You are on page 1of 10

Iron deficiency in women: assessment, causes and consequences

Jane Coada and Cathryn Conlonb


a
Institute of Food, Nutrition and Human Health, Purpose of review
Massey University, Palmerston North and bInstitute of
Food, Nutrition and Human Health, Massey University,
Iron deficiency is the most common nutritional disorder affecting about 20–25% of the
Albany, Auckland, New Zealand world’s population, predominantly children and women. There is emerging evidence that
Correspondence to Jane Coad, PhD, Institute of Food, depletion of iron stores may have adverse consequences for adults even in the absence
Nutrition and Human Health, Massey University, of anaemia. This raises issues about the most appropriate method of assessing iron
Palmerston North 4442, New Zealand
Tel: +64 6350 5962; e-mail: j.coad@massey.ac.nz status.
Recent findings
Current Opinion in Clinical Nutrition and
Metabolic Care 2011, 14:625–634
Although the effects of iron-deficiency anaemia are well characterized, emerging
evidence suggests that iron deficiency without anaemia can have negative
consequences in adults, particularly for neurocognitive outcomes. Iron deficiency is
more likely in women of reproductive age because of menstrual blood loss. However,
extremes of blood loss such as regular blood donation, diets of low bioavailability and
the challenges of pregnancy all markedly increase the risk of iron deficiency. In addition,
the physiological changes in pregnancy affect the normal reference ranges used in
laboratory assessment. The use of haemoglobin as a marker of iron deficiency is limited
by its low specificity and sensitivity and although the use of alternative biomarkers is
becoming more common, interpreting results in conditions of chronic inflammation,
including that associated with increased adiposity, needs more investigation.
Summary
By understanding the physiology of iron metabolism alongside the limitations and
interpretation of biomarkers of iron deficiency, clinicians and nutritionists are better
equipped to identify changes in iron balance and to further investigate the functional
outcomes of iron deficiency.

Keywords
biomarkers, iron deficiency, iron metabolism, risk factors

Curr Opin Clin Nutr Metab Care 14:625–634


ß 2011 Wolters Kluwer Health | Lippincott Williams & Wilkins
1363-1950

are reduced. Presentation of clinical symptoms is


Introduction variable depending on the rapidity of development;
Iron deficiency continues to be the most common nutri- slower progression allows more effective physiologi-
tional disorder and public health problem in the world cal adaptation.
[1,2], affecting about 20–25% of the world population,
disproportionately children and women. Iron deficiency Although iron deficiency is the major cause of anaemia,
is traditionally categorized into stages [3] (see Table 1): not all anaemia is IDA. Other forms of anaemia result
from other nutrient deficiencies (such as vitamin B12,
(1) mild deficiency or iron depletion characterized by folate and vitamin A), acute and chronic inflammation,
depleted stores with normal production of haemo- parasitic infections and disorders that affect Hb synthesis
globin (Hb) and iron-dependent proteins; or RBC production or survival (such as haemoglobino-
(2) marginal deficiency or iron-deficient erythropoiesis pathies).
characterized by depleted iron stores, decreased
iron-dependent protein production but normal Hb Women, particularly those of reproductive age, are at
concentrations; and much higher risk of iron deficiency. This has implications
(3) iron-deficiency anaemia (IDA), defined as a for not only their health but also that of their offspring.
decreased concentration of circulating red blood cells In addition to its role in the synthesis of Hb, iron has a
(RBCs) or decreased concentration of Hb within number of roles in tissue. Emerging evidence suggests
blood cells, resulting in compromised transport of that iron deficiency without anaemia (IDWA) may be
oxygen to tissues; iron stores are depleted and the associated with symptoms which result from impaired
concentrations of iron-dependent oxidative enzymes cellular metabolism. Although the measurement of Hb

1363-1950 ß 2011 Wolters Kluwer Health | Lippincott Williams & Wilkins DOI:10.1097/MCO.0b013e32834be6fd

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
626 Micronutrients

concentration has both low sensitivity and low specificity, Key points
Hb concentration is commonly used as the biomarker
to determine the prevalence of iron deficiency despite  Changes in Hb concentration are not specific or
sensitive in determining iron deficiency.
changes in Hb being a relatively late consequence of
 Iron deficiency without anaemia may compromise
iron deficiency. In order to both determine the effects
neurocognitive outcomes.
of preanaemic iron deficiency and identify women at
 Dietary forms of iron include ferritin as well as the
increased risk, it is important to consider the strengths
better characterized haem and nonhaem iron.
and limitations of alternative methods of assessing iron  Iron status and iron requirements in women of
status. reproductive age are significantly affected by men-
strual blood loss and can be further compromised by
other causes of blood loss including blood donation.
Physiology of iron metabolism related to  Laboratory assessment of iron status is complicated
assessment of iron deficiency by the normal physiological changes in pregnancy.
Identification of the most appropriate biomarker for  Chronic inflammation, which is common in hospi-
determining iron status is determined by the physiologi- talized patients, the elderly and in obese indi-
cal responses to adequate and inadequate iron supplied to viduals, may affect body sequestration of iron and
various tissues. iron absorption making biomarkers of iron status
difficult to interpret.
Dietary iron
Iron is present in food as haem iron and as nonhaem iron. proximal duodenum enterocytes via the divalent metal
Haem iron, from proteolytic digestion of Hb and transporter 1 (DMT1) is a process that requires proton co-
myoglobin, is absorbed efficiently (about 40%), whereas transport (see [6] for recent review of mechanisms
absorption of nonhaem iron is less efficient and strongly involved in iron metabolism). Absorption of nonhaem
influenced by other dietary components [4]. In iron is inhibited by phytic acid (in cereals and legumes)
addition, ferritin from plant (particularly legume) and and by polyphenols (in coffee, tea, wine and some
animal food sources and lactoferrin from milk contribute vegetables) (see [4] for a comprehensive review of
to dietary iron [4]. Absorption of dietary iron depends dietary factors affecting iron absorption and the mech-
primarily on the physiological status of the individual anisms involved). Enhancers of nonhaem iron absorption
(iron-deplete individuals absorb more iron) and on its include ascorbic acid, citric acid and some other organic
bioavailability in the diet (higher in diets rich in meat and acids, carotenes, alcohol and a yet-to-be-identified factor
ascorbic acid). Iron is unusual in that selective absorption in meat, poultry and fish.
of dietary iron is the primary homeostatic mechanism
involved in regulating iron balance. Dietary haem is transported into the enterocyte by less-
well characterized mechanisms, probably via a haem
Absorption carrier protein [7] followed by subsequent internalization
Dietary nonhaem iron is predominantly in the insoluble in cytoplasmic vesicles from which Fe2þ is liberated by
ferric form (Fe3þ) bound to components of food. Follow- haemoxygenase 1.
ing digestion, uptake of nonhaem iron requires reduction
to ferrous (Fe2þ) iron by ferrireductases such as duodenal Ferritin, the ubiquitous and highly conserved iron storage
cytochrome B (dcytB) [5] or by dietary ascorbic acid. molecule derived from both plant and animal foods which
Subsequent transport across the apical membrane of the is relatively resistant to proteolytic digestion, is probably

Table 1 Sequential stages of iron deficiency in women


Early negative Depletion of Iron-deficient
Normal iron balance storage iron erythropoiesis Iron-deficiency anaemia

Bone marrow iron 2–3 1 0–1 0 0


Score [1–3,4,5,6]
Erythrocytes Normal Normal Normal Normal Microcytic and hypochromic
Hb (g/l) 120–160 120–160 120–160 120–160 <120
Erythrocyte protoporphyrin (mg/dl) 30 30 30 <15 <15
Plasma iron (mg/dl) 115 þ 15 <120 115 <60 <40
TIBC(mg/dl) 330  30 330–360 360 390 410
Ferritin (mg/l) 100  60 <25 20 10 <10
Soluble transferrin receptors Normal Normal–high High Very high Very high
Transferrin saturation (%) 35  15 30 30 <15 <15
Iron absorption (%) 5–10 10–15 10–15 10–20 10–20
Adapted with permission from [3]. Hb, haemoglobin; TIBC, total iron-binding capacity.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Iron deficiency in women Coad and Conlon 627

taken up by the enterocyte by an independent mech- A proton pump results in acidification of the endocytic
anism probably mediated by a high-affinity receptor vesicle reducing the affinity of transferrin for iron, so it is
followed by endocytosis [8,9]. released into the cytosol [6]. The density of transferrin
receptors on the cell surface reflects cell requirements;
In the enterocyte, Fe2þ from all dietary sources enters a increased iron requirement (or iron deprivation) upregu-
common labile iron pool within the cytosol from which it lates synthesis of transferrin receptors (their expression is
may be used for cellular metabolism, sequestered within particularly high on dividing cells such as RBC precursors
the enterocyte as ferritin, where it may be retained until and placenta) [19]. A monomeric fragment is cleaved
the enterocyte is sloughed off into the lumen of the gut, off from the transferrin receptor resulting in very low
or exported into the bloodstream. Fe2þ is exported via concentrations of soluble transferrin receptors (sTfRs) in
the Fe2þ transporter, ferroportin [6], on the basolateral the serum; receptor number correlates well with the
membrane and subsequently oxidized to Fe3þ by number of erythroid precursors [20], thus measurement
hephaestin, a membrane-bound ferroxidase [10]. of sTfR indicates iron demand versus supply.

Ferroportin is involved in the efflux of iron into the Storage


blood-stream from both enterocytes and macrophages, Iron is stored largely in the reticuloendothelial cells of
release of iron from other tissues into the bloodstream and the liver, the spleen and bone marrow as ferritin which is
placental transfer of iron to the fetus. The ferroportin- a protein shell (apo-ferritin) made up of 24 heavy and
mediated egress of Fe2þ is negatively regulated by the light subunits, involved in both iron storage and iron
hepatic peptide, hepcidin, which binds to ferroportin detoxification. Up to 4500 Fe3þ ions are deposited in the
resulting in its internalization and subsequent degradation. core of apo-ferritin as insoluble ferric hydroxide phos-
phate forming holo-ferritin. Iron reserves act to buffer
Hepcidin also inhibits the expression of DMT1 [11] and increased physiological demands for iron such as in
dcytB on the apical membrane of the enterocyte, thus pregnancy and with acute blood loss. Minute quantities
abrogating iron uptake into the cell. Hepcidin levels of the light subunit of ferritin (free of iron) are present in
decrease in IDA, hypoxia and oxidative stress [12], so serum; serum ferritin levels correlate well with storage
intestinal iron absorption, release of recycled iron from iron which varies with age and sex. In women who are not
macrophages and mobilization of stored iron from pregnant or lactating, a serum ferritin concentration of
hepatocytes are all increased [13]. Hepcidin levels 1 mg/l is equivalent to about 7–8 mg mobilizable iron, so a
increase in response to inflammatory cytokines and infec- serum ferritin concentration of 50 mg/l indicates that iron
tion [14]; the increased hepcidin in response to infection stores are adequate (350–400 mg) for normal needs [21].
is important in depriving pathogens of essential iron
which inhibits their proliferation [15]. Conditions of Combinations of biomarkers are valuable; the sTfR :
systemic iron overload result from a lack of control by serum ferritin (R : F) ratio is a particularly useful marker
hepcidin of the ferroportin-mediated export of iron; of iron status which has been applied to population studies
excessively high hepcidin levels are responsible for the [22]; it gives a continuum across the sequential stages of
anaemias associated with chronic diseases, inflammation, iron deficiency. In addition, the sTfR : serum ferritin ratio
cancer and ageing. has been validated against serial phlebotomy to determine
a quantitative measure of body iron stores or tissue iron
Transport deficiency [20].
Iron exported from the enterocyte is transported to
all tissues as diferric transferrin (Tf); transferrin also Several biomarkers, including serum iron, serum ferritin
scavenges iron recycled from effete red cells by reti- and transferrin saturation, used to assess iron status are
culoendothelial cells. Plasma Tf has a high-binding acute-phase reactants to inflammatory cytokines (which
capacity and under normal physiological conditions, is are increased with a range of conditions including
hyposaturated (about 25–50%) and in excess of iron infection, inflammation, liver disease, hyperthyroidism,
concentration so there is a negligible amount of circulat- malignancy, alcohol consumption, obesity and use of oral
ing, nontransferrin-bound iron which has a high redox contraceptives). This means that inflammatory states are
activity and is potentially toxic [16]. In iron deficiency, associated with increased risk of iron deficiency and it is
Tf saturation may fall below 5% [17]; at Tf saturation less important to concurrently assess biomarkers of iron status
than 15%, delivery of iron to the bone marrow is unable to with acute phase protein biomarkers such as C-reactive
sustain normal rates of erythropoiesis, so Tf saturation is a protein (CRP) and/or a1-acid glycoprotein (AGP). Inter-
key factor for establishing iron deficiency [18]. pretation of test results presents some challenges; either
individuals with inflammation can be excluded from
Cells express specific transferrin receptors (TfRs); analysis of data or the cut-off definition of low ferritin
the ligand–receptor complex is taken up by endocytosis. concentration is increased, for example, from 12–15 to

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
628 Micronutrients

30–50 mg/l [23]. It is argued that measuring both CRP of the effete erythrocytes and phagocytose them.
and AGP presents the optimal picture of inflammatory The iron from the erythrocytes is then retained inside
time-course: CRP rises rapidly, peaking between 24 and the macrophages which act as an iron store until the iron is
48 h, whereas AGP increases over 4–5 days [24]; cut-off transported out of the cells by ferroportin. Iron liberated
levels for confirming inflammation are greater than 5 mg/l and recycled from senescent red cells contributes about
and greater than 1 g/l, respectively. 20–25 mg of iron each day which is about 90% of that
required for erythropoiesis, the remainder comes from
With the increasing prevalence of overweight and obesity the diet.
worldwide, use of biomarkers of iron status which can be
affected by inflammatory cytokines is challenging. Although most of the body iron (about 40 mg/kg body
It seems that increased body adipose tissue, particularly weight) is present in erythrocytes and myoglobin, iron has
visceral depots, is associated with increased risk of iron a myriad of other functions which derive from the ability
deficiency which may be masked by high serum ferritin of iron to donate electrons. A small proportion of body
levels [25], presumably because the cytokines increase iron is an essential component of the metalloenzymes
hepcidin synthesis resulting in increased macrophage involved in the formation of ATP and DNA synthesis.
sequestration and/or decreased intestinal iron absorption In addition, cellular proliferation requires iron; dividing
[26]. cells, such as erythrocyte precursors and immune cells,
have increased expression of TfR. Iron is used by
Utilization neutrophils and macrophages to generate the cytotoxic
Erythropoiesis is driven by erythropoietin and limited free radical oxygen burst.
by nutrient availability. Transferrin delivers most of
the circulating iron to the erythropoietic precursors in Iron concentrations in the brain are abundant with an
the bone marrow. Erythropoiesis takes about 7 days. The unusual and distinctive pattern of distribution [30] that
final stage is the release of reticulocytes which circulate in probably reflects differences in cell division, myelination,
the bloodstream for about a day before they mature into metabolism and neurotransmission. Iron uptake by the
erythrocytes; reticulocytes can be identified because they brain is regulated by the expression of transferrin recep-
are larger than RBC, contain remnants of nuclei and have tors on the endothelial cells of the brain microvasculature
a high surface concentration of TfR which are shed as [31]. How iron is redistributed within the brain is not well
the erythrocytes mature [18]. Usually reticulocyte understood, but iron continues to accumulate in humans
number comprises 1% of the total RBCs (50 000/ml of until the fourth decade of life and then plateaus [32].
blood); increased reticulocyte number reflects increased The amount of iron accumulated exceeds the amount
erythropoiesis [18]. Iron deficiency is associated with calculated to be required by the iron-dependent
increased release of reticulocytes which are larger processes by about 10-fold [33]; it is not clear what its
and paler than normal. Indices such as reticulocyte size role is but inappropriate accumulation, either excess or
(Ret-Y) and Hb content (ChR) are useful markers of mislocalization, of iron with age is associated with
functional iron deficiency [27]. neurodegenerative and movement disorders, including
Alzheimer and Parkinson’s diseases [30], age-related
The final step in Hb formation is the combination macular degeneration and restless legs syndrome [34].
of ferrous protoporphyrin (haem) and globin. Usually, The mechanism involved is probably iron-catalyzed
zinc protoporphyrin is produced in trace amounts during oxidative stress, particularly affecting mitochondria
haem synthesis; but in iron deficiency, zinc ions replace [35]; indeed, recent studies in mouse models suggest
some of the ferrous ions in the last stages of erythropoiesis that iron chelation therapy, even in the absence of iron
so more zinc protoporphyrin (ZPP) is formed and deregulation, may have neuroprotective potential [36].
incorporated into red cells [28]. Iron deficiency results
in compromised Hb synthesis and microcytosis; small, Animal studies suggest that although the developing
hypochromic and abnormally shaped cells are evident brain is vulnerable to iron deficiency [37], the adult brain
on a blood smear [29]. Although abnormalities in cell conserves iron moderately well even in the presence of
morphology, which can be detected by automated cell iron deficiency, although severe iron deficiency can cause
counters, are useful in differential diagnosis of the causes changes in brain fatty acid profile because iron is a
of anaemia, the slow turnover of RBCs means that these component of the fatty acid desaturase enzymes [38].
are only detected when iron deficiency has been present
for months. Erythrocytes contain 80% of the body’s
functional iron and have a lifespan of about 90–120 days, Pregnancy
so about 1% of erythrocytes are replaced each day. Pregnancy is marked by significant changes in the
Kupffer cells in the liver and other macrophages predo- haematological system [39]: the increased plasma volume
minantly in the spleen recognize the membrane changes is greater than the increased production of cellular

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Iron deficiency in women Coad and Conlon 629

components, so hypovolaemia causes progressive nosebleeds and foot-strike-haemolysis in athletes can


changes in many of the common haematological labora- also have an impact on iron balance.
tory values [40] (see Table 2). Blood volume increases
by 30–40% (about 1.5 l; more with multiple gestation) Pregnancy presents a significant physiological challenge
from 6 to 8 weeks, peaking at 28–34 weeks; increased to iron balance. The iron cost of pregnancy is about 1.2 g,
RBC number lags slightly behind. The expansion comprising approximately 270 mg in the fetus, approxi-
of plasma volume is positively associated with the mately 90 mg in the placenta, approximately 450 mg
outcome of pregnancy; lack of haemodilution is usually in maternal erythrocyte expansion and approximately
because of preeclampsia [39]. However, true com- 230 mg total basal losses [49]. Although approximately
promised iron status in pregnancy is associated with 600 mg can be recovered from cessation of menses and
adverse outcomes. recovery of the erythrocyte iron at the end of pregnancy, a
further approximately 600 mg is required predominantly
There is a physiological increase in inflammatory in the last two trimesters. Iron stores of 600 mg mean that
biomarkers in pregnancy, particularly in the first and the serum ferritin concentration in early pregnancy
third trimesters [43], so the cut off for serum ferritin in (before inflammatory cytokines increase) needs to be
pregnancy is higher than for nonpregnant women but 75–85 mg/l to prevent depletion of iron stores without
serum ferritin levels can markedly increase as a result of the need for supplementation.
cellular damage associated with preeclampsia [44].
The chances of achieving adequate iron status in
pregnancy are optimized if iron stores, assessed by serum
Causes of iron deficiency ferritin concentration, are adequate at the time of con-
Iron deficiency is due to increased requirement, such as ception, which can be affected by pregnancy spacing,
growth or pregnancy, increased blood loss and/or low and if the diet has a high bioavailability of iron. Animal
dietary intake of iron. Although iron balance is main- studies suggest the foetal liver regulates several com-
tained by iron acquisition, the ability of the body to ponents of iron balance including maternal absorption,
increase iron uptake across the gastrointestinal tract is maternal storage of iron and placental transfer [50].
finite. Worldwide, the most significant factors influencing Although an elegant classical study demonstrated marked
iron deficiency are increased blood loss due to parasitic increases in iron absorption (from 1 to 1.5 mg per day in
infection and limited dietary diversity due to poverty and the first trimester to 5 mg per day in second trimester and
low bioavailability of iron from a diet high in plants foods 9 mg per day in the third trimester) [51], the World
and low in animal-derived foods [45]. In developed Health Organization has not revised its recommendation
countries where the burden of infection is less, the usual of approximately 60 mg per day supplemental iron for the
causes of iron deficiency in premenopausal nonpregnant last 6 months of pregnancy [52]. However, routine iron
women are a low intake of dietary iron, menstrual losses supplementation is questioned in many countries and
and blood donation. Vegetarian diets have lower bio- concerns have been expressed about whether additional
availability of iron, so vegetarian women of reproductive iron may be positively harmful for iron-replete pregnant
age are recommended to increase their iron intake by women and associated with gestational diabetes and
1.8-fold [46] from 18 to 32 mg/day. preeclampsia [53].

Iron deficiency develops when absorption of dietary In older individuals, chronic blood loss, as a result of use
iron cannot match the obligate losses, which are predo- of NSAIDs (e.g. aspirin), gastrointestinal lesions or
minantly from desquamated skin (0.3 mg per day) and cancer, is the most common cause of iron deficiency
gastrointestinal cells (0.6 mg per day) plus small [54]. Decreased production of gastric acid (hypochlorhy-
amounts in sweat and urine (0.1 mg per day), that in dria or achlorydria), which can be either age related or
total account for approximately 1 mg iron/day, and drug induced, for example, by proton pump inhibitors,
variable blood loss. The average menstrual blood loss negatively affects iron absorption by increasing iron
is 35 ml per cycle, with a range of 25–60 ml [39]. It is insolubility and therefore decreasing the bioavailability
lower in women using oral contraception and higher in of iron in the proximal duodenum [55]. In addition,
women using intrauterine contraceptive devices [47] and malabsorption conditions, such as those associated with
can be in excess of 250 ml (average 60 ml) in women coeliac disease [56] and Helicobacter pylori infection [57],
of late reproductive age [48]. A blood loss of 35 ml per can also compromise iron status.
28-day cycle equates to a loss of 0.5–0.68 mg iron per day
in addition to other obligate iron losses. The usual blood
donation unit of about 450 ml equates to an additional Physiological response to iron depletion
loss of 1–1.35 mg per day, assuming 6 months between As iron balance becomes negative, there is increased iron
blood donations. Other sources of blood loss such as release from ferritin. Ferritin depots fall, so tissue and

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
630 Micronutrients

Table 2 Biomarkers of iron status in women and changes in pregnancy


Normal levels Changes in
Biomarker in women pregnancy Best use Advantages Limitations

Markers of functional iron status


Hb concentration 120–160 g/l First trimester: To determine severity Inexpensive and Not specific to iron
of anaemia and/or simple to deficiency
response to measure
treatment
115–140 g/l Low sensitivity
Second trimester: Indicates late stage of
iron depletion – does
not identify iron
deficiency. Influenced
by ethnicity, altitude,
smoking
97–148 g/l Unreliable in pregnancy
unless trimester
specific values are
used
Third trimester:
95–150 g/l
Note: normal or raised
Hb levels usually
indicate lack of
plasma expansion
RBC indices RBC count: RBC count: Abnormal Reflect availability Not specific to
4.2–5.4 million/ml 3.8–4.4 million/ml erythropoiesis. of iron over iron deficiency
previous
90–120 days
Identifies microcytic Late stage of iron
hypochromic depletion
anaemia; low SF
will confirm that
it is due to iron
deficiency
PCV: 37–47% PCV: 33–44% Note that increased Requires expensive
RDW usually automated flow
precedes other cytometry equipment –
markers of iron note that some of the
deficiency determinants of RBC
parameters can only be
measured on specific
brands of flow
cytometers
MCV: 84–99 MCV: 70–90
MCH: 27–32 pg MCH: 27–32 pg
(32–35 fl) (32–35 fl)
MCHC: 32–35 fl MCHC: 32–35 fl
Red cell distribution Red cell distribution
width (RDW): <14.5 width:
First trimester:

12.5–14.1%
Second trimester:
13.4–13.6%
Third trimester:
12.7–15.3%
Reticulocyte count: Reticulocyte count:
0.5–1% 1–2%
Markers of transport/supply
of iron to tissues
Serum iron 41–141 mmol/l First trimester: Short-term fluctuations:
diurnal variation and
increased after meat
ingestion
72–143 Serum iron is an acute
phase reactant to
inflammatory cytokines
Second trimester:
44–178
Third trimester:
30–193 mmol/l

Tf saturation 22–46% Values fall throughout Increases with


pregnancy but to iron overload
a lesser extent with
iron supplementation

(continued overleaf )

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Iron deficiency in women Coad and Conlon 631

Table 2. (continued )

Normal levels Changes in


Biomarker in women pregnancy Best use Advantages Limitations

sTfR 2.8–8.5 mg/l >8.5 mg/l Increased levels indicate Reflects iron supply Assays are not well
early iron deficiency to cells and standardized so
as stores start to erythroid activity reference ranges are
become depleted not robust. Emerging
evidence suggests
levels are increased
in infection
Note that the use of Ethnic differences are
this marker is apparent
contentious in
pregnancy.
ZPP <70 mmol/mol Hb 60 mmol/mol Reflects iron delivery Affected by inflammation
to the developing but not usually seen
erythrocytes. during acute infection
because of time-lag in
producing ZPP-containing
cells
or
<80 mg/dl red cells
% Hypochromic red cells <6% Data not available Indicates
compromised
iron supply during
erythropoiesis
Markers of iron in tissue
SF 15–200 mg/l 30–150 mg/l To confirm depletion Positively correlated Serum ferritin is an acute
of iron stores in the with iron stores phase reactant to
absence of inflammation inflammatory cytokines
so markers of inflammation
should always be measured
Threshold levels are set Note pregnancy is
higher in the presence associated
of infection (>30 mg/l) with increases in
inflammatory
markers, especially
in the first and third
trimester
Serum TIBC 40–80 mmol/l First trimester: Raised in iron-
deficiency anaemia
42–73 mmol/l
Second trimester:
54–93 mmol/l
Third trimester:
68–107 mmol/l
Bone marrow iron 2–3 1–2 To indicate depleted Considered to be Invasive and difficult
staining (Prussian blue) or absent body iron ‘gold standard’ of to collect
(scored on six-point stores. Research use body iron stores
scale: 0 is no detectable
iron, 1 is decreased iron,
2–3 is normal, 4–5
is iron increased)
Limited ability to detect
development of iron
deficiency
Therapeutic trial Confirms iron deficiency
if response to oral iron
therapy (usually 200 mg
of ferrous iron per day)
is an increase in Hb
concentration of 20 g/l
in 3 weeks
Potential markers
Hepcidin Induced by inflammatory
cytokines

Hb, haemoglobin; MCH, mean corpuscular haemoglobin; MCHC, mean corpuscular haemoglobin concentration; MCV, mean corpuscular volume; PCV, packed cell
volume; RBC, red blood cell; RDW, red cell distribution width; SF, serum ferritin; sTfR, soluble transferrin receptor; Tf, transferrin; TIBC, total iron-binding capacity; ZPP,
zinc protoporphyrin. Adapted with permission from [21,39,40,41,42].

serum ferritin levels decrease. However, concurrent upregulation of expression of DMT1 receptors and
infection or inflammation will independently increase ferroportin, so nonhaem iron transport is increased. Haem
serum ferritin levels. The raised ferritin level drives iron transport is also increased. As tissue iron stores
hepcidin production, so iron absorption is not increased become depleted, transferrin saturation falls below
despite a compromised iron status; this is the underlying 15%, and the tissues increase the expression of transferrin
pathology of anaemia of chronic disease [58]. In the receptors in an attempt to procure more iron so soluble
absence of infection, decreased serum ferritin results in TfR levels are elevated. With compromised delivery of

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
632 Micronutrients

iron to functional sites, normal erythropoiesis is disturbed assays [66], so Hb concentration and serum ferritin
with smaller RBCs which have a greater variation in size (and concurrent measurement of CRP) have remained
and reduced Hb content. In addition, there is increased the usual method of assessing iron status.
release of immature RBCs which contain more zinc
protoporphyrin (ZPP), so both ZPP and the ZPP : haem Chronic inflammatory conditions can result in reduced
ratio can be used to diagnose preanaemic iron deficiency iron absorption and increased sequestration of iron, thus
[59]. compromising iron status. That several biomarkers of
iron status are acute phase reactants means that caution
should be exercised in interpreting results in conditions
Consequences of iron deficiency of chronic inflammation. There is a particular need to
Research on the effects of IDWA has focussed on determine how the biomarker profile is altered with
children and effects of iron deficiency on myelination increased adiposity as the incidence of overweight
and neurotransmitter production, because it was assumed and obesity increases. In addition, the iron status of
that iron levels in the brain were predetermined at the hospitalized patients and the elderly may be similarly
time of closure of the blood brain barrier [60]. However, compromised and difficult to interpret.
animal studies have suggested that iron deficiency also
has adverse effects in the adult brain [61]. Recent studies
suggest an association of iron deficiency with neuropsy- Acknowledgements
chological consequences in women of reproductive age, Conflicts of interest
The authors have been the recipients of funding for hosting educational
such as emotions, quality of life and cognition, including events from Pfizer and JC has received funding from the
memory and learning (reviewed in Ref. [60]). Further- Nutricia Research Foundation for research on complementary feeding
more, iron supplementation in young women with and iron.
depleted tissue iron (defined as mild-to-moderate iron
deficiency in the absence of anaemia) has demonstrated
improvements in fatigue resistance, exercise perform- References and recommended reading
Papers of particular interest, published within the annual period of review, have
ance and muscle function [62–64]. been highlighted as:
 of special interest
 of outstanding interest
Iron deficiency in pregnancy, which is common, is Additional references related to this topic can also be found in the Current
associated with maternal and infant complications includ- World Literature section in this issue (pp. 658–659).
ing low birthweight, prematurity, perinatal mortality, 1 WHO/CDC. Worldwide prevalence of anaemia, 1993–2005. WHO global
increased risk of maternal infections and lowered database on anaemia. Geneva: World Health Organization; 2008.
tolerance to blood loss and infection. In addition, it is 2 McLean E, Cogswell M, Egli I, et al. Worldwide prevalence of anaemia, WHO
Vitamin and Mineral Nutrition Information System, 1993–2005. Public Health
evident that maternal iron status may affect mother– Nutr 2009; 12:444–454.
child interactions which can have implications for child 3 Clark SF. Iron deficiency anemia. Nutr Clin Pract 2008; 23:128–141.
development [65]. 4 Sharp PA. Intestinal iron absorption: regulation by dietary & systemic factors.
 Int J Vitam Nutr Res 2010; 80:231–242.
A recent discussion of how dietary components affect iron absorption by regulat-
ing iron solubility and oxidation state and by influencing the expression and function
Conclusion of the iron transporters of the enterocytes.

Despite elucidation of many components of iron meta- 5 McKie AT, Barrow D, Latunde-Dada GO, et al. An iron-regulated ferric
reductase associated with the absorption of dietary iron. Science 2001;
bolism, iron deficiency remains one the most common 291:1755–1759.
nutritional problems in the world. Women are particularly 6 Wang J, Pantopoulos K. Regulation of cellular iron metabolism. Biochem J
at risk because of the challenges of absorbing enough  2011; 434:365–381.
A recent review summarizing the current state of knowledge about the mechan-
additional iron to replace that lost in blood. Emerging isms by which iron metabolism is controlled through the iron responsive element–
evidence suggests that depletion of iron stores may iron-regulatory protein system.

affect neurocognitive function, even in adults before 7 Shayeghi M, Latunde-Dada GO, Oakhill JS, et al. Identification of an intestinal
heme transporter. Cell 2005; 122:789–801.
erythropoiesis is compromised. This means that appro- 8 San Martin CD, Garri C, Pizarro F, et al. Caco-2 intestinal epithelial cells
priate selection of biomarkers to determine early stages of absorb soybean ferritin by mu2 (AP2)-dependent endocytosis. J Nutr 2008;
138:659–666.
iron deficiency is essential not only to monitor effects of
9 Kalgaonkar S, Lonnerdal B. Receptor-mediated uptake of ferritin-bound
iron depletion but also for early recognition of negative iron by human intestinal Caco-2 cells. J Nutr Biochem 2009; 20:304–
iron balance and to identify the appropriate timing and 311.
assessment of interventions. Although the sTfR : serum 10 Frazer DM, Vulpe CD, McKie AT, et al. Cloning and gastrointestinal expression
of rat hephaestin: relationship to other iron transport proteins. Am J Physiol
ferritin ratio appears to be the biomarker of choice as Gastrointest Liver Physiol 2001; 281:G931–G939.
it reflects the prevalence and severity of IDA and is 11 Mena NP, Esparza A, Tapia V, et al. Hepcidin inhibits apical iron uptake in
intestinal cells. Am J Physiol Gastrointest Liver Physiol 2008; 294:G192–
sensitive to measuring the effectiveness of interventions, G198.
it is currently limited by cost and problems with the 12 Darshan D, Anderson GJ. Interacting signals in the control of hepcidin
standardization and inter-assay variation of commercial expression. Biometals 2009; 22:77–87.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Iron deficiency in women Coad and Conlon 633

13 Ganz T, Nemeth E. Hepcidin and disorders of iron metabolism. Annu Rev Med 36 Obolensky A, Berenshtein E, Lederman M, et al. Zinc-desferrioxamine
2011; 62:347–360. attenuates retinal degeneration in the rd10 mouse model of retinitis pigmen-
tosa. Free Radic Biol Med 2011. [Epub ahead of print]. doi: 10.1016/j.free-
14 Viatte L, Vaulont S. Hepcidin, the iron watcher. Biochimie 2009; 91:1223–
radbiomed.2011.07.014
1228.
37 Osendarp SJ, Murray-Kolb LE, Black MM. Case study on iron in mental
15 Nairz M, Schroll A, Sonnweber T, Weiss G. The struggle for iron: a metal at the
development: in memory of John Beard (1947–2009). Nutr Rev 2010; 68
 host–pathogen interface. Cell Microbiol 2010; 12:1691–1702.
(Suppl 1):S48–S52.
This study examines the immune-mediated strategies of the host which restrict
availability of iron to pathogens and discusses the iron-acquisition strategies 38 Badaracco ME, Siri MV, Pasquini JM. Oligodendrogenesis: the role of iron.
developed by pathogens to counter these. Biofactors 2010; 36:98–102.
16 Ozment CP, Turi JL. Iron overload following red blood cell transfusion and 39 Coad J, Dunstall M. Anatomy and physiology for midwives. 3rd ed. London:
its impact on disease severity. Biochim Biophys Acta 2009; 1790:694– Churchill Livingstone (Elsevier); 2011.
701.
40 Abbassi-Ghanavati M, Greer LG, Cunningham FG. Pregnancy and laboratory
17 Andrews NC. Iron metabolism: iron deficiency and iron overload. Annu Rev studies: a reference table for clinicians. Obstet Gynecol 2009; 114:1326–
Genomics Hum Genet 2000; 1:75–98. 1331.
18 Beard J, Han O. Systemic iron status. Biochim Biophys Acta 2009; 41 Geissler C, Singh M. Iron, Meat and Health. Nutrients 2011; 3:283–316.
1790:584–588. A summary of the ‘Iron and Health’ report carried out by the UK Scientific
Advisory Committee on Nutrition (SACN) which examines the uncertainties
19 Aisen P. Transferrin receptor 1. Int J Biochem Cell Biol 2004; 36:2137–
of the dose-response relationships and risks associated with iron deficiency
2143.
and excess.
20 Skikne BS, Flowers CH, Cook JD. Serum transferrin receptor: a quantitative
42 Walsh T, O’Broin SD, Cooley S, et al. Laboratory assessment of iron status in
measure of tissue iron deficiency. Blood 1990; 75:1870–1876.
pregnancy. Clin Chem Lab Med 2011; 49:1225–1230.
21 Wheeler S. Assessment and interpretation of micronutrient status during
43 Brewster JA, Orsi NM, Gopichandran N, et al. Gestational effects on host
pregnancy. Proc Nutr Soc 2008; 67:437–450.
inflammatory response in normal and preeclamptic pregnancies. Eur J Obstet
22 Mei Z, Cogswell ME, Looker AC, et al. Assessment of iron status in US Gynecol Reprod Biol 2008; 140:21–26.
 pregnant women from the National Health and Nutrition Examination Survey
44 Hubel CA, Bodnar LM, Many A, et al. Nonglycosylated ferritin predominates in
(NHANES), 1999–2006. Am J Clin Nutr 2011; 93:1312–1320.
the circulation of women with preeclampsia but not intrauterine growth
This study presents an analysis of the data assessing iron status from pregnant
restriction. Clin Chem 2004; 50:948–951.
women in the U.S. National Health and Nutrition Examination Survey, using serum
ferritin and soluble transferrin receptor concentrations to calculate total body iron. 45 Lynch SR. Why nutritional iron deficiency persists as a worldwide problem.
 J Nutr 2011; 141:763S–768S.
23 Beard JL, Murray-Kolb LE, Rosales FJ, et al. Interpretation of serum ferritin
A commentary reviewing the assessment of iron status and the limitations and
concentrations as indicators of total-body iron stores in survey populations:
advantages of various indicators of iron status.
the role of biomarkers for the acute phase response. Am J Clin Nutr 2006;
84:1498–1505. 46 Institute of Medicine Food and Nutrition Board. Dietary reference intakes
for vitamin A, vitamin K, arsenic, boron, chromium, copper, iodine, iron,
24 Thurnham DI, McCabe LD, Haldar S, et al. Adjusting plasma ferritin
manganese, molybdenum, nickel, silicon, vanadium, and zinc. Washington,
 concentrations to remove the effects of subclinical inflammation in the
DC: National Academy Press; 2001.
assessment of iron deficiency: a meta-analysis. Am J Clin Nutr 2010;
92:546–555. 47 Pynaert I, De Bacquer D, Matthys C, et al. Determinants of ferritin and soluble
A meta-analysis investigating the use of acute-phase proteins as biomarkers of transferrin receptors as iron status parameters in young adult women.
inflammation and how they can be used to correct serum ferritin concentrations in Public Health Nutr 2009; 12:1775–1782.
order to determine iron stores in the presence of concurrent inflammatory states.
48 Hale GE, Manconi F, Luscombe G, Fraser IS. Quantitative measurements
25 Zafon C, Lecube A, Simo R. Iron in obesity. An ancient micronutrient for a of menstrual blood loss in ovulatory and anovulatory cycles in middle- and late-
modern disease. Obes Rev 2010; 11:322–328. reproductive age and the menopausal transition. Obstet Gynecol 2010; 115
(2 Pt 1):249–256.
26 Cepeda-Lopez AC, Osendarp SJ, Melse-Boonstra A, et al. Sharply higher
 rates of iron deficiency in obese Mexican women and children are predicted by 49 Gambling L, Lang C, McArdle HJ. Fetal regulation of iron transport during
obesity-related inflammation rather than by differences in dietary iron intake.  pregnancy. Am J Clin Nutr 2011. [Epub ahead of print]. doi:10.3945/
Am J Clin Nutr 2011; 93:975–983. ajcn.110.000885
This study describes a study which identified that the risk of iron deficiency in This study examines the transfer of iron from the maternal to foetal compartment
obese women in Mexico was two-fold to four-fold higher than their normal-weight and the regulation of the molecular mechanisms involved.
counterparts and that as CRP, not iron intake, predicted this increased risk, a likely
50 Gambling L, Czopek A, Andersen HS, et al. Fetal iron status
explanation is that obesity-related inflammation is responsible.
regulates maternal iron metabolism during pregnancy in the rat. Am J Physiol
27 Urrechaga E, Borque L, Escanero JF. Erythrocyte and reticulocyte indices on Regul Integr Comp Physiol 2009; 296:R1063–R1070.
the LH 750 as potential markers of functional iron deficiency. Anemia 2010;
51 Barrett JF, Whittaker PG, Williams JG, Lind T. Absorption of nonhaem iron
Article ID:625919.
from food during normal pregnancy. BMJ 1994; 309:79–82.
28 Zimmermann MB. Methods to assess iron and iodine status. Br J Nutr 2008;
52 Stoltzfus R, Dreyfuss M. Guidelines for the use of iron supplements to prevent
99 (Suppl 3):S2–S9.
and treat iron deficiency anemia. Washington, DC: International Nutritional
29 WHO/CDC. Assessing the iron status of populations: report of a joint World Anemia Consultative Group/WHO/UNICEF; 1998.
Health Organization/Centers for Disease Control and Prevention technical
53 Weinberg ED. Are iron supplements appropriate for iron replete pregnant
consultation on the assessment of iron status at the population level (second
women? Med Hypotheses 2009; 73:714–715.
edition). Geneva: WHO/CDC; 2007.
54 Goddard AF, James MW, McIntyre AS, Scott BB. Guidelines for the manage-
30 Crichton RR, Dexter DT, Ward RJ. Brain iron metabolism and its perturbation
ment of iron deficiency anaemia. Gut 2011; 60:1309–1316.
in neurological diseases. J Neural Transm 2011; 118:301–314.
55 Sarzynski E, Puttarajappa C, Xie Y, et al. Association between proton pump
31 Mills E, Dong XP, Wang F, Xu H. Mechanisms of brain iron transport: insight
inhibitor use and anemia: a retrospective cohort study. Dig Dis Sci 2011;
into neurodegeneration and CNS disorders. Future Med Chem 2010; 2:51–
56:2349–2353.
64.
56 Hershko C, Skikne B. Pathogenesis and management of iron deficiency
32 Hulet SW, Powers S, Connor JR. Distribution of transferrin and ferritin binding anemia: emerging role of celiac disease, Helicobacter pylori, and autoimmune
in normal and multiple sclerotic human brains. J Neurol Sci 1999; 165:48– gastritis. Semin Hematol 2009; 46:339–350.
55.
57 Qu XH, Huang XL, Xiong P, et al. Does Helicobacter pylori infection play a role
33 Zecca L, Youdim MB, Riederer P, et al. Iron, brain ageing and neurodegen- in iron deficiency anemia? A meta-analysis. World J Gastroenterol 2010;
erative disorders. Nat Rev Neurosci 2004; 5:863–873. 16:886–896.
34 Connor JR, Ponnuru P, Wang XS, et al. Profile of altered brain iron acquisition 58 Weiss G. Iron metabolism in the anemia of chronic disease. Biochim Biophys
in restless legs syndrome. Brain 2011; 134 (Pt 4):959–968. Acta 2009; 1790:682–693.
35 Horowitz MP, Greenamyre JT. Mitochondrial iron metabolism and its 59 Rettmer RL, Carlson TH, Origenes ML, et al. Zinc protoporphyrin/
role in neurodegeneration. J Alzheimers Dis 2010; 20 (Suppl 2):S551– heme ratio for diagnosis of preanemic iron deficiency. Pediatrics 1999;
S568. 104:e37.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
634 Micronutrients

60 Murray-Kolb LE. Iron status and neuropsychological consequences in 63 Brownlie T, Utermohlen V, Hinton PS, Haas JD. Tissue iron deficiency without
 women of reproductive age: what do we know and where are we headed? anemia impairs adaptation in endurance capacity after aerobic training in
J Nutr 2011; 141:747S–755S. previously untrained women. Am J Clin Nutr 2004; 79:437–443.
A recent review from one of the late John Beard’s collaborators which reviews the
64 Hinton PS, Sinclair LM. Iron supplementation maintains ventilatory
emerging data on the consequences of iron deficiency without anaemia in women,
threshold and improves energetic efficiency in iron-deficient nonanemic
particularly on cognition and behaviour.
athletes. Eur J Clin Nutr 2007; 61:30–39.

61 Burhans MS, Dailey C, Beard Z, et al. Iron deficiency: differential effects on 65 Murray-Kolb LE, Beard JL. Iron deficiency and child and maternal health. Am J
monoamine transporters. Nutr Neurosci 2005; 8:31–38. Clin Nutr 2009; 89:946S–950S.
66 Lynch SR. Improving the assessment of iron status. Am J Clin Nutr 2011;
62 Brutsaert TD, Hernandez-Cordero S, Rivera J, et al. Iron supplementation  93:1188–1189.
improves progressive fatigue resistance during dynamic knee extensor A short editorial article which pleads for more extensive use of the
exercise in iron-depleted, nonanemic women. Am J Clin Nutr 2003; sTfR : serum ferritin (R : F) ratio in assessing iron status and summarizes the
77:441–448. reasons why it is not more widely used.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

You might also like