PreprintPDF Available

Immuno-epigenetic signature derived in saliva associates with the encephalopathy of prematurity and perinatal inflammatory disorders

Authors:

Abstract and Figures

Background: Preterm birth is closely associated with a phenotype that includes brain dysmaturation and neurocognitive impairment, commonly termed Encephalopathy of Prematurity (EoP), of which systemic inflammation is considered a key driver. DNA methylation (DNAm) signatures of inflammation from peripheral blood associate with poor brain imaging outcomes in adult cohorts. However, the robustness of DNAm inflammatory scores in infancy, their relation to comorbidities of preterm birth characterised by inflammation, neonatal neuroimaging metrics of EoP, and saliva cross-tissue applicability are unknown. Methods: Using salivary DNAm from 258 neonates (n = 155 preterm, gestational age at birth 23.28 – 34.84 weeks, n = 103 term, gestational age at birth 37.00 – 42.14 weeks), we investigated the impact of a DNAm surrogate for C-reactive protein (DNAm CRP) on brain structure and other clinically defined inflammatory exposures. We assessed i) if DNAm CRP estimates varied between preterm infants at term equivalent age and term infants, ii) how DNAm CRP related to different types of inflammatory exposure (maternal, fetal and postnatal) and iii) whether elevated DNAm CRP associated with poorer measures of neonatal brain volume and white matter connectivity. Results: Higher DNAm CRP was linked to preterm status (-0.0107 ± 0.0008, compared with -0.0118 ± 0.0006 among term infants; p < 0.001), as well as perinatal inflammatory diseases, including histologic chorioamnionitis, sepsis, bronchopulmonary dysplasia, and necrotising enterocolitis (OR range |2.00 | to |4.71|, p < 0.01). Preterm infants with higher DNAm CRP scores had lower brain volume in deep grey matter, white matter, and hippocampi and amygdalae (β range |0.185| to |0.218|). No such associations were observed for term infants. Association magnitudes were largest for measures of white matter microstructure among preterms, where elevated epigenetic inflammation associated with poorer global measures of white matter integrity (β range |0.206| to |0.371|), independent of other confounding exposures. Conclusions: Epigenetic biomarkers of inflammation provide an index of innate immunity in relation to neonatal health. Such DNAm measures complement biological and clinical metrics when investigating the determinants of neurodevelopmental differences.
Content may be subject to copyright.
1
Immuno-epigenetic signature derived in saliva associates with the
encephalopathy of prematurity and perinatal inflammatory
disorders
Authors: Eleanor L.S. Conole1,2,3*, Kadi Vaher4, Manuel Blesa Cabez4, Gemma Sullivan4, Anna
J. Stevenson2, Jill Hall4, Lee Murphy5, Michael J. Thrippleton3,5, Alan J. Quigley6, Mark E.
Bastin1,3, Veronique E. Miron4, Heather C. Whalley3, Riccardo E. Marioni2, James P.
Boardman4,3, Simon R. Cox1*
1 Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh
EH8 9JZ, UK.
2 Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of
Edinburgh, Edinburgh EH4 2XU, UK
3 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
4 MRC Centre for Reproductive Health, Queen’s Medical Research Institute, Edinburgh
BioQuarter, University of Edinburgh, Edinburgh, EH16 4TJ
5 Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh EH4 2XU, UK
6 Imaging Department, Royal Hospital for Children and Young People, Edinburgh, EH16 4TJ
*Correspondence to:
Simon. R. Cox
Email: simon.cox@ed.ac.uk
Eleanor. L. S. Conole
Email: eleanor.conole@ed.ac.uk
2
Abstract
Background: Preterm birth is closely associated with a phenotype that includes brain
dysmaturation and neurocognitive impairment, commonly termed Encephalopathy of Prematurity
(EoP), of which systemic inflammation is considered a key driver. DNA methylation (DNAm)
signatures of inflammation from peripheral blood associate with poor brain imaging outcomes in
adult cohorts. However, the robustness of DNAm inflammatory scores in infancy, their relation to
comorbidities of preterm birth characterised by inflammation, neonatal neuroimaging metrics of
EoP, and saliva cross-tissue applicability are unknown.
Methods: Using salivary DNAm from 258 neonates (n = 155 preterm, gestational age at birth
23.28 34.84 weeks, n = 103 term, gestational age at birth 37.00 42.14 weeks), we
investigated the impact of a DNAm surrogate for C-reactive protein (DNAm CRP) on brain
structure and other clinically defined inflammatory exposures. We assessed i) if DNAm CRP
estimates varied between preterm infants at term equivalent age and term infants, ii) how DNAm
CRP related to different types of inflammatory exposure (maternal, fetal and postnatal) and iii)
whether elevated DNAm CRP associated with poorer measures of neonatal brain volume and
white matter connectivity.
Results: Higher DNAm CRP was linked to preterm status (-0.0107  ±  0.0008, compared with -
0.0118  ±  0.0006 among term infants; p < 0.001), as well as perinatal inflammatory diseases,
including histologic chorioamnionitis, sepsis, bronchopulmonary dysplasia, and necrotising
enterocolitis (OR range |2.00 | to |4.71|, p < 0.01). Preterm infants with higher DNAm CRP
scores had lower brain volume in deep grey matter, white matter, and hippocampi and
amygdalae (β range |0.185| to |0.218|). No such associations were observed for term infants.
Association magnitudes were largest for measures of white matter microstructure among
preterms, where elevated epigenetic inflammation associated with poorer global measures of
white matter integrity (β range |0.206| to |0.371|), independent of other confounding exposures.
Conclusions: Epigenetic biomarkers of inflammation provide an index of innate immunity in
relation to neonatal health. Such DNAm measures complement biological and clinical metrics
when investigating the determinants of neurodevelopmental differences.
Key words
Perinatal; Inflammation; DNA methylation; Epigenetics; Encephalopathy of Prematurity; White
Matter; Preterm Birth; Necrotizing Enterocolitis; Neonatal Sepsis; Diffusion Tensor Imaging;
Neurodevelopment; Multiomics
3
1. Introduction
Preterm infants are at an increased risk of elevated inflammation, related health complications,
and adverse neurodevelopment compared to infants born at term (18). While the aetiology of
these outcomes is multifactorial, inflammation is considered to be a key component linking
preterm birth and poor neurodevelopmental and mental health outcomes via its effects on
cerebral maturational processes (912). Neonatal neuroimaging has identified neurostructural
hallmarks of preterm birth commonly referred to as Encephalopathy of Prematurity (EoP),
including dysmaturation of cortical and deep grey matter, atypical white matter development and
disrupted connectivity (13). Recent advances in epigenetics may permit new ways to
characterise sustained inflammation and reveal new insights into the relationship between
inflammatory exposures, inflammation and neonatal brain and health outcomes.
Preterm infants are more susceptible to sustained inflammation than term infants and can be
subject to multiple inflammatory stimuli during the perinatal period (14,15). Alongside maternal
lifestyle-related exposures (16), various complications during pregnancy such as preeclampsia
and histologic chorioamnionitis (17,18) can induce both maternal and fetal inflammatory
responses and increase the risk of a sustained pro-inflammatory state postnatally (1722).
Preterm infants are additionally at higher risk for developing severe inflammatory conditions in
the first few weeks of life, which may in turn perpetuate inflammation (8)including
bronchopulmonary dysplasia (BPD), necrotizing enterocolitis (NEC), severe retinopathy of
prematurity (ROP) and episodes of sepsis (23). Preterm infants often present with multiple
chronic conditions at once (24), putting them at higher risk of a greater allostatic load of
inflammation (14,2426).
Though numerous studies report associations between inflammation and cognitive outcomes in
preterm populations (8,2731), research linking inflammatory biomarkers with neurostructural
measures yield inconsistent findings (4,6,11,19,21,3237). Gaining a clearer understanding of
the pathways via which sustained inflammation in very early life may precipitate well
characterised cognitive and neurostructural outcomes requires novel approaches. The relative
inconsistency of work to date is likely due to heterogeneity in study design: there is substantial
variation in the demographic characteristics of study samples; the degree to which residual
confounding factors are controlled; and the presence or absence of term control groups.
Moreover, the relative nascency of the neonatal neuroimaging field (38) contributes to substantial
variation in the acquisition and selection of brain outcome measures, and there is marked
anatomic variation in early life, which can confound investigation of structure-function
relationships (39).
4
Above these factors, we argue that the measures used to characterise inflammation in the first
place may account for the greatest source of ambiguity in the inflammation-brain structure
literature. In both clinical and research settings, there is a historical reliance on sampling phasic
inflammation-related protein measures from blood to signpost inflammation. Of these, C-
Reactive Protein (CRP) is the most widely adopted (40), although there are criticisms of this
approach (41,42), particularly in newborn populations (40,43) where the CRP response to
inflammation is variable owing to an immature immune system (15). Accurate characterisation of
sustained (and not transient or acute) inflammation arguably requires repeated sampling or
follow up examinations, which few studies endeavour to profile (44). This, alongside uncertainty
of what exactly constitutes sustained inflammation in the preterm infant (15) calls for alternative,
more stable biomarkers which can accurately reflect baseline inflammation levels. There is a
clear precedent to identify tools to both circumvent the practical limitations of conventional
inflammatory measurement in the neonate, alongside study designs that necessitate detailed
brain and clinical information with adequate statistical power to detect small to moderate effect
sizes.
Our previous work demonstrated that DNA methylation (DNAm) markers of inflammation may
provide more stable readouts of cumulative inflammatory exposure (45,46) and shed greater
insight into the consequences of inflammation on brain structure (47,48). DNAm is an epigenetic
mechanism that can act as an interface by which environmental exposures influence gene
function. DNAm is dynamic during fetal development, both in terms of the developing immune
system (49) and brain (5052) and may mediate the impact of maternal, fetal and postnatal
exposures on brain development (5355). In the context of preterm birth, only a limited number
of studies have investigated DNAm changes (53,5665)of these, few examine DNAm in
relation to neonatal neuroimaging metrics (59,62,63). Additionally, though some of these studies
have examined DNAm in relation to postnatal health outcomes (53,65), no study to date has
examined inflammation, DNAm, and neuroimaging concurrently in the neonatal period. This
study is the first time an epigenetic measure of inflammation has been examined in a neonatal
cohort in relation to brain health outcomes.
Here, using a cohort of 258 infants (103 term, 155 preterm), we examine (1) how a salivary
DNAm signature of the inflammatory marker C-reactive protein (DNAm CRP) relates to preterm
birth (2) how this signature associates with maternal, fetal and postnatal inflammatory exposures
both individually and in aggregate, and (3) how variance in this measure relates to global
measures of MRI brain volume, diffusion MRI (dMRI) correlates of connectivity, and regional
variation in individual white matter tracts.
5
2. Methods
2.1 Study population
Preterm (gestational age at birth < 37 weeks) and term born infants delivered at the Royal
Infirmary of Edinburgh, UK were recruited to the Theirworld Edinburgh Birth Cohort, a
longitudinal study designed to investigate the effect of preterm birth on brain development (66).
Cohort exclusion criteria were major congenital malformations, chromosomal abnormalities,
congenital infection, overt parenchymal lesions (cystic periventricular leukomalacia, hemorrhagic
parenchymal infarction) or post-hemorrhagic ventricular dilatation. Ethical approval has been
obtained from the National Research Ethics Service, South East Scotland Research Ethics
Committee (11/55/0061, 13/SS/0143 and 16/SS/0154). Informed consent was obtained from a
person with parental responsibility for each participant. DNAm data were available from 258
neonates, 214 of whom also had successful structural and diffusion MRI acquisition.
2.2 Study variables
Inflammatory exposures were coded as binary variables (1 = present, 0 = absent) and were
grouped as follows: maternal (pertaining to mother / maternal exposure), fetal (affecting placenta
or fetus) or neonatal (affecting infant after birth). Table 1 presents participant characteristics of
these categories. Histologic chorioamnionitis (HCA) was defined via placental histopathology, as
reported previously (17,21). Incidence of any neonatal sepsis (either late onset or early onset
sepsis) was defined as detection of bacterial pathogen from blood culture, or physician decision
to treat for ≥5 days in the context of growth of coagulase negative staphylococcus from blood or
a negative culture. Necrotising enterocolitis (NEC) was defined as stages two or three according
to the modified Bell’s staging for NEC (67). Bronchopulmonary dysplasia (BPD) was defined by
the requirement for supplemental oxygen at 36 weeks gestational age. Birthweight z-scores were
calculated according to International Fetal and Newborn Growth Consortium for the 21st Century
(INTERGROWTH-21st) standards (68).
2.3 DNA extraction and methylation measurement and pre-processing
Saliva obtained at term equivalent age was collected in Oragene OG-575 Assisted Collection
kits, by DNA Genotek, and DNA extracted using prepIT.L2P reagent (DNA Genotek, Ontario,
Canada). DNA was bisulfite converted and methylation levels were measured using Illumina
HumanMethylationEPIC BeadChip (Illumina, San Diego, CA, USA) at the Edinburgh Clinical
Research Facility (Edinburgh, UK). The arrays were imaged on the Illumina iScan or HiScan
platform and genotypes were called automatically using GenomeStudio Analysis software
6
version 2011.1 (Illumina). Infants’ saliva samples were taken (for DNAm analysis) around the
same time as MRI acquisition. Details of DNAm pre-processing have been outlined previously
(63); for full details, refer to supplementary methods.
2.4 Inflammatory-related methylation signature
For each individual (n = 258), a weighted linear signature (DNAm CRP) was obtained by
multiplying the methylation proportion at a given CpG by the effect size from a previous
epigenome wide association study (EWAS) of CRP (69) (supplementary table 1), and then
summing these values. This method has been described previously (4648,70).
2.5 MRI acquisition
This study incorporates data from two phases of MRI acquisition which is reflected in the
flowchart of the study sample (supplementary figure 2). The data acquisition of this study has
been reported previously (63,66).
In the first phase (n = 93), structural and dMRI were performed in neonates using a MAGNETOM
Verio 3T clinical MRI scanner (Siemens Healthcare GmbH, Erlangen, Germany) and 12-channel
phased-array head coil. For dMRI, A protocol consisting of 11 baseline volumes (b = 0 s/mm2
[b0]) and 64 diffusion-weighted (b = 750 s/mm2) single-shot spin-echo echo planar imaging (EPI)
volumes acquired with 2 mm isotropic voxels (TR/TE 7300/106 ms) was used; 3D T1-weighted
(T1w) MPRAGE (TR/TE1650/2.43ms) with 1 mm isotropic voxels was acquired.
For the second phase (n=121), structural and dMRI were performed neonates using a
MAGNETOM Prisma 3T clinical MRI scanner (Siemens Healthcare GmbH, Erlangen, Germany)
and 16-channel phased-array pediatric head and neck coil. This was used to acquire dMRI in two
separate acquisitions: the first consisted of 8 b0 and 64 volumes with b = 750s/mm2; the second
consisted of 8 b0, 3 volumes with b = 200 s/mm2, 6 volumes with b = 500s/mm2 and 64 volumes
with b = 2500 s/mm2. An optimal angular coverage for the sampling scheme was applied (71). In
addition, an acquisition of 3 b0 volumes with an inverse phase encoding direction was
performed. All dMRI volumes were acquired using single-shot spin-echo planar imaging (EPI)
with 2-fold simultaneous multi-slice and 2-fold in-plane parallel imaging acceleration and 2 mm
isotropic voxels; all three diffusion acquisitions had the same parameters (TR/TE 3500/78.0ms).
Images affected by motion artifact were re-acquired multiple times as required; dMRI acquisitions
were repeated if signal loss was seen in 3 or more volumes. 3D T2-weighted SPACE images
(T2w) (TR/TE 3200/409 ms) with 1 mm isotropic voxels and 3D T1w MPRAGE (TR/TE 1970/4.69
ms) with 1 mm isotropic voxels were also acquired.
7
Infants were fed and wrapped and allowed to sleep naturally in the scanner without sedation.
Pulse oximetry, electrocardiography and temperature were monitored. Flexible earplugs and
neonatal earmuffs (MiniMuffs, Natus) were used for acoustic protection. All scans were
supervised by a doctor or nurse trained in neonatal resuscitation. Structural images were
reported by an experienced pediatric radiologist (A.J.Q), and each acquisition was inspected
contemporaneously for motion artefact and repeated if there had been movement while the baby
was still sleeping; dMRI acquisitions were repeated if signal loss was seen in 3 or more volumes.
As details on dMRI pre-processing have been previously outlined (72) please refer to
supplementary methods for specifics. T2w images from phase 2 were processed using the
dHCP pipeline (73). The T1w images from phase 1 were processed using specific software for
brain skull-stripping and tissue segmentation (74). The phase 1 pipeline relies on some atlases,
for these purposes, 10 subjects from the phase 2 that have both T1w and T2w were selected.
The volumes extracted include cortical grey matter, deep grey matter, white matter, hippocampi
and amygdalae, cerebellum, brainstem, cerebrospinal fluid (CSF) and ventricles.
From the diffusion images we calculated the tensor fractional anisotropy (FA), mean diffusivity
(MD), axial diffusivity (AD), and radial diffusivity (RD) and the NODDI (intracellular volume
fraction [NDI] maps) (75,76). All the subjects were registered to the Edinburgh Neonatal Atlas
(ENA50) using DTI-TK (75,77). The diffusion tensor derived maps of each subject (FA and MD)
were calculated after registration; NDI was then propagated to the template space using the
previously calculated transformations. The data was skeletonized using the ENA50 skeleton and
then multiplied by a custom mask. Finally, the peak width of the histogram of values computed
within the skeletonized maps was calculated as the difference between the 95th and 5th
percentiles (78). Global values of white matter microstructure reported in this study are the peak
width of skeletonised metrics (PSFA, PSMD, PSRD, PSAD, PSNDI), which have been derived
from the same pipeline previously used to characterise brain structural differences between
preterm and term infants (77).
2.6 Tract segmentation and extraction of tract-averaged dMRI metrics
As above, details of individual white matter tract segmentation and subsequent extract of tract-
averaged dMRI metrics have been outlined previously from infants from this study sample (79).
Briefly, FA and MD were derived for the left and right hemispheric tracts of the arcuate fasciculus
(AF), anterior thalamic radiation (ATR), cingulum cingulate gyrus (CCG), corticospinal tracts
(CST), inferior fronto-occipital fasciculus (IFOF), inferior longitudinal fasciculus (ILF), uncinate
fasciculus (UNC) and genu and splenium of the corpus callosum (CC).
8
2.7 Statistical Analysis
All statistical analyses were performed in R (version 4.0.5) (R Core Team, 2020).
2.7.1 Selection of covariates and confounding variables
In comparing participant demographics between term infants (n = 103) and preterm infants (n
=155), p-values derived from the t-test (for continuous variables) and Chi-square test or Fischer’s
Exact test if the count was below 5 (categorical variables) (Table 1) (80). In all models examining
the association between DNAm CRP with health outcomes and brain MRI metrics, we adjusted
for infant sex, gestational age at birth, gestational age at scan, and birthweight Z score. Scanner
variable was included when data included two phases of MRI acquisition. We also tested for an
interaction between gestational age × DNAm CRP and infant sex × DNAm CRP in sensitivity
analyses, where a significant interaction would indicate differences in association magnitudes at
different gestational ages / between males and females. To quantify the amount of variance in
each brain imaging biomarker accounted for by DNAm CRP, for all neuroimaging associations
we report both the adjusted R2 and the incremental R2 (81), the latter of which was calculated by
comparing the R2 of each model with that from a baseline model (reported as Model H0) R2 in
which the MRI measure was modelled with covariates only (e.g., Model H0 = MRI metric ~ sex +
birthweight + gestational age age + gestational age at scan + scanner).
After examining global associations, we wanted to control for variables that could either cause
the raised DNAm CRP (the exposure), variance in MRI metrics (the outcome), or both. We ran
bivariate correlations and from these we included all common correlates of the exposure and the
outcome in our second regression model (Model H2) to eliminate alternative explanations of the
outcome due to confounding (supplementary figure 1). In these models, neither antenatal
treatment of corticosteroids and MgSO4 for anticipated preterm birth were included to circumvent
issues of multicollinearity, since they were given to the majority of mothers (96% and 72%
respectively) in the preterm group and were highly correlated with preterm status (r = 0.71
0.93, p<0.001). The magnitude of effects are classified as small, medium, or large when the
standardized coefficients are 0.1, 0.3, or 0.5, respectively as classified by Cohen (82).
Finally, accounting for the fact that inflammatory risk factors are positively correlated, we
included all inflammatory risk factors in one multiple linear regression alongside DNAm CRP
signature for each MRI variable of interest. This allowed us to account for unique contribution of
DNAm CRP in the context of inflammatory-related exposures to variance in brain structural
outcomes.
9
2.7.2 Multiple inflammatory hits and DNAm CRP
We next performed investigations to assess whether DNAm was related to number of
inflammatory episodes experienced. Due to small numbers of individual inflammatory risk factors
and the frequent overlap of episodes experienced in the preterm group, we created binary
outcome measures based on combinations of inflammatory risk factors or conditions
experienced, combining infants that experienced three or more morbidities into a single group,
resulting in four possible levels for the risk score of 0, 1, 2, or 3 + alongside a term control
reference (0 inflammatory episodes). Results are presented firstly unadjusted (model H1), then
adjusted for gestational age at birth, infant sex and birthweight z-score (model H2), and then
adjusted for gestational age at birth, infant sex and birthweight z-score as well as administration
of MgSO4 and corticosteroids in pregnancy (model H3), given these have been identified as
potential confounders of the relationship between inflammation and health outcomes in previous
studies (8385). Results are presented as odds ratios (OR) and 95% confidence intervals (CI) for
categorical outcome measures.
2.7.3 DNAm CRP and global brain structure associations
To determine the effect of inflammation (DNAm CRP) on neuroimaging outcomes, data were
analysed using regression models, controlling for gestational age at birth, gestational age at
scan, infant sex, MRI scanner, and birthweight Z score. We aimed to contextualise these
associations with clinical health data. Inflammatory risk factors were added simultaneously as
covariates into a second model (model H2) in addition to the standard covariates of gestational
age at birth, gestational age at scan, birthweight Z score and sex (model H1). As no term infants
had postnatal inflammatory episodes (sepsis, NEC, ROP, BPD), analyses were stratified
according to term or preterm status. In models testing global brain structural metrics such as
brain volumes and PSMD and PSFA, MRI scanner was included as a binary covariate as MRI
data from both phases of data collection were included (refer to supplementary figure 2, study
sample flowchart). All continuous variables were standardised using z-score scaling to obtain
standardised effect sizes (β). P-values were corrected for multiple testing using the false
discovery rate (FDR) method and significance was deemed FDR corrected p-value (pFDR) <
0.05. 95% CIs are reported throughout.
2.7.4 DNAm CRP and dMRI White matter tract associations
dMRI measures of white matter appear to be highly correlated (e.g. high FA in an individual tract
such as the arcuate fasciculus is often accompanied by high FA across all other white matter
tracts in that individual), a property that persists from early infancy through to older age
(79,86,87). As a result of this, it is common to derive general factors (g-factors) of white matter
10
microstructure to characterise global white matter microstructure. One PCA was conducted for
FA and MD parameters across the 16 tracts to quantify the proportion of shared variance
between them; in each analysis, each subject was described by 16 features, computed as the
tract-averaged values of FA or MD across each tract (supplementary figure 5). The first
unrotated principal component (PC) scores were extracted as the single-metric g-factors, gFA
and gMD (scree plot and PCA variable contributions illustrated in supplementary figure 6).
As with global brain structural metrics, two models were used:
1. Model H1: dMRI metric ~ DNAm CRP + gestational age at scan + gestational age at birth
+ infant sex + birthweight.
2. Model H2: dMRI metric ~ DNAm CRP + gestational age at scan + gestational age at birth
+ infant sex + birthweight + all inflammatory risk factors
In comparison to global MRI volumetric metrics and PSFA, PSMD, PSAD and PSRD, all
individual tract associations, gFA, gMD and PSNDI were limited to neuroimaging data from
phase 2 of the study, hence no scanner variable was included in these analyses.
2.8 Data and code availability
Requests for original image and anonymised data will be considered through the BRAINS
governance process (www.brainsimagebank.ac.uk). Raw DNAm data are available upon request
from Theirworld Edinburgh Birth Cohort, University of Edinburgh
(https://www.tebc.ed.ac.uk/2019/12/data-access-and-collaboration), while DNAm and metadata
are not publicly available, generated DNAm CRP signatures are included alongside scripts for
data analysis. All brain volumetric metrics were obtained using the scripts provided in
https://github.com/amakropoulos/structural-pipeline-measures. The segmented tracts in the
ENA50 template space are available online: https://git.ecdf.ed.ac.uk/jbrl/ena. Code for primary
data analysis and figures are available at https://github.com/EleanorSC/TEBC_DNAmCRP and
code for tract propagation and average calculation are available at
https://git.ecdf.ed.ac.uk/jbrl/neonatal-gfactors.
11
3. Results
3.1 Participant characteristics
The study group consisted of 258 neonates: 155 participants were preterm and 103 were
controls born at full term, see Table 1 for participant characteristics and supplementary figure 1
for a flowchart of data acquisition. Among the preterm infants, 48 (31%) had bronchopulmonary
dysplasia, 10 (6%) developed necrotising enterocolitis, 8 (5%) developed ROP, 49 had HCA
(32%), 22 (15%) were born to women whose pregnancy was complicated by preeclampsia, and
36 (23%) had an episode of postnatal sepsis. Of the 258 participants with DNAm data, 214 also
had MRI data.
Characteristics & clinical features
Term infants
(n=103)
Preterm infants
(n=155)
P
value
Sex: Female (%)
44 (43)
75 (48)
0.2166
Gestational age at birth/weeks (range)
39.7 (37.00
42.14)
28.84 (23.28 34.84)
<0.001
Gestational age at scan/weeks (range)
42.27 (39.84
47.14)
40.56 (37.70 -45.14)
<0.001
Birth weight/g (range)
3482 (2346 4670)
1177 (500 2100)
<0.001
Birth weight z-score (range)
0.43 (-2.30 2.96)
-0.19 (-3.13 1.58)
<0.001
DNAm CRP (mean, SD)
-0.012 (0.001)
-0.011 (0.001)
<0.001
Maternal / fetal
Maternal age (years)
33.7 (1948)
31.1 (1744)
<0.001
Antenatal corticosteroid administration in
pregnancy, n (%)
2 (0.02)
148 (95.5)
<0.001
MgSO4 administration in pregnancy, n (%)
0 (0)
112 (72)
<0.001
Smoked during pregnancy (%)
2 (2)
29 (19)
<0.001
Histologic chorioamnionitis, n (%)
6 (0.06)
49 (31.6)
<0.001
Preeclampsia, n (%)
7 (0.07)
22 (14.2)
<0.001
Neonatal
Necrotizing enterocolitis, n (%)
0 (0)
10 (6)
N/A
Bronchopulmonary dysplasia, n (%)
0 (0)
48 (31)
N/A
Retinopathy of prematurity, n (%)
0 (0)
8 (5)
N/A
Sepsis, n (%)
0 (0)
36 (23)
N/A
Table 1. Demographic and clinical features of study sample (n=258). P values denote
significant difference between term and preterm groups.
12
3.2 Multiple inflammatory hits increase risk of elevated epigenetic inflammation
Preterm Infants in the sample for whom DNAm data and composite neonatal inflammatory risk
scores were available (n = 155), had high prevalence (n = 112, 72%) of experiencing at least one
of the documented inflammatory exposures (Figure 1B), which included incidence of smoking
during pregnancy, preeclampsia, HCA, sepsis, BPD, NEC or ROP. A small subset of these
infants experienced three or more of these exposures (n = 24, 15%).
There was an association between number of inflammatory episodes and the epigenetic
inflammation signature, with higher DNAm CRP in infants who had experienced greater
exposure to inflammation. DNAm CRP was associated with higher odds of several perinatal
morbidities including HCA, sepsis, BPD, and NEC. These relationships remained significant
following adjustment for gestational age at birth, birthweight, and infant sex as well as perinatal
variables of administration of corticosteroids and MgSO4 in pregnancy (Figure 1C,
supplementary table 3). The association of DNAm CRP with ROP was no longer significant
after controlling for MgSO4 and corticosteroid administration (model H3). DNAm CRP was also
associated with three or more inflammatory episodes. There was no significant association of
DNAm CRP with maternal smoking in pregnancy or preeclampsia. Infants with increasing
numbers of complications were more likely to have shorter gestational ages at birth and lower
birthweights (Figure 1D). Furthermore, preterm infants had significantly higher DNAm CRP
(Table 1, p < 0.001). When examining DNAm CRP alongside clinical inflammatory exposures
(supplementary table 4), there was no significant difference between term infants with no
inflammatory episodes vs those with one. The largest difference was found between term infants
with no inflammatory episodes and preterm infants with 3 or more inflammatory risk-factors (p <
0.001).
13
Figure 1. Multiple inflammatory hits associate with raised DNAm CRP (A) distributions of
DNAm CRP according to number of inflammatory episodes experienced by infant (B) Venn
diagram showing the overlap postnatal inflammatory morbidities in study sample (C) Odds ratios
and 95% confidence intervals for contribution of DNAm CRP to inflammatory exposures,
asterisks (*) indicate statistically significant (FDR-corrected p < 0.05) (D) Scatter plots of the
relationships between gestational age and birthweight, coloured according to number of
inflammatory episodes/exposures (top panel) and DNAm CRP (bottom panel).
1 inflammatory exposure
0 inflammatory exposures
2 inflammatory exposures
3+ inflammatory exposures
Term control
(0 inflammatory exposures)
A B
C D
3+ inflammatory exposures
2 inflammatory exposures
1 inflammatory exposure
term control
H2 adjusted OR model
DNAm CRP
+ gestational age at birth
+ birthweight (z-score)
+ infant sex
H1 OR model
DNAm CRP only
H3adjusted OR model
DNAm CRP
+ gestational age at birth
+ birthweight (z-score)
+ infant sex
+ MgSO4
+ corticosteroi ds
Models:
**
***
***
OR
Smoked in pregnancy
preeclampsia
HCA
Sepsis
BPD
NEC
***
***
***
*
*
ns
**
**
**
***
***
***
***
***
***
**
**
**
ns
ns
ns
ns
ns
ns
ns
ns
ns
ns
ns
ns
birthweight (g)
birthweight (g)
gestational age (weeks)
OR (95% CI)
14
3.3 DNAm CRP and brain volumes
Overall, magnitudes of associations between DNAm CRP and global MRI brain volumes were
modest, explaining a small amount of additional variance beyond covariates (of infant sex,
gestational age at birth, birthweight z-score, gestational age at scan, scanner variable). After
examining inflammation-brain structure associations across all infants (n = 214; supplementary
table 5), we stratified analyses into term (n = 87) and preterm (n = 127) subgroups to examine
group differences (Figure 2B). The incremental R2 upon adding DNAm CRP to a null model
varied by trait: global white matter volume = 3.7%, deep grey matter volume = 3.0%, hippocampi
and amygdalae volume = 2.7% and cerebellar volume = 1.6%. Visual inspection of diagnostic
plots suggested no regression assumptions were violated (an example is given in
supplementary figure 4). Term infants displayed no significant brain structural associations with
DNAm CRP, whereas preterm infants with higher DNAm CRP displayed brain volume reductions
in deep grey matter, white matter, and hippocampi and amygdalae (Figure 2B).
Figure 2. Association of DNAm CRP with brain volumes (A) schematic drawn by ggseg
package (Mowinckel & Vidal-Piñeiro, 2020) of reconstructed MRI brain volumetric measures (B)
Standardized regression coefficients for DNAm CRP associations between brain volumetric
measures for preterm infants (red circles) and preterm infants (blue circles). Points show
15
standardized coefficients and 95% confidence intervals. Asterisks (*) indicate statistically
significant (FDR-corrected p < 0.05). All models are controlled for infant sex, gestational age at
birth, gestational age at scan and birthweight Z score and scanner variable.
Analyses were repeated to include interactions between DNAm CRP and both sex and
gestational age (supplementary tables 6-9). While null findings were observed with the former
(p > 0.05; supplementary table 7), within the preterm cohort there was evidence for interactions
with gestational age at birth (supplementary table 8). Higher DNAm CRP was consistently
associated with lower brain volume in infants of lower gestational ages (i.e. extremely preterm
infants tended to have higher DNAm CRP and correspondingly smaller global brain volume
measures). In contrast, there was no significant interaction between gestational age and DNAm
CRP within the term sub-group (p > 0.05; supplementary table 9).
In fully adjusted models (Figure 3, supplementary table 10), there remained a significant
association of DNAm CRP with deep grey matter volume (β = -0.209, p = 0.008), white matter
volume (β = -0.304, p = 0.003), and cerebellum volume (β = -0.204, p = 0.013). For most brain
metrics, the strength of the association between DNAm CRP and MRI metric was increased
when additional inflammatory covariates were included in the model (percentage increase for
deep grey matter volume = 6%, white matter volume = 39%, and cerebellum volume = 27%).
Individually modelling risk factors revealed that this increase was mostly driven by controlling for
incidence of sepsis, whereas brain structural associations were most attenuated by controlling
for incidence of BPD (Figure 3).
16
Figure 3. Associations between DNAm CRP and brain volumes and the impact of
inflammatory risk factors on associations; Standardized regression coefficients for DNAm
CRP associations between brain volumetric measures for preterm infants (red circles) and
preterm infants (blue circles). Points show standardized coefficients and 95% confidence
intervals. All models are controlled for infant sex, gestational age at birth, gestational age at scan
and birthweight Z score and scanner variable; additional shapes show standardised regression
coefficients for different models (H2-H9, corresponding with supplementary table 10).
17
3.4 Global white matter microstructure associations with DNAm CRP
Preterm infants with higher DNAm CRP had poorer measures of white matter tract integrity. This
was seen at the global level for all peak width of skeletonised white matter microstructure metrics
(Table 2); PSFA, PSMD, PSRD, PSAD; β range |0.186| to |0.341|, incremental R2 2.7 9%). In
term infants, there were no significant associations (supplementary table 10).
As with global brain volumetric measures, there were significant interactions between gestational
age and DNAm CRP across measures of white matter integrity excepting PSRD: PSFA
(interaction β  =  0.225; main effect β  =  -0.212), PSMD (interaction β  =  -0.257; main effect β  = 
0.371) and PSAD (interaction β  =  -0.271; main effect β  =  0.232), indicating infants at younger
gestational ages were more likely to have poor white matter integrity with high DNAm CRP.
When controlling for inflammatory risk factors, DNAm CRP associations between PSRD and
PSAD were no longer significant.
Within a smaller subgroup of this sample, individual tract FA and MD as well as neurite density
index data was available (Phase 2, refer to supplementary figure 2 for study flow diagram).
PCA-derived single-metric g-factors, gFA and gMD (scree plot and PCA variable contributions
illustrated in supplementary figure 6) were almost exactly correlated with those previously
reported in a larger sample of Theirworld Edinburgh Birth cohort infants (79). When examining
the association between DNAm CRP and global white matter measures in this subsample, the
most striking association was seen with differences in a general factor of fractional anisotropy,
gFA (β = -0.52 [95% CI -0.304, -0.736], p = 1.48 x 10-5, incremental R2 = 22%) and mean
diffusivity, gMD (β =0.423 [95% CI 0.661, 0.191], p = 7.79 x 10-4, incremental R2 = 14%). These
effect sizes were attenuated by controlling for additional inflammatory risk factors (model H2) but
remained significant (β range |0.35| to |0.37|, p < 0.05). No significant associations were found
between DNAm CRP with PSNDI.
beta
lower CI
p
r2
additional r2
n
PSFA
-0.186
-0.324
0.009
0.540
0.027
127
PSMD
0.341
0.166
2.17E-04
0.256
0.090
127
PSRD
0.312
0.122
0.002
0.130
0.075
127
PSAD
0.201
0.030
0.023
0.294
0.031
127
gFA*
-0.520
-0.736
1.48E-05
0.441
0.216
64
gMD*
0.426
0.191
0.001
0.333
0.145
64
PSNDI*
-0.089
-0.321
0.452
0.354
0.006
64
PSFA
-0.215
-0.375
0.009
0.577
0.026
127
PSMD
0.206
0.009
0.042
0.357
0.024
127
PSRD
0.175
-0.041
0.115
0.222
0.017
127
18
PSAD
0.093
-0.095
0.336
0.414
0.005
127
gFA*
-0.371
-0.617
0.005
0.573
0.073
64
gMD*
0.355
0.082
0.014
0.477
0.067
64
PSNDI*
-0.123
-0.419
0.420
0.385
0.008
64
Table 2. Associations between DNAm CRP and global white matter microstructure and
the impact of inflammatory risk factors on associations in preterm infants; standardized
regression coefficients for DNAm CRP associations between global white matter microstructure
metrics for preterm infants. Betas (standardized coefficients) and 95% confidence intervals are
reported. Bold text indicate statistically significant association (FDR-corrected p < 0.05). Model
H1 controls for infant sex, gestational age at birth, gestational age at scan birthweight Z score
and scanner variable; model H2 additionally controls for inflammatory risk factors and associated
morbidities (maternal smoking in pregnancy, preeclampsia, HCA, sepsis, BPD, NEC and ROP)
*scanner variable is controlled for when examining PS metrics but not gFA, gMD and PSNDI
(single-scanner sample).
3.5 Individual white matter tract associations with DNAm CRP
We next examined associations between DNAm CRP and individual tract-averaged FA and MD.
In all models, term infants displayed no significant tract associations with DNAm CRP
(supplementary figure 7). In preterm infants, altered FA was present in both hemispheric tracts
of the AF, CST, IFOF, ILF, UNC and CCG (Figure 4 shows tract-averaged fractional anisotropy
for each of the 16 tracts for the term and preterm neonates). Some tract associations were
specific to hemisphere such as decreased FA in the left (but not right) ATR. Equally, altered FA
and MD was present in only the genu (and not splenium) of the corpus callosum. A breakdown of
all dMRI results is reported in supplementary table 11. However, after adjusting for additional
inflammatory risk factors, (in order of effect size) only FA in the right corticospinal tract (15.4%),
right AF (10%), and right CCG (8.7%) remained significant. For tract MD, bilateral increases in
MD were observed in the AF, CST, IFOF, ATR and CCG. Hemispheric specific associations
were found for the left ILF, left ATR and genu of the corpus callosum. Of these associations, AF
and ILF were no longer significant when accounting for additional inflammatory exposures.
19
Figure 4. DTI-tract associations with DNAm CRP. Standardized regression coefficients for
DNAm CRP associations between tract fractional anisotropy (FA) for preterm infants (squares),
preterm infants in models controlling for additionally inflammatory risk factors (circles) and term
infants (triangles). Filled shapes are left tracts and open shapes are right hemispheric tracts,
except in the case of the CC where filled shapes are the splenium and open shapes are the genu
of the corpus callosum. Points show standardized coefficients and 95% confidence intervals. All
models are controlled for sex, gestational age at birth, gestational age at scan and birthweight Z
score. Model H2 (circles) additionally controls for inflammatory risk factors (maternal smoking
during pregnancy, preeclampsia, HCA, neonatal sepsis, BPD, NEC, ROP). For MD associations
see supplementary figure 7.
20
4. Discussion
In this study, we integrated data from placenta, saliva and brain MRI in a large cohort of 258
infants to characterise the association of inflammation with brain structure. We demonstrate that
a composite buccal-cell DNA methylation measure of inflammation trained in adult peripheral
blood samples associates with comorbidities of preterm birth that are characterised by a pro-
inflammatory state and widespread differences in brain structure among preterm infants. The
epigenetic signature was particularly associated with white matter dysmaturation at term-
equivalent age both globally and at the level of individual white matter tract microstructure,
associations that largely remained significant when accounting for inflammatory exposures.
Using this epigenetic signature which has previously tracked with inflammation and brain
structural alterations in older age human cohorts, these data motivate further research into the
potential of immune-DNAm markers for translational medicine in the neonatal period as
diagnostic tools for identifying those at risk for inflammatory-related morbidities and
neurodevelopmental impairment.
4.1 Inflammatory-related DNAm biomarkers
There has been increasing interest in using methylation data to advance our understanding of
the causes and consequences of preterm birth as outlined in several reviews (10,8891). Only
recently has attention turned to exposures in the perinatal period, the role of epigenetics in utero
for neurodevelopment, and the potential of peripherally sampled DNAm to capture the impact of
environmental exposure in relation to brain and cognitive outcomes (92). Among these
developments are the use of poly-epigenetic scores of exposure, which integrate information
from multiple CpG sites to provide a record of exposure or to capture a complex trait (93). This
approach has been used to examine maternal smoking (9497) glucocorticoid, and prenatal
folate exposure during pregnancy (55,98), alongside environmental exposures such as pollution
(98). Examining DNAm proxies of inflammation is uncommon, but given the health associations
with inflammatory-related DNAm in adult cohorts (47,48,99), and the shared nature of epigenetic
changes between mother and infants (64,100), we hypothesised that variance in the neonatal
methylome could reflect a convergence of amassed inflammatory burden from different perinatal
origins.
4.2 DNAm CRP associates with gestational age and multiple inflammatory exposures
Our findings suggest that epigenetics offers a solution to the traditional limitations of assessing
inflammatory burden in infancy. Preterm infants displayed higher DNAm CRP than term infants,
and associations between DNAm CRP and postnatal health and brain outcomes were restricted
21
to the preterm infants. This novel finding that gestational age at birth correlates strongly with
epigenetic inflammation (r = -0.62, p <0.001) aligns with previous observations of elevated
inflammatory protein concentrations with lower gestational ages and prematurity (8,15), lending
further weight to the validity of this measure for carrying clinical significance. Equally, finding that
inflammation-related alterations in brain structure were reserved to preterm infants (and no
trends were seen in term infants) is likely because of the rapid developmental changes during
the second and third trimester of pregnancy for both the developing innate immune system and
brain in particular, the disruptive impact of inflammation on neurogenesis, neuronal migration,
synaptogenesis and myelination (3). As these processes are highly dynamic during these
periods and early postnatal life, preterm infants are both more susceptible to sustained
inflammation and neurodevelopmental disruption the mechanisms of which we outline below.
Our finding that multiple inflammatory hits contributed to higher DNAm CRP strengthens the
hypothesis that DNAm may index the allostatic load of inflammation during neonatal intensive
care. In both preclinical studies and cohort groups, preterm infants with multiple inflammatory
episodes or morbidities display an increased risk for brain structural abnormalities compared to
infants who had only one inflammatory episode or condition recorded (101103). The multi-hit
hypothesis of sustained inflammation (14,26,101) suggests that postnatal health complications
related to preterm birth can perpetuate a chronic inflammatory state, with timing of insults a key
factor for why preterm infants are more susceptible than term infants to sustained inflammation
(104). This could affect DNA methylation in the immune system, with studies demonstrating that
lower birthweight infants go on to display higher concentrations of CRP in adolescence and
adulthood (105,106), and that adversity-related changes in immune cell DNAm are related to
raised plasma inflammatory mediators (107). Epigenetic modifications are an essential
mechanism by which inflammatory risk factors could lead to long-term disruptions in both
immune and brain development (54,108), and this work highlights the utility of profiling such
changes alongside other clinical and biological data.
4.3 White and deep grey matter dysmaturation in preterms with elevated
inflammation
The most striking finding from this study is the association of DNAm CRP with widespread
variances in brain structure in preterm but not term infants (Figures 2-4). We observe larger
effect sizes for associations of DNAm CRP with global white matter microstructure (gFA and
gMD) than white matter volume in a sub-population of these infants. This may be because
diffuse white matter injury antedates overall reductions in white matter volume, with DNAm CRP-
DTI associations capturing a more subtle dysmaturation of programmed development (109).
Both global white matter volume, microstructure and regional white matter integrity were lower in
preterms with elevated DNAm CRP, with infants at younger gestational ages more prone to
22
elevated inflammation and related poor white matter integrity. These findings echo the results of
prior studies (25,33,110,111), and are overall consistent with the theory that alterations in white
matter microstructure are largely a consequence of dysregulation of white matter development
driven by inflammation (111).
In addition to white matter, volume reductions were observed in the hippocampi and amygdale
and deep grey matter with increased DNAm CRP, though the former did not remain significant
after accounting for additional inflammatory risk factors (supplementary table 10). The
association of elevated DNAm CRP with lower deep grey matter volume is consistent with
previous research that finds that preterms infants exhibit deep grey matter loss relative to term
infants (112114). Given the relationship we outline here between preterm birth and
inflammatory load, inflammation may be a key driver of these differences, both via its direct
effects on brain structure and its contribution to related damage such as sensitisation to hypoxia
ischaemia, excitotoxic insults and other early-life stressors (2,115). These widespread alterations
in brain structure are particularly interesting given the evidence base for inflammation relating to
cognitive impairment, as studies have shown that both hippocampal volume and thalamic volume
loss accompanying white matter microstructural alterations are linked to neurodevelopmental
outcomes in early childhood (116118). Inflammation-related grey matter loss is considered a
consequence of dysregulated neuronal development, with inflammatory mediators disrupting
processes such as dendritic arborization and cortico-thalamic connectivity (110). As
consolidation of thalamocortical connections happens in the third trimester of pregnancy, deep
grey matter structures may be vulnerable to inflammatory stimuli (119).
We also observed regional variance in how DNAm CRP associates with white matter tracts, a
finding consistent with previous studies that indicate that certain white matter tracts are more
vulnerable to inflammatory-adjacent events such as hypoxia ischemia (120,121) traumatic brain
injury (9), intraventricular haemorrhage (122) and cerebral palsy (5). Different white matter tracts
develop at different rates in utero and display distinct transient growth periods of increased
axonal development. These windows of plasticity have been outlined as particularly vulnerable to
perturbation (123,124), with inflammation disrupting the developmental lineage of
oligodendrocytes, resulting in hypomyelinated axons (1,110,125,126). Developmental growth
periods of certain white matter tracts may therefore underscore regional vulnerability to elevated
inflammation, with younger tracts likely to have higher proportions of pre-myelinating
oligodendrocytes vulnerable to inflammatory mediators. However, we caution that we lack the
statistical power to reliably detect differences between the magnitude of associations in regional
white matter structure, and instead interpret these findings as evidence of the pervasive and
widespread impact of inflammation on the development of white matter. Correspondingly, though
there were differences between the association significance for the left and right hemispheres for
23
several of the delineated tracts, these unilateral findings are in keeping with previous studies of
similar sample size (4,127); as the magnitudes were similar (with overlapping confidence
intervals), this did not indicate a strong basis for laterality of effects.
4.4 Strengths and limitations
To our knowledge, this is the first time an epigenetic measure of inflammation has been
examined in a preterm cohort in relation to brain health outcomes. The effect sizes reported in
this study are consistent with that of previous epidemiologic studies of DNAm and early life
outcomes (128). The sample size (n = 258 for inflammatory exposure, and n = 121-214 for
neuroimaging associations), is akin to that of previous work examining inflammation and brain
structure in preterm infant populations (6,11,36,37,129131), and in many cases more
substantial, with the vast majority of prior work conducted in sample sizes of less than 100
infants (4,19,21,3235,127,132). There is a distinct scarcity of detailed methylation alongside
multi-modal neuroimaging data (133,134), particularly in neonatal cohorts (61,62,135), making
this a valuable contribution to the DNAm-neuroimaging field.
Although the weights for the predictor were trained in adult blood samples, we observed similar
associations between DNAm CRP and brain structural outcomes to those in previous studies of
adults (47,48). Given we have now applied this method to buccal-cell DNAm, it is encouraging to
see similar associations between DNAm CRP and brain structural outcomes, especially given
that DNAm is highly tissue specific (136), and previous research has reported on cross-tissue
differences in magnitude and direction of effects for other traits (137). This cross-tissue approach
(where weights were originally created from blood-based DNAm, and later applied to saliva-
based composite signatures) has also been adopted in other studies (98,138). While future
studies would ideally measure CRP directly from serum or blood spots in infants to enable direct
cross-tissue comparisons, saliva has the advantage of being one of the most accessible tissue
samples for infant populations, and may be more suitable than other peripheral samples (such as
blood) when examining brain and cognitive outcomes owing to the brain and buccal cell shared
ectodermal origins (139142). In the absence of direct comparison with inflammatory mediators
from blood samples, the strong correlates with clinical inflammatory conditions (both fetal and
postnatal) is affirming, and we have taken steps to account for possible sources of confounding
(supplementary figure 1).
Neuroimaging studies are notoriously heterogenous in their design given the array of different
MRI acquisition techniques, processing pipelines and chosen outcome measures. The choice of
neuroimaging features is even more relevant in the context of preterm birth to adequately
address the motivating research questions (38). Here, our choice of neuroimaging features was
24
guided by established characterizations of EoP in preterm infants, namely water content and
dendritic/axonal complexity and dysmaturation within the white matter, and grey matter volume
(77,124). While we consider this comprehensive characterisation of brain structure from NODDI
and dMRI data a significant strength of this study, we acknowledge that microstructure measures
such as FA and MD in older cohorts are commonly considered surrogates of white matter
integrity or myelination, the white-matter pathways in this study are still developing at the time of
gestational age at scan (range 37.70 - 45.14 weeks), and as such may not reflect permanent
differences. Longitudinal follow up is therefore encouraged for future studies designed to
examine the implications of sustained inflammation in preterms for neurodevelopmental
outcomes and lifecourse brain health.
We do not attempt to discuss the causality of the relationship between DNAm CRP with brain
structure, though the causality of such associations is a persistent topic of debate in epigenetic
epidemiological research and has been discussed in depth in reviews (143). Future work
examining transcriptomic changes on the same peripheral samples from which DNAm data is
collected, as well as statistical approaches like two-step mendelian randomization, are important
developments to unpick causality of these relationships. Studies investigating whether these
differences in DNAm remain, amplify, or attenuate with age are advised, as well as how sensitive
these signatures are in the context of intervention (anti-inflammatory medications and
treatments, as well as lifestyle interventions such as the cessation of smoking in pregnancy).
There is also precedent to examine whether composite methylation proxies of inflammation differ
across psychopathology (144) or specific neurological cases such as Cerebral Palsy or
neurodevelopmental disorders such as Fragile X syndrome, autism and ADHD, given examples
of other poly-epigenetic signatures of psychiatric disorders (145). Future work that focuses on
such DNAm dynamics in relation to these outcomes in ongoing longitudinal studies of infants
born preterm is therefore of interest, as well as replication in different population samples.
Finally, DNAm was sampled in neonates postnatally. While this is rational when examining
variances in DNAm and brain structural differences in infants, future studies that examine both
maternal and infant DNAm could examine the degree to which exposures are shared or specific
to parent and offspring. Equally, multiple DNAm sampling during pregnancy could elucidate key
critical periods of susceptibility to inflammation by parsing out exposures specific to trimester or
months of pregnancy, affording new insights into the spatiotemporal patterning of brain
development in relation to dynamic immune changes in the perinatal period.
25
5. Conclusion
Inflammatory-related DNAm is associated with risk of postnatal health outcomes and brain
dysmaturation. Our results indicate that multiple inflammatory-related hits from different origins
(pertaining to maternal, fetal, and postnatal exposures) may be captured by changes to the DNA
methylation profiles of infants and may help to explain variances in brain structure in preterm
populations, circumventing limitations of traditional measures of inflammation. As early birth is
associated with sudden change in immune-related risks, which coincide with the developing
immune system and windows of neurodevelopmental plasticity, it is theorised that preterm
infants are at greater risk of inflammation-related disruption of white matter. Our work here
provides new layers to this theory, with epigenetic inflammation associating with diffuse and
global brain and particularly white matter alterations in preterm but not term infants, indicating
that sustained inflammation may be a key driver of neurodevelopmental disruption. In summary,
the association of an epigenetic signature with inflammatory outcomes, and inflammation-
relevant neural phenotypes, supports the use of methylation data in integrative, multimodal
approaches toward disease stratification in the perinatal period.
6. Funding
This research was funded in whole, or in part, by the Wellcome Trust (grant numbers
108890/Z/15/Z and 221890/Z/20/Z). For the purpose of open access, the authors have applied a
CC BY public copyright licence to any Author Accepted Manuscript version arising from this
submission.
7. Acknowledgements
Some of the participants were scanned in the University of Edinburgh Imaging Research MRI
Facility at the Royal Infirmary of Edinburgh which was established with funding from The
Wellcome Trust, Dunhill Medical Trust, Edinburgh and Lothians Research Foundation,
Theirworld, The Muir Maxwell Trust and other sources. We thank Thorsten Feiweier at Siemens
Healthcare for collaborating with dMRI acquisitions (Works-in-Progress Package for Advanced
EPI Diffusion Imaging). The authors are grateful to the families who consented to take part in the
study and to all the University’s imaging research staff for providing the infant scanning.
8. Declaration of interests
R.E.M has received a speaker fee from Illumina and is an advisor to the Epigenetic Clock
Development Foundation and Optima Partners
26
9. References
1. Back SA. Brain injury in the preterm infant: new horizons for pathogenesis and prevention.
Pediatric neurology. 2015;53(3):18592.
2. Bennet L, Dhillon S, Lear CA, van den Heuij L, King V, Dean JM, et al. Chronic inflammation
and impaired development of the preterm brain. Journal of Reproductive Immunology. 2018 Feb
1;125:4555.
3. Hagberg H, Mallard C, Ferriero DM, Vannucci SJ, Levison SW, Vexler ZS, et al. The role of
inflammation in perinatal brain injury. Nature Reviews Neurology. 2015;11(4):192208.
4. Inomata K, Mizobuchi M, Tanaka S, Iwatani S, Sakai H, Yoshimoto S, et al. Patterns of
increases in interleukin-6 and C-reactive protein as predictors for white matter injury in preterm
infants. Pediatrics International. 2014;56(6):8515.
5. Lin C, Chang Y, Wang S, Lee T, Lin C, Huang C. Altered inflammatory responses in preterm
children with cerebral palsy. Annals of neurology. 2010;68(2):20412.
6. Shah DK, Doyle LW, Anderson PJ, Bear M, Daley AJ, Hunt RW, et al. Adverse
neurodevelopment in preterm infants with postnatal sepsis or necrotizing enterocolitis is
mediated by white matter abnormalities on magnetic resonance imaging at term. The Journal of
pediatrics. 2008;153(2):1705.
7. Stoll BJ, Hansen NI, Adams-Chapman I, Fanaroff AA, Hintz SR, Vohr B, et al.
Neurodevelopmental and growth impairment among extremely low-birth-weight infants with
neonatal infection. Jama. 2004;292(19):235765.
8. Humberg A, Fortmann I, Siller B, Kopp M, Herting E, Göpel W, et al. Preterm birth and
sustained inflammation: consequences for the neonate. Seminars in Immunopathology. 2020;
42(4): 451468.
9. Malaeb S, Dammann O. Fetal inflammatory response and brain injury in the preterm newborn.
Journal of child neurology. 2009;24(9):111926.
10. Reiss JD, Peterson LS, Nesamoney SN, Chang AL, Pasca AM, Marić I, et al. Perinatal infection,
inflammation, preterm birth, and brain injury: A review with proposals for future investigations.
Experimental Neurology. 2022 May 1;351:113988.
11. Kelly CE, Cheong JLY, Gabra Fam L, Leemans A, Seal ML, Doyle LW, et al. Moderate and late
preterm infants exhibit widespread brain white matter microstructure alterations at term-
equivalent age relative to term-born controls. Brain Imaging and Behavior. 2016 Mar 1;10(1):41
9.
12. Favrais G, Van De Looij Y, Fleiss B, Ramanantsoa N, Bonnin P, Stoltenburg-Didinger G, et al.
Systemic inflammation disrupts the developmental program of white matter. Annals of
neurology. 2011;70(4):55065.
13. Boardman JP, Counsell SJ. Invited Review: Factors associated with atypical brain development
in preterm infants: insights from magnetic resonance imaging. Neuropathology and Applied
Neurobiology. 2020 Aug 1;46(5):41321.
14. Leviton A, Fichorova RN, O’Shea TM, Kuban K, Paneth N, Dammann O, et al. Two-hit model of
brain damage in the very preterm newborn: small for gestational age and postnatal systemic
inflammation. Pediatr Res. 2012/12/07 ed. 2013 Mar;73(3):36270.
15. Dammann O, Leviton A. Intermittent or sustained systemic inflammation and the preterm brain.
Pediatric research. 2014;75(3):37680.
27
16. Chahal N, McLain AC, Ghassabian A, Michels KA, Bell EM, Lawrence DA, et al. Maternal
Smoking and Newborn Cytokine and Immunoglobulin Levels. Nicotine Tob Res. 2017 Jul
1;19(7):78996.
17. Sullivan G, Galdi P, Borbye-Lorenzen N, Stoye DQ, Lamb GJ, Evans MJ, et al. Preterm Birth Is
Associated With Immune Dysregulation Which Persists in Infants Exposed to Histologic
Chorioamnionitis. Frontiers in Immunology [Internet]. 2021;12. Available from:
https://www.frontiersin.org/articles/10.3389/fimmu.2021.722489
18. Dammann O, Allred EN, Fichorova RN, Kuban K, O’Shea TM, Leviton A, et al. Duration of
Systemic Inflammation in the First Postnatal Month Among Infants Born Before the 28th Week
of Gestation. Inflammation. 2016 Apr 1;39(2):6727.
19. Yoon BH, Jun JK, Romero R, Park KH, Gomez R, Choi JH, et al. Amniotic fluid inflammatory
cytokines (interleukin-6, interleukin-1β, and tumor necrosis factor-α), neonatal brain white matter
lesions, and cerebral palsy. American journal of obstetrics and gynecology. 1997;177(1):1926.
20. Backes CH, Markham K, Moorehead P, Cordero L, Nankervis CA, Giannone PJ. Maternal
Preeclampsia and Neonatal Outcomes. Lewis DF, editor. Journal of Pregnancy. 2011 Apr
4;2011:214365.
21. Anblagan D, Pataky R, Evans MJ, Telford EJ, Serag A, Sparrow S, et al. Association between
preterm brain injury and exposure to chorioamnionitis during fetal life. Scientific Reports. 2016
Dec 1;6:37932.
22. Han X, Ghaemi MS, Ando K, Peterson LS, Ganio EA, Tsai AS, et al. Differential dynamics of the
maternal immune system in healthy pregnancy and preeclampsia. Frontiers in immunology.
2019;1305.
23. Bassler D, Stoll BJ, Schmidt B, Asztalos EV, Roberts RS, Robertson CM, et al. Using a count of
neonatal morbidities to predict poor outcome in extremely low birth weight infants: added role of
neonatal infection. Pediatrics. 2009;123(1):3138.
24. Singh JK, Wymore EM, Wagner BD, Thevarajah TS, Jung JL, Kinsella JP, et al. Relationship
between severe bronchopulmonary dysplasia and severe retinopathy of prematurity in
premature newborns. Journal of American Association for Pediatric Ophthalmology and
Strabismus. 2019;23(4):209-e1.
25. Korzeniewski SJ, Romero R, Cortez J, Pappas A, Schwartz AG, Kim CJ, et al. A ‘multi-hit’
model of neonatal white matter injury: cumulative contributions of chronic placental
inflammation, acute fetal inflammation and postnatal inflammatory events. J Perinat Med. 2014
Nov;42(6):73143.
26. Barnett ML, Tusor N, Ball G, Chew A, Falconer S, Aljabar P, et al. Exploring the multiple-hit
hypothesis of preterm white matter damage using diffusion MRI. NeuroImage: Clinical.
2018;17:596606.
27. Suleri A, Blok E, Durkut M, Rommel AS, Witte L de, Jaddoe V, et al. The long-term impact of
elevated C-reactive protein levels during pregnancy on brain morphology in late childhood.
Brain, Behavior, and Immunity. 2022 Jul 1;103:6372.
28. Andrews WW, Cliver SP, Biasini F, Peralta-Carcelen AM, Rector R, Alriksson-Schmidt AI, et al.
Early preterm birth: association between in utero exposure to acute inflammation and severe
neurodevelopmental disability at 6 years of age. American Journal of Obstetrics & Gynecology.
2008 Apr 1;198(4):466.e1-466.e11.
29. Dubner SE, Dodson CK, Marchman VA, Ben-Shachar M, Feldman HM, Travis KE. White matter
microstructure and cognitive outcomes in relation to neonatal inflammation in 6-year-old children
born preterm. NeuroImage: Clinical. 2019;23:101832.
28
30. Kuban KCK, Joseph RM, O’Shea TM, Heeren T, Fichorova RN, Douglass L, et al. Circulating
Inflammatory-Associated Proteins in the First Month of Life and Cognitive Impairment at Age 10
Years in Children Born Extremely Preterm. The Journal of Pediatrics. 2017 Jan 1;180:116-
123.e1.
31. O’Shea TM, Shah B, Allred EN, Fichorova RN, Kuban KC, Dammann O, et al. Inflammation-
initiating illnesses, inflammation-related proteins, and cognitive impairment in extremely preterm
infants. Brain, behavior, and immunity. 2013;29:10412.
32. Travis KE, Adams JN, Ben-Shachar M, Feldman HM. Decreased and Increased Anisotropy
along Major Cerebral White Matter Tracts in Preterm Children and Adolescents. PLOS ONE.
2015 Nov 11;10(11):e0142860.
33. Dubner SE, Dodson CK, Marchman VA, Ben-Shachar M, Feldman HM, Travis KE. White matter
microstructure and cognitive outcomes in relation to neonatal inflammation in 6-year-old children
born preterm. NeuroImage: Clinical. 2019 Jan 1;23:101832.
34. Lee ES, Kim EK, Shin S han, Choi YH, Jung YH, Kim SY, et al. Factors associated with
neurodevelopment in preterm infants with systematic inflammation. BMC Pediatrics. 2021 Mar
8;21(1):114.
35. Basu S, Agarwal P, Anupurba S, Shukla R, Kumar A. Elevated plasma and cerebrospinal fluid
interleukin-1 beta and tumor necrosis factor-alpha concentration and combined outcome of
death or abnormal neuroimaging in preterm neonates with early-onset clinical sepsis. Journal of
Perinatology. 2015;35(10):85561.
36. Sullivan G, Galdi P, Cabez MB, Borbye-Lorenzen N, Stoye DQ, Lamb GJ, et al. Interleukin-8
dysregulation is implicated in brain dysmaturation following preterm birth. Brain, Behavior, and
Immunity. 2020 Nov 1;90:3118.
37. Wu Y, Zhang H, Wang C, Broekman BFP, Chong YS, Shek LP, et al. Inflammatory modulation
of the associations between prenatal maternal depression and neonatal brain.
Neuropsychopharmacology. 2021 Jan 1;46(2):4707.
38. Korom M, Camacho MC, Filippi CA, Licandro R, Moore LA, Dufford A, et al. Dear reviewers:
Responses to common reviewer critiques about infant neuroimaging studies. Developmental
Cognitive Neuroscience. 2022 Feb 1;53:101055.
39. Dimitrova R, Pietsch M, Christiaens D, Ciarrusta J, Wolfers T, Batalle D, et al. Heterogeneity in
Brain Microstructural Development Following Preterm Birth. Cerebral Cortex. 2020 Jul
30;30(9):480010.
40. Brown J, Meader N, Cleminson J, McGuire W. C-reactive protein for diagnosing late-onset
infection in newborn infants. Cochrane Database of Systematic Reviews [Internet]. 2019;(1).
Available from: https://doi.org//10.1002/14651858.CD012126.pub2
41. Bower JK, Lazo M, Juraschek SP, Selvin E. Within-Person Variability in High-Sensitivity C-
Reactive Protein. Archives of Internal Medicine. 2012 Oct 22;172(19):151921.
42. DeGoma EM, French B, Dunbar RL, Allison MA, Mohler ER 3rd, Budoff MJ. Intraindividual
variability of C-reactive protein: the Multi-Ethnic Study of Atherosclerosis. Atherosclerosis.
2012/07/20 ed. 2012 Sep;224(1):2749.
43. Chiesa C, Natale F, Pascone R, Osborn JF, Pacifico L, Bonci E, et al. C reactive protein and
procalcitonin: Reference intervals for preterm and term newborns during the early neonatal
period. Clinica Chimica Acta. 2011 May 12;412(11):10539.
44. Macallister K, Smith-Collins A, Gillet H, Hamilton L, Davis J. Serial C-Reactive Protein
Measurements in Newborn Infants without Evidence of Early-Onset Infection. Neonatology.
2019;116(1):8591.
29
45. Stevenson AJ, McCartney DL, Harris SE, Taylor AM, Redmond P, Starr JM, et al. Trajectories of
inflammatory biomarkers over the eighth decade and their associations with immune cell profiles
and epigenetic ageing. Clin Epigenetics. 2018 Dec 20;10(1):159159.
46. Stevenson AJ, McCartney DL, Hillary RF, Campbell A, Morris SW, Bermingham ML, et al.
Characterisation of an inflammation-related epigenetic score and its association with cognitive
ability. Clinical Epigenetics. 2020 Jul 27;12(1):113.
47. Conole ELS, Stevenson AJ, Muñoz Maniega S, Harris SE, Green C, Valdés Hernández M del
C, et al. DNA Methylation and Protein Markers of Chronic Inflammation and Their Associations
With Brain and Cognitive Aging. Neurology. 2021 Dec 7;97(23):e2340.
48. Green C, Shen X, Stevenson AJ, Conole ELS, Harris MA, Barbu MC, et al. Structural brain
correlates of serum and epigenetic markers of inflammation in major depressive disorder. Brain,
Behavior, and Immunity. 2021 Feb 1;92:3948.
49. Martino DJ, Tulic MK, Gordon L, Hodder M, Richman TR, Metcalfe J, et al. Evidence for age-
related and individual-specific changes in DNA methylation profile of mononuclear cells during
early immune development in humans. Epigenetics. 2011;6(9):108594.
50. Spiers H, Hannon E, Schalkwyk LC, Smith R, Wong CC, O’Donovan MC, et al. Methylomic
trajectories across human fetal brain development. Genome research. 2015;25(3):33852.
51. Takizawa T, Nakashima K, Namihira M, Ochiai W, Uemura A, Yanagisawa M, et al. DNA
methylation is a critical cell-intrinsic determinant of astrocyte differentiation in the fetal brain.
Developmental cell. 2001;1(6):74958.
52. Fagiolini M, Jensen CL, Champagne FA. Epigenetic influences on brain development and
plasticity. Current opinion in neurobiology. 2009;19(2):20712.
53. Massaro AN, Bammler TK, MacDonald JW, Perez KM, Comstock B, Juul SE. Whole genome
methylation and transcriptome analyses to identify risk for cerebral palsy (CP) in extremely low
gestational age neonates (ELGAN). Scientific reports. 2021;11(1):110.
54. Ozanne SE, Constância M. Mechanisms of disease: the developmental origins of disease and
the role of the epigenotype. Nature clinical practice Endocrinology & metabolism.
2007;3(7):53946.
55. Suarez A, Lahti J, Lahti-Pulkkinen M, Girchenko P, Czamara D, Arloth J, et al. A polyepigenetic
glucocorticoid exposure score at birth and childhood mental and behavioral disorders.
Neurobiology of Stress. 2020 Nov 1;13:100275.
56. Konwar C, Price EM, Wang LQ, Wilson SL, Terry J, Robinson WP. DNA methylation profiling of
acute chorioamnionitis-associated placentas and fetal membranes: insights into epigenetic
variation in spontaneous preterm births. Epigenetics & Chromatin. 2018 Oct 29;11(1):63.
57. Liu Y, Hoyo C, Murphy S, Huang Z, Overcash F, Thompson J, et al. DNA methylation at imprint
regulatory regions in preterm birth and infection. American journal of obstetrics and gynecology.
2013;208(5):395-e1.
58. Merid SK, Novoloaca A, Sharp GC, Küpers LK, Kho AT, Roy R, et al. Epigenome-wide meta-
analysis of blood DNA methylation in newborns and children identifies numerous loci related to
gestational age. Genome Medicine. 2020 Mar 2;12(1):25.
59. Sparrow S, Manning JR, Cartier J, Anblagan D, Bastin ME, Piyasena C, et al. Epigenomic
profiling of preterm infants reveals DNA methylation differences at sites associated with neural
function. Translational Psychiatry. 2016 Jan 1;6(1):e716e716.
60. Winchester P, Nilsson E, Beck D, Skinner MK. Preterm birth buccal cell epigenetic biomarkers
to facilitate preventative medicine. Scientific Reports. 2022 Mar 1;12(1):3361.
30
61. Fumagalli M, Provenzi L, De Carli P, Dessimone F, Sirgiovanni I, Giorda R, et al. From early
stress to 12-month development in very preterm infants: Preliminary findings on epigenetic
mechanisms and brain growth. PloS one. 2018;13(1):e0190602.
62. Chen L, Pan H, Tuan TA, Teh AL, MacIsaac JL, Mah SM, et al. Brain-derived neurotrophic
factor (BDNF) Val66Met polymorphism influences the association of the methylome with
maternal anxiety and neonatal brain volumes. Development and psychopathology.
2015;27(1):13750.
63. Wheater ENW, Galdi P, McCartney DL, Blesa M, Sullivan G, Stoye DQ, et al. DNA methylation
in relation to gestational age and brain dysmaturation in preterm infants. Brain Communications.
2022 Apr 1;4(2):fcac056.
64. Camerota M, Graw S, Everson TM, McGowan EC, Hofheimer JA, O’Shea TM, et al. Prenatal
risk factors and neonatal DNA methylation in very preterm infants. Clinical Epigenetics. 2021
Sep 10;13(1):171.
65. Everson TM, O’Shea TM, Burt A, Hermetz K, Carter BS, Helderman J, et al. Serious neonatal
morbidities are associated with differences in DNA methylation among very preterm infants.
Clinical Epigenetics. 2020 Oct 19;12(1):151.
66. Boardman JP, Hall J, Thrippleton MJ, Reynolds RM, Bogaert D, Davidson DJ, et al. Impact of
preterm birth on brain development and long-term outcome: protocol for a cohort study in
Scotland. BMJ Open. 2020 Mar 4;10(3):e035854e035854.
67. Bell MJ, Ternberg JL, Feigin RD, Keating JP, Marshall R, Barton L, et al. Neonatal necrotizing
enterocolitis. Therapeutic decisions based upon clinical staging. Annals of surgery.
1978;187(1):1.
68. Villar J, Ismail LC, Victora CG, Ohuma EO, Bertino E, Altman DG, et al. International standards
for newborn weight, length, and head circumference by gestational age and sex: the Newborn
Cross-Sectional Study of the INTERGROWTH-21st Project. The Lancet. 2014;384(9946):857
68.
69. Ligthart S, Marzi C, Aslibekyan S, Mendelson MM, Conneely KN, Tanaka T, et al. DNA
methylation signatures of chronic low-grade inflammation are associated with complex diseases.
Genome biology. 2016;17(1):255.
70. Barker ED, Cecil CAM, Walton E, Houtepen LC, O’Connor TG, Danese A, et al. Inflammation-
related epigenetic risk and child and adolescent mental health: A prospective study from
pregnancy to middle adolescence. Development and Psychopathology. 2018;30(3):114556.
71. Caruyer E, Lenglet C, Sapiro G, Deriche R. Design of multishell sampling schemes with uniform
coverage in diffusion MRI. Magnetic resonance in medicine. 2013;69(6):153440.
72. Blesa M, Galdi P, Cox SR, Sullivan G, Stoye DQ, Lamb GJ, et al. Hierarchical Complexity of the
Macro-Scale Neonatal Brain. Cerebral Cortex. 2021 Apr 1;31(4):207184.
73. Makropoulos A, Counsell SJ, Rueckert D. A review on automatic fetal and neonatal brain MRI
segmentation. NeuroImage. 2018;170:23148.
74. Doshi J, Erus G, Ou Y, Gaonkar B, Davatzikos C. Multi-Atlas Skull-Stripping. Academic
Radiology. 2013 Dec 1;20(12):156676.
75. Zhang H, Schneider T, Wheeler-Kingshott CA, Alexander DC. NODDI: practical in vivo neurite
orientation dispersion and density imaging of the human brain. Neuroimage. 2012;61(4):1000
16.
31
76. Tariq M, Schneider T, Alexander DC, Gandini Wheeler-Kingshott CA, Zhang H. Bingham
NODDI: Mapping anisotropic orientation dispersion of neurites using diffusion MRI. NeuroImage.
2016 Jun 1;133:20723.
77. Blesa M, Galdi P, Sullivan G, Wheater EN, Stoye DQ, Lamb GJ, et al. Peak width of
skeletonized water diffusion MRI in the neonatal brain. Frontiers in neurology. 2020;11.
78. Baykara E, Gesierich B, Adam R, Tuladhar AM, Biesbroek JM, Koek HL, et al. A novel imaging
marker for small vessel disease based on skeletonization of white matter tracts and diffusion
histograms. Annals of neurology. 2016;80(4):58192.
79. Vaher K, Galdi P, Blesa Cabez M, Sullivan G, Stoye DQ, Quigley AJ, et al. General factors of
white matter microstructure from DTI and NODDI in the developing brain. NeuroImage. 2022 Jul
1;254:119169.
80. Kim HY. Statistical notes for clinical researchers: Chi-squared test and Fisher’s exact test.
Restor Dent Endod. 2017 May;42(2):1525.
81. Tzoulaki I, Liberopoulos G, Ioannidis JP. Assessment of claims of improved prediction beyond
the Framingham risk score. Jama. 2009;302(21):234552.
82. Cohen J. A power primer. Psychological Bulletin. 1992;112(1):1559.
83. Lingam I, Robertson NJ. Magnesium as a Neuroprotective Agent: A Review of Its Use in the
Fetus, Term Infant with Neonatal Encephalopathy, and the Adult Stroke Patient. Developmental
Neuroscience. 2018;40(1):112.
84. Odufalu FD, Long M, Lin K, Mahadevan U. Exposure to corticosteroids in pregnancy is
associated with adverse perinatal outcomes among infants of mothers with inflammatory bowel
disease: results from the PIANO registry. Gut. 2022 Sep 1;71(9):1766.
85. Schmidt AF, Kannan PS, Bridges J, Presicce P, Jackson CM, Miller LA, et al. Prenatal
inflammation enhances antenatal corticosteroidinduced fetal lung maturation. JCI Insight
[Internet]. 2021 Jan 20;5(24). Available from: https://doi.org/10.1172/jci.insight.139452
86. Cox SR, Lyall DM, Ritchie SJ, Bastin ME, Harris MA, Buchanan CR, et al. Associations between
vascular risk factors and brain MRI indices in UK Biobank. European Heart Journal. 2019 Mar
11;40(28):2290300.
87. Telford EJ, Cox SR, Fletcher-Watson S, Anblagan D, Sparrow S, Pataky R, et al. A latent
measure explains substantial variance in white matter microstructure across the newborn
human brain. Brain Structure and Function. 2017 Dec 1;222(9):402333.
88. Menon R, Conneely KN, Smith AK. DNA methylation: an epigenetic risk factor in preterm birth.
Reprod Sci. 2012 Jan;19(1):613.
89. Parets SE, Bedient CE, Menon R, Smith AK. Preterm birth and its long-term effects: methylation
to mechanisms. Biology (Basel). 2014 Aug 21;3(3):498513.
90. Perera F, Herbstman J. Prenatal environmental exposures, epigenetics, and disease.
Reproductive toxicology. 2011;31(3):36373.
91. Zuccarello D, Sorrentino U, Brasson V, Marin L, Piccolo C, Capalbo A, et al. Epigenetics of
pregnancy: looking beyond the DNA code. Journal of Assisted Reproduction and Genetics.
2022 Apr 1;39(4):80116.
92. Barker ED, Walton E, Cecil CAM. Annual Research Review: DNA methylation as a mediator in
the association between risk exposure and child and adolescent psychopathology. Journal of
Child Psychology and Psychiatry. 2018 Apr 1;59(4):30322.
32
93. Bakulski KM, Fallin MD. Epigenetic epidemiology: Promises for public health research.
Environmental and Molecular Mutagenesis. 2014 Apr 1;55(3):17183.
94. Odintsova VV, Rebattu V, Hagenbeek FA, Pool R, Beck JJ, Ehli EA, et al. Predicting Complex
Traits and Exposures From Polygenic Scores and Blood and Buccal DNA Methylation Profiles.
Frontiers in Psychiatry [Internet]. 2021;12. Available from:
https://www.frontiersin.org/articles/10.3389/fpsyt.2021.688464
95. Reese SE, Zhao S, Wu MC, Joubert BR, Parr CL, Håberg SE, et al. DNA methylation score as a
biomarker in newborns for sustained maternal smoking during pregnancy. Environmental health
perspectives. 2017;125(4):7606.
96. Richmond RC, Simpkin AJ, Woodward G, Gaunt TR, Lyttleton O, McArdle WL, et al. Prenatal
exposure to maternal smoking and offspring DNA methylation across the lifecourse: findings
from the Avon Longitudinal Study of Parents and Children (ALSPAC). Human molecular
genetics. 2015;24(8):220117.
97. Richmond RC, Suderman M, Langdon R, Relton CL, Davey Smith G. DNA methylation as a
marker for prenatal smoke exposure in adults. International Journal of Epidemiology. 2018 Aug
1;47(4):112030.
98. Bakulski KM, Fisher JD, Dou JF, Gard A, Schneper L, Notterman DA, et al. Prenatal Particulate
Matter Exposure Is Associated with Saliva DNA Methylation at Age 15: Applying Cumulative
DNA Methylation Scores as an Exposure Biomarker. Toxics. 2021;9(10).
99. Somineni HK, Venkateswaran S, Kilaru V, Marigorta UM, Mo A, Okou DT, et al. Blood-Derived
DNA Methylation Signatures of Crohn’s Disease and Severity of Intestinal Inflammation.
Gastroenterology. 2019 Jun 1;156(8):2254-2265.e3.
100. Sasaki A, Murphy KE, Briollais L, McGowan PO, Matthews SG. DNA methylation profiles in the
blood of newborn term infants born to mothers with obesity. PLOS ONE. 2022 May
2;17(5):e0267946.
101. Yanni D, Korzeniewski SJ, Allred EN, Fichorova RN, O’Shea TM, Kuban K, et al. Both antenatal
and postnatal inflammation contribute information about the risk of brain damage in extremely
preterm newborns. Pediatric Research. 2017 Oct 1;82(4):6916.
102. Fleiss B, Tann CJ, Degos V, Sigaut S, Van Steenwinckel J, Schang A, et al. Inflammation-
induced sensitization of the brain in term infants. Developmental Medicine & Child Neurology.
2015;57:1728.
103. Glass TJ, Chau V, Grunau RE, Synnes A, Guo T, Duerden EG, et al. Multiple postnatal
infections in newborns born preterm predict delayed maturation of motor pathways at term-
equivalent age with poorer motor outcomes at 3 years. The Journal of pediatrics. 2018;196:91
7.
104. Ophelders DRMG, Gussenhoven R, Klein L, Jellema RK, Westerlaken RJJ, Hütten MC, et al.
Preterm Brain Injury, Antenatal Triggers, and Therapeutics: Timing Is Key. Cells. 2020;9(8).
105. Tzoulaki I, Jarvelin MR, Hartikainen AL, Leinonen M, Pouta A, Paldanius M, et al. Size at birth,
weight gain over the life course, and low-grade inflammation in young adulthood: northern
Finland 1966 birth cohort study. European Heart Journal. 2008 Apr 1;29(8):104956.
106. McDade TW, Metzger MW, Chyu L, Duncan GJ, Garfield C, Adam EK. Long-term effects of birth
weight and breastfeeding duration on inflammation in early adulthood. Proceedings of the Royal
Society B: Biological Sciences. 2014 Jun 7;281(1784):20133116.
107. McDade TW, Ryan C, Jones MJ, MacIsaac JL, Morin AM, Meyer JM, et al. Social and physical
environments early in development predict DNA methylation of inflammatory genes in young
adulthood. Proceedings of the National Academy of Sciences. 2017 Jul 18;114(29):76116.
33
108. Fleiss B, Gressens P. Tertiary mechanisms of brain damage: a new hope for treatment of
cerebral palsy? The Lancet Neurology. 2012 Jun 1;11(6):55666.
109. Skiöld B, Horsch S, Hallberg B, Engström M, Nagy Z, Mosskin M, et al. White matter changes in
extremely preterm infants, a population-based diffusion tensor imaging study. Acta paediatrica.
2010;99(6):8429.
110. Volpe JJ. Dysmaturation of Premature Brain: Importance, Cellular Mechanisms, and Potential
Interventions. Pediatric Neurology. 2019 Jun 1;95:4266.
111. Favrais G, Van De Looij Y, Fleiss B, Ramanantsoa N, Bonnin P, Stoltenburg-Didinger G, et al.
Systemic inflammation disrupts the developmental program of white matter. Annals of
neurology. 2011;70(4):55065.
112. Padilla N, Alexandrou G, Blennow M, Lagercrantz H, Ådén U. Brain Growth Gains and Losses
in Extremely Preterm Infants at Term. Cerebral Cortex. 2015 Jul 1;25(7):1897905.
113. Inder TE, Warfield SK, Wang H, Hüppi PS, Volpe JJ. Abnormal Cerebral Structure Is Present at
Term in Premature Infants. Pediatrics. 2005 Feb 1;115(2):28694.
114. Boardman JP, Counsell SJ, Rueckert D, Kapellou O, Bhatia KK, Aljabar P, et al. Abnormal deep
grey matter development following preterm birth detected using deformation-based
morphometry. NeuroImage. 2006 Aug 1;32(1):708.
115. Lammertink F, van den Heuvel MP, Hermans EJ, Dudink J, Tataranno ML, Benders MJNL, et al.
Early-life stress exposure and large-scale covariance brain networks in extremely preterm-born
infants. Translational Psychiatry. 2022 Jun 18;12(1):256.
116. Boardman JP, Craven C, Valappil S, Counsell SJ, Dyet LE, Rueckert D, et al. A common
neonatal image phenotype predicts adverse neurodevelopmental outcome in children born
preterm. Neuroimage. 2010;52(2):40914.
117. Beauchamp MH, Thompson DK, Howard K, Doyle LW, Egan GF, Inder TE, et al. Preterm infant
hippocampal volumes correlate with later working memory deficits. Brain. 2008 Nov
1;131(11):298694.
118. Ball G, Boardman JP, Aljabar P, Pandit A, Arichi T, Merchant N, et al. The influence of preterm
birth on the developing thalamocortical connectome. Cortex. 2013;49(6):171121.
119. Volpe JJ, Kinney HC, Jensen FE, Rosenberg PA. The developing oligodendrocyte: key cellular
target in brain injury in the premature infant. International Journal of Developmental
Neuroscience. 2011 Jun 1;29(4):42340.
120. Kostović I, Kostović-Srzentić M, Benjak V, Jovanov-Milošević N, Radoš M. Developmental
dynamics of radial vulnerability in the cerebral compartments in preterm infants and neonates.
Frontiers in neurology. 2014;5:139.
121. Volpe JJ. Brain injury in premature infants: a complex amalgam of destructive and
developmental disturbances. The Lancet Neurology. 2009 Jan 1;8(1):11024.
122. Leviton A, Allred EN, Dammann O, Engelke S, Fichorova RN, Hirtz D, et al. Systemic
Inflammation, Intraventricular Hemorrhage, and White Matter Injury. J Child Neurol. 2013 Dec
1;28(12):163745.
123. Leviton A, Gressens P. Neuronal damage accompanies perinatal white-matter damage. Trends
in Neurosciences. 2007 Sep 1;30(9):4738.
124. Ment LR, Hirtz D, Hüppi PS. Imaging biomarkers of outcome in the developing preterm brain.
The Lancet Neurology. 2009 Nov 1;8(11):104255.
34
125. Back SA, Luo NL, Borenstein NS, Levine JM, Volpe JJ, Kinney HC. Late oligodendrocyte
progenitors coincide with the developmental window of vulnerability for human perinatal white
matter injury. Journal of Neuroscience. 2001;21(4):130212.
126. Majnemer A, Riley P, Shevell M, Birnbaum R, Greenstone H, Coates AL. Severe
bronchopulmonary dysplasia increases risk for later neurological and motor sequelae in preterm
survivors. Developmental medicine and child neurology. 2000;42(1):5360.
127. Alexandrou G, Mårtensson G, Skiöld B, Blennow M, Ådén U, Vollmer B. White matter
microstructure is influenced by extremely preterm birth and neonatal respiratory factors. Acta
paediatrica. 2014;103(1):4856.
128. Breton CV, Marsit CJ, Faustman E, Nadeau K, Goodrich JM, Dolinoy DC, et al. Small-
magnitude effect sizes in epigenetic end points are important in children’s environmental health
studies: the children’s environmental health and disease prevention research center’s
epigenetics working group. Environmental health perspectives. 2017;125(4):51126.
129. Glass HC, Bonifacio SL, Chau V, Glidden D, Poskitt K, Barkovich AJ, et al. Recurrent postnatal
infections are associated with progressive white matter injury in premature infants. Pediatrics.
2008;122(2):299305.
130. Wilson S, Pietsch M, Cordero-Grande L, Price AN, Hutter J, Xiao J, et al. Development of
human white matter pathways in utero over the second and third trimester. Proceedings of the
National Academy of Sciences. 2021 May 18;118(20):e2023598118.
131. Chau V, Brant R, Poskitt KJ, Tam EW, Synnes A, Miller SP. Postnatal infection is associated
with widespread abnormalities of brain development in premature newborns. Pediatric research.
2012;71(3):2749.
132. Nist MD, Shoben AB, Pickler RH. Early Inflammatory Measures and Neurodevelopmental
Outcomes in Preterm Infants. Nursing Research [Internet]. 2020;69(5S). Available from:
https://journals.lww.com/nursingresearchonline/Fulltext/2020/09001/Early_Inflammatory_Measur
es_and_Neurodevelopmental.3.aspx
133. Wheater ENW, Stoye DQ, Cox SR, Wardlaw JM, Drake AJ, Bastin ME, et al. DNA methylation
and brain structure and function across the life course: A systematic review. Neurosci Biobehav
Rev. 2020/03/06 ed. 2020 Jun;113:13356.
134. Lancaster K, Morris JP, Connelly JJ. Neuroimaging epigenetics: Challenges and
recommendations for best practices. Neuroscience. 2018;370:88100.
135. Sparrow S, Manning J, Cartier J, Anblagan D, Bastin M, Piyasena C, et al. Epigenomic profiling
of preterm infants reveals DNA methylation differences at sites associated with neural function.
Translational psychiatry. 2016;6(1):e716e716.
136. Davies MN, Volta M, Pidsley R, Lunnon K, Dixit A, Lovestone S, et al. Functional annotation of
the human brain methylome identifies tissue-specific epigenetic variation across brain and
blood. Genome Biology. 2012 Jun 15;13(6):R43.
137. Walton E, Hass J, Liu J, Roffman JL, Bernardoni F, Roessner V, et al. Correspondence of DNA
methylation between blood and brain tissue and its application to schizophrenia research.
Schizophrenia bulletin. 2016;42(2):40614.
138. Blostein FA, Fisher J, Dou J, Schneper L, Ware EB, Notterman DA, et al. Polymethylation
scores for prenatal maternal smoke exposure persist until age 15 and are detected in saliva in
the Fragile Families and Child Wellbeing cohort. Epigenetics. 2022 Aug 18;118.
139. Berko ER, Suzuki M, Beren F, Lemetre C, Alaimo CM, Calder RB, et al. Mosaic epigenetic
dysregulation of ectodermal cells in autism spectrum disorder. PLoS genetics.
2014;10(5):e1004402.
35
140. Lin X, Teh AL, Chen L, Lim IY, Tan PF, MacIsaac JL, et al. Choice of surrogate tissue
influences neonatal EWAS findings. BMC medicine. 2017;15(1):113.
141. Lowe R, Gemma C, Beyan H, Hawa MI, Bazeos A, Leslie RD, et al. Buccals are likely to be a
more informative surrogate tissue than blood for epigenome-wide association studies.
Epigenetics. 2013;8(4):44554.
142. Braun PR, Han S, Hing B, Nagahama Y, Gaul LN, Heinzman JT, et al. Genome-wide DNA
methylation comparison between live human brain and peripheral tissues within individuals.
Translational psychiatry. 2019;9(1):110.
143. Birney E, Smith GD, Greally JM. Epigenome-wide association studies and the interpretation of
disease-omics. PLoS genetics. 2016;12(6):e1006105.
144. Luo M, Meehan AJ, Walton E, Röder S, Herberth G, Zenclussen AC, et al. Neonatal DNA
methylation and childhood low prosocial behavior: An epigenome-wide association meta-
analysis. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics. 2021 Jun
1;186(4):22841.
145. Chen J, Zang Z, Braun U, Schwarz K, Harneit A, Kremer T, et al. Association of a reproducible
epigenetic risk profile for schizophrenia with brain methylation and function. JAMA psychiatry.
2020;77(6):62836.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The stressful extrauterine environment following premature birth likely has far-reaching and persistent adverse consequences. The effects of early “third-trimester” ex utero stress on large-scale brain networks’ covariance patterns may provide a potential avenue to understand how early-life stress following premature birth increases risk or resilience. We evaluated the impact of early-life stress exposure (e.g., quantification of invasive procedures) on maturational covariance networks (MCNs) between 30 and 40 weeks of gestational age in 180 extremely preterm-born infants (<28 weeks of gestation; 43.3% female). We constructed MCNs using covariance of gray matter volumes between key nodes of three large-scale brain networks: the default mode network (DMN), executive control network (ECN), and salience network (SN). Maturational coupling was quantified by summating the number of within- and between-network connections. Infants exposed to high stress showed significantly higher SN but lower DMN maturational coupling, accompanied by DMN-SN decoupling. Within the SN, the insula, amygdala, and subthalamic nucleus all showed higher maturational covariance at the nodal level. In contrast, within the DMN, the hippocampus, parahippocampal gyrus, and fusiform showed lower coupling following stress. The decoupling between DMN-SN was observed between the insula/anterior cingulate cortex and posterior parahippocampal gyrus. Early-life stress showed longitudinal network-specific maturational covariance patterns, leading to a reprioritization of developmental trajectories of the SN at the cost of the DMN. These alterations may enhance the ability to cope with adverse stimuli in the short term but simultaneously render preterm-born individuals at a higher risk for stress-related psychopathology later in life.
Article
Full-text available
Maternal obesity is an important risk factor for childhood obesity and influences the prevalence of metabolic diseases in offspring. As childhood obesity is influenced by postnatal factors, it is critical to determine whether children born to women with obesity during pregnancy show alterations that are detectable at birth. Epigenetic mechanisms such as DNA methylation modifications have been proposed to mediate prenatal programming. We investigated DNA methylation signatures in male and female infants from mothers with a normal Body Mass Index (BMI 18.5–24.9 kg/m ² ) compared to mothers with obesity (BMI≥30 kg/m ² ). BMI was measured during the first prenatal visit from women recruited into the Ontario Birth Study (OBS) at Mount Sinai Hospital in Toronto, ON, Canada. DNA was extracted from neonatal dried blood spots collected from heel pricks obtained 24 hours after birth at term (total n = 40) from women with a normal BMI and women with obesity matched for parity, age, and neonatal sex. Reduced representation bisulfite sequencing was used to identify genomic loci associated with differentially methylated regions (DMRs) in CpG-dense regions most likely to influence gene regulation. DMRs were predominantly localized to intergenic regions and gene bodies, with only 9% of DMRs localized to promoter regions. Genes associated with DMRs were compared to those from a large publicly available cohort study, the Avon Longitudinal Study of Parents and Children (ALSPAC; total n = 859). Hypergeometric tests revealed a significant overlap in genes associated with DMRs in the OBS and ALSPAC cohorts. PTPRN2 , a gene involved in insulin secretion, and MAD1L1 , which plays a role in the cell cycle and tumor suppression, contained DMRs in males and females in both cohorts. In males, KEGG pathway analysis revealed significant overrepresentation of genes involved in endocytosis and pathways in cancer, including IGF1R , which was previously shown to respond to diet-induced metabolic stress in animal models and in lymphocytes in the context of childhood obesity. These preliminary findings are consistent with Developmental Origins of Health and Disease paradigm, which posits that adverse prenatal exposures set developmental health trajectories.
Article
Full-text available
Importance Animal studies show that Maternal Immune Activation (MIA) may have detrimental effects on fetal brain development. Clinical studies provide evidence for structural brain abnormalities in human neonates following MIA, but no study has investigated the long-term effects of MIA (as measured with biomarkers) on human brain morphology ten years after the exposure. Objective Our aim was to evaluate the long-term impact of MIA on brain morphology in 10-year-old children, including the possible mediating role of gestational age at birth. Design We leveraged data from Generation R, a large-scale prospective pregnancy cohort study. Pregnant women were included between 2002 and 2006, and their children were invited to participate in the MRI study between 2013 and 2015. To be included, mother-child dyads had to have data on maternal C-reactive protein levels during gestation and a good quality MRI-scan of the child’s brain at age 10 years. Of the 3,992 children scanned, a total of 2,053 10-year-old children were included in this study. Exposure Maternal C-reactive protein was measured in the first 18 weeks of gestation. For the analyses we used both a continuous approach as well as a categorical approach based on clinical cut-offs to determine if there was a dose-response relationship. Main outcomes and measures High-resolution MRI brain morphology measures were used as the primary outcome. Gestational age at birth, established using ultrasound, was included as a mediator using a causal mediation analysis. Corrections were made for relevant confounders and multiple comparisons. Biological sex was investigated as moderator. Results We found a direct association between continuous MIA and lower cerebellar volume. In girls, we demonstrated a negative indirect association between continuous MIA and total brain volume, through the mediator gestational age at birth. We observed no associations with categorical MIA after multiple testing correction. Conclusion and relevance Our results suggest sex-specific long-term effects in brain morphology after MIA. Categorical analyses suggest that this association might be driven by acute infections or or other sources of severe inflammation, which is of clinical relevance given that the COVID-19 pandemic is currently affecting millions of pregnant women worldwide.
Article
Full-text available
Preterm birth is closely associated with diffuse white matter dysmaturation inferred from diffusion MRI and neurocognitive impairment in childhood. Diffusion tensor imaging (DTI) and neurite orientation dispersion and density imaging (NODDI) are distinct dMRI modalities, yet metrics derived from these two methods share variance across tracts. This raises the hypothesis that dimensionality reduction approaches may provide efficient whole-brain estimates of white matter microstructure that capture (dys)maturational processes. To investigate the optimal model for accurate classification of generalised white matter dysmaturation in preterm infants we assessed variation in DTI and NODDI metrics across 16 major white matter tracts using principal component analysis and structural equation modelling, in 79 term and 141 preterm infants at term equivalent age. We used logistic regression models to evaluate performances of single-metric and multimodality general factor frameworks for efficient classification of preterm infants based on variation in white matter microstructure. Single-metric general factors from DTI and NODDI capture substantial shared variance (41.8-72.5%) across 16 white matter tracts, and two multimodality factors captured 93.9% of variance shared between DTI and NODDI metrics themselves. General factors associate with preterm birth and a single model that includes all seven DTI and NODDI metrics provides the most accurate prediction of microstructural variations associated with preterm birth. This suggests that despite global covariance of dMRI metrics in neonates, each metric represents information about specific (and additive) aspects of the underlying microstructure that differ in preterm compared to term subjects.
Article
Full-text available
Epigenetics is the branch of genetics that studies the different mechanisms that influence gene expression without direct modification of the DNA sequence. An ever-increasing amount of evidence suggests that such regulatory processes may play a pivotal role both in the initiation of pregnancy and in the later processes of embryonic and fetal development, thus determining long-term effects even in adult life. In this narrative review, we summarize the current knowledge on the role of epigenetics in pregnancy, from its most studied and well-known mechanisms to the new frontiers of epigenetic regulation, such as the role of ncRNAs and the effects of the gestational environment on fetal brain development. Epigenetic mechanisms in pregnancy are a dynamic phenomenon that responds both to maternal–fetal and environmental factors, which can influence and modify the embryo-fetal development during the various gestational phases. Therefore, we also recapitulate the effects of the most notable environmental factors that can affect pregnancy and prenatal development, such as maternal nutrition, stress hormones, microbiome, and teratogens, focusing on their ability to cause epigenetic modifications in the gestational environment and ultimately in the fetus. Despite the promising advancements in the knowledge of epigenetics in pregnancy, more experience and data on this topic are still needed. A better understanding of epigenetic regulation in pregnancy could in fact prove valuable towards a better management of both physiological pregnancies and assisted reproduction treatments, other than allowing to better comprehend the origin of multifactorial pathological conditions such as neurodevelopmental disorders.
Article
Full-text available
Preterm birth is associated with dysconnectivity of structural brain networks and is a leading cause of neurocognitive impairment in childhood. Variation in DNA methylation is associated with early exposure to extrauterine life but there has been little research exploring its relationship with brain development. Using genome-wide DNA methylation data from saliva of 258 neonates, we investigated the impact of gestational age on the methylome and performed functional analysis to identify enriched gene sets from probes that contributed to differentially methylated probes or regions. We tested the hypothesis that variation in DNA methylation could underpin the association between low gestational age at birth and atypical brain development by linking differentially methylated probes with measures of white matter connectivity derived from diffusion MRI metrics: peak width skeletonised mean diffusivity, peak width skeletonised fractional anisotropy and peak width skeletonised neurite density index. Gestational age at birth was associated with widespread differential methylation at term equivalent age, with genome-wide significant associations observed for 8,870 CpG probes (p < 3.6 × 10−8) and 1,767 differentially methylated regions. Functional analysis identified 14 enriched gene ontology terms pertaining to cell-cell contacts and cell-extracellular matrix contacts. Principal component analysis of probes with genome-wide significance revealed a first principal component that explained 23.5% of variance in DNA methylation, and this was negatively associated with gestational age at birth. The first principal component was associated with peak width of skeletonised mean diffusivity (β=0.349, p = 8.37 × 10−10) and peak width skeletonised neurite density index (β=0.364, p = 4.15 × 10−5), but not with peak width skeletonised fraction anisotropy (β=-0.035, p = 0.510); these relationships mirrored the imaging metrics’ associations with gestational age at birth. Low gestational age at birth has a profound and widely distributed effect on the neonatal saliva methylome that is apparent at term equivalent age. Enriched gene ontology terms related to cell-cell contacts reveal pathways that could mediate the effect of early life environmental exposures on development. Finally, associations between differential DNA methylation and image markers of white matter tract microstructure suggest that variation in DNA methylation may provide a link between preterm birth and the dysconnectivity of developing brain networks that characterises atypical brain development in preterm infants.
Article
Full-text available
Preterm birth is the major cause of newborn and infant mortality affecting nearly one in every ten live births. The current study was designed to develop an epigenetic biomarker for susceptibility of preterm birth using buccal cells from the mother, father, and child (triads). An epigenome-wide association study (EWAS) was used to identify differential DNA methylation regions (DMRs) using a comparison of control term birth versus preterm birth triads. Epigenetic DMR associations with preterm birth were identified for both the mother and father that were distinct and suggest potential epigenetic contributions from both parents. The mother (165 DMRs) and female child (136 DMRs) at p < 1e−04 had the highest number of DMRs and were highly similar suggesting potential epigenetic inheritance of the epimutations. The male child had negligible DMR associations. The DMR associated genes for each group involve previously identified preterm birth associated genes. Observations identify a potential paternal germline contribution for preterm birth and identify the potential epigenetic inheritance of preterm birth susceptibility for the female child later in life. Although expanded clinical trials and preconception trials are required to optimize the potential epigenetic biomarkers, such epigenetic biomarkers may allow preventative medicine strategies to reduce the incidence of preterm birth.
Article
Full-text available
The field of adult neuroimaging relies on well-established principles in research design, imaging sequences, processing pipelines, as well as safety and data collection protocols. The field of infant magnetic resonance imaging, by comparison, is a young field with tremendous scientific potential but continuously evolving standards. The present article aims to initiate a constructive dialogue between researchers who grapple with the challenges and inherent limitations of a nascent field and reviewers who evaluate their work. We address 20 questions that researchers commonly receive from research ethics boards, grant, and manuscript reviewers related to infant neuroimaging data collection, safety protocols, study planning, imaging sequences, decisions related to software and hardware, and data processing and sharing, while acknowledging both the accomplishments of the field and areas of much needed future advancements. This article reflects the cumulative knowledge of experts in the FIT’NG community and can act as a resource for both researchers and reviewers alike seeking a deeper understanding of the standards and tradeoffs involved in infant neuroimaging.
Article
Prenatal maternal smoking is associated with low birthweight, neurological disorders, and asthma in exposed children. DNA methylation signatures can function as biomarkers of prenatal smoke exposure. However, the robustness of DNA methylation signatures across child ages, genetic ancestry groups, or tissues is not clear. Using coefficients from a meta-analysis of prenatal smoke exposure and DNA methylation in newborn cord blood, we created polymethylation scores of saliva DNA methylation from children at ages 9 and 15 in the Fragile Families and Child Wellbeing study. In the full sample at age 9 (n = 753), prenatal smoke exposure was associated with a 0.51 (95%CI: 0.35, 0.66) standard deviation higher polymethylation score. The direction and magnitude of the association was consistent in European and African genetic ancestry samples. In the full sample at age 15 (n = 747), prenatal smoke exposure was associated with a 0.48 (95%CI: 0.32, 0.63) standard deviation higher polymethylation score, and the association was attenuated among the European and Admixed–Latin genetic ancestry samples. The polymethylation score classified prenatal smoke exposure accurately (AUC age 9 = 0.77, age 15 = 0.76). Including the polymethylation score increased the AUC of base model covariates by 5 (95% CI: (2.1, 7.2)) percentage points, while including a single candidate site in the AHRR gene did not (P-value = 0.19). Polymethylation scores for prenatal smoking were portable across genetic ancestries and more accurate than an individual DNA methylation site. Polymethylation scores from saliva samples could serve as robust and practical biomarkers of prenatal smoke exposure.
Article
Preterm newborns are exposed to several risk factors for developing brain injury. Clinical studies have suggested that the presence of intrauterine infection is a consistent risk factor for preterm birth and white matter injury. Animal models have confirmed these associations by identifying inflammatory cascades originating at the maternofetal interface that penetrate the fetal blood-brain barrier and result in brain injury. Acquired diseases of prematurity further potentiate the risk for cerebral injury. Systems biology approaches incorporating ante- and post-natal risk factors and analyzing omic and multiomic data using machine learning are promising methodologies for further elucidating biologic mechanisms of fetal and neonatal brain injury.