Respirology, 22: 295–300. doi: 10.1111/resp.12905.

    3 Bjerregaard, A., Laing, I.A., Backer, V., Fally, M., Khoo, S.-K., Chidlow, G., Sikazwe, C.,

    Smith, D.W., Le Souëf, P. and Porsbjerg, C. (2017)-High fractional exhaled nitric oxide

    and sputum eosinophils are associated with an increased risk of future virus-induced

    exacerbations – a prospective cohort study. Clinical and Experimental Allergy

    4 The Quantitative Detection of Respiratory Syncytial Virus in Hospitalized Young South

    African Children Chisha T. Sikazwe, Alicia A. Annamalay, Glenys R. Chidlow, Salome

    Abbott, Siew-Kim Khoo, Joelene Bizzintino, Robin Green, Allison Imrie, Peter LeSouëf,

    David W. Smith. Submitted to Influenza and other Respiratory viruses (in submission)

    5 Reliable quantification of rhinovirus species C using real -time PCR. Sikazwe CT,

    Chidlow GR, Imrie A, Smith DW. J Virol Methods. 2016 May 20;235:65-72.

    doi:10.1016/j.jviromet.2016.05.014. [Epub ahead of print] PMID: 27216896

    6 Respiratory viruses in young South African children with acute lower respiratory

    infections and interactions with HIV. Annamalay AA, Abbott S, Sikazwe CT, Khoo SK,

    Bizzintino J, Zhang G, Laing I, Chidlow GR, Smith DW, Gern J, Goldblatt J, Lehmann D,

    Green RJ, Le Souëf PN. J Clin Virol. 2016 Aug;81:58-63. doi: 10.1016/j.jcv.2016.06.002.

    Epub 2016 Jun 4. PMID: 27317881

    7 Comparison of Droplet Digital RT-PCR to qPCR for the quantitative detection of

    Respiratory Syncytial Virus Chisha T. Sikazwe, Angela Fonceca, Avram Levy, Glenys R.

    Chidlow, Allison Imrie, Mark Everard, David W. Smith. RSV16: 10th International

    Respiratory syncytial virus symposium, Sept 2016, Patagonia, Argentina.

    8 Reliable Quantification of Rhinovirus Species C using a Taqman Real -time PCR Based

    Approach Chisha T Sikazwe · Glenys R. Chidlow · Allison Imrie · David W. Smith. 1st

    International Meeting for Respiratory Pathogens, Sep 2015 Singapore.

    9 The Relationship between RSV Load and Clinical Disease in South African Children

    Chisha T Sikazwe, Glenys R. Chidlow, Allison Imrie, David W Smith. 9th International

    Symposium on Respiratory syncytial virus Nov 2014, Cape Town, South Africa

  • xvii

    List of Abbreviations

    Abbreviation Definition

    µ Micro

    µL Microlitre

    µM Micromolar

    AAE Acute Asthma Exacerbation

    ALF Australian Lung Foundation

    ARTI Acute Respiratory Tract Infection

    BAL Bronchoalveolar Lavage

    BHQ Black Hole Quencher

    BLAST Basic Local Alignment Search Tool

    bp Base Pair

    cDNA Complementary DNA

    CFT Complement Fixation Test

    CMV Cytomegalovirus

    COPD Chronic Obstructive Pulmonary Disease

    CPE Cytopathic Effect

    Cq Cycle Quantification Value

    Ct Cycle Threshold

  • xviii

    CXCL- Chemokine Ligand

    ddPCR Digital Droplet PCR

    DNA Deoxyribonucleic Acid

    EIA Enzyme Immunoassay

    GINA Global Initiative For Asthma

    HAdV Human Adenovirus

    HBoV Human Bocavirus

    HCoV Human Coronavirus

    HMPV Human Metapneumovirus

    HPIV Human Parainfluenza virus

    GAPDH Glyceraldehyde 3-phosphate dehydrogenase

    IF Immunofluorescence

    IFAV Influenza A Virus

    IFBV Influenza B Virus

    IFN- Interferon

    IL- Interleukin

    LNA Locked Nucleic Acid

    LRTI Lower Respiratory Tract Infection

    MERS-CoV Middle Eastern Respiratory Syndrome Coronavirus

  • xix

    MGB Minor Groove Binding

    mL Milliliter

    NA Nasal Aspirate

    NAT Nucleic Acid Test

    NPA Nasopharyngeal Aspirate

    NPS Nasopharyngeal swab

    NT Neutralisation Test

    NW Nasal Wash

    PCR Polymerase Chain Reaction

    PMH Princess Margaret Hospital

    PWLM PathWest Laboratory Medicine WA

    qPCR Quantitative Real Time PCR

    RNA Ribonucleic Acid

    RSV Respiratory Syncytial Virus

    RT-ddPCR Reverse Transcription Digital Droplet PCR

    RT-PCR Reverse Transcription PCR

    RT-qPCR Reverse Transcription Quantitative Real Time PCR

    RV Rhinovirus

    SARS-CoV Severe Acute Respiratory Syndrome Coronavirus

  • xx

    Th T Helper Cells

    TKI Telethon Kids Institute

    TLR Toll Like Receptors

    TNF- Tumor Necrosis Factor

    URTI Upper Respiratory Tract Infection

    UTR Untranslated Region

    UWA The University Of Western Australia

    VL Viral Load

    WHO World Health Organisation

    α Alpha

    β Beta

  • 2

    1 Literature review

  • 1.1 Introduction

    Acute respiratory tract infections (ARTIs) contribute substantially to the global burden of

    illness from communicable pathogens. ARTIs are a leading cause of morbidity and mortality

    accounting for approximately four million deaths per year globally (Bryce et al., 2005).

    Children under the age of five years, adults over the age of sixty-five, individuals with an

    underlying chronic condition and immunocompromised individuals are population groups

    in whom poor outcomes occur following infection (Falsey et al., 2003; Graham and Gibb,

    2002; Nair et al., 2011a; Nair et al., 2010; Nair et al., 2013). Estimates of the global burden

    of acute respiratory illness indicate that there are vast differences between developed and

    developing countries. According to recent reports, ARTIs are the leading cause of childhood

    death in developing countries (Nair et al., 2011b). Pneumonia alone accounts for 1.4

    million childhood deaths per year in these regions. Conversely, in developed nations

    deaths due to ARTIs represent a negligible percentage of total deaths, accounting for less

    than two percent of deaths but are predominantly responsible for absenteeism and

    enormous financial costs to the healthcare system (Denny Jr., 2001). Reports from the US

    estimate that direct financial costs of ARTIs to the healthcare system approach $40 billion

    annually. Similarly, in Europe the expenditure on patients with ARTI is over €15 billion. The

    direct and indirect cost of ARTIs to the Australian health care system is estimated to be up

    to AUD 600 million each year (The Australian Lung Foundation, 2007).

    All groups of microbes are capable of establishing an infection in the respiratory tract.

    However, viruses predominate as aetiologic agents of ARTIs and can contribute to

    respiratory disease either directly through frank infection or indirectly by exacerbating

    pre-existing illness (Bafadhel et al., 2011; Bandi et al., 2003; Busse, Lemanske, and Gern,

    2010; Hosseini et al., 2015) and increasing the risk of secondary bacterial infection

    (Bellinghausen et al., 2016; Ewijk et al., 2007). Influenza virus (IFV), respiratory syncytial

  • 4

    virus (RSV), rhinovirus (RV), human metapneumovirus (hMPV), human parainfluenza virus

    (hPIV) and human adenovirus (hAdV) are the most prevalent viruses in hospitalised

    patients and are all linked to ALRI (Lukšić et al., 2013). The distribution of these viruses is

    influenced by season, geographic region and age group (Lukšić et al., 2013). They are at

    least 6 families and more than 150 viruses that are associated with ARTI. The clinical and

    seasonal profiles of the most common types are listed in Table 1.

  • Table 1-1: Seasonal and clinical profiles of the commonly detected viruses associated with acute respiratory tract infection

    Virus

    Incubation period (days)

    Seasonality Clinical manifestations Laboratory diagnostic method

    Respiratory syncytial virus (RSV) 2-8 Winter bronchiolitis, pneumonia Virus isolation in cell culture, RT-PCR, IF, EIA,

    Human metapneumovirus (HMPV) 2-8 Winter to spring bronchiolitis, pneumonia RT-PCR, IF

    Rhinovirus (RV) 2 All year round coryza, COPD and asthma exacerbation, bronchiolitis, pneumonia

    RT-PCR, NT and Virus isolation in cell culture (for some types)

    Coronavirus (HCoV) 2 All year round coryza, pneumonia (rarely in non-SARS or MERS infection)

    RT-PCR,ELISA,HA,IF

    Influenza virus (IFV) 2-8 Winter Virus isolation in cell culture, RT-PCR, CFT, HI,IF, EIA,NT

    Human parainfluenza viruses (HPIV) 2-8 Autumn to winter coryza, croup, bronchitis, bronchiolitis, and pneumonia

    Virus isolation in cell culture, RT-PCR, CFT, HI,IF, EIA,NT

    Adenovirus (AdV) 5-7 All year round pneumonia, bronchitis, pharyngitis, tonsillitis, and pharyngoconjunctival

    fever

    RT-PCR, HI, IF,EIA and NT

    Human bocavirus (HBoV) 5-7 All year round Coryza, bronchiolitis, pneumonia

    RT-PCR

    Abbreviations: RT-PCR; reverse transcription PCR, IF; immunofluorescence, EIA; enzyme immunoassay, HA; haemagglutination, NT; neutralisation test, HI; haemagglutination inhibition, CFT; complement fixation

    test.

  • 1.1.1 Respiratory syncytial virus

    RSV is a member of the paramyxoviridae family and has a single strand negative sense

    enveloped RNA genome. RSV genome encodes two non-structural proteins and nine

    structural proteins. The attachment (G) and fusion (F) glycoproteins are the

    immunodominant antigens of RSV and are essential in infectivity and antigenicity (Fodha et

    al., 2007). RSV is considered to be serologically monotypic, but consists of two genetic

    subgroups, RSV-A and RSV-B (Papadopoulos et al., 2004). RSV A is more prevalent than RSV

    B but there is no consensus on which of the subtypes is more pathogenic.

    RSV accounts for 40-50% of all viral infections requiring hospitalisation in young infants

    (Nair et al., 2011b). Virtually all children are exposed to RSV before the age of 3 years

    (Henderson et al., 2005). Acute bronchiolitis is the typical syndrome associated with RSV

    infection in young infants and is characterised by a predominantly neutrophilic pattern of

    inflammation and mucus in the airways, which often results in some degree of pulmonary

    obstruction. Young infants are predisposed to severe acute bronchiolitis because of the

    small diameter of their airways. Congenital heart disease, prematurity, bronchopulmonary

    dysplasia or cystic fibrosis are all risk factors for severe infection and possibly death

    (PREVENT, 1997).

    1.1.2 Rhinoviruses

    Rhinoviruses (RVs) are non-enveloped positive sense single stranded RNA viruses

    belonging to the family Picornaviridae. They are genetically and antigenically diverse

    consisting of three species (A, B and C). Historically, extensive cross neutralisation test

    were utilised for the classification of RV-A and B isolates into 100 serotypes but these

    isolates (types) are now assigned solely on genomic sequence (McIntyre, Knowles, and

  • 7

    Simmonds, 2013). In 2006, utilisation of molecular techniques led to the discovery of RV-C,

    which had been entirely unrecognised using traditional rhinovirus detection techniques

    (Arden et al., 2006). Cell lines that were conventionally used to isolate rhinovirus are not

    permissive to RV-C infection and as a result preclude serotype assignment. Sequence

    typing has revealed that RV-C is genetically more diverse than the other two species and

    currently consists of 65 distinct genotypes (McIntyre et al., 2013).

    Historically, RV-A and RV-B serotypes were stratified into two groups (major and minor) on

    the basis of cellular receptor utilisation (intracellular adhesion molecule [major] or low-

    density lipoprotein receptor [minor]) whereas the RV-C receptor remained unidentified

    until recently. A recent report has shown that the human cadherin-related family member

    3 (CDHR3), a member of the cadherin family of transmembrane proteins, facilitates RV -C

    attachment and replication (Bochkov et al., 2015). This receptor is highly expressed in the

    lower respiratory tract but its biological function remains unknown. A single nucleotide

    polymorphism in CDHR3 gene is associated with RV-C wheezing illness in infancy and has

    been shown to be risk factor for asthma inception (Bonnelykke et al., 2014).

    The spectrum of disease exhibited by RVs range from asymptomatic infections, mild upper

    respiratory infections (common cold) to severe or fatal lower respiratory tract infections

    (pneumonia and bronchiolitis). RV infection is also associated with the exacerbation of

    chronic respiratory conditions such as asthma, cystic fibrosis and other chronic obstructive

    pulmonary disorders (Blomqvist et al., 2002; Çalışkan et al., 2013; Camargo et al., 2012;

    Choi et al., 2015). In addition, RV induced wheeze in early childhood is associated with an

    increased risk of developing asthma later on in life (Jackson et al., 2008a). Recent studies

    have also revealed that RV-C species are more closely associated with asthma exacerbation

    and illness requiring hospitalization than the other RV species, suggesting that RV-C may be

    more pathogenic than RV-A and RV-B (Bizzintino et al., 2011a; Cox et al., 2013). Host and

  • 8

    viral factors associated with RV-C pathogenesis and illness severity are yet to be

    completely elucidated.

    1.1.3 Human Metapneumovirus

    HMPV is classified as a negative sense single stranded RNA virus belonging to the

    Paramyxoviridae family. Sequence analysis of the fusion (F) and attachment (G) genes has

    identified two genotypes namely A and B. Both genotypes may co-circulate but only one

    genotype is dominant during an epidemic (Tsukagoshi et al., 2013a; van den Hoogen et al.,

    2002).

    HMPV is associated with hospitalisation in young children, the elderly and individuals w ith

    an underlying chronic condition. It is distributed worldwide and mimics the seasonal

    distribution of influenza and RSV in that infection rates tend to peak in winter and early

    spring (Kahn, 2003). Much like other respiratory viruses the spectrum of clinical disease

    ranges from a mild upper respiratory tract infection to severe pneumonia. Observational

    studies report that elderly people infected with hMPV display more severe clinical

    symptoms compared to younger patients and those elderly infected with RSV or Influenza

    virus (Falsey et al., 2003; Widmer et al., 2012).

    1.1.4 Influenza virus

    The influenza viruses are a major causative agent of ARTI in both children and adults,

    Infection can cause mild to severe illness, and can occasionally lead to death (Pretorius et

    al., 2016). Influenza accounts for a substantial burden of ARTI and current estimates

    suggest that it is responsible for approximately five million severe cases and between

    291,000-646,000 deaths per annum (Iuliano et al., 2018). Three Influenza types (type A,

    type B and type C) are known to infect humans. Only the influenza A/H1N1, A/H3N2 and

    influenza B viruses are considered as being significant contributors to seasonal influenza.

  • 9

    Influenza C virus infection is seasonal but has limited genetic diversity and is not thought to

    cause epidemics (Nicholson, Wood, and Zambon, 2003).

    Genomes of influenza A and B viruses are composed of eight single-stranded negative

    sense RNA segments whereas the influenza C virus is composed of seven segments of

    single stranded negative sense (Nicholson et al., 2003).

    The genomes of influenza viruses specifically the Influenza A virus (IFAV) regularly undergo

    changes under immune pressure. The genes encoding surface glycoproteins

    haemagglutinin and neuraminidase may undergo subtle genetic changes (antigenic drift) or

    abrupt major changes (antigenic shift).

    Influenza therapeutics comes in the form of vaccines and anti -viral compounds. Vaccines

    are recommended for annual boosting especially in high risk groups (Fiore, Bridges, and

    Cox, 2009; Matsushita et al., 2017). Most Influenza antiviral compounds inhibit viral

    replication and release, and can be utilised for treatment and prophylaxis (Dobson et al.,

    2015; Jefferson et al., 2014b).However, adverse events reported from use of these

    compounds suggested that they should be administered judiciously(Jefferson et al.,

    2014a).

    1.1.5 Parainfluenza virus

    Human parainfluenza viruses (HPIV), of which they are four types (HPIV1-4) are negative

    sense RNA viruses of the Paramyxoviridae family. HPIVs typically cause mild, self-limited

    upper respiratory tract infections in adults, but can result in severe, life -threatening LRTIs

    in immunocompromised patients (Guzmán-Suarez et al., 2012). HPIVs 1-3 are a common

    cause of croup syndrome and are among the most commonly detected viruses in

    hospitalised young infants after RSV. HPIV-4 is known to cause mild upper respiratory tract

  • 10

    infections (Tsukagoshi et al., 2013b). However, due to a lack of epidemiological data on

    HPIV-4 the extent of its pathogenicity is largely unknown.

    1.1.6 Adenovirus

    Human Adenoviruses (HAdV) are a major cause of viral disease in both children and adults

    (Lynch, Fishbein, and Echavarria, 2011). HAdV have linear double-stranded DNA genomes

    and are classified into seven species (HAdV A to G) and a total of 55 types based on a

    combination of serological characteristics and phylogenetic analysis (Robinson et al., 2011).

    Certain species are known to cause respiratory illnesses including, acute febrile pharyngitis

    (HAdV-B and -C; types 1,3,5,7), pharyngoconjunctival fever (HAdV-B; types 3,7,14), acute

    respiratory tract infections (HAdV-B and -E; types 3,4,7,14,21), pneumonia (HAdV-B, -C,

    and -E; serotypes 1-4 and 7), pertussis-like syndrome (HAdV-C; type 5) (Demian et al.,

    2014). PCR is the method of choice in detecting adenoviruses.

    1.1.7 Human Coronaviruses

    Human coronaviruses (HCoVs) are enveloped viruses with a single-strand positive sense

    RNA genome. Coronaviruses possess the largest known genomes among any of the RNA

    viruses (Lau et al., 2013). Six species of HCoVs are known to infect humans; they are

    classified into four genera based on proteomic analysis (Lau et al., 2013). HCoV-229E and

    HCoV-NL63 belong to the Alphacoronavirus genus, HCoV-OC43 and HCoV-HKU1 belong to

    lineage A Betacoronavirus, while severe acute respiratory syndrome associated

    coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV)

    belong to lineages B and C Betacoronavirus, respectively (Annan et al., 2013). HCoV-OC43,

    HCoV-229E, hCoV-HKU1 and hCoV-NL63 are commonly associated with self-limiting mild

    upper respiratory tract infections but on some occasions may be causative agents of

    severe LRTIs especially in young infants, the elderly, and individuals with compromised

  • 11

    immunity (Lu et al., 2012). HCoVs are detected frequently with other respiratory viruses;

    the significance of this phenomenon in relation to illness severity is unclear.

    In the last decade SARS-CoV and MERS-CoV have been responsible for epidemics around

    the world (Cheng et al., 2007). SARS-CoV was identified as the causative agent of the SARS

    global epidemic from 2002 to 2003. MERS-CoV was initially reported in Saudi Arabia in

    2012 (Zaki et al., 2012). Since then it has caused illness in people in countries with links to

    the Arabian Peninsula (de Groot et al., 2013). Both MERS-CoV and SARS-CoV are emerging

    zoonotic pathogens that crossed the species barrier with case fatality rates of 35% and

    10% respectively (To et al., 2013). Bats appear to be the natural reservoir of both MERS-

    CoV and SARS-CoV, transmission to humans occurs via dromedary camels and civets (live

    market animals) respectively (Dijkman et al., 2013; Hu et al., 2015; Pfefferle et al., 2009;

    Yusof et al., 2015). Nucleic acid tests are the method of choice for the laboratory diagnosis

    of HCoVs (Gaunt et al., 2010) and MERS-CoV(Feikin et al., 2015).

    1.2 Laboratory Diagnosis of Respiratory Infections

    Laboratory diagnostic methods are the cornerstone in accurately attributing pathogen to

    clinical presentation. Early detection of the aetiologic agent is pertinent for providing

    optimal clinical management, which may include patient isolation, determining

    appropriate therapy and/or cessation of inappropriate therapeutic interventions. In

    addition, diagnostics are also essential for outbreak detection and response, and public

    health surveillance. Characteristics of the ideal diagnostic test include being accurate

    (between tests and laboratories), high throughput, cost-effective, suitable for a wide

    spectrum of clinical samples, excellent sensitivity and specificity. Current diagnostic tests

    fulfil some, but not all of these ideal characteristics.

  • 12

    They are several methods that are available for the identification of the aetiologic agent

    from patients with an ARTI. The traditional laboratory diagnostic methods such as virus

    isolation in cell culture and serology tests perform well but have some inherent limitations

    in aspects of sensitivity and/or specificity. Molecular based diagnostic techniques such as

    PCR have led to clinicians and scientists re-evaluating the role of certain viruses in disease

    outcome. PCR based tests are a relatively new tool in the laboratory diagnostics. These

    tests allow for rapid screening of clinical samples for multiple aetiologies with the added

    benefit of excellent sensitivity and specificity. Further, these diagnostic approaches

    facilitate further understanding of the viral kinetics in respiratory infections as well as the

    ability to identify respiratory viruses that have been entirely missed by traditional

    diagnostic approaches.

    1.2.1 Upper and Lower airway samples

    A variety of specimens have been used for directly sampling the respiratory tract including

    nasal swabs (NS), nasopharyngeal swabs (NPS), nasal washings (NW), nasal aspirates (NA),

    nasopharyngeal aspirates (NPA), sputum, bronchoalveolar lavage (BAL) or biopsies. The

    technique utilised to collect a sample is based on clinical presentation and the method to

    be utilised to identify the pathogen.

    NWs are conventionally used on children and are not well tolerated in adults. NW can be

    unpleasant to the patient, in addition, NW can be technically demanding, as the technique

    requires the use of a solution such as saline. A NS is commonly taken from the mid-inferior

    portion of the inferior turbinates for optimal virus recovery. Though this collection

    technique maximises virus recovery, it causes some discomfort to the patient.

    Nonetheless, both sampling techniques are suitable for detection of virus by cell culture.

    NPA is especially useful for virus culture, antigen detection, and polymerase chain reaction

    (PCR) based assays and is collected using a mucus trap, attached to a disposable suction

  • 13

    catheter. This technique requires a skilled operator, so it may be unsuitable for widespread

    clinical practice. Studies that have compared NS and NPA sampling techniques for

    detection by PCR report that the overall sensitivity of nasal swabs was inferior to that of

    NPAs, but the authors noted that obtaining a NPA was invasive, uncomfortable and

    significantly more distressful than a nasal swab (Heikkinen et al., 2002; Lambert et al.,

    2008). With the recent development of flocked nasal swabs and advanced molecular

    techniques, the sensitivities have increased to a level comparable to NPAs with the added

    advantage of the flocked nasal swab being less painful and more convenient than NPAs, as

    no supplementary tools are required (Ortiz de la Tabla et al., 2010). Sputum samples are

    more representative of lower respiratory tract and are collected non-invasively. Despite

    the aforementioned advantages of sputum collection, practical reasons preclude the

    effective collection of sputum from infants and children (Abdullahi et al., 2008; Grant et al.,

    2012). Children have difficulty producing sufficient sputum for laboratory evaluation

    compared to adults and are more inclined to swallow the specimen than expectorate it

    (Grant et al., 2012). Further the viscosity of sputum and the likely presence of

    contaminating bacteria as well as the toxic effects sputum can have on cell culture

    preclude its widespread use for viral diagnostic purposes (Falsey, Formica, and Walsh,

    2012). BAL and lung biopsy specimen are collected directly from lower respiratory tract

    sites. Previous studies on MERS-CoV have shown that quantification cycle (Cq) values were

    lower in lower respiratory tract samples compared to upper respiratory tract samples and

    more accurately reflect the apparent viral kinetics in the lower respiratory tract. However,

    these specimen types are usually collected from the severely ill (Falsey et al., 2012) and too

    few of these specimens would be available to conduct proper population level studies.

    1.2.2 Virus isolation in cell culture

  • 14

    Historically, virus isolation by cell culture was the diagnostic method used to screen

    respiratory samples for the presence of viruses. Virus isolation in cell culture is regarded as

    the "gold standard" to which all other detection methods have been compared since the

    isolation of virus in cell culture indicates the presence of an infectious, viable, and

    replication competent virus, an occurrence which is unachievable using other detection

    techniques (Leland and Ginocchio, 2007). This method is labour intensive, and often

    requires prolonged incubation periods (3-14 days) to provide results, limiting its use in the

    acute management of patients. A broad range of respiratory viruses can be grown by using

    at least 4 cell lines, which includes human epithelial cell lines (HEp-2, A-549, HeLa),

    human fibroblast cell lines (HLF, HELF, MCR-5, WI-38) and primary monkey kidney cells

    (Denny Jr., 2001). For most respiratory virus applications, the presence of virus is

    commonly detected by a characteristic cytopathic effect (CPE) under light microscopy. The

    combination of conventional cell culture and commercial viral -antigen detection systems

    can be used to accelerate diagnostic turn-around times, since viral antigens are detected in

    the cell monolayer by immunofluorescence, usually before a distinct CPE is observable

    (Terletskaia-Ladwig et al., 2008). In recent times, more rapid and powerful tools to detect

    the presence of virus in clinical samples collected from patients following ARTI have

    gradually superseded virus culture techniques.

    1.2.3 Direct detection of respiratory viruses by immunofluorescence

    Laboratory diagnosis can be performed by direct detection of virus in clinical specimen.

    Direct antigen tests assist in the diagnosis of respiratory infection by providing evidence of

    the antigen in respiratory specimen. These tests are instrumental in informing pertinent

    therapeutic decisions in a short time frame (Gomez et al., 2016). Direct and indirect

    immunofluorescence are the two sub-forms of the immunofluorescence test. The direct

    test is composed of a virus specific monoclonal antibody that is conjugated directly to

  • 15

    fluorescent dye. If the virus is present in the clinical sample the monoclonal antibody

    attaches to the targeted viral antigen and the conjugated fluorescent dye can be visualised

    under a fluorescent microscope. In the indirect method, two antibodies are used, a specific

    primary monoclonal antibody that attaches to the viral antigen and a secondary antibody

    labelled with a fluorescent dye to bind to the primary monoclonal antibody. The indirect

    method is more sensitive than the direct method because of the signal amplification from

    multiple secondary antibodies binding to a single primary antibody.

    Antigen detection methods such as the enzyme immunoassay (EIA) are performed by

    adding patient specimen onto a surface that is pre-coated with an antibody that captures

    the virus specific antigen if present in the specimen. EIA is a highly sensitive assay and

    utilises an enzyme system to produce a colour reaction that can be quantified.

    1.2.4 Nucleic Acid Tests

    Diagnostic laboratories detect respiratory viruses in clinical samples by using molecular

    techniques to detect virus genome. PCR based methods are the most favourable and the

    most widely used approach for rapid and accurate respiratory virus detection from clinical

    specimens. The advent of nucleic acid based tests has led to the identification of viruses

    that were entirely missed using conventional diagnostic techniques. Most respiratory

    viruses have their genetic information primarily stored in the form of RNA, with the

    notable exception of HAdV and HBoV. Amplification of an RNA target requires a reverse

    transcription (RT) step in order to convert RNA to cDNA followed by conventional PCR (RT-

    PCR). DNA targets do not require an RT step. The repetition of three successive reactions

    is the basis of PCR:

    1. Denaturation of double stranded DNA (dsDNA) into si ngle stranded DNA (ssDNA)

    between 94 and 95⁰C

  • 16

    2. Annealing of 2 synthetic oligonucleotides (primers) to each ssDNA at each termini at a

    variable temperature ranging from 37-72⁰C

    3. Synthesis of new DNA strands; the PCR method util izes a thermostable DNA polymerase

    to add nucleotides at the 3' of end of the primers at 68 - 72⁰C.

    These three steps entail a single PCR cycle, since each synthesized DNA strand becomes a

    template for amplification there is an exponential increase in amplicon at the end of each

    cycle. The length of the PCR product is equivalent to that of the two primers plus the

    distance separating the two primers. Several variations of PCR are applicable to the

    diagnosis of respiratory viral infection including nested PCR, real -time PCR (qPCR) and

    multiplex qPCR.

    1.2.4.1 Real time PCR (qPCR)

    Utilisation of modern molecular techniques has enabled detection of viral nucleic acid

    during the exponential phase of the reaction rather than waiting for the endpoint of the

    reaction to register detection. The fundamental principle of qPCR is based on the detection

    and quantification of fluorescent reporter molecules whose signal can be registered in

    each cycle of the PCR. The cycle number when the fluorescence becomes detectable is

    referred to as the cycle threshold/quantification value (Ct/Cq), and is proportional to the

    logarithm of the initial amount in the clinical sample. Unlike conventional PCR, qPCR does

    not require any post PCR amplification manipulations thereby minimizing any issues

    related to contamination. However, similar to conventional PCR is the addition of a reverse

    transcription step when screening clinical samples for respiratory viruses with an RNA

    genome.

    A DNA binding fluorescent dye, such as SYBR green represents the simplest method to

    detect amplified product using qPCR, given that this dye binds to any double stranded DNA

  • 17

    in the reaction. A shortfall of SYBR green chemistry is its lack of sequence specific DNA

    binding activity. So this type of chemistry will also bind to non-specific products that the

    qPCR reaction may generate and may lead to false positive results. Sequence specific

    hydrolysis probes are alternative detection chemistry and mitigate the problem of false

    positive results because fluorescence signal is only detected when the probe binds to the

    target area. Sequence specific hydrolysis probes are the most widely used and published

    detection chemistry available for qPCR. These probes are beneficial because they are

    designed to increase both the sensitivity and specificity of the assay. qPCR provides

    excellent sensitivity and a wide dynamic range and for this reason can be utilised to

    measure viral load in clinical samples.

    Viral load determination may increase current understanding of host viral interactions

    (Quinn, 2011) and may help predict disease progression (Jartti et al., 2013). Viral load

    determination from clinical samples is performed by interpolating the cycle quantification

    value (Cq) into a standard curve. The standard curve is constructed by standards of target

    nucleic acid that encompass a wide range (5-9 logs) of known concentrations. It is

    important to note that there is inherent variability in construction of a standard curve

    between batches. As such it is imperative that the reference standards used are scrutinised

    thoroughly for precision and reproducibility during the validation process , in order to

    understand the limitations of the assays and to provide valid standards for RNA

    quantification.

    Specimen collection is an important source of variability that may obscure real changes

    and consequently unreliable quantification results. The reliability of any PCR-based

    quantification experiment can be improved by including an invariant endogenous control

    (mRNA that is stably expressed and is not affected by experimental conditions) in the

    experiment to correct for sample to sample variation that may arise.

  • 18

    1.2.4.2 Multiplex PCR

    Multiplex PCR refers to the use of multiple sequence specific primers and probes to detect

    multiple targets in a single reaction tube. This type of assay is efficient and economical but

    requires substantial assay optimization to ensure optimal amplification of the different

    targets if present. The availability of multiplex PCR makes the detection of co-infections

    feasible (Franz et al., 2010). Multiplex PCR is an important tool in the diagnostic laboratory

    when screening for potential etiologic contributions to disease.

    1.2.4.3 Droplet digital PCR

    Droplet digital PCR (ddPCR) is a relatively new approach that improves on qPCR by making

    external standards unnecessary in viral quantification. In a similar approach to qPCR,

    ddPCR involves the detection of template sequence with either a SYBR green or hydrolysis

    probe reaction chemistry but quantification is conducted differently. It involves the

    generation of a large library of emulsion based droplets (~20,000), also termed partitions

    (Markey, Mohr, and Day, 2010). These partitions are generated from sample-reagent

    mixtures and are distributed in such a way that there may be either one or zero template

    molecules in the each partition (Mojtahedi, Fouquier d'Herouel, and Huang, 2014). This is

    the fundamental principle underpinning digital PCR. Further, and in contrast to qPCR the

    thermal cycling is performed to endpoint (Markey et al., 2010). The total initial number of

    template is obtained by tallying partitions in which the template is detected compared to

    the number of partitions in which the reaction is unreactive (Markey et al., 2010). A

    Poisson correction is applied to the final copy number to compensate for the possibility

    that more than one template molecule may be present in some partitions. ddPCR is highly

    sensitive, reproducible, provides enhanced accuracy and precision thus lending itself to a

    number of favourable applications such as low copy target detection, quantitative

    detection of minor genotypes. Moreover, ddPCR does not rely on factors such as

  • 19

    amplification efficiency and arbitrarily assigned threshold values and therefore reduces the

    amount of bias in the sample resulting in confident quantification (Hall Sedlak and Jerome,

    2014; Huggett et al., 2013).

    Recent work from a study comparing ddPCR to qPCR for quantitative detection of

    cytomegalovirus (CMV) demonstrated that ddPCR is equivalent for the accurate

    quantification of CMV load in clinical samples over a wide dynamic range (Hayden et al.,

    2013). Recent work that evaluated ddPCR for influenza vaccine development has

    demonstrated a high throughput ddPCR method for very precise and accurate influenza

    virus titre quantification (Palatnik de Sousa et al., 2015). The authors also noted several key

    issues that are determinants of variability in qPCR were circumvented with the ddPCR

    approach.

    1.3 Viral kinetics in acute respiratory tract infections

    Viral kinetic studies have improved our understanding of the interplay between host and

    virus in human immunodeficiency virus (HIV), CMV, hepatitis B and C infections.

    Experimental human infection studies on RSV and IFAV have shown the unique ways in

    which viral kinetics modulate illness severity and disease course (Bagga et al., 2013;

    DeVincenzo et al., 2010; El Saleeby et al., 2011). In addition, viral load studies have shed

    light on critical treatment time points used in order to minimise the risk of severe illness.

    The recent discovery of novel respiratory pathogens has stimulated investigations into viral

    load as a surrogate marker of disease severity in hospitalised patients. For ex ample,

    evidence from a recent study conducted in France reports that a high hMPV viral load is an

    important predictor of disease severity in young children (Roussy et al., 2014). Indeed,

    previous RSV studies indicate that high viral load is associated with symptom severity

    (DeVincenzo et al., 2010; El Saleeby et al., 2011; Houben et al., 2010) . Furthermore, a

  • 20

    reduction in RSV and influenza viral loads in patients as a consequence of antiviral therapy

    has been found to be associated with improved clinical outcomes (Boivin, Coulombe, and

    Wat, 2003; Shadman and Wald, 2011). This paradigm of direct virus damage to host tissue

    shaping clinical outcome has been supported further by investigations on SARS-CoV and

    MERS-CoV (Feikin et al., 2015; Hung et al., 2004; Min et al., 2016). In RV infection, evidence

    in the published literature suggests that viral load is predictive of the severity of clinical

    illness, given that individuals with a lower respiratory tract infection harbour higher viral

    loads that those with an upper respiratory tract infection (Utokaparch et al., 2011). This

    finding is further supported by an Italian study that reported that an abundant RV viral

    load (>106copies/mL) in the absence of other viral pathogens is strongly associated with

    LRTI (Piralla et al., 2012). However, other studies have not reproduced these findings (Jartti

    et al., 2008) and thus the relevance of RV viral load in lower respiratory tract infections is

    not yet completely understood.

    1.4 Viral respiratory infection and asthma

    Asthma is one of the most common chronic respiratory inflammatory conditions and a

    substantial contributor to morbidity worldwide (GINA, 2017). It is characterised by airway

    inflammation, remodelling, reversible airway blockage, and increased airway smooth

    muscle tone (Zhao et al., 2002). It can be difficult to diagnose asthma with certainty in

    children aged between 0-5 years because there are no standardised diagnostic criteria for

    asthma (GINA, 2017). In addition, respiratory infections predominant in childhood such as

    bronchiolitis share many clinical features with exacerbations of asthma, including

    wheezing, shortness of breath and respiratory distress (Sigurs et al., 2000). Diagnosis of

    asthma in children may involve the individual’s history of recurrent wheeze, family history

    of atopy and objective investigations that support the diagnosis. Several stimuli can trigger

  • 21

    exacerbations of asthma symptoms in young children including smoking, viral infections,

    air pollution and environmental allergens (Pelaia, Vatrella, and Maselli, 2012; Riccio et al.,

    2012; Saraya et al., 2014; Sykes et al., 2014). The advent of molecular methods has shown

    that respiratory viruses are present more commonly in acute asthma than previously

    acknowledged, and are detected in up to 85% of asthma exacerbations (Saraya et al.,

    2014). RV and RSV are the predominant viruses that appear to be involved with these

    exacerbations (Saraya et al., 2014; Sigurs et al., 2000; Soto-Quiros et al., 2012). It is still

    not yet clear if infection causes long term changes in the airways which subsequently

    increase the risk of developing asthma. An alternative hypothesis is that severe RSV and/or

    RV disease (bronchiolitis) may be an early marker of a predisposition for childhood asthma

    (Miller et al., 2011; Moore, Stokes, and Hartert, 2013; Moore et al., 2007). The children at

    high risk of developing asthma are those who wheeze and develop allergic sensitization

    before the age of 2 years (Jackson et al., 2008a; Sigurs et al., 2010; Sigurs et al., 2000). Long

    term prospective studies have linked severe RSV disease in young infants to subsequent

    recurrent wheeze and asthma in susceptible children (Sigurs et al., 2010). However other

    studies do not confirm the association between medically attended RSV disease and

    subsequent asthma development (Poorisrisak et al., 2010).

    RV infection in young infants is an independent risk factor for subsequent wheeze and

    childhood asthma (Jackson et al., 2008a). Several reports have shown that RVs are the

    most common virus detected in hospitalised young children with exacerbations of asthma,

    suggesting an etiologic role (Liu et al., 2016; Luchsinger et al., 2014; Message and Johnston,

    2002; Miller et al., 2011). Children classified as asthmatic are more likely to have poor

    outcomes following RV infection compared to normal individuals (Corne et al., 2002). RV-C

    appears to be the most commonly detected species in hospitalised asthmatic children with

    some studies suggesting that it is also the most pathogenic (Bizzintino et al., 2011a; Cox et

    al., 2013; Liu et al., 2016). However, determinants of RV-C associated asthma severity are

  • 22

    yet to be clearly defined, and the contribution of RV-C in asthma exacerbation severity is

    still not clear.

  • 23

    1.5 Innate immune response to viral respiratory infection

    In response to infection, germ line encoded receptors (pattern recognition receptors,

    PRRs) that are resident on sentinel cells of the immune system sense conserved pathogen-

    associated molecular patterns (PAMPs) on microbes (Gommerman and Ng, 2013; Kim and

    Lee, 2014; Malmgaard, 2004; Scagnolari et al., 2009). There are three known PRR families

    and they include toll-like receptors (TLRs), retinoic acid inducible gene 1 (RIG-1)-like RNA

    helicases (RLHs) and nucleotide-binding oligomerization domain (NOD) like receptors

    (NLRs). Signalling downstream of each PRR family results in the activation of important

    pathways that regulate the expression of chemokines, pro-inflammatory cytokines, type 1

    interferon (type 1 IFN) and antimicrobial peptides (Thompson and Locarnini, 2007). RLHs,

    TLR 3, 7, 8 and 9 identify viral nucleic acid and induce innate type 1 IFN responses with or

    without the production of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IL-12, IL-18).

    Type 1 IFN is known to block viral replication at an early step in replication and to be very

    important for the induction of an anti-viral state against common respiratory viruses

    (Scagnolari et al., 2009). Type I IFN deficient mice are more prone to delayed viral

    clearance and severe disease following viral challenge. Further, impaired IFN response to

    viral infection has been postulated as a pathogenic mechanism for poor outcomes in

    asthmatic patients (Baraldo et al., 2012) and MERS-CoV infection (Faure et al., 2014). In

    animal models of hMPV infection it has been shown that a TLR 4 mediated inflammatory

    response does not facilitate an adaptive immune response important for viral clearance

    and protection against reinfection but predicts the progression of clinical disease

    (Velayutham et al., 2013). In RSV infection, PRRs correlate with viral load and there is an

    increased pulmonary expression of PRRs compared to healthy controls or infants with RV

    or hBoV infection (Scagnolari et al., 2009).

  • 24

    Different viral infections can elicit different inflammatory responses, and these

    inflammatory responses can generally be grouped into T-helper 1 (Th1) and T-helper 2

    (Th2) responses. These inflammatory responses are classified based on the type of

    chemokines and cytokines produced. Th1 responses are characterised by production of

    IFN-γ, IL-1β, IL-2, IL-12, IL-18, and TNF-α. The Th1 response is pro-inflammatory and is

    important in the generation of immune responses necessary for the control and clearance

    of intracellular pathogens, thus it is the most suitable response to viral infection. The Th2

    response on the other hand is characterised by secretion of IL-4, IL-5, IL-6, IL-9, IL-10, and

    IL-13. This type of response is involved in antibody production (including IgE) and

    eosinophilic inflammation. This response is strongly associated with atopy and protection

    against parasitic infection. It has also been demonstrated that Th2 inflammation counter

    regulates Th1 mediated responses (Gill et al., 2010). Respiratory viral infections provoke

    differing inflammatory profiles. For instance, IFAV can induce the overproduction of Th1

    inflammatory mediators and this is associated with poor clinical outcomes. Dysregulated

    secretion of TNF-α and IL-1β maybe critical for pathogenicity and may also hinder the

    development of an effective adaptive immune response following infection (Han et al.,

    2014). In RSV disease, sequential increases of IFN-γ coincide with improved clinical

    outcomes and provide support for the protective role of IFN-γ following infection

    (Bermejo-Martin et al., 2007a). Conversely, severe acute viral respiratory infection in

    infants is associated with augmented Th2 responses (Bermejo-Martin et al., 2007b). In

    addition, individuals with an underlying Th2 bias, such as those with an allergic pulmonary

    condition, are more likely to have poor clinical outcomes compared with those without an

    underlying Th2 bias (Gern et al., 2000; Mahmutovic Persson et al., 2016; Monick et al.,

    2007).

    Many types of cells have been implicated in the pathogenesis of ARTI and of note is the

    quick and robust pulmonary and systemic neutrophil response following infection

  • 25

    (Nagarkar et al., 2009; Sugamata et al., 2012). This robust neutrophil response correlates

    with disease severity and is mediated primarily by IL-8 (Cortjens et al., 2016). Neutrophilia

    is a common response to both bacterial and viral infections. While there are clear

    protective roles for neutrophils against bacterial infection, the evidence for an anti-viral

    role is less clear. The role of neutrophils has been investigated in RSV infection and they

    are believed to play a role in pathogenesis (Cortjens et al., 2016; Linden, 2001;

    Mahmutovic Persson et al., 2016). The role of neutrophils in influenza mouse models is

    reported to be both protective and pathogenic. Some papers suggest neutrophils can

    reduce viral load by phagocytosis of infected cells and trapping of free virus in neutrophil

    extracellular traps. However, other reports have demonstrated that neutrophil

    myeloperoxidase facilitates acute respiratory distress syndrome following influenza

    infection and that mice lacking myeloperoxidase were more competent in reducing viral

    load (Sugamata et al., 2012). Neutrophil involvement has also been demonstrated in RV-

    induced asthma exacerbations and high counts are observed in the airways of individuals

    following fatal exacerbations (Fahy, 2009; Fahy et al., 1995; Linden, 2001). In a RV mouse

    model, IL-8 knock-out mice infected with RV showed significantly reduced neutrophil

    responses, and demonstrated reduced airway hyper-responsiveness (Nagarkar et al.,

    2009). Functional neutrophil activity has been demonstrated to be enhanced in the airways

    of asthmatics, particularly during exacerbations, and correlates with reductions in lung

    function and increases in symptom score (Proud, 2011). However, the precise contribution

    of neutrophils to asthma has yet to be established.

  • 26

    1.6 Aims of Project

    The general aim of this thesis was to develop reliable quantitative PCR (viral load) assays to

    investigate the contribution of certain respiratory viruses to clinical outcome of young

    children hospitalised with an acute viral respiratory tract infection.

    The specific aims were as follows:

    To develop and validate the use of reliable in-house qPCR (viral load) assays for

    different respiratory viruses including IFAV, hMPV, RSV-A&B, and RV-C.- Chapter 3

    and 4 (RV-C only)

    To compare the analytical performance of real time qPCR to digital PCR in the

    quantitative measurement of RSV load in clinical samples - Chapter 3

    To examine the contribution of RSV to clinical disease in young infants in a high HIV

    prevalence setting- Chapter 5

    To investigate the contribution of RV-C to disease severity in young children

    presenting to emergency department with acute asthma exacerbations - Chapter

    6.

    To understand the interplay between RV-C infection and the innate immune

    response in young children hospitalised with a RV-C associated wheezing illness -

    Chapter 7

  • 27

    2 Materials and Methods

    2.1 Sample collection

    Clinical specimens included flocked nasal swabs, per nasal aspirates, sputum and

    nasopharyngeal aspirates samples. Swabs were placed in virus transport medium (VTM)

    which contains Hanks balanced salt solution, with 50mg gentamicin, 0.1% bovine serum

    albumin, 0.1% sodium bicarbonate and 8mM HEPES buffer).

    Nasopharyngeal aspirates (NPAs) were collected from 105 ALRI cases and 53 controls from

    Pretoria, South Africa between July 2011 and November 2012 (Chapter 5). Children 0-2

    years of age hospitalized with an ALRI at the Steve Biko Academic Hospital or Tshwane

    District Hospital in Pretoria were enrolled as cases. A diagnosis of pneumonia (respiratory

    distress and either chest X-ray changes (e.g. consolidation or effusion), or auscultatory

    findings (e.g. crepitations or bronchial breathing) or bronchiolitis (respiratory distress and

    at least one of the following; wheeze, chest X-ray changes (e.g. hyperinflation) or

    Hoovers’s sign (inward movement of the lower rib cage during inspiration) was determined

    by the clinician in charge. Fifty-three age-matched children presenting to the same

    hospitals with a non-respiratory illness over the same period were enrolled as controls

    (NRD controls). The study protocol was approved by the University of Western Australia

    Human Ethics Committee and the University of Pretoria Ethics Committee. Written

    informed consent from parent/guardian was obtained prior to participation. NPAs were

    stored at -80°C in Pretoria, South Africa until transfer on dry ice to Telethon Kids Institute,

    Perth, Australia for processing and long term storage. An aliquot of each sample was

    transported on dry ice to PathWest Laboratory Medicine WA, Perth, Australia.

    For Chapter 6, flocked nasal swabs were collected from young children presenting with an

    acute asthma exacerbation as part of the MAVRIC (Mechanisms of Acute Viral Respiratory

  • 28

    Infection in Children) study carried out at Princess Margaret Hospital in Perth, Western

    Australia. Flocked nasal swab specimens were also collected from children with no

    evidence of respiratory disease. These children were selected to match the acute asthma

    exacerbation cases for age, and season of birth. An aliquot of each sample was

    transported on dry ice to PathWest Laboratory Medicine WA, Perth, Australia and stored

    at -80oC until further use.

    Study participants for the cytokine study (Chapter 7) had a flocked swab sample collected

    upon inpatient admission. These samples were placed on ice and transported to the

    PathWest laboratory as soon as practically possible for storage in -80oC freezer.

    2.2 Nucleic acid extraction and viral detection

    Total nucleic acids were extracted from 200µL of respiratory specimen using the MagMAX

    viral RNA isolation kit (Thermo-Fisher Scientific, Australia) according to the manufacturer’s

    instructions. All automated liquid transfer procedures utilised the CAS 1200 instrument

    and conductive tips (Corbett Life Science. Australia)

    An in-house multiplex respiratory pathogen assay was used to screen study samples for

    respiratory pathogens such as influenza A, B and C viruses (FLUAV, FLUBV,FLUCV),

    parainfluenza virus I-IV (PIV), RSV-A & B, Coronaviruses (hCoV- HKU1, -NL63, -OC-43, -

    229E), human metapneumovirus (hMPV), and human adenoviruses (hAdV) (Chidlow et al.,

    2009). Every multiplex PCR run performed included positive (PCR, inhibition and extraction

    positive controls) and negative controls (method blanks interspersed by 5 test samples).

    Nucleic acid extracts were stored at -80oC. A few minor adjustments were made to the

    multiplex PCR real-time assay (Chidlow et al., 2009). Adjustments included the addition of

    a 2.5 µM ROX reference dye (ThermoFisher), which was to be used for the 1:10 dilution of

    amplicon generated from the enrichment PCR assays. Further, extra primer pairs were

  • 29

    included to screen for hMPV (Chidlow et al., 2009). The primer pairs reported by Lee and

    colleagues (Lee et al., 2007) were used for RV identification and genotyping. Respective

    monoplex quantitative PCR assays were used to confirm any discordant results obtained

    from the respiratory multiplex PCR assay. Thermal cycling programmes for the enrichment

    PCR assays and the real time multiplex PCR assay were adopted from (Chidlow et al.,

    2009).

    2.3 Design of primers and probes

    Assay oligonucleotides were designed using primer express v3.0 software (primers and

    MGB probes) (ThermoFisher Scientific, Australia or the Exiqon microRNA PCR online

    service (LNA probes).Primers were synthesised by Integrated DNA Technologies (IDT,

    Australia), MGB probes were synthesized by Applied Biosystems (ThermoFisher Scientific,

    Australia) and Locked Nucleic Acid (LNA) probes were synthesized by Sigma-Aldrich (Sigma-

    Aldrich, Australia). The primers and probes used in the multiplex PCR to screen initially for

    viral nucleic acid in respiratory samples were as previously described (Chidlow, 2013;

    Chidlow et al., 2009).

    Viral load from clinical samples was determined by real-time PCR targeting specific regions

    of nucleoprotein gene of RSV (A&B) and HMPV, the matrix gene of IFAV and the

    5’untranslated region (UTR) of RV-C (Table 2.1). These sequences were used because they

    are highly conserved across all the respective strains investigated. Table 2.1 lists primers

    and probes details for IFAV, RSV (A&B), HMPV, RV-C and Glyceraldehyde-3-phosphate

    dehydrogenase (GAPDH) assay

  • 30

    Table 2.1: Primers and probes used in quantitative real-time PCR assays

    aLNA bases are underlined.

    Target Target region Oligonucleotide sequence (Position) a

    HMPV F (Forward Primer) Nucleoprotein gene 5’-ATCATCAGGYAAYATYCCACAAA-3’ (420 - 442)

    HMPV R (Reverse Primer ) 5’- TATTAARGCACCTACACATAATAA-3’ (518 -542)

    HMPV (Probe) 5’-FAM-CCTGCGTGGCTGCC-MGBNFQ-3’ (481- 497)

    RSV-A F (Forward Primer) Nucleoprotein gene 5’CAACTTCTGTCATCCAGCAAA3’(1117 -1137)

    RSV-A R (Reverse Primer) 5’TGCACATCATAATTAGGAGTATCAAT3’ (1166-1191)

    RSV-A Probe 5’-FAM-CACCATCCAACGGAGC`3’-BHQ-1 (1140 – 1155)

    RSV-B F (Forward Primer) Nucleoprotein gene 5’ATTCAACGTAGTACAGGAGATAATA3’ (1141 - 1165)

    RSV-B R (Reverse Primer ) 5’CCACATAGTTTGTTTAGGTGTTT’ (1193 -1214)

    RSV-B Probe 5’-FAM-TGACACTCCCAATTAT3’-BHQ-1 (1167 – 1182)

    IFAV (Forward Primer) Matrix gene 5’CTTCTAACCGAGGTCGAAACGTA3’ (7-29)

    IFAV (Reverse Primer ) 5’-GGTGACAGGATTGGTCTTGTCTTTA--3’ (137-161)

    IFAV (Probe) 5’FAM-TCAGGCCCCCTCAAAGCCGAG3’-BHQ-1 (49-69)

    RV-C IrlonS (Forward Primer) 5’UTR 5’-GCACTTCTGTTTCCCC-3’ (165 - 180)

    RV-C EntA (Reverse Primer) 5’- GCATTCAGGGGCCGGAG-3’ (461 -445)

    RV-C Probe 1 5’-FAM-CCTGCGTGGCTGCC-MGBNFQ-3’ (358 - 371)

    RV-C Probe 2 5’-FAM-CCCGCGTGGCTGCC3’-BHQ-1 (359 - 372)

    RV -C Probe 3 5’-FAM-CCCGCGTGGTGCCC-MGBNFQ-3’ (354 - 367)

    RV -C Probe 4 5’ -FAM-CCTGCGTGGTGCCC3’-BHQ-1 (354 – 367)

    GAPDH (forward) GAPDH mRNA 5’GAAGGTGAAGGTCGGAGTC3’ (7-25)

    GAPDH (reverse) 5’AAATCCCATCACCATCTTC3’ (213-231)

    GAPDH probe 5’-FAM-GGCTGAGAACGGGAAGCTTG-MGBNFQ

  • 31

    2.4 Production and quantification of transcribed RNA

    standards Nucleotide sequences matching a segment of the 5' UTR of RV-C2 [EF077280.1], RV-C6

    [EF582387], RV-C51 [JF317015],RV-C25 [JF317013], RSV A [KJ627348]&B [KU950585] NP gene,

    HMPV NP gene [AHV79765], IFAV matrix gene [CY056296] were incorporated into individual

    plasmid constructs manufactured by Integrated DNA Technologies (IDT,