assessing maternal, environmental and individual …

228
Assessing Maternal, Environmental and Individual Risk Indicators for Dental Caries in a Population of Children from Queensland, Australia Author Fernando, Surani Published 2017-08 Thesis Type Thesis (PhD Doctorate) School School of Dentistry&Oral Hlth DOI https://doi.org/10.25904/1912/859 Copyright Statement The author owns the copyright in this thesis, unless stated otherwise. Downloaded from http://hdl.handle.net/10072/371955 Griffith Research Online https://research-repository.griffith.edu.au

Upload: others

Post on 13-Mar-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Assessing Maternal, Environmental and Individual RiskIndicators for Dental Caries in a Population of Children fromQueensland, Australia

Author

Fernando, Surani

Published

2017-08

Thesis Type

Thesis (PhD Doctorate)

School

School of Dentistry&Oral Hlth

DOI

https://doi.org/10.25904/1912/859

Copyright Statement

The author owns the copyright in this thesis, unless stated otherwise.

Downloaded from

http://hdl.handle.net/10072/371955

Griffith Research Online

https://research-repository.griffith.edu.au

i

ASSESSING MATERNAL, ENVIRONMENTAL AND

INDIVIDUAL RISK INDICATORS FOR DENTAL CARIES IN

A POPULATION OF CHILDREN FROM QUEENSLAND,

AUSTRALIA

Surani Fernando

BDS, MSc, MD

School of Dentistry and Oral Health

Menzies Health Institute Queensland

Griffith University

Submitted in fulfillment of the requirements for the degree of Doctor of

Philosophy

August 2017

ii

STATEMENT OF ORIGINALITY

This work has not previously been submitted for a degree or diploma in any

University. To the best of my knowledge and belief, the thesis contains no

material previously published or written by another person except where due

reference is made in the thesis itself.

Surani Fernando

25.08.2017

iii

ACKNOWLEDGEMENTS

This doctoral thesis was carried out under the primary supervision of Professor Newell Johnson

and Professor Paul Scuffham with the assistance of, Professor Rodney Lea and Dr. David

Speicher as associate supervisors. I acknowledge their co-operation, time, and contributions,

which have helped to bring this work to its current form. I am thankful for their constant

motivation and encouragement when I needed it. I am grateful to each one of them for helping

me to sharpen my skills in scientific writing and critical thinking.

Particularly, I am indebted to Professor Newell Johnson and Professor Paul Scuffham for many

aspects of the oral health arm of the Environments for Healthy Living Study. I wish to thank

Professor Paul Scuffham for his kind and generous support during the stressful phase of

merging oral health data with the main EFHL data base and data analysis. I thank Professor

Rodney Lea for his continuous encouragement and support throughout the Epigenome–Wide

Association Study. I also acknowledge the advice and training given by Dr. David Speicher

during laboratory work.

A special thank you goes to Mr. Gabor Mihala, for all the advice and support given throughout

data management and analysis. I am thankful for the contributions of the EFHL Project

Manager, Rani Scott, and the current and past Database Managers. I thank Dr. Miles Benton

for helping with DNA sequencing and methylation assays and Dr. Ping Zhang for her advice

on bioinformatics analysis. I would also like to acknowledge the contribution of Professor Shu

Kay Ng and Dr. Santhosh Kumar for their statistical support. I am grateful for the help I have

received from Dr. Mahmoud Bakr during clinical data collection.

iv

I acknowledge the support of the Head of School, Dentistry and Oral Health, management of

Griffith Health Clinics and its team of Dental Assistants and Receptionists with the clinical

work.

Professional editor, Dr. John McAndrew, provided copyediting and proofreading services,

according to the guidelines laid out in the university-endorsed national Guidelines for Editing

Research Theses.

This study would not be possible without the co-operation of the mothers and children who

participated. I especially appreciate the mothers who were willing to spare their time to bring

their children for oral examinations at the Griffith Dental clinics. They agreed to participate

and they were kind enough to complete the questionnaires and patiently sit through the clinical

examination during late afternoons.

Although words cannot merely explain my gratefulness, I am thankful to my wonderful

husband, Shaun who lovingly and without complaining helped me with the housework and

children. He has been my constant support and pillar of strength throughout this journey. Thank

you, my lovely daughters, Kiyashi, Amaya, Emalshi, and Tiasha, for sacrificing many cuddles,

late-nights, weekends, and school holidays, so that I could remain focused on my work. I would

not have been able to complete this thesis without my family’s support and understanding.

I will be failing in my duties if I do not express my gratitude to my parents, Dawson and Dulcey,

for their sacrifices, support, love, and prayers without which I could not have come this far.

v

I am very grateful to Griffith University for scholarships and support. Lastly, I would like to

acknowledge and thank my examiners for the time and effort they have dedicated to reading

this thesis and providing valuable feedback to improve my work as a researcher.

vi

ABSTRACT

Oral diseases affect 3.9 billion people worldwide, and dental caries is the most prevalent oral

condition. Untreated caries in permanent and deciduous dentitions were reported among

approximately 35% and 9% of the world population, respectively, in 2010. Moreover, it is the

fourth-most expensive chronic disease to treat according to the World Health Organization.

The importance of various social, environmental, familial, and behavioural factors on

childhood caries has been identified by earlier researchers. In the past, epidemiological

research on caries has mainly focused on describing biological and dietary determinants of the

disease. During the past few decades, there has been an effort to explore children’s oral health

outcomes using a broader framework, incorporating behavioural, social, and environmental

predictors with biological and genetic factors. Despite the fact that these characteristics have

been found to be significantly associated with childhood caries experience in different

population groups, there has been scarce research exploring the whole range of putative risk

factors, and their associated risk indicators, and their possible interactions, simultaneously, in

a single child cohort. In particular, the impact of the epigenome and of the in utero environment

on susceptibility to dental caries has not been reported in children from a typical Western

industrialised country. Hence, this thesis explored a wide range of factors: environmental,

socio-economic, behavioural, maternal (including throughout pregnancy), children’s

individual factors (including well-established risks associated with diet and hygiene practices),

and a screen for epigenetic modifications, as indicators of risk for childhood caries.

Participants were 174, six- to seven-year-old children and their mothers from South East

Queensland, who were originally enrolled in the Environments for Healthy Living (EFHL)

Griffith University birth cohort study. Participants for the oral health sub-study were volunteers

vii

obtained from the EFHL database who were willing to come for an oral examination at Griffith

dental clinics.

Mothers completed a questionnaire on oral health knowledge and behaviours at the dental clinic

followed by anthropometric measurements. Trained and calibrated examiners conducted

detailed head and neck, and oral examinations, and recorded participant’s salivary

characteristics and dental caries scores. Total DNA was extracted from each participant’s saliva

for sequencing and methylation arrays to detect epigenetic changes. Descriptive statistics,

negative binomial regression, and path analyses were performed to evaluate the associations

between risk indicators and the lifetime dental caries experience of each child.

Maternal risk indicators included mother’s age at examination, her lifetime caries experience,

oral health knowledge and practices, body mass index, saliva characteristics of clinical oral

hydration, stimulated flow rate, pH, buffering capacity, and load of salivary mutans

Streptococci and Lactobacilli.

Results indicated that low annual household income was a risk indicator for dental caries

experience in this child population. Maternal behaviours: initiating child’s tooth brushing later

than six months of age and a high frequency of giving carbonated drinks to the child were

associated with increased caries experience in their children. In addition, high loads of maternal

oral Lactobacilli were related to the children’s increased risk of caries. Moreover, children’s

past caries experience, and increased levels of salivary mutans Streptococci were recognised

to be significant risk indicators for their dental caries experience. Children whose mothers took

iron supplements during pregnancy had low levels of caries (past and current) compared to

their counterparts.

viii

One highlight of the thesis has been the cutting-edge evidence of an association between

epigenetic modifications and caries experience of individuals. There were significant

differences in differentially methylated regions between persons with high and low caries

experience on chromosomes 1, 2, 5, 7, 12, 20 and 22. Chromosome 12, in particular, showed

the highest average methylation difference between the two groups. Further, functional

annotations of related genes revealed that there are two gene clusters related to zinc (Zn)

metabolism and membrane protein functions, which could indirectly be related to the caries

process.

In summary, the study found maternal, environmental, and children’s characteristics that were

risk indicators for caries experience in this child population. It was observed that low maternal

income and poor oral health behaviours had positive associations with children’s caries

experience. Also, bad oral health of the mother, expressed as maternal caries experience, placed

their children at risk of dental caries. Similarly, children with poor oral health with past caries

and high counts of cariogenic bacteria, along with reported low maternal iron supplements

during pregnancy were more at risk of continuing to develop dental caries than their

counterparts. The latter could reflect inadequate iron during pregnancy and consequent sub-

optimal development of foetal tissues and immune responses. The pilot study of 12 mother

child dyads to explore associations between epigenetic changes and lifetime caries experience

suggests that epigenetic modifications might, indeed, predispose individuals to dental caries.

However, with such a small sample size, this cannot be a firm conclusion. Nevertheless it leads

to the generation of hypotheses which can be tested in further studies.”

This thesis adds new knowledge to the current literature on local, systemic and environmental

factors influencing susceptibility and resistance to the process of childhood caries. The results

also offer new avenues for health promotion using a common risk factor approach to improve

ix

maternal behaviours and nutrition, which would eventually enhance the clinical caries

outcomes in children.

x

TABLE OF CONTENTS

STATEMENT OF ORIGINALITY ........................................................................................... ii

ACKNOWLEDGEMENTS ..................................................................................................... iii

ABSTRACT .............................................................................................................................. vi

TABLE OF CONTENTS ........................................................................................................... x

LIST OF TABLES ................................................................................................................... xv

LIST OF FIGURES ............................................................................................................... xvii

LIST OF APPENDICES ...................................................................................................... xviii

STATEMENT OF ETHICS APPROVAL.............................................................................. xix

LIST OF PUBLISHED AND SUBMITTED PAPERS FROM THE THESIS ....................... xx

ACKNOWLEDGEMNET OF PUBLICATIONS INCLUDED IN THE THESIS ............... xxii

Chapter 1: Introduction .............................................................................................................. 1

1.1 Background ...................................................................................................................... 1

1.2 Rationale for conducting the research in South East Queensland .................................... 5

1.3 Research questions and objectives ................................................................................... 7

1.4 Contribution of the study.................................................................................................. 9

1.5 The structure of the thesis .............................................................................................. 10

Chapter 2: Review of literature ................................................................................................ 12

2.1 Introduction .................................................................................................................... 12

2.2. The burden of dental caries in children ......................................................................... 13

2.3 Proposed Conceptual frameworks describing the process of dental caries .................... 15

2.4 Determinants of dental caries ......................................................................................... 19

2.4.1 Maternal and environmental determinants .............................................................. 20

2.4.1.1 Socio-economic status .......................................................................................... 20

2.4.1.2 Family structure and environment ........................................................................ 21

2.4.1.3 Maternal level of education, oral health knowledge and behaviours ................... 22

2.4.1.4 Maternal body mass index .................................................................................... 23

2.4.1.5 Maternal caries ..................................................................................................... 24

2.4.1.6 Maternal cariogenic bacteria ................................................................................ 25

2.4.1.7 Maternal salivary physiology ............................................................................... 26

2.4.2 Influence of children’s individual factors ................................................................ 28

xi

2.4.2.1 Intrauterine environment of child and birth weight .............................................. 28

2.4.2.2 Age and gender ..................................................................................................... 29

2.4.2.3 Past dental caries .................................................................................................. 30

2.4.2.4 Physicochemical properties of saliva ................................................................... 31

2.4.2.5 Oral cariogenic bacteria ........................................................................................ 32

2.5 Concluding remarks ....................................................................................................... 42

Chapter 3: Protocol for assessing maternal, environmental and epigenetic risk indicators for

dental caries in a population of Queensland children .............................................................. 43

3.1 Abstract .......................................................................................................................... 44

3.1.1Background ............................................................................................................... 44

3.1.2 Methods ................................................................................................................... 44

3.1.3 Discussion ................................................................................................................ 45

3.2 Background .................................................................................................................... 45

3.3 Methods .......................................................................................................................... 50

3.3.1 Study population ...................................................................................................... 50

3.3.2 Sample recruitment and examination ...................................................................... 51

3.3.3 Primary outcome variable ........................................................................................ 53

3.3.4 Main explanatory variables ..................................................................................... 53

3.3.5 Oral health knowledge and practices ....................................................................... 53

3.3.6 Anthropometric measurements ................................................................................ 54

3.3.7 Saliva characteristics ............................................................................................... 54

3.3.8 Periodontal status ..................................................................................................... 54

3.3.9 Demographic and environmental data ..................................................................... 55

3.3.10 Genetic and epigenetic markers ............................................................................. 55

3.4 Analysis .......................................................................................................................... 56

3.5 Discussion ...................................................................................................................... 59

Chapter 4: Maternal caries experience, household income, carbonated drinks and age at

commencement of tooth-brushing are associated with caries experience among 6- to 7-year-

old children in Australia........................................................................................................... 60

4.1 Abstract .......................................................................................................................... 61

4.2 Introduction .................................................................................................................... 62

4.3 Materials and methods ................................................................................................... 64

4.3.1 Study population ...................................................................................................... 65

4.3.2 Data collection ......................................................................................................... 65

xii

4.3.3 Outcome and risk indicators .................................................................................... 66

4.3.4 Statistical analyses ................................................................................................... 67

4.4 Results ............................................................................................................................ 68

4.5 Discussion ...................................................................................................................... 72

Chapter 5: Mothers who report taking iron supplements during pregnancy and child’s oral

carriage of mutans Streptococci are associated with a child’s lower and higher lifetime dental

caries experience, respectively................................................................................................. 78

5.1 Abstract .......................................................................................................................... 79

5.1.1 Objective .................................................................................................................. 79

5.1.2 Methods ................................................................................................................... 79

5.1.3 Results ..................................................................................................................... 79

5.1.4 Conclusions ............................................................................................................. 80

5.2 Introduction .................................................................................................................... 80

5.3 Methods .......................................................................................................................... 82

5.4 Results ............................................................................................................................ 87

5.5 Discussion ...................................................................................................................... 90

5.6 Conclusions .................................................................................................................... 93

Chapter 6: Influence of genes on the process of dental caries and what lies beyond .............. 94

6.1 Introduction .................................................................................................................... 94

6.2 Evidence that the host genome influences the natural history of dental caries .............. 95

6.3 Types of molecular genetic studies ................................................................................ 96

6.3.1 Genome-wide Studies .............................................................................................. 96

6.3.1.1 Genome-wide linkage studies ............................................................................. 96

6.3.1.2 Genome-wide association studies ....................................................................... 97

6.3.2 Candidate gene studies .......................................................................................... 101

6.3.2.1 Enamel formation genes ................................................................................... 105

6.3.2.2 Immune response genes .................................................................................... 107

6.3.2.3 Genes related to taste sensation ........................................................................ 110

6.3.2.4 Genes related to saliva ...................................................................................... 112

6.4 Genes, caries and environment..................................................................................... 114

6.4.1 Evidence from twin studies ................................................................................. 115

6.5 Epigenetics ................................................................................................................... 116

Mechanisms of Epigenetic modification ............................................................................ 117

6.6.1 DNA methylation .................................................................................................. 117

xiii

6.6.2 Histone modification ............................................................................................. 118

6.6.3 Non-coding RNA ................................................................................................... 119

6.7 Environmental stimuli which may result in epigenetic changes which could influence the

process of dental caries ...................................................................................................... 119

6.7.1 Diet and nutrition ................................................................................................... 120

6.7.2 Tobacco ................................................................................................................. 121

6.7.3 Bacteria and inflammation..................................................................................... 121

6.8 Future research ............................................................................................................. 122

Chapter 7: An Epigenome- Wide Association Study of mother-child dyads discordant for

dental caries: A pilot study .................................................................................................... 124

7.1 Abstract ........................................................................................................................ 125

7.1.1 Background ............................................................................................................ 125

7.1.2. Objective ............................................................................................................... 125

7.1.3 Methods ................................................................................................................. 125

7.1.4 Results ................................................................................................................... 126

7.1.5 Conclusion ............................................................................................................. 126

7.2 Introduction .................................................................................................................. 126

7.3 Methods ........................................................................................................................ 128

7.3.1 Study Population.................................................................................................... 128

7.3.2 Dental caries assessment ....................................................................................... 129

7.3.3 Sample Collection and Epigenetic analysis ........................................................... 129

7.4 Results .......................................................................................................................... 135

7.4.1 Methylation differences on chromosomes ............................................................. 136

7.4.2 Functions of identified genes ................................................................................. 136

7.5 Discussion, conclusion and limitations ........................................................................ 138

7.5.1 Discussion .............................................................................................................. 138

7.5.2 Conclusion ............................................................................................................. 141

7.5.3 Limitations ............................................................................................................. 141

Chapter 8: General Discussion............................................................................................... 143

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

8.2 Overview of the study .................................................................................................. 143

8.2.1 Background and research objectives ..................................................................... 143

8.3 Main empirical findings ............................................................................................... 145

8.3.1 Dental caries experience in the sample of children and their mothers .................. 145

xiv

8.3.2 Oral health knowledge and behaviours of mothers ............................................... 147

8.3.3 Maternal and environmental factors that influence the caries experience of children

........................................................................................................................................ 148

8.3.4 Children’s individual factors: prenatal environment, birth weight, and intra oral

characteristics as risk indicators for caries experience ................................................... 149

8.3.5 Epigenome-Wide Association Study of individuals discordant for dental caries: The

pilot study ....................................................................................................................... 151

8.4 Limitations ................................................................................................................... 154

8.5 Discussion and recommendations for future studies .................................................... 155

8.5.1Strengths of the study ............................................................................................. 155

8.5.2 Future research directions ...................................................................................... 156

8.6 Conclusions .................................................................................................................. 159

References .............................................................................................................................. 161

Appendix A : Oral Health Questionnaire............................................................................... 188

Appendix B: Kappa statistics for dental caries examination ................................................. 194

Appendix C: Search strategy used in litrrature search ........................................................... 196

Appendix D: Supplementary tables for differentially methylated CpG sites on chromosome 12

between cases and controls .................................................................................................... 199

Appendix E: Results of further pyrosequencing done on an extended sample of individuals

discordant for dental caries .................................................................................................... 201

Appendix E 1. Rationale .................................................................................................... 201

Appendix E 2. Results ........................................................................................................ 202

Appendix E 3. Future directions ........................................................................................ 205

xv

LIST OF TABLES

Table No: Title Page/s

Table 2.1 A summary of maternal, environmental and individual

determinants of childhood dental caries

34-41

Table 3.1 Percentage and of 6-7 year old children with caries experience in

Australia in 2011 ( 95% Confidence Interval )

46

Table 4.1 Frequency distribution of children’s and mothers’ socio

demographic and clinical characteristics.

70

Table 4.2 Frequency distribution of mother’s oral health related knowledge

and behaviours

71

Table 4.3 Risk indicators for children with and without dental caries using

logistic regression

71

Table 4.4 Risk indicators for childhood dental caries using negative binomial

regression

72

Table 5.1 Children’s individual characteristics explored as risk indicators for

dental caries experience and current activity

85

Table 5.2 Potential subject-based risk indicators for dental caries in children

analysed by structural equation modelling

86

Table 6.1 A summary of Genome-wide linkage and Genome-wide

association studies for human dental caries

99-100

Table 6.2 A summary of candidate genes studies for human dental caries 102-104

Table 7.1 Caries experience of controls and cases 132

Table 7. 2 Demographic and socio economic characteristics of controls and

cases

133

Table 7.3 Annotated data for the 16 identified Differentially Methylated

Regions between controls and cases

134

Table 7.4 Functions of gene clusters derived from DAVID 135

Table 7.5 Summary of Differentially Methylated Regions (with only one

CpG site) related to a gene promoter region.

137

Appendix B

Table 1

Calibration of examiner 1 for ICDAS criteria against a gold

standard

194

xvi

Appendix B

Table 2

Calibration of examiner 2 for ICDAS criteria against a gold

standard

195

Appendix C

Table 1

Search strategy used in PubMed 196

Appendix C

Table 2

Search strategy used in Scopus 197

Appendix C

Table 3

Search strategy used in Google Scholar 198

Appendix D

Table 1

Comparison of mean Differentially Methylated Regions on

chromosome 12 between controls and cases amongst children

199

Appendix D

Table 2

Comparison of mean Differentially Methylated Regions on

chromosome 12 between controls and cases amongst mothers.

200

Appendix E

Table 1

Percentage methylation analysis of CpG sites; Chr12:11700151,

Chr12:11700148 and Chr12:11700124 between individuals with

high and low caries experience

203

Appendix E

Table 2

Comparison of differential methylation of Chr12:11700151,

Chr12:11700148 and Chr12:11700124 between individuals with

high and low caries experience

204

Appendix E

Table 3

Correlation between individuals with high and low caries

experience and percentage methylation of CpGs;

Chr12:11700151, Chr12:11700148 and Chr12:11700124

204

xvii

LIST OF FIGURES

Figure Title Page

Figure 1.1 Community, maternal, child and epigenetic influences on

dental caries of children.

7

Figure 2.1 Child, family and community influences on oral health

outcome of children.

18

Figure 3.1 Conceptual framework for the possible interaction of risk

indicators under investigation.

48

Figure 3.2 Flowchart showing the process of data collection 52

Figure 5.1 Path analysis model with estimate of association between

dependant and independent variables

89

Figure 6.1 Environmental factors effecting epigenetic modification 115

Figure 6.2 A CpG island in relation to a gene promoter region 117

Figure 6.3 DNA methylation at CpG sites. 118

xviii

LIST OF APPENDICES

Appendix Title Page

Appendix A Oral Health Questionnaire 188

Appendix B Kappa statistics for dental caries examination 194

Appendix C: Search strategy used in literature search 197

Appendix D Supplementary tables for differentially methylated CpG sites

on chromosome 12 between cases and controls

199

Appendix E Results of further pyrosequencing done on an extended

sample of individuals discordant for dental caries

203

xix

STATEMENT OF ETHICS APPROVAL

I confirm that ethical clearance for this research was granted by the Griffith University Human

Research Ethics Committee, Australia (GU Ref No: DOH/20/12/HREC and GU Ref No:

OTH/25/13/HREC). I confirm that research was conducted in accordance with the approved

protocol.

Surani Fernando

Supervisor: Professor Newell Johnson Supervisor: Professor Paul Scuffham

xx

LIST OF PUBLISHED AND SUBMITTED PAPERS FROM

THE THESIS

Published papers

1. Fernando S, Speicher DJ, Bakr MM, Benton MC, Lea RA, Scuffham PA et al. (2015).

Protocol for assessing maternal, environmental and epigenetic risk indicators for dental

caries in children. BMC Oral Health 15(1):167. DOI.org/10.1186/s12903-015-0143-2

(29 December 2015).

Submitted papers

1 Surani Fernando, Gabor Mihala, Mahmoud Bakr, Shu-Kay Ng, Rodney Lea, Paul A

Scuffham, Newell W Johnson. Maternal caries experience, household income,

carbonated drinks and age at commencement of tooth-brushing are associated with

caries experience among 6-7 year old children in Australia. (Submitted to “Caries

Research” and under review- manuscript ID 201705020 )

2 Surani Fernando, Santosh Kumar, Mahmoud Bakr, David Speicher, Rodney Lea, Paul

A Scuffham, Newell W Johnson. Mothers who report taking iron supplements during

pregnancy and child’s oral carriage of mutans Streptococci are associated with a child’s

lower and higher lifetime dental caries experience, respectively (Submitted to “Journal

of Public Health Dentistry” and under review- manuscript JPHD-OA-08-17-0226).

xxi

3 Surani Fernando, Miles Benton, David Jeremiah Speicher, Mahmoud Bakr, Ping

Zhang, Rodney Lea, Paul A Scuffham, Newell Walter Johnson. An Epigenome Wide

Association Study of mother child dyads discordant for dental caries: A Pilot Study.

(Submitted to “BMC Oral Health” and under review-manuscript ID OHEA-D-17-

00215)

xxii

ACKNOWLEDGEMNET OF PUBLICATIONS INCLUDED IN

THE THESIS

The publications (published and submitted) included in this thesis are co-authored with other

researchers. These publications are in accordance with Section 9.1 of the Griffith University

Code for the Responsible Conduct of Research (“Criteria for Authorship”), Section 5 of the

Australian Code for the Responsible Conduct of Research, and Section 9.3 of the Griffith

University Code (“Responsibilities of Researchers”).

My contribution to each co-authored paper is outlined at the front of the relevant chapter.

Appropriate acknowledgments of those who contributed to the research but did not qualify as

authors are included in the thesis acknowledgments.

Surani Fernando

Supervisor: Professor Newell Johnson Supervisor: Professor Paul Scuffham

1

Chapter 1: Introduction

1.1 Background

Dental caries remains the most prevalent disease worldwide, burdening billions of people,

especially children, with pain and subsequently poor quality of life and general health

(Schwendicke et al., 2015; Kassebaum et al., 2015). According to the FDI World Dental

Federation, in 2010 44% of the world’s population was affected by tooth decay (Kieu et al.,

2017). Untreated dental caries is a major public health problem affecting 621 million children

worldwide (Kassebaum et al., 2015). Despite progress made in caries control by the protective

effects of fluoride, increased interventions in oral health promotion, widespread health

education, and advanced treatment options, dental caries still persists as a disease that places

a significant financial burden on both individuals and health care systems (Splieth et al., 2016).

The basics of the caries process are simple: acids derived from the fermentation of carbohydrate

foods dissolve enamel, and later dentine, if oral hygiene is poor and large volumes of microbial

biofilm remain on tooth surfaces. This means that, fundamentally, prevention of this disease is

simple and within the means of most ordinary members of society through self-care: restrict

the intake of readily fermentable carbohydrate and brush teeth thoroughly once or twice a day

with a fluoridated toothpaste (Cohen et al., 2017). Nevertheless, in real life, most people fail to

maintain their own oral health, and the predisposing susceptibility and resistance factors are

highly complex. In that sense, dental caries is a multifactorial disease, complexly modified by

genetic, behavioural, social, and environmental factors (Ditmyer et al., 2010; Splieth et al.,

2016). Historically, research was focused more on biological and dietary influences on the

disease. Recent years have seen interest growing in exploring a broader framework,

2

incorporating psychosocial and environmental influences, along with biological measures.

There remain many gaps in knowledge as to how these wider risk indicators, and possible risk

factors, interrelate, and why some children suffer a greater burden of disease than others.

It has been well researched that maternal education and income have a significant impact on a

child’s oral health. Socio-economic circumstances, mainly income (Schwendicke et al., 2015),

maternal level of education (Kumar et al., 2014), employment status (Tanaka et al., 2013), and

age (Niji et al., 2010), influence children’s susceptibility to, and experience of, dental caries,

as higher income is said to promote improved living conditions and reception of health care

(Capurro et al., 2015; Jepsen et al., 2017). Despite changing roles and responsibilities within

the family (Poutanen et al., 2007), the mother is still recognised as the key influencer of a

child’s eating and oral hygiene habits, and thus caries status, more than others (Saied-Moallemi

et al., 2008). Consequently, maternal oral health-related knowledge, beliefs, and attitudes

influence the oral health behaviour of their children (Poutanen et al., 2007; Saied-Moallemi et

al.; 2008, Szatko et al., 2004). Arguably, family circumstances reflect their socio-economic

status (SES); families provide support and role modelling to children so that their oral health

is influenced by their caregivers’ behaviours and perceptions. For example, single parent

families tend to be stressed, including financially, so that the principal role model may not be

setting a good example regarding health behaviours, with impact on the dental caries status of

the children (Hooley et al., 2012; Östberg et al., 2016; dos Santos Pinto et al., 2016). Given

that dental caries is a lifestyle disease and shares risk indicators with several other chronic

conditions like obesity, maternal body mass index could be expected to predict dental caries of

children to some degree (Wigen and Wang, 2011), especially because of the influence mothers

have on their children’s eating habits (Wigen and Wang, 2014). Thus, high levels of untreated

caries in mothers increased the odds of their children having untreated caries (Ersin et al., 2005;

3

Warren et al., 2016; Birungi et al., 2016) and were associated with more severe disease in the

children (Weintraub et al., 2010). Poor maternal oral health was a predictor of their children’s

caries experience both in childhood and adulthood (Shearer et al., 2011). Additionally,

mother’s oral carriage of cariogenic bacteria, mutans Streptococci (MS) and Lactobacilli (LB),

are associated with early oral colonisation of their children with these microorganisms and has

been used as a predictor of increased childhood caries occurrence (Chaffee et al., 2014). As

mentioned above, an individual’s risk for initiation of a carious lesion lies in an acidic

environment at tooth surfaces, (Kianoush et al., 2014, Varma et al., 2008), perhaps with low

flow rates of saliva (Diaz de Guillory et al., 2014), which may have poor buffering capacity

(Gao et al., 2016), and be reflected in low hydration of the mouth (Varma et al., 2008). The

homoeostasis of the oral environment is thus critical, and little attempt has been made to

explore maternal saliva characteristics and any association they might have with their child’s

oral environment and levels of dental disease. That is not to imply a direct effect on the child:

rather if the mother’s markers of caries risk in her saliva are high, it is likely that some of the

conditions which have produced this situation will also apply to her child.

It is evident that the prenatal circumstances of a child, which is strongly influenced by maternal

health and nutrition, will affect the development of children’s teeth because the deciduous

dentition is formed throughout most of the foetal period (Billings et al., 2004). Especially,

maternal intake of calcium, iron, folate, multivitamins, vitamin D, vitamin C, and fluoride

affect the development and mineralisation of their baby’s dentition (Tanaka et al., 2015;

Takahashi et al., 2015; Schroth et al., 2014). Vitamin and mineral deficiencies during

pregnancy are frequently accompanied by recurrent infections and have an adverse effect on

the daily functioning and well-being of both mother and foetus (De-Regil et al., 2016). It is of

interest, therefore, to understand the intra-uterine environment and to explore if any

4

abnormalities therein are related in any way to the caries experience of their offspring.

Moreover, low birth weight is a marker of foetal growth and has been linked to a variety of

chronic diseases in later life (Barker et al., 2002; Barker and Thornburg, 2013). Likewise,

compromised foetal nutrition and growth may affect the development of deciduous teeth, which

starts in utero. Low birth weight has especially been linked with enamel hypoplasia (Nelson et

al., 2013) and thereby with increased levels of caries (Bernabé et al., 2017).

As a child ages, he/she is exposed to many risk conditions. The mores of the wider community

and family influence the risk of developing caries, and risks will be reflected in intra-oral

conditions which play a direct role in the disease process. The physicochemical properties of a

child’s saliva, pH, buffering capacity, and flow rate play a major role in determining the levels

of MS and LB in saliva (Ramamurthy et al., 2014) and therefore in the susceptibility to caries

(Preethi et al., 2010). However, baseline (past) caries was the strongest single predictor of

accumulated carious lesions in children throughout the first one or two decades of their lives,

post-partum (Attaran et al., 2016). Thus caries experience in the deciduous dentition has been

found to predict caries experience in the permanent dentition (Casanova-Rosado et al., 2005).

Despite growing, detailed knowledge of the biochemical and biophysical mechanisms of the

caries process, and increasing attention paid to behavioural, social, and wider environmental

factors, dental caries persists as the main chronic disease among children worldwide

(Kassebaum et al., 2015). To this effect, The National Institute of Health Consensus

Development Program (USA) released a statement in 2001, specifying that there is a great

need for studies to identify genetic drivers, or at least genetic markers, which could be related

to diagnosis, prognosis, and therapy for dental caries

(http://consensus.nih.gov/2001/2001DentalCaries115html.htm). Even though the impact of

5

the human genome on dental caries has been researched for decades through twin (Boraas et

al., 1988), family (Bretz et al., 2005), genome-wide sssociation studies (GWAS; Vieira et al.,

2008), and candidate gene studies (Vieira et al., 2014), no potent effect variants have been

identified, indicating that factors other than the genome itself may have an impact on the

development of the disease: the epigenome may be one such. The epigenetic code, which is a

subject’s genome modified by environmental stimuli, determines when and where in the body

genes are expressed. Along with many other diseases, understanding the epigenome is

increasingly relevant to research in cariology (Williams et al., 2014; Seo et al., 2015).

1.2 Rationale for conducting the research in South East Queensland

Levels of dental caries in children have risen in Australia in recent years. From 1977 to 1995,

data from school dental services suggested that there was a drop in dental decay in deciduous

teeth (AIHW, 2014). However, by the year 2002, just over 47% of 5- to 6-year-old children

had one or more teeth with carious cavities measured with dmft index and, of these lesions,

80% were untreated. Out of all states and territories, Queensland (QLD) children aged six years

had the second highest caries scores (dmft 2.28) (Armfield et al., 2007). Moreover, by 2010,

55% of 6-year-old Australian children had experienced decay in their deciduous teeth

(Chrisopoulos et al., 2016). In 2010, out of the children who visited school dental services,

48% of 5-year-olds with caries had a mean decayed, missing or filled teeth index of 2.32

(AIHW, 2014). Oral health surveillance data from 2004-2006 revealed that there is a substantial

variation in children’s oral health across Australian states and territories (ARCPOH, 2009,

2014). With New South Wales as the reference state, QLD children who were 6-7 years old

had the highest caries experience (dmft 2.17) (Lucas et al., 2011,; Plonka et al., 2013). Mean

dmft scores of children varied between geographical regions in QLD: Among 5- to 6-year-olds,

6

it was 1.9 in Brisbane, 2.0 in the wider South-East, 2.4 in Northern Queensland, but only 1.4

in Townsville, which has had fluoridation of public water supplies since 1965, the only town

to receive this benefit in the State. Dental caries statistics in children, especially in the state of

QLD, show that this disease is a serious public health problem (Wong et al., 2012; Do and

Spencer, 2016).

Further, it has been highlighted that over a period of three years (from January 2008 to August

2010), the majority of emergency cases in children below 18 years who presented at community

dental clinics in Logan Beaudesert region were caries related, compared to other dental

emergencies (trauma and orthodontic related) (Wong et al., 2012). When compared with other

areas of QLD, the South East region has a higher proportion of children living in low-income

households (Do and Spencer, 2014). It was reported that the prevalence of untreated decay in

children from low-income areas such as Logan District (36.9%), was more than that of children

from medium-income areas (27.7%) and high-income areas (19.3%) (Do and Spencer, 2014).

Predictive and precise caries risk assessment allows the clinician to identify problems early

for effective preventive therapies and appropriate treatment planning, according to an

individual child’s level of risk. The literature on predictors of dental caries in children is

overwhelming, with numerous systematic reviews identifying socio-demographic, dietary,

and family-level as important risk indicators of childhood dental caries in different child

populations. Although the importance of understanding risk indicators of the process of dental

caries among Western industrialised countries is widely acknowledged, little attempt has been

made to explore the association between caries and a multitude of factors simultaneously

among the same population of children. Given this research gap, this thesis aims to gain

insights into associations between wider socio-economic, environmental, and family

7

variables, especially related to mothers of the children under study, and their child’s caries

history and current activity. We consider the intra-uterine environment of the child from

information on mothers during pregnancy. We then study the general health and oral

physiology of the children and search for epigenetic modifications which might be related to

the cariogenic process (Figure 1.1).

Figure 1.1: Community, maternal, child, and epigenetic influences on dental caries in children

1.3 Research questions and objectives

Based on the research gap identified in the literature on associations between children’s dental

caries and risk indicators, the main research theme of this thesis is centred on three main

research questions.

Wider socio-economic environment

Dental caries

Children’s in-utero environment

Epigenetic modifications

Maternal factors

Children’s intra oral Environment

8

1. Is there a relationship between maternal and environmental risk indicators, SES, mother’s

body mass index, intra oral characteristics, oral health knowledge, behaviours, and the caries

experience of their children?

2. What are the associations between children’s past dental caries experience, salivary

characteristics, prenatal conditions, birth weight, and both lifetime experience of dental caries

and current active dental caries?

3. Are there any differences in epigenetic modifications between individuals discordant for

lifetime, cumulative caries experience?

Answering these questions based on the caries status of a population of mother-child dyads in

South East Queensland (SEQ) is expected to fill the research gap identified. Specific objectives

of the thesis are to:

measure the level of dental caries in a sample of 6- to 7-year-old children and their mothers;

assess the oral health knowledge and behaviours of mothers of 6- to 7-year-old children in

the study sample;

develop risk factor models based on a proposed conceptual framework for dental caries in

a sample of 6- to 7-year-old children inclusive of maternal, environmental and children’s

individual characteristics as markers of risk; and

conduct a pilot study to screen for epigenetic modifications which might, conceivably, have

effects on cariogenesis.

9

1.4 Contribution of the study

The thesis makes two significant contributions to the literature around dental caries in general,

and childhood dental caries in SEQ in particular.

First, the thesis endeavours to narrow the gap in the existing literature on risk markers for

childhood dental caries in a typical Western industrialised population. Although there are

plenty of studies which document possible risk indicators for dental caries among children of

such populations, the amount of research which seeks to identify and quantify such a wide

range of risk factors, including epigenetic influences, is limited. This research explores the

impact of maternal, environmental, and children’s factors as potential risk indicators for

childhood dental caries. Possible interactions between variables were analysed to arrive at the

most significant correlations, and from these to fit hypothetical models of risk indicators. The

salient feature of these models is that they allow researchers to infer the findings in clinically

and biologically meaningful ways.

Second, the thesis adds new empirical findings to the literature on epigenetics and dental caries.

It examines the associations of epigenetic modifications, measured as differential methylations

in DNA, and lifetime experience of dental caries. In addition, new techniques are used to

sequence DNA with bisulfite treated DNA for methylation analysis on the SeqCap Epi 4 M

CpGIANT enrichment platform (Roche NimbleGen, USA). It examines, on a limited number

of dyads, as a pilot experiment, the differentially methylated CpG sites between individuals

with high and low caries experience. At the date of submission, this study may be the first of

its kind to assess epigenetic variations with regards to dental caries experience. Preliminary

10

experiments on deep sequencing of putative genetic loci associated with caries susceptibility

are described.

1.5 The structure of the thesis

The remainder of the thesis is organised into eight chapters. Chapter 2 provides an overview

of current understanding of the aetiopathogenesis of dental caries in otherwise healthy children,

on current Australian and global epidemiology, and on quantification of risk indicators for

dental caries in children. This chapter lays the foundation for the thesis and highlights the main

focus of the thesis.

Chapter 3 is presented as a published paper entitled, “Protocol for assessing maternal,

environmental and epigenetic risk indicators for dental caries in children”. It describes the

study design and methodological approach. Sample selection, sample size calculation, data

collection methods and proposed analyses are described in detail.

Chapter 4 is a results chapter and is presented as a submitted paper entitled, “Maternal caries

experience, household income, carbonated drinks and age at commencement of tooth-brushing

are associated with caries experience among 6- to 7-year-old children in Australia”. This

chapter gives the findings on risk indicators for dental caries in children concerning children’s

socio-economic environment and maternal characteristics, including behavioural and intra oral

attributes, and oral health knowledge.

Chapter 5 is a results chapter and is presented as a submitted paper entitled, “Mothers who

report taking iron supplements during pregnancy and child’s oral carriage of mutans

11

Streptococci are associated with a child’s lower and higher lifetime dental caries experience,

respectively.” The chapter explores the risk markers for children’s current caries experience,

measured with International Caries Detection and Assessment System (ICDAS > 0 at time of

examination), focusing on the child as an individual. Child’s prenatal environment and birth

weight, saliva physiology, and past caries experience (measured by the number of tooth

surfaces with restorations) are considered.

Chapter 6 provides an overview of the genetic predisposition to dental caries and explores what

lies beyond the current knowledge on genetics. It discusses the possible relationships between

caries and epigenetics, and it highlights the lack of empirical research in the area. Further, the

chapter provides the foundation for the next results chapter.

Chapter 7 is a results chapter presented as a submitted paper entitled, “An epigenome wide

association study of mother-child dyads discordant for dental caries: A Pilot Study”. It presents

the findings of a pilot study exploring epigenetic changes, detected in differential methylations

in DNA, between mother-child dyads with high and with low lifetime caries experience.

Chapter 8 is a regular thesis chapter. It concludes the study with an overview of the research,

focusing on research questions and objectives. The main findings regarding risk markers for

dental caries in a population of SEQ children are then discussed. Following this, the chapter

highlights the limitations of the study, future research directions and concluding remarks.

12

Chapter 2: Review of literature

2.1 Introduction

The purpose of this chapter is to examine the literature on current understanding of the

aetiopathogenesis of dental caries in children. The chapter also presents a brief description of

conceptual frameworks that have been proposed as means of understanding the risk indicators

for the process of childhood dental caries.

For this narrative review, PubMed via Medline and Google Scholar were used as data bases.

Key words used were children, caries, dental caries, risk, susceptibility, influence, maternal,

environmental, familial, individual, gender and caries history. Additional Boolean commands

were used to narrow down the search. Further the search was limited to full text journal articles

published between 1912/01/01 and 2017/08/01 on humans. Mesh terms for keywords were

used during the literature search in PubMed.

The chapter organisation is as follows. Section 2.2 outlines the burden of dental caries among

children over the last few decades. Section 2.3 briefly discusses the conceptual frameworks

proposed by researchers to identify risk indicators for dental caries. Section 2.4 reviews the

literature on determinants of dental caries in children from community level influences to

family level and child-level influences. Section 2.5 summarises the chapter with concluding

remarks.

13

2.2. The burden of dental caries in children

Since oral health is merely a part of the general wellbeing of individuals, compartmentalisation

of the mouth and the rest of the body regarding health conditions is not justified. Oral health

affects children’s general health by causing pain, suffering, and changing their dietary habits,

speech and quality of life (Sanders et al., 2009). Dental caries manifests as a continuum of

disease states of increasing severity and tooth destruction, ranging from subclinical changes to

lesions with dentinal involvement (Kidd and Fejerskov, 2004), and it is the fourth-most

expensive chronic disease to treat according to the World Health Organization (WHO)

(Petersen, 2008). If left untreated, caries augments oral infection (Selwitz et al., 2007) and

affect children’s school attendance and performance (Jackson et al., 2011). In 2010, untreated

caries in deciduous teeth was the 10th most prevalent condition affecting 9% of the global child

population (Kassebaum et al., 2015). In Asia Pacific high-income countries, the prevalence of

deciduous caries declined slightly from 10.8% in 1990 to 9.6% in 2010 (Kassebaum et al.,

2015). In Australasia, the decrease was from 5.5% to 4.9%, whereas in Europe (Central,

Eastern, and Western) the prevalence did not show a marked difference from 1990 to 2010

(Kassebaum et al., 2015). These data show that, worldwide, dental caries in the deciduous

dentition is not under control, in spite of our detailed knowledge of aetiopathogenesis. We

know how to prevent most caries in most individuals, but fail to apply these approaches at a

population level.

Decay of deciduous teeth (measured with decayed, missing, and filled teeth index; dmft, with

decay recorded at the level of cavitation) in children aged 6 attending school dental services in

Australia decreased from a mean dmft of 3.19 in 1978 to 1.45 in 1996, followed by a gradual

increase to a mean of 2.58 in 2010. According to National Child Oral Health Study in Australia

14

(2012-2014), one in every four children aged 5-to 10-years at that time had untreated decay in

their deciduous teeth (Do and Spencer, 2016). In 2013, approximately two-thirds (64.4%) of

children aged 5 and over had made a dental visit. Almost 4 in 5 (78.8%) children aged 5-14

had made a dental visit in the previous 12 months, with 90.6% having visited within the past

two years, although the nature of the visits were not explained (Chrisopoulos et al. 2016).

Furthermore, Chrisopoulos et al. (2016) report that mean dmft of Australian children in the

year 2015 was 1.83 for 5-year-olds, 1.74 for 6-year-olds and 2.63 for 8-year-olds (Chrisopoulos

et al., 2016). However, oral health surveillance data indicate significant variations in children’s

oral health and dental service attendance across Australian states and territories (ARCPOH,

2009; AIWH, 2014). In 2007, 5- to 6-year-old QLD children had the highest caries mean dmft

of 1.72 (Ha et al., 2011). Lucas et al. (2011) made the same observation in 2011 and report that

caries experience of 6- to 7-year-old children in QLD was the highest: mean dmft score 2.17,

followed by 1.68 in Northern Territory, and 1.53 in Western Australia. Dental caries is clearly

a serious public health problem, especially in QLD (Wong et al., 2012). Even within the state,

areas of high social disadvantage such as the Logan-Beaudesert district have the highest caries

prevalence among children (Plonka et al., 2013). In SEQ, dental caries was the number one

reason among children for attending hospital emergencies: in 2009 it accounted for 75% of all

emergency attendances and in 2010, 74% (Wong et al., 2012). In 2009, restoration of carious

teeth represented 26% of all treatment provided per month, and 20% in 2010. There was an

overall rise in the number of paediatric emergency visits in SEQ from 2008 to 2010, and the

most common cause was caries-related, for instance, dental pain and abscesses (Wong et al.,

2012). These observations show considerable geographical variations in population oral health,

which exist from a young age.

15

During the past few decades research into population oral health has focused on describing

biological and dietary determinants of disease. In more recent years there has been a growing

interest in exploring children’s oral health outcomes using a broader framework, incorporating

psychosocial, and environmental influences, together with biological (Crall et al., 1990) and

genetic (Boraas et al., 1988) variables. The effort towards a more comprehensive approach

stems partly from the recent shift in thinking about population health, in which academic

reports have proposed increasingly complex conceptualizations of oral health determinants.

Studies assessing risk factors for dental caries in children have reported different results, due

to variations in study designs, analytical methods, subjects’ ages, and definitions of risk factors

and caries phenotypes (Chankanka et al., 2011).

From a public health perspective, exploring consistent caries-related factors would be essential

for the prevention of the disease. However, the values of generalising such indicators among

different populations is debatable due to the ever present population diversities. Investigations

that look into associations of these varied characteristics and childhood caries are needed to

understand the disease trajectory, and thereby to reduce the disease burden.

2.3 Proposed Conceptual frameworks describing the process of dental caries

The common risk factor approach (CRFA) for oral diseases has been widely accepted and

endorsed globally by dental researchers and promoters of oral health, prompted to some extent

by policy recommendations from the WHO in the 1980s (Petersen and Kwan, 2010). This

concept was further developed and applied to oral health with an emphasis on directing action

at shared risk factors for chronic diseases including dental caries. Sheiham and Watt in 2000

proposed a conceptual framework addressing the broader shared social determinants of chronic

16

diseases, highlighting that a behavioural preventive approach alone would only widen the oral

health inequalities across populations (Sheiham and Watt, 2000). They placed a key emphasis

on socio-economic and political contexts which generate and maintain educational systems,

labour markets, welfare, and health systems leading to the maintenance of an individual’s

socio-economic position. They argued that this, in turn, would result in changes in behavioural,

biological factors, psychosocial, and health care utilisation leading to oral health inequalities.

However, they do not discuss possible interactions between the different levels, and the

proposed conceptual framework is unidirectional.

A more comprehensive CRFA approach was taken by Petersen in 2005, based on the risk factor

model in the promotion of oral health suggested by the WHO (Petersen, 2003). The design

incorporates evidence available on potential socio-behavioural risks of dental caries and some

widely used outcome measures. Interrelated health system and oral health services,

sociocultural factors, and environmental risk indicators are explained as dynamics that

influence the use of oral health services and risk behaviours of individuals, families, or

populations. The outcome; prevalence, and severity of dental caries, is described as a result of

these proximal factors (Petersen, 2005). Moreover, the model is based on empirical evidence

from both developed and developing countries. However, biological and physiological

characteristics have not been included in the model.

A unifying conceptual model proposed by Seow (2012) addresses broader social,

environmental, maternal, and child factors that are commonly associated with childhood dental

caries. This model is centred on the mother, coming from a socially disadvantaged background,

low SES, and a relatively low-level of education. The author describes in detail how the

external environment could affect maternal economic stress, dysfunctional parenting, health

17

beliefs, and behaviours which eventually lead children to a high risk of caries (Kim Seow,

2012). It also highlights possible interactions between the wider environment and the mother’s

characteristics. However, the model lacks a multi-level approach as child’s characteristics,

biological, and physiological risk factors have not been considered. According to the

conceptual framework proposed by Johnson in the early 1990s, dental caries is described as a

disease process that depends on a continually changing balance between external stimuli and

the host’s biological response to such stimuli. These stimuli act in an environment conditioned

by a range of local and general predisposing factors, and a range of local and general resistance

factors. The initiation of dental caries itself, as outlined in the model, is a relatively simple

series of physicochemical interactions at the tooth surface (Johnson, 1991). The balance

between demineralisation and remineralisation, which varies over time, is determined by

interactions amongst this wide range of predisposing and resistance factors.

A comprehensive model of children’s oral health, considering the wider social environment,

immediate family environment, children’s characteristics, and their possible interactions was

proposed by Fisher-owens, in 2007 (Figure 2.1) (Fisher-Owens et al., 2007). This conceptual

model of oral health is based on a solid foundation of social science and epidemiologic studies

of population health. The particular factors chosen to be included in the model were based on

research identifying key influences on children’s oral health, including genetic and biological,

social and physical environment, support system, health behaviours, and dental and medical

care. This conceptual model includes a broad range of genetic, social, and environmental risk

factors and appropriate multiple levels of influences, including a time or developmental

dimension.

18

Figure 2.1. Child, family and community influences on oral health outcome of children (Source: Fisher-Owens et al., 2007; Copy rights license

Number 4204590201918 https://s100.copyright.com/CustomerAdmin/PLF.jsp?ref=05f7487f-ec06-4b22-a903-9511ee5d81b5 )

19

A conceptual model directs researchers towards recognising risk indicators for disease and the

model’s greatest value would be when it can be used empirically, even though the relative

importance of each influence will vary between different populations. With the evolution of

social contexts and human behaviours, new risk markers and risk indicators evolve. Hence,

researchers need to be constantly aware of emerging trends. New analytical approaches guided

by the conceptual models are needed to reach the next stage of public health policy in order to

combat still high levels of dental caries.

2.4 Determinants of dental caries

The answer to the question ‘What causes dental caries in children?’ is an important, and

complex one. It concerns those in both developed and developing countries where many

children experience the adverse consequences of the disease. Understanding the aetiology and

pathogenesis of the disease would have a direct influence on the most appropriate preventive

and treatment interventions. They may include past caries experience, information about socio-

demography, socio-economy, oral hygiene, dietary habits, oral bacteria, and saliva

characteristics (Table 2.1). These comparatively objective parameters are often accompanied

by a subjective assessment of risk level. Children are especially vulnerable since they are

exposed to risk indicators for dental caries, notably high sugar-containing foods and treats,

provided by well-meaning adults and poor oral hygiene practices (Seow et al., 2009).

According to the conceptual frameworks described, dental caries is a social, cultural, and

behavioural condition that has a genetic interplay within the individual.

20

2.4.1 Maternal and environmental determinants

2.4.1.1 Socio-economic status

Socio-economic circumstances early in life help determine future health outcomes such as

dental caries. Low SES and neighbourhood poverty are significantly associated with a greater

risk of caries, unmet dental care needs, and poor oral health-related quality of life (OHRQOL)

(da Fonseca and Avenetti 2017). According to Vargas (1998), SES is a complex construct that

is operationally defined in many ways (Vargas, 1998). Most of the dental studies use parents’

education, income, and occupation as associated factors of SES (Nicolau et al., 2005). In

general, there is an inverse relationship between caries and SES: a high prevalence of childhood

caries has been shown to be related to low family income (dos Santos Junior et al., 2014).

Higher income promotes healthy living, better access to dental health care, and improved

conditions of life, although the reverse is often seen in developing countries (Fisher-Owens et

al., 2007). Aida et al., (2017) from their longitudinal survey of “Babies in the 21st Century”,

reported that in Japanese preschool children the rate of caries treatment at the age of

approximately 3 years was <10% for all SES groups and this increased to more than 30% at

around age 6. Children from lower SES received more frequent caries treatment compared to

their affluent counterparts (Aida et al., 2017). Similar observations were made in the USA

among children from low-income families (Ismail et al., 2008). Nevertheless, in rural settings

of developing countries, where traditional diets have low cariogenicity, it is the wealthier

classes, often urban, who have access to high sugar diets and hence higher caries experience

among children (Petersen, 2005). Not all studies agree: Chaffee et al. reports that dental caries

associated lower oral health-related-quality-of-life scores were similar across all social groups

among a population of Brazilian children (Chaffee et al., 2017). Moreover, Carvalho et al.,

21

(2014) did not find a significant association between caries development and household

monthly income in Brazil (Carvalho et al., 2014).

2.4.1.2 Family structure and environment

Family structure and stability are paramount for children to develop well and for the

establishment of healthy oral habits. Children in a family that experiences instability, for

instance, loss of resources, frequent moves, violence, child support issues, divorce,

incarceration, or maternal depression tends to have poor health behaviours (da Fonseca and

Avenetti, 2017; Piva et al., 2017). Parents play a pivotal role in the establishment of oral health

promoting behaviours that prevent the development of childhood dental caries (Hooley et al.,

2012). Mattila et al. (2000) reported that children in single-parent households are more likely

to have advanced levels of caries and have more periodic use of dental services (Mattila et al.,

2000), probably due to the lack of resources and social support being distributed among all

family members. Family structure and household crowding also have a significant impact on

children’s caries status, with single children showing a lower caries experience and a lower

impact of oral health on their quality of life (Kumar et al., 2016; Moimaz et al., 2014). Further,

lack of help in raising children is considered as a risk indicator for dental caries in children

since it can be a barrier for seeking oral health care for working mothers (Kim Seow, 2012).

Children from poorer functioning families on the dimensions responsiveness, communication,

organisation, and social network had higher levels of dental decay than children from normal

functioning families. Furthermore, they were also more likely to engage in less favourable oral

hygiene behaviours (Duijster et al., 2014). Reynolds et al. (2015) however, disagree that family

composition was not significantly associated with the oral health status of children. They report

that neighbourhood social capital plays a more prominent role in childhood dental caries than

22

the individual family structure (Reynolds et al., 2015). Nevertheless, studies linking the broader

home environment and family functioning to childhood dental caries are limited.

2.4.1.3 Maternal level of education, oral health knowledge and behaviours

When it comes to child rearing, the mother still plays the major role (Kavanaugh et al., 2006).

A systematic review reported that more studies are demonstrating a significant effect with a

child’s dental caries and mother’s level of education than that of the father’s (Hooley et al.,

2012). Maternal education level is an important indicator that reflects knowledge and skills for

making healthy behaviour choices (Laaksonen et al., 2005) For example, highly educated

mothers report more positive attitudes and stronger intentions to control children’s sugar intake

than low-educated mothers (Warren et al., 2016). In populations where the majority of people

have a low level of education, caries risk behaviours like frequent consumption of fermentable

carbohydrates and sweetened low pH beverages, and poor oral hygiene habits are more

common (Ashkanani and Al-Sane, 2012). Sweet food intake has a clear causal relationship

with dental caries, and maternal education has been reported as a risk indicator that affects

sweet food intake of children (Ju et al., 2016). Despite mothers’ high-level oral health

knowledge, perceptions of self and child oral health would ultimately determine the oral health

status of the offspring (Heaton et al., 2017). Research evidence suggests that the mother's

positive attitudes towards good oral health have an influence on her children's brushing habits,

diet, and oral diseases (Saied-Moallemi et al., 2008). Nevertheless, a study conducted in

Kuwait reported that maternal oral health knowledge was weakly correlated with practice,

suggesting that changing oral health-related behaviours takes more than just improving

knowledge (Ashkanani and Al-Sane, 2012). An added factor could probably be a child’s

temperament and the self-efficacy of the mother. A study on a population of Hispanic mothers

and children revealed that child’s resistance towards tooth brushing and lack of time in

23

supervising the children during brushing were some reasons for poor oral hygiene for the

children (Tiwari et al., 2017). It also has been revealed that negative child temperaments can

exacerbate parental stress and worsen parental feeding and oral health practices (Tiwari et al.,

2017). It also has been reported that negative child temperaments can exacerbate parental stress

and worsen parental feeding and oral health practices (Tiwari et al., 2017).

2.4.1.4 Maternal body mass index

Maternal body mass index (BMI) is an indication of health and lifestyle habits, such as diet,

physical activity, and attitudes to health and well-being, which ultimately influence the children

and the establishment of their oral health behaviours (Wigen and Wang, 2011). Children’s

dietary intake has been shown to be strongly associated with their mothers’ dietary intake, and

the children’s dietary habits are developed early in life (Oliveria et al., 1992). When exploring

the association between maternal diet and caries development in children, the results showed

that having a mother who consumed more sugar and fat than recommended, was associated

with caries development in children. Moreover, a mother with BMI classified as obese was

associated with caries development in their children before 5 years of age (Wigen and Wang,

2014). The association of BMI with dental caries is not necessarily causal and is complicated

by many intervening variables (Silva et al., 2013). For instance, dietary habits and income are

considered important factors for both dental caries and obesity. Dietary habits related to obesity

may provide a higher quantity and frequency of sucrose intake, which are important factors in

caries aetiology (Carvalho Sales-Peres et al., 2010). However, a study done in India reports

that maternal BMI is not significantly associated with dental caries in children as the

associations of BMI with dental caries varied across SES categories (Kumar et al., 2017).

24

2.4.1.5 Maternal caries

As far back as 1946, it was found that there can be a significant increase in dental caries among

children whose parents had untreated caries (Klein, 1946). The relationship between maternal

caries levels and children's caries experience has been found to be stronger than that of their

fathers (Weintraub et al., 2010). Mothers caries status has been related to preschool children’s

caries in Turkey (Ersin et al., 2005), Thailand (Thitasomakul et al., 2009), and the USA (Smith

et al., 2002). However, in a Japanese population of pre-schoolers, maternal caries experience

was not a risk indicator (Kawashita et al., 2009). Nevertheless, Moyer (2014) proposed that as

a prevention strategy for dental caries in American children, mothers caries levels should be

assessed as a risk factor (Moyer, 2014). Proença et al. (2015) reported that Brazilian children

who had mothers with increased levels of dental caries were more at risk of developing the

disease themselves, and that the family environment and maternal attitudes exert a significant

influence on the child and can affect children’s behaviour, thus reflecting on oral health

(Proença et al., 2015). Since mothers play a vital role in rearing children, certain maternal

characteristics expose the child to adverse environmental conditions that encourage caries

development and progression in children. For instance, frequency of dental visit, maternal

sugar consumption, oral hygiene practices, maternal attitudes towards oral health (Kumar et

al., 2016), and genetic predisposition (Boraas et al., 1988). This is evident by the conceptual

framework proposed by Fisher-Owens, as dental caries being a multi-level, multifactorial

disease having interactions within the oral cavity to include child, mother, and community-

level influences that vary over time (Fisher-Owens et al., 2007).

25

2.4.1.6 Maternal cariogenic bacteria

Other than the impact of unhealthy practices of caregivers, maternal-child transmission of oral

bacteria is one such potential mechanism, given the essential, albeit not sufficient, role of oral

infection in caries development. Despite conflicting evidence that has challenged the accepted

concept of vertical transmission as the mode of initial colonisation with MS, the mother as a

source is logical, due to closest contact when initial colonisation occurs (Douglass et al., 2008).

During and shortly after birth, the epithelial surfaces in the oral cavity of infants become

colonised by numerous bacterial species. This cross-establishment is usually from mother to

child, particularly when the mother has active caries and high levels of cariogenic bacteria in

their mouths (Hooley et al., 2012). Childers et al. (2017) report that the fidelity of MS genotype

between mother and child through vertical transmission was 58%, where, 40 out of 69 African

American mother-child dyads who participated in the study shared the same MS genotype

(Childers et al., 2017). Moreover, significant reductions in the incidence of MS carriage and

caries experience were observed in children whose mothers received preventive and restorative

interventions aimed at reducing the count of these organisms in their mouths before and shortly

after the birth of a child (Brambilla et al., 1998; Köhler and Andreen, 1994). Nonetheless, MS

(i.e., MS and Streptococcus sobrinus) have been implicated as the principal etiologic agent

involved in the initiation of human dental caries (Gross et al., 2010) and LB species as primary

late colonizers that are associated with advanced cavitation (Dige et al., 2014), both of which

could be taken as risk indicators for childhood dental caries.

26

2.4.1.7 Maternal salivary physiology

The interplay between microorganisms, diet, and host susceptibility determines whether dental

caries will occur. As teeth are constantly bathed in saliva, the constituents and properties of

this oral fluid play an essential role in the occurrence and progression of dental caries. Saliva

is believed to be one of the most important host factors and a key mediator controlling the

speed and direction of the cariogenic pathway (Fejerskov and Kidd, 2008). Saliva may protect

teeth through several mechanisms, such as clearance of food debris and sugar, aggregation and

elimination of microorganisms, buffering actions to neutralise acid, and maintaining

supersaturation on tooth mineral (Dawes, 2008). Salivary flow rate is the most important caries

preventive function of saliva for its “flushing/oral clearance” capacity by some researchers

(Lagerlöf, 1994), and the higher the flow rate, the faster the clearance and the more protection

for teeth from dental caries (Lagerlöf, 1994). However, low salivary flow rates (< 0.8 – 1.0

ml/min stimulated whole saliva) are found in pathological conditions and have reduced

sensitivity as a predictor for caries (Lenander-Lumikari and Loimaranta, 2000; Leone and

Oppenheim, 2001). Nevertheless, some research evidence suggests that there is no correlation

with caries process and salivary flow rate (Russell et al., 1991; Varma et al., 2008) while others

site salivary flow rate as a potential risk marker for dental caries (Gao et al., 2016). These

results cannot be generalised as dental caries phenotypes and methods used to measure flow

rate may vary within studies.

The carbonic acid/bicarbonate buffer is the primary buffer in stimulated saliva, with

bicarbonate acting mainly to neutralise acids produced by bacteria when they digest sugars in

the mouth or acids from the stomach (Lenander-Lumikari and Loimaranta, 2000). The pH of

the oral cavity is maintained at 6.3 to ensure the maintenance of the integrity of the tooth

structure with the acid neutralising capacity of the salivary bicarbonate buffering system

27

(Bardow et al., 2000). However, a recent study done in the USA among individuals with cystic

fibrosis gave conflicting results. It was observed that there was no significant association

between salivary flow rate or buffering capacity and caries prevalence among the participants.

However, the results cannot be generalised to the general population as the sample size was

small with n = 83 cystic fibrosis patients (Alkhateeb et al., 2017).

The pH of saliva is modulated by salivary bicarbonates, phosphates, and urea (Humphrey and

Williamson, 2001) maintaining the saliva pH between 6 and 7. The pH of saliva measured

directly from the ducts of individual salivary glands varies between 5.3 at low flow rates to 7.8

at maximum flow rates (Bardow et al., 2004; Humphrey and Williamson, 2001). A drop in pH

below 5.5 means that such acidic saliva has harmful effects on enamel and dentine (Aframian

et al., 2006; Bardow et al., 2004). There is sparse literature on the association between the pH

of resting saliva and dental caries (Varma et al., 2008), for it is plaque pH that contributes most

for the demineralization of the tooth surface and ultimately to the development of dental caries

(Lenander-Lumikari and Loimaranta, 2000). A dissimilar result was observed by Dos Santos

et al. in 2015 in a group of children with sialorrhea. Onabotulinum toxin given to control

sialorrhea reduces the salivary flow rate and alters salivary pH. The level of acidity, measured

by mean pH, among the group remained within neutrality range, and had not caused

demineralization of enamel surface. However, they observed a significant and positive

association with caries, stating that poor oral hygiene and cariogenic diet could have

contributed to an increase in caries activity even though the saliva pH remained neutral (Ferraz

dos Santos et al., 2016).

28

There is no published literature that explores the association of buffering capacity of saliva in

mothers, pH, and flow rate as risk indicators for children’s dental caries; hence, an investigation

is timely and would address the existing research gap.

2.4.2 Influence of children’s individual factors

2.4.2.1 Intrauterine environment of child and birth weight

The foetal environment is considered as a crucial indicator in the causality of diseases later in

life (Barker and Thornburg, 2013). Many studies are supporting the hypothesis that adult life

can be ‘modelled’ during intrauterine life, implying that intrauterine nutritional deficiencies

could be associated with adult diseases such as hypertension, diabetes, and obesity (De Boo

and Harding, 2006). Intrauterine growth is taken as an independent predictor of diseases in

childhood and throughout life (Barker, 1998; Cameron et al., 2012). Likewise, deciduous tooth

formation and mineralisation start during the foetal period. Hence the intrauterine environment,

which is affected by factors such as smoking and maternal nutrition during pregnancy, might

influence the development, formation, and mineralisation of children’s teeth (Winter, 1976).

Reported maternal intake of prenatal supplements by mothers has been associated with foetal

nutrition, lack of which linking to the development of conditions like autism (Schmidt et al.,

2011). Therefore, supplementation use is usually recommended during pregnancy to meet the

nutrition needs of both mother and foetus (Scholl, 2005). Maternal vitamin D deficiency in

pregnancy is reported to predispose children to developmental dental defects (specifically

hypermineralized enamel) since during prenatal development, vitamin D reaches the fetus only

through the placenta (Riggs et al., 2016). Reed et al. (2017) reported that children whose

mothers did not take vitamin D supplements during pregnancy were more at risk of developing

enamel hypoplasia (Reed et al., 2017). Hypoplastic enamel is more susceptible to colonisation

by cariogenic bacteria and is often hypersensitive, making adequate oral hygiene difficult and

29

hence increasing the risk of the child developing caries (Reed et al., 2017). Moreover, Bergel

et al., in 2010 showed that children whose mothers received calcium supplementation when

pregnant showed a significant reduction in dental caries compared to their corresponding

counterparts (Bergel et al., 2010). Research done with animal models have shown that vitamin

A deficiency during foetal life would result in failure of ameloblastic activity, which may lead

to enamel agenesis and hypoplasia (Armstrong, 1948). There is a dearth of literature on humans

that investigates the association between reported maternal prenatal supplement intake and

dental caries status of the mother’s offspring, hence the need to further investigations.

With, recently, attention shifting to early life factors for dental caries, the role of birth weight

is far from settled. Low birth weight, a marker of foetal growth, has been linked to a variety of

chronic diseases in later life (Barker and Thornburg, 2013). Even though birthweight was not

associated with baseline dental caries experience in a population of Scottish children, low birth

weight put them on a trajectory of increased dental caries over a period of 4 years (Bernabé et

al., 2017). There is evidence suggesting that infants with very low birth weight have an

increased risk for enamel hypoplasia and hence are placed at high risk of developing dental

caries (Nelson et al., 2013). These findings support the view that the prenatal and postnatal

environments are important for children’s dental status and also underscore the advantages of

focusing on accumulation of disease over time

2.4.2.2 Age and gender

One of the factors that are currently associated with dental caries in children is the lifetime of

dentition. Older aged children were more likely to have caries than their younger counterparts.

This difference is liable to be because caries experience measures the cumulative effects of

dental caries in the life time of a dentition (Mwakayoka et al., 2017). In a population of Nigerian

30

children, the proportion of children with caries increased significantly with increasing age up

to the age of 4 years, although there were other social and oral hygiene determinants associated

with the age (Folayan et al., 2015). A study conducted in China revealed that the prevalence of

caries in three age groups (36-47 months, 48-59 months, and 60-70 months) showed significant

differences, with the oldest group having the highest mean dmft of 5.54 (Li et al., 2011). These

observations are as expected because as age increases children have more teeth, they are

exposed to more cariogenic food and beverages, and if coupled with poor oral hygiene

behaviours are more predisposed to dental caries (Mwakayoka et al., 2017).

Though there are few studies undertaken specifically in children on the association of dental

caries and gender, the existing ones give contrasting evidence. Pecharki et al. (2016) showed

that caries was negatively associated with being male (Pecharki et al., 2016), whereas Declerck

et al. reported that boys were more at risk of developing caries than girls (Declerck et al., 2008).

Moreover, in a Chinese population of children aged 6 to 71 months, females were less likely

to have childhood caries (Li et al., 2011). Nevertheless, a study undertaken among a group of

Indian adolescents reported that females significantly made up most of the high caries cluster

(Warren et al., 2017). In Brazilian children who were aged 9 years, no significant association

could be observed with gender and dental caries experience (Casanova-Rosado et al., 2005).

This observation was also made by Chankanka et al., (2011) in a group of 5-year-old children

in the USA (Chankanka et al., 2011), and by Aida et al., (2008) in Japanese toddlers (Aida et

al., 2008).

2.4.2.3 Past dental caries

According to Attaran et al., (2016) baseline caries was shown as the strongest single predictor

of future caries in children (Attaran et al., 2016). Dental caries is diagnosed typically at the

31

tooth surface level, where both the shape and the depth of any potential lesions are scored on

an ordinal scale representing the stages of the disease process, ranging from sound (no lesion)

to lesions into the dental pulp. These observations enable epidemiological and clinical studies

to describe the intraoral susceptibility of dental caries and predict future risk through Bayesian

latent variable models (Zhang et al., 2011). Based on that, Pak et al. (2015) developed a

Bayesian approach to predict future transition probabilities of dental caries, given a child’s

observed caries life history data. The established model could predict future development of

caries not only for individual teeth but also at the mouth level (Pak et al., 2017). Whilst the

most reliable evidence comes from empirical research, although different population groups

may show unique predictive values (Attaran et al., 2016). Survival analysis revealed that

children without past caries experience in deciduous teeth at baseline remained caries free in

permanent teeth for a longer period than children with previous caries experience at baseline

(Cortellazzi et al., 2013). Piva et al. (2017) reported that dental caries progression in a cohort

of Brazilian pre-schoolers was directly associated with baseline caries (Piva et al., 2017). After

two years follow-up, the severity of baseline caries which included dentine lesions (ICDAS 4,

5 and 6) had almost doubled (Piva et al., 2017). According to Parisotto et al. (2012) even past

non-cavitated lesions (ICDAS 1 and 2) at baseline increased the risk of developing new caries

in a population of 3- to 4-year-old preschool children in Brazil (Parisotto et al., 2012),

indicating the importance of early screening for detection of dental caries.

2.4.2.4 Physicochemical properties of saliva

The saliva circulating in the mouth at any given time is termed as the whole saliva, and it

comprises a mixture of secretions from the major and minor salivary glands and traces from

the gingival crevicular fluid. Evaluating the causative factors in saliva of individual’s at risk to

dental caries can pay the way to make recommendations that will cater specifically to children’s

32

individual needs (Gopinath and Arzreanne, 2006). Salivary buffering capacity and pH have an

inverse relationship as buffering mechanisms in saliva (bicarbonate, phosphate, and protein)

act to neutralise salivary pH (Lenander-Lumikari and Loimaranta, 2000). A study undertaken

among 7- to 14-year-old children from India showed that salivary pH and buffering capacity

were low in a caries group compared to a caries free group (Preethi et al., 2010). However, the

associations were not significant, probably due to factors like microflora, diet and retention of

food might have dominated the buffering capacity and pH to initiate caries. Tulunoglu et al.

(2006) observed contradictory results in their study between caries free and caries active

groups. Salivary pH values were higher in the 7- to 10-year-old girls, in the caries free group

than in the caries-active group, whereas in the 11- to 15-year-old group, pH values were

significantly higher in caries active girls. They stated that individual and environmental

variations could be a major cause of the results. For instance, different foods containing varied

levels of dietary proteins and carbohydrates, can affect the salivary buffering capacity and

hence the observed differences in the sample of children (Tulunoglu et al., 2006). Nevertheless,

there is evidence that larger quantities and faster rates of acid production are present in caries

active children than that in caries-free children (El-kwatehy and Youssef, 2016). Even though

the evidence regarding salivary pH and buffering as a risk marker for dental caries is

inconclusive, it is still considered as a non-invasive and reliable risk assessment method for

caries in children (El-kwatehy and Youssef, 2016; Preethi et al., 2010).

2.4.2.5 Oral cariogenic bacteria

Even though dental caries is multifactorial, the crucial role of microorganisms is well

established in childhood dental caries (Palmer et al., 2010). According to Tanzer et al. (2001),

dental caries is a dieto-bacterial disease resulting from interactions between a susceptible host,

cariogenic bacteria, and cariogenic diets (Tanzer et al., 2001). The primary infection is by MS

33

and LB are found to play a role in the progression of caries (Samaranayake, 2012), and are part

of normal oral flora (Samaranayake, 2012) transmitted from parents or caregivers (Childers et

al., 2017). In an Indian child population (aged 3-5), a significant positive association was found

between salivary MS levels and caries experience and also between LB counts and caries

experience, where salivary loads of MS was the predominant bacterial species in the oral

microbiota (Ramamurthy et al., 2014). El-kwatehy and Youssef (2017) demonstrated that the

number of 6- to 12-year-old Egyptian children with MS or LB ≥ 105 colony forming units/ml

of saliva was significantly higher in caries affected children compared to caries free children

(El-kwatehy and Youssef, 2016). Even though MS and LB have been identified as the main

cariogenic bacteria, there are much more undetected genera in the oral cavity (Wade, 2013).

ElSalhy et al. (2016) demonstrated that there were more than 98 bacterial species in their

sample of 11- to 12-year-old children with carious lesions in Kuwait. Surprisingly, in their

sample of children, microbiota of the caries-affected vs. caries-free children was similar

(ElSalhy et al., 2013). Instead of MS and LB, they have found that the microbiota of the caries-

affected children was associated with higher levels of A. defective, A. meyeri, and A.

odontolyticus compared with caries-free children, and the levels also significantly correlated

with caries severity and the number of lesions (ElSalhy et al., 2013). A possible reason for this

observation could be that all children were initially selected for the study based on high MS

and LB levels in either plaque or saliva. Thus, an association might not have been detected.

Nevertheless, compared to other oral bacteria, MS possess an extraordinary ability to infect and

colonise teeth and promote the development of cariogenic biofilms in the presence of sucrose

(Leme et al., 2006). Early acquisition and colonization of MS (before 3 years of age) has

resulted in higher levels of oral MS and Decayed–Missing–Filed (DMF) index at the age of 19

years (Köhler and Andréen, 2012), indicating the importance of controlling infection by these

bacteria in the oral cavity of young children (Hajishengallis et al., 2017)

34

Table 2.1: A summary of maternal, environmental and individual determinants of childhood dental caries

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Maternal and environmental determinates

Nicolau et al. 2005 13 year old

Brazilian school

children (n=764)

Cross

sectional

Mother’s level of education, Father’s level of

education, Family income

Area of residence, Family structure, Paternal

levels of punishment, Birth order, Tooth

brushing frequency

Sugar consumption, Pattern of dental

attendance, Use of fluoride mouth rinse,

Height

DMFT Being second or later child (p=0.008),

Children from rural areas(OR = 2.74, 95% CI

=1.56–4.82), Mothers with lower levels of

education (OR = 2.10, 95% CI=1.03–4.27),

High levels of paternal punishment(OR

=1.60, 95% CI = 1.02–2.52)

Ismail et al. 2008 1021 African

American

caregiver-child (5-

year - old) dyads

Annual household income, Highest

education level, Caregivers’ relationship to

children, Frequencies of moving, Types of

insurance, Religiosity, Children’s

consumption of sugary foods, Depressive

symptoms of care givers, Parenting stress,

Perceived discrimination

dmfs Intake of soda beverages by children

(p=0.04), Caregivers’ fatalistic oral health

beliefs ( p=0.01) ), Religiosity ( p=0.02)

Carvalho et al.

2014

2,511 Brazilian 1-

to–5-year-olds

Cross

sectional

Mother’s education, Mother’s age, Monthly

family income, Parental practices to child’s

oral health,

Bottle-feeding the child during the night on

demand (p=0.0001), Not assisting the child in

tooth brushing (p=0.0001), Ensuring visit to

the dentist in case of troubles with teeth only

(p = 0.0001), Intake of sugary products 2–4

times daily at nursery (p = 0.026)

dos Santos Junior

et al. 2014

320 Brazilian

preschool children

Cross

sectional

Family income, Adolescent pregnancy

Birth weight, Gestational age, Nutritional

Risk

dmft Family income (p = 0.009), Birth weight (p <

0.001), Nutritional Risk (p < 0.001)

Aida et al. 2017 Japanese children

from 2.5 – years- to

-5.5 -years

(n=35260)

Longitudinal

(3-year

follow up)

SES measured by parental level of education Slope index of

inequality (SII) and

relative index of

inequality (RII) for

caries treatment

SII increased for ages 2.5 and 5.5 (95%

CI=3.16-5.09 and 13.68-17.32) respectively.

RII decreased for two age groups (95% CI,

1.59- 2.11 and 1.46- 1.61 respectively)

35

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Family structure

Mattila et al. 2000 1443 Finnish

mother-child (5 –

year- old) dyads

Longitudinal

(5-year

follow up)

Mother’s tooth brushing, Father’s caries

history, Marital status, Place of dwelling,

Mother’s age at child birth, Sugar

consumption at 18 months of age, Headache

episodes of child

dmft Mother's irregular tooth brushing (OR 2.2;

95% CI 1.4-3.5), Annual occurrence of

several carious teeth in the father (OR 2.6;

95% CI 1.9-3.6), Daily sugar consumption at

the age of 18 months (OR =2.4, 95% Cl=1.4-

4.1), Occurrence of child's headaches (OR

3.7; 95% CI 1.5-8.8), Parents' cohabitation

(OR 3.3; 95% CI 1.5-7.6), Rural domicile

(OR 2.4; 95% CI 1.2-4.5), Mother's young

age (OR 5.0; 95% CI 1.3-19.8).

Duijster et al. 2014 5- to- 6-year-old

Netherlands

children (n=630)

Cross

sectional

Location of dental clinic, Education level of

mother, Family functioning dimensions

Ethnicity, Relationship status of caregiver,

Birth order of the child, Oral hygiene

behaviours,

dmft Sociodemographic characteristics (p <

0.001), Oral hygiene behaviours (p < 0.001),

Family functioning dimensions (P < 0.05)

Moimaz et al.

2014

272 Brazilian

mother-child (1-to -

5 -year -old) dyads

Cross

sectional

Marital status, Number of children,

Education level, Employment, Income,

Frequency of visits to a dentist

Need for dental

treatment (NDT=

decayed teeth)

Maternal education (p<0.0001), Household

income (p<0.0001), Frequency of visits to a

dental professional (p<0.0018), Number of

children in the household (p<0.0001)

Maternal level of education, oral health knowledge and behaviours

Ju et al. 2016 1720 Australian

indigenous mother–

child (3- to -5 –

year- old) dyads

Longitudinal

(5-year

follow up)

Mother’s education level Maternal report of

child’s decay, or

extracted teeth due

to decay

0–9 years (β=1.21, 95% CI=1.05-1.39) , 10

years (β=1.06, 95% CI= 0.94–1.19) and 11–

12 years (β=1.06, 95% CI= 0.95–1.19

compared to > 12 years of education

36

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Maternal body mass index (BMI)

Wigen and Wang

2014

5 year old

Norwegian children

(n=1348)

Longitudinal

(5-year

follow up)

Oral health behaviour at 1.5 year of age,

Sugary drinks, Sugary drinks at night,

Maternal health and lifestyle, Maternal

dietary fat, Maternal BMI, Maternal

education, Parental origin, Family status

from pregnancy to age 5

dmft Having mother with non-western background

(β=5.4, 95% CI= 2.8–10.6), Having mother

with low education (β=1.5, 95% CI= 1.1–

2.3), A change in family situation from two

to one parent family (β=2.0, 95% CI= 1.1–

3.4), Having mother who consumed more

sugar and fat than recommended (β=1.6, 95%

CI= 1.1–2.5), Having mother with BMI

classified as obese (β=2.3, 95% CI= 1.3–4.1),

Low tooth brushing frequency at 1.5 year of

age (β=2.3, 95% CI= 1.4–3.7) , frequent

consumption of sugary drinks at 1.5 year of

age (β=1.7, 95% CI= 1.1–2.8)

Maternal caries

Smith et al. 2002 60 mother-child (3-

to- 5- year- old)

dyads from the

USA

Cross

sectional

Maternal high mutans Sreptococci level,

Maternal caries experience, reported sugar

consumption and demographic variables of

mother,

dmft Maternal high mutans Sreptococci levels (p <

0.001), Sugar consumption (p= 0.01),

Maternal active decay (p=0.004),

Ersin et al. 2005 101 Turkish

mother-child (15-

to- 36 months)

dyads

Cross

sectional

Maternal caries experience, Maternal

education, Breast feeding duration, Maternal

sharing of utensils, Child’s caregiver type,

First primary teeth eruption, Using a pacifier

dfs High maternal DMFS scores (OR=1.074;

95%

CI=1.0-1.15)

Kawashita et al.

2009

369 Japanese

mother-child (3 –

year- old) dyads

Cross

sectional

Residential area, Health insurance, Age of

the mother, Drinking alcohol, Smoking

habit, Knowledge of dental terminology,

Frequency of tooth brushing a day,

Frequency of eating between meals a day,

Maternal caries, Sex of child, Birth order,

Habit of feeding in bed, Frequency of eating

between meals a day, Frequency of

consuming sports drinks, Preventive dental

care

dmft Birth order of child (p=0.05), Habit of

feeding in bed (p=0.02), Higher frequency of

eating between meals a day (p<0.001),

Higher frequency of consuming increased

amounts of sports drinks (p=0.001),

Preventive dental care (p=0.002)

37

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Thitasomakul et al.

2009

406 Thailand

mother-child dyads

Longitudinal

(9 – to -18

months

follow up)

Maternal caries experience, Maternal

Educational level, Annual Income, Mother’s

decayed teeth, Calcium supplement during

pregnancy, Drinking milk during pregnancy,

Infant’s birthweight, Type of milk feeding,

Having commercial cereal at 3 months,

Having rice at 3 months, Mother started

sweetening food, Child start having snacks,

Child started having vegetables, Child

started having fish, Child started having

sugary snacks at 9 months, Child started

having local traditional dessert at 9 months,

Child started having soft drinks at 9 months,

Mother brushed the child’s teeth at 9 months

dmft Mother’s decayed teeth ≥ 10 (p ≤ 0.05), No

calcium supplement during pregnancy (p ≤

0.05), Mixed breast- and bottle-feeding (p ≤

0.001), No rice at 3 months (p ≤ 0.05), No

commercial cereal at 3 months (p ≤

0.001),Mother start sweetening food at ≤ 5

months (p ≤ 0.001),Child started snacking at

≤ 5 months (p ≤ 0.01), Child started having

sugary snacks at 9 months (p ≤ 0.05), Child

started having soft drinks at 9 months (p ≤

0.01), No daily tooth brushing at 9 months (p

≤ 0.001).

Weintraub et al.

2010

179 Hispanic

mothers and their <

18 year old children

(n=387) in the USA

Cross

sectional

Maternal untreated caries ds and DFS Maternal untreated caries (OR = 1.85, 95%

CI =1.12, 3.07)

Proença et al. 2015 77 Brazilian mother

child (1 - to-3 -

years) dyads

Cross

sectional

Children’s eating habits, Children’s oral

hygiene habits, Oral health conditions of

mother (dental caries, gingival bleeding and

plaque index), Oral health conditions of child

(caries, visible plaque, bleeding gums)

dmft High maternal caries experience (P < 0.001),

Maternal gingival bleeding (P < 0.001)

Maternal cariogenic bacteria

Chaffee et al. 2014 361 Hispanic

mother-child dyads

Cross

sectional

Maternal sociodemographic measures, Child

feeding and care practices, Maternal oral

health status,

Child’s relative

caries

incidence

feeding and care practices, and maternal

dental status, higher maternal salivary

challenge of both MS (p=0.01) and LB

(p=0.03) over the study period predicted

nearly double the child caries incidence

versus lower MS and LB

38

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Childers et al.

2017

69 African

American mother-

child (1-to -3- year

- old) dyads

Longitudinal

(3-year

follow up)

S mutans genotypes (GT) of mothers and

children.

dmfs Mother-child pair with S mutans GT match

(p=0.014), S mutans in children at age

18 months (>500 CFU/ml) (p=0.002), S

mutans in children at age 18 months (>500

CFU/ml) (p=0.014)

Intrauterine environment of child and birthweight

Bergel et al. 2010 12-year-old

Argentina children

and their mothers

(n=195)

Cross

sectional

Calcium supplementation during pregnancy dmft/DMFT Children whose mother received calcium

during pregnancy had a 27% reduction

in the risk of developing at least one

DMFT/dmft (RR: 0.73, CI 95%=0.62-0.87)

Nelson et al. 2013 Infant-mother

dyads from the

USA

Longitudinal

(2-year

follow up)

Child’s birth weight (normal, low, very low) ICDAS ≥ 2 -

Bernabé et al.

2017

1102 Scottish

children from birth

up to 4 years

Longitudinal

(4-year

follow up)

Child’s early life factors: Age, Gender, Birth

order, Birth weight, Maternal age at birth,

Maternal education, Breastfeeding duration,

Marital status, Maternal smoking, Parental

employment, Area deprivation, Child’s tooth

brushing frequency

dmfs Predicted mean difference in dmfs = 1.86

(95% CI= 0.03 - 3.74) between children with

low and normal birth weight, Predicted mean

difference in dmfs= 1.66 (95% CI=0.57 -

2.75) between children of smoking and non-

smoking mothers.

Child’s age and gender

Casanova-Rosado

et al. 2005

1,644 Brazilian

children aged 6–13

years

Cross

sectional

Social, Economic, Behavioural,

Demographic variables

DMFT,

dmft, SiC

Mother’s schooling (OR=0.95, 95% CI=

0.90-0.97), Caries experience in the primary

dentition (OR= 6.31, 95% CI=4.57-8.72)

39

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Declerck et al.

2008

3-year-old

(n=1250) and 1283

5-year-old

(n=1283) pre-

school children

from Belgium

Cross

sectional

Oral hygiene levels, Reported oral health

behaviours, Socio-demographic factors.

dmft In 3-year-olds,

Caries experience with presence of dental

plaque (OR = 7.93; 95% CI= 2.56–24.55),

Reported consumption of sugared drinks at

night (OR = 7.96; 95% CI=1.57–40.51),

In 5-year-olds,

Age (OR = 7.79; 95% CI= 2.38–25.43),

Gender (OR = 0.37 with 95% CI= 0.19–0.71

for girls), presence of visible dental plaque

(OR = 3.36; 95% CI= 1.64–6.89) Reported

habit of having sugar containing drinks in

between meals (OR = 2.60 with 95% CI=

1.16–5.84 and OR = 3.18 with 95% CI=

1.39–7.28, respectively for 1·/day and >

1·/day

Versus not every day).

Li et al. 2011 1523 Chinese

children aged < 71

months.

Cross

sectional

Age, Gender, Eating habits, Family status,

Childcare provider, Oral intervention

dmft/dmfs Rural residency (β= 0.78, AOR=2.2, 95%

CI=1.25-3.82, p=0.006), Private child care

facility (β = 0.73, AOR=2.1, 95% CI=1.34-

3.20, p=0.001), High frequency of carbonated

drinks (β = 0.52, AOR=1.6, 95% CI=1.02-

2.29, p=0.001), Using dairy products every

day (β = -0.53, AOR=0.59, 95% CI=0.35-

0.98, p=0.04), Bedtime eating (β = 0.88,

AOR=2.4, 95% CI=1.33-4.40, p=0.003), Age

(β = -0.92, AOR=0.40, 95% CI=0.24-0.65,

p=0.000)

Folayan et al. 2015 497 Nigerian

children aged 6

months- to – 6 -

years

Cross

sectional

Children’s ages, Sex, Socioeconomic status,

Tooth brushing habits, Sugary snacks

consumption, Use of fluoridated toothpaste,

Birth rank, Infant-feeding practices,

Breastfeeding practices, Maternal age at

childbirth, Maternal knowledge of oral

health

dmft Consumption of sugary snacks in between

meals more than three times a day (PR=0.05,

95% CI= 0.003 – 0.01, p = 0.04) , Child’s

oral hygiene status (PR: 0.05, 95 % CI=

0.005–0.10, p = 0.03)Child’s gender/female

(PR=−0.06, 95% CI=−0.10- -0.01, p= 0.02),

Mothers’ knowledge of oral health

(PR=−0.06, 95% CI=−0.11- -0.008,p= 0.02)

40

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Pecharki et al.

2016

687 Brazilian

students aged 12-

years-old

Cross

sectional

Individual level, School level, District level DMFT Female (OR=0.51, p=0.33-0.79).

Mwakayoka et al.

2017

525 Tanzanian

children

aged 2-4 - years

and their

parents/caregivers

Cross

sectional

Children’s demographic variables (location,

gender, age, parent/care giver education,

parent occupation), Children’s dietary

behaviours

dmft Older age (OR =2.722 p< 0.001), frequent

consumption of factory made sugary

foods/snacks at age 1-2 years (OR=3.061,

p=0.021).

Warren et al. 2017 396 children aged

5-to-17-years from

the Iowa Fluoride

Study (IFS) cohort

Longitudinal

(17-year

follow up)

Fluoride and demographic/behavioural data

from 9-to-17-years

DFS at age 13 and

age 17

The high caries cluster was associated with

lower maternal education level (p<0.001 ),

lower 100% juice consumption (p<0.01),

lower brushing frequency (p=0.02) and being

female (p=0.01).

Child’s past dental caries

Parisotto et al.

2012

179, 3-and-4-year-

old pre-schoolers

from Brazil

Longitudinal

(1-year

follow up)

Dental caries experience at baseline and at 1

year follow up

dmft Children with caries activity at baseline were

at a higher risk of developing new lesions

than caries-free children (OR=17.3 for early

caries lesion

development, OR=24.5 for

cavitations/fillings, p<0.001)

Cortellazzi et al.

2013

5-year-old Brazilian

preschool children

(n=427)

Longitudinal

(3-year

follow up)

Caries experience at baseline and after 3

years, Gingivitis, Gender, Monthly family

income, Number of people living in the

household, Father’s educational level,

Mother’s educational level, Home

ownership, Fluorosis, Car ownership

dmft Past caries experience (p=0.0004)

Piva et al. 2017 3-to-4-year-old

Brazilian children

(n=119)

Longitudinal

(2-year

follow up)

Socioeconomic variables, Caries at baseline,

Presence of Streptococcus mutans, and

Lactobacillus, with the progression of caries

lesions after

two years of follow-up

ICDAS=0, ICDAS

1, 2, 3 and ICDAS

4, 5, 6

Marital status (RR= 0.73, p=0.04),

MS(RR=0.71, p=0.03), Non-cavitated lesions

(RR=0.48, p <0.001)

41

Author/s Study setting Type of

study

Risk indicators investigated Outcome variable Significant risk indicators

Child’s oral cariogenic bacteria

Tulunoglu et al.

2006

80 children aged 7–

15 years from

Turkey.

Cross

sectional

Unstimulated salivary flowrates, pH,

Buffering, Calcium, Total proteins, Total

antioxidants

DMFS=0 (caries

free) and DMFS>0

(caries active)

Salivary pH (p<0.000), Calcium

concentrations (p<0.000) between 11-to-15-

year-old caries active and caries free girls

Preethi et al. 2010 120 Indian children

in 7-10 and 11-14

year age groups

Cross

sectional

Salivary pH, Buffering capacity, Total

protein, Calcium, Total antioxidant capacity

dmfs/DMFS Significant differences between caries free

and caries active groups; Total antioxidant

Capacity in 7-10-year-old girls (p<0.001),

Total protein (p<0.01), Total calcium

(p<0.05) and Total antioxidant capacity (

p<0.05) in -10-year-old boys, Buffering

capacity (p<0.05), Total protein (p<0.05),

Total calcium (p<0.05) and Total antioxidant

capacity (p<0.01) in 11-to-14-year old girls,

Total protein (p<0.01), Total calcium

(p<0.01), Total antioxidant capacity (p<0.05).

Köhler and

Andréen 2012

Children of mothers

with high salivary

MS levels followed

up at 3, 4, 7, 11, 15

and 19-years-of-age

Longitudinal

(19-year

follow up)

Salivary MS, MS in plaque from 12 selected

proximal surfaces.

Lesion prevalence

(DFS)

Early-colonised children (≤ 3 year olds who

had mothers with high MS levels) had higher

DFS at 19 years of age than those who were

non-MS-colonised at 3 years (p=0.003)

ElSalhy et al. 2013 122 children aged

11-to-12 yeas from

Kuwait

Cross

sectional

Saliva and plaque MS counts, Daily

brushing, Beverage consumption, Sweet

consumption, Rinsing and flossing frequency

ICDAS, CI (CI),

DMFT and dmft

Correlation between plaque MS and ICDAS

CI (p < 0.05). Children who brushed once a

day or more had significantly lower ICDAS

CI (p < 0.01). Children who consumed sweets

or drank soft drinks more than once a day had

significantly higher ICDAS CI (p < 0.05).

El-kwatehy and

Youssef 2016

80 Egyptian

children aged 6-to-

12-years

Cross

sectional

Saliva flow rate, pH, Total protein

concentration, Total antioxidant, Cariogenic

microorganisms.

DMFT and DMFS Saliva pH (p<0.000) and α-defensin (p=0.04)

between caries active and caries free children

Ramamurthy et al.

2014

50 children aged 2–

5-

years from India

Cross

sectional

Salivary MS and LB dmft/dmfs Correlation between salivary MS dmft

(r=0.34, p=0.02) and between MS and dmfs

(r=0.37, p=0.01). Correlation between

salivary LB and dmft (r=0.30, p=0.03).

Presence of MS and LB between caries free

and children with caries (p=0.004).

DMFT-Decayed Missing Filled Teeth index (for permanent teeth)/ dmfs-decayed missing filled surfaces index (for deciduous teeth)/ dmft- decayed missing filled

teeth index (for deciduous teeth/, dfs-decayed filled surfaces (for deciduous teeth)/ ds-decayed surfaces (for deciduous teeth)/ DFS-Decayed Filled Surfaces (for

permanent teeth)/ ICDAS-International Caries Detection and Assessment System/ SiC-Significant Caries index/ CI- Caries Index

42

2.5 Concluding remarks

This chapter provided an overview of the conceptual models used to determine risk factors for

dental caries and how they have evolved over the years with ever-changing sociobehavioural

and environmental characteristics. In particular, the chapter discussed the determinants of the

development and progression of dental caries in children, and conflicting results observed in

different populations. As evident by this overview, although a multitude of risk indicators for

caries exist, and empirical research has been done in various population groups with different

age limits, some results are contrasting, indicating that sets of unique risk indicators act upon

each population. The conceptual model proposed by Fisher-Owens (2006) describes the

interaction within and between factors which ultimately result in dental caries development.

This thesis will use the model suggested by Fisher-Owens as a guide to build a conceptual

framework in assessing risk indicators for childhood dental caries in a population of SEQ

children.

43

Chapter 3: Protocol for assessing maternal, environmental and

epigenetic risk indicators for dental caries in a population of

Queensland children

This chapter is a published paper (paper 1). The bibliographic details of the paper, including

all authors, are as follows:

Fernando S, Speicher DJ, Bakr MM, Benton MC, Lea RA, Scuffham PA, Mihala G Johnson

NW (2015). Protocol for assessing maternal, environmental and epigenetic risk indicators for

dental caries in children. BMC Oral Health 15(1):167. DOI.org/10.1186/s12903-015-0143-2

(29 December 2015).

My contribution to the paper involved the conception and design of the study, exploring data

collection and statistical analysis methods and drafting the article.

The journal in which this article has been published is an open access peer-reviewed journal

with an international readership. Permission has been provided by the publisher, Biomed

Central, under their open access policy, which states that “As an author of an article published

in BMC Oral Health, you retain the copyright of your article and you are free to reproduce and

disseminate your work”.

Surani Fernando

Corresponding author: Newell Johnson

Supervisor: Professor Newell Johnson Supervisor: Professor Paul Scuffham

44

3.1 Abstract

3.1.1Background

Expenditure on dental and oral health services in Australia is AUD $3.4 billion annually. This

is the sixth highest health cost and accounts for 7% of total national health expenditure.

Approximately 49% of Australian children aged 6 years have caries experience in their

deciduous teeth, and this is rising. The aetiology of dental caries involves a complex interplay

of individual, behavioural, social, economic, political, and environmental conditions, and there

is increasing interest in genetic predisposition and epigenetic modification.

3.1.2 Methods

The Oral Health Sub-study, a cross-sectional study of a birth cohort, began in November 2012

by examining mothers and their children who were six years old at the time of the initiation of

the study, which is ongoing. Data from detailed questionnaires of families from birth onwards

and data on mothers’ knowledge, attitudes, and practices towards oral health collected at the

time of clinical examination are used. Subjects’ height, weight and mid-waist circumference

are taken and BMI computed, using an electronic bioimpedance balance. Dental caries

experience is scored using the ICDAS. Saliva is collected for physiological measures. Salivary

DNA is extracted for genetic studies including epigenetics using the SeqCap Epi Enrichment

Kit. Targets of interest are being confirmed by pyrosequencing to identify potential epigenetic

markers of caries risk.

45

3.1.3 Discussion

This study will examine a wide range of potential determinants for childhood dental caries and

evaluate inter-relationships amongst them. The findings will provide an evidence base to plan

and implement improved preventive strategies.

Keywords Dental Caries, Children, Risk, Environment, Genetic, Epigenetic

3.2 Background

Dental caries affects children and adults alike all over the world. It is a disease of the poor as

well as of the rich. Largely preventable, it remains the most prevalent chronic disease among

children with a significant impact on individuals, families and society. It has recently been

reported that 2.4 billion people globally have untreated dental caries (Kassebaum et al., 2015).

More than 40% of preschool and primary school children in Western industrialised countries

and other middle-income countries, including children in the United States of America (Ismail

et al., 2008b), Sweden (Wendt et al., 1996), Brazil (Oliveira et al., 2008) and Australia (Seow

et al., 2009) experience a high prevalence of dental caries. Despite progress made in caries

control over the years by the protective effects of fluoride, increased efforts in oral health

promotion, widespread health education, and remarkable advances in treatment options, dental

caries remains the most common chronic childhood disease (Kassebaum et al., 2015). Among

Australian children aged 6 to 7 years the prevalence of dental caries (treated and untreated

combined) was 32.4% in 2011(Lucas et al., 2011). This figure varies by state from 26.5% in

New South Wales to 43.8%, in Queensland (Table 1) (Lucas et al., 2011). According to the

Queensland Child Oral Health Survey in 2012, 49.5% of children aged 5-10 had experienced

dental caries (Center, 2014). More alarmingly, among South East Queensland children this

46

figure was just over 50% (ARCPOH, 2014). The mean decayed missing and filled teeth index

(dmft) for the same child population was 2.0 with 0.8 teeth being untreated (ARCPOH, 2014).

This worsening situation is despite substantial resources being allocated for the prevention and

treatment of oral diseases (AIHW, 2014).

Table 3.1: Percentage of 6- to 7-year-old children with caries experience in Australia in

2011 (95% confidence Interval)

State Caries prevalence of

6- to 7- year-old children (95% CI)

Australian Capital Territory 32.4% (21.8-43.1)

South Australia 29.8 %(24.6-35.0)

Western Australia 35.5% (31.1-40.0)

Victoria 30.0% (27.3-32.7)

New South Wales 26.5% (28.2-46.3)

Tasmania 37.3% (40.6-47.1)

Queensland 43.8% (40.6-47.1)

Northern Territory 37.8% (15.7-42.1)

All States 32.4% (31.0-33.8)

Source Lucas et al. 2011

Oral health is determined by many factors: socio-economic (Vargas et al., 1998),

environmental (Van De Mheen et al., 1997), political (Petersen et al., 2005), availability of oral

health care facilities (Lave et al., 2002), and their level of utilization (Kim and Telleen, 2004).

Furthermore an individual’s health related behaviours (Petersen, 2005; Rossow, 1992), as well

as oral health knowledge (Ashkanani and Al-Sane, 2012), attitudes (Schultz et al., 2001) and

practices (Ashkanani and Al-Sane, 2012), and biological factors (Nicolau et al., 2005)

including the intrauterine environment (Smith et al., 2002) and genes (Harris et al., 2004) have

an impact on his/her oral health. Additionally, maternal factors like the mother’s level of caries

(Harris et al., 2004; Shearer et al., 2011), her oral health knowledge (Ashkanani and Al-Sane,

2012; Kim Seow, 2012; Saied-Moallemi et al., 2008; Shearer et al., 2011; Smith et al., 2002;

47

Traebert et al., 2011), salivary loads of cariogenic bacteria (Li et al., 2005; Ramamurthy et al.,

2014; Van Houte, 1993), salivary characteristics including pH, flow rate and buffering capacity

(Bardow et al., 2000; Bardow et al., 2004) as well as maternal genetics (Harris et al., 2004) are

significant risk indicators to be considered for childhood dental caries. Furthermore, an holistic

approach in identifying risk indicators for dental caries is required as oral health is but a part

of general health (Fisher-Owens et al., 2007). Investigating newly emerging risk factors and

risk indicators for dental caries is crucial, and these can be discovered by studying the natural

history of disease and by using modern molecular-based approaches (Johnson, 1991). A

conceptual model of how we envision that these factors might interact is presented in figure

3.1.

48

Figure 3.1 Conceptual framework for the possible interaction of risk indicators under

investigation (Permission has been provided by the publisher, Biomed Central, under their open

access policy, which states that “As an author of an article published in BMC Oral Health, you

retain the copyright of your article and you are free to reproduce and disseminate your work)

49

The importance of inherited factors in susceptibility to dental caries has been recognized for

decades (Klein and Palmer, 1940; Klein, 1946). Heritability is the proportion of phenotype

variation explained by genetic factors. These have been estimated to account for 30-60% (p ≤

0.01) of the variation in caries scores for the permanent dentition and 54-70% (p ≤ 0.01) for

deciduous caries scores (Wang et al., 2010). According to Shaffer et al. (2015), there is direct

evidence for a genetic component in the aetio-pathogenesis of dental caries (Shaffer et al.,

2015) but very little is known about the specific casual mechanisms (Olszowski et al., 2012).

There are many genes related to the composition and structure of dental enamel, inherited

alterations in sugar metabolism, and genetic regulation of salivary gland function.

Unsurprisingly, no single host gene that directly regulates dental caries initiation or progression

has been identified (Shaffer et al., 2012a). Epigenetics, however, may provide the missing link

to these unanswered questions (Barros and Offenbacher, 2009). Epigenetics investigates the

molecular mechanisms that link genetic and environmental cofactors to disease outcomes

(Williams et al., 2014). There are many complex mechanisms underlying epigenetic alterations,

such as DNA methylation, histone modification and gene regulation by non-coding RNA, of

which DNA methylation is the most common (Seo et al., 2015). Such changes may be heritable

or could be environmentally modulated. DNA methylation seems to be the primary mechanism

to suppress retro-transposable elements which are responsible for creating genetic variation

and, on occasion, disease-causing mutations within the human genome. Because these elements

remain partially methylated in sperm, there may be a prolonged period of transposition lasting

a few generations before gene expression (Tang et al., 2015). Possible epigenetic contributions

are currently being investigated in many diseases, but such studies are relatively novel in the

dental field (Seo et al., 2015) and are needed to fully understand DNA-based factors important

for development of oral diseases (Fisher-Owens et al., 2007).

50

3.3 Methods

3.3.1 Study population

The research described in this paper is the oral health arm of the main “Environments for

Healthy Living – the Griffith Birth Cohort Study” (EFHL), which, beginning in 2006, has

recruited some 3,000 families from South East Queensland. EFHL is a prospective, multi-year

longitudinal birth cohort study, collecting information during pregnancy through infancy and

adulthood (Cameron et al., 2012b). The EFHL study population includes all births from three

geographically defined adjoining Health Districts in Queensland (Logan, Beaudesert, and the

Gold Coast) and Northern Rivers/Tweed in NSW from 2006 to 2011. These four areas cover

30% of Queensland’s population. Women waiting for antenatal clinic appointments from the

three public maternity hospitals (Logan, Gold Coast, and Tweed) in the participating districts

were contacted by research-trained midwives, provided with a detailed explanation of the study

aims, and invited to participate (Cameron et al., 2012a). Written informed consent was obtained

from participants to access their information from hospital databases, to complete a maternal

baseline survey and for individual follow-up (Cameron et al., 2012b). The initiators of the

EFHL study expected to incorporate future sub-studies including research on dental and oral

health (Cameron et al., 2012a). The population for the oral health sub-study, which began in

2012, is a subset of 6- to 7-year-old EFHL children and their mothers who accepted our

invitation to participate.

Ethics approval to initiate the oral health sub-study was obtained from the Griffith University

Human Research Ethics Committee on 26.12.2012, based on a full National Ethics Approval

Form process, with N.W. Johnson as responsible Principal Investigator. A Variation to archive

and analyse DNA was approved on 17.07.2014 (GU Ref No: OTH/25/13/HREC). The ethics

51

approvals and informed consents obtained cover the clinical examination of mothers and their

child/children and parental consent for full participation for their child/children in all aspects

of the study.

3.3.2 Sample recruitment and examination

In 2012, mothers of the first cohort of children, who were 6 years old at the time, were sent

letters alerting them to the oral health study, followed by a telephone call to assess their level

of interest. Since then we went on to recruit participants from 2007 to 2009 cohorts and the

recruitments are still taking place. Participants who find it difficult to travel far, who have

serious illness and who are not willing to undergo a detailed oral examination are excluded

from the study. Those who agree to attend the dental clinic at Griffith Dental School are given

appointments for a free dental examination of the mother and child by two qualified dental

surgeons, which includes offer of free treatment for the child if required. Both mother and child

are asked not to brush their teeth 2-3 hours prior to examination, not to consume sweet food or

beverages, smoke, or use a mouthwash as per oral examination criteria. Mothers who are

pregnant or who are wearing cardiac pace makers are excluded from weighing on the

bioimpedance scale. Detailed medical histories are taken when subjects arrive at chair side

(Figure 3.2).

52

Figure 3.2: Flow chart showing the process of data collection (Permission has been provided

by the publisher, Biomed Central, under their open access policy, which states that “As an

author of an article published in BMC Oral Health, you retain the copyright of your article and

you are free to reproduce and disseminate your work)

A careful examination of the head and neck, visually and by palpation, is performed. Each

participant is then examined for salivary physiology, for the health of the oral soft tissues, for

dental status, and for experience of dental caries. Periodontal disease requirement (n = 147

dyads) was initially guided by the one-sample correlation test to determine whether a

correlation coefficient differs from zero, set at a low correlation level of r=0.4 (2-tailed alpha

=0.05, power=0.80), calculated using the “power one correlation” command of STATA v 13.1

data analysis statistical software (STATACorp, United States) (VanVoorhis and Morgan,

53

2007). In this study the sample size is limited by pragmatic factors (budget, time, and available

sample population).

3.3.3 Primary outcome variable

Each child’s dental caries experience is assessed using the International Caries Detection and

Scoring system (ICDAS) (CICDAS, 2005). The dentition is examined in detail, and every

surface of every tooth scored after drying. Lesions of dental caries on both smooth surfaces

and in pits and fissures, are scored from a range of 0 to 6 depending on severity. These scores

cover a range from non-cavitated “white spot” and “brown spot” lesions to overt cavities. A

special feature of ICDAS is the additional information it gives on the presence of, and type of,

any restorations on every surface which would be counted as past caries experience. Overall

caries experience per individual is calculated as a percentage of surfaces affected out of all

surfaces examined, and this can be set at any level of caries severity.

3.3.4 Main explanatory variables

Mother and child related variables are collected using questionnaires, clinical examinations and

laboratory testings.

3.3.5 Oral health knowledge and practices

After informed consent, a detailed questionnaire with 43 questions in total (developed for the

study) related to oral health is administered to mothers before the clinical examinations, to

collect data on knowledge, attitudes and practices towards oral health (Appendix A). Mothers

are questioned on their knowledge on oral diseases, the causes of dental caries in children and

adults and available oral health care services. With closed-ended questions, their dietary

54

practices, oral hygiene methods and frequency, use of oral health care services, and infant

feeding practices are assessed. A Likert scale measures the attitudes toward oral health, and the

importance mothers place on child’s oral health and deciduous teeth.

3.3.6 Anthropometric measurements

Height, waist circumference, and stride length of mother and child are measured. Participants

are weighed with an electronic bioimpedance balance, which gives measurements of body fat,

body water composition and BMI for adults and only weight for children. Mothers who are

pregnant or who have a cardiac pacemaker implanted are excluded from weighing on the

bioimpedance balance.

3.3.7 Saliva characteristics

We evaluate oral hydration and salivary viscosity (sticky/frothy, frothy/bubbly, or watery) by

visual observation. Simulated saliva is collected over a period of 5 minutes by chewing on

paraffin wax, and dribbling into a sterile cup. The expectorated volume is recorded. The pH

and buffering capacity of stimulated saliva are measured using Saliva-check BUFFER kits

(GC, United States of America) (Kitasako et al., 2008; Varma et al., 2008). Further, Caries

Risk Test (CRT) kits (Ivoclar Vivadent, Australia) are used to assess saliva buffering and

salivary counts of MS and LB (Kitasako et al., 2008).

3.3.8 Periodontal status

Mothers are examined for periodontal status using Periodontal Screening and Recording (PSR)

index measures, which measure the depth of periodontal pockets from the free gingival margin

to the bottom of the gingival sulcus (Khocht et al., 1995; Polk et al., 2012). Both upper and

55

lower teeth are divided in to sextants. Each and every tooth in the oral cavity is examined and

the highest score in each sextant is recorded. The scores vary from 0, denoting healthy

periodontium to 4, representing severe periodontitis (Landry and Jean, 2002).

3.3.9 Demographic and environmental data

Demographic, social, and environmental data were collected using detailed questionnaires

during recruitment and follow-up phases of the EFHL study from 2006 to 2011. All selected

factors are hypothesized predictors of the primary outcome. The present study will use the data

on each family’s socio-economic and demographic information of the mother, maternal age,

mother’s highest level of education, employment status, and total annual family income. The

child’s intrauterine environment will be assessed by maternal prenatal nutrition, mother’s

smoking habits, prescribed and illicit drug use, and alcohol. Data on breastfeeding and weaning

practices, types of food and beverages consumed, infant supplementary food, and bottle feeding

practices, will be used as covariates to include in the risk factor model. Oral and other health

care received by the child will be assessed from data on dental and medical check-ups (GP

visits), emergency department attendances, and availability of a medical insurance (health care

card).

3.3.10 Genetic and epigenetic markers

To identify genetic and epigenetic markers associated with dental caries, we will target host

genomic DNA obtained from saliva samples and use next-generation sequencing technology

to measure DNA-based variants on a genome-wide scale.

56

Specifically, stimulated saliva will be collected in 2 ml tubes and stored at -80 degrees C until

DNA is extracted with the High Pure PCR Template Preparation Kit (Roche Diagnostics,

Australia) using a method which was extensively modified in order to significantly increase

the DNA yield to a concentration needed for next-generation sequencing (unpublished

protocols). The extracted DNA will be sent to the Centre for Clinical Genomics at University

of Queensland to perform sequencing of bisulphite treated DNA for methylation analysis. This

approach will allow us to gain information on DNA sequences, variance, and CpG methylation

at ~ 450000 CpG sites spanning the human genome. These assays will be performed using the

SeqCap Epi 4 M CpGIANT Enrichment kit (Roache NimbleGen, Australia) and the HiSeq

2500 sequencer in (Illumina, United States) (Duhaime-Ross, 2014). A manuscript by Allum et

al., provides a detailed description of the assay protocols (Allum et al., 2015).

To date we have conducted a pilot study with 12 mother-child dyads; six with high caries

experience (average caries experience, 27.1% of surfaces affected in mothers and 12.4% in

children); and six with low caries experience (average caries experience, 11.2% in mothers,

2.5% in children), which was initiated using data collected within the oral health sub study of

EFHL. As a result of this pilot study, we have been able to establish the DNA extraction,

purification and sequencing protocols (in our hands) and have also established the necessary

bio informatics and statistical analysis methods for identifying target genes for dental caries.

3.4 Analysis

Observations will be stored as de-identified data, using a unique identifier for each participant.

The dataset for the analysis will be prepared in SPSS in a wide data format (one row per

57

participant). Data cleaning will be performed. Outliers will be investigated and corrected or

replaced with missing values if necessary. The pattern of missing values will be investigated,

and variables will not be used for further analysis if < 30% of values are found missing; missing

data will not be imputed.

The dependant variable (dental caries experience of children) is expected to be a ratio type

continuous variable, right-skewed with a mode of zero and which could be modelled using a

generalized linear model (GLM) with log-link and gamma family. The number of independent

variables will be reduced where possible (e.g. oral health knowledge and practices, periodontal

status) by using aggregate values. Independent variables without a known or hypothesized

association with the dependant variable will not be used in regression analysis. The

independent variable (covariates) will be prepared as follows: (a) dichotomous variables: coded

as 0/1, the zero category with the higher frequency, presented as number and proportion (%),

ignored for GLM if rare (frequency < 20); (b) categorical variables: coded as 0/1/2/ etc., sorted

by frequency in descending order, presented as number and proportion, for GLM the smaller

categories will be collapsed to form categories with frequency < 20, dummy coded for GLM;

(c) ordinal variables: coded as 0/1/2/ etc., sorted in a meaningful way, presented as number and

proportion, for GLM neighbouring categories will be collapsed to form categories with n <20;

and (d) continuous variables: re-scaled where necessary to ensure a change of a value of 1 can

be interpreted in a meaningful way, centred over mean where necessary (e.g. for age), presented

as mean and standard deviations (symmetrical distributions) or median and 25th / 75th

percentiles.

Descriptive statistics of participant characteristics (of values before re-categorization or

transformation) will be presented. Unadjusted associations between dependant variables and

58

covariates will be explored with bivariate GLMs. The covariates of interest for a multivariate

model will be shortlisted at p > 0.02. Correlations between the shortlisted covariates will be

investigated (using Pearson or Spearmen correlation and scatter plots), and covariates will not

be used together in a multivariate model if statistically significant correlation is found at |r| >

0.4. Scatter plots will be drawn to investigate the relationships between the dependant variables

and the covariates. Interaction terms will be created where necessary. The multivariate GLM

will be built by manually adding covariates one-by-one (forward method; sorted by descending

univariate p-values), whilst observing changes to the coefficients and errors for variables

already in the model. Covariates will be dropped from the multivariate model at p > 0.05.

Collinearity between covariates will be checked with the variance inflation factor. Model

building will be repeated using a backward method to check the final model. The number of

covariates in the model will be limited to avoid overfitting, as discussed in the sample size

section. Assumptions of GLM will be checked and the model adjusted if necessary.

Coefficients will be presented with the 95% confidence intervals, and evaluated considering

clinical significance. Intervals will be evaluated considering clinical significance. Statistical

significance will be declared at p > 0.05; the effect of multiple comparisons on overall error

rates will be considered during discussion of the results.

For epigenetic variation analysis, site-specific methylation levels will be defined as a

percentage methylation, with values ranging between 0 and 1. Each identified differentially

methylated site will be attributed a percentage methylation that will be included in the

regression modelling approach. Pyrosequencing on selected differentially methylated sites will

be done to validate the SeQCap Epi data.

59

3.5 Discussion

This cross-sectional study to assess maternal, environmental and individual risk indicators for

childhood dental caries will examine a wide range of determinants. It will evaluate the

interrelationship among independent variables and their relative effects on dental caries of

children. Furthermore, this research will pioneer research in epigenetic variation for dental

caries in children. The study will also potentially provide evidence on the interrelationship of

epigenetic variations with other social and environmental predictors for dental caries in the

participating child population. It is hoped that the evidence generated by the current study will

enable a more effective “risk factor modification approach” to overcome the problem of this

chronic disease that commences in childhood and continues throughout life. The data generated

in this thesis may possibly be extrapolated to similar populations worldwide, but further testing

on a much larger and more varied sample will be required. From thereon, we expect that

appropriate interventions will be recognized and implemented to tackle dental caries in children

and improve oral health and quality of life of the affected. However, we note that due to time

and logistic constraints, obtaining a larger sample size is one limitation of the study.

60

Chapter 4: Maternal caries experience, household income,

carbonated drinks and age at commencement of tooth-brushing

are associated with caries experience among 6- to 7-year-old

children in Australia.

This chapter is a submitted paper (paper 2). The bibliographic details of the paper, including

all authors, are as follows:

Surani Fernando, Gabor Mihala, Mahmoud Bakr, Shu-Kay Ng, Rodney Lea, Paul A Scuffham,

Newell W Johnson. Maternal caries experience, household income, carbonated drinks and age

at commencement of tooth brushing are associated with caries experience among 6- to 7-year-

old children in Australia.

My contribution to the paper involved the conception and design of the study, data collection,

statistical analysis, and drafting of the article.

The paper is submitted and under review with the journal Caries Research, which is a peer

reviewed journal (Manuscript ID 201705020).

Surani Fernando

Corresponding author: Surani Fernando

Supervisor: Professor Newell Johnson Supervisor: Professor Paul Scuffham

61

4.1 Abstract

Dental caries in children is a major public health problem worldwide, and research has shown

that there are a multitude of risk indicators acting upon children to different degrees in different

communities. The objective of this study was to determine the maternal and environmental

indicators which are associated with dental caries activity in a population of 6- to 7-year-old

children in SEQ, Australia. Maternal education, employment status, annual household income,

and marital status were used as proxies for environmental indicators; whereas maternal risk

markers included mother’s age at examination, lifetime caries experience, oral health

knowledge and practices, and BMI. Data were collected on mother’s saliva characteristics of

clinical oral hydration, stimulated flow rate, pH, buffering capacity, viscosity, and colony

forming units per ml of salivary MS and LB, which were included as covariates. These data

were analysed as follows: (i) using binary logistic regression with child’s lifetime cumulative

caries experience as the dependent variable; and (ii) using negative binomial regression with

child’s number of tooth surfaces affected with caries as dependent variable. Results indicated

that delayed child’s age when brushing started (p = 0.05), low annual household income (p

=0.03), child’s daily consumption of carbonated drinks (p = 0.02), and mother’s high caries

experience (p = 0.01) were directly associated with children’s caries experience. However,

maternal saliva LB levels were inversely associated with dental caries in children (p =0.01).

These data support the evidence of associations between maternal and environmental indicators

and caries in children in a typical western industrialised country.

Key words – Risk, Dental, Caries, Children, Mothers, Environment, Tooth-brushing,

Carbonated, Income

62

4.2 Introduction

Untreated caries in permanent teeth was the most prevalent disease condition worldwide in

2010, affecting 2.4 billion people. Untreated caries in deciduous teeth was the tenth most

prevalent condition, affecting 621 million children worldwide (Kassebaum et al., 2015) and

nearly 30% of 5- to 10-year-old Australian children (Do and Spencer, 2016). Dental caries

experience in children aged 5 to 6 years in Australia has varied over the last four decades: there

was a decline in prevalence during the 1970s and 1980s followed by an increase, captured by

both the Child Dental Health Survey 2014 and the National Oral Health Survey of Australia

1987–88 (Mejia et al., 2014). The prevalence of untreated dental caries [defined as decayed,

missing or filled surfaces (dmfs) more than zero] among 5- to 10-year-old children had

increased from 35% to 42% over two decades between 1988 and 2014 (Do and Spencer, 2016;

Slade and Spencer, 1998). Moreover, the Child Dental Health Survey 2014 reported that the

mean dmfs of deciduous teeth among the same age group of children was 3.1, of which 6% of

children had teeth missing due to caries, and 26% had filled tooth surfaces (Do and Spencer,

2016). Current caries prevalence in the deciduous dentition was higher among Queensland

children (50.2%) (Do and Spencer, 2016), compared to 1988 (48%) (Slade and Spencer, 1998).

It might be expected that with improvements in large-scale oral health promotion efforts,

increased exposure to fluoride from all sources, opportunities for better nutrition, and improved

treatment options, the incidence of caries would have declined in Australia in recent decades

(Do et al., 2017). However, individuals and populations are exposed all their lives to multiple

risk factors for dental diseases at different magnitudes (Johnson, 1991). Identifying the balance

or mix of factors affecting particular population groups and individuals is important for

63

prevention and management of the disease. No risk occurs in isolation, and many of these

happen in a complex series of events (Petersen, 2005).

Children from socially disadvantaged populations such as low socio-economic groups,

indigenous communities, and ethnic minorities have the highest risk for dental caries across

the globe (Seow et al., 2009; Tanaka et al., 2013; Zander et al., 2013). Since mothers play a

major role in rearing children, the maternal level of education (Kumar et al., 2014),

employment status (Kumar et al., 2014; Tanaka et al., 2013), oral health knowledge (Lee et al.,

2014), and behaviours (Nourijelyani et al., 2014) as well as maternal age (Ju et al., 2016;

Warren et al., 2016) are known to contribute to the health of her offspring. Because dental

caries is a lifestyle disease, chronic conditions such as obesity and diabetes share common risk

indicators with dental decay. Having mothers who consumed more than the daily recommended

sugar, fat and carbohydrate intake was found to be associated with caries development in young

Norwegian children (Wigen and Wang, 2014): children with obese mothers had more than

twice the risk of developing dental caries than the children of mothers who had a normal body

mass index BMI (Wigen and Wang, 2011). We hypothesise that caries experience in a

population of 6- to 7-year-old children living in SEQ is associated with maternal socio-

economic, demographic and behavioural factors.

Relationships between parental and children’s oral health status have been studied for decades

(Klein, 1946), and mothers’ caries status has been recognized as a stronger influence on caries

experience of children compared to fathers’ (Birungi et al., 2016; Mattila et al., 2000;

Weintraub et al., 2010). This is because certain parent-caregiver characteristics expose the child

to adverse environmental conditions that are conducive for caries development and

progression. Other than imitating unhealthy practices of caregivers, children are thought to be

64

at risk of acquiring cariogenic bacteria from their mothers. Maternal and child strains of MS

have been found to be similar, and higher salivary counts in mothers were associated with early

colonization of these organisms in children (Li et al., 2005). Mothers’ salivary MS (Warren et

al., 2009) and LB (Chaffee et al., 2014) levels have been able to predict children’s dental decay.

Putatively cariogenic bacteria thrive in environments such as low salivary flow rates (Diaz de

Guillory et al., 2014), low buffering capacity of saliva (Gao et al., 2016; Varma et al., 2008),

and low pH levels produced by cariogenic bacteria from dietary carbohydrates (Burne and

Marquis, 2000; Kianoush et al., 2014), increasing the risk of enamel and dentine

demineralization (Melo et al., 2016). Assuming that mothers associate more closely with their

children, we hypothesize that physiological features of maternal saliva could be taken as

predictors of the child’s dental caries experience.

From an epidemiological point of view, it is important to identify prominent indicators

contributing to dental caries development among children in each individual community.

Identifying risk indicators may substantially advance appropriate preventive and restorative

care. Hence, the objectives of the current study were to establish the prevalence of dental caries

in a population of children in SEQ, Australia, and to estimate associations of maternal and

environmental factors with dental caries experience in children.

4.3 Materials and methods

A protocol for the study was published in BMC Oral Health (2015) (Fernando et al., 2015).

65

4.3.1 Study population

The population for this cross-sectional study was 6- to 7-year-old children and their mothers

from three districts of SEQ (Logan, Beaudesert and Gold Coast), enrolled in the Griffith

University EFHL birth cohort study (Cameron et al., 2012b). According to Cameron et al the

EFHL cohort was compared with the rest of the SEQ and Australian demographics (Cameron

et al., 2012b). Participants of the oral health sub study was hence a representative sample of

SEQ. Recruiting participants for the oral health sub-study started in November 2012 from the

first cohort of children (2006), who were 6 years of age at the time, followed by participants

from 2007, 2009 and 2010 cohorts. It is a possibility that families which accepted our invitation

were better motivated towards health and were grateful for the opportunity to receive an oral

health examination and seized an opportunity for treatment. Data collection was completed in

April 2016 with a total sample of 174 mother-child dyads. However, data on oral health

knowledge and practices, salivary physiochemical characteristics and dental caries experience

was missing for one mother, hence the sample of 173 mother-child dyads.

4.3.2 Data collection

A questionnaire specifically developed for the oral health sub-study (non-validated) was

administered to mothers before clinical examinations. Data were collected on knowledge,

attitudes and practices related to oral health.

Mothers who were not pregnant or who did not have a cardiac pacemaker were weighed with

an electronic bioimpedance balance (Tanita, Australia), which gives measurements of body fat,

body water composition and BMI.

66

Oral examinations were carried out by trained and calibrated examiners at Griffith University

Dental clinics. Weighted Kappa values for two examiners against a gold standard were .810

(SE = 0.102, 95% CI = 0.414-0.813) and 0.928 (SE =0.093, 95% CI =0.628-0.992) (Appendix

B; Table 1 and 2). Dental caries experience was measured using the ICDAS (CICDAS, 2005),

with scores ranging from 0 (no disease) to 6 (overt caries) on each tooth surface. Presence of

restorations was considered as past caries experience. Saliva hydration was evaluated by visual

observation, and stimulated saliva was collected over 5 minutes whilst chewing on a piece of

paraffin wax and the volume measured. Saliva pH and buffering capacity were measured on

the expectorated sample with Saliva-check BUFFER kits (GC, Australia) and Caries Risk Test

kits (Ivoclar Vivadent, Australia). MS and LB colony forming units were measured according

to manufacturers’ instructions after 48 hours incubation at 370 C.

Demographic, socio-economic and environmental data were collected using detailed

questionnaires at recruitment for the EFHL study (Cameron et al., 2012a). In depth data of

participants and their families were merged with the oral health clinical data for analysis.

4.3.3 Outcome and risk indicators

The primary outcome variable was children’s dental caries experience, expressed as number of

tooth surfaces affected by dental caries (at the time of examination) including initial enamel

white spot lesions (ICDAS 1, 2 and 3) and dentinal caries (ICDAS 3, 4 5 and 6). The reason

being that the majority of children (61%) were caries free and the rest of the sample needed to

be included in the analysis for meaningful outcomes. This outcome was used as follows: (i) a

dichotomized variable: children with caries (ICDAS > 0) and children without caries (ICDAS

= 0); and (ii) as a continuous variable: total number of surfaces affected with caries (counts).

67

There were very few teeth extracted due to dental caries it was not considered for analysis,

since the influence would have been negligible.

Based on the questionnaire a total score (out of 100) for each participating mother was given

to evaluate their oral health knowledge attitudes and behaviours. Questions related to oral

hygiene practices, use of dental care services, feeding practices and consumption of cariogenic

food/beverages from the questionnaire were selected separately, to estimate mothers’ oral

health practices.

Moreover, mothers’ saliva hydration (normal, low), buffering capacity, pH, MS colony

forming units/ml of saliva (<105 = low, >105 = high), and LB colony forming units/ml of saliva

(<105 = low, >105 = high) were taken as saliva characteristics that could possibly be associated

with their children’s dental caries experience. Other explanatory variables included maternal

age, level of education, employment status, and annual household income.

4.3.4 Statistical analyses

Data were analysed using SPSS-21 (IBM, Chicago, IL). Measures of central tendency for

continuous variables, and frequency and percentages for categorical variables, were computed.

Categorical variables were recoded to ensure the frequency was at least 20 in each category.

Based on our research hypothesis, logistic regression was performed in GLM to explore

indicators that might have an impact on caries experience of children. Explanatory variables

were systematically tested in simple regression analyses, and shortlisted at p < 0.10 (Mota-

Veloso et al., 2016; Peres et al., 2005). Shortlisted explanatory variables were tested in

multivariable regression models with the significance level set at p < 0.05 and variables that

were insignificant were dropped to arrive at the final models. Final models were determined

68

by the significance levels of Omnibus test X2 (p ≤ .05) and clinically meaningful significant

associations between explanatory and outcome variables. The risk indicators for caries in

children were expressed with Adjusted Odds Ratios (AOR), Adjusted Rate Ratios (ARR) and

95% Confidence Intervals (CI).

4.4 Results

Of the 173 children, 88 (50.6%) were girls. Mean age for children without caries (n = 106) was

6.5 years (SD = 0.65) and 6.1 years (SD = 1.02) for children with caries (n = 67). Mean caries

experience (measured with the mean number of tooth surfaces affected with caries) was 8.8

(SD = 10.1) among children with caries. Mean maternal caries experience was 22.6 (SD =

13.13) within the caries free group and 25.29 (SD = 19.16) among the caries group. Mean

overall age for mothers on the day of clinical examination was 37.3 years (SD = 5.46). Other

maternal sociodemographic and clinical characteristics explored are given in Table 4.1.

Variables on maternal oral health knowledge, behaviour, and environment, explored as risk

indicators for childhood caries, are listed in Table 4.2.

Table 4.3 shows the variables in the multivariable binomial logistic regression model. Only

“age when child started brushing” was significantly associated with child’s caries experience

(p = 0.05). Children whose mothers initiated their children’s brushing after 6 months of age

were two times more at risk than their counterparts who initiated brushing at six months or

earlier (AOR = 2.23, 95% CI = 1.01-4.95). Initial frequency distribution showed that the

outcome variable was skewed with more children free of dental caries (61%), hence analytical

methods had to be improved according to the distribution.

69

As the total number of surfaces affected with caries were over dispersed (mean = 5.39, variance

= 80.2), negative binomial regression was used to assess risk indicators for dental caries.

Amongst all maternal independent variables entered into the model (listed in Tables 4.1 and

4.2), high levels of maternal LB was associated with low dental caries experience in children

(ARR = 0.61, 95% CI = 0.40-0.87, p = 0.01), whereas mother’s higher caries experience was

a risk marker for children’s caries (ARR = 1.02, 95% CI = 1.01-1.03, p = 0 .01). Low household

income (ARR = 0.67, 95% CI = 0.47-0.96, p = 0.03) and child’s consumption of carbonated

drinks once a day compared to never having such drinks (ARR = 2.03, 95% CI = 1.32-3.57, p

= 0.02) were significantly associated with high caries experience in our child population (Table

4.4).

70

Table 4.1: Frequency distribution of children’s and mother’s socio demographic and

clinical characteristics

Variable Children without

caries (n=106)

Children with

caries (n=67)

Mean SD Mean SD

Child age (years) 6.5 0.65 6.1 1.02

Child caries experience as number of tooth

surfaces affected with caries¥

0 0 8.8 10.10

Mother age 37.4 5.62 37.1 5.24

Mother saliva pH 7.0 0.41 7.1 0.53

Mother saliva buffering (GC*) 8.70 3.54 8.57 3.16

Mother caries experience as a percentage of

affected surfaces

22.6 13.1

3

25.29 19.16

Mother saliva volume in 5 minutes 5.21 2.30 4.87 3.36

N % N %

Child gender

Female 51 48 38 56

Male 55 52 29 44

Mother saliva hydration

Low 15 14 7 10

Normal 91 86 60 90

Mother saliva buffering capacity

(CRT**)

Low 20 19 16 23

High 86 81 51 77

Mother saliva mutans Streptococci

Low 33 31 27 40

High 73 69 40 60

Mother saliva Lactobacilli

Low 51 48 32 48

High 55 52 35 52

Mother level of education

Completed primary education 14 13 10 14

Completed high school 32 30 17 26

Completed tertiary education 60 57 40 60

Mother employment status

Unemployed 49 46 31 46

Employed 57 54 36 54

Mother marital status

Single 12 11 6 9

With partner/husband 94 89 61 91

Annual household income

Low (<$59999) 56 53 31 46

High (>$59999) 50 48 36 54 ¥ Caries experience was for both deciduous and permanent dentition in children, *Saliva-check BUFFER

kits (GC, United States of America), ** Caries Risk Test kits (Ivoclar Vivaden)t, Australia).

71

Table 4.2: Frequency distribution of mothers’ oral health related knowledge and

behaviours

Variable Children without

caries(n=106)

Children with

caries (n=67)

Mean SD Mean SD

Mothers oral health knowledge 72.6 9.65 72.6 10.8

N % N %

Mothers visits to dentist during pregnancy

Yes 72 68 48 72

No 34 32 19 28

Child’s age when brushing started

Six months 30 28 13 19

More than six months 76 72 54 81

Child’s dental visits since birth

Yes 75 74 48 72

No 25 26 19 28

Use of fluoridated tooth paste

Yes 63 59 33 49

No 43 41 34 51

Frequency of giving carbonated drinks per day

Never 65 61 49 73

Once a day 20 19 5 7

More than once a day 21 20 13 20

Mother BMI at beginning of pregnancy

Normal 62 58 38 57

Over weight 21 20 12 18

Obese 23 22 17 25

Main language spoken at home

English 93 88 61 91

Other 14 12 6 9

Table 4.3: Risk indicators for children with and without dental caries using logistic

regression

Omnibus test X2 for the model = 11.44, p=0.04, * Reference variables, AOR= Adjusted Odds Ratio, CI=

Confidence Interval, MS=mutans Streptococci

Variable AOR (95% CI) P value

Mother saliva MS levels 0.68 (0.34-1.36) 0.28

Mother caries experience 0.98 (0.96-1.00) 0.07

Use of fluoridated toothpaste at home (Yes*/No) 1.25 (0.88-1.77) 0.21

Age when child started brushing (6 months*/> 6 months) 2.23 (1.01-4.95) 0.05

Main language spoken at home (English*/non-English) 0.48 (0.16-1.37) 0.17

72

Table 4.4: Risk indicators for childhood dental caries using negative binomial regression

Variable Adjusted ARR (95% CI) P

value

Mothers’ levels of saliva LB

(Low*/High)

0.61 (0.40- 0.87) 0.01

Mothers’ saliva buffering capacity

(GC)

0.96 (0.91-1.01) 0.11

Mothers’ saliva pH 1.14 (0.75-1.73) 0.55

Mothers’ caries experience 1.02 (1.01-1.03) 0.01

Mothers’ annual income

(Low* /High) 0.67 (0.47-0.96) 0.03

Frequency of giving carbonated

drinks

Never*

Once a day 2.03 (1.32-3.57) 0.02

>Once a

day

1.06 (0.66-1.68) 0.82

Variance of outcome variable =80.25, Mean of outcome variable=5.39, Kolmogorov–Smirnov

statistic=1.075(p=0.198), Omnibus test X2 for the model=40.56, p=0.001, Dispersion parameter=2.57, ARR=

Adjusted Rate Ratio, CI= Confidence Interval, *Reference variables.

4.5 Discussion

The present study is based on our findings of caries prevalence and severity in a sample of 173

mother-child dyads in SEQ. We found that 34% boys and 42% of girls between 6 and 7 years

of age had active caries as per ICDAS criteria, whereas the Australian National Oral Health

survey 2012-2014 found it to be 30% for boys and 27% for girls in the same age group (Do

and Spencer, 2016). The higher rates in the current study could be due to the fact that volunteers

who presented at our clinics had a self-assessed treatment need. This is logical because

participants’ own perception of treatment need has been found to correlate significantly with

examiner ratings in a study conducted in Sweden with 1000 individuals aged 20-89 years

(Petersson et al., 2016). In the USA, around 37% of children aged 2 to 8 years had caries in

their deciduous teeth (Dye et al., 2015), and in Spain the prevalence of caries in 6-year-olds

was 30% (Almerich Silla et al., 2014). Bosnian 6-year-old children had a mean dmft of 6.7 (SD

±3.9) with decayed teeth contributing to 88.8% of the index (Markovic et al., 2013). A

73

longitudinal study in UK, to investigate caries progression through childhood to adolescence,

found that the mean caries prevalence of 7- to 9-year-olds’ was 16.7% which increased with

age (Hall-Scullin et al., 2017). These observations indicate that caries experience in the same

age group varies considerably in different communities, presumably because of differing

mixtures of risk indicators.

Evidence suggests that preventive interventions such as initiation of oral hygiene habits before

the age of 12 months are important to prevent the development of dental caries (Lee et al.,

2006). Children who started brushing at an early age were at comparatively low risk for

developing dental caries in a multicentre study from paediatric dentistry departments in the

Netherlands and Turkey (Özen et al., 2016). Moreover, caries prevalence was found to be

significantly higher (p = 0.04) in a population of Myanmar preschool children who did not start

brushing until after 2 years of age (Thwin et al., 2016). Similar results were observed by Huong

et al., (2017) in their study of Vietnamese pre-schoolers. Children who started brushing at the

eruption of their first deciduous tooth had less decayed surfaces than children who started

brushing later (p = 0.009) (Ngoc et al., 2017), and lack of tooth brushing past the age of 2 years

was a strong risk factor for dental caries in 6-year-old Cambodian children (Turton et al., 2016).

These findings are underlined by the results of the present study where we found that children

who started brushing at a later age were more likely to acquire caries than children who initiated

brushing earlier.

Socio-economic disparities in oral health are considered as a valid measure of progress or

deterioration of overall health status, and provide key information for health policies (Capurro

et al., 2015). Annual household income is an absolute measure of SES (Bernabe et al., 2009).

Children with one or more carious lesions were associated with lower family income, and our

74

findings are in line with others (Jepsen et al., 2017; Schwendicke et al., 2015). We report that

in our study population, children who had families with an annual income of more than AUD

59,999 were 33% less likely to develop dental decay than their lower income counterparts

(Table 4.4). In the USA, income-related inequalities were observed in untreated dental caries

among 2- to 12-year-old children in National Health and Nutrition Examination Surveys done

over three decades. It was found that children from low income families were three to five

times more likely to have untreated decay than children from high income families (Macek,

2016). Children from ethnic minorities with low income levels were reported to be more at risk

of dental caries than native children (van der Tas et al., 2016). For instance, an indigenous

population in Brazil exhibited that high income was a “protective factor” for dental caries,

whereas children from low income ethnic minority groups were more than twice as likely to

develop dental decay (Arantes and Frazão, 2016).

Alarmingly, in a review paper published a decade ago, it was reported that in many countries

22% of 1- and 2-year-old children consume high sugar carbonated beverages at an average of

1 cup/day, and this increases through childhood to adulthood (Tahmassebi et al., 2006). In

addition, these levels are likely to have risen since such drinks have become more accessible

(Lee and Giannobile, 2016). Findings in different studies are diverse, due to variations in study

designs, techniques of statistical analysis, and phenotype definitions. For instance, among 6-

year-old Korean children (n = 1214), “mild decay” had no significant association with

consuming carbonated drinks, whereas consuming soda drinks once a day was a risk factor for

“severe decay” (Han et al., 2014). Juices, frizzy drinks and smoothies were reportedly

associated with children’s past caries history (p = 0.04), and the number of “early” carious

lesions (p = 0.04) in 6- to 15-year-old Spanish children (Llena et al., 2015). López-Gómez et

al., (2016), reported that loss of deciduous teeth increased with high consumption of soft drinks

75

(OR = 1.89, 95% CI = 1.13–3.16): the variable was dichotomized into “≤ 1 times/day and >1

times/day”, and similar to our study, questionnaires were administered to mothers of 6- to 7-

year-old Mexican children (n = 833) to retrieve the information (López-Gómez et al., 2016).

Other authors have reported that consumption of 100% juices were associated with lower caries

incidence (Ghazal et al., 2015). Our observation that children who consumed carbonated drinks

more than once a day did not have greater caries could in part be due to participating mothers

having given their children 100% juice instead of carbonated drinks, and/or coupling fizzy

drink consumption with frequent tooth brushing (Asif et al., 2015). According to a dissimilar

observation by Cheng and Campbell (2016), in a sample of rural Ugandan children aged 8-10

years (n = 84), a relationship between cariogenic beverages (soft drinks and sweetened tea) and

dental caries could not be discovered (Cheng and Campbell, 2016). In their study, the frequency

of cariogenic beverages was taken on a weekly consumption, and there was a greater chance

of recall bias in children which might have influenced the findings.

The current study reports that this sample of SEQ children, whose mothers had high caries

experience, were only 2% more at risk of developing decay than children with mothers with

low caries experience, an overall weak association (Table 4.4). Similar results were found in a

study of American Indian children (OR = 1.04, 95% CI = 1.02-1.07); however, the children

were 3-year-olds and the environmental context was very different (Warren et al., 2016).

Nevertheless, Ersin et al., (2016), in their study of Turkish children, reported a much stronger

association: children of mothers with high DMFS scores had almost double the caries

experience of those whose mothers had low caries experience (Ersin et al., 2005). Another

study reported that caries prevalence was 22.5 times higher in children whose mothers had

severely decayed teeth than children whose mothers had low caries experience (PR = 22.5,

95% CI = 3.2-156.6) (Proença et al., 2015). In Cambodia, where caries prevalence is very high

76

among preschool children, maternal caries was found to be a strong predictor (Turton et al.,

2015). Correspondingly, higher maternal DMFS could predict child caries in a population of

Hispanic and African-American children (OR = 3.9, 95% CI = 1.1-14.6) (Smith et al., 2002).

Moreover, a five year follow up study among Ugandan mother child dyads provided further

evidence that caretaker’s caries experience was positively associated with childhood caries

(IRR = 2.0, 95% CI = 1.3-3.0) (Birungi et al., 2016). Kawashita et al., (2009), however,

reported contrasting findings as maternal caries was not found to be a predictor in a population

of Japanese children, probably due to higher socio-economic factors and level of education of

mothers (Kawashita et al., 2009).

Interestingly, we found that children who had mothers with high LB levels were 39% less likely

to get caries compared to mothers with low LB levels (Table 4.4). At a superficial level this

association is counter-intuitive, given the oft reported positive association with an individual’s

number of carious lesions and high counts of lactobacilli in saliva (Ahirwar et al., 2017) relating

to the acidogenic, and especially to the aciduric, properties of LB in general (Samaranayake,

2012). It has been established that children acquire cariogenic bacteria predominantly from

their mothers, and bacterial transmission from mothers with tooth decay to their infants takes

place earlier than in families where mothers have healthier mouths (Li et al., 2005). According

to Mattsson et al., (2007), naturally occurring oral LB inhibits the growth of autologous MS,

and the observation was more marked in caries-free subjects who harboured LB that suppressed

the growth of host’s oral MS (Simark‐Mattsson et al., 2007). Furthermore, LB species have

been recognized as later invaders of cavity formation, and infrequent colonizers of enamel and

early caries lesions (Guo and Shi, 2013; Samaranayake, 2012). In other words, invasion of LB

was seen only at advanced stages of dental caries which involve cavity formation and

destruction of dentine (Dige et al., 2014). In our study, 61% of the children were caries-free,

77

and a further 16% had carious lesions limited to enamel. There is evidence that MS species

play a significant role in the initiation of caries on smooth surfaces, fissures of crowns and on

root surfaces, and the role of LB in inducing caries is not well supported (Samaranayake, 2012).

Taking into account the evidence from previous studies, our finding is rational; nevertheless,

the significant association we observed needs to be explored in depth in similar populations

with larger sample size.

Limitations of this study are the modest sample size, which was compromised by practical

factors like budget, time limits, and number of volunteers willing to attend the dental clinics

for oral examinations (Fernando et al., 2015).

In conclusion, amongst the maternal and environmental factors explored in our study, low

income levels of mothers and giving carbonated drinks significantly increased the risk of their

6- to 7-year-old children developing dental caries, while maternal caries had a weakly positive

association with children’s caries experience. Maternal income presumably is related to

education, to attitudes to oral hygiene behaviours, and to dietary practices that ultimately

influence caries experience. Maternal saliva LB levels were inversely related to caries

experience in this child population; hence that cannot be taken as a risk marker for children’s

caries experience.

78

Chapter 5: Mothers who report taking iron supplements during

pregnancy and child’s oral carriage of mutans Streptococci are

associated with a child’s lower and higher lifetime dental caries

experience, respectively.

This chapter is a submitted paper (paper 3). The bibliographic details of the paper, including

all authors, are as follows:

Surani Fernando, Santosh Kumar, Mahmoud Bakr, David Speicher1, Rodney Lea, Paul A

Scuffham, Newell W Johnson. Mothers who report taking iron supplements during pregnancy

and child’s oral carriage of mutans Streptococci are associated with a child’s lower and higher

lifetime dental caries experience, respectively.

My contribution to the paper involved the conception and designing of the study, data

collection, statistical analysis and drafting the article.

The manuscript was submitted to the Journal of Public Health Dentistry, and is under review.

Manuscript ID is JPHD-OA-08-17-0226.

Surani Fernando

Corresponding author: Surani Fernando

Supervisor: Professor Newell Johnson Supervisor: Professor Paul Scuffham

79

5.1 Abstract

5.1.1 Objective

This study explored the association of children’s salivary characteristics, birth weight, and

reported maternal prenatal vitamin and mineral supplementation with the dental caries

experience of the child.

5.1.2 Methods

This cross-sectional study, a sub-study of Griffith University Environments for Healthy Living

birth cohort study, was conducted on 174 mother-child dyads. Mother’s prenatal usage of

vitamin and mineral supplements; child’s birthweight; salivary pH, buffering capacity, and

levels of salivary MS and LB were explored as risk indicators. Dental caries experience was

assessed using International Caries Detection and Assessment System criteria. Path analysis

was conducted to evaluate the association of risk indicators with children’s current and past

dental caries experience.

5.1.3 Results

Children’s past caries experience (β = 0.332, p = 0.018), and salivary MS counts (β = 0.215, p

= 0.032) were positively associated with current caries experience. With a trend towards

significance, children whose mothers had reported taking iron supplements during pregnancy

experienced lower levels of past (β = -0.137, p = 0.068) and current dental caries (β = -0.046,

p = 0.051).

80

5.1.4 Conclusions

This study confirms that a child’s current dental caries status is positively associated with their

past caries, and that it correlates with current levels of salivary MS. Children of mothers who

reported to have taken iron supplements during pregnancy experienced less caries throughout

their lives. These observations confirm the importance to offspring of monitoring maternal

health throughout pregnancy and of early monitoring of children’s oral health in preventing

future dental disease.

Key words: Birth weight, Caries, Children, Past caries, Prenatal supplements, Saliva

5.2 Introduction

Dental caries is the most common oral disease in Australian children, and understanding the

risk indicators of cariogenesis ultimately assists with improved early-life prevention.

According to the Australian National Child Oral Health study (2012-2014), 27% of children

aged 5-10 years had untreated caries in their deciduous dentition (Do and Spencer 2016). Dental

caries progresses very rapidly in deciduous teeth as they have relatively thin enamel, and

dentine and pulp involvement can occur quickly (dos Santos Junior et al. 2014). For a child,

treatment of caries is a stressful and often painful experience which frequently requires multiple

visits, extensive restorations, and extractions. It is also expensive for parents, and an added

burden on health expenditure for many countries. Identification of the factors which influence

dental caries in children allows the family, perhaps with advice from a clinician, to practice a

healthy life style and, where indicated, for a clinician to institute preventive interventions such

as topical fluorides. The literature surrounding dental caries in children is overwhelming, with

81

numerous systematic reviews identifying socio-demographic, dietary and family circumstances

and behaviours being important predictors of disease. However, the association of children’s

individual characteristics, particularly in terms of their foetal well-being, (dos Santos Junior et

al. 2014) and features of their own oral physiology, have not been studied simultaneously (Do

and Spencer 2016).

Since tooth formation starts during the foetal period, the intrauterine environment, which is

affected by maternal nutrition and supplement intake during pregnancy, influences the

development of children's teeth (Tanaka et al. 2015). Studies have shown that nutritional

supplements during pregnancy, like vitamin D (Tanaka et al. 2015), calcium, and iron,

influences tooth formation in children (Bergel et al. 2010). Low birth weight, a marker of sub-

optimal foetal nutrition and growth, is indicative of an increased risk for ill health throughout

life (Barker and Thornburg 2013), including the development of dental caries (Bernabé et al.

2017). On the other hand, it has long been established that low salivary pH and poor buffering

capacity, and a comparatively high load of salivary MS) and LB (Samaranayake 2012), help

drive cariogenesis in the child (Lenander-Lumikari and Loimaranta 2000). Age in general, is a

risk indicator for dental caries (Hunter 1988) as the teeth of older children have been at risk for

a longer period (Al-Shalan et al. 1997). Previous studies have been inconsistent in their findings

on the relation between caries and gender, with no consistent pattern favouring higher caries

propensity in one gender (Chankanka et al. 2011). Importantly, past dental caries experience is

widely recognised as the strongest single predictor of future dental caries (Attaran et al. 2016).

This study explored the association of children’s salivary characteristics, birth weight, and

maternal prenatal vitamin and mineral supplementation with the dental caries experience of the

child.

82

5.3 Methods

This study followed a previously published detailed protocol regarding sample size, participant

recruitment, and data collection (Fernando et al. 2015). In brief, the study cohort consisted of

174 children aged 6-7 years and their mothers from Logan, Beaudesert, and Gold Coast in SEQ,

Australia, who were enrolled in the Griffith University EFHL birth cohort study (Cameron et

al. 2012). Participant enrollment for the present oral health sub-study started in November 2012

with children who were 6 years of age at the time, followed by participants from 2007, 2009

and 2010 cohorts. Data collection was concluded in April 2016.

The primary outcome variable was each child’s dental caries level at the time of examination

expressed as the number of tooth surfaces affected by caries, including initial white spot

lesions, according to the ICDAS criteria (CICDAS, 2005; Fernando et al. 2015). Past caries

experience was measured by the number of tooth surfaces that had restorations (Fernando et

al. 2015). Explanatory variables included reported maternal intake of nutritional supplements

(iron, Zn folate, and multivitamins) during pregnancy. Prenatal nutrition was evaluated by

reported maternal intake of supplements, which is considered to be a valid and an

uncomplicated method of obtaining such data (Schmidt et al., 2011). Although information

related to substance use (tobacco, alcohol, and illicit drugs) and calcium supplemets were

collected through the EFHL study, these variables could not be considered in the analysis as

most of the mothers participating in the oral health substudy had not answered the relevant

questions. Child’s salivary pH, buffering capacity, counts of MS and LB, past dental

experience, age, and gender were other independent variables under study. Salivary buffering

capacity was assessed using Saliva-check BUFFER kits (GC, Australia), salivary MS and LB

levels as measured with Caries Risk Test kits (Ivoclar Vivadent, Australia). Salivary data of

83

the children were collected during the oral health substudy (Fernando et al. 2015). Data on

maternal, iron, Zn, folate and multivitamin supplementation during pregnancy, and child’s

birth weight were collated from the main EFHL birth cohort database, and merged with the

oral health data (Cameron et al. 2012).

Mean salivary pH and buffering capacity, mean birth weight and past caries experience were

compared between children with no current or past caries (ICDAS = 0) and those with any

caries experience (ICDAS > 0). Moreover, the percentage of mothers who have reported to

taking nutritional supplements during pregnancy, proportions of children with low and high

salivary bacterial levels, were compared between the two groups of children.

Data analysis was performed using SPSS-21 (IBM, Chicago, IL) and AMOS (IBM SPSS

AMOS 22.0, Meadville, PA). Measures of central tendency for continuous variables and

frequency and percentages for categorical variables were computed. Normality of the data was

assessed using the Shapiro-Wilk test. As the data were not normally distributed, non-parametric

tests were used for univariate analysis. Mann-Whitney U test was used to compare the mean

age, past caries experience, and salivary pH between children with caries and those with no

caries. For comparison of proportions, Fisher’s Exact and Chi-square tests were used according

to the type of data. Based on the research hypothesis, path analysis, which describes the directed

dependencies among a set of variables, was used as it facilitated simultaneous assessment of

associations between several independent and dependent variables (Kumar et al. 2017). In the

path analysis, whether or not mothers took the particular nutritional supplements and birth

weight of the particular child, were hypothesised to have a direct relationship to the child’s

cumulative past dental caries experience and both direct and an indirect association to current

caries experience. Moreover, while a child’s current salivary characteristics, age, and gender

84

were hypothesised to have direct association with current caries levels (figure 5.1). In addition,

past dental caries experience was hypothesised to have a relationship to current dental caries

experience. All the variables related to maternal intake of supplements and child’s salivary

characteristics were allowed to covary in the path analysis. The model has 2 endogenous

(dependent) and 11 exogenous (independent) variables. The minimum required sample size for

the path model, as determined by N > 50 + 8m where N is the number of participants and m is

the number of predictor variables, with 12 predictors, was 146 children. As two variables (past

and present caries experience) deviated significantly from normality, Bollen-Stine

bootstrapping was used to estimate the fit indices (Kim and Millsap 2014). Further,

bootstrapping and bias-corrected confidence intervals were used to analyse direct and mediated

parameter estimates, and the bootstrapping sample was set to the generally accepted value of

250 (Nevitt and Hancock 1998). The model was considered to have a good fit if Root Mean

Square Error of Approximation (RMSEA) and Standardized Root Mean Square

Residual (SRMR) were < 0.08, and Comparative Fit Index (CFI) and Tucker Lewis Index

(TLI) were ≥ 0.9 (Hoyle and Panter 1995).

85

Table 5.1: Children’s individual characteristics explored as risk indicators for dental

caries experience and current activity

Variable Cases (n = 67) Controls (n = 107) Significance

Mean (SD) Mean (SD) t (p value)

Age at examination 6.07(1.02) 6.49(0.65) 2.6 (0.09)

Saliva pH 7.17(0.45) 7.26 (0.49) 1.3 (0.19)

Saliva buffering capacity (*GC) 8.26(3.85) 9.41(3.15) 1.4 (0.16)

Child’s birthweight (g) 3438 (438.82) 3487(481.85) 0.8 (0.42)

Past caries experience (number

of surfaces with restorations)

2.10 (5.52) 0.21 (0.86) 4.1 (0.0001)

Current caries experience

(number of surfaces with caries)

8.26 (9.96) 0 8.9 (0.0001)

N (%) N (%) X2 (p value)

Gender 2.75 (0.25)

Female 37(55) 51 (48)

Male 30(45) 56 (52)

Saliva �MS levels 8.33 (0.004)

<105 CFU/ml of saliva 52 (78) 60 (56)

>105 CFU/ml of saliva 15 (22) 47 (44)

Saliva ∞LB levels 0.14 (0.71)

<105 CFU/ml of saliva 37 (55) 56 (52)

>105 CFU/ml of saliva 30 (45) 51 (48)

Maternal Folate

supplementation during

pregnancy

0.27 (0.61)

No 17 (25) 31 (29)

Yes 50 (75) 76 (71)

Maternal Iron

supplementation during

pregnancy

0.001 (0.97)

No 28 (42) 45 (42)

Yes 39 (58) 62 (58)

Maternal Multi Vitamin

supplementation during

pregnancy

3.8 (0.05)

No 23 (34) 53 (49)

Yes 44 (66) 54 (51)

Maternal Zn intake during

pregnancy

0.32 (0.57)

No 46 (69) 69 (64)

Yes 21 (31) 38 (36)

*Saliva-check BUFFER kits (GC, Australia), � mutans Streptococci, ∞Lactobacilli

86

Table 5.2: Standardised boot strapped estimates with 95% confidence intervals of the

path model with past and current dental caries experiences as the endogenous variables

Effect β Bias corrected

95% CI

P

DIRECT EFFECTS

Predicting past dental caries

Maternal Zn supplementation during

pregnancy 0.132 -0.066 - 0.298 0.210

Maternal Folate supplementation during

pregnancy 0.042 -0.089 - 0.140 0.582

Maternal Iron supplementation during

pregnancy -0.137 -0.241- 0.018 0.068

Maternal Multi vitamin supplementation

during pregnancy 0.076 -0.076- 0.234 0.339

Child’s birth weight -0.048 -0.151-0.058 0.303

Predicting current caries

Age 0.006 -0.167-0.141 0.95

Gender 0.036 -0.11-0.181 0.566

Maternal Zn intake during pregnancy 0 -0.175-0.211 0.977

Maternal Folate intake during pregnancy 0.026 -0.231-0.208 0.911

Maternal Iron intake during pregnancy 0.013 -0.164-0.26 0.882

Maternal Multi vitamin intake during

pregnancy -0.054 -0.219-0.107 0.585

Child’s birth weight -0.005 -0.178- 0.145 0.839

Child’s past caries 0.332 0.131- 0.503 0.018

Child’s Salivary pH 0.086 -0.008-0.19 0.06

Child’s Salivary MS 0.215 0.02-0.382 0.032

Child’s Salivary LB 0.097 -0.067-0.224 0.254

Child’s Buffering capacity (*GC) -0.034 -0.219-0.131 0.722

INDIRECT EFFECTS

Predicting current caries

Iron -0.046 -0.1 – 0.001 0.051

Quantified influence not mediated by other variables in the model, Chi-square = 83.730, p = 0.348, TLI – 0.954,

CFI – 0.969, RMSEA – 0.028 and SRMR = 0.0592, *Saliva-check BUFFER kits (GC, Australia)

87

5.4 Results

Of the children enrolled, 88/174 (51%) were girls. The mean age for children with caries was

6.1 years (SD = 1.02) and for children without caries was 6.5 years (SD = 0.65). At time of

examination, 67/174 (38%) children had caries, of which 37/67 (55%) were girls. Mean number

of tooth surfaces affected with caries in the total sample of children at the time of examination

was 5.44 (SD = 8.95), and mean number of tooth surfaces with past caries was 1.37 (SD =

4.45). In the subset with caries the mean was 8.26 (SD = 9.96). Children with at least one

carious tooth surface currently, had significantly greater caries experience in the past (Mean:

2.1, SD = 5.52, p = 0.0001) than those who were completely caries free (Mean: 0.21, SD =

0.86, p = 0.0001) at the time of examination (Table 5.1). Salivary loads of MS (p = 0.005) and

reported maternal multivitamin intake during pregnancy (p = 0.05) were significantly different

between caries and caries free groups in univariate analysis, even though reported multivitamin

intake during pregnancy was not associated with caries experience of children in the path

analysis. Our hypothesised model, which provides estimates of the magnitude and the

significance of association between explanatory and the outcome variables, demonstrated a

good fit with the data (Chi-square = 83.730, p = 0.348, TLI = 0.954, CFI = 0.969, RMSEA

=0.028 and SRMR = 0.0592) and shows a relationship and degree of association between

dependant and independent variables (figure 5.1). Table 5.2 gives the 95% CI and the level of

significance (p) against the β values (associations between independent and dependant

variables) obtained and indicated in figure 5.2 There was a significant association between the

past and current dental caries experience (β = 0.332, p = 0.018), with children who had more

carious tooth surfaces in the past tending to have a greater number of new carious surfaces at

the time of examination (Table 5.2). A positive association was also observed in path analysis

between child’s salivary MS levels and current dental caries experience (β = 0.215, p = 0.032).

88

Although only approaching statistical significance, children whose mothers reported to have

taken prenatal iron supplements had fewer tooth surfaces affected with caries in the past (β = -

0.137, p = 0.068). Maternal intake of iron supplements during pregnancy was indirectly

associated with current dental caries experience with marginal significance (β = -0.046, p =

0.051), while there was no association between reported intake of Zn, and folate supplements

during pregnancy and dental caries experience. Loads of LB in saliva and other salivary

characteristics were not associated with current dental caries experience in this child

population.

89

Figure 5.1. Path analysis with standardised bootstrapped estimates (β) of the associations

between maternal intake of nutritional supplements during pregnancy, salivary characteristics,

age, and gender with dental caries experience of children. (Significant standardised estimates

(*p ≤ 0.05) are in bold font. Error terms and covariances between exogenous variables are not

presented for ease of understanding. Β values of the associations are written on the connected

line arrows.

Maternal Multi-vitamin intake in Pregnancy

Maternal Folate intake in Pregnancy Age at examination

Maternal Zn intake in Pregnancy

Maternal Iron intake in Pregnancy

Gender

Saliva pH

Saliva MS

levels

Saliva buffering capacity

-0.05

Saliva LB

levels

Birth weight

Past caries experience Current caries experience

0.04

-0.05

-0.005

0.33*

-0.03 0.10

0.22*

0.09

0.006

0.04

0.03

0

0.01

0.08 -0.14

0.13

90

5.5 Discussion

The present study confirms that children with a history of dental caries are more at risk of

developing future caries. This is consistent with much of the published literature. A recent

systematic review reports that baseline caries was the strongest predictor for future dental caries

in very high human development countries (Attaran et al. 2016). Similarly, progression of

carious lesions was positively associated with baseline caries among a population of Brazilian

children (Piva et al. 2017). Furthermore, according to Brennan and Spencer, greater caries

experience at age 13 was associated with more missing or filled teeth at 30 years of age among

Australians (Brennan and Spencer 2014). These findings emphasise the importance of early

and regular screening for caries and associated risk indicators in families so that appropriate

preventive approaches to control future disease can be put in place. This is best achieved by

educating families in oral hygiene and healthy eating habits, although occasional referral to a

clinician for preventive interventions such as topical fluorides and/or fissure sealants may be

necessary.

Both longitudinal and cross-sectional studies have shown that their oral MS levels are

positively associated with caries experience in children (Piva et al. 2017). In accordance, we

found that children who had high salivary MS counts (> 105 cfu/ml saliva), were more likely

to have greater caries experience than children who harboured lower MS counts (< 105 cfu/ml

of saliva). Levels of MS in saliva were identified as a useful and a quick method to evaluate

caries risk among a small sample of children from India, where mean MS scores for children

with caries were significantly higher than for children without caries (p ≤ 0.001) (Nanda et al.

2015). From a cohort of American 6-year-old children, Edelstein et al. reported that even within

families with high-income and who were accessing preventive care, the most useful predictor

91

of current caries was salivary MS count (Edelstein et al. 2016). Contrastingly, Widita et al.

found that the predictive value of MS counts for scores of dental caries among Indonesian pre-

schoolers was high only when combined with other predictive factors like cavitated teeth,

unhealthy feeding practices, and poor oral hygiene, demonstrating the importance of

identifying risk indicators within individual families and communities for effective public

health initiatives (Widita et al. 2017).

We have observed that the children of mothers who took iron supplements during pregnancy

had comparatively low levels of dental caries at age approximately 6 years. Supplementation

with iron is generally recommended during pregnancy to meet the iron needs of both mother

and foetus (Scholl 2005); there can be depletion of iron stores during pregnancy and oral

supplements can prevent maternal iron stores from reaching deficient levels (Fenton et al.

1977) although there was no way of knowing whether nutritional supplements taken by any

individual pregnant mother were prescribed because of a measured deficiency, as routine

treatment or were self-administered. Early iron supplementation, even during foetal period and

in breastfed human infants, is feasible and increases iron status which is carried into early

childhood (Lima et al. 2006). A possible mechanism supporting our observation was proposed

by Alves et al, where they demonstrated in an in vitro study that higher concentrations of Iron

reduced the demineralization of enamel (Alves et al. 2011). Martinhon et al, reported similar

findings in bovine enamel. Their results showed that ferrous sulphate reduced the

demineralization of enamel blocks and altered the ionic composition of the dental biofilm

formed in situ (Martinhon et al. 2006). A recent randomized control study investigating the

effect of ferrous sulphate on deciduous teeth have found that ferrous sulphate indeed reduces

the demineralization of enamel and has a cariostatic effect (De Gauw et al. 2017).

92

Moreover, iron supplementation could be a proxy or marker for a wider set of psychosocial

and health behavioural variables which are associated with childhood caries. For instance,

serum haemoglobin levels, mean corpuscular volume and packed cell volume (Haematocrit)

have been shown to be significantly low in children with caries compared to caries free children

in studies from India, with a low socio economic background (Bansal et al. 2016) and in a

similar background in Egypt (Abed et al. 2014). Tang et al. (2013) reported that severe early

childhood caries was strongly associated with anaemia of the child, which could be because of

significant illness of the child or, again, be a result of poor diet and related to economic status

(Tang et al. 2013)

Our univariate analysis showed that, reported maternal multivitamin intake during pregnancy

showed a significant difference between children with and without caries, though a significant

association could not be found in path analysis. Even though this is worth investigating further

we cannot overlook the fact of ambiguity in the data. Perhaps some of this variable is correlated

strongly with other variables that are in the final model and have been masked by other

variables. For example reported intake of other supplements. This is hardly surprising as the

types of vitamins included in the multivitamin “pill” or “capsule” were not asked in the

questionnaire.

Our study has some limitations. Participants were volunteers and may not be representative of

the general population. Further, information on prenatal supplements is as reported by

participants themselves, and serum levels were unavailable. The study was cross-sectional,

hence the associations found cannot be presumed as causal. Moreover, past caries experience

was assessed based on the number of surfaces with restorations at examination; thus, the

periods of life during which a child had active caries is not known, and may not be comparable.

93

5.6 Conclusions

This study confirms that a child’s current activity of dental caries is positively associated their

past caries experience, and both are likely to relate to future caries unless there is improved

behaviour and/or preventive interventions. It also confirms that current levels of salivary MS

are associated with both past and present levels of caries, and likely also future disease. Mothers

who have reported to taking nutritional supplements throughout pregnancy could be associated

with lower caries experience in their 6-year-old child, consistent with the Barker hypothesis,

in that a healthy foetus predisposes to a healthy child, and vice versa. This study is the first to

explore associations between mothers’ reported intake of nutritional supplements during

pregnancy and their dental caries experience in a path model adjusting for the effects of salivary

and demographic variables. Further longitudinal studies are required to explore if these

associations have true predictive value and if any of the attributes studied might be regarded as

part of the causal chain of disease, and can be regarded as true risk factors.

These findings may be typical of Western industrialised countries and indicate the importance

of promoting good family health throughout the lifespan, including pregnant mothers.

94

Chapter 6: Influence of genes on the process of dental caries and

what lies beyond

6.1 Introduction

The purpose of this chapter is to review the literature on the influence of genetic and epigenetic

contributions to susceptibility to dental caries

The organization is as follows. Section 6.2 outlines the evidence reported over the last few

decades on the genetic influences on dental caries; particular attention is paid to research

conducted with humans irrespective of age, gender, and other population characteristics.

Section 6.3 briefly discusses the types of molecular genetic studies conducted and their

outcome in relation to susceptibility to dental caries, paying attention to possible biological

pathways that promote or prevent progression of the disease. Section 6.4 briefly describes

possible reasons why a dominant genetic influence could not be observed with dental caries

and describes the landmark twin studies. Section 6.5 briefly reviews the field of epigenetics

with an emphasis on mechanisms of DNA modification. Section 6.6 focuses on potential

environmental stimuli that might initiate or inhibit DNA modifications and eventually

influence dental caries. Section 6.7 describes the importance of future research on epigenetics

in relation to dental caries, acknowledging the dearth of empirical research in the area.

95

6.2 Evidence that the host genome influences the natural history of dental

caries

Following completion of the Human Genome Project in 2003 (www.genome.gov), interest

grew to understand the underlying genetic mechanisms influencing susceptibility to dental

caries. The high prevalence of caries among certain ethnic groups and observations that

fluoride exposure is not protective for all individuals motivated research seeking possible

genetic contributors (Slade et al., 2013; Vieira et al., 2014). Whilst genome-wide and candidate

gene studies have delivered some successes, they have also exposed challenges, such as the

need for robust phenotype definitions and sample sizes that allow enough statistical power to

detect modest genetic effects.

In 1988, Borass et al. showed evidence of a genetic contribution to caries susceptibility by

studying twins raised together and apart (Boraas et al., 1988). More recent studies involving

both monozygotic (MZ) and dizygotic (DZ) twins (Bretz et al., 2005; Bretz et al., 2006), and

family studies (Shaffer et al., 2015; Wang et al., 2010), estimated the heritability component

(proportion of phenotype variation due to genetic factors) for caries experience, measured by

decayed, missing, and filled teeth/surfaces index (dmft/s, DMFT/S) as ranging from 45%-64%

in the deciduous dentition, and an apparently lower heritability of 35%-55% in the permanent

dentition. However, heritability studies alone cannot fully quantify a genetic component in a

disease because shared behaviour and environmental factors contribute to covariance within

families and populations.

There are also important methodological considerations. On the one hand it is currently

unknown which methods of quantifying dental caries best reflect underlying genetic risk, so

96

research to identify phenotypes associated with specific genetic information is important

(Shaffer et al., 2013; Wang et al., 2010). On the other hand, better understanding of the structure

and function of DNA has generated a new era in genetics, namely the field of epigenetics. This

is now emerging as a significant platform in searching for answers to questions that were left

blank with earlier genetic studies (Seo et al., 2015). Electronic databases Medline via PubMed,

Scopus and Google scholar were used to search literature on genetic and epigenetic influences

on caries. The search strategy applied is described in Appendix C Table 1. The following is a

review of the cariology literature involving molecular genetic studies, and it emphasises how

epigenetics, through environmental modifications of the host genome, might ultimately be

important in determining risk and in preventing dental caries.

6.3 Types of molecular genetic studies

There are two main types of molecular genetic studies: genome-wide studies and candidate

gene studies (Vieira et al., 2014).

6.3.1 Genome-wide Studies

6.3.1.1 Genome-wide linkage studies

Genome-wide linkage studies constitute a powerful tool to investigate the genetic construction

of complex diseases (Hardy and Singleton, 2009; Manolio et al., 2009) and are based on

exploring for segregation of alleles within a recorded pedigree, investigating the transmission

to offspring of alleles that are physically close or linked to each other in relation to disease

traits within families (Kemper et al., 2012). In respect to the caries process, published literature

has shown three gene loci (5q13.3, 14q11.2, and Xq27.1) associated with low caries experience

97

(DMFT 1 and 2) implying a degree of resistance, and two loci (at 13q31.1 and 14q24.3)

associated with high caries experience (DMFT 3 and 4), implying susceptibility in such

individuals (Riley et al., 2007; Vieira et al., 2008; Wang et al., 2013a). The former loci

appeared to be associated with the functional properties of a psycho-stimulant; the latter pair

with taste and flavour (Vieira et al., 2008), attributes which, intuitively, would influence diet,

especially the intake of sugars (Table 6.1). However, a strong and statistically significant

association with dental caries experience was not found, perhaps because the separation of the

disease groups was not marked.

6.3.1.2 Genome-wide association studies

Genome-wide association studies (GWAS) search for associations between genetic variants

and phenotypic differences within a population and analyse genome-wide structural variations,

which are detected by screening the whole genome (Kemper et al., 2012). Several GWAS have

been performed on dental caries (Table 6.1). In 2012, Shaffer et al. reported a GWAS screening

study related to caries experience in the deciduous dentition of 3- to 12-year-old children in the

USA. That study found several genomic regions suggestive of association, although no specific

loci were statistically significant (Shaffer et al., 2011). Vieira et al. subsequently found loci not

previously described which did have a significant association with disease history in the

deciduous dentition (Vieira et al., 2014). In 2012, Wang et al. published a GWAS in relation

to experience of dental caries in the permanent dentition, and although, again, no statistically

significant associations were found, several “statistically suggestive” loci were identified

(Wang et al., 2012a). With adults, advancing age increases the influence of behavioural

(including diet and oral hygiene) and environmental influences, and confounding diseases such

as diabetes, perhaps operating over decades, will all have cumulative effects which may

overshadow genuine genetic effects (Wang et al., 2012b). As is evident from the results

98

presented by Shaffer et al. (2012), where the heritability of caries scores in the permanent

dentition was 35%-55% (p < 0.01) compared to 70% in deciduous dentition (Shaffer et al.,

2012a). However it is possible that different caries phenotypic definitions might have given the

observed variations in results as indices like DMFT might not be sensitive enough to precisely

detect these effects. Moreover, the different biological samples (blood, mouthwash, buccal

swab, saliva) used to obtain DNA, and various genotyping techniques used might have given

dissimilar study findings.

99

Table 6.1 A summary of genome-wide linkage and genome-wide association studies for human dental caries

Reference Study design and study

population

Phenotype Results

Suggestive loci (gene) Significant loci (gene)

Genome-wide linkage studies

Vieira et al. , 2008 Cross sectional

642 Philippine individuals

from 46 families

Decayed Missing Filled Teeth

index (DMFT), permanent

dentition

For low caries experience:

5q13.3, 14q11.2,

Xq27.1

For high caries experience:

13q31.1, 14q24.3

-

-

Genome-wide association studies

Shaffer et al., 2011 Cross sectional

1305 US children aged 3-12

years

decayed missing filled teeth

index (dmft), primary dentition

1q42–q43 (ACTN2, MTR,

EDARADD), 11p13

(MPPED2), 17q23.1 (LPO)

-

Wang et al., 2012a Cross sectional

7443 adults from US

DMFT, permanent dentition 1q42.11–q42.3

(RHOU), 4q13.3

(ADAMTS3), 8p21.1

(PTK2B)

4q32 (TLR2), 5q11.1 (ISL1), 6q27

(RP56KA2), 7q21 (FZD1), 14q22

(CDKN3, CNIH, GMFB,

CGRRF1, BMP4)

Shaffer et al., 2013

Cross sectional

920 US adults aged 17-75

years

Decayed Missing Filled

Surface index (DMFS) scores

1-5, permanent dentition

DMFS2: 4q31.22 (EDNRA)

DMFS5:

1p36 (SPSB1),

2p24 (TRIB2),

4p15 (PROM1), 4q22

(ABCG2, PKD2,

SCPP), 4q31.22

(EDNRA), 7q22

(ATXN7), 17q11

(WSB1)

DMFS2: 10p11.23 (LYZL2)

DMFS5: 1p36 (AJAP1)

100

Reference Study design and study

population

Phenotype Results

Suggestive loci (gene) Significant loci (gene)

Zeng et al., 2013 Cross sectional

1017 US adults aged 14-56

years

DMFS scores-pit and fissure

and smooth surface caries in

permanent dentition

For pit and fissure:

7p15 – 13 (INHBA), 8q21.3,

Xp11.4 (BCOR)

For smooth surface:

2q35 (CXCR1,

CXCR2), Xq26.1

(BCORL1)

Smooth surface: 8q21.3 (no gene)

Zeng et al., 2014 Cross sectional

1006 US children aged 3-12

years

dfs scores-pit and fissure and

smooth surface caries in

deciduous dentition

For pit and fissure:

11p14.1 (MPPED2)

For smooth surface:

1p36 (AJAP1),

6q27 (RP56KA2),

16p11.2 (ITGAL),

20q11.21 (PLUNC)

Pit and fissure: 3q26.1 (KPNA4)

Genome-wide association with gene set enrichment Gene sets Gene sets for;

Wang et al., 2013b Cross sectional

1142 US children aged 3-12

years

dfs scores in deciduous

dentition

Several suggestive gene sets,

refer Wang et al, 2013 (61).

I. cytokine secretion (18, including

INS)

II. ligase activity forming carbon

nitrogen bonds (68, including

WWP2, RNF217)

III. ubiquitin protein ligase activity (49)

IV. protein secretion (32)

V. regulation of protein secretion (22)

VI. regulation of cytokine secretion (16)

VII. regulation of axonogenesis (10)

VIII. regulation of neurogenesis (14,

including ROBO2, SLIT2)

IX. central nervous system

development (122)

X. small conjugating protein ligase

activity (51)

XI. glycoprotein catabolic process (12)

XII. axonogenesis (43)

XIII. cell matrix junction (16; includes

ACTN2)

GWAS on dental caries can be accessed at https://www.ebi.ac.uk/gwas/search?query=dental%20caries

101

6.3.2 Candidate gene studies

Selection of candidate genes is based on extant knowledge of the biology of the caries process,

and are, therefore, limited to genes that have already been identified and characterised. There

are several advantages to this approach as these studies may provide adequate power to detect

relative low risk for dental caries (Daly and Day, 2001). As most candidate gene studies focus

directly on a single gene and frequently look directly at functionally significant

polymorphisms, concerns about the extent of linkage disequilibrium and the adequacy of SNP

markers to detect associations are not relevant (Daly and Day, 2001). Different expressions of

dental caries phenotypes between candidate gene studies and the diversity of populations

studied, however, make it difficult to generalize the findings.

Major candidate gene categories reported to date in relation to the process of dental caries

include genes involved in enamel formation, immune response genes, genes related to salivary

function, and genes related to taste (Kutsch et al., 2013; Opal et al., 2015; Vieira et al., 2014)

(Table 6.2). Since these candidate genes are associated with the process of dental caries,

researchers have shown interest in investigating several of these gene loci simultaneously in

their respective studies. However most of these associations have not been replicated in each

independent study, perhaps due to population differences, contrasting case definitions, or issues

with sample size and statistical power.

102

Table 6.2 A summary of candidate genes studies for human dental caries

Gene Gene Function Results References

Enamel formation genes

Tuftelin (TUFT1) Expressed in the developing and

mineralized tooth and non-

mineralizing soft tissues

Associated with higher caries experience;

this association can be dependent of the

presence of Streptococcus mutans

Slayton et al., 2005, Deeley et al. 2008,

Patir et al., 2008, Shimizu et al., 2012

Amelogenin (AMELX) Involved in the mineralization

during tooth enamel development

Associated with higher caries experience Deeley et al., 2008, Patir et al., 2008,

Kang et al., 2011, Shimizu et al., 2012,

Slayton et al., 2005, Olszowski et al.,

2012

Enamelin (ENAM) Involved in the mineralization and

structural organization of the

enamel

Associated with higher caries experience Patir et al., 2008, Shimizu et al., 2012,

Slayton et al., 2005, Olszowski et al.,

2012

Tuftelin-interacting

protein 11 (TFIP11)

Interact with tuftelin and can play

a role in spliceosome disassembly

in Cajal bodies

Associated with initiation of carious lesions

and higher

caries experience

Shimizu et al., 2012 , Slayton et al., 2005,

Deeley et al., 2008, Patir et al., 2008,

Shimizu et al., 2012

Ameloblastin (AMBN) Involved in enamel matrix

formation and mineralization

Associated with higher caries experience Patir et al., 2008, Shimizu et al., 2012,

Slayton et al., 2005, Deeley et al., 2008

Matrix metalloproteinase

20 (MMP20)

Degrades amelogenin Associated with higher caries experience in

Whites with poor oral health habits

Tannure et al., 2012b

Matrix metalloproteinase

13 (MMP13)

Involved in endochondral

ossification and bone remodelling

Associated with lower caries experience Tannure et al., 2012a

103

Gene Gene Function Results References

Immune response genes

CD14 molecule (CD14) Mediates innate immune response

to bacterial lipopolysaccharide

Absent in the saliva of individuals with

active carious lesions

Bergandi et al., 2007

Human leukocyte antigen;

major histocompatibility

complex, class II, DR beta

1 (HLA-DRB1) and DQ

beta 1 (HLA-DQB1)

Presents peptides derived from

extracellular proteins

Frequency of allele 4 of DRB1 is increased

in children with early childhood caries; also

allele 2 of DQB1 is increased in adolescents

affected by caries; DRB1 allele 1 and DQB1

allele 3 frequencies are increased in the

presence of Streptococcus mutans

Altun et al., 2008, Bagherian et al., 2008,

Valarini et al., 2012, Wallengren et al.,

1997

Beta defensin 1 (DEFB1) Implicated in the resistance of

epithelial surface to microbial

colonization

Distinct DEFB1 haplotypes are associated

with low and high caries experience

Ozturk et al., 2010, Krasone et al., 2014

Lactotransferrin (LTF) Major iron-binding protein in milk

and body secretions with

antimicrobial activity

Associated with lower caries experience

Azevedo et al., 2010

Mucin 7 (MUC7) Facilitates the clearance of

bacteria in the oral cavity

Associated with higher caries experience

with poor oral hygiene

Pol, 2010

Mannose-binding lectin

(protein C) 2, soluble

(MBL2)

Recognizes mannose and N-

acetylglucosamine on many

microorganisms

Associated with higher caries experience

Olszowski et al., 2012

Genes related to saliva composition and function

Carbonic anhydrase VI

(CA6)

Maintains pH homeostasis in body

tissues

No association with caries but positive

association with buffer capacity

Peres et al., 2010

Proline-rich protein

HaeIII subfamily 1 (PRH1)

Involved in oral biofilm formation,

tissue

Homeostasis and immunological

investigation

Associated with higher caries experience

and

colonization by Streptococcus mutans

Zakhary et al., 2007

Cysteine rich glycoprotein

(gp-340 I)

Adhesion of MSto the dental bio

film

Associated with high caries experience Jonasson et al., 2007

104

Gene Gene Function Results References

Basic proline rich proteins

(PRB1 and PRB2)

Regulates plaque pH Associated with low caries experience Ayad et al., 2000

TIMP metallopeptidase

inhibitor 2 (TIM2)

Important to the maintenance of

tissue homeostasis by suppressing

the proliferation of quiescent

tissues in response to angiogenic

factors, and inhibit protease

activity in tissues undergoing

remodelling of the extracellular

matrix.

No evidence of association

Tannure et al., 2012a

Genes related to taste sensation

Taste receptor, type 2,

member 38 (TAS2R38)

Controls the ability to taste

glucosinolates

Associated with lower caries experience Wendell et al., 2010, Pidamale et al.,

2012

Taste receptor, type 1,

member 2 (TAS1R2)

Sweet taste receptor Associated with both lower and higher caries

experience

Wendell et al., 2010, Kulkarni et al., 2013

Guanine nucleotidebinding

protein, alpha

transducing 3 (GNAT3)

Involved in dietary preferences

No evidence of association

Wendell et al., 2010

Solute carrier family 2

(facilitated glucose

transporter), member 2

(SLC2A2)

Mediates bidirectional glucose

transport

Associated with higher caries experience

Kulkarni et al., 2013

Taste receptors TAS1R2 and

TAS1R3

Sweet taste receptors Associated with high caries experience Fushan et al., 2009

105

6.3.2.1 Enamel formation genes

Calcium phosphate hydroxyapatite crystals form the bulk of enamel. Enamel formation is

regulated through the interaction of a number of organic matrix molecules including

amelogenin, enamelin, ameloblastin and tuftelin. The amelogenin gene (AMELX) is located

on the p arm of the X chromosome, at Xp22.31-p22.1 and promotes the formation of a scaffold

for enamel crystallites and controls their growth (Patir et al., 2008; Rauth et al., 2009). The

ameloblastin gene (AMBN), which controls the functions of ameloblastin, is nested in

chromosome 4q21 between D4S409 (4q13) and D4S400 (4q21) (MacDougall et al., 1997).

Ameloblastin is a vital adhesion molecule for enamel formation, while playing an important

role in binding and maintaining the secretory ameloblasts (Sasaki et al., 2008). Tuftelin plays

a vital role during the development and mineralization of enamel, facilitating the initial stages

of enamel mineralisation (Deutsch et al., 2002). Functions of this phosphorylated glycoprotein

are regulated by TUFTI gene and an over expression of the gene will cause imperfections in

both enamel prisms and crystallite structure (Luo et al., 2004), making the enamel vulnerable

to acid challenges causing demineralisation (Cai et al., 2007). However, Shimizu et al. (2012)

reported that variations in TUFT1 were associated with the ability of the enamel surface to

uptake fluoride in very low concentrations, thus lowering the individual susceptibility to

demineralization at subclinical levels (Shimizu et al., 2012). A systematic removal of the

organic matrix and replacement with mineral is fundamental to generate an enamel layer that

is harder and unstained by retained enamel protein. Matrix metalloprotease 20 (regulated by

MMP-20) and kalikrien 4, (regulated by KLK4 gene), synchronize this process, making the

enamel layer harder, less porous, and unblemished by retained enamel proteins (Lu et al.,

2008). Hence, defects of these genes can result in the fabrication of abnormal proteins or reduce

the amount of MMP-20 and KLK 4 in developing teeth. Defective mineralisation caused by

106

these malformations could influence both bacterial adherence and resistance of enamel to acid,

increasing the susceptibility of tooth surfaces to dental caries (Opal et al., 2015).

Shimizu et al. (2012) showed that variations in AMELX, TUFT1, AMBN, and ENAM have

associations with caries experience by studying microhardness changes under simulated

cariogenic challenges (Shimizu et al., 2012). They found that the frequency of T allele of

AMELX rs946252 and C allele of AMBN rs4694075 was significantly higher in a group of

Filipino families with high caries experience. Comparable results were observed in a Korean

population, where two SNPs of AMELX; rs5933871 (p = 0.02) and rs5934997 (p = 0.02) were

significantly associated with increased dental caries experience in individuals living/lived in

fluoridation areas (Kang et al., 2011). Genotype of the tuftelin rs3790506 marker was reported

to be overrepresented in cases with higher dmft scores especially in the presence of MS (Patir

et al., 2008). Similarly, Slayton et al. (2005) observed that tuftelin, a gene involved in enamel

mineralization, when combined with high levels of MS increased the susceptibility of dental

caries significantly (R2 = 0.268) and they were able to explain 27% of the variation in the dmfs

scores by this interaction (Slayton et al., 2005). Deeley et al. (2008), on the other hand, reported

an association between the tuftelin-1 rs2337360 marker and high caries experience only when

individuals with DMFT≤ 2 were compared with DMFT ≥ 3 (p = 0.03), ≥4 (p = 0.02), ≥ 5 (p

= 0.042), and ≥ 6 (p = 0.049) (Deeley et al., 2008). Further, polymorphism in MMP-13 (OR =

0.538, 95% CI = 0.313-0.926) revealed a significantly reduced caries risk, and the result

remained significant even after adjusting for other candidate genes, type of dentition, and

dietary factors in a population of children and adolescents 3- to 21-years-old (Tannure et al.,

2012b). In addition, Shaffer et al. showed that, candidate genes have an effect on enamel

according to the surface features of teeth, such as smooth surface or pits and fissures (Shaffer

et al., 2012b). They observed that heritability score for caries in pit and fissure (h 2 = 19–53%;

107

p < 0.001) and smooth surface caries (h 2 = 17–42%; p < 0.001) in deciduous teeth was more

than for their permanent counterparts. A suggestive association was observed on chromosome

Xq26.1 (gene BCORL1) with smooth surface caries and BCOR with pit and fissure caries

(Zeng et al., 2013). Both are x-linked genes with sequence similarity, providing evidence for

the hypothesis proposed by Vieira et al. (2008) that x-linked genetic variants may partly explain

gender differences in dental caries susceptibility (Vieira et al., 2008; Zeng et al., 2013; Zeng et

al., 2014).

These studies report the role of specific genes in increasing susceptibility to caries, as well as

differential effects on the two dentitions and on specific surfaces. However, the complexity in

dental caries is further complicated by genetic phenotypes, which manifest differently in

different populations, races, dentition, or surfaces being studied. Most studies have compared

individuals who are caries free to individuals with one or more tooth surfaces affected, meaning

that extremes of disease experience, and thus presumptively of disease susceptibility, would be

difficult to detect. Further research is needed to identify the interactions of different phenotypes

and other environmental factors.

6.3.2.2 Immune response genes

Genes control the immune response, as all bodily development and function. Hitherto, CD14,

Human leukocyte antigen (HLA), Beta defensin (DEFB), Lactotransferrin (LTF), Mucin

(MUC), Mannose-binding lectin (MBL), and Manna-binding lectin serine peptidase (MASP)

have been studied extensively as genes that have the potential to make the host susceptible to

dental caries (Vieira et al., 2014). These genes operate by maintaining the immune system in

promoting or inhibiting oral cariogenic bacteria. The innate immune system relies on the

recognition of pathogen-associated molecules, such as bacterial cell wall components. CD14,

108

a 55-kD membrane glycoprotein, is expressed predominantly on the surface of monocytes,

macrophages, and neutrophils. They play a crucial role in the recognition of several microbial

products, such as lipopolysaccharide (LPS, endotoxin) and peptidoglycan, which are major

components of gram-negative and gram-positive bacterial surfaces (Lien and Ingalls, 2002).

CD14 is a multifunctional protein and has been suggested to reduce endotoxin-induced

activities of salivary pathogens.

The presence of salivary soluble CD14 has been found to reveal an inverse relationship with

the carious lesions; that is, CD14 was completely absent in the saliva of patients affected by

two to eight carious lesions (Bergandi et al., 2007). Moreover, HLA and major

histocompatibility complex (MHC) molecules have important roles in immune responsiveness.

Polymorphisms in the molecules may cause some variations in immune responses against oral

colonization levels and may influence an individual’s susceptibility to caries (Opal et al., 2015).

For instance, being positive for HLA-DRB1FNx0104 gene was reported to increase the risk of

developing caries by ten times (p = 0.03) in a group of 1- to 6-year-old children from Iran

(Bagherian et al., 2008). According to Altun et al. (2008), an increased frequency in HLA-

DRB1*01(p = 0.02) and HLA-DQB1*O3 (p = 0.009) were significantly associated with the

salivary MS among a group of children, though no association could be observed with caries

experience (Altun et al., 2008). Nevertheless, a population of adolescents in Brazil who were

positive for HLA-DQ2 allele were less likely to have dental caries than their counterparts who

were negative for this allele (OR = 0.33, CI = 0.16–0.66) (Valarini et al., 2012). A similar trend

was found towards a relationship between the presence of HLA-DR4 and high levels of MS;

however, the results were not statistically significant, probably due to the small number (28)

of subjects involved (Wallengren et al., 1997). DEFB 1 on the other hand is an oral anti-

microbial peptide, which provides the first line of defence against a wide spectrum of pathogens

109

(Krasone et al., 2014). Two haplotypes in the DEFB 1 promoter were linked with high and low

caries experience: a high-caries-experience haplotype (GCA), which were associated with

DMFT scores 2-folds higher, and a low-caries-experience haplotype (ACG), with DMFT

scores 2-folds lower (Ozturk et al., 2010). Variation in the promoter region of DEFB 1 plays a

role in the aetiology of caries since it has been proven that carrying a copy of the variant allele

of the DEFB 1 marker rs11362 (G-20A) was associated with a more than 5-folds higher DMFT

and DMFS scores (Krasone et al., 2014). Azevedo et al. (2010) observed that LTF, a major

iron-binding protein in milk and body secretions with antimicrobial activity, was associated

with lower caries experience in a population of 12-year-old Brazilian children (Azevedo et al.,

2010).

MBL (protein) is a lectin that is instrumental in innate immunity in humans as the protein

recognises mannose and N-acetylglucosamines on oral microorganisms. MBL is capable of

binding to carbohydrate moieties predominantly present on the surfaces of bacteria (Eddie Ip

et al., 2009). High serum levels and high activity of MBL, regulated by MBL2 gene, have been

associated with inflammatory diseases, transplant rejection, and diabetic nephropathy. Further,

MBL2 polymorphisms have been linked with bacterial infections such as Pseudomonas

aeruginosa and mycobacteria (Bouwman et al., 2006). According to Olszowski et al. (2012),

MBL2 gene polymorphism was significantly associated with increased caries experience in

Polish children. However, the direction of the association was found to be opposite in

deciduous and permanent dentition (Olszowski et al., 2012). Defensins are small cysteine-

rich cationic proteins localized in oral cavities and are an important component of the innate

immune system. Most defensins function by binding to the microbial cell membrane, and once

embedded, form pore-like membrane defects that allow efflux of essential ions and nutrients

(Fellermann and Stange, 2001). Ozturk et al. (2010) investigated three promoter

110

polymorphisms of the beta defensin 1 gene (DEFB1) and found that the presence of G-20A

(DEFB1 marker rs11362) variant were associated with DMFT and DMFS scores 5-folds

higher, while the G-52A (DEFB1 marker rs179946) variant was associated with lower DMFT

scores (Ozturk et al., 2010). This observation is most probably due to defensins playing a

greater or lesser role in bacterial aggregation, after which they will be swallowed, as first line

of defence, and on colonisation of tooth surfaces.

6.3.2.3 Genes related to taste sensation

It has been hypothesized that food preferences of children are learned although the desire to

select one food over another is more closely linked to taste and other sensory properties of

foods (Pidamale et al., 2012). Given the influence of dietary habits on dental caries, it can be

theorized that taste pathway genes, such as genes for taste preference, might influence caries

risk. Although taste preferences based on chemical tastants have been proven useful for the

assessment of dietary habits, studies have demonstrated the importance of dietary habits, and

nutritional status coupled with genetically determined taste sensitivity with respect to caries

risk (Bretz et al., 2006; Wendell et al., 2010). Inappropriate intakes of fermentable

carbohydrates, particularly sucrose, are well established as dominant factors in the

pathogenesis of caries. The cariogenic flora can metabolise sucrose moieties in the oral

environment in a unique manner, synthesising extracellular polysaccharides which help form

oral biofilms, and producing acids that can demineralize the tooth structure (Loesche, 1986).

Human sweet taste perception is mediated by the heterodimeric G protein-coupled receptor

encoded by the TAS1R2 and TAS1R3 genes (Fushan et al., 2009). Fushan et al. found that two

C/T SNPs located at positions 21572 (rs307355) and 21266 (rs35744813) upstream of the

TAS1R3 coding sequence strongly correlate with human taste sensitivity to sucrose (Fushan et

111

al., 2009). Further, they suggested that there were strong deviations from evolutionary neutral

genetic drift for variation in the coding regions of TAS1R2 and, to a lesser extent, TAS1R3,

which could contribute to sensitivity to sweet compounds. People who carried T alleles

displayed decreased sucrose sensitivity scores (using quantitative stimuli of sucrose solutions

with different concentrations) compared to those who carried C alleles (Fushan et al., 2009).

Kulkarni et al. (2013) observed similar outcomes in a group of Caucasians aged 21-32 years

by studying polymorphisms in the sweet taste receptor (TAS1R2) and glucose transporter

(GLUT2). They found that subjects with GLUT2 and TAS1R2 genotypes, both individually

and in combination, were associated with caries risk. A higher DMFT score was observed

(mean DMFT 4.3 vs 6.1, p = 0.04) among carriers of the Ile allele for GLUT2 (regarded as a

high/higher risk group), whereas carriers of the Val allele for TAS1R2 showed lower DMFT

scores (mean DMFT 4.1 vs 5.8, p = 0.05) and were regarded as a resistant group (Kulkarni et

al., 2013). Pidamale et al. (2012) demonstrated that the sensitivity to bitter taste, which is

mediated by the TAS2R38 gene, was associated with dental caries. Children aged 36-71

months who carried TAS2R38 gene could taste bitter sensation more and had less affinity

towards sweet sensation and had low dmft scores compared to non-tasters (Pidamale et al.,

2012). By hypothesizing that genetic variation in taste pathway genes may be associated with

dental caries, Wendell et al. investigated TAS2R38, TAS1R2 and GNAT3 genes in families

recruited by the Centre for Oral Health Research in Appalachia. Results revealed that

TAS2R38 and TAS1R2 are associated with experience of, and presumptively with

susceptibility to, dental caries, while the taste gene TAS2R38 (bitter taste receptors) appears to

be “protective” in the primary dentition: certain alleles of taste genes TAS1R2 (sweet taste

receptors) were associated with caries risk in the mixed dentition group (Wendell et al., 2010).

When the concern is taste genes and dental caries, it is logical to accept the fact that genetic

112

variations contribute to differences in dietary habits which in turn influence the caries process,

rather than having a direct relationship with caries.

6.3.2.4 Genes related to saliva

Numerous studies bear evidence that individuals who lack the ability to produce adequate

saliva volumes to maintain equilibrium in pH and buffering capacity suffer from several

diseases. It has long been understood that saliva has important properties that protect the

dentition: volume and flow which can dilute acids and flush food debris and microorganisms

from the mouth, a pH which can neutralise acids, and a significant buffering ability (Lenander-

Lumikari and Loimaranta, 2000). Although these properties of saliva are known to modify the

caries process, the role of genetic polymorphism remains relatively unexplored. There is hardly

any literature that shows an association between genetic variations in salivary physiology and

dental caries (Opal et al., 2015). Peres et al. in 2009 investigated the allele and genotype

distribution of three polymorphisms in the coding sequence of the Carbonic anhydrase VI

(CA6) gene and checked possible associations with saliva buffering capacity, plaque pH,

plaque index, and dental caries experience (Peres et al., 2010). They observed that among the

245 children studied there was a positive association between buffer capacity and the CA6 gene

rs2274327 (C/T) polymorphism. The allele T (p = 0.023) and genotype TT (p = 0.045) were

less frequent in children with higher buffer capacity. Since the association between CA6 gene

polymorphism and caries experience was not found to be statistically significant (Peres et al.,

2010), it is probable that the effects are overshadowed by other genetic variables, diet, and oral

hygiene practices.

The acidic proline-rich proteins (PRPs) account for 25%-30% of all proteins in saliva, and these

influence the attachment of cariogenic bacteria to tooth surfaces (Lenander-Lumikari and

113

Loimaranta, 2000). Human PRPs are encoded by six closely linked genes on chromosome

12p13.2 (Kim et al., 1990). Zakhary et al. (2007) studied one of the three acidic PRP alleles of

PRH1 locus (Db) to determine its association with dental caries (Zakhary et al., 2007). Results

revealed that in Caucasians, the Db gene frequency was 14%, similar to Db protein from parotid

saliva, and in African-Americans the similarity was 37%. All Caucasians, when compared to

African-Americans had significantly high caries experience along with greater MScounts. They

concluded that racial differences in caries experience, and by implication caries susceptibility,

might be explained in part by the differences in Db alleles (Zakhary et al., 2007). Similarly, the

salivary scavenger receptor cysteine-rich glycoprotein gp-340, which facilitates adhesion of

Streptococcus mutans, shelters three major size variants, designated gp-340 I, II and III, each

specific to an individual’s saliva. The gp-340 I was found to correlate positively with caries

experience in a group of 12-year-old Swedish children (Jonasson et al., 2007). Moreover, the

allelic acid PRP protein variant Db positively correlated with high caries experience, and the

gp-340 I protein was more common among Db+ subjects than among Db- subjects. Hence, the

phenotypes positive for both gp-340 I and Db experienced more caries than those negative for

both proteins (Jonasson et al., 2007). Similar findings were reported by Ayad et al. (2000),

where the phenotype of PRPs, PS1 encoded by PRB1 and con1 encoded by PRB2, were

significantly higher in caries free individuals (Ayad et al., 2000).

Owing to the fact that caries is a disease modified by a multitude of factors in different

population groups the variation of which cannot be explained by genetic endowment alone.

Genetic polymorphisms in a large number of genes have been shown to quantitatively and

qualitatively affect dental caries, with effects on susceptibility and severity of diverse

communities. Yet the human genome is diverse and when looked at individually, important

variations such as different SNPs and haplotypes can be observed. Multiple scans for these

114

variations across the genome have pointed to genes involved in behavioural traits as well as to

those that may predict deferred disease liability. Stimuli and mechanisms that influence these

inter-individual differences may help in explaining personal susceptibility to dental caries.

6.4 Genes, caries and environment

Although genetic studies have indicated some associations of DNA variants to dental caries

phenotypes, there have been no truly "potent" effect variants identified with causal biological

relevance. This could be due to the influence of environmental factors having a long-term

influence on genetic modifications; in other words, epigenetic modifications could have an

effect on gene expression leading to altered dental caries phenotypes (figure 6.1). Williams et

al. (2014) emphasized this following the difficulty to establish causal relationships between

specific genes and a number of complex diseases, many of which manifest later in life (Eichler

et al., 2010; Williams et al., 2014).

115

Figure 6.1 Environmental factors affecting epigenetic modification (Source: Seo et al., 2015)

(This is from an open-access article distributed under the terms of the Creative Commons

Attribution-Noncommercial-Share Alike 3.0 Unported, which permits unrestricted use,

distribution, and reproduction in any medium, provided the original work is properly cited

http://creativecommons.org/licenses/by-nc-nd/3.0/ )

6.4.1 Evidence from twin studies

Investigating the influence of environmental factors on the epigenome is challenging because

it is difficult to establish whether the variations observed in gene expression are caused purely

by changes in genes or influenced by other factors. However, the twin model displayed that

monozygotic (MZ) twins who have the same genome (but a different epigenome, particularly

if they were reared apart) differ significantly in susceptibility to many diseases (Machin, 1996;

Petronis et al., 2003). Epigenome-wide studies of phenotypically discordant, genetically

Environmental factors

Diet Smoking Inflammation Stimuli Age

Genetics Epigenetics

Epigenetic modification

DNA methylation Non-coding RNA Histone modification

Gene Expression

Functions: Imprinting, Development, Disease,

Cancer

116

‘identical’ MZ twins have provided information about gene-environment interaction and

variable penetrance; however, the study lacked balanced MZ and DZ comparison groups

(Boraas et al., 1988). In 2005, Fraga et al. demonstrated that MZ twins could differ significantly

in patterns of both methylation and histone modification of their genomes, with these

differences increasing with age (Fraga et al., 2005). Twins who had spent less of their lifetime

together (i.e. different environmental exposures) showed the greatest differences in histone

modification (Fraga et al., 2005). These epigenetic modifications were associated with, or

caused by, environmental factors like smoking, diet, age, and lifestyle factors, for example,

unhealthy BMI (Fraga et al., 2005; Seddon et al., 2011). Such findings support the current

hypothesis that epigenetic changes increase with age and that epigenetic biomarkers can predict

disease long before clinical onset (Craig, 2013; Fernando et al., 2015). Hence, the need for

investigations in the field of oral diseases especially in the area of dental caries is further

emphasised; we have not detected any publications to date on the epigenetics of cariogenesis.

6.5 Epigenetics

Epigenetics is the study of alterations in gene regulation that are not necessarily caused by

changes in the DNA sequence (Seo et al., 2015). Historically “epigenetics” was used to

describe inherited influences that could not be described solely by genetic principles (Goldberg

et al., 2007). These include chemical and environmental alterations in the DNA and associated

proteins, with remodelling of the chromatin and dysregulation of specific gene function, thus

influencing pathogenesis (Gomez et al., 2009; Su et al., 2011; Wilson, 2008).

117

Mechanisms of Epigenetic modification

6.6.1 DNA methylation

DNA methylation is the most studied epigenetic alteration of DNA in humans (Barros and

Offenbacher, 2009; Leenen et al., 2016) and the most characteristic type of chromatin

modification (Seo et al., 2015). The process of DNA methylation is by the covalent transfer of

a methyl group from S-adenosyl methionine (SAM) to a cytosine present in CpG dinucleotides

of the DNA chain (Williams et al., 2014). In the normal cell, gene promoter-associated CpG

islands are predominantly unmethylated, whereas CpG sites within gene bodies are generally

methylated (Patterson et al., 2011) (figure 6.2).

Figure 6.2 A CpG island in relation to a gene promoter region (Source: Patterson et al., 2011).

(This is from an open-access article distributed under the terms of the Creative Commons

Attribution-Noncommercial-Share Alike 3.0 Unported, which permits unrestricted use,

distribution, and reproduction in any medium, provided the original work is properly cited

http://creativecommons.org/licenses/by-nc-nd/3.0/ )

Promoter-associated

CpG Island

118

Hypermethylation of CpG inlands causes inhibition of gene transcription in the area and

hypomethylation results in activation of these genes, both of which have been implicated in the

development and progression of diseases such as cancer (Mani and Herceg, 2011) (figure 6.3).

Figure 6.3. DNA methylation at CPG sites (Source: Seo et al., 2015). (This is from an open-

access article distributed under the terms of the Creative Commons Attribution-

Noncommercial-Share Alike 3.0 Unported, which permits unrestricted use, distribution, and

reproduction in any medium, provided the original work is properly cited

http://creativecommons.org/licenses/by-nc-nd/3.0/ )

6.6.2 Histone modification

The nucleosome is the fundamental subunit of chromatin. Each nucleosome is composed of

less than two turns of DNA wrapped around a set of eight proteins called “histones” (Seo et

al., 2015). The structure of histones determines how the DNA is condensed, ultimately

affecting gene expression (Önder et al., 2015; Williams et al., 2014). The commonest form of

histone modification is through acetylation of its eight sub-units (Seo et al., 2015). Hyper- and

hypo-acetylation can affect chromatin condensation either allowing or preventing gene

transcription respectively (Williams et al., 2014). Differences in gene expression associated

with differential acetylation will affect disease outcomes.

CPG Gene

CPG Gene

Unmethylated Transcription Activation

Methylated Transcription Inactivation

119

6.6.3 Non-coding RNA

RNA makes up coding units which produce proteins. RNAs are broadly categorized into coding

RNA, RNAs with an open reading frame (ORF) which are translated to proteins; and non-

coding RNA without an ORF, which do not produce proteins, though they are active

themselves (Williams et al., 2014). Non-coding RNA includes important RNAs like tRNA

(transfer RNA), rRNA (ribosome RNA), miRNA (mitochondrial RNA) and siRNA (small

interfering RNA), which can regulate gene expression without altering DNA sequence

(Goldberg et al., 2007; Seo et al., 2015). These non-coding RNAs have the ability to control

epigenetic mechanisms like gene silencing (Goldberg et al., 2007) and have been shown to

contribute significantly to the progression of oral mucosal lesions through a combination of

oral RNA molecules (oral transcriptome) derived from oral premalignant lesions (Perez et al.,

2014).

6.7 Environmental stimuli which may result in epigenetic changes which

could influence the process of dental caries

Dental caries is known to be a disease with many risk modifiers, both environmental and

intrinsic, to an individual’s biological make up. Environmental stressors, including an

unbalanced diet, microbial exposures which cause a shift from a healthy ecology to a

potentially pathogenic ecology, tobacco smoking, and high alcohol intake can change

epigenetic patterns and thereby effect changes in gene activation and cell phenotypes and

functions (figure 6.1). However, confounders like age, sex, and ethnicity might mask the real

effect. As epigenetic changes are frequently retained following cell division, altered patterns

120

in tissues can accumulate over time and lead to variations phenotypes, reflecting a molecular

consequence of gene-environment interaction, which can then be inherited by following

generations.

6.7.1 Diet and nutrition

Alterations in DNA methylation due to environmental stimuli begin even before birth.

Methylation of foetal DNA in utero due to low dietary levels of folate, methionine, and/or

selenium can change epigenetic programming, and this can persist into adulthood (Zaina et al.,

2005). However, established epigenetic patterns during the foetal period can become altered in

adulthood by environmental factors (Su et al., 2011). Folate is a critical methyl donor for S-

Adenosyl Methionine (SAM), which is used by the enzyme DNA methyltransferase (DNMT)

to methylate CpG residues selectively during embryonic development (Razin and Shemer,

1995). Hence, a decreased folate intake by the mother during pregnancy would cause a

subsequent decrease in DNA methylation in the foetus (Okano et al., 1999), although the level

“decreased” is subjective. Furthermore, nutritional components like vitamin D, betaine and

methionine can also cause epigenetic changes and consequently lead to disease not only at the

time of ingestion but even during intrauterine life (Seddon et al., 2011). Low intake of folate

and high alcohol consumption has been associated with altered methylation patterns in global

DNA, presenting an increased risk for colon cancer (Seddon et al., 2011). Studies like these

need to be interpreted with caution as there can always be recall bias. Nevertheless, high

consumption of alcohol was found to be associated with changes in methylation patterns of

squamous cells in the colon, promoting colon cancer (Schernhammer et al., 2010). This was

further emphasized by Sue et al., who elaborated that a wide range of dietary factors including

both macro-nutrients and micro nutrients have the potential to alter methylation of DNA.

Through diet an individual can be exposed not only to the nutritional components but also to

121

toxins that can cause hypomethylation of cancer cells and embryonic stem cells, for example,

arsenic, cadmium, mercury, and nickel (Davis and Uthus, 2004; Su et al., 2011); however, these

are rare.

6.7.2 Tobacco

Tobacco use is another cause for differential methylation of DNA. Tobacco smoke induces a

reduction in monoamine oxidase B (MAOB) promoter methylation and has long-lasting effects

on MAOB transcription (Launay et al., 2009). According to Launay et al. smoking causes long-

term hypo- and hyper-methylation changes in platelet and peripheral blood mononuclear cells

of smokers. These changes are present not only in current smokers but also in former smokers

(Belinsky et al., 2002; Launay et al., 2009). Hillemacher et al. reported that smoking has long

term effect on global DNA methylation across multiple tissues, and these changes are

demonstrable in leukocytes of offspring if parents smoked (Hillemacher et al., 2008).

6.7.3 Bacteria and inflammation

The human microbiomes, including the oral microbiota, comprise another set of influences on

the human genome, with potential to generate significant epigenetic changes. Complex

interactions between oral bacteria and host genome are the basis of susceptibility to

periodontitis (Pihlstrom et al., 2005). For example, specific polymorphisms of the serum

Interleukin-6 gene have been shown in patients with severe forms of destructive periodontitis

and correlate with the presence of Aggregatibacter actinomycetemcomitans and

Porphyromonas gingivalis (Nibali et al., 2008). A similar result was observed by Wu et al.,

where loss of function of the adenine methyltransferase (DAM) gene via DNA methylation by

reduced production of leokotoxins (Wu et al., 2006). In the cariology literature, Cardoso et al.,

122

in 2010, reported that differential methylation of the promoter region of the IFN-ɤ gene was

associated with human pulpal tissue affected with pulpitis. Either partial methylation or un-

methylation was observed in 93.75% of samples of inflamed pulp tissue (Cardoso et al., 2010).

6.8 Future research

Many studies have shown the importance of epigenetics in oral health. This has been shown in

relation to periodontal diseases (Zhang et al., 2010), oral squamous cell carcinoma (Sandoval

and Asteller, 2012), malocclusion (Carlson, 2005; Dave Singh and Callister, 2013), and cleft

lip/palate (Beaty et al., 2011; Marazita, 2012). However, there is little information related to

the pathogenesis of dental caries. Epigenetics, the potential conduit between phenotype and

genotype, has much promise in becoming increasingly relevant to cariology because of its role

in gene expression. Dental caries has been studied for decades as a multifactorial disease but

is still among the ten most common chronic diseases among children (Fisher-Owens et al.,

2007; Kassebaum et al., 2015). In this context, the notion that gene-environment interactions

(i.e. epigenetics) are important in caries pathogenesis is appealing as it could prove to be the

missing link to understanding of the role of genetics in caries initiation and progression.

Future studies utilizing epigenetics are important to the individual as well as for society, as

findings regarding the interplay of environmental and genetic factors can be used in

identification of individuals and groups at high risk of developing dental caries. This can help

to identify new preventive strategies and treatment approaches. As epigenetic patterns and

modifications are found to be reversible, it may also be possible to use this knowledge to devise

approaches to reduce risk (Azad et al., 2013). Combined clinical and longitudinal

123

epidemiological studies are needed to quantify the extent to which environmental factors may

influence epigenetic changes that can predispose to dental caries.

124

Chapter 7: An Epigenome- Wide Association Study of mother-

child dyads discordant for dental caries: A pilot study

This chapter is the fourth paper in this thesis. The bibliographic details of the paper, including

all authors, are as follows:

Surani Fernando, Miles Benton, David Jeremiah Speicher, Mahmoud Bakr, Ping Zhang,

Rodney Lea, Paul A Scuffham, Newell Walter Johnson. An epigenome wide association study

of mother child dyads discordant for dental caries: A pilot study.

My contribution to the paper involved the conception and designing of the study, clinical data

collection, DNA extraction and sample preparation, statistical analysis and drafting the article.

The manuscript has been submitted to the journal BMC Oral Health, which is a peer reviewed

journal. Manuscript ID is OHEA-D-17-00215.

Surani Fernando

Corresponding author: Surani Fernando

Supervisor: Professor Newell Johnson Supervisor: Professor Paul Scuffham

125

7.1 Abstract

7.1.1 Background

Dental caries is a major public health problem affecting millions of children worldwide. GWAS

have examined the gene structure associated with dental caries, but few have investigated

environmental effects on the epigenome. Whilst the environment can significantly impact the

epigenome, there are currently no published epigenome-wide association studies (EWAS) on

dental caries.

7.1.2. Objective

To conduct a pilot study to identify potential differentially methylated regions associated with

dental caries experience in a cohort of mother-child dyads.

7.1.3 Methods

For this pilot study, 12 mother-child dyads from SEQ, Australia were selected from within the

Griffith University EFHL birth cohort study based on high (6 dyads) and low (6 dyads) caries

experience according to ICDAS. Epigenetic screening was conducted on total DNA extracted

from saliva using the SeqCap Epi 4M CpGIANT Enrichment Kit and HiSeq2500 Sequencer.

CpG counts were analysed using the methylKit package in R. The statistical significance

threshold was set using Bonferroni correction based on testing 435508 CpG sites (p = 1.1E10-

7).

126

7.1.4 Results

Differentially methylated regions (DMR) between cases and controls were found on

chromosomes 1, 2, 5, 7, 12, 20 and 22. Chromosome 12 showed the highest average

methylation difference (41%) between case and control groups (p < 2.17E-103). DAVID gene

function analysis of all DMR showed two clusters related to Zn metabolism and membrane

protein functions.

7.1.5 Conclusion

This pilot EWAS identified significantly differentially methylated sites discordant for dental

caries on chromosome 12 closely related to a gene promoter region responsible for cell

membrane function and Zn metabolism. Further work is required to understand the mechanisms

by which these epigenetically modified sites might influence susceptibility to dental caries.

Key words Dental caries, Epigenomics, Methylation, CpG, EWAS

7.2 Introduction

Dental caries remains a major public health problem in most low and middle income countries,

affecting more than 621 million children and 2.4 billion adults worldwide (Kassebaum et al.,

2015). It is an almost ubiquitous disease throughout the life course, with peak prevalence at

ages 6, 25 and 70 years. The burden of untreated dental caries is greatest in developing low-

middle income countries, particularly in Africa (Kassebaum et al., 2015), but is a problem in

some population groups within all countries. It is unsurprising that dental scientists are turning

127

their attention to the role of epigenetic changes, most commonly through gene methylation, in

susceptibility to, and the pathogenesis of, oral diseases (Hughes et al., 2013; Seo et al., 2015).

Recently, GWAS have explored associations between the structure and function of particular

genes and the presence or absence of many diseases, whilst EWAS have revealed genetic

changes associated with environmental factors and disease. DNA methylation, characterized

by covalent transfer of a methyl group from SAM in cytosine located in CpG islands, is the

most studied epigenetic modification and the most distinctive category of chromatin changes

(Barros and Offenbacher, 2009; Schübeler, 2015; Williams et al., 2014). DNA methyl-

transferases mediate DNA methylation and stabilise the transmission of differential

methylation changes during cell division (Bestor, 2000; Okano et al., 1999). In a normal cell,

CpG sites associated with gene promoters are usually unmethylated, whereas CpG sites within

genes are generally methylated (Patterson et al., 2011). Hyper-methylation of CpG sites inhibits

gene transcription, whereas hypomethylation results in gene activation (Mani and Herceg,

2011). Availability of high-throughput genotyping methods and high-density reference panels

of the human methylome have made it feasible to conduct EWAS at population levels (Sun,

2014). Seo et al. (2015) argued that DNA methylation patterns associated with inflammatory

and immune responses potentially affect the pathogenesis of oral diseases.

Emerging evidence highlights the importance of epigenetic regulatory mechanisms in

controlling the inflammatory response in inflamed dental pulps of carious teeth. Recently,

Cardoso et al., reported that 93.75% of inflamed pulps showed unmethylation in the promoter

regions of the IFN-ɤ gene (Cardoso et al., 2010), whereas hypomethylated TLR2 and CD14

genes were a regular characteristic of healthy dental pulps (Cardoso et al., 2014). An in-vitro

study of human dental pulp cells showed that, even though no direct association with dental

128

caries could be found, the DNA methyltransferase inhibitor 5-Aza_CdR could inhibit

proliferation and heighten odontogenic differentiation of cells (Zhang et al., 2015). This

suggests that methylome status could influence the production of reparative dentine in response

to a carious lesion or other injury. During amelogenesis, pre-ameloblasts express high levels

of DNAMT1, a regulator of DNA methylation, whilst in their undifferentiated state (Yoshioka

et al., 2015). If hypermethylation were to occur, amelogenesis could be damaged through

expression of DNMT1, producing hypomineralisation of the enamel. In this pilot study, we

conducted an EWAS of 12 mother-child dyads with high and low experience of dental caries

to detect associations of differentially methylated regions (DMR) with dental caries experience.

Given the expensive nature of the techniques involved, the study was designed to identify

potential sites for further investigation in a larger population.

7.3 Methods

7.3.1 Study Population

Six- to eight-year-old children and their mothers who were enrolled in the Griffith University

EFHL (Cameron et al., 2012a) were recruited for the oral health sub study of EFHL following

ethical approval (Griffith University: OTH/25/13/HREC). A protocol for the main oral health

sub study was published in BMC Oral Health (2015), which describes enrolment of subjects

and data collection in detail (Fernando et al., 2015). For this pilot study 12 mother-child dyads

discordant for dental caries were enlisted to screen for changes in the epigenome which were

associated with low or high caries experience.

129

7.3.2 Dental caries assessment

The severity and extent of dental caries were measured using ICDAS criteria, where all surfaces

of all teeth of participating individuals were recorded by trained examiners (CICDAS, 2005).

Past caries experience (surfaces of teeth missing due to caries and restored tooth surfaces) and

current caries was recorded as diseased surfaces ranging from demineralisation without

cavitation (Score 1) to overt cavities (Score 6). Teeth that were not erupted and extracted for

reasons other than caries were excluded from the calculation. Cases and controls were

determined according to the caries experience of both mother and child: controls, low caries

experience of both; and cases, high caries experience of both. The criteria used here for

classification of caries experience took the percentage of surfaces affected with caries and age

into consideration, since it is expected that caries experience will increase in the general

population with age, and also because our pilot study sample lacked totally caries-free mother-

child dyads (Küchler et al., 2014). Hence average caries experience for both mothers and

children were calculated; children who had positive caries scores of more than 6% of their tooth

surfaces and mothers with caries in more than 20% of their tooth surfaces were cases and the

rest controls (Table 7.1).

7.3.3 Sample Collection and Epigenetic analysis

Saliva is an effective, efficient and non-invasive source of host DNA (Kaczor-Urbanowicz et

al., 2017), which may be especially relevant to studies of the epigenetics of oral diseases as it

bathes the tissues of interest. Hence saliva was collected from the 12 mother-child dyads and

stored at -80oC (Fernando et al., 2015). Genomic DNA was extracted from saliva using the

High Pure PCR Template Preparation Kit (Roche Diagnostics, Australia). Total DNA was

130

quantified on the NanoDrop® ND-1000 Spectrophotometer (ThermoFisher Scientific,

Australia). DNA was considered pure if optical density at 260/280 was 1.70–2.00.

DNA sequencing was performed with bisulfite treated DNA for methylation analysis. Assays

were performed using the SeqCap Epi 4 M CpGIANT Enrichment Kit (Roche NimbleGen,

USA) and the HiSeq2500 Sequencer (Illumina®, USA) at the University of Queensland Centre

for Clinical Genomics (UQCCG) (Brisbane, Australia). This approach obtained both DNA

sequence variants and CpG methylation simultaneously at ~450,000 CpG sites across the whole

genome.

Initial read quality assessment was performed using FastQC (Andrews, 2010) while adapter

trimming was implemented using the Trimmomatic software

(http://www.usadellab.org/cms/?page=trimmomatic). Due to the bisulfite treatment, reads

were required to be aligned to an in-silico bisulfite converted reference genome. This was

achieved by performing an in-silico conversion of hg19 using bowtie2 (http://bowtie-

bio.sourceforge.net/bowtie2/index.shtml). Read alignment was then carried out using Bismark,

a software suite specifically designed for bisulfite sequence mapping

(www.bioinformatics.bbsrc.ac.uk/projects/bismark/). After alignment, all bam files for each

individual were merged using bamtools (https://github.com/pezmaster31/bamtools).

Deduplication of reads was conducted using the ‘MarkDuplicates’ function within Picard

Tools (http://broadinstitute.github.io/picard/command-line-overview.html#MarkDuplicates). Finally,

the software PileOMeth (https://github.com/dpryan79/PileOMeth) was used to extract all CpG

sites and read counts formatted for the R analysis package methylkit

(http://code.google.com/p/methylkit).

131

Site specific methylation levels were defined as β values, which is the ratio of methylated probe

signal over methylated plus unmethylated probe signals. Each identified differentially

methylated site was attributed a percentage methylation that was included in the statistical

analyses. A DMR was defined as a genomic region that contained ≥ 3 directly adjacent

differentially methylated CpGs.

General statistical analysis was performed using SPSS Statistics V21.0 (IBM, USA).

Parametric tests were used as the data were normally distributed and where relevant chi-square

tests were used to compare the two groups. To evaluate the differences between cases and

controls with respect to DMR changes in CpGs on Chr12, t-tests were performed.

Analysis of CpG counts was performed in R using the methylKit package (Team, 2013). Data

were imported and treated as a case/control setup. CpG sites were filtered out if they exhibited

read count coverage lower than 10. Differential analysis was performed using the methylkit

‘calculateDiff’ function, which calculates differential methylation statistics between two

groups of samples using logistic regression. A custom R script was written to identify DMR.

Briefly, this method uses a sliding window to identify CpG sites showing significant

differential methylation in a similar direction within a defined region. An overall DMR p-value

is assigned by assessing all individual p-values of CpG sites contained within a DMR,

combining these using a modified version of Fisher's approach (Zaykin, 2011). All CpG and

DMR objects were annotated using Granges and R tracklayer, based on hg19 annotation data

available from Bioconductor (http://www.bioconductor.org). Statistical significance (i.e. alpha

level) was set based on Bonferroni correction for the number of CpGs tested. In this study, we

observed and tested 435508 CpG sites and thus set the empirical alpha-level to

0.05/435508=1.1E10-7. Annotation and functional analysis of the list of genes that are

132

associated with DMR or selected CpGs were identified through the DAVID bioinformatics

functional annotation tool (Huang et al., 2009a; b).

Table 7.1: Caries experience of controls and cases

Child #ID Child’s caries

*experience

Mother ID Mother’s caries

*experience

Controls Controls

FAM1C 0 FAM1M 7.69

FAM2C 0 FAM2M 4.29

FAM3C 2.61 FAM3M 10.00

FAM4C 3.00 FAM4M 0

FAM5C 3.33 FAM5M 19.85

FAM6C 6.00 FAM6M 15.71

Cases Cases

FAM7C 6.50 FAM7M 24.29

FAM8C 9.00 FAM8M 26.43

FAM9C 11.00 FAM9M 39.49

FAM10C 13.33 FAM10M 22.14

FAM11C 16.00 FAM11M 33.79

FAM12C 19.00 FAM12M 25.00 # Identity, *Caries experience was measured as a percentage of tooth surfaces affected with dental caries

(including early enamel caries)

133

Table 7. 2: Demographic and socio-economic characteristics of controls and cases

Variable Controls Cases X2 P value

N (%) N (%)

Annual household income 4.2 0.24

$0-59,999 3 (50) 3 (50) $60000-89999 3 (50) 1 (13) $>90000 0 2 (37) Mother’s employment status 2.0 0.57

Full time 1 (17) 1 (17) Part time 2 (33) 1 (17) Unemployed 3 (50) 4 (66) Mother’s level of education 6.0 0.11

Primary school 2 (33) 0 High school 1 (17) 0 Tertiary education 3 (50) 6 (100) Mean (SD) Mean

(SD)

t value

Mother’s age in years at examination 37.6 (5.1) 36.8 (5.4) 0.27 0.79

Mother’s caries experience as a % of affected

surfaces

9.7 (7.5) 28.5 (6.6) 4.60** 0.01

Child’s caries experience as a % of affected

surfaces

2.4 (2.3) 12.48

(4.6)

4.80** 0.01

Child’s age in years at examination 6.3 (5.2) 6.5 (1.1) -0.35 0.73 Considered level of significance p≤0.05, **p=0.01

134

Table 7.3: Annotated data for the 16 identified Differentially Methylated Regions between controls and cases

¥Chr Number of §CpG

Start End ɤDMR

length

ɤDMR

P value

Average

different

methylation

Entrez ID Gene

Symbol

Promoter

chr1 14 1.47E+08 1.47E+08 6798 2.38E-107 16.31 644861 NBPF13P FALSE

chr2 3 88802707 88802733 26 4.53E-11 17.89 200523 TEX37 FALSE

chr2 3 1.28E+08 1.28E+08 25 1.38E-17 15.57 84826 SFT2D3 FALSE

chr2 4 2.32E+08 2.32E+08 121 1.96E-15 19.757 348761 SPATA3-AS1 FALSE

chr5 3 23951427 23951496 69 1.12E-13 -20.60* 56979 PRDM9 FALSE

chr5 20 28928022 28928448 426 6.44E-266 -32.58* 729862 LSP1P3 FALSE

chr5 7 1.77E+08 1.77E+08 62 1.26E-75 20.58 10636 RGS14 FALSE

chr5 3 1.78E+08 1.78E+08 46 1.10E-13 -18.08* 6940 ZNF354A FALSE

chr5 3 1.8E+08 1.8E+08 75 6.89E-08 17.81 255426 RASGEF1C FALSE

chr5 3 1.8E+08 1.8E+08 191 1.77E-09 19.58 9945 GFPT2 FALSE

chr7 3 120449 120480 31 2.99E-11 -16.11* 56975 FAM20C FALSE

chr7 3 1.55E+08 1.55E+08 55 5.98E-13 -18.26* 3638 INSIG1 FALSE

chr12 8 11700085 11700343 258 2.17E-103 -41.17* 338817 LINC01252 TRUE

chr20 4 61590651 61591049 398 3.72E-15 -16.22* 63910 SLC17A9 FALSE

chr22 4 32600622 32601130 508 8.11E-15 19.56 10739 RFPL2 TRUE

chr22 3 45832692 45832871 179 3.38E-13 -18.60* 26150 RIBC2 FALSE ¥Chr – Chromosome, *A negative average different methylation indicates that cases have lower methylation on average compared to controls, §CpG - Cytosine

Guanine sites, ɤDMR - Differentially Methylated Regions

135

Table 7.4: Functions of gene clusters derived from DAVID

Term Gene symbols Function

Cluster1

*GO:0008270~Zn ion

binding

ZNF354A, RFPL2,

PRDM9

Interacting selectively and non-covalently with Zn

ions. GO:0046872~metal ion

binding

FAM20C, ZNF354A,

RFPL2, PRDM9

Interacting selectively and non-covalently with Zn

and any metal ion

GO:0043169~cation

binding

FAM20C, ZNF354A,

RFPL2, PRDM9

Interacting selectively and non-covalently with Zn

ad other cations, charged atoms or groups of atoms

with a net positive charge

GO:0043167~ion

binding

FAM20C, ZNF354A,

RFPL2, PRDM9

Interacting selectively and non-covalently with

ions, charged atoms or groups of atoms

Zn binding ZNF354A, RFPL2,

PRDM9

Protein which binds at least one Zn atom or protein

whose function is Zn dependent.

Cluster2

GO:0016021~integral

to membrane

INSIG1, SFT2D3,

SLC17A9

The component of a membrane consisting of the

gene products and protein complexes having at

least some part of their peptide sequence embedded

in the hydrophobic region of the membrane.

GO:0031224~intrinsic

to membrane

INSIG1, SFT2D3,

SLC17A9

The component of a membrane consisting of the

gene products having some covalently attached

portion, for example part of a peptide sequence or

some other covalently attached group such as a GPI

anchor, which spans or is embedded in one or both

leaflets of the membrane.

Transmembrane INSIG1, SFT2D3,

SLC17A9

Protein with at least one transmembrane domain, a

membrane-spanning helical or beta-stranded

domain embedded in a membrane. *GO– Gene Ontology

7.4 Results

The six dyads with low caries experience and six dyads with high caries experience were

similar with respect to their demographic and socio-economic characteristics (Table 7.2).

Children had mean ages of 6.3 years (SD = 1.1) and 6.5 years (SD = 5.2) in controls and cases

respectively. In the control group, the mean ICDAS scores of the mothers and children were

9.7 (SD = 7.5; p ≤ 0.01) and 2.4 (SD = 2.3; p ≤ 0.01), respectively. In the case group, the mean

ICDAS scores of the mothers and children were 28.5 (SD = 6.6; p ≤ 0 .01) 12.48 (SD = 4.6; p

136

≤ 0.01), respectively. Mean total DNA concentration for controls was 132.26 ng/μL (range:

76.95 ng/μL – 224.66 ng/μL) and it was 178.58 ng/μL (range: 77.58 ng/μL – 370.32 ng/μL)

and for cases.

7.4.1 Methylation differences on chromosomes

Epigenetic analysis identified 435508 CpG sites with methylation that differed significantly

between controls and cases in DMR on chromosomes 1, 2, 5, 7, 12, 20 and 22 (p < 1.1E10-7;

Table 7.2). Chr12 had the highest average methylation difference (41%) between the two

groups for one CpG site (28928022 to 28928448) with eight differentially methylated CpGs in

relation to the promoter region of LINCO1252 (p = 2.17E-103). DMRs between cases and

controls on Chr12 were analysed separately for children (Appendix D, Table 1) and mothers

(Appendix D, Table 2).

7.4.2 Functions of identified genes

Analysis via DAVID showed that only the DMR on Chr12 (LINC01252, p = 2.17E-103) and

on Chr22 (RFPL2, p = 8.11E-15) were related to a gene promoter region (Table 7. 3). Chr12

showed the highest average methylation difference between control and case groups and thus

had the highest possibility of being linked with dental caries. Associated genes on Chr12 fell

into two functional clusters: (1) Zn or other metal ion binding; and (2) proteins of cell

membranes (Table 7.4). Moreover, chromosomes that had a single CpG cite differentially

methylated and associated with a gene promoter region were also explored through DAVID

gene function classification: BTNL8 on Chr 5, PFKFB3 on Chr10, ENOSF1 on Chr18, and

RFPL2 on Chr 22 could be related to possible functions related to dental caries (Table 7.5).

137

Table 7.5: Summary of Differentially Methylated Regions (with only one CpG site) related to a gene promoter region.

* A negative average different methylation indicates that cases have lower methylation on average compared to controls

Chromosome Start End Transcript Entrez ID Gene Symbol PROMOTER P-value Q value Methylation

difference

chr1 25292420 25292420 uc009vrl.1 864 RUNX3 TRUE 4.10572E-06 0.001587897 -15.08*

chr1 95556966 95556966 uc021oqe.1 148534 TMEM56 TRUE 9.81552E-08 7.95521E-05 19.17

chr1 101003634 101003634 uc001dth.3 54112 GPR88 TRUE 1.22669E-08 1.34245E-05 -16.53*

chr1 110254662 110254662 uc010ovu.1 2949 GSTM5 TRUE 2.66108E-09 3.56719E-06 22.56

chr1 182922582 182922582 uc001gpu.3 81626 SHCBP1L TRUE 5.00135E-05 0.009360155 -16.75*

chr1 182922588 182922588 uc001gpu.3 81626 SHCBP1L TRUE 1.32452E-06 0.000669004 -19.29*

chr1 223887895 223887895 uc010puy.2 824 CAPN2 TRUE 0 0 42.13

chr2 139538610 139538610 uc002tvi.3 11249 NXPH2 TRUE 3.06171E-10 5.31894E-07 -16.26*

chr2 139538640 139538640 uc002tvi.3 11249 NXPH2 TRUE 2.0518E-07 0.0001472 -16.51*

chr3 101658926 101658926 uc003dvr.1 152225 LOC152225 TRUE 1.86141E-08 1.94372E-05 -15.78*

chr5 6447161 6447161 uc003jdp.4 134111 UBE2QL1 TRUE 1.82771E-07 0.000134358 -16.83*

chr5 135418248 135418248 uc021ydy.1 100126299 VTRNA2-1 TRUE 0 0 -16.02*

chr5 180324777 180324777 uc003mmp.3 79908 BTNL8 TRUE 2.79604E-09 3.73517E-06 -15.40*

chr6 168198215 168198215 uc003sic.3 26238 C6orf123 TRUE 3.18527E-05 0.007035302 16.16

chr7 103849116 103849116 uc003vcb.3 5001 ORC5 TRUE 7.36078E-14 2.45828E-10 -21.03*

chr8 74007003 74007003 uc003xzf.3 157869 SBSPON TRUE 0.000000014 1.51094E-05 -20.19*

chr10 4868202 4868202 uc010qam.2 83592 AKR1E2 TRUE 2.02326E-06 0.000932887 21.52

chr10 6185422 6185422 uc001ijd.3 5209 PFKFB3 TRUE 8.36163E-06 0.002745206 -16.80*

chr10 82296143 82296143 uc001kck.1 387694 SH2D4B TRUE 2.50535E-06 0.001094318 20.68

chr11 300286 300286 uc001low.2 387733 IFITM5 TRUE 5.96865E-10 9.2863E-07 -15.90*

chr11 65327098 65327098 uc010rok.1 4054 LTBP3 TRUE 0.000000211 0.000150287 -15.39*

chr11 124439829 124439829 uc010san.2 390275 OR8A1 TRUE 0 0 -37.99*

chr15 89155014 89155014 uc010upm.2 407044 MIR7-2 TRUE 1.66276E-11 3.72348E-08 -19.64*

chr17 5674423 5674423 uc002gcm.3 339166 LOC339166 TRUE 6.28094E-06 0.002210056 -16.33*

chr17 16593135 16593135 uc002gqk.1 9720 CCDC144A TRUE 0.000013245 0.003834968 15.85

chr17 16593243 16593243 uc002gqk.1 9720 CCDC144A TRUE 5.74144E-06 0.002069085 18.14

chr18 713049 713049 uc002kkt.4 55556 ENOSF1 TRUE 1.34432E-06 0.000678121 15.51

chr19 36800807 36800807 uc021utf.1 100134317 LOC100134317 TRUE 8.93061E-08 0.000074244 15.89

chr19 36800819 36800819 uc021utf.1 100134317 LOC100134317 TRUE 1.32736E-10 2.53314E-07 17.38

chr22 22874959 22874959 uc002zwe.3 129025 ZNF280A TRUE 3.66237E-07 0.000236866 15.28

chr22 32601130 32601130 uc003amh.3 10739 RFPL2 TRUE 1.60469E-07 0.000120245 17.07

chr22 37256261 37256261 uc003apy.4 4689 NCF4 TRUE 1.32181E-05 0.003831219 19.21

138

7.5 Discussion, conclusion and limitations

7.5.1 Discussion

Though epigenetic examination of dental caries is in its infancy, it is increasingly understood

that environmental, physiological, and pathological events can affect the methylation status of

genes and thus regulate gene expression. This pilot EWAS explored the epigenome of

individuals discordant for dental caries in 12 mother-child dyads, and identified significantly

differentially methylated regions on chromosomes 1, 2, 5, 7, 12, 20 and 22 between cases and

controls. DAVID gene function analysis identified two clusters of genes related to DMR.

Genes identified in cluster 1 (ZNF354A, RFPL2, PRDM9 and FAM20C) mainly interact with

Zn and other metal ions at the cellular level. Zn fingers are clustered into more than 20 classes

of physically different components and are known to interact with a variety of proteins, lipids,

and nucleic acids (Lupo et al., 2013), whilst playing a significant role in metabolism of enzymes

and other proteins (Fatima et al., 2016). None of the genes identified in our study could be

directly functionally linked with the process of dental caries, though Zn is an important

component in health and disease, including in relation to oral diseases.

Zn is an essential co-enzyme in mineral metabolism that has long been recognized as an

element that influences all aspects of tooth development and repair, such as the

demineralisation/remineralisation equilibrium of dental hard tissues. Zn oxide is widely

recognized as a substance that inhibits dentine demineralization (Takatsuka et al., 2005). Zn, a

divalent cation (Zn+2), is electronically attracted to negatively charged oral hard tissues. In this

scenario, cariogenic bacteria like MS may have difficulty attaching to enamel surfaces, and as

a result have difficulty in acquiring nutrients essential for their survival (Burguera-Pascu et al.,

2007).

139

Zn favours the precipitation of calcium phosphate and is important in both the development of

enamel and its remineralisation after damage during cariogenesis (Lynch, 2011). The in-vitro

caries-simulating model of Mohammed et al. (2014), demonstrates significant reductions in

calcium release from enamel after exposure to Zn. Nevertheless, the adsorption of Zn and

formation of Zn phosphate depends on Zn concentration and the available surface area for solid

liquid interactions (Mohammed et al., 2014). Zn is an essential co-enzyme for matrix MMP,

which are fundamental to the progression of the caries process in dentine, notably in destroying

organic matrix after demineralisation by bacterial acids (Chaussain-Miller et al., 2006).

However, some studies have shown that metals including Zn can inhibit MMPs, thereby

reducing MMP-mediated collagen degradation in carious dentine (Henn et al., 2012; Santos et

al., 2004). Toledano et al., found that when Zn was added to dentine beams made from

extracted non-carious molars, which were then demineralised in phosphoric acid, degradation

in demineralised dentine was strongly reduced, as Zn served as an effective inhibitor of MMP-

mediated collagen degradation (Toledano et al., 2012). An in-vitro study investigating the local

environment of Zn atoms in dentine has reported that treatment with Zn compounds made

dentin more acid resistant (Takatsuka et al., 2005).

Abnormalities of Zn availability and metabolism may thus influence susceptibility to dental

caries, justifying further investigation with a larger sample size and specific phenotype

definitions for cases and controls of disease.

The cluster 2 genes, INSIG1, SFT2D3 and SLC17A9, predominantly code for proteins that are

embedded in membranes. Integral (intrinsic) and transmembrane proteins are permanently

attached to cell membranes by interactions between their hydrophobic dimer units and the

phospholipids of the membrane and play vital roles in cellular functions (Mishra et al., 2014).

The passage of solutes such as ions and small molecules through biological membranes are

140

mediated by these proteins, which are also called membrane transport proteins (Mishra et al.,

2014). As far as we are aware, direct associations of this group of genes with dental tissues or

with dental caries have not been reported in the literature. Nevertheless, genes identified in our

study code for proteins that have general functions, such as transportation of small vesicles,

vesicular uptake, storage and secretion of ATP, and fusion of transport vesicles with the Golgi

complex. Whilst there is no published evidence of a direct association with cariogenesis,

general protein functions coded by this group of genes might be involved in the caries process

as proper membrane function is fundamental to the formation and repair of all tissues, and the

secretion of enamel matrix through the ruffled border of ameloblasts could be compromised.

Amongst the genes that had only “one CpG site differentially methylated” between cases and

controls (Table 7.5) and were closer to a gene promoter, two genes, PFKFB3 and ENOSF1,

are part of fructose and mannose metabolism pathways respectively (Ogata et al., 1999). These

pathways are involved in the synthesis, utilization and/or degradation of these sugars (Musen

et al., 2012; Whetzel et al., 2011). Another two genes from this list, BTNL8 and RFPL2, are

associated with PRY and butyrophylin-like domains as defined by SMART (simple modular

architecture research tool) (Schultz et al., 1998). Whist the function of PRY is unknown, its

distant homologues are protein domains (a sealed part of a given protein sequence that can

change, function, and exist independently of the rest of the protein chain) in butyrophilin, the

major protein associated with fat droplets in the milk, and pyrin, a neutrophil protein (Finn et

al., 2017). Interestingly, a group of proteins identified by the PRY domain contain a ring-finger

domain that binds two atoms of Zn (Doerks et al., 2002; Schultz et al., 1998). Several proteins

in the butyrophylin-like domain also contain RING fingers and a well-conserved 40-residue

cysteine-rich domain termed a B-box Zn finger (Borden, 1998).

141

7.5.2 Conclusion

To the best of our knowledge, this is the first EWAS study performed on the human genome

in relation to dental caries. The aim was to identify specific areas differentially methylated

among individuals with high and low dental caries experience. Our research provides evidence

for an association between epigenetic changes and dental caries evaluated through DMR. The

two sets of genes that were identified associated with the DMR between high-caries and low--

caries dyads are of high interest and warrant further investigation of the biological mechanisms

mediated through Zn and membrane proteins. Findings of our pilot study highlight a further

need for investigations on larger and diverse populations to clarify the role of epigenetic factors

in cariogenesis, the mechanisms involved, and how these might be manipulated in caries

prevention on an individual and population level.

Further pyrosequencing on an extended sample of 38 mother-child dyads was conducted in

order to validate the region of interest on chromosome 12 (chromosome region between

11700080 and 11700350). However, there were technical difficulties and the proposed region

could not be replicated. Hence, three different CpG sites within the discovery region were

pyrosequenced. The observed results, however, did not show any significant association

between individual with high and low lifetime cumulative caries experience (Appendix E).

7.5.3 Limitations

The study was unfunded, and hence a larger number of subjects could not be recruited for this

pilot EWAS. Because caries experience in our population was a continuous variable, trait

definitions for cases and controls had to be based on extremes of burden of disease, precluding

142

a standard case-control study design. A very large and expensive study would be necessary to

analyse epigenomic differences across a full range of caries experience.

143

Chapter 8: General Discussion

8.1 Introduction

The purpose of this final chapter is to fulfil three main tasks. First, it summarises the research

background and objectives (Section 8.2). Second, it discusses the findings of the study and the

implications for the prevention of dental caries in children (Section 8.3). Section 8.4 briefly

discusses the limitations of the study. Finally, it discusses strengths and recommendations for

future studies in Section 8.5 and gives concluding remarks in Section 8.6.

8.2 Overview of the study

8.2.1 Background and research objectives

Despite marked improvements over the past century, especially in industrialised countries, poor

oral health is still a significant problem, not least among children (Kassebaum et al., 2015).

Caries is the commonest chronic disease of childhood; nevertheless, oral health researchers

primarily examine influences on the disease process from within the oral cavity, with

consideration of limited individual-level factors to identify risks for the disease (Fisher-Owens

et al., 2007). Individuals are exposed to a multitude of risk indicators throughout their life,

placing them at varying magnitudes of risk. Different communities with their unique

behaviours and practices face different risk conditions; especially this variation is more marked

between industrialised and non-industrialised countries (Attaran et al., 2016). Conceptual

models have been proposed to help in understanding these inequalities, though many do not

144

incorporate individual, family, and community level factors with a multilevel approach which

considers possible interactions among the variables. In the methodology of the thesis we have

proposed a conceptual framework based on genetic and biological factors, the social

environment, health behaviours, and dental and medical care utilisation and their possible

interactions (Figure 3.1) (Fernando et al., 2015). As yet, not many studies reported in the

literature have focused on empirically investigating this concept by recognising the presence

of a complex interplay of causal factors. Therefore, this study was conducted with the objective

of filling this knowledge gap by testing the proposed conceptual framework and identifying

risk indicators in connection to childhood dental caries in a typical industrialised country.

The main objective of this thesis was to identify relationships between maternal,

environmental, and individual-level parameters and the severity and extent of dental caries in

children. This included exploring pathways and interactions of these parameters within a

broader framework containing mothers’ socio-economic situation, oral health status,

knowledge, attitudes and behaviours towards their own and their families’ oral and general

health, children’s intraoral pathophysiology, in utero circumstances, birth weight, and possible

epigenetic modifications.

The study has specifically answered three central research questions:

1. Are there relationships between maternal and environmental risk indicators, SES, BMI,

salivary characteristics, dental caries experience, oral health knowledge and behaviours, and

the caries experience of their children?

2. What are the associations between children’s past dental caries experience, salivary

characteristics, prenatal environment, birth weight, and current experience of dental caries?

145

3. Are there any differences in epigenetic modifications between individuals discordant for

lifetime, cumulative, experience of dental caries?

In the process of addressing these research questions, the study expected to discover a range of

risk indicators, which may or may not be risk factors that influence the process of dental caries

in children, and to quantify this effect in a clinically and epidemiologically meaningful way.

Negative Binomial Regression Modelling (Mota-Veloso et al., 2016) and Structural Equation

Modelling (SEM) (Jeon, 2015) were used to test these associations, as they allow simultaneous

assessment of direct and mediating effects between dependent as well as between independent

variables.

8.3 Main empirical findings

The series of results chapters contain the specific research findings related to the research

questions covering the research gap identified in Chapters 1 and 6. A summary of these results

is presented next.

8.3.1 Dental caries experience in the sample of children and their mothers

The sample of children was aged between 6 to 7 years, (mean 6.3, SD = 0.83) years, and just

over half (51%) were girls. Caries experience was measured using ICDAS criteria. Hence it

was possible to report cumulative caries experience from initial white spot lesions to pulp-

exposed extended cavities. The assumption was made – as is often the case in the literature,

but is not entirely valid – that “white spot” lesions represented “initial” or “early” lesions, and

might thus represent current caries activity. Of the 174 children, 61% were caries-free, and

146

16% had carious lesions limited to enamel (ICDAS = 1 and ICDAS = 2). As the majority of

the children were caries free, exploring risk markers for caries in this population was

challenging. Mean cumulative caries experience, the percentage of surfaces affected by past

and current caries, was 6.5 (SD = 11.07) in the total sample. Mean current caries experience,

the number of surfaces affected with caries (ICDAS > 0) at the time of examination, was 5.39

(SD = 8.90). Mean past caries experience (the number of surfaces with restorations), was 1.29

(SD =4.52) in the population. Out of all children who had caries, 56% were girls. Do and

Spencer (2016) reported that 27% of girls in the same age group in Australia were experiencing

the disease in 2014, and just over 27% of children had untreated caries in their deciduous

dentition (Do and Spencer, 2016). This thesis reports much higher caries prevalence (38%) in

the same age group; however, the prevalence is for deciduous and permanent teeth combined.

It was observed that the mean caries experience of mothers, measured with ICDAS (both

current and past) was 23.64 (SD = 28.28) in the total sample. Mean caries experience of mothers

with children who had caries was 25.29 (SD = 19.16) whereas mothers of children who were

caries free had lower lifetime experience of the disease: 22.60 (SD = 13.13). A similar

observation was made by Do et al. (2017), where 52% of the females aged 35 to 44 years had

experienced caries and mean DMFS was 24.8 (SD = 76.74) (Do et al., 2017). However, the

study was done to evaluate the effectiveness of water fluoridation in the prevention of dental

caries in adults and comparisons between maternal and childhood caries were not reported.

There is no recent information from Australia comparing maternal and children’s caries

experience; hence, findings from this thesis fill this gap in the literature.

147

8.3.2 Oral health knowledge and behaviours of mothers

In the sample investigated, the mean oral health knowledge, attitudes and practice (KAP) score

of mothers was 72.64 (SD =10.10). Surprisingly, mothers’ mean KAP score in both groups,

children with and without caries, was the same (72.6). This is a reasonably high score (Ahmed,

2015), yet the caries experience of the cohort of children was higher than the Australian average

for the same age group. It is acknowledged that the questionnaire used for the study was a self-

administered one and participating mothers had the freedom to answer as they wish. Hence it

can be assumed that even though they did not practice optimal oral hygiene habits, they would

still gave socially acceptable answers to indicate that they practiced good oral hygiene (Ju et

al., 2017).

It is understood that good oral health can be achieved and enjoyed by all through the application

of known preventive regimens and following good dietary and oral health practices.

Nevertheless, the theoretical model that reinforces oral health knowledge and its role in oral

health outcomes are that without functional, applied, and contextual understanding of both oral

health behaviours and oral health services, optimal oral health cannot be reached (Baur et al.,

2005). However, it is questionable if oral health services are designed to establish health since

dentists are paid and evaluated according to the number of treatment procedures performed.

Arguably, dentists can worsen oral health conditions, especially diseases like caries, whereas

early establishment of good diet and good hygiene practices can easily prevent, and even

reverse non-cavitated lesions, through good home care. This concept was advocated in the La

Cascada Declaration, which argues that most individuals are well able to care for their own

oral health, perhaps supported by a cadre of primary health care professionals who would

integrate health promotion, health education, and necessary interventions like vaccination and

148

perhaps provide topical fluoride applications, throughout particular communities. This could

be integrated with other primary health services such as mother and child services, and

vaccination (Cohen et al., 2017). Even though it is a good concept, a policy for integrating oral

health as a part of general health would take time, because health professionals need to be

educated and empowered with the notion to care for the individual without compartmentalising.

8.3.3 Maternal and environmental factors that influence the caries experience of children

One of the objectives of this thesis was to evaluate the effects of maternal and environmental

factors on dental caries experience of children. Based on the literature, maternal SES, measured

with annual household income (Zander et al., 2013), mother’s level of education (Kumar et al.,

2014) and employment status (Kumar et al., 2014), have negative associations with children’s

caries experience. Moreover, marital status (Piva et al., 2017), mother’s poor oral health

knowledge (Lee et al., 2014), practices (Nourijelyani et al., 2014), and age (Warren et al., 2016)

were significant indicators of children’s poor oral health. Maternal BMI, which is a reflection

of personal and lifestyle behaviours such as dietary and health promotion activities has been

associated with dental caries in children in an indirect pathway, through the common risk factor

approach (Wigen and Wang, 2014). Also, mothers who harbour increased levels of oral

cariogenic bacteria have been found to be associated with increased levels of the same bacterial

species (Li et al., 2005) and higher levels of dental caries in their children’s oral cavities

(Chaffee et al., 2014). Furthermore, mothers’ caries status was recognised as one of the main

risk indicators for dental caries in their children (Birungi et al., 2016). Even though there is no

literature available on the association between maternal salivary characteristics (pH, buffering,

and stimulated saliva flow rate) and children’s caries experience, it was assumed that since

mothers associate closely with their children, these features would reflect their children’s caries

levels. Hence it was hypothesised that caries experience in a child population of a typical

149

Western industrialised country is associated with maternal socio-economic, behavioural, and

demographic and intra oral indicators.

Our negative binomial regression model demonstrated that low maternal levels of salivary LB

(ARR = 0.61, p = 0.01), high caries experience (ARR = 1.02, p = 0.01), low annual income

(ARR = 0.67, p = 0.03), and high frequency of giving carbonated drinks to children (ARR =

2.03, p = 0.02) were associated with high caries experience in their children. Observed results

were in line with current literature with regards to high maternal caries experience (Birungi et

al., 2016), low income (Jepsen et al., 2017), high frequency of giving carbonated drinks

(López-Gómez et al., 2016), and initiation of tooth brushing practices (Ngoc et al., 2017). One

possible reason for all selected variables not being significantly associated with childhood

caries could be because only 67 out of 174 children had dental caries, and that number might

not have been adequate to provide enough statistical power. This was a limitation of using an

existing cohort. Moreover, it could be that other mediating factors which were not included in

the models have an impact on childhood dental caries. For example, parenting style (Kumar et

al., 2017), maternal stress, coping strategies (Wendt et al., 1995), and child temperament (Kim

Seow, 2012) have been reported to be risk indicators of caries in children. These data were not

included in the risk factor model because over 50% of participating mothers had not answered

the relevant questions.

8.3.4 Children’s individual factors: prenatal environment, birth weight, and intra oral

characteristics as risk indicators for caries experience

Literature reveals that children’s prenatal environment determined by maternal nutrition and

supplement intake during pregnancy impacts tooth development for the children. For instance,

lack of vitamin D, calcium, and iron during foetal life caused hypoplastic teeth in offspring

150

(Bergel et al., 2010). Moreover, birth weight, an indicator of foetal growth and nutrition, has

been associated with dental caries, where children with low birth weight were more at risk of

developing the disease (Bernabé et al., 2017). As children get older, they are exposed to

environmental risk indicators, and if coupled with poor oral hygiene practices, are more

predisposed to caries (Plaka et al., 2017). Progression of caries is synergized with increased

loads of oral cariogenic bacteria, like MS and LB (Samaranayake, 2012). In addition, it has

been long established that intra oral salivary characteristics, like low pH and low buffering

capacity, create a caries inducing environment (Lenander-Lumikari and Loimaranta, 2000).

Literature is inconsistent with evidence for an association between gender and dental caries

(Chankanka et al., 2011). However, past caries experience has been identified as the strongest

single predictor of current and future caries in children (Attaran et al., 2016). Therefore, to

assess these effects on dental caries experience a pathway model was constructed by

hypothesising that these individual factors: maternal supplement intake during pregnancy (as a

surrogate for prenatal environment), birth weight of the child, salivary buffering, pH, and past

caries experience would influence current dental caries status.

It was observed that children’s previous dental caries experience was significantly associated

with current dental caries experience (β = 0.332, p = 0.018), reinforcing the literature (Attaran

et al., 2016). Moreover, a positive association was also detected between child’s salivary MS

levels and current dental caries experience (β = 0.215, p = 0.032). Edelstein et al. (2017)

reported similar findings in a group of 6-year-old American children: they observed that even

within high SES groups, the most useful predictor of current caries was high salivary MS count

(Edelstein et al., 2016). Interestingly, in our study population, children whose mothers had

reported to have taken iron supplements during pregnancy had lower past caries experience,

which was a direct effect in the path model (β = -0.137, p = 0.068), although the results were

151

not statistically significant. In addition, it was observed that reported maternal intake of iron

supplement during pregnancy was indirectly associated with higher current dental caries

experience of their offspring (β = -0.046, p = 0.051). Mothers of the EFHL study answered

questions during the initial recruitment about prenatal supplement intake (including iron), and

the information was used as an indicator of intra uterine growth and foetal environment

(Cameron et al., 2012). Schmidt et al. followed the same method of data collection during

preconception and prenatal periods to evaluate the maternal nutrition status during gestation

(Schmidt et al., 2011). Supplementation with iron is generally recommended during pregnancy

to meet the iron needs of both mother and foetus (Scholl, 2005) because there is a decrease in

maternal iron stores during pregnancy (Fenton et al., 1977), and oral iron supplements have

been observed to prevents the stores from reaching deficient levels in mothers and thereby in

the foetus (Fenton et al., 1977). To the best of the knowledge of the investigators, there are no

previous studies that have found a link between the reported prenatal maternal intake of iron

and dental caries in their offspring. The observed results in the thesis should be interpreted with

caution, since serum iron levels were not measured in participating mothers and it is not proven

in humans that foetal iron deficiency is carried into the childhood. However, researchers have

linked iron deficiency anaemia to children with caries experience (Abed et al., 2014; Bansal et

al., 2016; Tang et al., 2013).

8.3.5 Epigenome-Wide Association Study of individuals discordant for dental caries: The

pilot study

Another focus of the thesis was to investigate associations between epigenetic changes and

dental caries experience. Despite decades of preventive and curative interventions and

researchers identifying various risk indicators, dental caries continues to remain a public health

problem globally (Kassebaum et al., 2015), and this has motivated investigators to search for

152

possible genetic contributors to the disease (Vieira et al., 2014). In 1988, Borass et al. showed

evidence of a genetic contribution to caries susceptibility by studying monozygotic twins who

were raised apart, showing that there was a significant resemblance to monozygotic twins

raised together with regards to lifetime caries experience. He thereby suggested a genetic

influence on susceptibility to dental caries (Boraas et al., 1988), which was further tested and

confirmed by other scientists (Bretz et al., 2005; Wang et al.; 2012, Shaffer et al., 2012).

Nevertheless, the association between dental caries and a potent single genotype with causal

biological relevance was not established. Hence, a few investigators became keen on exploring

epigenetic modifications, brought about by influences of environmental factors having a long-

term impact on the epigenome (Seo et al., 2015).

A pilot study was conducted to evaluate the epigenetic changes between individuals with high

and low caries experience. To the best of our knowledge, this was the first ever, EWAS,

conducted on dental caries and the results are very promising. In this study, 435508 CpG sites

were tested epigenome-wide. It was identified that there were significant DMR between

persons – both mothers and their child – with high and low caries experience on chromosomes

1, 2, 5, 7, 12, 20, and 22, out of which Chromosome 12 showed the highest average methylation

difference (41%) between high and low caries experience groups (p < 2.17E-103). There were

8 CpG sites differentially methylated between the region of 11700085 and 11700343 on

chromosome 12 within the promoter region of the gene, Long Intergenic Non-Protein Coding

RNA 1252 (LINC01252), which regulates non-coding RNA in humans

(https://www.ncbi.nlm.nih.gov/gene/?term=LINC01252). Annotation and functional analysis

of DMR related to all genes, which was significantly differentially methylated between high

and low caries experience groups, were investigated through the DAVID bioinformatics

functional annotation tool (Huang et al., 2009a).

153

It was observed that the associated genes fell into two functional clusters regulating (i) Zn or

other metal binding ions and (ii) cell membrane proteins. Zn is an essential co-enzyme in

mineral metabolism that regulates demineralization/remineralization equilibrium of dental hard

tissues. Moreover, it is an element that inhibits dentine demineralization (Takatsuka et al.,

2005). Zn acts as a co-enzyme for MMP, which destroy the organic matrix after

demineralization by bacterial acids (Chaussain-Miller et al., 2006). An opposing evidence was

described by Henn et al. (2012) and Santos et al. (2004), where Zn was reported to inhibit MMP

thereby reducing MMP-mediated collagen degradation in carious dentine (Santos et al., 2004;

Henn et al., 2012). Moreover, Zn enhances calcium phosphate precipitation on and in enamel,

both during tooth development and after an attack of caries (Lynch, 2011; Mohammed et al.,

2014). Also, as a divalent cation, Zn is electronically attracted to negatively charged oral hard

tissue surfaces, causing cariogenic bacteria like MS to detach from enamel (Burguera-Pascu et

al., 2007).

The second cluster of genes identified in the current study predominantly code for membrane

proteins that have general functions, such as transportation of small vesicles, vesicular uptake,

storage, and secretion of ATP, and fusion of transport vesicles with the Golgi complex. Even

though a direct relationship with dental caries could not be established, these intrinsic proteins

play a vital role in cellular functions by mediating the passage of solutes (ions, molecules)

through biological membranes (Mishra et al., 2014).

Among the DMR which showed differential methylation of a single CpG between individuals

discordant for caries, and were within proximity of a gene promoter region, two genes,

PFKFB3 and ENOSF, were found to be part of fructose and mannose metabolism pathways

respectively (Ogata et al., 1999). These pathways are implicit in the synthesis, utilisation, and

154

degradation of these sugars (Musen et al., 2011), which might be of importance to the process

of caries by means of providing a substrate for bacterial growth, though direct evidence for

such is not available.

Further deep pyrosequencing of the significant DMR on chromosome 12, between sites

11700080 and 11700350, was conducted on DNA from saliva of an extended sample (n = 76).

There were 38 mother child dyads: 19 dyads with high caries experience and 19 with low caries

experience. The results, however, did not give conclusive evidence of discovery analysis

(Appendix E, Table 1). The targeted sites were found to be located in a non-specific long

terminal repeat region, hence designing primers for the region failed. Therefore, three substitute

target sites with specific primers were selected (Chr12:11700151, Chr12:11700148, and

Chr12:11700124) for analysis. Nevertheless, there was no significant variation in methylation

between individuals with high and low caries experience on the alternative sites chosen

(Appendix E, Table 2 and Table 3).

8.4 Limitations

There are several limitations to highlight. One of the main limitations is that due to restrictions

of time and logistics, a larger sample of mother-child dyads could not be recruited. Moreover,

the response rate of volunteer participants was low, and to achieve the required sample size

data collection continued for over three years. Further, mothers’ oral health related behaviours

were assessed by a self-administered questionnaire with closed-ended questions. Hence,

practices were not observed directly by the investigator. More variables related to maternal

behaviours would have been reliable and perhaps significantly associated with childhood caries

155

if properly observed and objectively recorded by investigators. These limitations are inherent

when tapping into an existing cohort study where questionnaires have been designed and used

for several years, and substantial volumes of data have been collected. Included with this

limitation is that data on the prenatal environment were also based on answers provided by

participants and serum levels of prenatal supplements were unavailable to support the data.

Many variables related to the prenatal environment could not be included in data analysis as

most of the participating mothers have answered “not relevant” to the use of certain

supplements, alcohol consumption, and smoking during pregnancy. Finally, there were only

12 dyads (24 participants) selected for the pilot study of EWAS, due to lack of funding. Further

research using a larger representative sample is required to validate the discovered findings.

8.5 Discussion and recommendations for future studies

8.5.1Strengths of the study

Despite the limitations, the study explored pathways linking maternal characteristics,

children’s socio-economic and family environment, children’s prenatal environment, birth

weight, and epigenetic changes with childhood caries are the novel contributions of this thesis.

Whilst several associations of maternal, environmental, and child factors with regards to dental

caries in children have already been established (Ersin et al., 2005; Ju et al. 2016; Kim Seow,

2012), this thesis adds to knowledge on risk indicators for dental caries experience in children

and emphasises the possible role of prenatal environment on susceptibility to childhood caries.

The thesis followed the conceptual frame work proposed in chapter three, incorporating

community level, family level, and child level including epigenetic endowment and intra oral

156

characteristics of children (Fernando et al., 2015). The findings are imperative for a population

of children in a Western industrialised country since the study area is culturally and

geographically similar to other states of Australia and to many similar populations across the

globe. Further, the study adds much-needed evidence for an association between epigenetic

changes and dental caries experience. However, the study findings might not be generalizable

to a child population in low- and middle-income countries. Further, it needs to be understood

that the associations extrapolated in this thesis might not be causal owing to the cross-sectional

design of the study.

8.5.2 Future research directions

The findings from the thesis indicate that the effect of maternal oral health related behaviours

on children’s caries levels is persistent throughout infancy and childhood as maternal

perseverance and responsiveness influence oral health-related behaviours of their children

(Tiwari et al., 2017). Therefore, future research needs to consider collecting behavioural data

by naturalistic observation, where the participants’ behaviours are observed in their natural

environment (Jha et al., 2017) or by the direct observational method (Dart et al., 2016), since

these methods provide unbiased information.

Maternal caries status has been found in this thesis to be associated with children’s oral health

status; this effect has been observed to continue into adulthood by others (Shearer et al., 2011).

These pathways of association could be validated by conducting studies that are prospective

and longitudinal that examine multi-level determinants of oral diseases (Cameron et al., 2012).

The best method would be to recruit participants from early pregnancy and follow them through

childhood to adulthood. However, this is clearly a very expensive operation, and one which

cannot be accomplished within a PhD timespan. An alternative would be to use existing data

157

on potential determinants of dental caries (such as records from hospitals with dental outpatient

facilities), and explore associations with current disease status by conducting retrospective

cohort studies (Lai et al., 1997; Sedgwick, 2014).

An interesting finding from the thesis was that reported maternal intake of iron during

pregnancy has an association with their children’s caries experience: these children had

comparatively low levels of caries. Nevertheless, there is a need to use serum levels of these

supplements to reaffirm data obtained via information provided by participants. There is further

evidence for a positive relationship with other supplement use during pregnancy, for example,

vitamin A, vitamin D, and calcium and childhood dental caries (Bergel et al., 2010), indicating

the need to integrate dental and non-dental health care providers to reduce the burden of oral

diseases. However, research shows limited emphasis being placed on oral health by non-dental

professionals in Australia, and hence there is a need for further education and empowering of

general medical practitioners in this area (George et al., 2014). Therefore, there is a requisite

to integrate dentistry with medicine and the health care system as a whole to provide greater

research support for an oral-systemic health connection including paediatricians, pedodontists,

physicians, gynaecologists, nurses, and midwives (Cohen et al., 2017). Furthermore, since

dental caries shares common risk factors with a number of systemic conditions (Williams,

2011), and the interventions for improving prenatal nutrition of mothers have a positive effect

in preventing systemic diseases of the offspring (Roseboom et al., 2001, De Boo and Harding,

2006, Barker et al., 2002), there is a high likelihood that these strategies can also be used in

preventing dental caries. Therefore, it is logical to have a common risk factor approach rather

than oral disease-specific approach, which would be a more effective and cost-effective method

of utilising human and financial resources.

158

A highlight of the thesis was the results observed for associations between epigenetic changes

and caries experience of individuals. However, the pilot study used only 24 individuals for this

discovery. In EWAS, power depends on several factors: study design, sample size, effect size,

and correction for multiple testing. Also the longitudinal stability of the epigenetic markers and

their variance within a biological sample also matter, as epigenetic signals in a biological

sample from one individual represent frequency measures from a population of cells (Tsai and

Bell, 2015). To date EWAS have predominantly focused on DNA methylation, identifying

many differentially methylated positions (DMPs), DMR, and allele-specific methylation

(ASM) regions related to complex diseases (Liu et al., 2013). To achieve an 80% power to

detect DMRs at epigenome-wide significance, at least 100 cases and 100 controls are required

(Tsai and Bell, 2015). Therefore, future research is needed to confirm the findings of the thesis

from a population with similar demographic and geographic backgrounds. However, site-

specific DNA methylation analysis does not provide a global picture of DNA methylation

changes within a genome. Hence future researchers need to focus on global assays that could

study multiple DNA repetitive elements, such as Alu, long interspersed nucleotide elements

(LINE 1) (Yang et al., 2004), or SAT2 (Delgado-Cruzata et al., 2014), as these global assays

cover multiple transposable regions throughout the genome. Moreover, successful genetic

studies require careful measurement of caries phenotypes that define cases and controls. For

example, caries measured by ICDAS criteria to delineate a control as a person who has ICDAS

= 0 on all their tooth surfaces and a case with ICDAS > 0 on one or more of their tooth surfaces.

159

8.6 Conclusions

In conclusion, the findings from this study of SEQ children and their mothers demonstrate that

maternal oral health behaviours, income, and maternal caries experience are significantly

associated with, and probably, directly and/or indirectly, exert a significant influence on caries

susceptibility of their children. Children, in particular, are more at risk of developing caries if

they had high loads of salivary MS, past caries experience, and if their mothers have reported

not to take prenatal iron supplements. One of the main findings of the study was discovering

an association between epigenetic modifications and dental caries experience. The results of

this study are relevant to oral health policy makers in order to make decisions to reduce the

burden of dental caries in children, and especially for health care managers who have to decide

how to use scarce resources in combination with non-dental professionals while maximising

chances of preventing dental caries in children. The findings are also relevant for dental

professionals who seek risk indicators for childhood caries to help them design preventive

interventions. Oral health professionals who wish to reduce the burden of caries in children

need to involve mothers from the prenatal period by providing advice on the importance of

supplement intake and maintaining a healthier lifestyle. The concept of epigenetics in the field

of caries research is in its infancy, and dental professionals are unaware of the epigenetic

endowment on caries progression and the underlying biology. They need to be educated on the

environmental stimuli that impact the epigenome and ultimately influence the development of

dental caries. Hence, proper history taking during clinical procedures and explicit maintenance

of records are critical in identifying high-risk mothers and thereby high-risk children.

Moreover, the whole family need to be engaged in oral health friendly behaviours as the

children’s behaviours are influenced by parents. Clinicians can use the knowledge thus

160

obtained in assessing the family circumstances that would increase the risk of dental caries in

children.

161

References

Abed NT, Aly IA, Deyab SM, Ramoon FM (2014). The relation between early dental caries

and iron-deficiency anaemia in children. Med Res J 13(2):108-114.

Aframian D, Davidowitz T, Benoliel R (2006). The distribution of oral mucosal pH values in

healthy saliva secretors. Oral Dis 12 (4):420-423.

Ahirwar SS, Gupta M, Gupta G, Singh V (2017). Screening, Isolation and Identification of

Lactobacillus Species from Dental Caries of Children. Int J Curr Microbiol App Sci 6(1):497-

503.

Ahmed MS (2015). Oral health knowledge and attitude among primary school teachers of

Madinah, Saudi Arabia. J Contemp Dent Pract 16 (4):275-279.

Aida J, Ando Y, Oosaka M, Niimi K, Morita M (2008). Contributions of social context to

inequality in dental caries: a multilevel analysis of Japanese 3‐ year‐ old children. Community

Dent Oral Epidemiol 36(2):149-156.

Aida J, Matsuyama Y, Tabuchi T, Komazaki Y, Tsuboya T, Kato T et al. (2017). Trajectory of

social inequalities in the treatment of dental caries among preschool children in Japan.

Community Dent Oral Epidemiol DOI: 10.1111/cdo e.12304 (2 April 2017).

[AIHW] Australian Institute of Health and Welfare (2014). Oral health and dental care in

Australia: key facts and figures trends. Canberra. http://www.aihw.gov.au/publication-

detail/?id=60129548265 (Accessed on 10 December 2016)

Alves KMRP, Fancoa KS, Sassaki KT, Rabelo AM, Buzala MAR, DElbem ACB (2011). Effect

of iron on enamel demineralization and remineralization in vitro. Arch Oral Biol 56(11): 1192-

119.

[ARCPOH] Australian Research Centre for Population Oral Health (2009) Geographic

variation in oral health and use of dental services in the Australian population 2004–06. (Dental

Statistics and Research Unit: Adelaide, SA)

http://www.aihw.gov.au/WorkArea/DownloadAsset.aspx?id=6442455420 (Accessed on 25

April 2014).

[ARCPOH] Australian Research Centre for Population Oral Health (2014). The Beginning of

Change- Queensland Child Oral Health Survey 2010-2012.

https://publications.qld.gov.au/storage/f/2014-08-06T03%3A11%3A44.862Z/oral-health-

survey-2010-12.pdf (Accessed on 18 June 2015)

Al-Shalan T, Erickson P, Hardie N (1997). Primary incisor decay before age 4 as a risk factor

for future dental caries. Pediatr Dent 19(1):37-41.

Alkhateeb AA, Mancl LA, Presland RB, Rothen ML, Chi DL (2017). Unstimulated Saliva-

Related Caries Risk Factors in Individuals with Cystic Fibrosis: A Cross-Sectional Analysis of

Unstimulated Salivary Flow, pH, and Buffering Capacity. Caries Res 51(1):1-6.

162

Allum F, Shao X, Guénard F, Simon M-M, Busche S, Caron M et al. (2015). Characterization

of functional methylomes by next-generation capture sequencing identifies novel disease-

associated variants. Nat Commun: DOI: 10.1038/ncomms8211 (29 May 2015).

Almerich Silla JM, Boronat Ferrer M, Iranzo Cortés JE (2014). Caries prevalence in children

from Valencia (Spain) using ICDAS II criteria, 2010. Med Oral Patol Oral Cir Bucal 19

(6):574-580.

Altun C, Guven G, Orkunoglu F, Cehreli ZC, Karaaslan A, Basak F et al. (2008). Human

leukocyte antigen class II alleles and dental caries in a child population. Pediatr Dent

30(2):154-159.

Andrews S (2013). FastQC: A quality control tool for high throughput sequence data. Available

at: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/

Arantes R, Frazão P (2016). Income as a Protective Factor for Dental Caries among Indigenous

People from Central Brazil. J Health Care Poor Underserved 27(1):81-89.

Armstrong WD (1948). An Evaluation of the Role of Vitamins and Minerals in the Control of

Caries. J Dent Res 27(3):376-396.

Ashkanani F, Al-Sane M (2012). Knowledge, attitudes and practices of caregivers in relation

to oral health of preschool children. Med Princ Pract 22(2):167-172.

Asif Z, Raza SM, Rana N, Zafar T, Zehra A (2015). An Assessment of Association between

Carbonated Drink Consumption and Dental Caries Prevalence: A Cross-Sectional Study. J Isl

Int Med Col 10(2):168-172.

Attaran N, Khoshnevisan MH, Ghorbani Z, Pakkhesal M, Dehghanian D (2016). Dental Caries

Predictors in Countries with Different Human Development Index: A Review of Articles. J Int

Oral Health 8(2):182-190.

Ayad M, Van Wuyckhuyse B, Minaguchi K, Raubertas R, Bedi G, Billings R et al. (2000). The

association of basic proline-rich peptides from human parotid gland secretions with caries

experience. J Dent Res 79(4):976-982.

Azad N, Zahnow CA, Rudin CM, Baylin SB (2013). The future of epigenetic therapy in solid

tumours—lessons from the past. Nat Rev Clin Oncol 10(5):256-266.

Azevedo LF, Pecharki GD, Brancher JA, Cordeiro CA, Jr., Medeiros KG, Antunes AA et al.

(2010). Analysis of the association between lactotransferrin (LTF) gene polymorphism and

dental caries. J Appl Oral Sci 18(2):166-170.

Bagherian A, Nematollahi H, Afshari JT, Moheghi N (2008). Comparison of allele frequency

for HLA-DR and HLA-DQ between patients with ECC and caries-free children. J Indian Soc

Pedod Prev Dent 26(1):18-21.

Bansal K, Goyal M, Dhingra R (2016). Association of severe early childhood caries with iron

deficiency anemia. J Indian Soc Pedod Prev Dent 34(1):36.

163

Bardow A, Moe D, Nyvad B, Nauntofte B (2000). The buffer capacity and buffer systems of

human whole saliva measured without loss of CO2. Arch Oral Biol 45(1):1-12.

Bardow A, Pedersen A, Nauntofte B (2004). Saliva. In: MilesTS, NauntofteB, SvenssonP eds.

Clinical Oral Physiology. Quintessence: Copenhagen, pp. 17–18, 30–33.

Barker D, Thornburg K (2013). Placental programming of chronic diseases, cancer and

lifespan: a review. Placenta 34(10):841-845.

Barker DJ, Eriksson JG, Forsén T, Osmond C (2002). Fetal origins of adult disease: strength

of effects and biological basis. Int J Epidemiol 31(6):1235-1239.

Barker DJP (1998). Mothers, babies and health in later life. Churchill Livingstone, Edinburg.

Barros S, Offenbacher S (2009). Epigenetics: connecting environment and genotype to

phenotype and disease. J Dent Res 88(5):400-408.

Baur C, Comings J, Evans C, Garcia R, Horowitz A, Ismail A et al. (2005). The invisible

barrier: literacy and its relationship with oral health. J Public Health Dent 65(3):174-182.

Beaty TH, Ruczinski I, Murray JC, Marazita ML, Munger RG, Hetmanski JB et al. (2011).

Evidence for gene‐ environment interaction in a genome wide study of nonsyndromic cleft

palate. Genet Epidemiol 35(6):469-478.

Belinsky SA, Palmisano WA, Gilliland FD, Crooks LA, Divine KK, Winters SA et al. (2002).

Aberrant promoter methylation in bronchial epithelium and sputum from current and former

smokers. Cancer Res 62(8):2370-2377.

Bergandi L, Defabianis P, Re F, Preti G, Aldieri E, Garetto S et al. (2007). Absence of soluble

CD14 in saliva of young patients with dental caries. Eur J Oral Sci 115(2):93-96.

Bergel E, Gibbons L, Rasines MG, Luetich A, Belizán JM (2010). Maternal calcium

supplementation during pregnancy and dental caries of children at 12 years of age: follow-up

of a randomized controlled trial. Acta Obstet Gynecol Scand 89(11):1396-1402.

Bernabé E, Sheiham A, Sabbah W (2009). Income, income inequality, dental caries and dental

care levels: an ecological study in rich countries. Caries Res 43(4):294-301.

Bernabé E, MacRitchie H, Longbottom C, Pitts N, Sabbah W (2017). Birth weight,

breastfeeding, maternal smoking and caries trajectories. J Dent Res 96(2):171-178.

Bestor TH (2000). The DNA methyltransferases of mammals. Hum Molec Genet 9(16):2395-

2402.

Billings RJ, Berkowitz RJ, Watson G (2004). Teeth. Pediatrics 113(Supplement 3):1120-1127.

Birungi N, Fadnes LT, Nankabirwa V, Tumwine JK, Åstrøm AN (2016). Caretaker’s caries

experience and its association with early childhood caries and children’s oral health-related

quality of life: A prospective two-generation study. Acta Odontol Scand 74(8):605-612.

164

Boraas J, Messer L, Till M (1988). A genetic contribution to dental caries, occlusion, and

morphology as demonstrated by twins reared apart. J Dent Res 67(9):1150-1155.

Borden KL (1998). RING fingers and B-boxes: Zn-binding protein-protein interaction

domains. Biochem Cell Biol 76(2-3):351-358.

Bouwman LH, Roep BO, Roos A (2006). Mannose-binding lectin: clinical implications for

infection, transplantation, and autoimmunity. Hum Immunol 67(4):247-256.

Brambilla E, Felloni A, Gagliani M, Malerba A, Strohmemger D (1998). Caries prevention

during pregnancy. J Am Den Assoc 129(871-877.

Brennan D, Spencer A (2014). Childhood oral health and SES predictors of caries in 30-year-

olds. Caries Res 48(3):237-243.

Bretz WA, Corby PM, Schork NJ, Robinson MT, Coelho M, Costa S et al. (2005). Longitudinal

analysis of heritability for dental caries traits. J Dent Res 84(11):1047-1051.

Bretz WA, Corby PM, Melo MR, Coelho MQ, Costa SM, Robinson M et al. (2006).

Heritability estimates for dental caries and sucrose sweetness preference. Arch Oral Biol

51(12):1156-1160.

Burguera-Pascu M, Rodríguez-Archilla A, Baca P (2007). Substantivity of Zn salts used as

rinsing solutions and their effect on the inhibition of Streptococcus mutans. J Trace Elem Med

Biol 21(2):92-101.

Burne RA, Marquis RE (2000). Alkali production by oral bacteria and protection against dental

caries. FEMS Microbiol Lett 193(1):1-6.

Cai F, Manton D, Shen P, Walker G, Cross K, Yuan Y et al. (2007). Effect of addition of citric

acid and casein phosphopeptide-amorphous calcium phosphate to a sugar-free chewing gum

on enamel remineralization in situ. Caries Res 41(5):377-383.

Cameron CM, Scuffham PA, Shibl R, Ng S, Scott R, Spinks A et al. (2012a). Environments

For Healthy Living (EFHL) Griffith birth cohort study: characteristics of sample and profile of

antenatal exposures. BMC Public Health 12(1):1080. DOI. 10.1186/1471-2458-12-1080 (15

December 2012).

Cameron CM, Scuffham PA, Spinks A, Scott R, Sipe N, Ng S et al. (2012b). Environments for

healthy living (EFHL) griffith birth cohort study: background and methods. Matern Child

Health J 16(9):1896-1905.

Capurro DA, Iafolla T, Kingman A, Chattopadhyay A, Garcia I (2015). Trends in income‐related inequality in untreated caries among children in the United States: findings from

NHANES I, NHANES III, and NHANES 1999–2004. Community Dent Oral Epidemiol

43(6):500-510.

Cardoso FP, Viana MB, Sobrinho APR, Diniz MG, Brito JAR, Gomes CC et al. (2010).

Methylation Pattern of the IFN-γ Gene in Human Dental Pulp. J Endod 36(4):642-646.

165

Cardoso FP, de Faria Amormino SA, Dutra WO, Sobrinho APR, Moreira PR (2014).

Methylation pattern of the CD14 and TLR2 genes in human dental pulp. J Endod 40(3):384-

386.

Carlson DS (2005).Theories of craniofacial growth in the postgenomic era. Semin Orthod

11(4):172-183.

Carvalho JC, Silva EF, Vieira EO, Pollaris A, Guillet A, Mestrinho HD (2014). Oral health

determinants and caries outcome among non-privileged children. Caries Res 48(6):515-523.

Carvalho Sales-Peres SHd, Goya S, Moreira de Freitas Sant'Anna R, Mendes Silva H, Carvalho

Sales-Peres Ad, Pianta Rodrigues da Silva R et al. (2010). Prevalência de sobrepeso e

obesidade e fatores associados em adolescentes na região centro-oeste do estado de São Paulo

(SP, Brasil). Ciência & Saúde Coletiva 15(2):3175-3184.

Casanova-Rosado AJ, Medina-Solís CE, Casanova-Rosado JF, Vallejos-Sánchez AA,

Maupomé G, Ávila-Burgos L (2005). Dental caries and associated factors in Mexican

schoolchildren aged 6–13 years. Acta Odontol Scand 63(4):245-251.

Chaffee B, Gansky S, Weintraub J, Featherstone J, Ramos-Gomez F (2014). Maternal oral

bacterial levels predict early childhood caries development. J Dent Res 93(3):238-244.

Chaffee BW, Rodrigues PH, Kramer PF, Vítolo MR, Feldens CA (2017). Oral health‐ related

quality‐ of‐ life scores differ by socio-economic status and caries experience. Community

Dent Oral Epidemiol 45(3):216-224.

Chankanka O, Cavanaugh JE, Levy SM, Marshall TA, Warren JJ, Broffitt B et al. (2011).

Longitudinal associations between children's dental caries and risk factors. J Public Health

Dent 71(4):289-300.

Chaussain-Miller C, Fioretti F, Goldberg M, Menashi S (2006). The role of matrix

metalloproteinases (MMPs) in human caries. J Dent Res 85(1):22-32.

Cheng J, Campbell K (2016). Caries and dental erosion: are Soroti children and adolescents at

risk from increased soft-drink availability in Uganda? Afr Health Sci 16(4):943-946.

Childers NK, Momeni SS, Whiddon J, Cheon K, Cutter GR, Wiener HW et al. (2017).

Association Between Early Childhood Caries and Colonization with MSGenotypes From

Mothers. Pediatr Dent 39(2):130-135.

Chrisopoulos S, Harford J, Ellershaw A (2016). Oral health and dental care in Australia: key

facts and figures.

https://digital.library.adelaide.edu.au/dspace/bitstream/2440/98160/2/hdl_98160.pdf

(Accessed on 11 November 2016).

Cohen L, Dahlen G, Escobar A, Fejerskov O, Johnson NW, Manji F (2017). La Cascada

Declaration Colombia. Aust Dent J: DOI: 10.1111/adj.12546 (9 August 2017). Available at

https://lacascada.pressbooks.com

166

[CICDAS] Committee International Caries Detection and Assessment System (2005). Criteria

Manual; International Caries Detection and Assessment System (ICDAS II) Baltimore,

Maryland, USA

https://www.icdas.org/uploads/Rationale%20and%20Evidence%20ICDAS%20II%20Septem

ber%2011-1.pdf (Accessed on September 2012).

Cortellazzi KL, da Silva Tagliaferro EP, Pereira SM, Bovi Ambrosano GM, Guerra LM, de

Lima Vazquez F et al. (2013). A cohort study of caries incidence and baseline socio-economic,

clinical and demographic variables: a Kaplan-Meier survival analysis. Oral Health Prev Dent

11(4): 349-358.

Craig JM (2013). Epigenetics in Twin Studies. Medical Epigen 1(1):78-87.

Crall JJ, Edelstein B, Tinanoff N (1990). Relationship of microbiological, social, and

environmental variables to caries status in young children. Pediatr Dent 12(4):233-236.

Da Fonseca MA, Avenetti D (2017). Social Determinants of Pediatric Oral Health. Dent Clin

N Am 61: 519–532.

Daly AK, Day CP (2001). Candidate gene case‐ control association studies: advantages and

potential pitfalls. Br J Clin Pharmacol 52(5):489-499.

Dart EH, Radley KC, Briesch AM, Furlow CM, Cavell HJ (2016). Assessing the Accuracy of

Classwide Direct Observation Methods: Two Analyses Using Simulated and Naturalistic Data.

Behav Disord 41(3):148-160.

Dave Singh G, Callister JD (2013). Effect of a maxillary appliance in an adult with obstructive

sleep apnea: A case report. Cranio 31(3):171-175.

Davis CD, Uthus EO (2004). DNA methylation, cancer susceptibility, and nutrient interactions.

Exp Biol Med (Maywood) 229(10):988-995.

Dawes C (2008). Salivary flow patterns and the health of hard and soft oral tissues. J Am Dent

Assoc 139(supplement 2):18S-24S.

De-Regil LM, Harding KB, Roche ML (2016). Preconceptional Nutrition Interventions for

Adolescent Girls and Adult Women: Global Guidelines and Gaps in Evidence and Policy with

Emphasis on Micronutrients. J Nutr : DOI: 10.3945/jn.115.223487 (8 June 2016).

De Boo HA, Harding JE (2006). The developmental origins of adult disease (Barker)

hypothesis. Aust N Z J Obstet Gynaecol 46(1):4-14.

Declerck D, Leroy R, Martens L, Lesaffre E, Garcia‐ Zattera MJ, Broucke SV et al. (2008).

Factors associated with prevalence and severity of caries experience in preschool children.

Community Dent Oral Epidemiol 36(2):168-178.

Deeley K, Letra A, Rose E, Brandon C, Resick J, Marazita M et al. (2008). Possible association

of amelogenin to high caries experience in a Guatemalan-Mayan population. Caries Res

42(1):8-13.

167

De Gauw JH, Costa LMM ,Silva RN, Santos NB, Tenorio MDH (2017). Evaluation of the

effect of ferrous sulphate on enamel deminaralization of human deceduous teeth : an in vitro

study. Journals Bahiana DOI: 10.17267/2238-2720revbahianaodonto.v8i3.1549( 09/29/2017)

Delgado-Cruzata L, Vin-Raviv N, Tehranifar P, Flom J, Reynolds D, Gonzalez K et al. (2014).

Correlations in global DNA methylation measures in peripheral blood mononuclear cells and

granulocytes. Epigenetics 9(11):1504-1510.

Deutsch D, Leiser Y, Shay B, Fermon E, Taylor A, Rosenfeld E et al. (2002). The human

tuftelin gene and the expression of tuftelin in mineralizing and nonmineralizing tissues.

Connect Tissue Res 43(2-3):425-434.

Diaz de Guillory C, Schoolfield JD, Johnson D, Yeh CK, Chen S, Cappelli DP et al. (2014).

Co‐ Relationships between glandular salivary flow rates and dental caries. Gerodontology

31(3):210-219.

Dige I, Grønkjær L, Nyvad B (2014). Molecular studies of the structural ecology of natural

occlusal caries. Caries Rres 48(5):451-460.

Ditmyer M, Dounis G, Mobley C, Schwarz E (2010). A case-control study of determinants for

high and low dental caries prevalence in Nevada youth. BMC Oral Health 10(1):24.

Do L, Spencer AJ (2014). The Beginning of Change: Queensland Child Oral Health Survey

2010-2012: Australian Research Centre for Population Oral Health, The University of Adelaide

Press.

Do L, Spencer A (2016). Oral health of Australian children. The National Child Oral Health

Study 2012–14 Adelaide (Australia): University of Adelaide Press.

Do L, Ha D, Peres MA, Skinner J, Byun R, Spencer AJ (2017). Effectiveness of water

fluoridation in the prevention of dental caries across adult age groups. Community Dent Oral

Epidemiol 45(3):225-232.

Doerks T, Copley RR, Schultz J, Ponting CP, Bork P (2002). Systematic identification of novel

protein domain families associated with nuclear functions. Genome Res 12(1):47-56.

dos Santos Junior VE, de Sousa RMB, Oliveira MC, de Caldas Junior AF, Rosenblatt A (2014).

Early childhood caries and its relationship with perinatal, socio-economic and nutritional risks:

a cross-sectional study. BMC Oral Health 14(1):47.

dos Santos Pinto G, de Ávila Quevedo L, Britto Correa M, Sousa Azevedo M, Leão Goettems

M, Tavares Pinheiro R et al. (2016). Maternal Depression Increases Childhood Dental Caries:

A Cohort Study in Brazil. Caries Res 51(1):17-25.

Douglass JM, Li Y, Tinanoff N (2008). Association of mutans streptococci between caregivers

and their children. Pediatr Dent 30(5):375-387.

Duhaime-Ross A (2014). Revved-up epigenetic sequencing may foster new diagnostics. Nat

Med 20(1):2-2.

168

Duijster D, Verrips G, Loveren C (2014). The role of family functioning in childhood dental

caries. Community Dent Oral Epidemiol 42(3):193-205.

Dye BA, Thornton-Evans G, Li X, Iafolla TJ (2015). Dental caries and sealant prevalence in

children and adolescents in the United States, 2011-2012: US Department of Health and

Human Services, Centers for Disease Control and Prevention, National Center for Health

Statistics.

Eddie Ip W, Takahashi K, Alan Ezekowitz R, Stuart LM (2009). Mannose‐ binding lectin and

innate immunity. Immunol Rev 230(1):9-21.

Edelstein BL, Ureles SD, Smaldone A (2016). Very High Salivary MSPredicts Caries

Progression in Young Children. Pediatr Dent 38(4):325-330.

Eichler EE, Flint J, Gibson G, Kong A, Leal SM, Moore JH et al. (2010). Missing heritability

and strategies for finding the underlying causes of complex disease. Nat Rev Genet 11(6):446-

450.

El-kwatehy W, Youssef A (2016). Salivary Biomarkers in Caries Affected and Caries Free

Children. Int J Dentistry Oral Sci 3(10):348-352.

ElSalhy M, Honkala S, Söderling E, Varghese A, Honkala E (2013). Relationship between

daily habits,< i> Streptococcus mutans</i>, and caries among schoolboys. J Dent 41(11):1000-

1006.

Ersin NK, Eronat N, Cogulu D, Uzel A, Aksit S (2005). Association of maternal-child

characteristics as a factor in early childhood caries and salivary bacterial counts. J Dent Child

(Chic) 73(2):105-111.

Fatima T, Rahim ZBHA, Lin CW, Qamar Z (2016). Zn: A precious trace element for oral health

care? J Pak Med Assoc 66:1019-1023.

Fejerskov O, Kidd E (2008). Dental caries: the disease and its clinical management. 2nd Edn.

ed. New Jersey: Wiley-Blackwell.

Fellermann K, Stange EF (2001). Defensins–innate immunity at the epithelial frontier. Eur J

Gastroenterol Hepatol 13(7):771-776.

Fenton V, Cavill I, Fisher J (1977). Iron stores in pregnancy. Br J Haematol 37(1):145-149.

Fernando S, Speicher DJ, Bakr MM, Benton MC, Lea RA, Scuffham PA et al. (2015). Protocol

for assessing maternal, environmental and epigenetic risk factors for dental caries in children.

BMC Oral Health 15(1):167. DOI.org/10.1186/s12903-015-0143-2 (29 December 2015).

Ferraz dos Santos B, Dabbagh B, Daniel SJ, Schwartz S (2016). Association of onabotulinum

toxin A treatment with salivary pH and dental caries of neurologically impaired children with

sialorrhea. Int J Paediatr Dent 26(1):45-51.

Finn RD, Attwood TK, Babbitt PC, Bateman A, Bork P, Bridge AJ et al. (2017). InterPro in

2017—beyond protein family and domain annotations. Nucleic Acids Res 45(1):D190-D199.

169

Fisher-Owens SA, Gansky SA, Platt LJ, Weintraub JA, Soobader M-J, Bramlett MD et al.

(2007). Influences on children's oral health: a conceptual model. Pediatrics 120(3):e510-e520.

DOI: 10.1542/peds.2006-3084 (2 March 2007).

https://s100.copyright.com/AppDispatchServlet copyright order link

Folayan MO, Kolawole KA, Oziegbe EO, Oyedele T, Oshomoji OV, Chukwumah NM et al.

(2015). Prevalence, and early childhood caries risk indicators in preschool children in suburban

Nigeria. BMC Oral Health 15(1):72.

Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML et al. (2005). Epigenetic

differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A

102(30):10604-10609.

Fushan AA, Simons CT, Slack JP, Manichaikul A, Drayna D (2009). Allelic polymorphism

within the TAS1R3 promoter is associated with human taste sensitivity to sucrose. Curr Biol

19(15):1288-1293.

Gao X, Jiang S, Koh D, Hsu CYS (2016). Salivary biomarkers for dental caries. Periodontol

2000 70(1):128-141.

Garcia CA, Ahmadian A, Gharizadeh B, Lundeberg J, Ronaghi M, Nyrén P (2000). Mutation

detection by pyrosequencing: sequencing of exons 5–8 of the p53 tumor suppressor gene. Gene

253(2):249-257.

George A, Duff M, Johnson M, Dahlen H, Blinkhorn A, Ellis S et al. (2014). Piloting of an

oral health education programme and knowledge test for midwives. Contemp Nurse 46(2):180-

186.

Ghazal T, Levy SM, Childers NK, Broffitt B, Cutter GR, Wiener HW et al. (2015). Factors

associated with early childhood caries incidence among high caries‐ risk children. Community

Dent Oral Epidemiol 43(4):366-374.

Goldberg AD, Allis CD, Bernstein E (2007). Epigenetics: a landscape takes shape. Cell

128(4):635-638.

Gomez RS, Dutra WO, Moreira PR (2009). Epigenetics and periodontal disease: future

perspectives. Inflammation Res 58(10):625-629.

Gopinath V, Arzreanne A (2006). Saliva as a diagnostic tool for assessment of dental caries.

Arch Orofac Sci 1:57-59.

Gross EL, Leys EJ, Gasparovich SR, Firestone ND, Schwartzbaum JA, Janies DA et al. (2010).

Bacterial 16S sequence analysis of severe caries in young permanent teeth. J Clin Microbiol

48(11):4121-4128.

Guo L, Shi W (2013). Salivary biomarkers for caries risk assessment. J Calif Dent Assoc

41(2):107.

170

Ha D, Roberts-Thomson KF, Armfield JM (2011). The child dental health surveys Australia,

2005 and 2006: Australian Institute of Health and Welfare, Dental Statistics and Research Unit.

http://www.aihw.gov.au/WorkArea/DownloadAsset.aspx?id=10737420632 (Accessed on 8

February 2014).

Hajishengallis E, Parsaei Y, Klein MI, Koo H (2017). Advances in the microbial etiology and

pathogenesis of early childhood caries. Mol Oral Microbiol 32(1):24-34.

Hall-Scullin E, Whitehead H, Milsom K, Tickle M, Su T-L, Walsh T (2017). Longitudinal

study of caries development from childhood to adolescence. J Dent Res 96(7):762-767.

Han DH, Kim DH, Kim MJ, Kim JB, Jung‐ Choi K, Bae KH (2014). Regular dental checkup

and snack–soda drink consumption of preschool children are associated with early childhood

caries in Korean caregiver/preschool children dyads. Community Dent Oral Epidemiol

42(1):70-78.

Hardy J, Singleton A (2009). Genomewide association studies and human disease. N Engl J

Med 360(17):1759-1768.

Harris R, Nicoll AD, Adair PM, Pine CM (2004). Risk factors for dental caries in young

children: a systematic review of the literature. Community Dent Health 21(1):71-85.

Heaton B, Crawford A, Garcia RI, Henshaw M, Riedy CA, Barker JC et al. (2017). Oral health

beliefs, knowledge, and behaviors in Northern California American Indian and Alaska Native

mothers regarding early childhood caries. J Public Health Dent: DOI: 10.1111/jphd.12217 (27

April 2017).

Henn S, de Carvalho RV, Ogliari FA, de Souza AP, Line SRP, da Silva AF et al. (2012).

Addition of Zn methacrylate in dental polymers: MMP-2 inhibition and ultimate tensile

strength evaluation. Clin Oral Investig 16(2):531-536.

Hillemacher T, Frieling H, Moskau S, Muschler MA, Semmler A, Kornhuber J et al. (2008).

Global DNA methylation is influenced by smoking behaviour. Eur Neuropsychopharmacol

18(4):295-298.

Hooley M, Skouteris H, Boganin C, Satur J, Kilpatrick N (2012). Parental influence and the

development of dental caries in children aged 0–6 years: a systematic review of the literature.

J Dent 40(11):873-885.

Hoyle R, Panter A (1995). Writing about structural equation modeling. In: Structural equation

modeling Concepts issues and application Hoyle RH editor. Thousand Oaks, CA: Sage.

Huang DW, Sherman BT, Lempicki RA (2009a). Systematic and integrative analysis of large

gene lists using DAVID bioinformatics resources. Nat Protoc 4(1):44-57.

Huang DW, Sherman BT, Lempicki RA (2009b). Bioinformatics enrichment tools: paths

toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37(1):1-

13.

171

Hughes T, Bockmann M, Mihailidis S, Bennett C, Harris A, Seow WK et al. (2013). Genetic,

epigenetic, and environmental influences on dentofacial structures and oral health: ongoing

studies of Australian twins and their families. Twin Res Hum Genet 16(1):43-51.

Humphrey SP, Williamson RT (2001). A review of saliva: normal composition, flow, and

function. J Prosthet Dent 85(2):162-169.

Hunter PB (1988). Risk factors in dental caries. Int Dent J 38(4):211-217.

Ismail AI, Lim S, Sohn W, Willem JM (2008a). Determinants of early childhood caries in low-

income African American young children. Pediatr Dent 30(4):289-296.

Ismail AI, Sohn W, Tellez M, Willem JM, Betz J, Lepkowski J (2008b). Risk indicators for

dental caries using the International Caries Detection and Assessment System (ICDAS).

Community Dent Oral Epidemiol 36(1):55-68.

Jackson SL, Vann Jr WF, Kotch JB, Pahel BT, Lee JY (2011). Impact of poor oral health on

children's school attendance and performance. Am J Public Health 101(10):1900-1906.

Jeon J (2015). The Strengths and Limitations of the Statistical Modeling of Complex Social

Phenomenon: Focusing on SEM, Path Analysis, or Multiple Regression Models. Int J Soc

Behav Educ Econ Bus Ind Eng 9(1559-1567.

Jepsen S, Blanco J, Buchalla W, Carvalho JC, Dietrich T, Dörfer C et al. (2017). Prevention

and control of dental caries and periodontal diseases at individual and population level:

consensus report of group 3 of joint EFP/ORCA workshop on the boundaries between caries

and periodontal diseases. J Clin Periodontol 44(S18):S85-S93.

Jha A, Tiwari G, Mohan D, Mukherjee S, Banerjee S (2017). Analysis of pedestrian movement

on Delhi roads by using naturalistic observation techniques. Transp Res Rec 2634:95-100.

Johnson NW (1991). Introduction: the nature of the caries process and the need for markers of

risk. In: Risk Markers for oral diseases NW Johnson editor. New York: Cambridge University

press, pp. 1-11.

Jonasson A, Eriksson C, Jenkinson HF, Kallestal C, Johansson I, Stromberg N (2007). Innate

immunity glycoprotein gp-340 variants may modulate human susceptibility to dental caries.

BMC Infect Dis 7:57: DOI:10.1186/1471-2334-7-57 (11 June 2007).

Ju X, Jamieson LM, Mejia GC (2016). Estimating the effects of maternal education on child

dental caries using marginal structural models: The Longitudinal Study of Indigenous

Australian Children. Community Dent Oral Epidemiol 44(6):602-610.

Ju X, Brennan D, Parker E, Mills H, Kapellas K, Jamieson L (2017). Efficacy of an oral health

literacy intervention among Indigenous Australian adults. Community Dent Oral Epidemiol

DOI: 10.1111/cdoe.12305 (19 May 2017).

Kaczor-Urbanowicz KE, Martin Carreras-Presas C, Aro K, Tu M, Garcia-Godoy F, Wong DT

(2017). Saliva diagnostics–Current views and directions. Exp Biol Med (Maywood)

242(5):459-472.

172

Kang S, Yoon I, Lee H, Cho J (2011). Association between AMELX polymorphisms and dental

caries in Koreans. Oral Dis 17(4):399-406.

Kassebaum N, Bernabé E, Dahiya M, Bhandari B, Murray C, Marcenes W (2015). Global

Burden of Untreated Caries A Systematic Review and Meta-regression. J Dent Res 94(5):650-

658.

Kavanaugh M, Halterman JS, Montes G, Epstein M, Hightower AD, Weitzman M (2006).

Maternal depressive symptoms are adversely associated with prevention practices and

parenting behaviors for preschool children. Ambulatory Pediatr 6(1):32-37.

Kawashita Y, Fukuda H, Kawasaki K, Kitamura M, Hayashida H, Furugen R et al. (2009).

Dental Caries in 3‐ Year‐ Old Children is Associated More with Child‐ Rearing Behaviors

than Mother‐ Related Health Behaviors. J Public Health Dent 69(2):104-110.

Kemper KE, Daetwyler HD, Visscher PM, Goddard ME (2012). Comparing linkage and

association analyses in sheep points to a better way of doing GWAS. Genet Res 94(04):191-

203.

Khocht A, Zohn H, Deasy M, Chang K-M (1995). Assessment of periodontal status with PSR

and traditional clinical periodontal examination. J Am Dent Assoc 126(12):1658-1665.

Kianoush N, Adler CJ, Nguyen K-AT, Browne GV, Simonian M, Hunter N (2014). Bacterial

profile of dentine caries and the impact of pH on bacterial population diversity. PLoS One 9(3)

DOI:10.1371/journal.pone.0092940 (27 March 2014).

Kidd E, Fejerskov O (2004). What constitutes dental caries? Histopathology of carious enamel

and dentin related to the action of cariogenic biofilms. J Dent Res 83(1):35-38.

Kieu TQ, Ngo LTQ, Pham TAAV (2017). Dental Caries and Related Factors in Vietnamese

Dental Patients. UIP Health Med 1(1):148-154.

Kim H-S, Smithies O, Maeda N (1990). A physical map of the human salivary proline-rich

protein gene cluster covers over 700 kbp of DNA. Genomics 6(2):260-267.

Kim H, Millsap R (2014). Using the Bollen-Stine Bootstrapping Method for Evaluating

Approximate Fit Indices. Multivariate Behav Res 49(6):581-596.

Kim Seow W (2012). Environmental, maternal, and child factors which contribute to early

childhood caries: a unifying conceptual model. Int J Paediatr Dent 22(3):157-168.

Kim Y, Telleen S (2004). Predictors of the utilization of oral health services by children of

low-income families in the United States: beliefs, cost, or provider? Taehan Kanho Hakhoe

Chi 34(8):1460-1467.

Kitasako Y, Burrow M, Stacey M, Huq L, Reynolds E, Tagami J (2008). Comparative analysis

of three commercial saliva testing kits with a standard saliva buffering test. Aust Dent J

53(2):140-144.Klein H, Palmer CE (1940). Dental caries in brothers and sisters of immune and

susceptible children. Milbank Mem Fund Q 8(1):67-82.

173

Klein H (1946). The family and dental disease; dental disease (DMF) experience in parents and

offspring. J Am Dent Assoc 33(11):735-743.

Köhler B, Andreen I (1994). Influence of caries-preventive measures in mothers on cariogenic

bacteria and caries experience in their children. Arch Oral Biol 39(10):907-911.

Köhler B, Andréen I (2012). Mutans streptococci and caries prevalence in children after early

maternal caries prevention: a follow-up at 19 years of age. Caries Res 46(5):474-480.

Krasone K, Lace B, Akota I, Care R, Deeley K, Kuchler EC et al. (2014). Genetic variation in

the promoter region of beta-defensin 1 (DEFB 1) is associated with high caries experience in

children born with cleft lip and palate. Acta Odontol Scand 72(3):235-240.

Küchler EC, Feng P, Deeley K, Fitzgerald CA, Meyer C, Gorbunov A et al. (2014). Fine

mapping of locus Xq25. 1-27-2 for a low caries experience phenotype. Arch Oral Biol

59(5):479-486.

Kulkarni GV, Chng T, Eny KM, Nielsen D, Wessman C, El-Sohemy A (2013). Association of

GLUT2 and TAS1R2 genotypes with risk for dental caries. Caries Res 47(3):219-225.

Kumar S, Kroon J, Lalloo R (2014). A systematic review of the impact of parental socio-

economic status and home environment characteristics on children’s oral health related quality

of life. Health Qual Life Outcomes 12:41. DOI:10.1186/1477-7525-12-41 (21 March 2014).

Kumar S, Tadakamadla J, Kroon J, Johnson NW (2016). Impact of parent-related factors on

dental caries in the permanent dentition of 6–12-year-old children: A systematic review. J Dent

46:1-11. DOI:10.1016/j.jdent.2015.12.007 (26 December 2015).

Kumar S, Kroon J, Lalloo R, Kulkarni S, Johnson NW (2017a). Relationship between body

mass index and dental caries in children, and the influence of socio‐ economic status. Int Dent

J 67(2):91-97.

Kumar S, Tadakamadla J, Zimmer‐ Gembeck M, Kroon J, Lalloo R, Johnson N (2017b).

Parenting practices and children's dental caries experience: A structural equation modelling

approach. Community Dent Oral Epidemiol DOI: 10.1111/cdoe.12321 (8 June 2017).

Kumar S, Zimmer-Gembeck M, Kroon J, Lalloo R, Johnson N (2017c). The role of parental

rearing practices and family demographics on oral health-related quality of life in children.

Qual Life Res 26(8): 2229–2236.

Kutsch VK, Kois JC, Chaiyabutr Y, Milicich G (2013). Reconsidering remineralization

strategies to include nanoparticle hydroxyapatite. Compend Contin Educ Dent 34(3):2-6.

Laaksonen M, Rahkonen O, Karvonen S, Lahelma E (2005). Socio-economic status and

smoking: analysing inequalities with multiple indicators. Eur J Public Health 15(3):262-269.

Lagerlöf F (1994). Caries-protective factors in saliva. Adv Dent Res 8(2):229-238.

Lai P, Kim Seow W, Tudehope D, Rogers Y (1997). Enamel hypoplasia and dental caries in

very-low birthweight children: a case-controlled, longitudinal study. Pediatr Dent 19:42-49.

174

Landry R, Jean M (2002). Periodontal Screening and Recording (PSR) Index: precursors,

utility and limitations in a clinical setting. Int Dent J 52(1):35-40.

Launay J-M, Del Pino M, Chironi G, Callebert J, Peoc’h K, Mégnien J-L et al. (2009). Smoking

induces long-lasting effects through a monoamine-oxidase epigenetic regulation. PLoS One

4(11): DOI:10.1371/journal.pone.0007959 (23 November 2009).

Lave JR, Keane C, Lin C, Ricci E (2002). The impact of dental benefits on the utilization of

dental services by low-income children in western Pennsylvania. Pediatr Dent 24(3):234-240.

Lee J, Giannobile W (2016). Taxes on sugar-sweetened beverages a strategy to reduce

epidemics of diabetes, obesity, and dental caries? J Dent Res 95(12): 1325–1326.

Lee JY, Bouwens TJ, Savage MF, Vann Jr WF (2006). Examining the cost-effectiveness of

early dental visits. Pediatri Dent 28(2):102-105.

Lee JY, Divaris K, DeWalt DA, Baker AD, Gizlice Z, Rozier RG et al. (2014). Caregivers’

health literacy and gaps in children’s Medicaid enrollment: findings from the Carolina Oral

Health Literacy Study. PLoS One 9(10): DOI:10.1371/journal.pone.0110178 (10 October

2014).

Leenen FA, Muller CP, Turner JD (2016). DNA methylation: conducting the orchestra from

exposure to phenotype? Clin Epigenetics 8:92 DOI 10.1186/s13148-016-0256-8 (6 September

2016).

Leme AP, Koo H, Bellato C, Bedi G, Cury J (2006). The role of sucrose in cariogenic dental

biofilm formation—new insight. J Dent Res 85(10):878-887.

Lenander-Lumikari M, Loimaranta V (2000). Saliva and dental caries. Adv Dent Res 14(1):40-

47.

Leone CW, Oppenheim FG (2001). Physical and chemical aspects of saliva as indicators of

risk for dental caries in humans. J Dent Educ 65(10):1054-1062.

Li Y, Caufield P, Dasanayake A, Wiener H, Vermund S (2005). Mode of delivery and other

maternal factors influence the acquisition of MSin infants. J Dent Res 84(9):806-811.

Li Y, Zhang Y, Yang R, Zhang Q, Zou J, Kang D (2011). Associations of social and

behavioural factors with early childhood caries in Xiamen city in China. Int J Paediatr Dent

21(2):103-111.

Lima AC, Lima MC, Guerra MQ, Romani SA, Eickmann SH, Lira PI (2006). Impact of weekly

treatment with ferrous sulfate on hemoglobin level, morbidity and nutritional status of anemic

infants. J Pediatr 82(6):452-7. DOI: 10.2223/JPED.1568.

Liu Y, Aryee MJ, Padyukov L, Fallin MD, Hesselberg E, Runarsson A et al. (2013).

Epigenome-wide association data implicate DNA methylation as an intermediary of genetic

risk in rheumatoid arthritis. Nat Biotechnol 31(2):142-147.

175

Llena C, Leyda A, Forner L, Garcet S (2015). Association between the number of early carious

lesions and diet in children with a high prevalence of caries. Eur J Paediatr Dent 16(1):7-12.

Loesche WJ (1986). Role of MSin human dental decay. Microbiol Rev 50(4):353-380.

López-Gómez SA, Villalobos-Rodelo JJ, Ávila-Burgos L, Casanova-Rosado JF, Vallejos-

Sánchez AA, Lucas-Rincón SE et al. (2016). Relationship between premature loss of primary

teeth with oral hygiene, consumption of soft drinks, dental care, and previous caries experience.

Sci Rep 6: 21147 DOI: 10.1038/srep21147 (26 February 2016).

Lu Y, Papagerakis P, Yamakoshi Y, Hu JC-C, Bartlett JD, Simmer JP (2008). Functions of

KLK4 and MMP-20 in dental enamel formation. Biol Chem 389(6):695-700.

Lucas N, Neumann A, Kilpatrick N, Nicholson J (2011). State‐ level differences in the oral

health of Australian preschool and early primary school‐ age children. Aust Dent J 56(1):56-

62.

Luo W, Wen X, Wang H-J, MacDougall M, Snead ML, Paine ML (2004). In vivo

overexpression of tuftelin in the enamel organic matrix. Cells Tissues Organs 177(4):212-220.

Lupo A, Cesaro E, Montano G, Zurlo D, Izzo P, Costanzo P (2013). KRAB-Zn finger proteins:

a repressor family displaying multiple biological functions. Curr Genomics 14(4):268-278.

Lynch RJ (2011). Zn in the mouth, its interactions with dental enamel and possible effects on

caries; a review of the literature. Int Dent J 61(3):46-54.

MacDougall M, DuPont BR, Simmons D, Reus B, Krebsbach P, Kärrman C et al. (1997).

Ameloblastin gene (AMBN) maps within the critical region for autosomal dominant

amelogenesis imperfecta at chromosome 4q21. Genomics 41(1):115-118.

Macek MD (2016). Although the Magnitude Varied, Income-Related Inequalities in Untreated

Dental Caries were Consistently Found among Young Children in 3 US Surveys. J Evid Based

Dent Pract 16(2):142-144.

Machin GA (1996). Some causes of genotypic and phenotypic discordance in monozygotic

twin pairs. Am J Med Genet A 61(3):216-228.

Mani S, Herceg Z. DNA methylation changes in cancer. In: Craig JM, Wong NC, eds.

Epigenetics: a reference manual. Norfolk, UK: Caister Academic Press, 2011:195–210.

Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ et al. (2009). Finding

the missing heritability of complex diseases. Nature 461(7265):747-753.

Marazita ML (2012). The evolution of human genetic studies of cleft lip and cleft palate. Annu

Rev Genomics Hum Genet 13:263-283.

Markovic N, Muratbegovic AA, Kobaslija S, Bajric E, Selimovic-Dragas M, Huseinbegovic A

(2013). Caries prevalence of children and adolescents in Bosnia and Herzegovina. Acta Med

Acad 42(2):108-116.

176

Martinhon CCR, Italiani FM, Padilha PM, Bijella MFTB, Delbem ACB, Buzalaf MAR (2006).

Effect of iron on bovine enamel and on the composition of the dental biofilm formed “in situ”.

Arch Oral Biol 51 (6): 471-475

Mattila M-L, Rautava P, Sillanpää M, Paunio P (2000). Caries in five-year-old children and

associations with family-related factors. J Dent Res 79(3):875-881.

Mejia G, Jamieson L, Ha D, Spencer A (2014). Greater inequalities in dental treatment than in

disease experience. J Dent Res 93(10):966-971.

Melo M, Weir M, Passos V, Powers M, Xu H (2016). pH-activatable nano-amorphous calcium

phosphate to reduce dental enamel demineralization. Dent Mater 32(1):e64

DOI:10.1016/j.dental.2016.08.132 (August 2016).

Mishra NK, Chang J, Zhao PX (2014). Prediction of membrane transport proteins and their

substrate specificities using primary sequence information. PLoS One 9(6):

10.1371/journal.pone.0100278 (26 June 2014).

Mohammed N, Mneimne M, Hill R, Al-Jawad M, Lynch R, Anderson P (2014). Physical

chemical effects of Zn on in vitro enamel demineralization. J Dent 42(9):1096-1104.

Moimaz SAS, Fadel CB, Lolli LF, Garbin CAS, Garbin AJÍ, Saliba NA (2014). Social aspects

of dental caries in the context of mother-child pairs. J Appl Oral Sci 22(1):73-78.

Mota-Veloso I, Soares MEC, Alencar BM, Marques LS, Ramos-Jorge ML, Ramos-Jorge J

(2016). Impact of untreated dental caries and its clinical consequences on the oral health-related

quality of life of schoolchildren aged 8–10 years. Qual Life Res 25(1):193-199.

Moyer VA (2014). Prevention of dental caries in children from birth through age 5 years: US

Preventive Services Task Force recommendation statement. Pediatrics 133(6):1102-1111.

Musen MA, Noy NF, Shah NH, Whetzel PL, Chute CG, Story M-A et al. (2011). The national

center for biomedical ontology. J Am Med Inform Assoc 19(2):190-195.

Musen MA, Noy NF, Shah NH, Whetzel PL, Chute CG, Story M-A et al. (2012). The national

center for biomedical ontology. J Am Med Inform Assoc 19(2):190-195.

Mwakayoka H, Masalu JR, Kikwilu EN (2017). Dental Caries and Associated Factors in

Children Aged 2-4 Years Old in Mbeya City, Tanzania. J Dent 18(2):104-111.

Nanda J, Sachdev V, Sandhu M, Deep-Singh-Nanda K (2015). Correlation between dental

caries experience and mutans streptococci counts using saliva and plaque as microbial risk

indicators in 3-8 year old children. A cross Sectional study. J Clin Exp Dent 7(1): e114 –e118.

Nelson S, Albert J, Geng C, Curtan S, Lang K, Miadich S et al. (2013). Increased enamel

hypoplasia and very low birthweight infants. J Dent Res 92(9):788-794.

177

Nevitt J, Hancock GR (1998). Relative Performance of Rescaling and Resampling Approaches

to Model Chi Square and Parameter Standard Error Estimation in Structural Equation Modeling

http://files.eric.ed.gov/fulltext/ED420711.pdf (Accessed on 28 February 2017).

Ngoc VTN, Chu D-T, Le D-H (2017). Prevalence of early childhood caries and its related risk

factors in preschoolers: Result from a cross sectional study in Vietnam. Pediatr Dent J

27(2):79-84.

Nibali L, Tonetti MS, Ready D, Parkar M, Brett PM, Donos N et al. (2008). Interleukin-6

polymorphisms are associated with pathogenic bacteria in subjects with periodontitis. J

Periodontol 79(4):677-683.

Nicolau B, Marcenes W, Allison P, Sheiham A (2005). The life course approach: explaining

the association between height and dental caries in Brazilian adolescents. Community Dent

Oral Epidemiol 33(2):93-98.

Niji R, Arita K, Abe Y, Lucas ME, Nishino M, Mitome M (2010). Maternal age at birth and

other risk factors in early childhood caries. Pediatr Dent 32(7):493-498.

Nourijelyani K, Yekaninejad MS, Eshraghian MR, Mohammad K, Foroushani AR, Pakpour A

(2014). The influence of mothers’ lifestyle and health behavior on their children: an exploration

for oral health. Iran Red Crescent Med J 16(2): DOI: 10.5812/ircmj.16051 (5 February 2014).

Ogata H, Goto S, Sato K, Fujibuchi W, Bono H, Kanehisa M (1999). KEGG: Kyoto

encyclopedia of genes and genomes. Nucleic Acids Res 27(1):29-34.

Okano M, Bell DW, Haber DA, Li E (1999). DNA methyltransferases Dnmt3a and Dnmt3b

are essential for de novo methylation and mammalian development. Cell 99(3):247-257.

Oliveira LB, Sheiham A, Bönecker M (2008). Exploring the association of dental caries with

social factors and nutritional status in Brazilian preschool children. Eur J Oral Sci 116(1):37-

43.

Oliveria SA, Ellison RC, Moore LL, Gillman MW, Garrahie EJ, Singer MR (1992). Parent-

child relationships in nutrient intake: the Framingham Children's Study. Am J Clin Nutr

56(3):593-598.

Olszowski T, Adler G, Janiszewska-Olszowska J, Safranow K, Kaczmarczyk M (2012).

MBL2, MASP2, AMELX, and ENAM gene polymorphisms and dental caries in Polish

children. Oral Dis 18(4):389-395.

Önder Ö, Sidoli S, Carroll M, Garcia BA (2015). Progress in epigenetic histone modification

analysis by mass spectrometry for clinical investigations. Expert Rev Proteomics 12(5):499-

517.

Opal S, Garg S, Jain J, Walia I (2015). Genetic factors affecting dental caries risk. Aust Dent J

60(1):2-11.

178

Östberg AL, Skeie MS, Skaare AB, Espelid I (2016). Caries increment in young children in

Skaraborg, Sweden: associations with parental sociodemography, health habits, and attitudes.

Int J Paediatr Dent 27(1):47-55.

Özen B, Van Strijp A, Özer L, Olmus H, Genc A, Cehreli SB (2016). Evaluation of possible

associated factors for early childhood caries and severe early childhood caries: a multicenter

cross-sectional survey. J Clin Pediatr Dent 40(2):118-123.

Ozturk A, Famili P, Vieira A (2010). The antimicrobial peptide DEFB1 is associated with

caries. J Dent Res 89(6):631-636.

Pak D, Li C, Todem D, Sohn W (2017). A multistate model for correlated interval‐ censored

life history data in caries research. J R Stat Soc Ser C Appl Stat 66(2):413-423.

Palmer C, Kent Jr R, Loo C, Hughes C, Stutius E, Pradhan N et al. (2010). Diet and caries-

associated bacteria in severe early childhood caries. J Dent Res 89(11):1224-1229.

Parisotto TM, Santos MN, Rodrigues LK, Costa LS (2012). Behavior and progression of early

carious lesions in early childhood: a 1-year follow-up study. J Dent Child (Chic) 79(3):130-

135.

Patir A, Seymen F, Yildirim M, Deeley K, Cooper ME, Marazita ML et al. (2008). Enamel

formation genes are associated with high caries experience in Turkish children. Caries Res

42(5):394-400.

Patterson K, Molloy L, Qu W, Clark S (2011). DNA methylation: bisulphite modification and

analysis. J Vis Exp (56): DOI: 10.3791/3170 (21 October 2011).

Pecharki GD, Brancher JA, Olandoski M, Doetzer AD, Moyses SJ, Trevilatto PC (2016).

Multilevel modeling for dental caries among adolescents in a Brazilian large city. Dent 3000

4(1):43-54.

Peres MA, Latorre MdRDd, Sheiham A, Peres KG, Barros FC, Hernandez PG et al. (2005).

Social and biological early life influences on severity of dental caries in children aged 6 years.

Community Dent Oral Epidemiol 33(1):53-63.

Peres RCR, Camargo G, Mofatto LS, Cortellazzi KL, Santos MCLG, Nobre-dos-Santos M et

al. (2010). Association of polymorphisms in the carbonic anhydrase 6 gene with salivary buffer

capacity, dental plaque pH, and caries index in children aged 7-9 years. Pharmacogenomics J

10(2):114-119.

Perez P, Jang SI, Alevizos I (2014). Emerging landscape of non‐ coding RNAs in oral health

and disease. Oral Dis 20(3):226-235.

Petersen PE (2003). The World Oral Health Report 2003: continuous improvement of oral

health in the 21st century–the approach of the WHO Global Oral Health Programme.

Community Dent Oral Epidemiol 31(s1):3-24.

Petersen PE (2005). Sociobehavioural risk factors in dental caries–international perspectives.

Community Dent Oral Epidemiol 33(4):274-279.

179

Petersen PE, Bourgeois D, Ogawa H, Estupinan-Day S, Ndiaye C (2005). The global burden

of oral diseases and risks to oral health. Bull World Health Organ 83(9):661-669.

Petersen PE (2008). World Health Organization global policy for improvement of oral health‐World Health Assembly 2007. Int Dent J 58(3):115-121.

Petersson GH, Åkerman S, Isberg P-E, Ericson D (2016). Comparison of risk assessment based

on clinical judgement and Cariogram in addition to patient perceived treatment need. BMC

Oral Health 17:13. DOI 10.1186/s12903-016-0238-4 (7 July 2016).

Petronis A, Gottesman II, Kan P, Kennedy JL, Basile VS, Paterson AD et al. (2003).

Monozygotic twins exhibit numerous epigenetic differences: clues to twin discordance?

Schizophr Bull 29(1):169-178.

Pidamale R, Sowmya B, Thomas A, Jose T (2012). Genetic sensitivity to bitter taste of 6-n

Propylthiouracil: A useful diagnostic aid to detect early childhood caries in pre-school children.

Indian J Hum Genet 18(1):101-105.

Pihlstrom BL, Michalowicz BS, Johnson NW (2005). Periodontal diseases. The Lancet

366(9499):1809-1820.

Piva F, Pereira JT, Luz PB, Hashizume LN, Hugo FN, Araujo FBd (2017). A Longitudinal

Study of Early Childhood Caries and Associated Factors in Brazilian Children. Braz Dent J

28(2):241-248.

Plaka K, Ravindra K, Mor S, Gauba K (2017). Risk factors and prevalence of dental fluorosis

and dental caries in school children of North India. Environ Monit Assess 189:40. DOI

10.1007/s10661-016-5684-6 (26 December 2016).

Plonka K, Pukallus M, Holcombe T, Barnett A, Walsh L, Seow W (2013a). Randomized

controlled trial: a randomized controlled clinical trial comparing a remineralizing paste with

an antibacterial gel to prevent early childhood caries. Peadiatr Dent 35(1):e8-e12.

Plonka KA, Pukallus ML, Barnett A, Holcombe TF, Walsh LJ, Seow W (2013b). A controlled,

longitudinal study of home visits compared to telephone contacts to prevent early childhood

caries. Int J Paediatr Dent 23(1):23-31.

Polk DE, Wang X, Feingold E, Shaffer JR, Weeks DE, Weyant RJ et al. (2012). Effects of

smoking and genotype on the PSR index of periodontal disease in adults aged 18–49. Int J

Environ Res Public Health 9(8):2839-2850.

Poutanen R, Lahti S, Seppä L, Tolvanen M, Hausen H (2007). Oral health-related knowledge,

attitudes, behavior, and family characteristics among Finnish schoolchildren with and without

active initial caries lesions. Acta Odontol Scand 65(2):87-96.

Preethi B, Reshma D, Anand P (2010). Evaluation of flow rate, pH, buffering capacity,

calcium, total proteins and total antioxidant capacity levels of saliva in caries free and caries

active children: an in vivo study. Indian J Clin Biochem 25(4):425-428.

180

Proença MAM, Franco MM, Rodrigues VP, Costa JF, Costa EL (2015). Association between

early childhood caries and maternal caries status: A cross-section study in São Luís, Maranhão,

Brazil. Eur J Dent 9(1):122-126.

Ramamurthy P, Swamy H, Bennete F, Rohini M, Nagarathnamma T (2014a). Relationship

between severe-early childhood caries, salivary mutans streptococci, and lactobacilli in

preschool children of low socio-economic status in Bengaluru city. J Indian Soc Pedod Prev

Dent 32(1):44-47.

Rauth RJ, Potter KS, Ngan AY-W, Saad DM, Mehr R, Luong VQ et al. (2009). Dental enamel:

genes define biomechanics. J Calif Dent Assoc 37(12):863-868.

Razin A, Shemer R (1995). DNA methylation in early development. Hum Molec Genet

4(1):1751-1755.

Reed SG, Voronca D, Wingate JS, Murali M, Lawson AB, Hulsey TC et al. (2017). Prenatal

vitamin D and enamel hypoplasia in human primary maxillary central incisors: A pilot study.

Pediatr Dent J 27(1):21-28.

Reynolds JC, Damiano PC, Glanville JL, Oleson J, McQuistan MR (2015). Neighborhood and

family social capital and parent‐ reported oral health of children in Iowa. Community Dent

Oral Epidemiol 43(6):569-577.

Riggs E, Slack-Smith L, Yelland J, Chadwick B, Robertson L, Kilpatrick N (2016).

Interventions with pregnant women andnew mother s for preventing caries in children.

Cochrane Database Syst Rev (4): CD012155. DOI:10.1002/14651858.CD012155 (21 April

2016).

Riley B, Schultz R, Cooper M, Goldstein‐ McHenry T, Daack‐ Hirsch S, Lee K et al. (2007).

A genome‐ wide linkage scan for cleft lip and cleft palate identifies a novel locus on 8p11‐23. Am J Med Genet A 143(8):846-852.

Ronaghi M (2001). Pyrosequencing sheds light on DNA sequencing. Genome Res 11(1):3-11.

Roseboom TJ, van der Meulen JH, van Montfrans GA, Ravelli AC, Osmond C, Barker DJ et

al. (2001). Maternal nutrition during gestation and blood pressure in later life. J Hypertens

19(1):29-34.

Rossow I (1992). Intrafamily influences on health behavior. J Clin Periodontol 19(10):774-

778.

Russell JI, MacFarlane TW, Aitchison TC, Stephen KW, Burchell CK (1991). Prediction of

caries increment in Scottish adolescents. Community Dent Oral Epidemiol 19(2):74-77.

Saied-Moallemi Z, Virtanen J, Ghofranipour F, Murtomaa H (2008). Influence of mothers’ oral

health knowledge and attitudes on their children’s dental health. Eur Arch Paediatr Dent

9(2):79-83.

Samaranayake L (2012). Essential microbiology for dentistry Edinburgh; Fourth ed. New

York: Churchill Livingstone Elsevier Health Sciences.

181

Sanders AE, Slade GD, Lim S, Reisine ST (2009). Impact of oral disease on quality of life in

the US and Australian populations. Community Dent Oral Epidemiol 37(2):171-181.

Sandoval J, Asteller M (2012). Cancer epigenomics: beyond genomics. Curr Opin Genet Dev

22(1):50-55.

Santos M, De Souza A, Gerlach R, Trevilatto P, Scarel‐ Caminaga R, Line S (2004). Inhibition

of human pulpal gelatinases (MMP‐ 2 and MMP‐ 9) by Zn oxide cements. J Oral Rehabil

31(7):660-664.

Sasaki T, Takagi M, Yanagisawat T (2008). Structure and function of secretory. In: Dental

enamel 785:32-50.

Schernhammer ES, Giovannucci E, Kawasaki T, Rosner B, Fuchs CS, Ogino S (2010). Dietary

folate, alcohol and B vitamins in relation to LINE-1 hypomethylation in colon cancer. Gut

59(6):794-799.

Schmidt RJ, Hansen RL, Hartiala J, Allayee H, Schmidt LC, Tancredi DJ et al. (2011). Prenatal

vitamins, one-carbon metabolism gene variants, and risk for autism. Epidemiology 22(4):476.

Scholl TO (2005). Iron status during pregnancy: setting the stage for mother and infant. Am J

Clin Nutr 81(5):1218S-1222S.

Schroth RJ, Lavelle C, Tate R, Bruce S, Billings RJ, Moffatt ME (2014). Prenatal vitamin D

and dental caries in infants. Pediatrics 133(5):e1277-e1284.

Schübeler D (2015). Function and information content of DNA methylation. Nature

517(7534):321-326.

Schultz J, Milpetz F, Bork P, Ponting CP (1998). SMART, a simple modular architecture

research tool: identification of signaling domains. Proc Natl Acad Sci 95(11):5857-5864.

Schultz ST, Shenkin J, Horowitz A (2001). Parental perceptions of unmet dental need and cost

barriers to care for developmentally disabled children. Pediatr Dent 23(4):321-325.

Schwendicke F, Dörfer C, Schlattmann P, Page LF, Thomson W, Paris S (2015). Socio-

economic inequality and caries a systematic review and meta-analysis. J Dent Res 94(1):10-

18.

Seddon JM, Reynolds R, Shah HR, Rosner B (2011). Smoking, dietary betaine, methionine,

and vitamin D in monozygotic twins with discordant macular degeneration: epigenetic

implications. Ophthalmology 118(7):1386-1394.

Sedgwick P (2014). Retrospective cohort studies: advantages and disadvantages. BMJ (Online)

348: DOI: 10.1136/bmj.g1072 (24 January 2014)

Selwitz RH, Ismail AI, Pitts NB (2007). Dental caries. The Lancet 369(9555):51-59.

182

Seo JY, Park YJ, Yi YA, Hwang JY, Lee IB, Cho BH et al. (2015). Epigenetics: general

characteristics and implications for oral health. Restor Dent Endod 40(1):14-22.

Seow W, Clifford H, Battistutta D, Morawska A, Holcombe T (2009). Case-control study of

early childhood caries in Australia. Caries Res 43(1):25-35.

Shaffer J, Wang X, Feingold E, Lee M, Begum F, Weeks D et al. (2011). Genome-wide

association scan for childhood caries implicates novel genes. J Dent Res 90(12):1457-1462.

Shaffer JR, Feingold E, Wang X, Lee M, Tcuenco K, Weeks D et al. (2013). GWAS of dental

caries patterns in the permanent dentition. J Dent Res 92(1):38-44.

Shaffer JR, Feingold E, Wang X, Tcuenco KT, Weeks DE, DeSensi RS et al. (2012a). Heritable

patterns of tooth decay in the permanent dentition: principal components and factor analyses.

BMC Oral Health 12:7. DOI:10.1186/1472-6831-12-7 (9 March 2012).

Shaffer JR, Wang X, Desensi RS, Wendell S, Weyant RJ, Cuenco KT et al. (2012b). Genetic

susceptibility to dental caries on pit and fissure and smooth surfaces. Caries Res 46(1):38-46.

Shaffer JR, Wang X, McNeil DW, Weyant RJ, Crout R, Marazita ML (2015). Genetic

susceptibility to dental caries differs between the sexes: a family-based study. Caries Res

49(2):133-140.

Shearer D, Thomson W, Broadbent J, Poulton R (2011). Maternal oral health predicts their

children’s caries experience in adulthood. J Dent Res 90(5):672-677.

Sheiham A, Watt RG (2000). The common risk factor approach: a rational basis for promoting

oral health. Community Dent Oral Epidemiol 28(6):399-406.

Shimizu T, Ho B, Deeley K, Briseno-Ruiz J, Faraco IM, Jr., Schupack BI et al. (2012). Enamel

formation genes influence enamel microhardness before and after cariogenic challenge. PLoS

One 7(9): DOI:10.1371/journal.pone.0045022 (24 September 2012).

Silva AER, Menezes AMB, Demarco FF, Vargas-Ferreira F, Peres MA (2013). Obesity and

dental caries: systematic review. Rev Saude Publica 47(4):799-812.

Simark‐ Mattsson C, Emilson CG, Håkansson EG, Jacobsson C, Roos K, Holm S (2007).

Lactobacillus‐ mediated interference of mutans streptococci in caries‐ free vs. caries‐ active

subjects. Eur J Oral Sci 115(4):308-314.

Slade G, Sanders A, Do L, Roberts-Thomson K, Spencer A (2013). Effects of fluoridated

drinking water on dental caries in Australian adults. J Dent Res 92(4): 376-382.

Slade G D, Spencer A J (1988). Population estimates, standard errors and hypothesis tests from

the 1987–88 National Oral Health Survey of Australia. AIHW Dental Statistics and Research

Series No. 14. Adelaide: The University of Adelaide.

Slayton RL, Cooper ME, Marazita ML (2005). Tuftelin, mutans streptococci, and dental caries

susceptibility. J Dent Res 84(8):711-714.

183

Smith R, Badner VM, Morse DE, Freeman K (2002). Maternal risk indicators for childhood

caries in an inner city population. Community Dent Oral Epidemiol 30(3):176-181.

Splieth CH, Christiansen J, Foster Page L (2016). Caries Epidemiology and Community

Dentistry: Chances for Future Improvements in Caries Risk Groups. Outcomes of the ORCA

Saturday Afternoon Symposium, Greifswald, 2014. Part 1. Caries Res 50(1):9-16.

Su LJ, Mahabir S, Ellison GL, McGuinn LA, Reid BC (2011). Epigenetic contributions to the

relationship between cancer and dietary intake of nutrients, bioactive food components, and

environmental toxicants. Front Genet 2:91. DOI: 10.3389/fgene.2011.00091 (9 January 2011).

Sun YV (2014). The influences of genetic and environmental factors on methylome-wide

association studies for human diseases. Curr Genet Med Rep 2(4):261-270.

Szatko F, Wierzbicka M, Dybizbanska E, Struzycka I, Iwanicka-Frankowska E (2004). Oral

health of Polish three-year-olds and mothers' oral health-related knowledge. Community Dent

Health 21(2):175-180.

Tabachnick BG, Fidell LS (2007). Using multivariate statistics (5th ed.) Boston: USA: Allyn

& Bacon, Inc.

Tahmassebi J, Duggal M, Malik-Kotru G, Curzon M (2006). Soft drinks and dental health: a

review of the current literature. J Dent 34(1):2-11.

Takahashi R, Ota E, Hoshi K, Naito T, Toyoshima Y, Yuasa H et al. (2015). Fluoride

supplementation in pregnant women for preventing dental caries in the primary teeth of their

children. Cochrane Libr : DOI: 10.1002/14651858.CD011850 (28 August 2015).

Takatsuka T, Hirano J, Matsumoto H, Honma T (2005). X‐ Ray absorption fine structure

analysis of the local environment of Zn in dentine treated with Zn compounds. Eur J Oral Sci

113(2):180-183.

Tanaka K, Miyake Y, Sasaki S, Hirota Y (2013). Socio-economic status and risk of dental

caries in Japanese preschool children: the Osaka Maternal and child health study. J Public

Health Dent 73(3):217-223.

Tanaka K, Hitsumoto S, Miyake Y, Okubo H, Sasaki S, Miyatake N et al. (2015). Higher

vitamin D intake during pregnancy is associated with reduced risk of dental caries in young

Japanese children. Ann Epidemiol 25(8):620-625.

Tang R-S, Huang M-C, Huang S-T (2013). Relationship between dental caries status and

anemia in children with severe early childhood caries. Kaohsiung J Med Sci 29(6):330-336.

Tang WW, Dietmann S, Irie N, Leitch HG, Floros VI, Bradshaw CR et al. (2015). A unique

gene regulatory network resets the human germline epigenome for development. Cell

161(6):1453-1467.

Tannure PN, Kuchler EC, Falagan-Lotsch P, Amorim LMF, Raggio Luiz R, Costa MC et al.

(2012a). MMP13 polymorphism decreases risk for dental caries. Caries Res 46(4):401-407.

184

Tannure PN, Küchler EC, Lips A, Costa MdC, Luiz RR, Granjeiro JM et al. (2012b). Genetic

variation in MMP20 contributes to higher caries experience. J Dent 40(5):381-386.

Tanzer JM, Livingston J, Thompson AM (2001). The microbiology of primary dental caries in

humans. J Dent Educ 65(10):1028-1037.

Team RDC (2013) R: A language and environment for statistical computing. Vienna, Austria:

R Foundation for Statistical Computing. Available: http://www.R-project.org/ ISBN 3-

900051-07-0, Available: http://www.R-project.org/ .

Thitasomakul S, Piwat S, Thearmontree A, Chankanka O, Pithpornchaiyakul W, Madyusoh S

(2009). Risks for early childhood caries analyzed by negative binomial models. J Dent Res

88(2):137-141.

Thwin KM, Zaitsu T, Ueno M, Kawaguchi Y (2016). Early Childhood Caries and Related Risk

Factors among Myanmar Preschool Children. Int J Clin Prev Dent 12(4):229-236.

Tiwari T, Rai N, Colmenero E, Gonzalez H, Castro M (2017). A Community-Based

Participatory Research Approach to Understand Urban Latino Parent’s Oral Health Knowledge

and Beliefs. Int J Dent 2017: DOI:10.1155/2017/9418305 (29 March 2017).

Toledano M, Yamauti M, Osorio E, Osorio R (2012). Zn-inhibited MMP-mediated collagen

degradation after different dentine demineralization procedures. Caries Res 46(3):201-207.

Traebert J, Jinbo Y, de Lacerda JT (2011). Association between maternal schooling and caries

prevalence: a cross-sectional study in southern Brazil. Oral Health Prev Dent 9(1):47-52

Tsai P-C, Bell JT (2015). Power and sample size estimation for epigenome-wide association

scans to detect differential DNA methylation. Int J Epidemiol 44(4):1429-1441.

Tulunoglu Ö, Demirtas S, Tulunoglu I (2006). Total antioxidant levels of saliva in children

related to caries, age, and gender. Int J Paediatr Dent 16(3):186-191.

Turton B, Durward C, Manton D, Bach K, Yos C (2016). Socio-behavioural risk factors for

early childhood caries (ECC) in Cambodian preschool children: a pilot study. Eur Arch

Paediatr Dent 17(2):97-105.

Turton BJ, Durward CS, Manton DJ (2015). Early childhood caries and maternal caries

experience in a convenience sample of Cambodian pre-schoolers. Pediatr Dent J 25(1):14-18.

Valarini N, Maciel S, Moura S, Poli-Frederico R (2012). Association of dental caries with HLA

class II allele in Brazilian adolescents. Caries Res 46(6):530-535.

Van De Mheen H, Stronks K, Van Den Bos J, Mackenbach JP (1997). The contribution of

childhood environment to the explanation of socio-economic inequalities in health in adult life:

a retrospective study. Soc Sci Med 44(1):13-24.

van der Tas JT, Kragt L, Veerkamp JJ, Jaddoe VW, Moll HA, Ongkosuwito EM et al. (2016).

Ethnic disparities in dental caries among six-year-old children in the Netherlands. Caries Res

50(5):489-497.

185

Van Houte J (1993). Microbiological predictors of caries risk. Adv Dent Res 7(2):87-96.

VanVoorhis CRW, Morgan BL (2007). Understanding power and rules of thumb for

determining sample sizes. Tutor Quant Methods Psychol 3(2):43-50.

Vargas CM, Crall JJ, Schneider DA (1998). Sociodemographic distribution of pediatric dental

caries: NHANES III, 1988-1994. J Am Dent Assoc 129(9):1229-1238.

Varma S, Banerjee A, Bartlett D (2008). An in vivo investigation of associations between saliva

properties, caries prevalence and potential lesion activity in an adult UK population. J Dent

36(4):294-299.

Vieira A, Marazita M, Goldstein-McHenry T (2008). Genome-wide scan finds suggestive

caries loci. J Dent Res 87(5):435-439.

Vieira AR, Modesto A, Marazita ML (2014). Caries: review of human genetics research.

Caries Res 48(5):491-506.

Wade WG (2013). The oral microbiome in health and disease. Pharmacol Res 69(1):137-143.

Wallengren ML, Johnson U, Ericson D (1997). HLA-DR4 and number of mutans streptococci

in saliva among dental students and staff. Acta Odontol 55(5):296-298.

Wang Q, Jia P, Cuenco KT, Feingold E, Marazita ML, Wang L et al. (2013a). Multi-

Dimensional Prioritization of Dental Caries Candidate Genes and Its Enriched Dense Network

Modules. PLoS One 8(10): DOI:10.1371/journal.pone.0076666e76666 (11 October 2013).

Wang Q, Jia P, Cuenco KT, Zeng Z, Feingold E, Marazita ML et al. (2013b). Association

signals unveiled by a comprehensive gene set enrichment analysis of dental caries genome-

wide association studies. PLoS One 8(8): DOI:10.1371/journal.pone.007265372653 (14

August 2014).

Wang X, Shaffer J, Weyant R, Cuenco K, DeSensi R, Crout R et al. (2010). Genes and their

effects on dental caries may differ between primary and permanent dentitions. Caries Res

44(3):277-284.

Wang X, Shaffer JR, Zeng Z, Begum F, Vieira AR, Noel J et al. (2012a). Genome-wide

association scan of dental caries in the permanent dentition. BMC Oral Health 12:57.

DOI:10.1186/1472-6831-12-57 (21 December 2012).

Wang X, Willing MC, Marazita ML, Wendell S, Warren JJ, Broffitt B et al. (2012b). Genetic

and environmental factors associated with dental caries in children: the Iowa Fluoride Study.

Caries Res 46(3):177-184.

Warren JJ, Weber‐ Gasparoni K, Marshall TA, Drake DR, Dehkordi‐ Vakil F, Dawson DV et

al. (2009). A longitudinal study of dental caries risk among very young low SES children.

Community Dent Oral Epidemiol 37(2):116-122.

186

Warren JJ, Blanchette D, Dawson DV, Marshall TA, Phipps KR, Starr D et al. (2016). Factors

associated with dental caries in a group of American Indian children at age 36 months.

Community Dent Oral Epidemiol 44(2):154-161.

Warren JJ, Van Buren JM, Levy SM, Marshall TA, Cavanaugh JE, Curtis AM et al. (2017).

Dental caries clusters among adolescents. Community Dent Oral Epidemiol: DOI:

10.1111/cdoe.12317 (3 July 2017).

Watt RG, Sheiham A (2012). Integrating the common risk factor approach into a social

determinants framework. Community Dent Oral Epidemiol 40(4):289-296.

Weintraub J, Prakash P, Shain S, Laccabue M, Gansky S (2010). Mothers’ caries increases

odds of children’s caries. J Dent Res 89(9):954-958.

Wendell S, Wang X, Brown M, Cooper ME, DeSensi RS, Weyant RJ et al. (2010). Taste genes

associated with dental caries. J Dent Res 89(11):1198-1202.

Wendt L-K, Hallonsten A-L, Koch G, Birkhed D (1996). Analysis of caries-related factors in

infants and toddlers living in Sweden. Acta Odontol 54(2):131-137.

Wendt L, Svedin C, Hallonsten A, Larsson I (1995). Mental health, family interaction, and life

events in infants and toddlers with caries. Swed Dent J 19(1-2):17-27.

Whetzel PL, Noy NF, Shah NH, Alexander PR, Nyulas C, Tudorache T et al. (2011). BioPortal:

enhanced functionality via new Web services from the National Center for Biomedical

Ontology to access and use ontologies in software applications. Nucleic Acids Res 39(2):W541-

W545.

Widita E, Pamardiningsih Y, Vega CA (2017). Caries Risk Profiles amongst Preschool Aged

Children Living in the Sleman District of Yogyakarta, Indonesia. J Dent Indon 24(1):1-6.

Wigen TI, Wang NJ (2011). Maternal health and lifestyle, and caries experience in preschool

children. A longitudinal study from pregnancy to age 5 years. Eur J Oral Sci 119(6):463-468.

Wigen TI, Wang NJ (2014). Health behaviors and family characteristics in early childhood

influence caries development. A longitudinal study based on data from MoBa. Nor Epidemiol

24(1-2):91-95.

Williams DM (2011). Global oral health inequalities: The research agenda. J Dent Res

90(5):549-551.

Williams S, Hughes T, Adler C, Brook A, Townsend G (2014). Epigenetics: a new frontier in

dentistry. Aust Dent J 59(1):23-33.

Wilson AG (2008). Epigenetic regulation of gene expression in the inflammatory response and

relevance to common diseases. J Periodontol 79(8):1514-1519.

Winter G (1976). Maternal nutritional requirements in relation to the subsequent development

of teeth in children. J Hum Nutr 30(2):93-99.

187

Wong N, Tran C, Pukallus M, Holcombe T, Seow W (2012). A three‐ year retrospective study

of emergency visits at an oral health clinic in south‐ east Queensland. Aust Dent J 57(2):132-

137.

Wu H, Lippmann J, Oza J, Zeng M, Fives‐ Taylor P, Reich N (2006). Inactivation of DNA

adenine methyltransferase alters virulence factors in Actinobacillus actinomycetemcomitans.

Oral Microbiol Immunol 21(4):238-244.

Yang AS, Estécio MR, Doshi K, Kondo Y, Tajara EH, Issa JPJ (2004). A simple method for

estimating global DNA methylation using bisulfite PCR of repetitive DNA elements. Nucleic

Acids Res 32(3): DOI: 10.1093/nar/gnh032 (21 January 2004).

Yoshioka H, Minamizaki T, Yoshiko Y (2015). The dynamics of DNA methylation and

hydroxymethylation during amelogenesis. Histochem Cell Biol 144(5):471-478.

Yun KH, Lee HS, Nam OH, Moon CY, Lee JH, Choi SC (2017). Analysis of bacterial

community profiles of endodontically infected primary teeth using pyrosequencing. Int J

Paediatr Dent 27(1):56-65.

Zaina S, Lindholm MW, Lund G (2005). Nutrition and aberrant DNA methylation patterns in

atherosclerosis: more than just hyperhomocysteinemia? J Nutr 135(1):5-8.

Zakhary G, Clark R, Bidichandani S, Owen W, Slayton R, Levine M (2007). Acidic proline-

rich protein Db and caries in young children. J Dent Res 86(12):1176-1180.

Zander A, Sivaneswaran S, Skinner J, Byun R, Jalaludin B (2013). Risk factors for dental caries

in small rural and regional Australian communities. Rural Remote Health 13(3):2492.

Zaykin DV (2011). Optimally weighted Z‐ test is a powerful method for combining

probabilities in meta‐ analysis. J Evol Biol 24(8):1836-1841.

Zeng Z, Shaffer JR, Wang X, Feingold E, Weeks DE, Lee M et al. (2013). Genome-wide

association studies of pit-and-fissure- and smooth-surface caries in permanent dentition. J Dent

Res 92(5):432-437.

Zeng Z, Feingold E, Wang X, Weeks DE, Lee M, Cuenco KT et al. (2014). Genome-wide

association study of primary dentition pit-and-fissure and smooth surface caries. Caries Res

48(4):330-338.

Zhang D, Li Q, Rao L, Yi B, Xu Q (2015). Effect of 5-Aza-2′-deoxycytidine on Odontogenic

Differentiation of Human Dental Pulp Cells. J Endod 41(5):640-645.

Zhang S, Barros S, Niculescu M, Moretti A, Preisser J, Offenbacher S (2010). Alteration of

PTGS2 promoter methylation in chronic periodontitis. J Dent Res 89(2):133-137.

Zhang Y, Todem D, Kim K, Lesaffre E (2011). Bayesian latent variable models for spatially

correlated tooth-level binary data in caries research. Stat Modelling 11(1):25-47.

188

Appendix A : Oral Health Questionnaire

STUD Study ID

0

MATERNAL, ENVIRONMENTAL AND EARLY CHILDHOOD INFLUENCES ON DENTAL CARIES

IN CHILDREN

Questionnaire assessing the oral health Knowledge, Attitude and Practices

of mothers giving birth to infants within the EFHL study

Please answer all of the following questions on a scale of 1 to 5 (1=Strongly Agree; 2=Agree;

3=Neutral/Not Sure; 4=Disagree; 5=Strongly Disagree), and place a circle around the

corresponding number.

Strongly

Agree Agree

Neutral /

Not Sure Disagree

Strongly

Disagree

I feel that it is important to acquire

information on oral health care for my child. 1 2 3 4 5

I understand what the term ‘dental decay’

means. 1 2 3 4 5

I believe my oral health could affect my

child’s oral health. 1 2 3 4 5

I believe that baby teeth are important. 1 2 3 4 5

I believe that problems with baby teeth will

affect adult teeth. 1 2 3 4 5

I believe that rotten baby teeth could affect a

child’s health. 1 2 3 4 5

189

It is important to clean an infant’s gums with

a damp washcloth after meals, prior to first

teeth erupting

1 2 3 4 5

Strongly

Agree Agree

Neutral /

Not Sure Disagree

Strongly

Disagree

Cleaning of an infant’s teeth should start from

the day the first tooth appears in the mouth 1 2 3 4 5

I believe that breastfeeding is important for a

newborn child’s teeth. 1 2 3 4 5

I believe that it is OK for a child to go to bed

with a bottle of milk. 1 2 3 4 5

I believe it is OK to clean my child’s dummy

in my own mouth. 1 2 3 4 5

I believe there are no problems with sharing a

feeding spoon with my child. 1 2 3 4 5

It is a good idea to give a baby a bottle to

comfort them while teething. 1 2 3 4 5

Babies who do not have bottles will cry more. 1 2 3 4 5

Frequently giving my child soft drinks is OK

for their teeth. 1 2 3 4 5

Frequently giving my child fruit juice is OK

for their teeth. 1 2 3 4 5

Frequently feeding my child milk or formula

is OK for their teeth. 1 2 3 4 5

190

Using fluoride toothpaste helps to prevent

tooth decay.

1 2 3 4 5

Strongly

Agree Agree

Neutral /

Not Sure Disagree

Strongly

Disagree

I believe it is safe for infants to use fluoride

toothpaste. 1 2 3 4 5

I believe that regular visits to the Dentist will

prevent problems with my baby’s teeth. 1 2 3 4 5

Children should see a Dentist or Dental

Therapist by the time they are 1 year old. 1 2 3 4 5

I believe that maintaining both my and my

infant’s oral health costs too much. 1 2 3 4 5

I believe that keeping baby teeth until they are

due to fall out is not that important. 1 2 3 4 5

I am afraid of going to the Dentist because of

possible pain. 1 2 3 4 5

I believe that good oral health is important to

my overall health and well-being. 1 2 3 4 5

26. Did you visit the Dentist during your pregnancy? (Please tick one response only)

Yes

No

27. Which of the following did you give your new born child?

I practiced exclusive breast feeding up to 6 months for my new born

child

I only bottle-fed my new born child

I both breastfed and bottle-fed my new born child beyond 12 months

of age

28. Prior to my infant’s first teeth erupting, I cleaned my infant’s gums with a damp wash

cloth before bed.

191

Yes

No

29. How often have you taken your child to a Dentist since he/she was born?

Never

Once

Twice

Three times

More than 3 times

30. How old was your child when you started brushing his/her teeth with tooth paste?

Six months

Nine months

One year

After one year

After two years

31. Do you use fluoridated toothpaste for your child?

Always

Sometimes

Never

Don’t know

32. At what age did you leave your child with the responsibility of brushing his/her own

teeth?

33. Does your child brush his teeth on his/her own?

Yes

No

Two years

Three years

Four years

Five years

I still brush my child’s teeth

192

34. Do you check your child’s teeth after cleaning them? (Please tick one response only)

Yes

No

Don’t know

35. How often do you give your child carbonated drinks?

Never

Once a day

Twice a day

Three times a day

More than 3 times a day

36. What best describes your child’s consumption of sweets/lollies or sugary food habit?

37. How many times in the past 4 years have you seen a Dentist? (Please tick one

response Only)

Never

Once

Twice

Three times

More than 3 times

38. If once or more – what was the main reason for that visit? (Please tick one response

only)

Pain

Hole in tooth

Broken tooth

Dental check-up

Have teeth cleaned

39. I have untreated tooth decay myself

Doesn’t like sweets

Prefers sweets over fruits &

vegetables

Has sweets more than three times

a day

Gets sweets/lollies after only after

meals

Has the freedom to have sweets

whenever he/she wants

193

Yes

No

40. How often do you brush your teeth? (Please tick one response only)

Once a day

Two or more times a day

Never

Once a week

Other (specify)

41. Which of the following do you use to clean your teeth? (Please tick as many as you use)

Tooth brush

Dental floss

Tooth picks

Interdental brush

Chew sticks

Other (specify)

42. Do you use toothpaste containing fluoride? (Please tick one response only)

Yes

No

Don’t know

43. Cleaning of my infant’s teeth should start from (Please tick one response only)

The day the first tooth appears in the mouth

Once the first 4 teeth appear

Once the first 6 teeth appear

After my child becomes two years old

END

194

Appendix B: Kappa statistics for dental caries examination

Appendix B: Table 1 Calibration of examiner 1 for ICDAS criteria against a gold

standard

Mothers and children were included as participants for the calibration exercise. Table

indicates the number of surfaces examined by each examiner.

Number of observed agreements: 139 (92.67% of the observations)

Number of agreements expected by chance: 121.6 (81.04% of the observations)

Weighted Kappa= 0.810

SE of kappa=0.102

95% CI: From 0.414 to 0.813

Gold standard ICDAS 0 ICDAS 1 ICDAS 2 ICDAS 3 ICDAS 4 ICDAS 5 ICDAS 6 Total

surfaces

Ex

am

iner

1

ICDAS 0 130 0 0 0 0 0 0 130

ICDAS 1 6 2 0 0 0 0 0 8

ICDAS 2 4 0 0 0 0 0 0 4

ICDAS 3 0 0 2 0 0 0 0 2

ICDAS 4 0 0 0 0 1 0 0 1

ICDAS 5 0 0 0 0 0 3 0 3

ICDAS 6 0 0 0 0 1 0 1 2

Total

surfaces

140 2 0 2 2 3 1 150

195

Appendix B: Table 2 Calibration of examiner 2 for ICDAS criteria against a gold

standard

Mothers and children were included as participants for the calibration exercise. Table

indicates the number of surfaces examined by each examiner.

Number of observed agreements: 146 (97.33% of the observations)

Number of agreements expected by chance: 129.0 (85.97% of the observations)

Weighted Kappa= 0.928

SE of kappa = 0.093

95% confidence interval: From 0.628 to 0.992

Gold standard ICDAS 0 ICDAS 1 ICDAS 2 ICDAS 3 ICDAS 4 ICDAS 5 ICDAS 6 Total

surfaces

Ex

am

iner

2

ICDAS 0 137 1 0 0 0 0 0 138

ICDAS 1 2 1 0 0 0 0 0 3

ICDAS 2 1 0 1 0 0 0 0 2

ICDAS 3 0 0 0 2 0 0 0 2

ICDAS 4 0 0 0 0 1 0 0 1

ICDAS 5 0 0 0 0 0 3 0 3

ICDAS 6 0 0 0 0 0 0 1 1

Total

surfaces

140 2 1 2 1 3 1 150

196

Appendix C

Appendix C Table 1: Search strategy used in PubMed

# 1 dental caries [MeSH Terms]

# 2 dental [All Fields] AND caries [All Fields]

#3 dental caries [All Fields]

#4 Susceptibility [All Fields]

# 5 (#1 or #2 or #3 or #4)

# 6 Genetic [MeSH Term]

# 7 Genetic [All fields]

# 8 #7 AND #3

# 9 #5 AND #8

#10 epigenetics [MeSH Terms]

# 11 epigenetics [All Fields]

#12 #3 AND #11

#13 candidate genes [All Fields]

# 14 genome wide [All Fields]

#15 (#3 AND #7 or 11or #13 or #14)

#16 linkage [All Fields]

#17 association [All Fields]

#18 (#13 or #14 or #16 or #17 AND #4 AND #3)

Search limited to Journal Article[ptyp] AND ("1912/01/01"[PDAT] : "2015/12/31"[PDAT])

AND "humans"[MeSH Terms] AND English[lang] AND dental caries [Mesh Terms])

197

Appendix C Table 2: Search strategy used in Scopus

Data base selection

#1 Health

#2 Dentistry

#3 Oral Health

Subject area in Scopus

#4 Health Sciences

Key word search with additional search fields

# 5 dental caries [All Fields]

# 6 dental AND caries [All Fields]

#7 dental caries [All Fields]

#8 Susceptibility [All Fields]

#9 Genetic [All Fields]

# 10 (#5 or #6 or #7 or #8 and#9)

# 11 #5 AND #9

# 12 #10 AND #11

# 13 epigenetics [All Fields]

#14 #5 AND #13

#15 candidate genes [All Fields]

# 16 genome wide [All Fields]

#17 (#5 AND #9 or 13 or #15 or #16)

#18 association [All Fields]

#19(#15 or #16 or #17 or #18 AND #5 AND #8)

Search limited to Journal Article AND ("1912/01/01"[PDAT] : "2015/12/31"[PDAT]) AND

AND English[lang]

198

Appendix C Table 3: Search strategy used in Google Scholar

Key word search with additional search fields

# 1 dental caries

# 2 dental AND caries

#3 Susceptibility

#4 Genetic

# 5 (#1 or #2 or #3 or #1 and#4)

# 6 #1 AND #4

# 7 #5 AND #6

# 8 epigenetics

#9 #1 AND #8

#10 candidate genes

# 11 genome wide

#12 (#1 AND #4 or 8 or #10 or #11)

#13 association

#14 (#10 or #11 or #12 or #13 AND #1 AND #3)

Search limited to Journal Article AND ("1912/01/01"[PDAT] : "2015/12/31"[PDAT]) AND

AND English[lang] AND humans in all subject areas.

199

Appendix D: Supplementary tables for differentially methylated

CpG sites on chromosome 12 between cases and controls

Appendix D Table 1: Comparison of mean Differentially Methylated Regions on

chromosome 12 between controls and cases amongst children.

*DMR Location Controls Cases

Mean SD Mean SD P value

chr12_740087 27.787 26.31351 44.2081 29.1216

chr12_2690385 17.8054 31.74555 46.0709 23.2653

chr12_3333760 3.5088 8.5947 41.1555 22.28168 0.05

chr12_4303130 42.6501 17.17231 23.8803 28.69158

chr12_6929513 80.107 23.86684 59.122 36.98673

chr12_11700085 59.6729 15.06503 35.8385 22.20227

chr12_11700254 76.0531 4.57678 42.3561 36.46868

chr12_11700256 78.3614 2.93485 42.7813 36.24977

chr12_11700258 76.2923 4.99635 41.417 37.31218

chr12_11700277 79.5354 4.61533 45.0658 38.06553

chr12_11700292 80.6821 3.56008 44.5347 37.99269

chr12_11700303 83.743 6.17952 44.8866 38.21628 0.05

chr12_11700343 67.8906 9.03084 43.9098 37.80618

chr12_14974998 75.7742 28.12442 47.7288 19.24457

chr12_18845488 78.5426 13.75798 58.4449 38.35139

chr12_20960555 92.4863 5.89851 51.9749 14.75764 0.001

chr12_22408980 51.3718 8.81698 31.0428 7.8338 0.01

chr12_26351890 20.0206 15.5864 46.651 32.75298

chr12_27622729 68.6844 22.28025 32.7252 23.32907 0.05

inchr12_30309985 75.5484 14.37837 61.5824 19.80425

chr12_31821824 63.3135 30.45111 80.7474 29.0153

chr12_33139261 72.0545 17.1057 49.504 39.61165

chr12_55805945 88.2937 8.54696 64.8014 16.62681 0.05

chr12_64503585 83.0138 7.16783 67.0699 23.15768

chr12_90418121 50.303 30.44237 64.4311 23.76368

chr12_117363165 16.7824 26.26586 33.547 36.76977

chr12_117482887 20.2448 15.75705 37.2619 24.33456

chr12_117482900 21.532 16.36244 32.4161 20.41894

chr12_119594418 64.6671 16.36121 82.2457 12.68901

chr12_121198298 46.5608 20.15452 30.3233 37.48128

chr12_124421877 84.2063 13.21861 64.2233 15.49226 0.05

chr12_125448960 63.3316 35.20476 55.4548 35.54152

chr12_130498324 28.4407 5.41868 48.238 8.59233 0.001

chr12_130604735 69.6955 12.57381 82.0722 12.55787

chr12_131118432 7.6121 9.45917 35.0168 22.99084 0.05

chr12_131606752 20.8859 23.74313 34.5049 39.39545

chr12_132333255 62.1149 20.15986 75.2436 8.07253

chr12_132639381 67.8602 7.46409 85.6348 8.94127 0.01

chr12_133324690 96.3769 4.40792 74.1954 12.43519 0.01

*DMR - Differentially Methylated Regions, Considered level of significance p≤0.05

200

Appendix D Table 2: Comparison of mean Differentially Methylated Regions on

chromosome 12 between controls and cases amongst mothers.

*DMR location Controls Cases

Mean SD Mean SD P value

chr12_740087 28.7508 27.51415 50.1137 32.32284

chr12_2690385 28.2883 32.13572 39.1446 28.16432

chr12_3333760 5 12.24745 6.25 13.37234

chr12_4303130 49.5769 13.01096 28.0724 29.54887

chr12_6929513 87.5966 9.25084 64.5569 25.07899

chr12_11700085 59.2928 26.7023 35.2861 22.80244

chr12_11700254 73.6554 28.35755 31.2699 26.36198 0.05

chr12_11700256 72.3923 25.99988 36.3805 34.77748

chr12_11700258 71.6581 27.07353 32.9962 28.11834 0.05

chr12_11700277 74.4628 29.60109 34.9669 31.33842 0.05

chr12_11700292 74.538 25.69358 34.1378 32.14766 0.05

chr12_11700303 72.7728 25.13668 34.5742 30.25465 0.05

chr12_11700343 65.0265 23.73455 32.9658 29.54845

chr12_14974998 69.5373 19.44996 41.7707 29.72958

chr12_18845488 74.4713 28.25694 57.2818 34.80051

chr12_20960555 92.8054 6.91488 89.7376 6.57752

chr12_22408980 54.3388 4.14064 39.8481 7.02251 0.01

chr12_26351890 38.2243 22.83675 47.5197 27.34149

chr12_27622729 53.6066 25.94518 44.3527 26.83579

chr12_30309985 80.9757 10.24072 69.5668 20.0663

chr12_31821824 67.4206 35.95397 80.5296 22.04074

chr12_33139261 84.7545 8.59528 59.4302 26.02042

chr12_55805945 84.4266 8.29618 78.8481 19.20707

chr12_64503585 89.4536 7.13107 74.9638 18.28488

chr12_90418121 42.1258 22.25222 59.0858 23.00297

chr12_117363165 10.3509 17.49291 44.2665 37.37407

chr12_117482887 18.4112 9.21666 38.4728 19.28807 0.05

chr12_117482900 10.1832 5.89391 38.0391 21.16146 0.05

chr12_119594418 65.8448 8.36885 81.9041 10.87612 0.05

chr12_121198298 42.3358 32.86024 14.2857 22.5877

chr12_124421877 86.6468 12.72739 63.8745 25.35636

chr12_125448960 80.8952 24.52015 52.3284 40.61875

chr12_130498324 26.4647 9.57459 43.5969 9.41409 0.01

chr12_130604735 85.5972 9.51844 28.6111 24.88549 0.01

chr12_131118432 16.4997 17.5476 23.5311 13.14025

chr12_131606752 0.7576 1.85567 19.5076 30.88248

chr12_132333255 64.6186 2.62998 84.8906 8.44676 0.001

chr12_132639381 68.8844 12.76626 87.5738 10.10195 0.05

chr12_133324690 85.2131 13.45641 76.4431 13.03145

*DMR - Differentially Methylated Regions, Considered level of significance p≤0.05

201

Appendix E: Results of further pyrosequencing done on an

extended sample of individuals discordant for dental caries

Appendix E 1. Rationale

Based on the results of differentially methylates CpG sites between cases and controls in the

12 mother child dyads, further 26 DNA samples from mother child dyads (52 individual

samples) with high and low dental caries experience, were analysed for epigenetic variations.

It was decided to investigate Chromosome 12 was further, since it gave the highest differential

methylation between individuals with high and low dental caries experience. Eight CpGs

between sites 11700085 and 11700343 showed 41% methylation difference between cases and

controls where on average, cases had significantly lower methylation differences than cases (p

< 2.17E-103) (Table 7.3). Therefore, this particular CpG site on Chr12 was selected for further

pyrosequencing to support and validate the initial discovery results.

High-throughput pyrosequencing technique is a widely applicable, alternative technology for

the detailed characterization of nucleic acids. Pyrosequencing is a DNA sequencing technique

that is based on the detection of released pyrophosphate (PPi) during DNA synthesis

(http://www.biology-pages.info/P/Pyrosequencing.html). Pyrosequencing has the potential

advantages of accuracy, flexibility, parallel processing, and can be easily automated (Yun et

al., 2017). Furthermore, the technique dispenses with the need for labelled primers, labelled

nucleotides, and gel-electrophoresis (Ronaghi, 2001). Pyrosequencing is currently the fastest

method for sequencing a PCR product. Due to the fact that, pyrosequencing generates an

accurate quantification of the mutated nucleotides, the resequencing of PCR-amplified disease

genes for mutation scanning is one of the more interesting applications of the technique

202

(Ronaghi, 2001). Using this technique for resequencing results in longer read length than de

novo sequencing because nucleotide delivery can be specified according to the order of the

sequence (Garcia et al., 2000).

Appendix E 2. Results

Our initial pyrosequencing test runs on the targeted CpG sites indicated that the sites were

located in a highly repetitive region of the chromosome with much non-specific amplifications.

Hence there was a difficulty in designing precise primers with adequate specificity for reliable

results. Therefore, three different CpGs that had good amplification were selected from the

proposed region (Chr 12 between genomic positions 11700080 and 11700350) although they

were not included in our discovery results. It was expected that the three CpG sites might carry

the same association signal by way of being directly adjacent in the DNA sequence for this a

frequent observation for strings of CpGs in a DNA region. The first CpG site

(Chr12:11700151) was very accurate and could be validated with specific primers. The second

CpG site (Chr12:11700148) was polymorphic and not present in all sample types, so the data

for this site were variable. The third CpG site (Chr12:11700124) was downstream of a very

variable region, and the results at this site depended on the particular samples and the sequence

variation was observed in the region (Appendix D Table 1).

However, according to the data there appears to be very little variation among all the samples

tested (Appendix D Table 1). This implies that there was no association signal in the cohort,

which is contrary to our initial results and indicates that our expectation of association-by-

proxy was not realised (Appendix D Table 2).

203

Percentage methylation in individuals with high caries experience Percentage methylation in individuals with low caries experience

Sample

ID

*Caries

experience

Chr12:11700151 Chr12:11700148 Chr12:11700124 Mean

methylation for

the three sites

Sample

ID

*Caries

experience

Chr12:11700151 Chr12:11700148 Chr12:11700124 Mean

methylation for

the three sites

Children Children

4 6.00 57.37 17.91 33.47 36.25 1 3.00 52.40 18.81 27.98 33.06 8 19.00 53.50 18.27 34.02 35.26 7 2.11 52.83 17.70 31.54 34.02

52 9.00 53.91 18.47 35.95 36.11 14 1.65 56.50 19.79 35.45 37.25

64 11.00 54.42 17.52 35.32 35.75 34 2.61 51.34 17.51 30.71 33.19 71 13.33 54.53 18.50 34.45 35.83 48 .00 56.31 18.60 36.14 37.02

105 16.00 56.38 17.46 34.50 36.11 58 .00 55.82 17.06 34.55 35.81

114 64.80 58.09 18.17 36.28 37.51 61 .00 55.11 19.61 35.52 36.75 115 16.00 52.84 16.70 33.74 34.43 67 .00 53.90 18.35 35.21 35.82

125 22.22 52.85 18.02 35.72 35.53 112 1.25 59.34 17.82 36.39 37.85

129 7.50 54.89 18.94 37.19 37.01 122 3.33 57.66 18.45 35.57 37.23 138 6.00 57.05 18.96 36.68 37.56 149 .00 59.48 19.07 36.76 38.44

152 8.24 54.47 15.38 33.36 34.40 153 .83 53.61 16.20 33.79 34.53

176 21.11 56.67 17.98 35.31 36.65 170 .00 60.13 18.87 38.19 39.06 223 28.42 55.33 17.69 36.07 36.36 191 .00 62.61 19.19 37.89 39.90

243 20.00 58.42 17.90 36.24 37.52 211 .00 52.36 17.53 33.34 34.41

247 6.67 56.16 19.80 37.01 37.66 233 1.25 56.44 18.07 35.83 36.78 249 18.82 51.12 18.29 32.85 34.09 239 1.00 57.37 17.28 35.31 36.65

257 6.00 61.21 17.51 37.23 38.65 255 .00 53.34 17.61 33.01 34.65

96 6.00 53.50 17.98 35.11 35.53 164 4.21 61.56 16.54 37.29 38.46

Mothers Mothers

5 15.71 52.38 19.05 34.37 35.27 2 .00 56.79 19.59 33.55 36.64

9 25.00 59.14 18.45 33.89 37.16 6 .95 58.82 17.88 35.14 37.28 51 26.43 61.23 18.46 37.13 38.94 15 15.00 56.00 17.11 34.81 35.97

63 39.49 56.51 19.58 35.30 37.13 33 10.00 52.05 17.87 30.60 33.51

70 22.14 51.97 16.91 32.76 33.88 47 7.69 52.04 17.06 32.99 34.03 104 45.00 60.20 17.77 36.40 38.12 57 15.48 60.70 19.66 34.41 38.26

113 64.80 58.36 17.63 36.19 37.39 62 9.29 59.51 18.71 36.25 38.16

116 33.79 50.83 16.93 31.24 33.00 82 4.29 56.02 19.05 35.93 37.00 124 32.37 60.40 18.04 37.44 38.63 110 2.58 56.75 17.38 34.13 36.09

128 29.17 57.44 18.72 36.17 37.44 121 20.65 54.70 18.73 31.82 35.08

137 46.15 54.71 18.45 35.02 36.06 148 14.38 57.67 20.13 37.09 38.30 150 23.08 55.37 18.72 34.56 36.22 154 16.55 57.90 18.16 35.71 37.26

174 75.56 54.51 18.49 37.15 36.72 171 10.00 55.77 18.58 36.67 37.01

221 37.78 52.46 17.28 33.84 34.53 190 6.54 60.24 18.22 37.59 38.68

244 26.32 57.81 17.98 35.85 37.21 210 8.75 56.08 15.78 35.21 35.69 248 30.71 57.60 19.32 36.66 37.86 234 10.00 53.45 17.22 34.97 35.21

250 37.50 51.18 16.59 33.57 33.78 240 6.00 60.63 18.16 38.67 39.15 258 27.74 51.10 18.15 34.03 34.43 256 10.40 57.02 16.91 35.37 36.43

95 24.29 58.33 17.83 35.18 37.11 166 15.00 54.04 17.71 34.21 35.32

Appendix E Table 1: Percentage methylation analysis of CpG sites; Chr12:11700151, Chr12:11700148 and Chr12:11700124 between

individuals with high and low caries experience

*Caries experience was measured as a percentage of tooth surfaces affected with dental caries (including early enamel caries)

204

Appendix E Table 2: Comparison of differential methylation of Chr12:11700151,

Chr12:11700148 and Chr12:11700124 between individuals with high and low caries

experience

Individuals with high caries experience (n=38) Individuals with low caries experience (n=38)

Mean (SD) Mean (SD) t P

Chr12:11700151 (CpG#139) 55.63(2.9) 56.42 (2.9) 1.17 0.25

Chr12:11700148 (CpG#139A) 18.04(0.86) 18.10 (1.02) 0.26 0.79

Chr12:11700124 (CpG#140) 35.19 (1.49) 34.88 (2.24) 0.70 0.49

For all three different CpG sites analysed, there was no marked difference in the mean

percentage methylation and a significant difference between individuals with high and low

caries experience could not be observed.

Appendix E Table 3: Correlation between individuals with high and low caries experience

and percentage methylation of CpGs; Chr12:11700151, Chr12:11700148 and

Chr12:11700124

CpG site Correlation coefficients

Individuals with high

caries experience (n=38)

Individuals with low caries

experience (n=38)

Chr12:11700151 (CpG#139) 0.10 -0.01

Chr12:11700148

(CpG#139A)

0.01 0.01

Chr12:11700124 (CpG#140) 0.15 -0.07

Similarly, as described in table 3, there were no significant correlations between high and low

caries individuals with regards to percentage methylation of the three selected CpG sites

(Appendix E Table 3). Since the methylation values were consistent across all individuals (ie

partially methylated), it can be suggested that these CpGs are not associated with dental caries.

The possible reasons could be lack of specificity in this repetitive region, lack of totally caries

free and high caries individuals to compare with as cases and controls, and possible low quality

of DNA.

205

Appendix E 3. Future directions

As the initial discovery assay failed due to lack of specificity and high repetitiveness, further

repetitive assays for instance, LINE1, ALU, SAT2, or HERV could be performed, for these are

global assays that cover repetitive regions throughout the genome. Moreover, pyrosequencing

could be performed on the second best target of the discovery analysis, by selecting target

regions which do not belong to a repetitive region.