Proposal summaries

These are research proposals that have been approved by the ALSPAC exec. The titles include a B number which identifies the proposal and the date on which the proposals received ALSPAC exec approval.

Click here to export results in Word format.

B1460 - In-silico detection of deletions in KLK3 from the ALSPAC raw SNP data - 03/12/2012

B number: 
B1460
Principal applicant name: 
Santiago Rodriguez (University of Bristol, UK)
Co-applicants: 
Prof Ian Day (University of Bristol, UK), Dr Osama Al-Ghamdi (University of Bristol, UK)
Title of project: 
In-silico detection of deletions in KLK3 from the ALSPAC raw SNP data.
Proposal summary: 

Aim: To estimate the frequency of KLK3 copy number variants in the general population.

KLK3 encodes PSA, the most widely used biomarker in the early detection of prostate cancer. Genetic variants in KLK3 may influence serum PSA levels. From a pilot of selected controls within the ProtecT biorepository, we have identified three subjects with very low serum PSA levels (less than 1 ng/micro-L) with evidence of heterozygous deletions that entirely encompassed KLK3 (Rodriguez et al., Clinical Chemistry, in press)

Hypothesis: That deletions in the KLK3 region can be identified from SNP raw data available from ALSPAC.

Background: SNP arrays were originally designed to genotype SNPs; however they have been adapted for structural variant discovery. Several tools to analyse data from SNP arrays have been developed recently, however they tend to be platform-specific (i.e. able to process either Affymetrix or Illumina array data).

Algorithms that handle data from Affymetrix arrays include programmes as Birdsuite (1), and ITALICS (2). Other algorithms were developed to analyse data from Illumina arrays, such as SCIMM (3) and TriTyper (4). Some software applications are "versatile"; capable of handling data from both platforms (e.g.) PennCNV (5) and QuantiSNP (6).

Approach: Access to the fluorescence SNP raw data for a genomic interval of ~18Mb on chromosome 19 is required; (Chromosome 19: [41,000,000 - 59,128,983] Human GRCh37 build). This interval includes the entire human kallikrein locus, comprised of 15 kallikrein genes -in tandem-, including KLK3 that encodes the prostate specific antigen (PSA). Indeed serum PSA of normal women is typically undetectable (around 1 pg/mL) (7), however, deletions in KLK3 could be transmitted from mothers to children. KLK3 deletions from the general population, and from prostate cancer cases are considered. ALSPAC mothers will fit in as a "control" group to data from the 1958 cohort and from the ProtecT cohort (males with prostate cancer and controls). Copy number variants with high confidence scores will be assembled from multiple scans using several algorithms, filtering out false positive/discrepant signals in the process. A list of copy number variants called from this study will be compared across other studies to estimate the frequency of deletion events in KLK3 among males vs. females, and male controls vs. prostate cancer cases. We will also request access to the SNP genotype data for the same genomic region for children. This will enable us to study deletion transmission patterns from mothers to children.

References:

Santiago Rodriguez, Osama A Al-Ghamdi, Kimberley Burrows, Philip A.I. Guthrie, J. Athene Lane, Michael Davis, Gemma Marsden, Khalid K Alharbi, Angela Cox, Freddie C Hamdy, David E Neal, Jenny L Donovan, and Ian N. M. Day (2012) Very low PSA levels and deletions of the KLK3 gene. Clin Chem, -in press-.

(1) Korn, J. M., F. G. Kuruvilla, S. A. McCarroll, A. Wysoker, J. Nemesh, S. Cawley, E. Hubbell, J. Veitch, P. J. Collins, K. Darvishi, C. Lee, M. M. Nizzari, S. B. Gabriel, S. Purcell, M. J. Daly & D. Altshuler (2008) Integrated genotype calling and association analysis of SNPs, common copy number polymorphisms and rare CNVs. Nat Genet, 40, 1253-60.

(2) Rigaill, G., P. Hupe, A. Almeida, P. La Rosa, J. P. Meyniel, C. Decraene & E. Barillot (2008) ITALICS: an algorithm for normalization and DNA copy number calling for Affymetrix SNP arrays. Bioinformatics, 24, 768-74.

(3) Kelley, D. R. & S. L. Salzberg (2010) Clustering metagenomic sequences with interpolated Markov models. BMC Bioinformatics, 11, 544.

(4) Franke, L., C. G. de Kovel, Y. S. Aulchenko, G. Trynka, A. Zhernakova, K. A. Hunt, H. M. Blauw, L. H. van den Berg, R. Ophoff, P. Deloukas, D. A. van Heel & C. Wijmenga (2008) Detection, imputation, and association analysis of small deletions and null alleles on oligonucleotide arrays. Am J Hum Genet, 82, 1316-33.

(5) Wang, K., M. Li, D. Hadley, R. Liu, J. Glessner, S. F. Grant, H. Hakonarson & M. Bucan (2007) PennCNV: an integrated hidden Markov model designed for high-resolution copy number variation detection in whole-genome SNP genotyping data. Genome Res, 17, 1665-74.

(6) Colella, S., C. Yau, J. M. Taylor, G. Mirza, H. Butler, P. Clouston, A. S. Bassett, A. Seller, C. C. Holmes & J. Ragoussis (2007) QuantiSNP: an Objective Bayes Hidden-Markov Model to detect and accurately map copy number variation using SNP genotyping data. Nucleic Acids Res, 35, 2013-25.

(7) Chang, Y. F., S. H. Hung, Y. J. Lee, R. C. Chen, L. C. Su, C. S. Lai & C. Chou (2011) Discrimination of breast cancer by measuring prostate-specific antigen levels in women's serum. Anal Chem, 83, 5324-8.

Date proposal received: 
Thursday, 8 November, 2012
Date proposal approved: 
Monday, 3 December, 2012
Keywords: 
Genetics, Cancer
Primary keyword: 

B1472 - NMR metabolomics analysis of ALSPAC Child Mother and Fathers samples - 22/11/2012

B number: 
B1472
Principal applicant name: 
Prof George Davey Smith (University of Bristol, UK)
Co-applicants: 
Prof Mika Ala-Korpela (University of Oulu, Europe), Prof Debbie A Lawlor (University of Bristol, UK), Dr Susan Ring (University of Bristol, UK)
Title of project: 
NMR metabolomics analysis of ALSPAC Child, Mother and Father's samples.
Proposal summary: 

The development of metabolomics research in population health science has been driven by mass spectrometry (MS) and proton nuclear magnetic resonance (NMR) spectroscopy as the two key experimental technologies. NMR spectroscopy is increasingly used because of its capability to simultaneously detect a wide range of metabolites, thus providing a "snapshot of the metabolite composition" in biofluids. NMR-based applications can also offer fully automated and highly reproducible high-throughput experimentation in a very cost-effective manner. During the last 5 years Prof Mika Ala-Korpela's Computational Medicine team has focused on developing an NMR metabolomics platform for analysing human serum - 1H NMR spectroscopy (Tukiainen et al 2008). This novel methodology has been used to analyse serum from over 100,000 individuals (in less than 4 years). The method uses three molecular windows, (two applied to native serum and one to serum lipid extracts requiring minimal preparation) to quantify the 216 metabolomic traits, including 14 lipoprotein subclasses with detailed molecular information on serum lipid extracts including free and esterified cholesterol, sphingomyelin, degree of saturation and omega-3 fatty acids, 117 metabolites (including 80 lipoproteins, 15 lipids and 22 low molecular weight metabolites), 99 derived metabolic measures indexing amino acid metabolism, gluconeogenesis, ketogenesis, kidney function, and fatty acid saturation, as well as measures such as apolipoprotein A-I and B (see list in Kettunen et.al 2012) . The majority of analyte data is reported as a molar concentration, with derived variables reported as ratios . 1H NMR spectroscopy-based quantification reduces measurement error for main blood lipid fractions, compared to the usual enzymatic methods, and also allows detection of associations with lipid and metabolic sub-phenotypes that may be proximal consequences of genetic variation and environmental risk factors.

Genome-wide associations with novel metabolic pathways are likely to be identified with greater statistical efficiency using phenotypes obtained from this method, and estimates from non-genetic association studies are also likely to be more precise. For example, in a GWAS of 8,330 adults using 216 serum metabolic phenotypes measured using 1H NMR spectroscopy 31 GWAS loci with p less than 10-10 were identified, 20 of which replicated previously known associations obtained in much larger sample sizes and 11 of which were novel(Kettunen et. al. 2012). In a second study of 9,179 adults, 6 novel metabolites associated with CVD mortality, which were independent of established risk factors, were identified. (al-hussaini et.al. 2012)

We plan to analyse all suitable plasma and serum samples collected from ALSPAC participants, their mothers and fathers at as many timepoints as possible. Methodology exists for analysis of EDTA plasma and serum samples from adults and children. Methodology will be developed for analysis of cord blood plasma and/or serum samples and may be developed for heparin plasma samples.

Data generated will become part of the ALSPAC resource and utalised for a numerous association studies which will be covered by separate applications to ALSPAC Executive.

Samples will initially be analysed in Prof Ala-Korpela's Computational Medicine laboratories,

University of Oulu & University of Eastern Finland (Kuopio), (details of first sets on samples in Appendix 1b below).

UoB has recently invested in core metabolomics facilities, including a new 500 MHz instrument NMR system 2, which will be overseen by Prof Ala-Korpela. In the future, analysis will be completed in these new UoB facilities.

Tukiainen T, Tynkkynen T, Makinen VP et al. A multi-metabolite analysis of serum by 1H NMR spectroscopy: early systemic signs of Alzheimer's disease. Biochem Biophys Res Commun 2008;375:356-361

Kettunen J, Tukiainen T, Sarin AP et al. Genome-wide association study identifies multiple loci influencing human serum metabolite levels. Nat Genet 2012;44:269-276

al-hussaini A, Sehmi J, Tan T, Ala-Korpela M, Kooner J, Chambers J. Identification of novel metabolic biomarkers for cardiovascular mortality. Journal of the American College of Cardiology 2012;59:E1649-doi:10.1016/S0735-1097(12)61650-7

Date proposal received: 
Thursday, 22 November, 2012
Date proposal approved: 
Thursday, 22 November, 2012
Keywords: 
Metabolomics
Primary keyword: 

B1471 - Developmental origins of mood symptoms in children and adolescents The role of genes maltreatment and social process - 22/11/2012

B number: 
B1471
Principal applicant name: 
Dr Erin Dunn (Massachusetts General Hospital, USA)
Co-applicants: 
Dr Jordan Smoller (Harvard School of Public Health, USA), Prof Ezra Susser (Columbia University, New York, USA)
Title of project: 
Developmental origins of mood symptoms in children and adolescents; The role of genes, maltreatment, and social process.
Proposal summary: 

SPECIFIC AIMS

This research proposal aims to identify mechanisms explaining why early adversity, namely child maltreatment, is consistently associated with an elevated risk for mental disorder 1-7. While there has been much emphasis on the long-term effects of adversity, including how genes and adversity jointly shape risk 8, there is limited research on how timing of adversity shapes trajectories of development and risk for internalizing symptoms.

The overarching concept we seek to test is that there are "sensitive periods" in development 9-11, or windows of time in the lifespan when the developing brain is particularly vulnerable or sensitive to experience, including adversity. Sensitive periods are characterized by phases of high plasticity and rapid brain development in a given domain 12; 13. First articulated by Hubel and Wiesel 14, a robust literature in animals 15; 16 and now in humans has identified sensitive periods in visual 17 and auditory system processes, including language development 18-22. Several genes (e.g., GAD1, GAD2, GABA1 alpha subunit, and BDNF) and environmental exposures (e.g., exercise, psychotropic medications) have been shown in animal studies to manipulate sensitive periods, either triggering their opening or closing 23. Importantly, recent literature also suggests sensitive periods are not fixed or limited solely to the early years of life and brain development. Instead, sensitive periods can be reopened throughout the lifecourse, even in adulthood, as a result of genetically-mediated factors and environments that "remove the brakes on plasticity" 24; 25.

Currently, we lack an in-depth understanding of the time-dependent effects of adversity on the Research Domain Criteria (RDoC) functional domains 26-29. This is especially true for social system processes, including attachment, social cognition, and social communication, which are likely precursors of internalizing symptoms. Although understudied, extant research suggests there are sensitive periods in attachment 30 and social cognition 31. Child abuse and neglect are known to affect social system processing, including perception of emotional expression32. However, it is unclear whether these effects are the same for all maltreated children or whether there is individual variation, depending on the age when children are first exposed. To that end, early evidence from epidemiological studies now suggests the effects of child maltreatment on internalizing symptoms differ depending on age at first exposure, which is suggestive of a sensitive period 33-35.

Based on prior research and our preliminary studies, the main hypothesis we are testing is that disruptions in social system processes explain the elevated risk of internalizing symptoms among children exposed to maltreatment and that the magnitude of this relationship will depend on the developmental timing of exposure to adversity and what sets of genes are expressed at that point in time.

HYPOTHESES

This proposal will test the following aims/hypotheses:

Aim 1: Explore the time-dependent effect of maltreatment on social domains/internalizing symptoms

Hypothesis 1: Children exposed to maltreatment during a sensitive period (age 3-5) will have lower functioning in social system domains and higher internalizing symptoms when compared to both non-exposed children and children exposed outside of the sensitive period, even after adjusting for maltreatment features (e.g., duration).

Aim 2: Examine the association between age at first onset of exposure to child maltreatment, social function, and internalizing symptoms in adolescents

Hypothesis 2: Social functioning will partially mediate or explain the association between age at first onset of exposure to child maltreatment and internalizing symptoms.

Aim 3: Investigate whether developmental genes modify the association between age at first exposure to child maltreatment and both social domain functioning and internalizing symptoms

Hypothesis 3: Variants in genes shown in animal studies to regulate sensitive periods, and sets of genes shown in humans to be differentially expressed across the lifespan, will modify the effect of adversity timing on domains of social functioning and internalizing symptoms.

The expected outcome of this proposal will be the development of a more nuanced approach to defining sensitive periods and the time-dependent effects of adversity on domains relevant to RDoC. This knowledge will have major impacts on the field, including greater insights into the biology of and mechanisms explaining susceptibility to internalizing symptoms and lead to the development of novel preventions and treatments.

EXPOSURE VARIABLES

Caregiver Reported Childhood Adversities: At yearly intervals between ages 1 and 8, caregivers provided information about whether the child was exposed to the different adversities in the past 12 to 18 months, including being taken into some form of foster care; physically hurt by someone else; sexually abused; separated from mother; and separated from father. This was included in the section entitled "Upsetting Events."

Child Reported Childhood Adversity: At age 16 children were asked to report whether they had been exposed to 23 different types of stressors since age 12.

Documented Reports of Child Maltreatment: We are also seeking information from the 329 children who were invested by social services for suspected maltreatment prior to age 6; this includes 162 children who were placed on local child protection registries as having confirmed cases of maltreatment.

MEDIATOR VARIABLES

Diagnostic Assessment of Non-Verbal Accuracy (DANVA): We are requesting data from the DANVA, which provides information about the offspring's ability to accurately detect facial displays of emotion. This task was administered at age 8.

Emotion Triangles Task: The child's ability to attribute emotional states to other individuals, a component of theory of mind, was assessed at age 13, using the emotion triangle task.

Separation Anxiety, as measured by the Development and Wellbeing Assessment (DAWBA), was asssessed at ages 7, 10, 13, 15.

Social and Communication Disorders Checklist: We are requesting data from this measure, which taps domains of social cognition. It appears this measure was administered to children at ages 7 and 15 (and possibly 10 and 13).

OUTCOME VARIABLES

We are proposing to examine the following outcome variables: (1) Rutter parent scale for preschool children; (2) Strengths and difficulties questionnaire; (3) Development and Wellbeing Assessment; (4) Short Mood and Feelings Questionnaire. For all of these outcomes, we will examine symptoms of mood and anxiety disorders (as a continuous measure) as well as extremes in the symptom distribution or diagnoses (from DAWBA).

EFFECT MODIFIERS

We are proposing to examine sets of genes as effect modifiers of the association between maltreatment and both social system processing and mood and anxiety disorders. Please see Appendix 1B for further details on the genetic information we are requesting.

COVARIATES

We would also be interested in receiving basic demographic variables to use as covariates in our analysis. This includes information about: age, gender, race/ethnicity, socioeconomic status.

Date proposal received: 
Thursday, 22 November, 2012
Date proposal approved: 
Thursday, 22 November, 2012
Keywords: 
GWAS, Mental Health, Social Science
Primary keyword: 

B1470 - Investigation of the mapping from genetic markers to facial features - 22/11/2012

B number: 
B1470
Principal applicant name: 
Dr Colin Campbell (University of Bristol, UK)
Co-applicants: 
Prof Stephen Richmond (University of Cardiff, UK), Dr Dave Evans (University of Bristol, UK), Dr Lavinia Paternoster (University of Bristol, UK)
Title of project: 
Investigation of the mapping from genetic markers to facial features.
Proposal summary: 

Aim: we aim to investigate the mapping from genetic variants to normal facial variation measures using data from the Avon Longitudinal Study of Parents and Children (ALSPAC). 3D high-resolution images have been obtained using two laser scanners for 4747 children. The images were merged, aligned and 22 important facial landmarks were identified. Their x, y and z co-ordinates were used to generate 54 3D distances reflecting facial features. These children also have genome-wide single nucleotide polymorphism (SNP) data available for ~2.5 million genetic markers.

Dr. Campbell has a long standing interest in machine learning and the posed problem of mapping from genetic data to a set of facial feature measures is a classic problem within machine learning. We will start by looking at some Bayesian ARD algorithms, previously devised by Dr. Campbell and collaborators (ARD = automatic relevance determination). These algorithms have an inbuilt mechanism to discard features (in this case genetic variant data) irrelevant to the considered problem. They derive a regression function (since the mapped facial feature will be continuously-valued). Performance evaluation will be on test data by ranked performance i.e. we make a prediction from the input data, evaluate the distance between this prediction and the corresponding facial feature measures of members of the test group, hence determining the position of the correct matching value in the rank list. For n members of the test set, an average rank of n/2 indicates null predictive performance. We will experiment with other types of classifier and other feature selection strategies, including predictors which give a posterior spread for the prediction (a mode and a spread for the predicted facial features). We also aim to look at other machine learning tools, for example, sparseCCA (canonical correlation analysis) which may indicate the correlates between sets of input (genetic) features and facial features.

Dr. Campbell has already contacted Dr. Lavinia Paternoster (MRC CAiTE centre) and will work closely with her and the rest of the team generating and analysing this data. We only envisage an initial look-see investigation to determine the level of predictive performance which may be achievable. It is possible the size of the input data (~2.5 million genetic markers) may be too prohibitive in computational cost for the anticipated Bayesian ARD algorithms. Also predictive performance may be too low. However, should positive results be obtained, we will communicate this to Lavinia and collaborators to discuss extensions and any possible scope for publications.

Date proposal received: 
Thursday, 22 November, 2012
Date proposal approved: 
Thursday, 22 November, 2012
Keywords: 
GWAS
Primary keyword: 

B1469 - Long-term effects of infant sleeping position - 22/11/2012

B number: 
B1469
Principal applicant name: 
Prof Jean Golding (University of Bristol, UK)
Co-applicants: 
Dr Peter Blair (University of Bristol, UK), Prof Alan Emond (University of Bristol, UK)
Title of project: 
Long-term effects of infant sleeping position.
Proposal summary: 

Aims: To determine whether sleeping position in the first 6 months is associated with long-term phenotypic changes.

Hypotheses:

1. That there is no adverse association of long-term physical health associated with sleeping on the back or side.

2. That a consequence of increased risk of chest infection associated with prone sleep position is a reduction in lung function in childhood.

3. That prone sleeping may be associated with improved motor ability and coordination.

4. That sleeping position has no long-term effect on cognitive abilities, behaviour or educational attainment.

Exposure variables:

Sleeping position, and other features of bedding and parenting up to 18 months

Outcome variables

All phenotypes on the database

Confounding variables

Gender; socio-economic features; housing; diet; parental education; child care; birthweight; gestation; etc.

Statistical approach will use a phenome scan, taking account of multiple testing. Regression analyses will be used to account for confounders as appropriate.

Date proposal received: 
Thursday, 22 November, 2012
Date proposal approved: 
Thursday, 22 November, 2012
Keywords: 
Sleeping Positions, Moto Co-ordination, Motor Co-ordination
Primary keyword: 

B1468 - The causal role of the nutritionally-regulated IGF system in prostate cancer Mendelian randomization study - 22/11/2012

B number: 
B1468
Principal applicant name: 
Prof Richard Martin (University of Bristol, UK)
Co-applicants: 
Prof George Davey Smith (University of Bristol, UK), Prof Jeff Holly (University of Bristol, UK), Prof Jenny Donovan (University of Bristol, UK), Prof David Gunnell (University of Bristol, UK), Dr Sarah J Lewis (University of Bristol, UK), Dr Tom Palmer (University of Bristol, UK), Dr Mari-Anne Rowlands (University of Bristol, UK)
Title of project: 
The causal role of the nutritionally-regulated IGF system in prostate cancer: Mendelian randomization study.
Proposal summary: 

Background:

The main component of this investigation is based in the ProtecT prostate cancer trial, co-ordinated from the School of Social and Community Medicine. This study is funded by a World Cancer Research Fund grant. We are applying to use ALSPAC data in order to conduct a validation exercise, and to perform a genome-wide association study (GWAS).

The nutritionally-regulated insulin-like growth factor (IGF) system, which includes two IGFs (IGF-I and IGF-II) and six IGF-binding proteins (IGFBP-1 to -6) play a key role in somatic growth, and activate potentially carcinogenic intracellular signalling networks. Many epidemiological studies observe positive associations of circulating IGF-I with prostate cancer but our recent systematic review highlights inconsistencies,[1] and it is unclear whether elevated IGF-I causes prostate cancer or reflects confounding or reverse causality. We have completed the largest single study we are aware of to investigate IGFs/IGFBPs in prostate cancer (ProtecT study), revealing strong positive associations of IGF-II, IGFBP-2 and IGFBP-3 with screen-detected prostate cancer (all p trendless than 0.001) but no association of serum IGF-I.[2] Our findings are consistent with other much smaller investigations for IGF-II, IGFBP-2 and IGFBP-3, but causality remains to be established.

Recently discovered genetic associations with prostate cancer are linked to the IGF-signalling pathway. In a genome-wide association study (GWAS),[3] 4 of 7 genotypes linked to prostate cancer are also involved in the IGF system: homozygous variants at insulin-IGF2 and NKX3.1 (an androgen-regulated tumour suppressor gene that regulates IGFBP-3) had up to a 50% increased prostate cancer risk, while homozygous variants at ITGA6 (encodes integrins that interact with IGFBPs) and PDLIM5 (inter-connected to an IGF-1R signaling protein that regulates migration) had up to a 65% decreased risk. Other common IGF-related genes (e.g. IGF-1R; PI3K; INS; IRS-1/-2) are associated with prostate or other cancers.[4-9]

Some genotypes are, simultaneously, related to both IGF/IGFBP levels and cancer; e.g. men homozygous for IGFBP-3 -202A/C promoter variants have lower circulating IGFBP-3,[10] a 2.5-fold increased prostate cancer risk[11] and a four-fold increased risk of metastatic disease.[12] IGF-1-variants are associated with both circulating IGF-I[8] and prostate (OR=1.46) [8] and breast (OR=1.4) [13] cancer; IGFBP-3 rs2270628 is associated with IGF-I and ovarian cancer (ORs=1.18-1.36).[14] There are very few studies relating genetic variants to IGF-II or IGFBP-2 levels. One GWAS relating genetic loci to circulating IGF-I and IGFBP-3 has been carried out in middle-aged adults, and identified several loci that were associated with IGFBP-3 and IGF-I concentrations.[15].

Aims:

We aim to clarify the role of the IGF system in prostate cancer, by using genetic variants as 'instruments' for measured circulating IGF-I, IGF-II, IGFBP-2 and IGFBP-3 and conducting formal Mendelian randomization (MR) instrumental variables (IV) analyses to obtain causal (unconfounded and unbiased) odds ratios for associations of circulating IGFs/IGFBPs with prostate cancer initiation and progression. Application of MR will identify IGFs/IGFBPs that are causally important in prostate cancer initiation or progression, which could be manipulated for primary or secondary prostate cancer prevention. Some trials of dietary interventions to reduce IGFs/IGFBPs in men at high risk of cancer are already underway, but should only be warranted if a causal link is demonstrated, particularly given potential unintended IGF-related effects (e.g. on insulin resistance, cardiovascular disease and cognitive decline). If we determine, instead, that raised IGFs/IGFBPs are a consequence of prostate cancer (reverse causality), the focus of future research would shift to assessing these ligands as nutritionally-related biomarkers of diagnosis or cancer monitoring/surveillance.

Objectives:

1) To obtain unbiased estimates of the diection and magnitude of the the causal associations of circulating IGF-I, IGF-II, IGFBP-2 and IGFBP-3 with prostate cancer prevalence, stage and grade, and disease progression. This will involve i) deriving genetic allele scores based on single nucleotide polymorphisms (SNPs) associated with IGF/IGFBP levels, in approximately 1000 controls from the ProtecT prostate cancer cohort; ii) validating the allele scores in ASLPAC children (or deriving new allele scores if we observe differences in SNP-IGF associations in adults versus children).

2) To conduct a genome-wide association study to identify SNPs associated with circulating IGF-I, IGF-II, IGFBP-2 and IGFBP-3 during childhood, among children in ALSPAC.

Hypothesis:

Raised levels of circulating IGF-I, IGF-II, IGFBP-2 and IGFBP-3 are causally associated with increased risk of prostate cancer overall; advanced stage and grade; and with disease progression.

Exposure:

GWAS (which will include selected variants in IGF-related genes)

Outcome:

Circulating IGF-I, IGF-II, IGFBP-2 and IGFBP-3 levels in all children in whom it was measured.

Confounding variables:

Age at time of IGF/IGFBP measurement, sex, 10 principal components for population stratification.

Reference List

1 Rowlands M, Gunnell D, Harris R, Vatten LJ, Holly JMP, Martin RM: Circulating insulin-like growth factor peptides and prostate cancer risk: A systematic review and meta-analysis. Int J Cancer 2009;124:2416-2429.

2 Rowlands MA, Holly JMP, Gunnell D, Donovan J, Lane JA, Hamdy F, Neal DE, Oliver S, Smith GD, Martin RM: Circulating Insulin-Like Growth Factors and IGF-Binding Proteins in PSA-Detected Prostate Cancer: The Large Case-Control Study ProtecT. Cancer Res 2012;72:503-515.

3 Eeles RA, Kote-Jarai Z, Al Olama AA, et al. Identification of seven new prostate cancer susceptibility loci through a genome-wide association study. Nat Genet 2009;41:1116-1121.

4 Schumacher FR, Cheng I, Freedman ML, et al. A comprehensive analysis of common IGF1, IGFBP1 and IGFBP3 genetic variation with prospective IGF-I and IGFBP-3 blood levels and prostate cancer risk among Caucasians. Hum Mol Genet 2010;19:3089-3101.

5 Koutros S, Schumacher FR, Hayes RB, et al. Pooled Analysis of Phosphatidylinositol 3-Kinase Pathway Variants and Risk of Prostate Cancer. Cancer Res 2010;70:2389-2396.

6 Neuhausen SL, Slattery ML, Garner CP, Ding YC, Hoffman M, Brothman AR: Prostate cancer risk and IRS1, IRS2, IGF1, and INS polymorphisms: strong association of IRS1 G972R variant and cancer risk. Prostate 2005;64:168-174.

7 Ho GY, Melman A, Liu SM, Li M, Yu H, Negassa A, Burk RD, Hsing AW, Ghavamian R, Chua SC, Jr.: Polymorphism of the insulin gene is associated with increased prostate cancer risk. British Journal of Cancer 88(2):263-9, 2003.

8 Johansson M, Mckay JD, Stattin P, Canzian F, Boillot C, Wiklund F, Adami HO, Balter K, Gronberg H, Kaaks R: Comprehensive evaluation of genetic variation in the IGF1 gene and risk of prostate cancer. Int J Cancer 2007;120:539-542.

9 Cheng I, Stram DO, Penney KL, Pike M, Le ML, Kolonel LN, Hirschhorn J, Altshuler D, Henderson BE, Freedman ML: Common genetic variation in IGF1 and prostate cancer risk in the Multiethnic Cohort. J Natl Cancer Inst 2006;98:123-134.

10 Gu F, Schumacher FR, Canzian F, Allen NE, et al. Eighteen Insulin-like Growth Factor Pathway Genes, Circulating Levels of IGF-I and Its Binding Protein, and Risk of Prostate and Breast Cancer. Cancer Epidemiology Biomarkers & Prevention 2010;19:2877-2887.

11 Hernandez W, Grenade C, Santos ER, Bonilla C, Ahaghotu C, Kittles RA: IGF-I and IGFBP-3 gene variants influence on serum levels and prostate cancer risk in African-Americans. Carcinogenesis 2007;28:2154-2159.

12 Wang LZ, Habuchi T, Tsuchiya N, et al. Insulin-like growth factor-binding protein-3 gene-202 A/C polymorphism is correlated with advanced disease status in prostate cancer. Cancer Res 2003;63:4407-4411.

13 Al-Zahrani A, Sandhu MS, Luben RN, et al. IGF1 and IGFBP3 tagging polymorphisms are associated with circulating levels of IGF1, IGFBP3 and risk of breast cancer. Hum Mol Genet 2006;15:1-10.

14 Terry KL, Tworoger SS, Gates MA, Cramer DW, Hankinson SE: Common genetic variation in IGF1, IGFBP1 and IGFBP3 and ovarian cancer risk. Carcinogenesis 2009;30:2042-2046.

15 Kaplan RC, Petersen AK, Chen MH, et al. A genome-wide association study identifies novel loci associated with circulating IGF-I and IGFBP-3. Hum Mol Genet 2011.

Date proposal received: 
Thursday, 22 November, 2012
Date proposal approved: 
Thursday, 22 November, 2012
Keywords: 
GWAS, IGF, Mendelian, Mendelian Randomisation
Primary keyword: 

B1467 - Does TRPA1 modify the effects of prenatal paracetamol and tobacco smoke exposure on childhood respiratory outcomes - 22/11/2012

B number: 
B1467
Principal applicant name: 
Prof Seif Shaheen (Barts & The London School of Medicing & Dentistry, UK)
Co-applicants: 
Prof John Henderson (University of Bristol, UK), Prof George Davey Smith (University of Bristol, UK), Prof John Holloway (University of Southampton, UK), Dr Susan Ring (University of Bristol, UK), Dr Dave Evans (University of Bristol, UK)
Title of project: 
Does TRPA1 modify the effects of prenatal paracetamol and tobacco smoke exposure on childhood respiratory outcomes?
Proposal summary: 

Background:

We have previously reported an association between prenatal paracetamol exposure and asthma in ALSPAC and shown that this relation is modified by maternal antioxidant gene polymorphisms [1,2]. It was recently reported that the analgesic properties of paracetamol may be mediated by the transient receptor potential ankyrin-1 (TRPA1) channel [3]. Furthermore, animal studies have suggested that TRPA1 plays a role in airway inflammation in asthma [4] and, of relevance to our epidemiological observations, there is evidence that paracetamol can cause neurogenic inflammation in the airways through stimulation of TRPA1 [5]. Cigarette smoke induced neurogenic inflammation is also thought to be mediated by TRPA1 [6].

Aims:

To analyse the relation between maternal and child TRPA1 variants and asthma and lung function in ALSPAC and to explore interactions between TRPA1 and prenatal paracetamol and tobacco smoke exposure on childhood asthma and lung function in order to potentially strengthen causal inference and shed light on mechanisms.

Hypotheses:

1) Maternal and child TRPA1 genotype is associated with asthma and lung function in the offspring.

2) TRPA1 modifies the effects of prenatal paracetamol exposure on childhood asthma and of maternal smoking in pregnancy on childhood small airway function.

Analyses:

It has been proposed that TRPA1 alleles in ALSPAC could be inferred using SNP imputation; a list of 129 imputed SNPs has been generated (all SNPs except one which have been mentioned in the literature associated with pain phenotypes are included in this list) and the plan is to work with Dave Evans to see how best to analyse the data.

A DTA covering analysis of genetic data is already in place with QMUL.

NB: We have all other variables (maternal exposures, confounders and childhood atopic and respiratory outcomes) needed to carry out these analyses.

References:

1) Shaheen SO, Newson RB, Henderson AJ, Headley JE, Stratton FD, Jones RW, Strachan DP, and the ALSPAC Study Team. Prenatal paracetamol exposure and risk of asthma and elevated IgE in childhood. Clin Exp Allergy 2005; 35:18-25.

2) Shaheen SO, Newson RB, Rose-Zerilli M, Ring SM, Holloway JW, Henderson AJ. Prenatal and infant acetaminophen exposure, antioxidant gene polymorphisms and childhood asthma. J Allergy Clin Immunol 2010; 126: 1141-1148.

3) Andersson DA et al. TRPA1 mediates spinal antinociception induced by acetaminophen and the cannabinoid delta9-tetrahydrocannabiorcol. Nature Communications 2011; 2:551.

4) Caceres AI et al. A sensory neuronal ion channel essential for airway inflammation and hyperreactivity in asthma. PNAS 2009; 106: 9099-9104.

5) Nassini R et al. Acetaminophen, via its reactive metabolite N-acetyl-p-benzo-quinoneimine and transient receptor potential ankyrin-1 stimulation, causes neurogenic inflammation in the airways and other tissues in rodents. FASEB J 2010; 24:4904-16.

6) Andre E et al. Cigarette smoke-induced neurogenic inflammation is mediated by alpha, beta-unsaturated adehydes and the TRPA1 receptor in rodents. J Clin Invest 2008; 118: 2574-82.

Date proposal received: 
Thursday, 22 November, 2012
Date proposal approved: 
Thursday, 22 November, 2012
Keywords: 
Asthma, Genetics, Smoking, Drugs
Primary keyword: 

B1466 - Exploring the development of hallucinations paranoia and first rank delusions using latent transition models - 22/11/2012

B number: 
B1466
Principal applicant name: 
Dr Fraenze Kibowski (University of Ulster, Northern Ireland)
Co-applicants: 
Prof Mark Shevlin (University of Ulster, Northern Ireland), Dr Jamie Murphy (University of Ulster, Northern Ireland)
Title of project: 
Exploring the development of hallucinations, paranoia, and 'first rank' delusions using latent transition models
Proposal summary: 

A psychotic-like experience in itself does not confer a specific enough risk in regards to clinically relevant psychosis (4). However, it has been shown that the co-occurrence of hallucinations and delusions increases the risk of subsequent clinical outcomes. The temporal ordering of the occurrence and co-occurrence of specific psychotic-like symptoms has not been examined. This project also aims to establish the association between particular patterns of occurrence and co-occurrence with established risk factors such as a family history of mental illness, substance use, and childhood victimisation. There are three interconnected parts to the analysis which correspond to intended paper publications.

Demographics of developmental trajectories of psychosis

(1) Do homogeneous patterns of psychotic-like symptoms exist for responses at 11.6, 13, 14, and 16.5 years via the PLIKS questionnaire?

(2) Are parental histories of mental illness and substance use disorder, adverse events during pregnancy, and socio-economic variables able to predict 'stayers' and 'movers' in the latent transition model?

Trauma-based antecedents of the developmental trajectories of psychosis

While controlling for previously identified demographic confounders:

(1) When adjusting for IQ and early childhood psychotic disorders, do childhood adversities (such as bullying, death of relative or friend, and emotional cruelty) predict transition?

(2) When adjusting for IQ, early childhood psychotic disorders and childhood adversities, do parental childhood adversities (up to the age of 17) predict transition?

(3) Are there significant associations with any of the transitions when entering time-varying exposure variables into the model, such as social skills, substance use, schizotypy, attachment and moods?

Outcomes and risk profiles for the developmental trajectories of psychosis

(1) What sort of outcomes are associated with specific transitions?

References

1. Bebbington P, Jonas S, Kuipers E, King M, Cooper C, Brugha T, et al. Childhood sexual abuse and psychosis: data from a cross-sectional national psychiatric survey in England. The British journal of psychiatry?: the journal of mental science [Internet]. 2011 Jul [cited 2011 Oct 26];199:29-37. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21508437

2. Johns LC, Cannon M, Singleton N, Murray RM, Farrell M, Brugha T, et al. Prevalence and correlates of self-reported psychotic symptoms in the British population. The British journal of psychiatry?: the journal of mental science [Internet]. 2004 Oct [cited 2011 Nov 17];185:298-305. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15458989

3. Bendall S, Jackson HJ, Hulbert C a, McGorry PD. Childhood trauma and psychotic disorders: a systematic, critical review of the evidence. Schizophrenia bulletin [Internet]. 2008 May [cited 2011 Aug 7];34(3):568-79. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2632421&tool=p...

4. Linscott RJ, van Os J. Systematic reviews of categorical versus continuum models in psychosis: evidence for discontinuous subpopulations underlying a psychometric continuum. Implications for DSM-V, DSM-VI, and DSM-VII. Annual review of clinical psychology [Internet]. 2010 Apr 27 [cited 2011 Aug 23];6:391-419. Available from: http://www.ncbi.nlm.nih.gov/pubmed/20192792

5. Wigman JTW, van Winkel R, Raaijmakers Q a W, Ormel J, Verhulst FC, Reijneveld S a, et al. Evidence for a persistent, environment-dependent and deteriorating subtype of subclinical psychotic experiences: a 6-year longitudinal general population study. Psychological medicine [Internet]. 2011 Nov [cited 2011 Oct 27];41(11):2317-29. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21477418

Date proposal received: 
Thursday, 22 November, 2012
Date proposal approved: 
Thursday, 22 November, 2012
Keywords: 
Psychosis
Primary keyword: 

B1462 - The exploration of heritability of facial features Fathers and offspring - 08/11/2012

B number: 
B1462
Principal applicant name: 
Prof Stephen Richmond (University of Cardiff, UK)
Co-applicants: 
Title of project: 
The exploration of heritability of facial features: Fathers and offspring
Proposal summary: 

Aim: The exploration of heredibility of facial features: Fathers and offspring

Hypothesis: Fathers face shape has no influence on the face shape of his offspring

Twin, family and animal studies have suggested that inheritance plays a role in the determination of craniofacial morphology.1-4 Traditional two-dimensional measuring techniques on photographs or lateral skull radiographs tend to be imprecise as landmarks are subject to rotational, positional and magnification errors. However, recent advances in high resolution three-dimensional imaging technologies has provided the opportunity in detailing the spatial relationship of facial landmarks and investigating which genetic variants may be influencing these. However, the evidence for heritability of facial features is not robust and often poorly researched using small samples exhibiting weak associations between parental and offspring with mid father/mother estimates reported as a best predictor for offspring facial parameters.5,6 We will initially focus on facial parameters reported to be associated with known genes. 7,8

1. Kohn, LAP. The Role of Genetics in Craniofacial Morphology and Growth. Annual Review of Anthropology 20, 261-278(1991).

2. Saunders SR, Popovich F, Thompson GW, A family study of craniofacial dimensions in the Burlington Growth Centre sample, American Journal of Orthodontics, Volume 78, Issue 4, October 1980, Pages 394-403.

3. W. Stuart Hunter, Daniel R. Balbach, Donald E. Lamphiear, The heritability of attained growth in the human face, American Journal of Orthodontics, Volume 58, Issue 2, August 1970, Pages 128-134.

4. Johannsdottir B, Thorarinsson F, Thordarson A, Magnusson TE, Heritability of craniofacial characteristics between parents and offspring estimated from lateral cephalograms, American Journal of Orthodontics and Dentofacial Orthopedics, Volume 127, Issue 2, February 2005, Pages 200-207.

5. Nakata M, Yu P, Davis B, Nance WE, The use of genetic data in the prediction of craniofacial dimensions, American Journal of Orthodontics, Volume 63, Issue 5, May 1973, Pages 471-480

6. Nakasima A, Ichinose M, Nakata S, Takahama Y, Hereditary factors in the craniofacial morphology of Angle's Class II and Class III malocclusions, American Journal of Orthodontics, Volume 82, Issue 2, August 1982, Pages 150-156

7. Paternoster L, Zhurov AI, Toma AM, Kemp JP, St Pourcain B, Timpson NJ, McMahon G, McArdle W, Ring SM, Smith GD, Richmond S, Evans DM. Genome-wide association study of three-dimensional facial morphology identifies a variant in PAX3 associated with nasion position. Am J Hum Genet. 2012 Mar 9;90(3):478-85.

8. Liu F, van der Lijn F, Schurmann C, Zhu G, Chakravarty MM, Hysi PG, Wollstein A, Lao O, de Bruijne M, Ikram MA, van der Lugt A, Rivadeneira F, Uitterlinden AG, Hofman A, Niessen WJ, Homuth G, de Zubicaray G, McMahon KL, Thompson PM, Daboul A, Puls R, Hegenscheid K, Bevan L, Pausova Z, Medland SE, Montgomery GW, Wright MJ, Wicking C, Boehringer S, Spector TD, Paus T, Martin NG, Biffar R, Kayser M. A genome-wide association study identifies five Loci influencing facial morphology in europeans. PLoS Genet. 2012 Sep;8(9):e1002932. doi:10.1371/journal.pgen.1002932.

Date proposal received: 
Thursday, 8 November, 2012
Date proposal approved: 
Thursday, 8 November, 2012
Keywords: 
Face Shape
Primary keyword: 

B1461 - Association study of ANTXR2 capillary morphogenesis gene and TNFR1 with ankylosing spondylitis - 08/11/2012

B number: 
B1461
Principal applicant name: 
Prof Bryan Paul Wordsworth (University of Oxford, UK)
Co-applicants: 
Ms Tugce Karaderi (University of Oxford, UK), Dr Dave Evans (University of Bristol, UK)
Title of project: 
Association study of ANTXR2 (capillary morphogenesis gene) and TNFR1 with ankylosing spondylitis.
Proposal summary: 

Aims - To replicate and refine previously reported genetic associations of the genes ANTXR2 (otherwise known as CMG2 - capillary morphogenesis factor) and TNFRSF1A (encoding the type 1 TNF receptor) with ankylosing spondylitis (AS).

Hypothesis

1) Variants of ANTXR2 have previously been associated with AS in 2 previous genome-wide association studies (GWAS) (Australo-Anglo-American Spondyloarthritis Consortium - TASC 2010, rs4333130, p=9.3 x 10^-8; Wellcome Trust Case Control Consortium - WTCCC2-TASC meta-analysis, rs4389526, p=9.4 x 10^-8). However, the replication set in the latter study only examined the single top SNP and did not conduct a systematic study of ANTXR2 variants. The discovery set in this study consisted of ~2,000 caucasian AS cases and the majority of significant "hits" were followed up in a large number of cases both of caucasian and other ethnicities using the customised "Immunochip" targeting 200k SNPs of largely immunologic or inflammatory function. Unfortunatley, the Immunochip contained no SNPs covering the ANTXR2 locus. We therefore hypothesise that there should be detectable associations at this locus in an independent set of patients and controls. We have therefore already formally genotyped 9 tagging SNPs across the ANTXR2 gene in a further 3,000 UK cases with AS and now wish to confirm association in an independent set of controls - the ALSPAC cohort. From these data, we shall also be able to impute associations with rare alleles and to refine the likely primary associations of AS with variants at ANTXR2.

2) Variants in LTBR-TNFRSF1A region have already been associated with AS in GWAS (TASC 2010, rs1800693, p=6.9 x 10^-5, WTCCC2-TASC meta-analysis, rs11616188, p=4.1 x 10^-12). Furthermore, variants in TNFRSF1A have also been associated with multiple sclerosis (MS) (Gregory et al., 2012). However, the variant rs1800693 has opposite effects in AS and MS. The G allele of this SNP leads to a splice variant that appears to have an anti-TNF effect. This allele (G) of rs1800693 is therefore, unsurprisingly, protective in AS, whereas it is predisposing in MS (a condition that can be provoked by anti-TNF therapy). It is important to confirm the association of this SNP with AS and analyse its effects further, particularly with respect to responses of AS to anti-TNF therapy and effects on disease severity.

Exposure variable

We have already typed about greater than 5000 cases for SNP across ANTXR2 and have undertaken scrupulous quality checks on the data. We will compare the allele and genotype frequencies in this case dataset to those in the 8365 controls obtained from the ALSPAC cohort. Genotypes of SNPs in regions containing genes ANTXR2 and TNFRSF1A will be used to do statistical tests to detect/confirm possible associations with AS. The precise regions for which data are required for us to be able to undertake this analysis are listed below:

ANTXR2 region (chr4) - 80,000,000 - 88,700,000 (Genome build 37)

LTBR-TNFRSF1A region (chr12) - 6,000,000 - 7,000,000 (Genome build 37)

Date proposal received: 
Thursday, 8 November, 2012
Date proposal approved: 
Thursday, 8 November, 2012
Keywords: 
Bone, Genetics, Bones
Primary keyword: 

B1459 - Replication of genetic variants 11 and genes 2 influencing relational processing reasoning working memory and IQ - 08/11/2012

B number: 
B1459
Principal applicant name: 
Dr Margaret Wright (Queensland Institute of Medical Research, ROW)
Co-applicants: 
Prof Nick Martin (Queensland Institute of Medical Research, ROW)
Title of project: 
Replication of genetic variants (11) and genes (2) influencing relational processing, reasoning, working memory and IQ
Proposal summary: 

BACKGROUND: Our primary measure of interest is relational complexity, a measure that encapsulates both an individuals's processing capacity and task complexity and is proposed to account for capacity limits found in both reasoning and working memory. At a variance components level, we show that a genetic factor influencing relational complexity accounts for most of the genetic covariation between measures of reasoning and working memory, and further, that there is a strong overlap with IQ. A criterion of our genetic variants of interest was that they must influence variation in all measures (i.e. relational complexity, IQ, reasoning, and working memory), while our genes of interest were the top results for relational complexity and for IQ.

AIM: To assess our most promising genetic variants and genes in independent samples. Two independent groups (the LBC1936 Scottish sample and the NCNG Norwegian sample) have shown some consistency with our findings, but results from a third group (the NTR Netherlands sample) were inconsistent.

HYPOTHESIS: Replicating genetic association and gene-based analyses in multiple independent samples increases the likelihood of avoiding Type 1 (and Type II) error and thereby increases the value of our manuscript.

INDEPENDENT VARIABLES: genotypes for 99 single nucleotide polymorphisms (SNPs) - 11 SNPs are specific genetic variants of interest and the remaining 88 SNPs represent the two genes of interest (list of 99 rs numbers to be provided if application is successful). Note that our genotyping was done using the Illumina HumanHap 610 quad chip (versus HumanHap 550 used by ALSPAC) and it is possible we may need to supply proxies for some SNPs.

DEPENDENT VARIABLES: Matrix Reasoning, WASI IQ (measures available for the other samples: LBC1936 and NCNG both had measures of Matrix Reasoning and IQ, while NTR had a measure of Raven's Progressive Matrices)

(possible) CONFOUNDING VARIABLES: Sex, Age, and Multidimensional Scaling (MDS) components as appropriate to the ALSPAC sample to adjust for population stratification

ANALYSES: Genetic association for 99 SNPs for the measures Matrix Reasoning and WASI IQ, with sex, age, and MDS components included as covariates. (NOTE: for a subset of these SNPs (i.e. 88 of 99) the association results will be used to run a gene-based analysis for 2 genes - this analysis to be conducted at QIMR)

Date proposal received: 
Thursday, 8 November, 2012
Date proposal approved: 
Thursday, 8 November, 2012
Keywords: 
Genetics, Memory
Primary keyword: 

B1453 - Investigating the prenatal origins of ADHD - 26/10/2012

B number: 
B1453
Principal applicant name: 
Prof Anita Thapar (University of Cardiff, UK)
Co-applicants: 
Dr Stephan Collishaw (University of Cardiff, UK), Dr Kate Langley (University of Cardiff, UK), Dr Ajay Thapar (University of Cardiff, UK), Prof Peter Holmans (University of Cardiff, UK), Prof Michael O'Donovan (University of Cardiff, UK), Prof Michael Owen (University of Cardiff, UK), Dr Caroline Relton (University of Bristol, UK), Prof George Davey Smith (University of Bristol, UK), Dr Kate Northstone (University of Bristol, UK), Dr Susan Ring (University of Bristol, UK), Dr Jonathan Mill (King's College London, UK)
Title of project: 
Investigating the prenatal origins of ADHD.
Proposal summary: 

Aims:

1. Test for associations between prenatal exposures to adversity (toxins and diet) and ADHD (age 7/8) traits.

2. Test for associations between germline genetic variation (identified in large scale case control genetic discovery studies of ADHD diagnosis) and ADHD (age 7/8 traits).

3. Identify epigenetic signatures at birth associated with ADHD.

4. Test if epigenetic signatures index/mediate link between prenatal exposures, genes and ADHD.

5. Use epigenetic signatures to form hypotheses about relevant environmental exposures for ADHD.

6. Identify those at high ADHD risk (those at high genetic/prenatal environmental/epigenetic risk at birth).

7. Identify preschool enrichment factors (diet, care) that modify early ADHD risk.

Hypotheses

1. Genetic risks and prenatal exposure to environmental hazards that are indexed by epigenetic signatures alter brain development and increase ADHD risk (at age 7) .

2.Epigenetic and genomic signatures provide clues on biological and environmental risks that might be future targets for early intervention.

3. Birth risks (captured by germline genetic risks, prenatal environment and epigenetic profile) on ADHD are modifiable by postnatal enrichment.

Exposures

1.Foetal exposure to lead, mercury, other toxins (atrazine available, PCBs to be assayed), maternal cigarette smoking (reported and cotinine levels), alcohol

2.Maternal diet and dietary supplementation

3. Child germline genetic variation-composite of common and rare variants known to be associated with ADHD (via GWAS then exome sequencing).

4. Epigenetic array data from ARIES at birth -cord blood (additional epigenetic micrarray data to be obtained on ADHD cases in ALSPAC who are not in ARIES-this will be costed in)

Outcomes

1. Trait ADHD aged 7/8 asssesed by DAWBA

Modifiers

1. Child diet

2. Early maternal care

Confounders

1. Sex of child

2. Social class

3. IQ

4. Comorbid conduct/mood problems.

Date proposal received: 
Thursday, 11 October, 2012
Date proposal approved: 
Friday, 26 October, 2012
Keywords: 
ADHD, Epigenetics
Primary keyword: 

B1458 - RNA-Seq Deep Sequencing and In Vivo Chromatin Studies Identifying Functional Elements Relevant to Reading and Language - 25/10/2012

B number: 
B1458
Principal applicant name: 
Prof Jeffrey Gruen (Yale University, USA)
Co-applicants: 
Dr Natalie Powers (Yale University, USA), Dr John Eicher (Yale University, USA)
Title of project: 
RNA-Seq, Deep Sequencing, and In Vivo Chromatin Studies Identifying Functional Elements Relevant to Reading and Language.
Proposal summary: 

Aims and Hypotheses

Learning disabilities are disorders characterized by unexpected difficulty with a specific mode of learning, generally with normal IQ and educational opportunity. The most common learning disabilities involve language; the NICHD estimates that 15-20% of Americans have a language-based learning disability. This high prevalence makes academic remediation of these disorders a costly burden to the educational system. The most common learning disabilities by far are dyslexia and language impairment (LI), which are specific deficits in processing and expressing written and spoken language, respectively. Both disorders are highly heritable, and genetic studies over the past three decades have identified a number of risk loci and genes. Because genetic methods have been used almost exclusively, however, nothing is known about how most risk variants exert their effects. Additionally, these risk variants account for little of the known heritability of these disorders; much of it is still 'missing,' which precludes development of a gene-based diagnosis. An effective gene-based diagnostic tool would be useful for early detection of affected individuals, as interventions are far more effective if administered earlier in life. In our current funding period, we reported compelling evidence that BV677278, a polymorphic tandem repeat within an intron of the dyslexia risk gene DCDC2, is a regulatory element that substantially influences reading and verbal language skills. We showed that this element can modulate expression from the DCDC2 promoter, that it binds specifically to the potent transcription factor ETV6, that at least two of its alleles significantly reduce mean reading or language performance, and that these alleles show a synergistic genetic interaction with a known risk allele of KIAA0319 (another known dyslexia risk gene in the same locus, DYX2). We also observed associations between other DYX2 regions, such as C6orf62 and THEM2, indicating that other DYX2 elements contribute to reading and language along with the risk variants in DCDC2 and KIAA0319. Based on these results, we hypothesize that BV677278 directly regulates KIAA0319 and other genes via a regulatory complex containing ETV6, and that the deleterious effects of specific BV677278 alleles are due to differences in target gene expression. We also hypothesize that BV677278 and KIAA0319 are not the only functional variants in the DYX2 locus that influence reading and language. To test these hypotheses, we will address the following specific aims:

Specific Aim 1) Examine the effect of BV677278 on gene expression. Because a BV677278 deletion exists naturally in humans, we can examine the effect of having two, one, or zero copies of BV677278 on gene expression, as well as the effects of different BV677278 genotypes. To this end, we will correlate extant expression microarray data from ~1,000 lymphoblastoid cell lines from ALSPAC subjects with various BV677278 genotypes. Allele frequency of the deletion suggests that there will be at least 7 BV677278-null cell lines. We will also perform RNA-seq on ~100 of these cell lines selected for genotypes of interest, including all BV677278-nulls.

Specific Aim 2) Identify putative regulatory targets that interact with the ETV6-BV677278 complex. We will use chromatin interaction analysis with paired end tagging (ChIA-PET) to find putative regulatory targets that physically interact with the ETV6-BV677278 complex. ChIA-PET allows for an unbiased scan of the genome for physical interactions with this complex, and positive results can be confirmed by chromatin conformation capture (3C). 3C will be used as a backup if ChIA-PET fails, though it will require prior identification of candidate target sequences, informed by our expression study in Aim 1. These experiments will be performed in 20 ALSPAC lymphoblastoid cell lines selected for two, one, and zero BV677278 copies.

Specific Aim 3) Identify additional DYX2 variants independent of the BV677278 regulatory element and the KIAA0319 risk haplotype. Our data suggest that, although BV677278 and the KIAA0319 risk haplotype are important sources of the DYX2 linkage and association with dyslexia, there are other DYX2 variants that contribute risk. To identify these variants and to elucidate their functions, we will exploit the single-base resolution provided by next-generation sequencing, and deep sequence the entire DYX2 locus in 1,000 ALSPAC subjects. We will select these subjects using an extreme phenotypes approach: 250 each of the worst performers on reading and language tasks, respectively, and 250 each of the best performers on those same tasks, respectively. Risk variants will accumulate in specific regions in severely affected individuals and will be assessed for functional implications in gene expression, protein function, and histone modifications.

The overall goal of this proposal is to explore the molecular mechanisms by which genetic variants in the DYX2 locus influence written and verbal language skills and impart heritable risk to dyslexia and LI. Our proposed experiments will elucidate the mechanism of action of BV677278, characterize its synergistic interaction with KIAA0319, identify other regulatory and gene targets of BV677278 that may not have been captured by genetic approaches, and detect other variants in the DYX2 locus that influence reading and language. By expanding our understanding of the biological basis of complex reading and language processes, we hope to open future options for diagnosis and treatment of language-based learning disabilities.

Exposure Variables

RNA, DNA and lymphoblastoid cell lines from ALSPAC subjects will be chosen on the basis of their BV677278 genotype, expression data from the microarray analyses of ALSPAC cell lines, performance on verbal language and reading tasks, IQ, and ancestry.

BV677278 is a hypermutable compound short tandem repeat with more than 30 alleles. A 2,445bp deletion, encompassing BV677278 in its entirety, also exists naturally in humans-individuals heterozygous for this deletion are hemizygous (have only one copy) for BV677278, while individuals homozygous for the deletion are completely BV677278-null (no copies of BV677278 in their genome). Our analyses of the ALSPAC thus far have shown that BV677278 allele 5 is associated with dyslexia and lowered reading skills and that BV677278 allele 6 is associated with language impairment and lowered language skills. Not only did these alleles show association with their respective phenotypes, their effects were strong enough to significantly reduce mean performance on reading tasks in the case of allele 5, and language tasks in the case of allele 6, in carriers vs. non-carriers. Additionally, we found that BV677278 interacts synergistically with a known risk variant in the other dyslexia gene in DYX2, KIAA0319, to adversely affect several reading, language, and cognitive phenotypes [Powers et al., submitted]. We also showed that BV677278 specifically binds a nuclear protein and is capable of modulating expression from the DCDC2 promoter [Meng et al., 2011]. We identified this protein that binds to BV677278 as the potent transcription factor and proto-oncogene ETV6 [Powers et al., submitted]. In our aims, we propose to take advantage of the BV677278 microdeletion and the various BV677278 genotypes by comparing RNA-sequencing, deep sequencing of the DYX2 locus, and chromatin studies in lymphoblastoid cell lines with two, one, or no copies (control) of BV677278, and with various alleles that we have so far found to be deleterious to language and reading (alleles 5 and 6).

Outcome Variables

For Aim 1, the outcome of RNA-sequencing will show quantitative expression data (number of cDNA transcripts) from subjects with different BV677278 genotypes. We will be able to compare expression from subjects with two, one, or no copies of BV677278, as well as with BV677278 genotypes of interest, especially alleles 5 and 6. By sequencing 100 selected ALSPAC subjects, we will be able to adjust for intra-subject and inter-subject variation. We will also be able to compare RNA-sequence results with the extant ALSPAC microarray data. We intend to perform RNA-sequence at a high resolution (approximately two samples per flow-cell lane on the Illumina Hi-Seq platform), which should enable us to detect differences in splice variants as well.

For Aim 2, the outcome of the CHiA-PET chromatin studies will be next-generation sequence data of fusion fragments. If successful, one half of the fragment should contain sequence from BV677278, and the other half from putative target genes (under BV677278 transcriptional control) located elsewhere in the DYX2 locus (cis) and beyond (trans). Bioinformatics analysis will be able to differentiate cis from trans elements. Within the DYX2 locus we will be especially looking for fusion fragments containing sequence from the KIAA0319 risk haplotype or promoter region, as well as other possible regulatory elements in the region. Outside DYX2, we will use existing annotation from publically available resources such as JASPAC, ENCODE, and ENSEMBL and in vitro transcriptional reporter assays to inform us whether we have hit transcriptional control elements and target genes that may be relevant to language and reading.

For Aim 3, the outcome of deep sequencing of the 1.5MB DYX2 locus will be variants, known and unknown, distributed throughout the locus. Here we will be looking for clusters of variants in putative regulatory regions such as the DCDC2 promoter, the KIAA0319 promoter, and promoters from other genes that appear to influence reading and language. Analysis for this outcome will be somewhat empirical in terms of setting cluster boundaries and significance, and will depend on the frequency of variants that we see, and whether hits are in areas of LD with SNPs previously shown to be associated with reading, language, or cognition, or that have putative functional roles or show evolutionary conservation.

Confounding Variables

A significant confounder for these experiments will be the heterogeneous genetic background of the subjects, and consequently the lymphoblastoid cell lines, we will use. We will try to minimize the effects of genetic background by restricting studies to subjects of European ancestry, by comparing outcomes from subjects that have the same BV677278 genotypes, and by utilizing population structure metrics already calculated in genome-wide association studies completed at ALSPAC.

There are inherent biases in the techniques we have selected to use in our studies. Therefore,we intend to confirm all novel variants identified by next-generation sequencing results with Sanger sequencing, and all putative physical interactions identified via CHiA-PETwith chromatin conformation capture (3C). We will also use luciferase-reporter experiments to confirm function of novel putative regulatory elements identified by sequencing or the chromatin studies

Materials Requested and Selection Criteria:

1) RNA: We request 5 micrograms (concentration ~50 nanogram/microliter) of RNA from 100 selected subjects for RNA-sequencing. Subject selection will be informed by the results of our genotyping of the BV677278 element and the analysis of the microarray expression data (pending). The goal in subject selection is to identify subjects with BV677278 genotypes of interest, specifically alleles previously related to reading and language performance and the presence/absence of the BV677278 element (del/del and del/+), and to correlate their relationship with global gene expression.

2) Lymphoblastoid Cell Lines: We request 20 lymphoblastoid cell lines from selected subjects for in vivo chromatin studies. Subject selection will be informed by BV677278 genotype, particularly the presence/absence of the BV677278 element and risk alleles (e.g. alleles 5 and 6).

3) DNA: Depending on availability, we request 1-3 micrograms (concentration ~100 nanogram/microliter) of cell line-derived DNA from 1,000 selected subjects for deep sequencing of the 1.5MB of the DYX2 locus. Subject selection will be contingent on past performance on selected language and reading tasks in the ALSPAC at ages 7, 8 and 9 years. We will select subjects using an extreme phenotypes approach: 250 each of the worst performers on reading and language tasks, respectively, and 250 each of the best performers on those same tasks, respectively.

Performance Sites

All non-sequencing experiments will be performed in Dr. Gruen's lab at the Yale Child Health Research Center. This includes all the chromatin studies (CHiA-PET and 3C) and any cell culture that will be required, DNA extraction, and PCR.

All sequencing, next-generation and conventional Sanger sequencing, sequence alignment and annotation, will be performed at the Yale Center for Genome Analysis and The W.M. Keck Foundation Biotechnology Resource Laboratory at Yale. Analysis of aligned and annotated sequence results will be performed in Dr. Gruen's lab at the Yale Child Health Research Center.

Security and Storage of Information and Materials

ALSPAC phenotype, genotype, and sequence information are stored on Yale networked/secured desktop computers only. No ALSPAC data are downloaded to computers that are not connected to the Yale network. No ALSPAC data are downloaded onto thumb drives, peripheral drives, or laptop computers that are not directly connected to the Yale network. No ALSPAC data may be shared with collaborators other than those that have been approved by ALSPAC. In addition, there is no identifying information with phenotype, genotype or sequence information that could link them with a specific ALSPAC subject.

RNA and DNA are stored in -70degree freezers in Dr. Gruen's lab in the Yale Child Health Research Center (464 Congress Avenue). Doors to the lab are locked and accessible by key entry only. Access to the Yale Child Health Research Center is restricted to Yale employees, students, and post-docs who work in the Center. In addition there is no identifying information recorded on the RNA/DNA tubes that could link the material with a specific ALSPAC subject.

Lymphoblastoid cell lines (LCL's) are stored in liquid nitrogen freezers in the Yale Child Health Research Center. Active cultures are stored in CO2 incubators in dedicated facilities in the Center. Access to the Yale Child Health Research Center is restricted to Yale employees, students, and post-docs who work in the Center. In addition there is no identifying information recorded on the cell culture flasks or storage vials that could link the material with a specific ALSPAC subject.

Data Sharing

Per ALSPAC protocol, all data generated in the course of this research will be returned to the ALSPAC within 12 months of its generation.

Ethical Considerations

The main ethical concerns of this proposed research are 1) protection of subject privacy, 2) security and storage of information and materials, 3) serendipitous identification of potentially important health information, and 4) subject withdrawal from the study.

1) Protection of Subject Privacy

Per ALSPAC protocol, all materials and information from ALSPAC is stripped of any identifiers that could possibly link them with a specific subject prior to being sent to collaborators. There is a theoretical possibility that whole genome sequencing or whole exome sequencing could identify an exceedingly rare phenotype that could be traced to a specific subject in the ALSPAC. However, we will not be performing either whole genome or whole exome sequencing on any subject DNA in this proposal. Furthermore, there are no known syndromes, rare or common, that could become known to us by deep sequencing the DYX2 locus. Therefore the risk of violating subject privacy through this research is minimal.

2) Security and Storage of Information and Materials

All information and data obtained during the proposed studies will be stored on Yale-maintained servers with security maintained by Yale University networks. Please see details in the above security section.

3) Serendipitous Identification of Potentially Important Health Information

In the course of deep sequencing the 1.5 megabases (MB) of the DYX2 dyslexia locus on 6p22, it is theoretically possible that we could identify a translocation that could suggest risk of malignancy. The 1.5MB we propose to sequence has been well defined in terms of coding regions and genes. It does not contain any oncogenes or proto-oncogenes or sequences previously described as causing malignancies. However, a recent report by Longoni et al. (2012) suggests that DCDC2 could be a novel oncogenic target of the ETS transcription factor ESE3/EHF. In this study the authors found that DCDC2 was aberrantly expressed in 53 malignant prostate tumors, but absent in 10 normal control prostates. They conclude that the ETS transcription factor ESE3/EHF, which is expressed in normal prostate and frequently lost in prostate tumors, maintained DCDC2 repressed by binding to a novel identified ETS binding site in the DCDC2 gene promoter. The authors do not report that genomic rearrangements, mutations, or RNA splice variants involving DCDC2 were the cause of prostatic malignancies or contributed to drug resistance, but a translocation could theoretically remove DCDC2 repression by separating DCDC2 from its regulatory element.

The risk of serendipitously finding a genomic rearrangement or mutation in the DYX2 locus that could increase the risk of malignancy is extremely small. However, should we find any genomic rearrangement, mutation, or splice variant, in the course of deep sequencing DYX2 that could suggest even a small increased risk of malignancy, we will report it to the ALSPAC within 6 months of identifying it. It will then be ALSPAC's responsibility to decide whether to share the information with a subject or for any further action.

4) Subject Withdrawal from the Study

In the event that we are notified by the ALSPAC that a donor-subject has withdrawn from the study, we will destroy any biomaterials we have received, including DNA, RNA, or lymphoblastoid cell lines. Since all information, genotypes, and variables are de-identified we don't expect that these would need to be deleted. However, we will delete any data if directed to do so by the ALSPAC. The Yale Human Investigation Committee will oversee this procedure.

Oversight of Protection of Human Subjects

All human subject research at Yale is under the oversight of the Yale Human Investigation Committee (HIC). Protocols describing protection of human subjects in detail must be reviewed and approved by the Yale HIC prior to applying for any funding, federal or private, or submission of results for publication. All prior research with the ALSPAC, including this proposal, has been submitted for review by the Yale HIC.

References:

Cho K, Frijters JC, Zhang H, Miller LL, Gruen JR. Prenatal exposure to nicotine and impaired reading performance. The Journal of Pediatrics, in press.

Eicher JD, Powers NR, Cho K, Miller LL, Mueller K, Ring SM, Tomblin JB, Gruen JR. Associations of prenatal nicotine exposure and the dopamine related genes ANKK1/DRD2 to verbal language, manuscript in preparation.

Longoni N, Kunderfranco P, Pellini S, Albino D, Mello-Grand M, Pinton S, D'Ambrosio G, Sarti M, Sessa F, Chiorino G, Catapano CV, Carbone, GM. (2012) Aberrant expression of the neuronal-specific protein DCDC2 promotes malignant phenotypes and is associated with prostate cancer progression. Oncogene doi: 10.1038/onc.2012.245 (epub ahead of print)

Meng H, Smith SD, Hager K, Held M, Liu J, Olson RK, Pennington B, DeFries JC, Gelernter J, O'Reilly-Pol T, Somlo S, Skudlarski P, Shaywitz SE, Shaywitz BA, Marchione K, Wang Y, Paramasivam M, LoTurco JJ, Page GP, Gruen JR. DCDC2 is associated with reading disability and modulates neuronal development in the brain, Proc Natl Acad Sci USA, 102: 17053-17058, 2005. PMID 16278297

Meng H, Powers NR, Tang L, Cope NA, Zhang P-X, Fuleihan R, Gibson C, Page GP, Gruen JR. A dyslexia-associated variant in DCDC2 changes gene expression. Behavior Genetics 2011 Jan;41(1):58-66.

Powers NR, Eicher JD, Butter F, Kong Y, Miller LL, Ring SM, Mann M, Gruen JR. Alleles of a Rapidly-Evolving ETV6 Binding Site in DCDC2 Confer Risk of Reading and Language Impairment, submitted.

Date proposal received: 
Thursday, 25 October, 2012
Date proposal approved: 
Thursday, 25 October, 2012
Keywords: 
Genetics, Speech and Language, Speech & Language
Primary keyword: 

B1457 - How do individual and area deprivation get under the skin place effects epigenetic changes in respiratory health - 25/10/2012

B number: 
B1457
Principal applicant name: 
Dr Bruna Galobardes (University of Bristol, UK)
Co-applicants: 
Prof George Davey Smith (University of Bristol, UK), Dr Caroline Relton (University of Bristol, UK), Prof Kate Tilling (University of Bristol, UK), Prof John Henderson (University of Bristol, UK)
Title of project: 
How do individual and area deprivation get under the skin: "place" effects & epigenetic changes in respiratory health.
Proposal summary: 

OBJECTIVES

The objectives of this proposal are:

1) Determine when an independent (of individual socioeconomic characteristics) association between early life area deprivation and lung function, doctor diagnosis of asthma and atopy phenotypes in children at ages 7-9 and 15-17 years emerges.

2) Determine specific area deprivation domains and the individual level life course exposures that mediate an association found in objective 1.

3) Determine whether there is a higher or differential DNA methylation in children of low compared to high parental SEP, and in children living in the worse compared to the least deprived areas (independent of individual level SEP) at 3 points in time (birth, 7, 15-17 years).

4) To investigate whether epigenetic changes found in objective 3 could mediate the association between individual and area deprivation with respiratory-related health outcomes

METHODS

Study participants. ALSPAC is a prospective birth cohort that followed offspring of 14,541 pregnant women resident in the county of Avon with an expected date of delivery between 1st April 1991 and 31st December 1992, from the time of the pregnancy to age 15-17 for this project) (http://www.bristol.ac.uk/alspac/). The study catchment area has a population of 1 million and includes de city of Bristol (0.5 million) with a mixture of rural areas, inner city deprivation, affluent suburbs and moderate sized towns. The cohort of Alspac has been extensively phenotyped and there is a detailed characterization of risk factors and exposures through medical and other administrative records, questionnaires and clinic visits carried out from pregnancy: 4 maternal and 2 partner's questionnaires during pregnancy; 17 mother's, 15 partner's and 23 child-based questionnaires during childhood; with children starting to respond questionnaire form the age of 7y. Since the age of 7y all children were invited to biannual hand-on assessments. DNA has been collected at multiple time points from parents and children. We detail here the measures that are relevant to this project.

Assessment of respiratory health and allergies: The main outcome measures are: 1) Lung function obtained through spirometry at ages 8-9y and 15-17y (FVC, FEV1, FEF25-75); 2) asthma (doctor's diagnosis of asthma at ages 8y and 15y); 3) atopy (Skin Prick test at age 7y and 15y); and, 3) combined asthma and atopy phenotypes which classify children into No asthma and no atopy (reference group), asthma alone, asthma with atopy and atopy alone (see work leading to this project for the socioeconomic patterning at individual level of these phenotypes). Secondary outcomes will include: wheezing phenotypes from birth until age 7 years which classify children based on their patterns of wheezing over time, bronchial reactivity measured with methacoline challenge at age 8y and fractional exhaled nitric oxide (FeNO) concentrations to asses airway inflammation at age 15y.

Assessment of individual socioeconomic position (SEP) and area-level deprivation: These are the main exposures in this project and will be indexed with a variety of indicators aiming to capture all different aspects of this construct67,68. Individual-level SEP is based on education (maternal and paternal), occupational class (maternal and paternal), household income, housing tenure and car access measured during pregnancy, ages 7-8y and 18y. We will analyze specific indicators to evaluate distinct pathways and a composite SEP index, aimed at capturing all different aspects of this construct, obtained with factor analysis. Area-level deprivation measured during 1) Pregnancy: Townsend index of area deprivation derived form 1990 census data at ward level already available in Alspac data (includes indicators of unemployment, overcrowding, car access and home ownership); 2) Two occasions in childhood at ages 8 and 16y based on the Index of multiple deprivation (IMD) 2004 (mostly constructed with 2000 and 2001 data) and 2010 (mostly based with 2008 data) at the Lower layer Super Output Area which has a minimum population of 1000 and a mean population of 1500. The Health domain of the IMD will be excluded to avoid having a marker of health in the exposure variable.

Specific area deprivation domains. The role of specific area attributes will be assessed analysing separately the deprivation domains included in the 2004 and 2010 IMD: Living environment (includes 'indoors' living environment with quality of housing and the 'outdoors' living environment through measures of air quality and road traffic accidents); barriers to housing and services (overcrowding, homelessness, difficulty to access owner occupation; distance to GP, supermarkets, and other services); income domain (including the Income Deprivation Affecting Children Index (Department for Communities and Local Government, 2008), employment domain (considers people of working age who are involuntarily excluded from the world of work, either through unemployment. ill health or family circumstances) (indexed through training and education domain, employment domain), education skills and training domain (which includes two sub-domains: one relating to lack of attainment among children and young people and one relating to lack of qualifications in terms of skills and captures the educational disadvantage of an area); Crime domain (rate of recorded crime for four major crime themes - burglary, theft, criminal damage and violence).

Other area characteristics: Population density (small-area measure of population density, in quintiles); rurality using DEFRA categorisations: 1) urban, rural types (town and fringe, village or hamlet and dispersed); 2)Predominantly urban, predominantly rural, significant rural69; and, length of residence in an area.

Perceived neighbourhood characteristics obtained from maternal self-reported of neighbourhood characteristics will include perceived neighbourhood quality, perceived social problems (crime and disorder), opinion of neighbourhood (good/fairly good/not good/bad) measured during gestation, 2, 5 7, 10 and 17 years.

Assessment of potential confounding, mediating or effect modifying individual level factors: a unique characteristic of Alspac is the extensive information available on a wide range of potential mediating and confounding factors measured since pregnancy and repeatedly throughout the life course of the participating children and their parents. Confounders will include child's age and assay/batch number for epigenetic analysis. All other variables will be considered, a priori, factors that can mediate the association between deprivation and the health outcome. These will include: birth weight; life course exposure to tobacco smoke will be characterized with "in utero" exposure (self-report of maternal smoking during pregnancy), exposure to environmental tobacco smoke (ETS) during childhood through maternal report of child's ETS exposure and blood cotinine levels at age 8y and 15y, child's self-report of smoking initiation at 15y; height and body mass index trajectories throughout childhood; physical activity measured with uniaxial accelerometer at 11, 13, 15 & 17y follow-up; housing characteristics including mould, humidity and temperature in several rooms of the house through maternal and child self-report questionnaire; pet ownership and exposure to pests measured through maternal and child self-report at several points in childhood.

Asssessment of DNA epigenetic changes. DNA has been stored from mothers and children at multiple time points. This is being used by ARIES, a BBSRC-funded resource, to generate epigenomic information based on peripheral blood leucocyte DNA at multiple time points across the lifecourse (http://www.ariesepigenomics.org.uk/). ARIES generates large scale genome-wide DNA methylation analysis of 1000 mother-child pairs measured at pregnancy, birth, age 7y and 15-17y through a Human Illumina 450K array. Illumina 450K data is processed using Illumina's Genome Studio Software. In-assay controls for each stage of the Infinum process are included on the array and analysed for quality prior to data export. Summary statistics of array-wide beta and intensity value are checked to ensure the assay is performing as required. The betas (proportion methylated) for each probe location and probe intensities are then exported. In addition to the measures available through ARIES we seek funding in this project to measure methylation in xxx children in total. To maximize the statistical power to find a difference in the proportion of methylation we will sample xxx children living in the most deprived quartile area and xxx living in the least deprived area at each 3 points in time (see power calculations: pending linking with methylation data). These measures will be obtained following the same ARIES protocol described above.

Date proposal received: 
Thursday, 25 October, 2012
Date proposal approved: 
Thursday, 25 October, 2012
Keywords: 
Childhood Adversity, Epigenetics , Respiratory
Primary keyword: 

B1456 - Is television viewing associated with depression and antisocial behaviour in adolescents - 25/10/2012

B number: 
B1456
Principal applicant name: 
Dr Markand Patel (University of Bristol, UK)
Co-applicants: 
Prof Glyn Lewis (University of Bristol, UK), Dr Nicola Wiles (University of Bristol, UK), Prof Matt Hickman (University of Bristol, UK)
Title of project: 
Is television viewing associated with depression and antisocial behaviour in adolescents?
Proposal summary: 

Background

Television Exposure

A systematic review in 2006 reported that young people watched on average 1.8-2.8 hours of TV per day, however 28% watched more than 4 hours and these were more likely to remain in the high viewing category at older ages.(1) Despite the development of various media technologies over the past 10 years, television content remains at the forefront of total media exposure in young people. It is now accessible through live and scheduled broadcasting, 'On Demand', recorded time shifted programming, online content via computers, mobile phones or other portable devices. More recent statistics have shown that television content exposure accounts for a daily average of 4 hours and 39 minutes across all these platforms. (2)

In the UK there are an average of 2.4 televisions per household,(3) which has risen over the last 10 years, with over 96% of homes having access to digital television.(4) In 2002 58% of UK children aged between 4-9 years had a television set in their bedroom, which rose to 79% of those aged 10-15 years old.(5)

Effects of television

Studies have shown that television viewing in adolescence can provide information on safe health practices, encourage social connections and behaviours. However there are concerns about its effects on aggression, sexual behaviour, substance use, self-image, social behaviour, physical health, life satisfaction and academic attainment. It has been recognised that television viewing does not require any special physical or cognitive abilities and does not require coordination with other people, therefore has a low or non-existent entry barrier compared to other leisure activities. It provides entertainment which is seen as an immediate benefit and the immediate costs appear negligible or are not predicted at all. Many of the associated costs do not result immediately, rather they take longer to appear for example the effects on sleep, material aspirations and consequences of a lack of investment in social contacts, education or career.(6)

Much of the knowledge we obtain about the world is indirect, through fictional or true accounts and experiences of others on television rather than our own experiences.(7) It has been suggested through Cultivation theory that messages obtained through television exposure on a frequent basis, whether accurate or not, makes up an individual's knowledge and affects their perceptions and behaviours.(7-9) Children and adolescents are particularly vulnerable to these messages, which can comprise of violence, sexuality, body image distortion, objectification and adverse health messages including alcohol, tobacco and illicit substance use, and it is more difficult for them to discriminate between what they see and reality.(10-13) However, children and adolescents are also able to learn pro-social content from television viewing and then generalise that learning to real-life situation, to produce helping behaviour, friendliness, imagination, racial and ethnic tolerance and respect for elders.(14-16)

A high amount of television viewing also displaces time spent in important developmental activities such as reading, problem-solving, homework, hobbies and interactions with parents, siblings and friends, and shown to be associated with attachment problems, social ability issues and attention issues.(17) In adults, it is suggested that excessive television viewing promotes social isolation and therefore hinders the development of social support networks and coping abilities, affecting mental health of individuals.(18)

Effects on mental health and behaviour

One longitudinal study has shown an association between television exposure in adolescence and increased depressive symptoms in young adulthood.(19) Television was most closely linked to depression compared to other media content such as videocassettes, computer games or radio, however this study did not adjust for physical activity.(19) Depressive symptoms have been associated with increased television viewing in other studies, although data shows that physical activity can be unrelated to depressive symptoms,(20) whereas it has also shown to be a protective factor.(21) In children, one study has shown that a greater amount of television viewing was associated with psychological difficulties irrespective of physical activity or sedentary time.(22)

Adolescents with a television set in their bedroom were shown to have fewer family meals, poorer school performance and had higher screen time exposure.(23) This exposure is negatively associated with health-promoting behaviours such as life appreciation, health responsibility, social support and exercise behaviour(24) and a risk factor for school life dissatisfaction and anxiety.(21) Anxiety and symptoms of psychological trauma has also been seen in children with greater television exposure.(25) A greater percentage of nightmares and separation anxiety was experienced in children witnessing distressing content on television, with 38% of them experiencing an increase in distress symptoms.(26) Several studies have shown that an increase in television viewing and in particular before going to bed affected sleep times, disturbances and sleepiness in children,(10, 27-30) which may contribute to mental and cognitive dysfunction.(31)

It is proposed that unrealistic ideals of attractiveness are transmitted through the media resulting in body dissatisfaction; a study concluded that idealised commercials led to an immediate impact on negative mood and appearance comparison for both sexes, with an increased body dissatisfaction for girls.(32) These recurrent episodes of dissatisfaction may accumulate over time leading to a longer-term body image dissatisfaction.(33) Music videos in particular have a high content of visual and auditory messages and have shown to bring about body dissatisfaction.(12) A meta-analysis by Groesz showed that body image was more strongly negative for female adolescents after viewing slender ideals in the media.(34) Another study showed that those who watched television content about cosmetic surgery wanted to alter their own appearance using the similar methods more than those not exposed to this content.(35)

High levels of television viewing leads to crowding out of activities which have a positive effect on happiness, such as time spent with friends and collegues.(36) In adults, it has been associated higher levels of loneliness, hopelessness, shyness, feelings of failure and guilt, depression and eating disorders; in addition to lower levels of self-esteem, weight satisfaction, perceived attractiveness and life satisfaction.(30,37) Particular television content also appears to directly affect mental health, especially news content which individuals are exposed to frequently,(38) which have shown to intensify depressed moods.(39) Heavy television viewers tended to overestimate crime rates, show more anxiety,(40) have less trust in others,(41,42) overestimate the affluence of others,(43) have higher material aspirations(44) and rate their own relative income lower,(46) leading to a lower subjective well-being.(47)

In adults who are experiencing stress, mood management theory predicts television is used to ameliorate moods or block anxious thoughts.(39,48) Stress is associated with an increase in self-reported television viewing in and adults(48) and in adolescents as a coping strategy.(49) In children, stress has been associated with an increased amount of television viewing in children who usually watched a greater amount of television.(50)

Effects on physical health

Television viewing in children and adolescents has shown to be associated with poor fitness, being overweight, smoking and raised cholesterol in adulthood, thus having long-term adverse effects on health.(51) Those adolescents with a television set in their bedroom undertook less physical activity, and had poorer dietary habits including higher consumption of foods containing sugar on a daily basis while watching television or as a result of advertisements.(23,52) The act of sitting itself had been studied and has been shown to lead to changes in resting glucose levels, blood pressure and biomarkers of cardiovascular disease and cancer.(53)

Anti-social behaviour

It has been shown that media exposure is associated with increased violence and aggressive behaviour, more high-risk behaviours such as substance use and earlier onset of sexual activity.(54) In children as young as three years of age, television exposure has shown an increased tendency to exhibit aggressive behaviour.(25,55) A television violence study has shown that nearly two-thirds of all programmes contained violence, with children's shows containing the most.(56) A longitudinal study found that watching violent television content as a young child was also associated with antisocial behaviour several years later in boys 7-10 years old.(57) One RCT showed that an intervention to reduce television viewing decreased aggressive behaviour in school children, supporting the causal influences of television content on aggression.(58) In adolescents the time spent viewing television content increased the likelihood of subsequent aggressive acts against others.(59) Nearly a quarter of music videos portrayed violence and weapon carrying,(60) and higher alcohol consumption and violent behaviour has been observed in teens from the effect of music videos, particularly from violent lyrics.(61,62) However, studies are not entirely consistent and some show no association between television violence and youth violence and aggression.(63,64)

Aims:

1. To examine the association between amount of television viewing in childhood/adolescence and depression in adolescents.

2. To examine the association between amount of television viewing in childhood/adolescence and antisocial behaviour in adolescents.

Hypotheses:

We hypothesise that adolescents who watched a greater amount of television in childhood or adolescence are more likely to show depressive symptoms and exhibit antisocial behaviour in adolescence. There are likely to be multiple reasons for the association of depression with television viewing including the displacement of time from more important developmental and social activities, certain television content causing distress/anxiety and poor sleep, unreal ideals of attractiveness and life dissatisfaction. Television viewing may also be used to ameliorate mood, block anxious thoughts or as a coping mechanism for stress. Regarding antisocial behaviour, the Cultivation theory predicts that through television content, frequent exposure to messages of violence can affect perceptions and behaviour in children and adolescents.

Exposure and outcome variables:

Exp: Self-reported television viewing at age 16

Out: Depressive symptoms and anti-social behaviour at age 18

Exp: Self-reported television viewing at ages 14 and 16

Out: Depressive symptoms and antisocial behaviour at ages 16 and 18

Exp: Depressive symptoms and antisocial behaviour at ages 14 and 16

Out: Self-reported television viewing at age 16

Exp: Parent-reported television viewing at ages 4, 5, 6, and 9 - Weekday, weekend, school day and holidays to create daily average

Out: Depressive symptoms and antisocial behaviour at ages 14, 16 and 18

Confounding variables:

Physical activity, BMI, previous depression, socio-economic indicators, major life events, substance misuse, gender, maternal depression, completion of school and educational achievement, parental education, neglect, parental abuse, early violence, neighbourhood violence, early antisocial behaviour.

Analysis

The main outcomes could be treated either as a binary or continuous variable. However, both depressive symptom scores and antisocial behaviour do not have normally distributed frequencies so our primary analysis will use logistic regression with binary outcomes. We will use the logistic regression model to adjust for confounding variables.

References

1. Marshall SJ, Gorely T, Biddle SJ. A descriptive epidemiology of screen-based media use in youth: a review and critique. Journal of adolescence. 2006;29(3):333-49. Epub 2005/10/26.

2. Generation M2: Media in the Lives of 8- to 18-Year-Olds - Kaiser Family Foundation [Internet]. [cited 2012 Oct 7]. Available from: http://www.kff.org/entmedia/mh012010pkg.cfm

3. Office for National Statistics and Department for Environment, Food and Rural Affairs, Living Costs and Food Survey, 2010 [computer file]. 2nd Edition. Colchester, Essex: UK Data Archive [distributor], July 2012. SN: 6945, http://dx.doi.org/10.5255/UKDA-SN-6945-2

4. Ofcom - Independent regulator and competition authority for the UK communications industries. Facts and figures. [Online] Available from: http://media.ofcom.org.uk/facts/

5. Independent Television Commission, Television: the Public's View, 2002 [computer file]. Colchester, Essex: UK Data Archive [distributor], March 2005. SN: 5137, http://dx.doi.org/10.5255/UKDA-SN-5137-1

6. Kubey RW, Csikszentmihalyi M. Television and the quality of life : how viewing shapes everyday experiences. Hillsdale, N.J.: L. Erlbaum Associates; 1990.

7. Shanahan J, Morgan MS. Television and its viewers : cultivation theory and research. Cambridge: Cambridge University Press; 1999.

8. Bandura A. Social Cognitive Theory of Mass Communication. Media Psychology. 2001;3(3):265-99.

9. Gerbner G. The analysis of communication content. Developments in scientific theories and computer techniques. Edited by George Gerbner [and others]: New York, etc.: John Wiley & Sons; 1969.

10. Kappos AD. The impact of electronic media on mental and somatic children's health. International journal of hygiene and environmental health. 2007;210(5):555-62. Epub 2007/09/18.

11. Gerbner G, Gross L, Morgan M, Signorielli N. Growing up with television: the cultivation perspective. In Bryant J, Zillmann D, eds. Media Effects: Advances in Theory and Research. Hillsdale, NJ: Lawrence Erlbaum; 1994:17-41

12. Tiggemann M, Slater A. Thin ideals in music television: a source of social comparison and body dissatisfaction. The International journal of eating disorders. 2004;35(1):48-58. Epub 2004/01/06.

13. Education Committe on Public Education. Children, Adolescents, and Television. Pediatrics. 2001 Jan 2;107(2):423-6.

14. Friedrich LK, Stein AH. Prosocial Television and Young Children: The Effects of Verbal Labeling and Role Playing on Learning and Behavior. Child Development. 1975 Mar 1;46(1):27-38.

15. Strasburger VC. Children, adolescents, and the media. Current Problems in Pediatric and Adolescent Health Care. 2004 Feb;34(2):54-113.

16. Liebert RM, Sprafkin J. The early window: Effects of television on children and youth (3rd ed.). Elmsford, NY, US: Pergamon Press; 1988.

17. Christakis DA, Zimmerman FJ, DiGiuseppe DL, McCarty CA. Early Television Exposure and Subsequent Attentional Problems in Children. Pediatrics. 2004 Jan 4;113(4):708-13.

18. Hamer M, Stamatakis E, Mishra GD. Television- and Screen-Based Activity and Mental Well-Being in Adults. American Journal of Preventive Medicine. 2010 Apr;38(4):375-80.

19. Primack BA. Association Between Media Use in Adolescence and Depression in Young Adulthood, A Longitudinal Study. Archives of General Psychiatry. 2009 Feb 1;66(2):181.

20. Hume C, Timperio A, Veitch J, Salmon J, Crawford D, Ball K. Physical activity, sedentary behavior, and depressive symptoms among adolescents. Journal of Physical Activity. 2011 Feb;8(2):152-6.

21. Cao H, Qian Q, Weng T, Yuan C, Sun Y, Wang H, et al. Screen time, physical activity and mental health among urban adolescents in China. Preventive Medicine. 2011 Oct;53(4-5):316-20.

22. Page AS, Cooper AR, Griew P, Jago R. Children's screen viewing is related to psychological difficulties irrespective of physical activity. Pediatrics. 2010;126(5):e1011-7. Epub 2010/10/13.

23. Barr-Anderson DJ, Berg PVD, Neumark-Sztainer D, Story M. Characteristics Associated With Older Adolescents Who Have a Television in Their Bedrooms. Pediatrics. 2008 Jan 4;121(4):718-24.

24. Chen M-Y, Liou Y-M, Wu J-Y. The Relationship Between TV/Computer Time and Adolescents' Health-Promoting Behavior: A Secondary Data Analysis. Journal of Nursing Research March 2008. 2008;16(1):75-85.

25. Singer MI, Slovak K, Frierson T, York P. Viewing preferences, symptoms of psychological trauma, and violent behaviors among children who watch television. J Am Acad Child Adolesc Psychiatry. 1998 Oct;37(10):1041-8.

26. Kennedy C, Charlesworth A, Chen J-L. Disaster at a distance: Impact of 9.11.01 televised news coverage on mothers' and children's health. Journal of Pediatric Nursing. 2004 Oct;19(5):329-39.

27. Gaina A, Sekine M, Hamanishi S, Chen X, Wang H, Yamagami T, et al. Daytime Sleepiness and Associated Factors in Japanese School Children. The Journal of Pediatrics. 2007 Nov;151(5):518-522.e4.

28. Van den Bulck J. Television viewing, computer game playing, and Internet use and self-reported time to bed and time out of bed in secondary-school children. Sleep. 2004 Feb 1;27(1):101-4.

29. Qidwai W, Ishaque S, Shah S, Rahim M. Adolescent lifestyle and behaviour: a survey from a developing country. PLoS ONE [Electronic Resource] [Internet]. 2010 [cited 2012 Jun 25];5(9).

30. Hammermeister J, Brock B, Winterstein D, Page R. Life Without TV? Cultivation Theory and Psychosocial Health Characteristics of Television-Free Individuals and Their Television-Viewing Counterparts. Health Communication. 2005;17(3):253-64.

31. Dworak M, Schierl T, Bruns T, Struder HK. Impact of singular excessive computer game and television exposure on sleep patterns and memory performance of school-aged children. Pediatrics. 2007 Nov;120(5):978-85.

32. Hargreaves DA, Tiggemann M. Idealized media images and adolescent body image: 'comparing' boys and girls. Body Image. 2004 Dec;1(4):351-61.

33. Hargreaves D, Tiggemann M. Longer-term implications of responsiveness to 'thin-ideal' television: support for a cumulative hypothesis of body image disturbance? European Eating Disorders Review. 2003;11(6):465-77.

34. Groesz LM, Levine MP, Murnen SK. The effect of experimental presentation of thin media images on body satisfaction: A meta-analytic review. International Journal of Eating Disorders. 2002;31(1):1-16.

35. Markey CN, Markey PM. A correlational and experimental examination of reality television viewing and interest in cosmetic surgery. Body Image. 2010 Mar;7(2):165-71.

36. Bruni L, Stanca L. Watching alone: Relational goods, television and happiness. Journal of Economic Behavior & Organization. 2008 Mar;65(3-4):506-28.

37. Dittmar ML. Relations among depression, gender, and television viewing of college students. J Soc Behav Pers, 9 (1994), pp. 317-328.

38. Lemal M, Van den Bulck J. Television news exposure is related to fear of breast cancer. Preventive Medicine. 2009 Feb;48(2):189-92.

39. Potts R, Sanchez D. Television viewing and depression: No news is good news. Journal of Broadcasting & Electronic Media. 1994;38(1):79-90.

40. Gerbner G, Gross L, Morgan M, Signorielli N, Shanahan J. Growing up with television: Cultivation processes. In: Bryant J, Zillmann D, editors. Media effects: Advances in theory and research (2nd ed.). Mahwah, NJ, US: Lawrence Erlbaum Associates Publishers; 2002. p. 43-67.

41. Gerbner G, Gross L, Signorielli N, Morgan M. Aging with television: images on television drama and conceptions of social reality. The Journal of communication. 1980;30(1):37-47. Epub 1980/01/01.

42. Signorielli N, Gerbner G, Morgan M. Standpoint: Violence on television: The cultural indicators project. Journal of Broadcasting & Electronic Media. 1995;39(2):278-83.

43. O'Guinn TC, Shrum LJ. The Role of Television in the Construction of Consumer Reality. Journal of Consumer Research. 1997 Mar 1;23(4):278-94.

44. Bruni L, Stanca L. Income Aspirations, Television and Happiness: Evidence from the World Values Survey. Kyklos. 2006;59(2):209-25.

45. Shrum LJ, Burroughs JE, Rindfleisch A. Television's Cultivation of Material Values. Journal of Consumer Research. 2005 Dec 1;32(3):473-9.

46. Layard R. Happiness: Lessons from a New Science (Second Edition). Penguin UK; 2006.

47. Korkeila J, Markkula J, Korhonen P. P02-190 - High level of TV viewing is associated with silent inflammation. European Psychiatry. 2010;25, Supplement 1(0):814.

48. Anderson DR, Collins PA, Schmitt KL, Jacobvitz RS. Stressful Life Events and Television Viewing. Communication Research. 1996 Jan 6;23(3):243-60.

49. Kurdek LA. Gender Differences in the Psychological Symptomatology and Coping Strategies of Young Adolescents. The Journal of Early Adolescence. 1987 Jan 12;7(4):395-410.

50. Balantekin KN, Roemmich JN. Children's coping after psychological stress. Choices among food, physical activity, and television. Appetite. 2012 Oct;59(2):298-304.

51. Hancox RJ, Milne BJ, Poulton R. Association between child and adolescent television viewing and adult health: a longitudinal birth cohort study. The Lancet. 364(9430):257-62.

52. Vereecken C, Maes L. Television viewing and food consumption in Flemish adolescents in Belgium. Sozial- und Praventivmedizin/Social and Preventive Medicine. 2006;51(5):311-7.

53. Katzmarzyk PT, Lee I-M. Sedentary behaviour and life expectancy in the USA: a cause-deleted life table analysis. BMJ Open [Internet]. 2012 Jan 1 [cited 2012 Oct 7];2(4). Available from: http://bmjopen.bmj.com/content/2/4/e000828

54. Villiani, S. Impact of Media on Children and Adolescents: A 10-Year Review of the Research. Journal of the American Academy of Child & Adolescent Psychiatry. 2001 Apr;40(4):392-401.

55. Manganello JA, Taylor CA. Television Exposure as a Risk Factor for Aggressive Behavior Among 3-Year-Old Children. Archives of Pediatrics & Adolescent Medicine November 2009. 2009;163(11):1037-45.

56. Federman J, ed. National Television Violence Study. Vol 3. Thousand Oaks, CA: Sage; 1998

57. Christakis DA, Zimmerman FJ. Violent Television Viewing During Preschool Is Associated With Antisocial Behavior During School Age. Pediatrics. 2007 Jan 11;120(5):993-9.

58. Robinson TN WM. Effects of reducing children's television and video game use on aggressive behavior: A randomized controlled trial. Arch Pediatr Adolesc Med. 2001 Jan 1;155(1):17-23.

59. Johnson JG, Cohen P, Smailes EM, Kasen S, Brook JS. Television Viewing and Aggressive Behavior During Adolescence and Adulthood. Science. 2002 Mar 29;295(5564):2468-71.

60. DuRant RH, Rich M, Emans SJ, Rome ES, Allred E, Woods ER. Violence and weapon carrying in music videos: a content analysis. Arch Pediatr Adolesc Med. 1997;151:443-448

61. Kunkel D, Cope KM, Colvin C. Sexual Messages on Family Hour Television: Content and Context. Menlo Park, CA: Henry J Kaiser Family Foundation; 1996

62. Brummert Lennings HI, Warburton WA. The effect of auditory versus visual violent media exposure on aggressive behaviour: The role of song lyrics, video clips and musical tone. Journal of Experimental Social Psychology. 2011 Jul;47(4):794-9.

63. Savage J. Does viewing violent media really cause criminal violence? A methodological review. Aggression and Violent Behavior. 2004 Nov;10(1):99-128.

64. Ferguson C, San Miguel C, Hartley R. A multivariate analysis of youth violence and aggression: the influence of family, peers, depression, and media violence. Journal of Pediatrics. 2009 Dec;155(6):904-8.

Date proposal received: 
Thursday, 25 October, 2012
Date proposal approved: 
Thursday, 25 October, 2012
Keywords: 
Depression
Primary keyword: 

B1455 - Nutritional genetic and epigenetic contributions to blood pressure and cardiovascular disease - 25/10/2012

B number: 
B1455
Principal applicant name: 
Dr Kaitlin Wade (University of Bristol, UK)
Co-applicants: 
Dr Nic Timpson (University of Bristol, UK), Prof George Davey Smith (University of Bristol, UK)
Title of project: 
Nutritional, genetic and epigenetic contributions to blood pressure and cardiovascular disease.
Proposal summary: 

Background

Cardiovascular disease (CVD) is the leading cause of world-wide mortality, with more than 42 million people1 affected by forms of the disease including stroke, atherosclerosis, and hypertension. In 2008, an estimated 17.3 million people died from CVD, representing more than 30% of all global deaths2. Hypertension is a major risk factor for CVD3 and causes ~50% of ischemic heart disease and increases the risk of stroke4. The World Health Organisation estimates hypertension to contribute to 7.1 million deaths every year2, where the prevalence was estimated to be 40% in adults over 25 years old in 20082. Hypertension becomes a greater risk for CVD with age, where systolic blood pressure (SBP) becomes of an important predictor of CVD risk4.

Dietary sodium intake is one of the most common and important risk factors for hypertension5-8. Combinations of observational studies9,10, clinical trials11,12 and meta-analyses13-15 have shown a positive association between salt intake and sodium excretion with blood pressure (BP) and hypertension risk16.

Conducted in 1988, the clinical study INTERSALT17 found that sodium excretion was positively associated with SBP within 10,079 men and women aged 20-59 across 48 centres from 32 countries. The estimated effect of 100mmol/d lower dietary sodium intake corresponded to a 2.2mmHg lower average population SBP17-19. The World Heart Federation estimates that a universal reduction in dietary sodium intake of about 1g a day (approximately 3g of salt) would lead to a 50% reduction in required hypertensive treatment, 22% fewer deaths resulting from strokes and 16% fewer deaths from coronary heart disease (CHD)20.

A recent meta-analysis of randomized controlled trials (RCTs)13 identified seven studies where reductions in sodium excretion of between 27-39mmol/24h were associated with a decrease in SBP of 1-4mmHg. However, there was no strong evidence supporting decreases in CVD morbidity with salt restriction.

Animal models have shown an association between high salt intake and risk of hypertension. For example, chimpanzees fed a diet containing 35 versus 120mmol of sodium per day had significantly lower BP21. And after introducing a diet containing ~248mmol of sodium per day for 2 years, subsequent reduction to ~126mmol of sodium per day reduced BP compared to animals maintained on an increased salt diet21. Additionally, Dahl salt-sensitive rats fed on a high NaCl diet have a greater rise in BP compared with salt-resistant rats fed on the same diet22. Within 3 generations of selection, the salt-sensitive rats and salt-resistant rat strains show clear differences, suggesting that salt sensitivity is inherited.

However, some studies have also shown negative results suggesting that low salt intake could even be harmful23-25, highlighting the inconsistencies within this area. Moreover, observational studies are known to suffer from bias, confounding and reverse causation, potentially providing misleading results, which could prove costly when identifying specialised drug targets for reducing the prevalence of CVD. CVD is a complex trait, where the maintenance of BP is influenced by multiple environmental and genetic determinants, as well as their interactions.

The mechanisms underlying the relationship between dietary salt intake and BP are many and complex, achieved by paracrine, neural, endocrine and systematic control. For example, once plasma volume decreases, renin is released from the juxtaglomerular cells (JGCs) within the kidney and activates the renin-angiotensin system (RAS). Renin transforms angiotensinogen to angiotensin I (ANG I), which is then converted to ANG II by the angiotensin-converting enzyme (ACE)26. ANG II then stimulates the adrenal cortex to release aldosterone (Aldo), which stimulates sodium and water absorption through many mechanisms, restoring plasma volume. Another pathway involves transforming growth factor-beta (TGF-beta), where excess salt intake rapidly increases endothelial production of TGF-beta, causing arterial stiffness, peripheral vasoconstriction and arterial hypertension27. Nitric oxide (NO) normally counterbalances the effects of TGF-beta, but is depleted in salt-sensitive individuals, increasing susceptibility to hypertension. Salt-sensitivity involves a genetic defect, combined with renal injury and other environmental factors. Therefore, in genetically predisposed individuals, high salt intake reduces renal vascular function and increases the risk of hypertension27.

The prevalence and complexity of CVD has driven genetic studies that identify genes associated with sodium/water homeostasis and exploit the properties of these variants with the hope to develop greater understanding the pathways of salt-induced high BP and salt-sensitive hypertension.

GenSalt28,29 is a prospective intervention study in rural China of 3,153 participants in 658 families, with untreated pre-hypertension or hypertension. The study has identified many candidate genes for the underlying pathway explaining the association between salt intake and BP. These genes encompass the renin-angiotensin system (REN, AGT, AT2R1/2, ACE, RENBP, ACE2, APLN and AGTRL1), the aldosterone system (CYP11B1/2, MLR and HSD11B1/2), and the endothelial system (ET1/EDN1, NOS3 and SELE), epithelial sodium channels (ENaC, SCNN1B and SCNN1G)29. Additional genome-wide association studies (GWAS) have identified more than 47 genetic variants at 40 loci associated with BP and hypertension risk30-41. Additionally, 8 genetic variants have been associated with BP in more than 25,000 individuals: at the MTHFR-NPPB, AGT, NPR3, HFE, NOS3, LSP1/TNNT3, SOX6 and ATP2B1 loci3. Using a multi-stage design in 200,000 individuals of European descent, the International Consortium for Blood Pressure GWAS on SBP, DBP and hypertension risk identified 29 loci robustly associated with BP, 16 of which were novel. Of these, six loci contained genes previously thought to regulate BP: GUCY1A3-GUCY1B3, NPR3-c5orf23, ADM, FURIN-FES, GOSR2 and GNAS-EDN3) and revealed ten new loci providing clues on BP aetiology including SLC4A7, SLC39A8, MOV10, EBF1 and JAG1. However, these only explain a small amount of the heritability of this trait.

Much effort has focused on understanding the 'missing heritability' in complex traits such as CVD, which can be attributable to small effects of genetic variants, rare variants of moderate penetrance and gene-environment interactions. Additionally, there is a considerable lack of causal and mechanistic analyses on the association between dietary sodium intake and BP. Evidence suggests that the salt/water balance is in part mediated by epigenetic mechanisms, specifically histone modification and DNA methylation (in animals and plants), which alters expression levels of important regulatory genes such as epithelial Na+ channels (ENaCs) in response to varying salt intake42-46. These epigenetic mechanisms also play a role in the prenatal imprinting of postnatal-specific feeding behaviours and intergenerational transmission of salt appetite from mother to offspring47, a pathway of which is evident in rats and has been verified in human newborn infants47. Furthermore, data suggests that environmental, particularly nutritional, factors during pregnancy can lead to changes in DNA methylation of the offspring48, supporting the hypothesis that origins of diseases in adults begin in utero48-50.

Epigenetic association studies are beginning to accumulate51-53. Importantly, differentiating between causative and consequential differential epigenetic modifications is difficult but necessary, as epigenetic marks are more similar to phenotypes than genotypes, and can thus be confounded by other environmental factors.

Our knowledge in this area is far from complete and inconsistencies between previous studies drives the importance of understanding the causal and mechanistic pathways underlying this known association between dietary sodium intake and BP. Therefore, I intend to examine the longitudinal, intergenerational, intrauterine, and epigenetic contributions of dietary sodium intake to BP regulation, assessing the underlying causal pathways and mechanisms.

Objectives

1. Identify genes involved in sodium regulation and excretion through GWAS, focusing on genes with the greatest diversity of effect on circulating sodium levels.

2. Use a novel MR approach to assess the causal links between dietary sodium intake and BP with known genetic variants.

3. Multi-level model of how BP changes longitudinally in children as a result of dietary sodium intake, in terms of genetic and epigenetic pathways.

4. Examine the intergenerational and intra-uterine effects of maternal diet and offspring BP, taking into account paternal, maternal-prenatal and maternal-postnatal dietary intake and the challenges with this type of analysis.

5. Examine the differential methylation in BP of children and mothers, prospectively comparing BP of children with varying diets.

6. Assess differential transcription/expression using mRNA levels due to differential methylation found in (5).

7. Identify differences in BP due to varying diets in urban vs. rural populations and exploit the genetic information available on BP to assess rural vs. urban interaction.

Study Design

Participants and Variables

GWAS will be based on the ~10,000 children within ALSPAC54 who have been genotyped on the 610K Illumina SNP Chip. Averages from repeated BP measurements will be used to increase the reliability and sensitivity of this trait. BP has been collected from children at 37, 49 and 61 months and 7, 9, 10, 11, 12, 13, 15, and 17 years at rest and after exercise. Maternal BP measurements were collected at each pregnancy trimester, and when the children were 11, 13, 15 and 17 years old. Information on family history of hypertension, CHD and stroke from parents is also available.

Salt intake will be derived from 3-day nutritional diaries and food frequency questionnaires (FFQ) from a sub-sample from ALSPAC children collected at 4, 8, 18, 43, and 61 months, and at 7.5, 10.6 and 13.9 years and from mothers and 32 weeks gestation, and at 47 months, 97 months and 13 years. Nutrient intakes and food groups consumed will be derived from the FFQs and diet diaries using similar methods to previous studies16,55. Information on sodium excretion and sodium-related metabolites56 will be obtained from the ~1,000 children who have urine spot samples taken at age 10, 15 and 17. Epigenomic information will be obtained from ARIES in 1,000 ALSPAC mothers and children, of whom have phenotypic data. Intergenerational data will primarily use ALSPAC (and COCO90s, if available). Assessment of urban vs. rural interaction will use the Indian Migration Study57.

Rare variants analysis will utilize ~2,000 ALSPAC children who have whole-genome sequencing data and will be used as a reference set for imputing the rest of the cohort. Significant associations will be performed for replication in other cohorts where suitable data are available.

Significant associations will be followed up in available data from cohorts including EPIC, BWHHS, BRHS, Birth to Twenty, Indian Migration Study, Hellenic Isolated Cohorts (HELIC), Born in Bradford, Danish National Birth Cohort, Generation R and the German Infant Study on the Influence of Nutrition Intervention.

Statistical Analysis

Genome-wide data will initially be imputed to HapMap Phase II. Rare variant (1-5% MAF) will be using genome-wide SNP data from ~10,000 ALSPAC children imputed into UK10K dataset (or 1,000 genomes if UK10K is not available).

Multivariate regression will be used to assess associations between exposures and outcomes, where analyses will be adjusted for appropriate confounders (eg. age, sex, BMI). Intergenerational associations between maternal diet and offspring BP will take into account additional covariates (eg. maternal nutrition before pregnancy, supplementation, birthsize, intake after birth, child growth from birth and sex). Longitudinal multivariate modelling of BP in ALSPAC children will use MLwiN to fit multi-level models, which describe BP variation over time as well as covariation between BP phenotypes. MR methodology will be used to assess causal associations between salt intake and BP, using dietary data and sodium-related metabolites as the exposures. Associations between paternal, maternal post-natal and maternal pre-natal dietary information and offspring BP will be examined to assess whether associations between maternal diet and offspring BP is due to intrauterine effects.

Assessment of epigenetic associations will involve examining differential methylation and using the two-step MR approach58. Expression levels will be investigated using genome-wide expression data on 1,000 ALSPAC children measured from lymphoblastoid cell lines using the Illumina Human-6 v2 Beadchip (48,000 transcripts). All analysis will be using STATA, R, PLINK and MLwiN.

Date proposal received: 
Thursday, 25 October, 2012
Date proposal approved: 
Thursday, 25 October, 2012
Keywords: 
Blood Pressure, Epigenetics , Genetics, Nutrition
Primary keyword: 

B1454 - The association between birth weight infant growth and asthma until young adulthood in the ALSPAC cohort - 11/10/2012

B number: 
B1454
Principal applicant name: 
Dr Agnes Sonnenschien-van der Voort (Erasmus University Medical Center, Rottterdam, the Netherlands, Europe)
Co-applicants: 
Prof John Henderson (University of Bristol, UK), Dr Raquel Granell (University of Bristol, UK)
Title of project: 
The association between birth weight, infant growth and asthma until young adulthood in the ALSPAC cohort.
Proposal summary: 

AIMS

Various different asthma definitions are used in research and clinic. It is important to identify specific underlying mechanisms for the associations of early life exposures with different asthma related outcomes, which might reflect different specific structural and functional adaptations. Major potential early life risk factors are a low birth weight, and change in infant's growth. In a large population-based cohort study followed from birth until the age of 17 years, our aim is to identify potential risk factors for the different asthma phenotypes in young adulthood.

HYPOTHESIS

A recent paper on fetal and infant growth and asthma symptoms in preschool children showed an association between infant weight gain and asthma symptoms, independent of fetal growth, suggesting that early infancy might be critical for the development of asthma. The effect of infant growth on asthma phenotypes and lung function measurements at older ages needs to be explored. If adverse infant growth is also associated with respiratory outcomes in young adulthood, a valuable and modifiable risk factor is identified which could be potentially used for new preventive, diagnostic and therapeutic approaches.

Low birth weight is associated with increased risks of asthma, chronic obstructive lung disease, and impaired lung function in adults and with increased risks of respiratory symptoms in the first 7 years. Low birth weight per se is not likely to be the causal factor for asthma. The same birth weight might be the result of various growth patterns. The developmental plasticity hypothesis suggests that the associations between low birth weight and diseases in later life are explained by early adaptation mechanisms in response to various adverse exposures, including smoke exposure. Previous studies showed inconclusive results about fetal growth patterns and fetal smoke exposure and the risk of wheezing in childhood. The associations of longitudinally measured fetal and early childhood growth patterns, and their interactions with wheezing phenotypes, asthma, bronchial hyperresponsiveness and lung function in later life need to be studied in detail.

EXPOSURE VARIABLES

Birth weight and infant growth.

POTENTIAL CONFOUNDER VARIABLES

parental age, body mass index, education and occupation, history of asthma or atopy, smoking during pregancy, gestational age at birth, children's gender, crowding, siblings, breastfeeding status, ethnicity, tobacco smoke exposure, housing, pet keeping and body mass index at time of outcome.

OUTCOME VARIABLES

(1) recently identified asthma phenotypes: never/infrequent wheeze, transient early wheeze, prolonged early wheeze, intermediate onset wheeze, late onset wheeze, persistent wheeze; (2) doctor diagnosed asthma; at ages 8 and 15 years (2) Lung function (spirometry, bronchial responsiveness to methacholine, exhaled nitric oxide; at ages 8 and 15 years).

Date proposal received: 
Thursday, 11 October, 2012
Date proposal approved: 
Thursday, 11 October, 2012
Keywords: 
Asthma, Growth
Primary keyword: 

B1450 - Genetic and Environmental Predictors of ADHD Symptom Trajectories The Role of Symptom Onset and Psychiatric Co-morbidity - 11/10/2012

B number: 
B1450
Principal applicant name: 
Dr Kate Langley (University of Cardiff, UK)
Co-applicants: 
Prof Anita Thapar (University of Cardiff, UK), Dr Kimberley Rhoades (Oregon Social Learning Center, USA), Mrs Joanna Martin (University of Bristol, UK)
Title of project: 
Genetic and Environmental Predictors of ADHD Symptom Trajectories: The Role of Symptom Onset and Psychiatric Co-morbidity.
Proposal summary: 

Attention deficit hyperactivity disorder (ADHD) is a common, extremely disabling disorder which has major adverse squalae in childhood and later life. Available evidence suggests that both genetic and environmental risk factors are important in the aetiology of ADHD; how these influences, and their interplay, impact the course (i.e., trajectories) of ADHD symptoms is poorly understood.

The aim of this proposal is to further explore individual trajectories of ADHD symptoms over time and genetic and family environmental predictors of those trajectories. In addition, we aim to explore the influence of symptom age of onset and psychiatric comorbidity on the course of ADHD.

ADHD symptoms trajectories, on average, show declines from middle childhood through early adolescence (Langberg et al., 2008), although environmental stressors (in this study, the transition to secondary school) can lead to temporary reversals of these declines. Previous research using the ALSPAC sample has identified 4 latent class trajectories of ADHD symptoms over time: low risk, intermediate, childhood limited, and persistently impaired (Pourcain et al., 2011). Other research has found evidence for three symptoms trajectory classes: low; increasing in childhood, then decreasing; and decreasing during childhood and then increasing later in development (Malone et al., 2010). Individual trajectories of ADHD symptoms have implications for later adjustment; in the Malone et al., study, participants with the 3rd trajectory (decreasing symptoms during childhood, then later increases) reported an earlier onset of illicit drug use than the stable low trajectory class.

Little to no research has examined either genetic or family environmental influences on ADHD trajectories over time. Increased knowledge about the aetiology of these symptom trajectories in epidemiological samples is crucial for prevention efforts and provides preliminary foci for future research in clinical samples.

1). First, we propose to explore individual trajectories of ADHD symptoms over time and associations among initial symptoms and longitudinal course. Predictors of these trajectories will then be examined, including the family environment, genetic risk, age of symptom onset, and psychiatric comorbidity, specifically symptoms of CD and ODD.

2). We also propose to look at the COMT val158met genotype which has been shown to be associated with CD in samples with comorbid ADHD (Langley et al., 2010) and a (previously generated) polygenic risk score, following work finding significant association between risk score and comorbid CD in those with ADHD (Hamshere et al., submitted).

3). Using this genetic information, we are interested in looking at parental transmission of risk alleles to their child; if a child is exposed to a dysfunctional family environment and the mother has the risk genotype but does not pass that genotype on to her child, the course of ADHD symptoms over time should be less pathological if shared genetic liability were important, but not if environmental mediation was at work.

4). We additionally plan to examine moderation of genetic risk on longitudinal trajectories of ADHD symptoms by the family environment. The expectation is that children who both have genetic risk for ADHD and are exposed to dysfunctional environments will evidence steady high and/or increasing trajectories of ADHD symptoms over time.

These analyses would help elucidate predictors of the course of ADHD. Furthermore, we aim to replicate our findings from ALSPAC in our clinical sample of (n=800) children with ADHD which should provide further insight.

The polygenic risk score for ADHD and the COMT vala58met genotype that we propose to study are strong candidates for association with ADHD trajectories and associated conduct problems and are also available in the ALSPAC data set, the two main reasons why they have been proposed here. We recognise that other variants in these and other genotypic regions may also be linked to ADHD symptoms and, should funds be available for future genotyping, would be interested in extending our analyses.

Because our proposed study is similar to current work by Barbara Maughan on CD trajectories, we have been in consultation with Dr. Maughan and will maintain a close collaboration during this project to both enhance the quality of the proposed research and to ensure the distinctness and complementarity of our studies. The proposed project with also be conducted with the collaboration of Prof. Gordon Harold and Dr. Stephan Colinshaw at the University of Cardiff.

The proposed study will utilize the polygenic risk score for ADHD currently under development by Joanna Martin (ALSPAC project # B1342; planned completion date for deriving the polygenic risk score is Nov. 1st, 2012). The polygenic risk score is calculated by using the results (below a chosen threshold) of a genome-wide association study (GWAS) of ADHD cases vs. population controls to create a polygenic score for each individual in the target dataset (i.e. in ALSPAC). Thus, we propose to examine the effect of common genetic variants implicated in ADHD, in the form of polygenic scores calculated in ALSPAC based on a discovery sample of children diagnosed with ADHD, on trajectories of ADHD during childhood and adolescence.

Current data required:

1) Parent & Teacher reports DAWBA symptoms and diagnoses of ADHD, CD and ODD at all available assessment points

2) SDQ - subscale summary scores at all available time points

3) Child- and parent-report of antisocial behavior

4) Child and parent report of parental monitoring, parenting, and the parent-child relationship at all available assessments

Items: 6 months: KB554-KB586; 18 months: KD270-KD305, KD366-KD422; 24 months: KE010-KE021; 30 months: KF300-KF326, KF358-KF394, KF430-KF431; 42 months: KJ250-KJ268, KJ298-KJ334, KJ370-KJ459; 77 months: KP6000-KP6050, KP6210-KP6230, KP6270-KP6282; 103 months: KT3030-KT3098; 115 months: KU280-KU329, CCF104, 111, 118, 125, 133, 141, 149, 157, 165; 140 months: KW4000-KW4050, KW9020-KW9088; 169 months: CCR500-CCR550; 16.5 years: TC1000-TC1180

5) Age (parents & child at assessments)

6) Gender

7) BWT

8) Derived variables of Partner Affection, and Partner Aggression at each available time point (e.g., F593-F595 & F596-F598 from 8 week Mother Questionnaire)

9) SES & Mothers education

10) Mother & Father psychopathology - ADHD, CD, Anx. & Depression

11) Mother & Child genotypes

References:

Langberg, J. M., Epstein, J. N., Altaye, M., Molina, B. S. G., Arnold, L. E., Vitiello, B. (2008). The transition to middle school is associated with changes in the developmental trajectory of ADHD symptomatology in young adolescents with ADHD. Journal of Clinical Child and Adolescent Psychology, 37, 651-663.

St. Pourcain, B., Mandy, W. P., Heron, J., Golding, J., Smith, G. D., & Skuse, D. H. (2011). Links between co-occurring social-communication and hyperactive-inattentive trait trajectories. Journal of the American Academy of Child & Adolescent Psychiatry, 50, 892-902.

Malone, P. S., Van Eck, K., Flory, K., & Lamis, D. A. (2010). A mixture model approach to linking ADHD to adolescent onset of illicit drug use. Developmental Psychology, 46, 1543-1555.

Langley, K., Heron, J., O'Donovan, M. C., Owen, M. J., & Thapar, A. (2010). Genotype link with extreme antisocial behavior. Archives of General Psychiatry, 67, 1317-1323.

Hamshere, M.L., Langley, K. et al., High loading of polygenic risk for ADHD in those with comorbid aggression. Submitted.

Date proposal received: 
Thursday, 11 October, 2012
Date proposal approved: 
Thursday, 11 October, 2012
Keywords: 
ADHD, Genetics
Primary keyword: 

B1449 - Genomewide array Illumina platforms testing using the iScan technology at Oakfield House ALSPAC laboratories - 11/10/2012

B number: 
B1449
Principal applicant name: 
Dr Nic Timpson (University of Bristol, UK)
Co-applicants: 
Dr Wendy McArdle (University of Bristol, UK), Dr Susan Ring (University of Bristol, UK)
Title of project: 
Genomewide array (Illumina platforms) testing using the iScan technology at Oakfield House, ALSPAC laboratories.
Proposal summary: 

The iScan technology housed within the ALSPAC laboratories provides an opportunity to collect valuable genetic data on specific samples in small scale projects. However, where dealing with small sample sizes and potentially valuable,limited, DNA sources, one needs to be confident in the efficiency and reliability of the lab based pipelines for data collection. This proposal simply seeks permission to use a small number (likely sample number for each test being 16 samples, though this may vary by platform) of DNA samples from the ALSPAC immortalised cell lines (which have a theoretically infinite supply of DNA) to test new arrays. Data generated from this testing will then be used to verify the performance of particular arrays before use on samples with a finite reserve.

Date proposal received: 
Thursday, 11 October, 2012
Date proposal approved: 
Thursday, 11 October, 2012
Keywords: 
Genetics, Methods
Primary keyword: 

B1447 - The maturational air-bone gap Trends in behavioural air- and bone- conduction hearing thresholds in children - 11/10/2012

B number: 
B1447
Principal applicant name: 
Dr Amanda J Hall (University of Bristol, UK)
Co-applicants: 
Ms Noelle Conheady (University of Bristol, UK)
Title of project: 
The maturational air-bone gap: Trends in behavioural air- and bone- conduction hearing thresholds in children.
Proposal summary: 

Background

For the purposes of hearing assessment, thresholds to air conducted and bone conducted stimuli are converted into comparable units, dBHL at each frequency. This calibration of air and bone- conducted stimuli allows for comparison between air and bone conduction hearing thresholds. The difference between the air conduction threshold and the bone conduction threshold is termed the "air-bone gap" and generally indicates the presence of a conductive (i.e affecting the outer or middle ear) hearing loss. However air and bone conduction thresholds for infants with normal hearing using physiological measures have identified an air-bone gap in the low frequencies that does not result from conductive hearing losses but rather from maturational difference and sensitivity (Small and Stapells 2008 and Vander Werff et al. 2009). Studies to date have suggested that this maturational air bone gap exists up to 3 years of age on both physiological and behavioural studies, however studies specifically examining the patterns of behavioural air conduction and bone conduction thresholds in children older than 3 years of age have not been carried out. It is therefore not known if the maturational air-bone gap exists in children older than three years of age and at what stage it starts to close.

Research Questions:

1. Does a maturational air-bone gap exist in the ALSPAC cohort at age 5, 7, 9 and 11 years?

2. If a maturational air-bone gap is found to exist, at what age does this close/ reduce?

3. Are there frequency dependent trends in behavioural air and bone conduction thresholds for children aged 5, 7, 9 and 11 years?

Objectives:

1. Describe the air conduction hearing thresholds for each stimulus frequency at age 5, 7, 9 and 11.

2. Describe the bone conduction hearing thresholds for each stimulus frequency at age 5, 7, 9 and 11.

3. Identify if there are a) age and b) frequency dependent trends in air conduction and bone conduction hearing thresholds across the different age groups.

4. Quantify the air-bone gap at each age and examine changes over time.

Hypothesis:

It is hypothesised that the air-bone gap decreases with age.

Exposure variable(s): Age of child

Outcome variable(s): Air conduction and bone conduction hearing thresholds

Confounding variable(s): Sex and socioeconomic status.

Date proposal received: 
Thursday, 11 October, 2012
Date proposal approved: 
Thursday, 11 October, 2012
Keywords: 
Hearing
Primary keyword: 

Pages