Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Hypertension

REVIEW

Developmental and Early Life Origins of


Cardiometabolic Risk Factors
Novel Findings and Implications
Empar Lurbe, Julie Ingelfinger

ABSTRACT: The intent of this review is to critically consider the data that support the concept of programming and its
implications. Birth weight and growth trajectories during childhood are associated with cardiometabolic disease in adult life.
Both extremes, low and high birth weight coupled with postnatal growth increase the early presence of cardiometabolic risk
factors and vascular imprinting, crucial elements of this framework. Data coming from epigenetics, proteomics, metabolomics,
and microbiota added relevant information and contribute to better understanding of mechanisms as well as development of
biomarkers helping to move forward to take actions. Research has reached a stage in which sufficiently robust data calls for
new initiatives focused on early life. Prevention starting early in life is likely to have a very large impact on reducing disease
incidence and its associated effects at the personal, economic, and social levels.

Key Words:  cardiovascular disease ◼ coronavirus ◼ birth weight ◼ proteomics ◼ traction

RELEVANCE-WHY IS THE TOPIC EARLY ORIGINS OF CARDIOMETABOLIC


Downloaded from http://ahajournals.org by on January 15, 2023

IMPORTANT RISK FACTORS


Before the present pandemic of the severe acute respi- The concept of the developmental origins of health and
ratory syndrome coronavirus 2, noncommunicable dis- disease (DOHaD) and the knowledge contributing to it
eases caused more deaths than infectious diseases amassed during the latter part of the 20th century has
worldwide; once the pandemic recedes, that is likely to consistently attracted intense interest.1,2 There is grow-
be the case again. Over the past several decades, the ing evidence from observational studies indicating an
concept that perinatal conditions, both intrauterine and association between early life and later cardiometabolic
immediately after birth, increase the risk of many dis- risk factors. While initial studies emphasized intrauterine
eases later in life, has gained traction, based both on epi- conditions as the main factor, further research has pos-
demiological studies and experimental studies in animal tulated that postnatal growth patterns have an important
models. Strong epidemiological data suggest that low additional impact on cardiovascular disease (CVD).3–6
birth weight (LBW) and prematurity, surrogates for intra- Currently, the concept that a life course perspective may
uterine conditions, are associated with adverse health be the optimal way to understand adult health and dis-
conditions later in life, and even transgenerationally. ease is recognized.
The intent of this review is to critically consider the Although in childhood it is extremely rare to suffer the
data that support the concept of programming and its adverse CV events experienced by adults, cardiovascu-
implications. Why is this important? Obviously, if adverse lar and metabolic disease risk factors may already be
perinatal conditions can be avoided or ameliorated, present and related to the development of future adult
then perhaps the implied legacy of higher risk can be CVD.7 The challenge is to elucidate the main risk factors,
diminished. know when and how to assess these factors and discern


Correspondence to: Empar Lurbe, Department of Pediatrics, Consorcio Hospital General, University of Valencia, Avda. Tres Cruces s/n, 46014 Valencia, Spain. Email
empar.lurbe@uv.es
For Sources of Funding and Disclosures, see page 315.
© 2020 American Heart Association, Inc.
Hypertension is available at www.ahajournals.org/journal/hyp

308   February 2021 Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

between LBW with systolic blood pressure13,14 and type


Nonstandard Abbreviation and Acronyms 2 diabetes risk.15–17 Aside from analyses of the impact of
LBW, the impact of different BW categories on cardio-
ACE-1 angiotensin-converting enzyme 1 metabolic risk patterns in both children and adults has

Review
AGA appropriate for gestational age become recognized.
AT1b angiotensin type 1b receptor Data in children from the National Health and Nutri-
AT2 angiotensin type 2 receptor tion Examination Survey from 1999 through 2014, which
BMI body mass index included 28 153 subjects from birth to 15 years, were
CVD cardiovascular disease divided into 5 BW categories (<2.5, 2.5–3.0, 3.0–3.5, 3.5–
4.2, and ≥4.2 kg), with <2.5 kg as the LBW group (8.2%)
DOHaD developmental origins of health and
disease and ≥4.2 kg as the HBW group (10.4%).18 Over the entire
population, with an increasing BW, the results showed a
HBW high birth weight
significant rising trend for the risk of general and central
IMT intima-media thickness
obesity. In contrast, BW was significantly and inversely
LBW low birth weight
associated with high systolic blood pressure, high glycated
miRNA microRNA hemoglobin, and low high-density lipoprotein cholesterol,
SGA small for gestational age independent of current body size. The relation between
BW and childhood cardiometabolic risk factors showed
different patterns, depending on ethnic background.18
how to manage them in children and adolescents. Birth
While reports consistently note that upon reaching
weight, growth trajectories during childhood, and the
adult life there is an association between BW and type
cardiometabolic risk factors themselves are crucial ele-
2 diabetes,15–17 the link with CVD has been inconsis-
ments of this framework.
tent.19–23 Both of these issues were evaluated in a meta-
analysis of almost 8 million participants from 135 studies
The Importance of Birth Weight stratified into 6 BW groups from <2.5 kg to >4.5 kg.24
The findings suggested that BW is associated with the
Birth weight is a sentinel marker of fetal health, reflect-
risk of type 2 diabetes and CV disease in a J-shaped
ing both intrauterine growth and length of gestation. The
trend with increments of risk at both extremes. In con-
World Health Organization defines LBW as <2.5 kg, with
Downloaded from http://ahajournals.org by on January 15, 2023

trast, the risk of hypertension was inversely associated


the prevalence ranging from 2% to 12%. Low BW has
with BW—the lower the BW the higher the risk. Further-
been rising due to the increment in the number of mul-
more, among those at the higher end of the BW distribu-
tiple births, partly as a result of the increase in fertility
tion (>4.5 kg), BW associated more strongly with type 2
treatments, to the increasing age of mothers at child-
diabetes and CVD in females than in males. Considering
birth and to the rise in smoking among young mothers.8
these data and given that the prevalence of both low and
Despite the birth of more newborns with LBW, medical
HBW is increasing, these findings are highly relevant to
care has been successful in reducing infant mortality,
population health and risk estimates for chronic disease.
raising the number of persons at risk. Moreover, sex may act as an effect modifier rather than
Not only does LBW merit consideration but so does as a confounder at the upper tail of the BW distribution.24
high BW (HBW) as a consequence of intrauterine over-
nutrition, since that occurrence also has been related to
cardiometabolic risk factors. One of the main causes of The Case of Prematurity
fetal overnutrition is maternal obesity, with its inherent Studies concerning DOHaD have predominantly been
increased risk of complications in pregnancy, among performed in full-term births, even when prematurity has
them, gestational diabetes. Newborns of obese moth- become a relatively common reason for LBW in devel-
ers are generally heavier, with more body fat9 and higher oped countries. Of concern, however, adults who were
body mass index (BMI) in childhood compared with born prematurely have an increased prevalence of car-
children of nonobese mothers.10,11 As mothers become diometabolic risk factors in comparison with persons who
heavier, which is occurring in almost all populations, the were born after a full-term pregnancy.25,26 Epidemiologi-
incidence of diabetes during pregnancy is increasing,12 cal data have focused mainly on prematurity per se, often
which is likely to exacerbate the burden of obesity and without accounting for BW contingent on gestational
diabetes in the succeeding generation. age, with a paucity of knowledge concerning whether all
Following original observations by Barker et al,1 persons born prematurely have a tendency for higher risk
epidemiological studies demonstrated an association in adult life or whether the risk is increased only in the
between LBW and adverse cardiometabolic risk fac- presence of fetal growth restriction.
tors. Meta-analyses and systematic reviews with a large In a longitudinal study among participants of the pro-
number of studies supported an inverse association spective Cardiovascular Risk in Young Finns Study for

Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592 February 2021   309


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

whom there was information on BW and gestational age, childhood, that does not necessarily connote childhood
together with longitudinal data on cardiovascular risk obesity, also increases the risk, which underscores the
markers from age 3 to 18 years in 1980 to age 34 to 49 importance of regular growth monitoring.
years in 2011, the authors observed increased BP lev-
Review

els among those participants born prematurely and who


were also small for gestational age (SGA). In contrast,
Identifying Cardiometabolic Risk Factors
preterm appropriate for gestational age (AGA) did not The interest in assessing the presence of cardiometabolic
have elevated BP levels when compared with those born risk factors during childhood is based on identifying peo-
at term. The difference between those born at term and ple at increased risk of developing adult CVD, to estab-
those born preterm but at normal weights (AGA) only lish timely strategies for CVD prevention. Examination of
became evident when they reached the mean age of 41 the conventional risk factors obesity, hypertension, insu-
years, not earlier.27 For other cardiovascular risk factors, lin resistance, and dyslipidemia31 show heterogeneous
no significant differences have been observed based on results due to age at which they have been analyzed, the
birth characteristics.27 A recent systematic review and variables used and the design of the studies. The dis-
meta-analysis has pointed out that while only the preterm crepancies, according to age and conditions under which
AGA population was included, instead of the total pre- the variables are examined, derive from changes in body
term population, the significant preterm versus term dif- proportions in growing children and the absence of the
ferences of outcomes assessed remained significant.28 stability of risk factors across different ages. Addition-
Overall, multiple confounders make it difficult to ally, physiological changes over time imply variations in
reach firm conclusions, and it remains controversial as to the classification of relevant parameters. Fluctuations in
whether prematurity constitutes an independent risk fac- cardiometabolic risk factor levels during puberty are well
tor for the subsequent development of CVD. A marked known.32 Moreover, considering all of these factors, along
racial disparity is also persistent, even though the rates with the fact that most studies are cross-sectional and
of adverse birth outcomes have declined for all groups retrospective, the disparate results are understandable.
over the last century. Adjusting for maternal characteris- To better comprehend the effect of fetal life and post-
tics, income, race, and race plus income concentrations natal growth on cardiometabolic risk factors, longitudinal
remained negatively associated with preterm birth.29 studies may add relevant information, even though the
length of follow-up and the focus on different outcomes
have been limitations up to now. The major focus of
Downloaded from http://ahajournals.org by on January 15, 2023

The Role of Postnatal Growth longitudinal studies has been the association with later
Early childhood growth trajectories and rapid catch-up obesity. New findings on the predictors of obesity from
growth have also been shown to independently influence the Family Life Project, assessing longitudinal patterns of
the development of risk factors and the incidence of obesity, were that by age 6 stable obesity was associated
chronic disease. In a study of the Helsinki Birth Cohort by with both LBW and HBW.33 Repeating BMI measure-
Barker et al,3 adults with a coronary event were noted to ments, from childhood into adolescence, the UK Mille-
be relatively small at birth and thin at 2 years of age and nium Cohort Study showed that rapid weight gain in early
thereafter gained weight rapidly. This pattern of growth life was associated with higher BMI at 5 years and more
during childhood was associated with insulin resistance rapid BMI gain subsequently from 5 to 14 years, espe-
in later life. Furthermore, the risk of coronary events was cially in those large for gestational age at birth.34 Further
more related to the pace of childhood BMI gain than to follow-up of these cohorts will be needed to examine if
the BMI achieved at any particular age.3 the increased risk of obesity associated with HBW and
The impact of BW and postnatal growth on the pres- LBW is accompanied by obesity-related markers of dis-
ence of overweight or obesity, BP values, metabolic ease such as insulin resistance or high BP.
parameters, and diseases such as hypertension and type A prospective study went a step further, reaching
2 diabetes, was the topic of a systematic review that the end of the first decade of life and analyzing car-
included children, adolescents, and adults.30 While some diometabolic risk factors.35,36 Healthy, full-term infants
studies reviewed had a significant association with BW of uncomplicated pregnancies were stratified as SGA,
and postnatal growth, in others no association was found. AGA, or large for gestational age. At age 5 years, fast-
The most frequent association was seen with BP values ing insulin levels were higher in all children who had
and fasting insulin. Furthermore, the greatest adverse become heavy by that time, and the levels were high-
levels of cardiometabolic risk factors in children were est among the SGA group. Even the SGA infants who
consistently present in those who started life as LBW remained small at 5 years of age had measures of insulin
newborns but then became relatively heavy.30 It is worth resistance comparable to those who became heavy at
noting that the use of obesity criteria to detect children at 5 years, suggesting metabolic programming in the SGA
risk of future cardiometabolic risk will miss many at risk group.35 Among participants reexamined at 10 years of
persons. An upward crossing of BMI percentiles during age, children with fasting insulin levels >15 U/L had a

310   February 2021 Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

clustering of cardiometabolic risk factors: higher office pediatric normative data, this method remains a research
systolic blood pressure, higher plasma triglyceride and tool rather than a predictor in pediatrics.
uric acid levels, and lower high-density lipoprotein cho-
lesterol.36 Such findings are especially relevant, consid-

Review
ering that insulin resistance does not resolve in youth MOLECULAR TECHNIQUES AND THEIR
who are entering puberty, which may result in increased CONTRIBUTION TO UNDERSTANDING OF
cardiometabolic risk. Notably, puberty is associated with
PROGRAMMING
a marked decrease in insulin sensitivity. Little is known
about the underlying pathophysiology of insulin resis- The molecular era brought the capacity to perform rapid
tance in puberty, and how it might contribute to increased nucleic acid sequencing and microarray studies, as well
disease risk (eg, type 2 diabetes).37 as other molecular techniques and has provided the
potential for far more understanding of DOHaD. Molec-
ular and proteomic techniques have made it feasible to
Vascular Imprinting investigate the mechanisms underlying both epidemio-
Using the life course perspective concept of the con- logical data and experimental studies that have been
tinuum of aging, new elements need to be considered. heretofore phenomenological. Ultimately, such studies
There is a constant debate over whether the assess- may be able to define causality for developmental ori-
ment of arterial wall function and structure can add gins. For example, one of the most logical explanations
relevant information for further lowering the cardio- for programming presently lies in the many observa-
vascular risk beyond that from assessing conventional tions that epigenetic mechanisms play an important role
risk factors. Though there are an increasing number in DOHaD.45
of methods currently available to evaluate arterial wall An early molecular approach involved seeking variants
function and structure, those used the most in pedi- (mutations) induced by perinatal stressors. Indeed, some
atrics are arterial stiffness, intima-media thickness toxins may lead to mutations. However, finding germ-
(IMT), and endothelial function. However, information line mutations or somatic mutations in promoter regions
that links early life programming to arterial function and of genes, or in coding or noncoding areas and relating
structure is still scarce. these to DOHaD has not resulted in cogent explanations.
Arterial stiffness is a dynamic parameter that depends Much of the data that suggest that other changes,
on the arterial wall, its function and concomitant BP val- termed epigenetic changes, which do not alter DNA
Downloaded from http://ahajournals.org by on January 15, 2023

ues. An increment in arterial stiffness may be an early sequence, but, rather, accessibility to genes, in con-
marker of higher cardiovascular risk. Considered as a trast, may play a substantial role in both maternal and
core component in the concept of early vascular aging, paternal influences on the fetus. Such alterations may
it has been associated with early life factors, BW, and persist and exert not only effects on offspring but on
postnatal growth patterns.38,39 future generations.
Arterial IMT is an acknowledged marker of subclini- Epigenetics as a concept (and term) was coined by
cal vascular damage in both adults and children. Even Waddington nearly 80 years ago (1942) and was envi-
though carotid IMT has been the most frequently ana- sioned broadly.45–48 It has evolved as a field with mul-
lyzed as a target, previous studies have suggested that tiple dimensions with potential application to the area
aortic IMT may be a more discriminating measure in of DOHaD.46–48 The known manner in which epigenetic
childhood.40,41 Aortic wall thickening is increased in SGA changes occur include DNA methylation, histone modifi-
neonates compared with that of healthy newborns.40,41 cations, and changes through noncoding RNAs such as
A recent study found increased IMT both at the carotid microRNAs (miRNAs), IncRNAs, and piRNAs—which are
and thoracic aorta in 6- to -8-year-old children born SGA small, noncoding RNAs that contribute greatly to tissue-
as compared to that for those born AGA, somewhat not specific phenotype and function. Beyond that, other phe-
explained by differences in body size.42 nomena are considered epigenetic as well.
A few studies that have tested the impact of BW on Within the cell (and there are over 200 cell types in
endothelial function, measured by flow-mediated vaso- mammals), chromatin stores the DNA—packaged either
dilation induced by ischemia, indicated that children with as heterochromatin, which is closed and tightly wound,
a history of LBW show impaired endothelial function.43 and euchromatin, which is relatively open. Studies in
More recently, LBW has been associated with a decrease the 1970s showed that DNA lies within nucleosomes,
in the circulating/functional capacity of endothelial pro- where it is wrapped around dimers of histone (classified
genitor cells among healthy children 7 to 11 years of age, as H2A, H2B, H3, and H4; histone H1 does not form
independent of traditional cardiovascular risk factors.44 dimers).49 The structure of chromatin changes substan-
There are still many unanswered questions regarding the tially both during development and cell differentiation.
effect of BW on endothelial function. Indeed, because Further, processes such as acetylation (which is the
of the time requirements and the absence of reliable most extensively studied), methylation, ubiquitylation, as

Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592 February 2021   311


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

well as by ribosylation, phosphorylation, citrullination, and and hydrophobic properties of DNA. Methylation gener-
sumoylation all modify histone.45–48 ally impacts gene expression—hypermethylation lowers
Both maternal and paternal epigenetic changes expression, while hypomethylation increases it. Within
impact the fetus—the mother, through changes from the body of a gene, methylation generally increases tran-
Review

the inception of fertilization to birth (and beyond in scription by changing how compact chromatin is through
breast fed infants) and via epigenetic changes in sperm attracting proteins that complex with the methyl group
that fertilize the egg. The exposures in the environ- and, then, the DNA, silencing or enhancing transcription
ment reported to affect male germ cells include stress, factors.53 Further, modifications of histones can alter the
heavy metals, bisphenols, and endocrine disruptors, as N-terminal region of histones, which affects the conden-
well as some foods, and more, shown directly in animal sation of chromatin.
models (Figure 1). The genome’s methylation profile undergoes changes
There is now a growing literature concerning the early in embryogenesis with demethylation in the pater-
role of epigenetics in DOHaD, the majority of which has nal genome (except for paternally imprinted genes), and
been performed in experimental models, particularly in remethylation occurs.54–56 Most animal studies of epi-
rodents.50,51 There are, however, some epidemiological genetics have focused on changes in maternal protein-
data, as well as epigenome-wide association studies. calorie intake during gestation.
Taken together, such investigation is presently consid-
ered highly relevant to the observational data amassed
over the past decades.52
Methylation and Perinatal Programming of
Methylation (and demethylation) of cytosine nucleo- Hypertension
tides within the promoter region in a gene alters gene In a low protein model of induced programming Bogda-
expression. The most commonly methylated cytosines are rina et al,57 found that the angiotensin type 1b receptor
adjacent to guanosine and the event impacts hydrophilic promoter was undermethylated in the adrenal, which they
Downloaded from http://ahajournals.org by on January 15, 2023

Figure 1. Epigenetic changes and developmental origins of health and disease.


Fetal DNA sequences are not altered by epigenetic changes, yet these profoundly influence DNA configuration. The processes of DNA
methylation, post-translational histone modification, RNA modulation are involved and can affect gene availability and activation over the lifespan.

312   February 2021 Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

took as a link between fetal insults and epigenetic modi- state of the microbiome influences BP and salt-sen-
fication. The investigators hypothesized that low protein sitive hypertension. Increasingly, the concept that the
diet might decrease methyl donors, but, more plausibly, microbiome may play a role in DOHaD with respect
that stress would influence glucocorticoid production to hypertension seems appealing.74 While a number

Review
and release, affecting methylation. Goyal et al,58 using a of animal studies hint that the microbiome may play a
murine model, also found evidence that maternal protein role in the development of hypertension, such data in
deprivation led to changes in the methylation in genes humans are lacking.
of the renin-angiotensin system in the brain. They found
that CpG islands in the ACE-1 (angiotensin-converting
enzyme 1) promoter were hypomethylated, and, further
Proteomics and Perinatal Programming
found that miRNAs that influence ACE-1 mRNA trans- The structure and function of the large number of pro-
lation (mmu-mir-27a and -27b) were hypomethylated, teins within cells, organs, and whole beings is elucidated
along with decreased levels of a miRNA that regulates by proteomics; in human beings there are over 100 000
AT2 (angiotensin type 2 receptor) translation (miRNA known proteins, and presently, the use of high through-
mmu-mir-330). put techniques makes it possible to examine differen-
A number of studies that may be related to metabolic tial expression of proteins, which are emblematic of
syndrome are reviewed in Bianco-Miotto et al,59 none of physiology, pathophysiology, and treatment. Proteomic
which related to directly to the programming of hyperten- techniques are time intensive and difficult to perform in
sion per se. The degree of adiposity in adult life has been large numbers of patients, rendering widespread clinical
correlated with changes in methylation of DNA at birth or applications difficult. These techniques involve 2-dimen-
early in life, suggesting that epigenetic markers might be sional gel electrophoresis, or 2-dimensional dimensional
sought as a predictor.60–64 difference gel electrophoresis, the use of Western blots,
Payton et al65 studied miRNA in the placenta and and surface-enhanced laser desorption/ionization time-
related that to BW using placental samples from the of-flight mass spectrometry.75
Extremely Low Gestational Age Newborns cohort. They While the promise of finding biomarkers or a mean-
observed that mRNA was upregulated for particular ingful signature of protein changes that signify risk of
pathways, especially glycoprotein VI, as well as hepa- later disease in possibly at risk persons seems worthy,
tocyte growth factor and human growth factors. Using there have not been many studies of proteomics with
respect to perinatal programming.
Downloaded from http://ahajournals.org by on January 15, 2023

networking approaches, they found that BW-associated


miRNAs in human placental tissues predicted which pro- Romero et al76 studied 43 women with normal preg-
teins would be associated with individual BW. nancies that culminated in term deliveries at 3 to 6 time
In still other studies investigators found that intra- points during the pregnancy and profiled the levels of
uterine growth retardation led to methylation abnor- 1125 proteins with the use of multiplex techniques and
malities in the offspring. Maternal diabetes has been then modeled protein abundance and correlated it with
observed to result in growth restriction and epigen- gestational age. Ten percent of proteins changed signifi-
etic modulation of the Srebf2 gene in rat fetuses.66 In cantly between 8 weeks of gestation and term. Proteins
the mouse, Radford reported that intrauterine growth considered to be significantly changed were defined
retardation changed the methylation of adult sperm, as having the following45: >1.5-fold change between
affecting the metabolism in subsequent generations.67 8 and 40 weeks of gestation; and46 a false discovery
Further, Broholm et al68 found that persons with LBW rate-adjusted value of P<0.1. Gene ontology enrich-
had evidence of epigenetic changes (see recent review ment analysis was used to identify biological processes
by Arima and Fukuoka69). Recently, Felix and Cecil70 overrepresented among the proteins that changed with
have suggested that DNA methylation may explain advancing gestation. Proteins that are recognized as
much of the mechanism by which cardiovascular important in embryogenesis, growth, and angiogenesis,
changes take place through developmental program- as well as in immunoregulation and inflammation and
ming, and Chehade et al71 has summarized epigenetic host defense response changed more than 5-fold (these
changes observed in premature birth. were glypican-3, sialic acid-binding immunoglobulin-type
lectin-6, placental growth factor, C-C motif-28, carbonic
anhydrase 6, prolactin, interleukin-1 receptor 4, dual-
The Microbiome and Programming specificity). The implications for the fetus are not known,
Much recent attention has focused on the microbiome but one may speculate that such changes during preg-
and its contribution to health and disease. Within the nancy are important.
intestine, microbes interact with signaling pathways— Wang et al77 found aberrant proteomic signals in an
in the neural system, endocrine system, and general intrauterine growth retardation study in pigs, suggesting
physiology.72 The food we eat and medications we take that maternal nutrition can influence the type and abun-
alter the microbiome.73 It has been suggested that the dance of proteins that regulate gut development.

Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592 February 2021   313


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

Again, examining protein-calorie malnutrition models a metabolomic platform kit that quantified 163 metabo-
and growth-restricted kidneys Guo et al78 examined pro- lites that included free carnitine, 40 acylcarnitines, 14
files of ion channel and transporter proteins. They looked amino acids (13 proteinogenic+ornithine), hexoses
at 367 proteins that were differentially expressed and (sum of hexoses—about 90%–95% glucose), 92 glyc-
Review

then used Ingenuity Pathway Analysis. These investiga- erophospholipids (15 lysophosphatidylcholines and 77
tors found that in the kidneys of growth-restricted fetuses, phosphatidylcholines), and 15 sphingolipids. In another
a number of carriers were differentially expressed, and, study, Horgan et al88 found that the metabolic profile
they hypothesized, dysregulated. In analyzing the data, during pregnancy led to a potential phenotypic signa-
they noted 22 differentially expressed proteins that are ture for fetuses who would become SGA neonates.
known to be involved with transporters and ion channels. Such studies hint that there may be value in metabo-
They took the results to indicated that such dysregulation lomic studies that could ultimately hone down the rea-
may be involved with changes in kidney function in fetal sons for programming of hypertension; however, these
growth-restricted offspring. presently constitute, at best, pilot data.
Overall, a variety of noncoding events impact the
future health of offspring. Some of the presently known
Metabolomics and Programming factors are epigenetics, proteomics, metabolomics, the
The examination of low molecular weight compounds in microbiome and other impactful events (Figure 2).
a given biological sample to attain a picture of the com-
plete metabolites is called metabolomics.79 The involved
methodology, which requires high-resolution techniques THE AT RISK CHILD AND A
such as mass spectrometry, nuclear magnetic resonance PERSONALIZED MEDICINE APPROACH-
studies, and Fourier transform infrared spectrometry
are time and funding intensive.79 As the metabolome
FEASIBILITY
changes quickly (within seconds to minutes), the choice Presently, the phenotype of LBW for gestational age
of sampling is crucial. With respect to DOHaD, metab- and prematurity (with either LBW for gestational age,
olomic profiling has been performed in some animal birth weight appropriate for age, or high for gestational
metabolome studies and on clinical samples from reposi- age) are easily determined. The data linking individual
tories.80 Such studies have the potential to contribute a children to future risk, however, are lacking.84 However,
physiological signature to changes that may contribute to a common-sense approach is feasible, as suggested
Downloaded from http://ahajournals.org by on January 15, 2023

programming.81–84 by Bassareo et al89 in which it was suggested that both


Few perinatal programming studies have been pub- growth retardation and prematurity should be considered
lished to date. In an animal model of uterine ligation to in patient management. It would be useful to develop a
induce intrauterine growth retardation, Seferovic et al85 perinatal period of risk calculator, but the input data are
reported alterations in the metabolome that become lacking. That being said, there are some simple mea-
heritable, and that such changes occurred before the sures to apply. It is reasonable to note a list of potential
onset of metabolic syndrome in adult life. They show historical risks: BW (if low for gestational age or high
that in this model, dietary intervention may prevent for gestational age); gestational age at birth; perinatal
the metabolic syndrome. The investigators profiled the course; occurrence of severe illness or accident in child-
serum metabolome in the F2 generation at weaning and hood; the presence of obesity or overweight; the use of
later, in adulthood to see whether metabolomic altera- medications that impact kidney function or perfusion, or
tions preceded development of metabolic syndrome. cardiac and vascular health. Then, current risks can be
Indeed, such changes occurred and were alterable to assessed—current BP for age, weight for age, activity
dietary changes. level, and stress.
In a 2014 study Bassareo et al86 examined hematic If the potential risks are multiple, then such a child
asymmetrical dimethylarginine levels in a small group should be monitored carefully. It makes sense to fol-
of former extremely LBW prematures (BW <1 kg) low children and youth with increased numbers of red
and compared these to controls of term normal BW flag factors more closely and to avoid nephrotoxins and
young adults. They found that in the ex-premies the cardiotoxins.
asymmetrical dimethylarginine levels correlated with On the contrary, there are not currently sufficient data
their metabolic profiles, whereas this was not found for an individualized approach, unless there are known
among the controls.86 Such pilot studies may indicate genetic and familial risk factors.90 In thinking about
the future feasibility of such an approach, but there is hypertension and kidney disease, Barry Brenner has
not presently a signature that could be used clinically. promulgated90 that the single most important question to
In another study of 226 mother: child pairs Lu et al87 ask a patient is, what was your birth weight? For now, that
examined cord blood lysophophatidylcholine levels and question, followed by the assessment of additional (and
found that these correlated with BW. The authors used additive) risk factors would make sense.

314   February 2021 Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

Review
Downloaded from http://ahajournals.org by on January 15, 2023

Figure 2. OMICS and programming.


A variety of noncoding events impact the future health of offspring. Some of the presently known factors are epigenetics, proteomics,
metabolomics, the microbiome, and other impactful events.

see potential risk from that angle, we should resolve to


FUTURE DIRECTIONS use the developmental perspective to devise interven-
Research into the developmental origins of cardiomet- tions in time to help the next generation. Prevention
abolic risk factors and disease has reached a stage starting early in life is likely to have a very large impact
in which sufficiently robust data call for new initiatives on reducing disease incidence and its associated effects
focused on early life. New insights associated with a at the personal, economic, and societal levels.
life course perspective demand longitudinal rather than
cross-sectional studies. It is essential to design new
preconception and birth cohorts with precise pheno- ARTICLE INFORMATION
typic observations and long-term perspectives. After Affiliations
that, it will be necessary to pair them with molecu- From the Pediatric Department, Consorcio Hospital General, University of Va-
lencia (E.L.); CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto
lar studies. The identification of molecular changes, de Salud Carlos III, Spain (E.L.); and Department of Pediatrics, Harvard Medi-
together with later phenotypes, is critical to better cal School, Mass General Hospital for Children, Massachusetts General Hospital,
understanding the underlying mechanisms and to Boston (J.I.).

delineating sensitive and specific biomarkers. This can Acknowledgments


facilitate the detection of risk and monitor the efficacy We wish to thank Rob Flewell for editing the figures.
of preventive interventions to reduce the occurrence of Sources of Funding
future disease. This work has been financially supported by: Ministerio de Ciencia, Innovación
The current epidemic of cardiometabolic disease y Universidades (Grant Number PI17/01517), Instituto de Salud Carlos III and
reveals that existing interventions are insufficient, and cofinanced by the European Regional Development Fund (to E. Lurbe).

a new approach to disease prevention with initiatives Disclosures


emphasizing on early development is required. When we None.

Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592 February 2021   315


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

REFERENCES 22. Eriksson JG, Forsén T, Tuomilehto J, Osmond C, Barker DJ. Early growth,
adult income, and risk of stroke. Stroke. 2000;31:869–874. doi: 10.1161/
1. Barker DJ, Osmond C, Golding J, Kuh D, Wadsworth ME. Growth in utero, 01.str.31.4.869
blood pressure in childhood and adult life, and mortality from cardiovascular 23. Lawlor DA, Davey Smith G, Ebrahim S. Birth weight is inversely associated
disease. BMJ. 1989;298:564–567. doi: 10.1136/bmj.298.6673.564 with coronary heart disease in post-menopausal women: findings from the
Review

2. Barker JP, Osmond C, Winter PD, Margetts B, Simmonds SJ. Weigh in British women’s heart and health study. J Epidemiol Community Health.
infancy and death from ischaemic heart disease. Lancet. 1989;2:577–580. 2004;58:120–125. doi: 10.1136/jech.58.2.120
doi: 10.1016/s0140-6736(89)90710-1 24. Knop MR, Geng TT, Gorny AW, Ding R, Li C, Ley SH, Huang T. Birth weight
3. Barker DJ, Osmond C, Forsén TJ, Kajantie E, Eriksson JG. Trajectories of and risk of type 2 diabetes mellitus, cardiovascular disease, and hyperten-
growth among children who have coronary events as adults. N Engl J Med. sion in adults: a meta-analysis of 7 646 267 participants from 135 studies.
2005;353:1802–1809. doi: 10.1056/NEJMoa044160
J Am Heart Assoc. 2018;7:e008870. doi: 10.1161/JAHA.118.008870
4. Gluckman PD, Hanson MA, Cooper C, Thornburg KL. Effect of in utero
25. Sipola-Leppänen M, Vääräsmäki M, Tikanmäki M, Matinolli HM, Miettola S,
and early-life conditions on adult health and disease. N Engl J Med.
Hovi P, Wehkalampi K, Ruokonen A, Sundvall J, Pouta A, et al. Cardiometa-
2008;359:61–73. doi: 10.1056/NEJMra0708473
bolic risk factors in young adults who were born preterm. Am J Epidemiol.
5. Andersen LG, Angquist L, Eriksson JG, Forsen T, Gamborg M, Osmond C,
2015;181:861–873. doi: 10.1093/aje/kwu443
Baker JL, Sørensen TI. Birth weight, childhood body mass index and risk of
26. Parkinson JR, Hyde MJ, Gale C, Santhakumaran S, Modi N. Preterm birth
coronary heart disease in adults: combined historical cohort studies. PLoS
and the metabolic syndrome in adult life: a systematic review and meta-anal-
One. 2010;5:e14126. doi: 10.1371/journal.pone.0014126
ysis. Pediatrics. 2013;131:e1240–e1263. doi: 10.1542/peds.2012-2177
6. Fujita Y, Kouda K, Nakamura H, Iki M. Association of rapid weight gain dur-
27. Juonala M, Cheung MM, Sabin MA, Burgner D, Skilton MR, Kähönen M,
ing early childhood with cardiovascular risk factors in Japanese adolescents.
Hutri-Kähönen N, Lehtimäki T, Jula A, Laitinen T, et al. Effect of birth weight
J Epidemiol. 2013;23:103–108. doi: 10.2188/jea.je20120107
on life-course blood pressure levels among children born premature: the
7. Lurbe E, Agabiti-Rosei E, Cruickshank JK, Dominiczak A, Erdine S,
cardiovascular risk in young finns study. J Hypertens. 2015;33:1542–1548.
Hirth A, Invitti C, Litwin M, Mancia G, Pall D, et al. 2016 European soci-
doi: 10.1097/HJH.0000000000000612
ety of hypertension guidelines for the management of high blood pres-
28. Markopoulou P, Papanikolaou E, Analytis A, Zoumakis E, Siahanidou T.
sure in children and adolescents. J Hypertens. 2016;34:1887–1920. doi:
Preterm birth as a risk factor for metabolic syndrome and cardiovascu-
10.1097/HJH.0000000000001039
lar disease in adult life: a systematic review and meta-analysis. J Pediatr.
8. Prevalence and change from 1990-2017 of low birth weight in the OECD
2019;210:69–80.e5. doi: 10.1016/j.jpeds.2019.02.041
countries. OCDE Family Database http://www.oecd.org/els/family/data-
29. Chambers BD, Baer RJ, McLemore MR, Jelliffe-Pawlowski LL. Using
base.htm Updated July 2019. OECD-Social Policy Division-Directorate of
index of concentration at the extremes as indicators of structural racism
Employment, Labour and Social Affair. Accessed October 9, 2020.
to evaluate the association with preterm birth and infant mortality-Cal-
9. Fall CHD, Kumaran K. Metabolic programming in early life in humans.
ifornia, 2011-2012. J Urban Health. 2019;96:159–170. doi: 10.1007/
Philos Trans R Soc Lond B Biol Sci. 2019;374:20180123. doi: 10.1098/
s11524-018-0272-4
rstb.2018.0123
30. Kelishadi R, Haghdoost AA, Jamshidi F, Aliramezany M, Moosazadeh M.
10. Godfrey KM, Reynolds RM, Prescott SL, Nyirenda M, Jaddoe VW,
Low birthweight or rapid catch-up growth: which is more associated with
Eriksson JG, Broekman BF. Influence of maternal obesity on the long-
term health of offspring. Lancet Diabetes Endocrinol. 2017;5:53–64. doi: cardiovascular disease and its risk factors in later life? A systematic review
10.1016/S2213-8587(16)30107-3 and cryptanalysis. Paediatr Int Child Health. 2015;35:110–123. doi:
10.1179/2046905514Y.0000000136
Downloaded from http://ahajournals.org by on January 15, 2023

11. Gaillard R, Santos S, Duijts L, Felix JF. Childhood health consequences


of maternal obesity during pregnancy: a narrative review. Ann Nutr Metab. 31. Falkner B, Lurbe E. Primordial prevention of high blood pressure in child-
2016;69:171–180. doi: 10.1159/000453077 hood: an opportunity not to be missed. Hypertension. 2020;75:1142–1150.
12. International Diabetes Federation. 2017 IDFDiabetes Atlas, 8th edn. Brus- doi: 10.1161/HYPERTENSIONAHA.119.14059
sels, Belgium: International Diabetes Federation. 32. Steinberger J, Daniels SR, Hagberg N, Isasi CR, Kelly AS, Lloyd-Jones D,
13. Mu M, Wang SF, Sheng J, Zhao Y, Li HZ, Hu CL, Tao FB. Birth weight Pate RR, Pratt C, Shay CM, Towbin JA, et al. American heart associa-
and subsequent blood pressure: a meta-analysis. Arch Cardiovasc Dis. tion atherosclerosis, hypertension, and obesity in the young commit-
2012;105:99–113. doi: 10.1016/j.acvd.2011.10.006 tee of the council on cardiovascular disease in the young; council on
14. Gamborg M, Byberg L, Rasmussen F, Andersen PK, Baker JL, Bengtsson C, cardiovascular and stroke nursing; council on epidemiology and pre-
Canoy D, Drøyvold W, Eriksson JG, Forsén T, et al; NordNet Study Group. vention; council on functional genomics and translational biology; and
Birth weight and systolic blood pressure in adolescence and adulthood: stroke council. cardiovascular health promotion in children: challenges
meta-regression analysis of sex- and age-specific results from 20 Nordic and opportunities for 2020 and beyond: a scientific statement from the
studies. Am J Epidemiol. 2007;166:634–645. doi: 10.1093/aje/kwm042 American Heart Association. Circulation. 2016;134:e236–e255. doi:
15. Harder T, Rodekamp E, Schellong K, Dudenhausen JW, Plagemann A. Birth 10.1161/CIR.0000000000000441.
weight and subsequent risk of type 2 diabetes: a meta-analysis. Am J Epi- 33. O’Connor TG, Williams J, Blair C, Gatzke-Kopp LM, Francis L, Willoughby MT.
demiol. 2007;165:849–857. doi: 10.1093/aje/kwk071 Predictors of developmental patterns of obesity in young children. Front
16. Whincup PH, Kaye SJ, Owen CG, Huxley R, Cook DG, Anazawa S, Pediatr. 2020;8:109. doi: 10.3389/fped.2020.00109
Barrett-Connor E, Bhargava SK, Birgisdottir BE, Carlsson S, et al. Birth weight 34. Yi L, Pearce A, Li L. Weight gain in early years and subsequent body mass
and risk of type 2 diabetes: a systematic review. JAMA. 2008;300:2886– index trajectories across birth weight groups: a prospective longitudinal
2897. doi: 10.1001/jama.2008.886 study. Eur J Public Health. 2020;30:316–322. doi: 10.1093/eurpub/ckz232
17. Tian G, Guo C, Li Q, Liu Y, Sun X, Yin Z, Li H, Chen X, Liu X, Zhang D, et 35. Lurbe E, Garcia-Vicent C, Torro MI, Aguilar F, Redon J. Associations
al. Birth weight and risk of type 2 diabetes: a dose-response meta-anal- of birth weight and postnatal weight gain with cardiometabolic risk
ysis of cohort studies. Diabetes Metab Res Rev. 2019;35:e3144. doi: parameters at 5 years of age. Hypertension. 2014;63:1326–1332. doi:
10.1002/dmrr.3144 10.1161/HYPERTENSIONAHA.114.03137
18. Sun D, Wang T, Heianza Y, Huang T, Shang X, Lv J, Li S, Harville E, Chen W, 36. Lurbe E, Aguilar F, Álvarez J, Redon P, Torró MI, Redon J. Determi-
Fonseca V, et al. Birthweight and cardiometabolic risk patterns in multiracial nants of cardiometabolic risk factors in the first decade of life: a lon-
children. Int J Obes (Lond). 2018;42:20–27. doi: 10.1038/ijo.2017.196 gitudinal study starting at birth. Hypertension. 2018;71:437–443. doi:
19. Ferrie JE, Langenberg C, Shipley MJ, Marmot MG. Birth weight, compo- 10.1161/HYPERTENSIONAHA.117.10529
nents of height and coronary heart disease: evidence from the Whitehall II 37. Kelsey MM, Zeitler PS. Insulin resistance of puberty. Curr Diab Rep.
study. Int J Epidemiol. 2006;35:1532–1542. doi: 10.1093/ije/dyl184 2016;16:64. doi: 10.1007/s11892-016-0751-5
20. Hubinette A, Cnattingius S, Johansson AL, Henriksson C, Lichtenstein P. 38. Sperling J, Nilsson PM. Does early life programming influence arterial stiff-
Birth weight and risk of angina pectoris: analysis in Swedish twins. Eur J ness and central hemodynamics in adulthood? J Hypertens. 2020;38:481–
Epidemiol. 2003;18:539–544. doi: 10.1023/a:1024686625528 488. doi: 10.1097/HJH.0000000000002292
21. Osler M, Lund R, Kriegbaum M, Andersen AM. The influence of birth weight 39. Stock K, Schmid A, Griesmaier E, Gande N, Hochmayr C, Knoflach M,
and body mass in early adulthood on early coronary heart disease risk Kiechl-Kohlendorfer U; Early Vascular Aging (EVA) Study Group. The impact of
among Danish men born in 1953. Eur J Epidemiol. 2009;24:57–61. doi: being born preterm or small for gestational age on early vascular aging in ado-
10.1007/s10654-008-9301-z lescents. J Pediatr. 2018;201:49–54.e1. doi: 10.1016/j.jpeds.2018.05.056

316   February 2021 Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

40. Skilton MR, Evans N, Griffiths KA, Harmer JA, Celermajer DS. Aortic BMI and gestational weight gain, offspring DNA methylation and later off-
wall thickness in newborns with intrauterine growth restriction. Lancet. spring adiposity: findings from the Avon longitudinal study of parents and
2005;365:1484–1486. doi: 10.1016/S0140-6736(05)66419-7 children. Int J Epidemiol. 2015;44:1288–1304. doi: 10.1093/ije/dyv042
41. Sehgal A, Doctor T, Menahem S. Cardiac function and arterial biophysical 63. Tobi EW, Goeman JJ, Monajemi R, Gu H, Putter H, Zhang Y, Slieker RC,
properties in small for gestational age infants: postnatal manifestations Stok AP, Thijssen PE, Müller F, et al. DNA methylation signatures link prena-

Review
of fetal programming. J Pediatr. 2013;163:1296–1300. doi: 10.1016/j. tal famine exposure to growth and metabolism. Nat Commun. 2014;5:5592.
jpeds.2013.06.030 doi: 10.1038/ncomms6592
42. Muñiz M, Oulego I, Revilla D, Muñoz A, Rodriguez A, Lurbe E. Thoracic aortic 64. Dominguez-Salas P, Moore SE, Baker MS, Bergen AW, Cox SE, Dyer RA,
intima-media thickness in preschool children born small for gestational age. Fulford AJ, Guan Y, Laritsky E, Silver MJ, et al. Maternal nutrition at concep-
J Pediatr. 2019;208:81–88.e2. doi: 10.1016/j.jpeds.2018.12.037 tion modulates DNA methylation of human metastable epialleles. Nat Com-
43. Franco MC, Christofalo DM, Sawaya AL, Ajzen SA, Sesso R. Effects of low mun. 2014;5:3746. doi: 10.1038/ncomms4746
birth weight in 8- to 13-year-old children: implications in endothelial func- 65. Payton A, Clark J, Eaves L, Santos HP Jr, Smeester L, Bangma JT,
tion and uric acid levels. Hypertension. 2006;48:45–50. doi: 10.1161/01. O’Shea TM, Fry RC, Rager JE. Placental genomic and epigenomic signa-
HYP.0000223446.49596.3a tures associated with infant birth weight highlight mechanisms involved in
44. Souza LV, De Meneck F, Oliveira V, Higa EM, Akamine EH, Franco MDC. collagen and growth factor signaling. Reprod Toxicol. 2020;96:221–230.
Detrimental impact of low birth weight on circulating number and functional doi: 10.1016/j.reprotox.2020.07.007
capacity of endothelial progenitor cells in healthy children: role of angiogenic 66. Golic M, Stojanovska V, Bendix I, Wehner A, Herse F, Haase N, Kräker K,
factors. J Pediatr. 2019;206:72–77.e1. doi: 10.1016/j.jpeds.2018.10.040 Fischer C, Alenina N, Bader M, et al. Diabetes mellitus in pregnancy leads
45. Vinci G, Buffat C, Simoncini S, Boubred F, Ligi I, Dumont F, Le Bonniec B, to growth restriction and epigenetic modification of the Srebf2 gene
Fournier T, Vaiman D, Dignat-George F, et al. Gestational age-related patterns in rat fetuses. Hypertension. 2018;71:911–920. doi: 10.1161/
of AMOT methylation are revealed in preterm infant endothelial progenitors. HYPERTENSIONAHA.117.10782
PLoS One. 2017;12:e0186321. doi: 10.1371/journal.pone.0186321 67. Radford EJ, Ito M, Shi H, Corish JA, Yamazawa K, Isganaitis E, Seisenberger S,
46. McKay JA, Waltham KJ, Williams EA, Mathers JC. Folate depletion dur- Hore TA, Reik W, Erkek S, et al. In utero effects. In utero undernourishment
ing pregnancy and lactation reduces genomic DNA methylation in perturbs the adult sperm methylome and intergenerational metabolism. Sci-
murine adult offspring. Genes Nutr. 2011;6:189–196. doi: 10.1007/ ence. 2014;345:1255903. doi: 10.1126/science.1255903
s12263-010-0199-1 68. Broholm C, Olsson AH, Perfilyev A, Hansen NS, Schrölkamp M, Strasko KS,
47. Kunes J, Zicha J. The interaction of genetic and environmental factors in the Scheele C, Ribel-Madsen R, Mortensen B, Jørgensen SW, et al. Epigenetic
etiology of hypertension. Physiol Res. 2009;58(suppl 2):33-41. programming of adipose-derived stem cells in low birthweight individuals.
48. Nadeau JH. Transgenerational genetic effects on phenotypic variation and dis- Diabetologia. 2016;59:2664–2673. doi: 10.1007/s00125-016-4099-9
ease risk. Hum Mol Genet. 2009;18:R202–R210. doi: 10.1093/hmg/ddp366
69. Arima Y, Fukuoka H. Developmental origins of health and disease theory in
49. Sperling R, Bustin M. Histone dimers: a fundamental unit in histone assem-
cardiology. J Cardiol. 2020;76:14–17. doi: 10.1016/j.jjcc.2020.02.003
bly. Nucleic Acids Res. 1976;3:1263–1275. doi: 10.1093/nar/3.5.1263
70. Felix JF, Cecil CAM. Population DNA methylation studies in the Develop-
50. Spadafora C. Transgenerational epigenetic reprogramming of early
mental Origins of Health and Disease (DOHaD) framework. J Dev Orig
embryos: a mechanistic model. Environ Epigenet. 2020;6:dvaa009. doi:
Health Dis. 2019;10:306–313. doi: 10.1017/S2040174418000442
10.1093/eep/dvaa009
71. Chehade H, Simeoni U, Guignard JP, Boubred F. Preterm birth: long term
51. Briffa JF, Wlodek ME, Moritz KM. Transgenerational programming of neph-
cardiovascular and renal consequences. Curr Pediatr Rev. 2018;14:219–
ron deficits and hypertension. Semin Cell Dev Biol. 2020;103:94–103. doi:
226. doi: 10.2174/1573396314666180813121652
10.1016/j.semcdb.2018.05.025
Downloaded from http://ahajournals.org by on January 15, 2023

72. Elijovich F, Laffer CL, Sahinoz M, Pitzer A, Ferguson JF, Kirabo A. The gut
52. Adli M, Parlak M, Li Y, El-Dahr SS. Epigenetic states of nephron progeni-
microbiome, inflammation, and salt-sensitive hypertension. Curr Hypertens
tors and epithelial differentiation. J Cell Biochem. 2015;116:893–902. doi:
Rep. 2020;22:79. doi: 10.1007/s11906-020-01091-9
10.1002/jcb.25048
73. Mischke M, Plösch T. The gut microbiota and their metabolites: potential
53. Yang X, Han H, De Carvalho DD, Lay FD, Jones PA, Liang G. Gene body
implications for the host epigenome. Adv Exp Med Biol. 2016;902:33–44.
methylation can alter gene expression and is a therapeutic target in cancer.
doi: 10.1007/978-3-319-31248-4_3
Cancer Cell. 2014;26:577–590. doi: 10.1016/j.ccr.2014.07.028
74. Simeoni U, Armengaud JB, Siddeek B, Tolsa JF. Perinatal origins of adult
54. Gluckman PD, Hanson MA, Buklijas T, Low FM, Beedle AS. Epigenetic
disease. Neonatology. 2018;113:393–399. doi: 10.1159/000487618
mechanisms that underpin metabolic and cardiovascular diseases. Nat Rev
Endocrinol. 2009;5:401–408. doi: 10.1038/nrendo.2009.102 75. Vora N, Kalagiri R, Mallett LH, Oh JH, Wajid U, Munir S, Colon N, Raju VN,
55. Raychaudhuri N, Raychaudhuri S, Thamotharan M, Devaskar SU. Histone Beeram MR, Uddin MN. Proteomics and metabolomics in pregnancy-an
code modifications repress glucose transporter 4 expression in the intra- overview. Obstet Gynecol Surv. 2019;74:111–125. doi: 10.1097/OGX.
uterine growth-restricted offspring. J Biol Chem. 2008;283:13611–13626. 0000000000000646
doi: 10.1074/jbc.M800128200 76. Romero R, Erez O, Maymon E, Chaemsaithong P, Xu Z, Pacora P,
56. Nistala R, Hayden MR, Demarco VG, Henriksen EJ, Lackland DT, Chaiworapongsa T, Done B, Hassan SS, Tarca AL. The maternal plasma
Sowers JR. Prenatal programming and epigenetics in the genesis of the proteome changes as a function of gestational age in normal pregnancy:
cardiorenal syndrome. Cardiorenal Med. 2011;1:243–254. doi: 10.1159/ a longitudinal study. Am J Obstet Gynecol. 2017;217:67.e1–67.e21. doi:
000332756 10.1016/j.ajog.2017.02.037
57. Bogdarina I, Welham S, King PJ, Burns SP, Clark AJ. Epigenetic modification 77. Wang X, Lin G, Liu C, Feng C, Zhou H, Wang T, Li D, Wu G, Wang J. Tem-
of the renin-angiotensin system in the fetal programming of hypertension. Circ poral proteomic analysis reveals defects in small-intestinal development
Res. 2007;100:520–526. doi: 10.1161/01.RES.0000258855.60637.58 of porcine fetuses with intrauterine growth restriction. J Nutr Biochem.
58. Goyal R, Goyal D, Leitzke A, Gheorghe CP, Longo LD. Brain renin- 2014;25:785–795. doi: 10.1016/j.jnutbio.2014.03.008
angiotensin system: fetal epigenetic programming by maternal pro- 78. Guo Y, Lu Y, Wang J, Zhu L, Liu X. Dysregulated ion channels and trans-
tein restriction during pregnancy. Reprod Sci. 2010;17:227–238. doi: porters activate endoplasmic reticulum stress in rat kidney of fetal growth
10.1177/1933719109351935 restriction. Life Sci. 2020;259:118276. doi: 10.1016/j.lfs.2020.118276
59. Bianco-Miotto T, Craig JM, Gasser YP, van Dijk SJ, Ozanne SE. Epigenetics 79. Hodyl NA, Muhlhausler B. Novel insights, challenges and practical implica-
and DOHaD: from basics to birth and beyond. J Dev Orig Health Dis. tions of DOHaD-omics research. Med J Aust. 2016;204:108–10.e1. doi:
2017;8:513–519. doi: 10.1017/S2040174417000733 10.5694/mja14.01626
60. Clarke-Harris R, Wilkin TJ, Hosking J, Pinkney J, Jeffery AN, Metcalf BS, 80. Hivert MF, Perng W, Watkins SM, Newgard CS, Kenny LC, Kristal BS,
Godfrey KM, Voss LD, Lillycrop KA, Burdge GC. PGC1α promoter methyla- Patti ME, Isganaitis E, DeMeo DL, Oken E, et al. Metabolomics in the devel-
tion in blood at 5-7 years predicts adiposity from 9 to 14 years (EarlyBird opmental origins of obesity and its cardiometabolic consequences. J Dev
50). Diabetes. 2014;63:2528–2537. doi: 10.2337/db13-0671 Orig Health Dis. 2015;6:65–78. doi: 10.1017/S204017441500001X
61. Godfrey KM, Sheppard A, Gluckman PD, Lillycrop KA, Burdge GC, 81. Holmes E, Loo RL, Stamler J, Bictash M, Yap IK, Chan Q, Ebbels T, De Iorio M,
McLean C, Rodford J, Slater-Jefferies JL, Garratt E, Crozier SR, et al. Epi- Brown IJ, Veselkov KA, et al. Human metabolic phenotype diversity and its
genetic gene promoter methylation at birth is associated with child’s later association with diet and blood pressure. Nature. 2008;453:396–400. doi:
adiposity. Diabetes. 2011;60:1528–1534. doi: 10.2337/db10-0979 10.1038/nature06882
62. Sharp GC, Lawlor DA, Richmond RC, Fraser A, Simpkin A, Suderman M, 82. Sulek K, Han TL, Villas-Boas SG, Wishart DS, Soh SE, Kwek K, Gluckman PD,
Shihab HA, Lyttleton O, McArdle W, Ring SM, et al. Maternal pre-pregnancy Chong YS, Kenny LC, Baker PN. Hair metabolomics: identification of fetal

Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592 February 2021   317


Lurbe and Ingelfinger Early Life Origins of Cardiometabolic Risk Factors

compromise provides proof of concept for biomarker discovery. Theranos- 87. Lu YP, Reichetzeder C, Prehn C, Yin LH, Yun C, Zeng S, Chu C,
tics. 2014;4:953–959. doi: 10.7150/thno.9265 Adamski J, Hocher B. Cord blood lysophosphatidylcholine 16: 1 is positively
83. Crisafulli A, Bassareo PP, Kelleher S, Calcaterra G, Mercuro G. Factors predis- associated with birth weight. Cell Physiol Biochem. 2018;45:614–624. doi:
posing to hypertension in subjects formerly born preterm: renal impairment, 10.1159/000487118
arterial stiffness, endothelial dysfunction or something else? Curr Hypertens 88. Horgan RP, Broadhurst DI, Walsh SK, Dunn WB, Brown M, Roberts CT,
Review

Rev. 2020;16:82–90. doi: 10.2174/1573402115666190627140523 North RA, McCowan LM, Kell DB, Baker PN, et al. Metabolic profil-
84. Pintus R, Bassareo PP, Dessì A, Deidda M, Mercuro G, Fanos V. Metabo- ing uncovers a phenotypic signature of small for gestational age
lomics and cardiology: toward the path of perinatal programming and per- in early pregnancy. J Proteome Res. 2011;10:3660–3673. doi:
sonalized medicine. Biomed Res Int. 2017;2017:6970631. doi: 10.1155/ 10.1021/pr2002897
2017/6970631 89. Bassareo PP, Marras AR, Cugusi L, Zedda AM, Mercuro G. The reasons
85. Seferovic MD, Goodspeed DM, Chu DM, Krannich LA, Gonzalez-Rodriguez why cardiologists should consider prematurity at birth and intrauterine
PJ, Cox JE, Aagaard KM. Heritable IUGR and adult metabolic syndrome growth retardation among risk factors. J Cardiovasc Med (Hagerstown).
are reversible and associated with alterations in the metabolome follow- 2016;17:323–329. doi: 10.2459/JCM.0000000000000338
ing dietary supplementation of 1-carbon intermediates. Faseb J. 2015; 90. Luyckx VA, Perico N, Somaschini M, Manfellotto D, Valensise H, Cetin I,
29:2640–2652. doi: 10.1096/fj.14-266387 Simeoni U, Allegaert K, Vikse BE, Steegers EA, et al; writing group of the
86. Bassareo PP, Fanos V, Noto A, Solla P, Barberini L, Flore G, Puddu M, Low Birth Weight and Nephron Number Working Group. A developmental
Mercuro G. Clinical metabolomics and hematic ADMA predict the future onset approach to the prevention of hypertension and kidney disease: a report
of cardiorenal syndrome in young grown-up subjects who were born preterm. from the low birth weight and nephron number working group. Lancet.
Clin Biochem. 2014;47:423–426. doi: 10.1016/j.clinbiochem.2013.11.018 2017;390:424–428. doi: 10.1016/S0140-6736(17)30576-7
Downloaded from http://ahajournals.org by on January 15, 2023

318   February 2021 Hypertension. 2021;77:308–318. DOI: 10.1161/HYPERTENSIONAHA.120.14592

You might also like