Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Expert Reviews ajog.

org

Iron deficiency in pregnancy


Michael K. Georgieff, MD

the unit. Beyond oxygen delivery, iron in


Iron is essential for the function of all cells through its roles in oxygen delivery, electron cytochromes catalyzes the generation of
transport, and enzymatic activity. Cells with high metabolic rates require more iron and ATP at a time when the fetal oxygen con-
are at greater risk for dysfunction during iron deficiency. Iron requirements during sumption rate is very high, driven largely
pregnancy increase dramatically, as the mother’s blood volume expands and the fetus by the structural development of fetal or-
grows and develops. Thus, pregnancy is a condition of impending or existing iron gans.2 Of these, the brain is particularly
deficiency, which may be difficult to diagnose because of limitations to commonly used “greedy,” accounting for an astounding
biomarkers such as hemoglobin and ferritin concentrations. Iron deficiency is associated 60% of the total fetal oxygen consumption
with adverse pregnancy outcomes, including increased maternal illness, low birthweight, rate.2 This high human fetal brain oxygen
prematurity, and intrauterine growth restriction. The rapidly developing fetal brain is at consumption rate is due to the structural
particular risk of iron deficiency, which can occur because of maternal iron deficiency, development of both neurons and glia and
hypertension, smoking, or glucose intolerance. Low maternal gestational iron intake is far exceeds the brain oxygen consumption
associated with autism, schizophrenia, and abnormal brain structure in the offspring. rate of other mammalian species.2 The
Newborns with iron deficiency have compromised recognition memory, slower speed of fetal brain develops from a smooth bilobed
processing, and poorer bonding that persist despite postnatal iron repletion. Preclinical structure at 24 weeks of gestation to a
models of fetal iron deficiency confirm that expected iron-dependent processes such as complex sulcated and gyrated structure at
monoamine neurotransmission, neuronal growth and differentiation, myelination, and term that looks far more like a mature
gene expression are all compromised acutely and long term into adulthood. This review adult brain than like its earlier fetal coun-
outlines strategies to diagnose and prevent iron deficiency in pregnancy. It describes the terpart.3 The increased surface complexity
neurocognitive and mental health consequences of fetal iron deficiency. It emphasizes reflects the massive cellular differentiation
that fetal iron is a key nutrient that influences brain development and function across the of the brain in multiple brain regions and
lifespan. processes that include the hippocampus,
the striatum, and the process of
Key words: anemia, biomarkers, brain, brain development, epigenetics, ferritin, fetal myelination.4
growth restriction, fetus, gestational diabetes, hemoglobin, hepcidin, hippocampus, iron,
iron deficiency, mental health, metabolism, nutrition neurodevelopmental disorder, Why Does Pregnancy Increase the
placenta, pregnancy, prematurity, preterm birth Need for Iron?
The onset of pregnancy can be consid-
ered a case of impending iron deficiency,
Why Is Iron Important in Biology and from the extracellular environment by as evidenced the high rate of the disorder
MaternaleFetal Medicine? plants, yeast, and mammals is remark- during gestation. In low- and middle-
Iron is so fundamental to cellular func- ably similar. Mammalian cells incorpo- income countries, the rate approaches
tions that its uptake by cells is a highly rate iron into the porphyrin ring of 80%, whereas estimated rates in well-
conserved process across species. The hemoproteins as well as into enzymes, resourced countries approach 45%
mechanism by which iron is taken up where the iron moiety is essential for depending on the sensitivity of the
those enzymes’ activities. Examples of markers used to detect it.5,6 The serum
the former include hemoglobin, concentration of hepcidin, which is
From the Division of Neonatology, Department myoglobin, and cytochromes. Examples regulated by maternal iron status, is
of Pediatrics, University of Minnesota Medical
of the latter encompass a wide range of extremely low during pregnancy.1 Since
School, Minneapolis, MN.
enzymes from hydroxylases that mediate hepcidin is a negative regulator of in-
Received Jan. 24, 2020; revised Feb. 28, 2020;
accepted March 3, 2020.
fundamental intermediary cellular testinal iron absorption, a low level in-
metabolism to demethylases that modify dicates a high requirement for iron.1
The authors report no conflict of interest.
the chromatin of DNA and thus gene Pregnancy increases maternal iron
The work was supported by grants 5R01 HD-
29421, R01 NS-099178, R01 HD-089989, and
expression. demand for 3 reasons. Maternal plasma
R01 HD-094809 from the National Institutes of Iron sufficiency is essential for oxygen and blood volumes are increased during
Health. delivery to the maternaleplacentalefetal pregnancy.1 Each extra gram of hemo-
Corresponding author: Michael K. Georgieff, unit to support the increased oxygen globin that the mother synthesizes re-
MD. georg001@umn.edu consumption demand of pregnancy.1 quires an addition 3.46 mg of elemental
0002-9378/$36.00 Maintenance of adequate maternal he- iron. In addition, the fetus requires iron
ª 2020 Elsevier Inc. All rights reserved. moglobin concentrations supports the for its own metabolic and oxygen de-
https://doi.org/10.1016/j.ajog.2020.03.006
oxygen demands of the 3 components of livery needs as well as the loading of its

516 American Journal of Obstetrics & Gynecology OCTOBER 2020


ajog.org Expert Reviews

comparatively large endogenous iron compartments; red cells, storage iron including the brain (Figure 1).34e37
stores that will be used in the first 6 mostly in the liver, and non-heme, non- Consequently, anemia represents the
months of postnatal life.7,8 The placenta storage tissue iron.7 Red cells contain 55 end stage of the iron deficiency process
is a highly metabolically active organ mg of iron per kilogram body weight as and is not a sensitive marker of tissue
with large iron requirements. It has the hemoglobin and this represents the level iron deficiency including brain iron
capacity to store iron in resident reticu- largest compartment. The amount of deficiency.36 Because of the prioritiza-
loendothelial cells to provide a buffer fetal iron in hemoglobin can be esti- tion of fetal iron to the red cells, the iron
against periods of low maternal iron mated by assuming 3.46 mg of iron per status of these tissues must be inferred
supply.7e9 Overall, pregnancy requires gram of hemoglobin and a blood volume from the serum ferritin.34 Iron status of
an additional 1 g of iron, relatively of 85 mL per kilogram body weight.7 the fetal and neonatal brain can be
equally divided between the mother and Normal values for hemoglobin concen- assessed from the serum ferritin con-
the fetus. tration in term babies range from 133 to centration and the demonstrated rela-
184 g/L.23 Risks for fetal anemia include tionship between reduced liver iron
Postnatal Iron Status During Early acute fetalematernal or fetaleplacental stores and compromised brain iron sta-
Childhood Depends on Fetal Iron hemorrhage and severe maternal iron tus.30,37 Fundamentally, tissue iron is at
Loading deficiency when maternal hemoglobin risk once fetal iron stores have been
Postnatal iron deficiency is extremely concentration is less than 90 g/L.24 exhausted.30,34,37 To that end, newborn
common in infants and toddlers, with Iron stores are found mostly in the ferritin concentrations <76 mg/L (25th
rates approaching 80% in low- and liver at the end of gestation and represent percentile) in humans have been asso-
middle-income countries. In well-resource 12 mg per kilogram body weight.7 Iron ciated with abnormal neonatal neuro-
countries in Europe and in the United stores are proportionately larger in ne- logic function including poorer
States, the rate of iron deficiency in chil- onates than in older infants, children, recognition memory15,17 and slower
dren ranges from 9% to 45%.10,11 Post- and adults. The mean cord serum speed of processing (longer interwave
natal iron deficiency has been convincingly ferritin concentration of the term infant latencies) on brainstem evoked
linked to short- and long-term neuro- 170 mg/L and the fifth percentile is responses.16
developmental abnormalities.12 59.13,23 Storage iron can be estimated The maternal gestational conditions
Until recently, postnatal iron defi- from the serum ferritin concentration by that compromise fetal and neonatal iron
ciency was thought to be due to a com- using nomograms that relate them to status are remarkably common,26 yet
bination of poor dietary iron intake and liver iron content.25 Negative fetal iron routine measurement of iron status in the
blood loss (due to intestinal infections). balance can be recognized clinically by a neonate is not performed. Conversely, the
However, a recent large randomized trial low cord serum ferritin concentration, fully iron loaded neonate, who receives
of pregnant women in a Chinese popu- which indicates loss of fetal iron stores.26 the benefit of delayed cord clamping and
lation with a moderate rate of iron Reduced ferritin concentrations occur in is growing at the rate described by the
deficiency demonstrated that postnatal infants born to iron-deficient mothers World Health Organization growth
iron deficiency in the offspring was with serum ferritin concentrations curves, has enough iron to maintain
driven to a large degree by the iron status <13.4 mg/L,13 iron sufficient mothers adequate iron delivery to supply the
of the neonate and thus was a function of with hypertension during gestation,27 growing tissues and expanding red cell
fetal iron loading.13 Previous studies had mothers who smoke cigarettes,28 and mass for up to 4e6 months.38 Delayed
shown that neonatal non-anemic iron mothers with glucose intolerance/dia- cord clamping improves hemoglobin
deficiency, as defined by low cord serum betes mellitus during pregnancy.29,30 status at 2 months of age.39
ferritin concentrations, negatively affects Twins can have discordant iron loading
neurologic and behavioral functions in where one twin may be profoundly iron Biomarkers of Iron Status and
the neonatal period and confers long- deficient.31 Diagnosis of Iron Deficiency in
term risks to neurodevelopment.14e19 The smallest compartment is made up Pregnant Women
Children born with lower fetal iron of the non-heme, non-storage tissues, Traditionally, the measurement of he-
loading have lower iron stores at 9 months including the brain and the heart, and moglobin concentration has been used
of age and a greater risk of postnatal iron comprises approximately 8 mg per ki- for patients of all ages to screen for iron
deficiency.20,21 Thus, proper fetal iron logram body weight.7 No biomarkers status because so much of anemia
loading has taken on greater importance index this compartment, yet much of the worldwide is due to iron deficiency. The
in public health strategies to combat symptomatology of iron deficiency, wide availability, ease of measurement,
postnatal iron deficiency and its long- including fatigue and altered brain and low cost of hemoglobin measure-
term neurodevelopmental risks. function, stem from iron deficiency at ment are advantages particularly in low-
The fetus maintains a relatively con- the tissue level.32,33 resource areas. Normal values for
stant 75 mg of iron per kilogram of body An important concept of fetal iron hemoglobin concentration are published
iron during the last trimester.7,22 Fetal metabolism is that iron is prioritized to and are easily accessed so that the diag-
iron can be divided into 3 red cells over all other compartments nosis of anemia is not problematic.

OCTOBER 2020 American Journal of Obstetrics & Gynecology 517


Expert Reviews ajog.org

biomarker of inflammation, such as C-


FIGURE 1
reactive protein, if interpretation of
Iron prioritization during fetal and neonatal iron deficiency34e37 serum ferritin in the setting of inflam-
mation or infection.42 Measurement of
serum ferritin requires specialized
equipment and more blood than mea-
surement of hemoglobin, although the
global availability of the test is expanding
rapidly. Overall, serum ferritin, if avail-
able, should be used in conjunction with
hemoglobin concentration to more
precisely define iron status in
pregnancy.42
Other iron biomarkers show promise
in refining the diagnosis of iron defi-
ciency in pregnant women. %TSAT is
computed by dividing the serum iron
level by the total iron-binding capacity
The relative distributional flow of iron is indicated by the thickness of the blue arrows. The red blood and multiplying times 100. It reflects the
cells receive the primary allocation followed sequentially by the brain, the heart, and skeletal percentage of iron binding sites on
muscle.34-37 As negative iron balance progresses over time (red arrow), iron-dependent metabolic serum iron transport proteins that are
dysregulation of the skeletal muscle and heart is first noted by alterations in the serum metab- occupied by iron molecules. Serum iron
olome.36 Progressive worsening of iron deficiency subsequently negatively affects brain metabolism concentrations decrease early during
at approximately the same time that serum iron panels (eg, ferritin, %TSAT) become abnormal.36 Iron negative iron balance, thereby decreasing
deficiency results in anemia only in the final stage of the process.34-37 the %TSAT. In stage 2 iron deficiency,
CSF, cerebrospinal fluid; RBC, red blood cell; %TSAT, % total iron binding capacity saturation.
transferrin concentrations increase to
Georgieff. Iron deficiency in pregnancy. Am J Obstet Gynecol 2020. optimize iron binding capacity for
transport, which drives the %TSAT even
lower. All of these changes occur before
Nevertheless, there are significant for iron-dependent enzymes and he- microcytosis or anemia. Measurement of
concerns with relying solely on hemo- moproteins. Both the liver and the %TSAT has confirmed the tenuous iron
globin screening. Because iron incorpo- placenta have remarkable ability to store status of a large number of pre-anemic
ration into hemoglobin is a highly iron as ferritin to buffer the mother and women with iron deficiency.6 Reticulo-
prioritized process, anemia is the end- the fetus from wide swings in iron cyte hemoglobin concentration also
stage result of negative iron balance. availability. Ferritin also acts as an identifies pre-anemic, pre-microcytic
Thus, hemoglobin concentration lacks intracellular regulator of the labile iron iron deficiency and thus can serve as an
the sensitivity to detect early stages of pool by releasing iron when needed via early biomarker of iron deficiency. Its
iron deficiency, when there are already regulated chaperone molecules such as advantage is that most equipment that
physiologic effects at the tissue level. In NCOA4.40 Clinically, the finding of low measures hemoglobin concentration
addition, conditions other than iron serum ferritin concentration is highly also has the capacity to measure reticu-
deficiency cause anemia, including he- diagnostic for iron deficiency.41 Stan- locyte hemoglobin.
moglobinopathies, hemolysis, and dards for normal ferritin concentrations In the future, measurement of serum
inflammation/chronic disease. Reliance in pregnant women are available and hepcidin concentrations may provide
on hemoglobin concentration alone easily accessed in practice. the most information about who should
lacks sufficient sensitivity and specificity However, elevated or even normal and should not receive iron supple-
to diagnose iron deficiency in pregnancy. serum ferritin concentrations may be mentation. Hepcidin is the master
Iron-specific biomarkers, such as serum difficult to interpret because ferritin is an regulator of intestinal iron absorption
ferritin, % total iron binding capacity acute phase reactant, rising when sys- and iron distribution from the reticulo-
saturation (%TSAT), and hepcidin can temic inflammation is present.42 A high endothelial cells.43 Hepcidin is synthe-
be used to distinguish iron deficiency ferritin may represent true total body sized by the liver in response to iron
anemia from other causes of anemia. iron overload or simply a shift of iron status and inflammation. Hepcidin is a
The serum ferritin concentration in- into ferritin as a response to inflamma- negative regulator, which means that
dexes the body’s storage iron capacity. tion.42 A normal ferritin concentration high concentrations reduce intestinal
Ferritin is stored in reticuloendothelial may mask an iron-deficient state if iron absorption and promote iron
cells and released to the red cells for inflammation is present. Current rec- sequestration, whereas low levels in-
hemoglobin synthesis and to the tissues ommendations are to measure a crease intestinal iron absorption and

518 American Journal of Obstetrics & Gynecology OCTOBER 2020


ajog.org Expert Reviews

iron release from reticuloendothelial anemia. Genetic models of neuronal- The Short- and Long-Term Effects of
cells. High %TSAT indicating iron suf- specific iron deficiency without anemia Gestational Iron Deficiency on the
ficiency or high levels of intraleukin-6, a confirm that most of the neurocognitive Fetus
pro-inflammatory cytokine, increase phenotype of iron deficiency anemia is Fortunately, during mild maternal iron
hepcidin synthesis and release. Low % not due to the anemia.33,45 deficiency, iron is prioritized to the
TSAT is found in iron deficiency and fetus.8,24 However, during moderate
suppresses hepcidin. Patients with low Iron Supplementation During and severe iron deficiency, the entire
hepcidin levels likely require iron and Pregnancy maternaleplacentalefetal unit becomes
will respond to iron therapy, whereas The major reasons to maintain an iron- iron deficient, with significant short-
those with high hepcidin levels due to sufficient state during pregnancy are to and long-term consequences to the
iron sufficiency or iron overload do not protect the health of the mother, fetus. These consequences also occur in
require additional iron and will not improve pregnancy outcomes, and foster fetuses of iron-sufficient mothers who
absorb dietary iron.43 fetal development. A recent systematic nevertheless have gestational conditions
The beginning of pregnancy is review demonstrates that iron deficiency that compromise fetal iron delivery.
characterized by very low hepcidin in the first and second trimesters is These maternal conditions include hy-
concentrations, indicating a state of associated with increased maternal pertension, smoking, diabetes mellitus,
negative iron balance, and providing morbidity and an increased risk of and twinning.26e31 Insufficient fetal
strong evidence that iron needs during adverse pregnancy outcomes defined as iron, whether due to maternal iron
pregnancy are high.1 Women with low birthweight, prematurity, or intra- deficiency or to gestational conditions
marginal or even adequate iron status uterine growth restriction.46 Random- that compromise maternal fetal
in the prepregnancy state are thus ized controlled trials of iron iron delivery, results in 3 risks to the
prone to iron deficiency when they supplementation of iron-deficient preg- fetus.
become pregnant. nant women show that iron therapy re- The immediate risk is to fetal brain
duces the rates of these morbidities.46 development. Low maternal iron intake
Iron Deficiency and Iron Deficiency Little debate exists as to whether iron- in the third trimester results in altered
Anemia deficient pregnant women should have neonatal brain structure, including
Iron deficiency and iron deficiency ane- their iron deficiency treated.47,48 alterations in gray matter that index less-
mia are not synonymous. Iron deficiency Nearly 2 billion of the 7.5 billion complex dendritic architecture.52 Non-
is a state of negative iron balance where people on the planet are iron deficient, anemic iron-deficient newborns with
iron supply does not meet iron demand, making it the most common micro- cord serum ferritin concentrations <40
whereas iron deficiency anemia defines nutrient deficiency in the world.5 Iron mg/L have compromised recognition
the condition when hemoglobin syn- deficiency predominantly affects memory processing15 as do 2-month-old
thesis has been limited by the lack of iron women, their fetuses, and their infants with cord serum ferritin concen-
and the person is anemic as a conse- offspring.5,49 Such stark statistics com- trations <76 mg/L.53 The early age at
quence. Because iron is prioritized to red bined with the evidence that pregnancy which these abnormalities are detectable
blood cells for hemoglobin synthesis, induces negative iron balance1 has led strongly implicate abnormal fetal as
iron deficiency proceeds through mul- governing bodies to consider whether opposed to postnatal brain development
tiple non-anemic stages before anemia universal supplementation with iron due to iron deficiency. Recognition
ensues. Iron deficiency anemia can thus should be policy.5,50 The Office of Di- memory is mediated in large part by the
be thought of as the end stage of the iron- etary Supplements at the National In- hippocampus, which is a highly meta-
deficiency process. Iron stores are stitutes of Health recently held a bolic and rapidly developing structure
mobilized during the first stage of iron workshop to consider the question during the late fetal and early neonatal
deficiency, resulting in reduced %TSAT whether iron-sufficient pregnant women period, making it more vulnerable to the
and ferritin concentrations, respectively. should receive iron supplementation.48 lack of a critical substrate that supports
This process reflects the body’s attempt The question was considered because energy metabolism.54
to maintain iron delivery to the red cells many women are not iron deficient and The second risk is to long-term brain
and non-heme tissues including the because a small body of emerging development. Fetal/neonatal iron defi-
brain. However, when placed in direct research suggests an increased risk of ciency is associated with long-term
competition for iron, the red cells will be glucose intolerance in iron-sufficient neurocognitive dysfunction despite
prioritized over the brain.35e37 Children pregnant women treated with iron.51 spontaneous repletion of iron stores by 9
with pre-anemic iron deficiency This area deserves further, adequately months of age.18,20 Low neonatal iron
demonstrate abnormal brain and powered studies and exploration of po- status reduces recognition memory
behavioral function,44 indicating the tential underlying mechanisms (eg, performance at 3.5e4 years of age, even
likely presence of brain tissueelevel iron oxidative stress, microbiome alterations) though the infants are no longer iron
deficiency before the appearance of before policy decisions can be made. deficient.18

OCTOBER 2020 American Journal of Obstetrics & Gynecology 519


Expert Reviews ajog.org

The third risk is postnatal iron defi- second decade of life and into young newborns reduces oxygen consumption
ciency in infancy and toddlerhood and adulthood.58 The resultant loss of edu- rate by 60%, which in turn results in a
its attendant neurodevelopmental cation and job potential can be consid- more primitive and simple neuronal
sequelae. Infants who were iron under- ereable.49 Thus, ensuring fetal structure and poorer brain mitochon-
loaded as fetuses have poorer iron status nutritional and in particular iron health drial health.65,72 Among the brain re-
at 9 months of age and thus a greater risk can be seen as an investment in the next gions that are most rapidly developing in
of becoming iron deficient.20 Iron defi- generation and society going forward. the last trimester, the hippocampus is
ciency during infancy and toddlerhood foremost. The hippocampus resides at
is associated with slower speed of pro- The Mechanisms Underlying Iron the center of the recognition memory
cessing, poorer motor function and Deficiency Effects on the Young Brain system that mediates spatial and fact
increased social dysfunction during the Iron is essential for the proper func- learning. The hippocampus in humans
acute phase of iron deficiency. It confers tioning of all cells, including those that begins to differentiate rapidly at 28
a risk of significant long-term neuro- comprise the brain. The negative effects weeks gestation and continues to be
morbidities including depression and of developmental iron deficiency on the highly active in its development until
anxiety in adulthood.55e58 One reason brain have been well described in pre- approximately 2 years of age.4 Fetal iron
proper fetal iron loading is important is clinical models over the past 45 years,12 deficiency in preclinical models com-
that human milk is very low in iron. particularly in the domains of mono- promises hippocampal synaptic plas-
Breast milk may be the only dietary amine neurotransmitter meta- ticity and structural gene expression,73,74
source for the infant in the first 6 post- bolism,62,63 neuronal and glial energy metabolism,64 electrophysiologic capac-
natal months and thus the infant relies metabolism,64,65 myelination,66 and ity,75,76 and recognition memory
on the iron stored during fetal life to gene regulation.67e69 The alterations in behavior acutely and into adulthood
support hemoglobin synthesis and organ the brain induced by iron deficiency are despite iron repletion in the neonatal
development for that period.37,38 highly consistent with the acute and period.33,45 These findings provide
long-term neurobehavioral deficits that biological plausibility for the neuro-
The Link Between Fetal or Early have been described in humans.12 cognitive changes documented in iron-
Postnatal Iron Status and Long-Term Monoamine neurotransmitters, eg, deficient newborns.15,17,18
Neurocognitive and Mental Health dopamine, serotonin, and norepineph- In the human, rapid myelination of
Maternalefetal iron status has been rine, are synthesized by the iron con- the brain begins at 32 weeks of gestation
linked to a number of neurocognitive taining hydroxylases-tyrosine and extends into the first 2 years of
and mental health disorders in the hydroxylase and tryptophan hydroxylase. postnatal life. Iron-containing enzymes
offspring, driven in part by the timing of In the human, the regulation of these are important in the synthesis of fatty
iron deficiency during pregnancy. Low neurotransmitters is highly shaped be- acids integral to myelin and early life
maternal iron intake at the time of tween mid-gestation and 3 years’ post- iron deficiency alters the fatty composi-
conception is associated with a greater natal age. Research dating from the mid- tion of myelin and results in a hypo-
risk of autism in the offspring.59 Low 1970s to the present in preclinical models myelinated nervous system.66 Hypo- and
iron intake during the second trimester indicates that early life iron deficiency dysmyelination would result in slower
increases the risk of schizophrenia in the alters the concentrations of the neuro- speed of neuronal processing, a finding
offspring by 30%.60 Infants born with transmitters, their receptors, and their that is present in iron deficient neonates
low serum ferritin concentrations, reuptake mechanisms acutely while iron and infants.16,55,61 Myelin effects persist
reflecting reduced fetal iron loading, deficient,62,63 and more importantly, into adulthood in the preclinical
have poorer recognition memory at long-term into adulthood despite iron models.68
birth,15 2 months of age,53 and 3.5 years repletion.70 These neurochemical More recently, iron’s role in fetal and
of age.18 They have poorer school per- changes underlie behavioral changes in neonatal gene expression has been
formance at 5e6 year of age14 and dif- the preclinical models and the behaviors demonstrated in preclinical models and
ficulty with planning and attention at 10 are similar to those seen in humans, has been used as a way of demonstrating
years of age.19 Infants with onset of iron where anxiety, hesitancy, and mood dis- a mechanism underlying the long-term
deficiency in early postnatal life, likely orders have been documented.12,71 effects of early-life iron deficiency. Ro-
driven by inadequate fetal iron As noted previously, the fetal brain is a dents with fetal iron deficiency modeling
loading,13 demonstrate slower speed of highly metabolic organ, consuming 60% the degree of brain iron deficiency seen
neural processing at the time of their of the total fetal oxygen consumption in humans demonstrate a reduction in
deficiency.55 The slower speed of neural rate.2 This high metabolic rate serves the the expression of individual synaptic
processing persists beyond the period of purpose of cellular differentiation and plasticity genes essential for develop-
iron deficiency and despite treatment.61 function. Iron is essential in this process ment and function,73,74,77 as well as
The infants have a greater risk of cogni- through its role in cytochromes in the networks of genes underlying critical
tive and socioemotional problems generation of ATP. Neuronal iron defi- neural processes and mental health
including anxiety and depression in their ciency to the degree found in human conditions, including autism,

520 American Journal of Obstetrics & Gynecology OCTOBER 2020


ajog.org Expert Reviews

FIGURE 2
Gene networks affected by gestational iron deficiency and choline supplementation69

Georgieff. Iron deficiency in pregnancy. Am J Obstet Gynecol 2020.

schizophrenia and mood disorders.69 constitutes a critical period,79 wherein driven in part by fetal conditions.84 Fetal
These long-term gene changes are likely lack of substrates necessary for proper iron status appears to follow this pattern.
epigenetically driven, particularly by al- development result in permanent struc- Low maternal iron intake in the second
terations to iron containing histone tural abnormalities.45,79,80 Such a period trimester is associated with a 30%
demethylases of the JARID family.78 has been identified in the fetal/neonatal increased risk of schizophrenia.60 Pre-
Based on the understanding of the period for the mouse hippocampus.45 clinical models demonstrate that a
biology, it should not be surprising that Iron deficiency that is not corrected network of genes underlying schizo-
iron deficiency compromises brain during that period results in long-term phrenia are activated by fetal iron defi-
acutely in the fetus and newborn. How- structural deficits accompanied by neu- ciency.69 These gene networks continue
ever, it is the long-term effects that are rocognitive abnormalities.45 to show abnormal expression in the
concerning from a societal perspective The epigenetic mechanism is rooted formerly iron deficiency adult brain,
and thus the etiologies of and potential in the concept of the Developmental providing biological plausibility to the
therapies for the long-term effects are a Origins of Adult Health and Disease.81,82 epidemiologic finding in humans.60,69
topic of intense investigation. Many regulatory setpoints for genes that The finding that these changes to gene
The long-term effects can be concep- control multiple metabolic processes are networks are prevented by feeding a diet
tualized by 2 basic mechanisms that are set in late fetal life, a concept popularized rich in a methyl donor (choline) support
not mutually exclusive: the critical by David Barker and known initially as the hypothesis that the long-term effects
period mechanism and the epigenetic the “Barker Hypothesis” in the 1990s.83 may be epigenetic in nature (Figure 2).69
modification of chromatin mechanism. The observation that similar principles A similar line of research demonstrates a
The brain is not a homogenous organ; of “fetal setpoint anticipation” apply to link between low first-trimester iron
instead, its various areas and processes the brain became evident in the early intake and the risk of autism in
are distinct entities that develop on 2000s with the demonstration that the humans,59 with elucidation of the
different trajectories.4 A period of rapid risk for several adult mental health involved gene networks in the preclinical
growth and differentiation often conditions including schizophrenia are fetal iron deficiency model.69

OCTOBER 2020 American Journal of Obstetrics & Gynecology 521


Expert Reviews ajog.org

In summary, the preclinical science development of the offspring. Peri- health from solely focusing on postnatal
across multiple investigative platforms, conceptional iron and folate iron intake nutrition to focusing on prenatal and
including molecular biology, cellular are both related to the subsequent risk of even preconceptional nutrition. In this
biology, electrophysiology, structure, autism in the offspring with lower in- way, it argues for policies that promote
and behavior, have provided a takes associated with higher risks.59,85 maternal nutritional health heading into
convincing plausibility to the role of Undernutrition is not the only prob- pregnancy, followed by close attention to
fetal iron in brain development and lem. Overnutrition in the form of provision of critical nutrients such as
the role of fetal iron deficiency in maternal obesity is a risk factor. Elevated iron during pregnancy. This life-cycle
the short- and long-term neuro- prepregnancy body mass index inde- approach to nutrition can be seen as an
behavioral abnormalities observed in pendent of gestational weight gain in- investment in the mental health of the
humans.82 creases the risk of mental health and next generation and an investment in
neurodevelopmental disorders in the society. -
Pre- and Periconceptional Iron and offspring.86
the Argument for a Life-Cycle Effect
of Nutrients Conclusion REFERENCES
Ongoing research indicates that a wom- The information presented in this review 1. Fisher A, Nemeth E. Iron homeostasis during
an’s nutritional status heading into shifts the conversation about the role of pregnancy. Am J Clin Nutr 2017;106(suppl):
pregnancy influences the brain nutrition in ensuring childhood brain 1567S–74S.
2. Kuzawa CW. Adipose tissue in human infancy
and childhood: an evolutionary perspective. Am
J Phys Anthropol 1998;27(suppl):177–209.
GLOSSARY OF TERMS: 3. Cowan MW. The development of the brain.
C-reactive protein: A protein generated by the liver during conditions of inflammation, such Sci Am 1979;241:113–33.
4. Thompson RA, Nelson CA. Developmental
as infection. science and the media. Early brain development.
Dysmyelination: Abnormal biochemical composition of myelin. Am Psychol 2001;56:5–15.
Ferritin: The intracellular storage protein for iron. Measurement of ferritin in the serum or 5. World Health Organization. Iron deficiency
plasma likely comes from ferritin shed from senescent cells and reflects the intracellular anaemia: assessment, prevention and control: a
storage pool. guide for programme managers. 2015. Available
at: www.who.int/nutrition/publications/en/ida_
Glia: Non-neuronal cells in the brain including oligodendrocytes that make myelin, as- assessment_prevention_control.pdf 2015.
trocytes that serve multiple functions including nutrient delivery, and microglia that traffic Accessed April 10, 2020.
neurons during development and can generate pro-inflammatory responses. 6. Auerbach M, Abernathy J, Juul S, Short V,
Hepcidin: A protein generated by the liver in response to iron sufficiency or inflammation Derman R. Prevalence of iron deficiency in first
that inhibits iron absorption from the intestine and sequesters iron in macrophages. trimester, nonanemic pregnant women.
J Matern Fetal Neonatal Med 2019;3:1–4.
Labile iron pool: The intracellular iron that is not bound in active enzymes or hemoproteins 7. Oski FA. The hematologic aspects of the
or stored in ferritin and thus is available for use. maternal-fetal relationship. In: Oski FA,
Methyl donor: Compounds that through their metabolism can donate a methyl group that Naiman JL, eds. Hematologic problems in the
can bind to DNA components such as cytosineeguanine islands or histones and thereby newborn, ed 3. Philadelphia: WB Saunders;
alter gene expression. Multiple nutrients such as choline, betaine, folate can act as methyl 1982. p. 32–3.
8. Cao C, O’Brien KO. Pregnancy and iron ho-
donors. meostasis: an update. Nutr Rev 2013;71:35–51.
Monoamine neurotransmitters: Dopamine, serotonin, and norepinephrine are the neu- 9. McArdle HJ, Gambling L, Kennedy C. Iron
rotransmitters that are synthesized by iron containing enzymes tyrosine hydroxylase and deficiency during pregnancy: the consequences
tryptophan hydroxylase where iron is critical to the enzymatic function and thus the for placental function and fetal outcome. Proc
concentration of neurotransmitter produced. Nutr Soc 2014;73:9–15.
10. van der Merwe LF, Eussun SR. Iron status of
Neurocognitive: Brain functions related to reasoning, learning and memory, and higher young children in Europe. Am J Clin Nutr
functions (eg, multi-tasking, attention, inhibition, working memory). 2017;106(suppl):1663S–71S.
Neurodevelopment: The process of rapid brain growth and differentiation that takes place 11. Gupta PM, Perrine CG, Mei Z, Scanlon KS.
in the period of time from conception through early adulthood. Iron, anemia and iron deficiency anemia among
Transferrin saturation: Transferrin is the major binding protein for iron circulating in the young children in the United States. Nutrients
2016;8:E330.
serum. Under normal physiologic conditions, up to two ferric iron molecules are bound to 12. Lozoff B, Beard J, Connor J, Felt B,
transferrin molecule. Georgieff M, Schallert T. Long-lasting neural and
Oxygen consumption rate: The cellular metabolic rate of oxygen in the process of ATP behavioral effects of early iron deficiency in in-
generation as set by the mitochondria. fancy. Nutr Rev 2006;64:S34–43.
Synaptic plasticity: The ability of neurons to respond to external influences and thereby 13. Shao J, Lou J, Rao R, et al. Maternal serum
ferritin concentration is positively associated
modify their anatomic structure and functional output. The term is often used to reflect the with newborn iron stores in women with low
flexibility of the functioning brain. ferritin status in late pregnancy. J Nutr 2012;142:
2004–9.

522 American Journal of Obstetrics & Gynecology OCTOBER 2020


ajog.org Expert Reviews

14. Tamura T, Goldenberg RL, Hou J, et al. influences on fetal iron status at term using cord relationships between iron deficiency with or
Cord serum ferritin concentrations and mental blood transferrin receptors. Arch Dis Child Fetal without anemia and infant social-emotional
and psychomotor development of children at Neonatal Ed 2001;84:F40–3. behavior. J Pediatr 2008;152:696–702.
five years of age. J Pediatr 2002;140:165–70. 29. Georgieff MK, Landon MB, Mills MM, et al. 45. Fretham SJB, Carlson ES, Wobken J,
15. Siddappa AM, Georgieff MK, Wewerka S, Abnormal iron distribution in infants of diabetic Tran PV, Petryk A, Georgieff MK. Temporal
Worwa C, Nelson CA, deRegnier R-A. Auditory mothers: spectrum and maternal antecedents. manipulation of transferrin receptor-1 depen-
recognition memory in iron-deficient infants of J Pediatr 1990;117:455–61. dent iron uptake identifies a sensitive period in
diabetic mothers. Pediatr Res 2004;55: 30. Petry CD, Eaton MA, Wobken JD, Mills MM, mouse hippocampal neurodevelopment. Hip-
1034–41. Johnson DE, Georgieff MK. Iron deficiency of pocampus 2012;22:1691–702.
16. Amin SB, Orlando M, Eddins A, liver, heart, and brain in newborn infants of dia- 46. Dewey KG, Oaks BM. U-shaped curve for
MacDonald M, Monczynski C, Wang H. In utero betic mothers. J Pediatr 1992;121:109–14. risk associated with maternal hemoglobin, iron
iron status and auditory neural maturation in 31. Ru Y, Pressman EK, Guillet R, Katzman PJ, status or iron supplementation. Am J Clin Nutr
premature infants as evaluated by auditory Vermeylen F, O’Brien KO. Umbilical cord hep- 2017;106:1694S–702S.
brainstem response. J Pediatr 2010;156: cidin concentrations are positively associated 47. Georgieff MK, Krebs NF, Cusick S. The
377–81. with the variance in iron status among multiple benefits and risks of iron supplementation in
17. Geng F, Mai X, Zhan J, Xu L, Georgieff M, birth neonates. J Nutr 2018;148:1716–22. pregnancy and childhood. Ann Rev Nutr
Shao J, Lozoff B. Timing of iron deficiency and 32. Blayney L, Bailey-Wood R, Jacobs A, et al. 2019;39:121–46.
recognition memory in infancy. Nutr Neurosci The effects of iron deficiency on the respiratory 48. Brannon PM, Stover PJ, Taylor CL. Inte-
2020 [Epub ahead of print]. function and cytochrome content of rat heart grating themes, evidence gaps, and research
18. Riggins T, Miller NC, Bauer PB, mitochondria. Circ Res 1976;39:744–8. needs identified by workshop on iron screening
Georgieff MK, Nelson CA. Consequences of low 33. Carlson ES, Tkac I, Magid R, et al. Iron is and supplementation in iron-replete pregnant
neonatal iron status due to maternal diabetes essential for neuron development and memory women and young children. Am J Clin Nutr
mellitus on explicit memory performance in function in mouse hippocampus. J Nutr 2017;106:1703S–12S.
childhood. Dev Neuropsychol 2009;34:762–79. 2009;139:672–9. 49. Walker SP, Wachs TD, Gardner JM,
19. Jabès A, Thomas KM, Langworthy S, 34. Georgieff MK. Iron assessment to protect Lozoff B, Wasserman GA, Pollitt E, Carter JA;
Georgieff MK, Nelson CA. Functional and the developing brain. Am J Clin Nutr 2017;106: International Child Development Steering
anatomic consequences of diabetic pregnancy 1588S–93S. Group. Child development: risk factors for
on memory in 10-year-old children. J Dev Behav 35. Zamora TG, Guiang SF III, Georgieff MK, adverse outcomes in developing countries.
Pediatr 2015;36:529–35. Widness JA. Iron is prioritized to red blood cells Lancet 2007;369:145–57.
20. Georgieff MK, Wewerka SW, Nelson CA, over the brain in phlebotomized anemic 50. Siu AL; US preventative Services Task
deRegnier R-A. Iron status at 9 months of infants newborn lambs. Pediatr Res 2016;79:922–8. Force. Screening for iron deficiency anemia and
with low iron stores at birth. J Pediatr 2002;141: 36. Rao R, Ennis K, Lubach G, Lock E, iron supplementation in pregnant women to
405–9. Georgieff MK, Coe C. Metabolomic analysis of improve maternal health and birth outcomes: US
21. Zhao G, Guobin X, Zhou M, et al. Prenatal CSF indicates brain metabolic impairment pre- Preventative Services Task Force Recommen-
iron supplementation reduces maternal anemia, cedes hematological indices of anemia in the dation Statement. Ann Intern Med 2015;163:
iron deficiency, iron deficiency anemia in a ran- iron-deficient infant monkey. Nutr Neurosci 529–36.
domized clinical trial in rural china, but iron defi- 2016;6:1–9. 51. Zhang C, Rawal S. Dietary iron intake, iron
ciency remains widespread in mothers and 37. Georgieff MK, Schmidt RL, Mills MM, status and gestational diabetes. Am J Clin Nutr
neonates. J Nutr 2015;145:1916–23. Radmer WJ, Widness JA. Fetal iron and cyto- 2017;106:1672S–80S.
22. Ziegler EE, O’Donnell AM, Nelson SE, chrome c status after intrauterine hypoxemia 52. Monk C, Georgieff MK, Xu D, et al. Maternal
Fomon SJ. Body composition of the reference and erythropoietin administration. Am J Physiol prenatal iron status and tissue organization in the
fetus. Growth 1976;40:329–41. 1992;262:R485–91. neonatal brain. Pediatr Res 2015;79:482–8.
23. Lorenz L, Peter A, Poets CF, Franz AR. 38. Lonnerdal B. Excess iron intake as a factor in 53. Geng F, Mai X, Zhan J, et al. Impact of fetal-
A review of cord blood concentrations of iron growth, infections, and development of infants neonatal iron deficiency on recognition memory
status parameters to define reference ranges for and young children. Am J Clin Nutr 2017;106: at 2 months of age. J Pediatr 2015;167:
preterm infants. Neonatology 2013;104: 1681S–7S. 1226–32.
194–202. 39. Grajeda R, Perez-Escamilla R, Dewey KG. 54. Georgieff MK, Innis S. Controversial nutri-
24. O’Brien KO, Zavaleta N, Abrams SA, Delayed clamping of the umbilical cord improves ents in the perinatal period that potentially affect
Caulfield LE. Maternal iron status influences hematologic status of Guatemalan infants at 2 neurodevelopment: essential fatty acids and
iron transfer to the fetus during the third mo of age. Am J Clin Nutr 1997;65:425–31. iron. Pediatr Res 2005;57:99R–103R.
trimester of pregnancy. Am J Clin Nutr 40. Philpott CC, Jadhav S. The ins and outs of 55. Roncagliolo M, Garrido M, Walter t,
2003;77:924–30. iron: escorting iron through the mammalian Peirano P, Lozoff B. Evidence of altered central
25. Saarinen UM, Siimes MA. Serum ferritin in cytosol. Free Rad Biol Med 2019;133:112–7. nervous system development in infants with iron
assessment of iron nutrition in healthy infants. 41. Mast AE, Blinder MA, Gronowski AM, deficiency anemia at 6 mo: delayed maturation
Acta Paediatr Scand 1978;67:745–51. Chumley C, Scott MG. Clinical utility of the sol- of auditory brainstem responses. Am J Clin Nutr
26. Siddappa AJ, Rao R, Long JD, Widness JA, uble transferrin receptor and comparison with 1998;68:683–90.
Georgieff MK. The assessment of newborn iron serum ferritin in several populations. Clin Chem 56. Angulo-Kinzler RM, Peirano P, Lin E,
stores at birth: a review of the literature and 1998;44:45–51. Algarin C, Garrido M, Lozoff B. Twenty-four-
standards for ferritin concentrations. Neona- 42. Suchdev PS, Williams AM, Mei Z, et al. hour motor activity in human infants with and
tology 2007;92:73–82. Assessment of iron status in settings of inflam- without iron deficiency anemia. Early Hum Dev
27. Chockalingam UM, Murphy E, Ophoven JC, mation: challenges and potential approaches. 2002;70:85–101.
Weisdorf SA, Georgieff MK. Cord transferrin and Am J Clin Nutr 2017;106:1626S–33S. 57. Lozoff B, Jimenez E, Smith JB. Double
ferritin levels in newborn infants at risk for pre- 43. Coffey R, Ganz T. Iron homeostasis: an burden of iron deficiency in infancy and low
natal uteroplacental insufficiency and chronic anthropocentric perspective. J Biol Chem socioeconomic status: a longitudinal anal-
hypoxia. J Pediatr 1987;111:283–6. 2017;292:12727–34. ysis of cognitive test scores to age 19
28. Sweet DG, Savage G, Tubman TR, 44. Lozoff B, Clark KM, Jing Y, Armony-Sivan R, years. Arch Pediatr Adolesc Med 2006;160:
Lappin TR, Halliday HL. Study of maternal Angelilli ML, Jacobson SW. Dose-response 1108–13.

OCTOBER 2020 American Journal of Obstetrics & Gynecology 523


Expert Reviews ajog.org

58. Lukowski AF, Koss M, Burden MJ, et al. Iron altered developmental expression of genes efficacy in area CA1 of the developing rat
deficiency in infancy and neurocognitive func- mediating energy metabolism and neuronal hippocampus. Hippocampus 2005;15:
tioning at 19 years: evidence of long-term defi- morphogenesis in hippocampus. Hippocampus 1094–102.
cits in executive function and recognition 2007;17:679–91. 76. Pisansky MT, Wickham RJ, Su J, et al. Iron
memory. Nutr Neurosci 2010;13:54–70. 68. Clardy SL, Wanx X, Zhao W, et al. Acute deficiency with or without anemia impairs pre-
59. Schmidt RJ, Tancredi DJ, Krakowiak P, and chronic effects of developmental iron pulse inhibition of the startle reflex. Hippocam-
Hansen RL, Ozonoff S. Maternal intake of sup- deficiency on mRNA expression patterns in pus 2013;23:952–62.
plemental iron and risk of autism spectrum dis- the brain. J Neural Transm Suppl 2006;71: 77. Tran PV, Fretham SJB, Carlson ES,
order. Am J Epidemiol 2014;180:890–900. 173–96. Georgieff MK. Long-term reduction of hippo-
60. Insel BJ, Schaefer CA, McKeague IW, 69. Tran PV, Kennedy BC, Pisansky MT, campal BDNF activity following fetal-neonatal
Susser ES, Brown AS. Maternal iron and the risk Won K-J, Gewirtz JC, Simmons RA, iron deficiency in adult rats. Pediatr Res
of schizophrenia in offspring. Arch Gen Psychi- Georgieff MK. Prenatal choline supplementation 2009;65:493–8.
atry 2008;65:1136–44. diminishes early elife iron deficiency induced 78. Tran PV, Kennedy BC, Lien YC,
61. Algarin C, Peirano P, Garrido M, Pizzaro F, preprogramming of networks associated with Simmons RA, Georgieff MK. Fetal iron deficiency
Lozoff B. Iron deficiency anemia in infancy: long- behavioral abnormalities in the adult rat hippo- induces chromatin remodeling at the Bdnf locus
lasting effects on auditory and visual system campus. J Nutr 2016;146:484–93. in adult rat hippocampus. Am J Physiol Integr
functioning. Pediatr Res 2003;53:217–23. 70. Unger EL, Hurst AR, Georgieff MK, et al. Comp Phys 2015;308:R276–82.
62. Youdim MB, Yehuda S. The neurochemical Behavior and monoamine deficits in pre- and 79. Hensch TK. Critical period regulation. Annu
basis of cognitive deficits induced by brain iron peri-natal iron deficiency are not corrected by Rev Neurosci 2004;27:549–79.
deficiency: involvement of dopamine-opiate early postnatal moderate or high iron diet in rats. 80. Jorgenson LA, Wobken JD, Georgieff MK.
system. Cell Mol Biol 2000;46:491–500. J Nutr 2012;142:2040–9. Perinatal iron deficiency alters apical dendritic
63. Felt BT, Beard JL, Schallert T, et al. Persis- 71. Wachs TD, Pollitt E, Cueto S, Jacoby E, growth in hippocampal CA-1 pyramidal neu-
tent neurochemical and behavioral abnormalities Creed-Kanashiro H. Relation of neonatal iron rons. Dev Neurosci 2003;25:412–20.
in adulthood despite early iron supplementation status to individual variability in neonatal 81. Hanson MA, Poston L, Gluckman PD.
for perinatal iron deficiency anemia in rats. Behav temperament. Dev Psychobiol 2005;46: DOHaD—the challenge of translating science
Brain Res 2006;171:261–70. 141–53. to policy. J Dev Orig Health Dis 2019;10:
64. Rao R, Tkac I, Townsend EL, Gruetter R, 72. Bastian T, Von Hohenberg W, Georgieff MK, 263–7.
Georgieff MK. Perinatal iron deficiency alters the Lanier L. Chronic energy depletion due to iron 82. Barks AK, Hall AM, Tran PV, Georgieff MK.
neurochemical profile of the developing rat hip- deficiency impairs dendritic mitochondrial Iron as a model nutrient for understanding the
pocampus. J Nutr 2003;133:3215–21. motility during hippocampal neuron develop- nutritional origins of neuropsychiatric disease.
65. Bastian TW, von Hohenberg WC, ment. J. Neurosci 2019;39:802–13. Pediatr Res 2019;85:176–82.
Mickelson DJ, Lanier LM, Georgieff MK. Iron 73. Tran PV, Carlson ES, Fretham SJ, 83. Curran GC, Chertow GM, Willett WC, et al.
deficiency impairs developing hippocampal Georgieff MK. Early-life iron deficiency anemia Birth weight and adult hypertension and obesity
neuron gene expression, energy metabolism alters neurotrophic factor expression and hip- in women. Circulation 1996;94:1310–5.
and dendrite complexity. Dev Neurosci 2016;38: pocampal neuron differentiation in male rats. 84. Bale TL, Baram TZ, Brown AS, et al. Early life
264–76. J Nutr 2008;138:2495–501. programming and neurodevelopmental disor-
66. Ortiz E, Pasquini JM, Thompson K, Felt B, 74. Brunette KE, Tran PV, Wobken JD, ders. Biol Psychiatry 2010;68:314–9.
Butkus G, Beard J, Connor JR. Effect of Carlson ES, Georgieff MK. Gestational and 85. Schmidt RJ. Maternal folic acid supple-
manipulation of iron storage, transport, or avail- neonatal iron deficiency alters apical dendrite ments associated with reduced autism risk in
ability on myelin composition and brain iron structure of CA1 pyramidal neurons in adult rat the child. Evid Based Med 2013;18:e53.
content in three different animal models. hippocampus. Dev Neurosci 2010;32:238–48. 86. Rivera HM, Christiansen KJ, Sullivan EL.
J Neurosci Res 2004;77:681–9. 75. Jorgenson LA, Sun M, O’Connor M, The role of maternal obesity in the risk of
67. Carlson ES, Stead JDH, Neal CR, Petryk A, Georgieff MK. Fetal iron deficiency disrupts neuropsychiatric disorders. Front Neurosci
Georgieff MK. Perinatal iron deficiency results in the maturation of synaptic function and 2015;9:194.

524 American Journal of Obstetrics & Gynecology OCTOBER 2020

You might also like