Next Article in Journal
Lingonberries—General and Oral Effects on the Microbiome and Inflammation
Next Article in Special Issue
Maternal Diet, Infection, and Risk of Cord Blood Inflammation in the Bangladesh Projahnmo Pregnancy Cohort
Previous Article in Journal
A Novel UC Exclusion Diet and Antibiotics for Treatment of Mild to Moderate Pediatric Ulcerative Colitis: A Prospective Open-Label Pilot Study
Previous Article in Special Issue
Impact of Fetal Growth Restriction on the Neonatal Microglial Proteome in the Rat
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Iron and Neurodevelopment in Preterm Infants: A Narrative Review

Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
*
Author to whom correspondence should be addressed.
Nutrients 2021, 13(11), 3737; https://doi.org/10.3390/nu13113737
Submission received: 22 September 2021 / Revised: 19 October 2021 / Accepted: 21 October 2021 / Published: 23 October 2021
(This article belongs to the Collection Nutrition for Brain Development and Repair)

Abstract

:
Iron is critical for brain development, playing key roles in synaptogenesis, myelination, energy metabolism and neurotransmitter production. NICU infants are at particular risk for iron deficiency due to high iron needs, preterm birth, disruptions in maternal or placental health and phlebotomy. If deficiency occurs during critical periods of brain development, this may lead to permanent alterations in brain structure and function which is not reversible despite later supplementation. Children with perinatal iron deficiency have been shown to have delayed nerve conduction speeds, disrupted sleep patterns, impaired recognition memory, motor deficits and lower global developmental scores which may be present as early as in the neonatal period and persist into adulthood. Based on this, ensuring brain iron sufficiency during the neonatal period is critical to optimizing neurodevelopmental outcomes and iron supplementation should be targeted to iron measures that correlate with improved outcomes.

1. Introduction

Iron is an essential mineral and iron deficiency is the most common micronutrient deficiency worldwide, affecting approximately 2 billion individuals globally [1]. While the majority of iron deficiency occurs in low-income countries, reproductive-age women and children represent particularly at-risk groups even in middle- and high-income countries [2] and due to decreasing intake of iron-rich foods [3], the rate of iron deficiency within this population may be increasing [4].
The primary role of iron in the body is to facilitate blood oxygen-carrying capacity and delivery. Approximately 70% of iron in the body is found in red blood cells as a component of hemoglobin. In addition to its important role in erythropoiesis, iron also plays key roles in the brain including brain growth, myelination, neurotransmitter production and energy production [5,6]. Based on the critical importance of iron for oxygen delivery, animal studies have shown that iron is prioritized for red blood cell production over other uses for iron, including in the brain [7]. Based on this, changes in red blood cell indices, such as anemia, represent a late marker of iron deficiency. When severe, iron deficiency with inadequate supplementation leads to iron deficiency anemia. The brain, which undergoes rapid development in the neonatal period and is reliant on iron for much of this development, appears to be particularly vulnerable to iron deficiency states. Iron deficiency during key periods of development may lead to disrupted development in areas of the brain that are sensitive to iron status and are undergoing rapid development during these periods. Iron deficiency during these periods (which appear to be the pre-natal and neonatal/infancy period) may have long-lasting neurodevelopmental effects which are not reversible with late supplementation [8,9]. As brain iron stores are depleted before the erythropoietic pool, these neurodevelopmental effects may be seen at iron deficiency levels prior to the emergence of anemia. Conversely, iron excess may also have detrimental effects, highlighting the importance of optimizing iron status, particularly in developing infants.
In this review, we will focus on the importance of iron for brain function and development in the neonatal population. Additionally, we will briefly review key hurdles to overcoming iron imbalance in neonates.

2. Iron in Neonates

Maintaining a normal iron balance is particularly important in the neonatal population, and infants are at higher risk than adults for both iron deficiency and overload. Given that the bulk of iron transfer occurs during the third trimester of pregnancy [10], preterm neonates are at particular risk for deficiency if they do not receive adequate supplements. Maternal iron deficiency and conditions that lead to impaired placental function or inflammation, such as diabetes, smoking and obesity, may also lead to impaired fetal stores as iron is actively transported across the placenta [11,12,13]. Iron stores are most critical during the first 4–6 months of life until infants take in iron through supplementary foods, as breastmilk is low in iron (though what limited iron is present is readily absorbed as lactoferrin) [14,15]. Additionally, rapid somatic growth and red cell expansion that occurs following preterm birth leads to increased iron demand in the newborn period. Iron demand is further increased in critically ill neonates due to repeated phlebotomy.

2.1. Iron Metabolism

Due to the risks attributable to both iron deficiency and iron overload, iron status is highly regulated in children and adults. Hepcidin, a polypeptide hormone produced in the liver, is the primary regulator of iron absorption and availability. It acts on the transmembrane protein ferroportin. Ferroportin is present on the basal surface of intestinal cells and on cells that commonly store iron such as macrophages and hepatocytes. Ferroportin allows absorption of iron from intestinal epithelial cells into the bloodstream where it is transported to sites of need bound to transferrin. During periods of iron sufficiency, hepcidin values increase. Hepcidin causes the internalization and degradation of ferroportin leading to decreased iron absorption from the gut and sequestration of iron in storage cells such ass hepatocytes and macrophages [16,17]. During periods of iron deficiency or with high iron need, such as during pregnancy, hepcidin is suppressed, thus increasing iron absorption and mobilization [17,18]. The role of hepcidin regulation during pregnancy has been shown to be critical with postnatal iron deficiency being highly correlated with newborn iron values and thus fetal iron loading. Thus, dysregulation of the iron-hepcidin–ferroportin axis in both neonates and pregnant women can significantly impact neonatal iron status and neurodevelopmental outcomes [19,20]. Further, both iron excess and deficiency during pregnancy have been associated with worse neonatal and maternal outcomes [21,22]. For example, maternal anemia has been associated with preterm birth and growth restriction [23,24], highlighting the importance of targeting optimization of iron status in pregnant women to improve neurodevelopmental outcomes of neonates.
Hepcidin levels have also been shown to rise with inflammation [25]. Theoretically this aids to sequester iron from siderophilic bacteria.
Emerging studies have suggested that hepcidin regulation may be intact in the neonatal population [26,27,28].

2.2. Iron Supplementation

Due to the high prevalence of iron deficiency worldwide, the majority of research in the field of iron metabolism and supplementation has focused on iron deficiency and iron deficiency anemia. However, free iron is a potential pro-oxidant and iron-mediated ferroptosis has been implicated as a potential cause of brain injury in neonates. Because preterm infants have immature anti-oxidant mechanisms, they may be at particular risk of iron overload. Adverse effects of iron supplementation in iron-sufficient infants and children have been shown, though results are inconsistent and depend upon risk factors of the population. For example, iron supplementation in populations at high risk of malaria has been associated with increased risk of infection, morbidity and mortality from malaria [29,30]. In a prospective study of iron fortification in infancy, Lozoff et al. found that iron-sufficient infants with normal hemoglobin levels who received iron-fortified formula had worse outcomes, compared to the improved outcomes for iron deficient infants in this cohort who received supplements [31]. By contrast, Singhal et al. randomized infants to unfortified cows milk vs. low or high iron-fortified formula in a large cohort in the UK. They did not find any adverse effects on gastrointestinal symptoms, morbidity or infection in those treated with high-iron formula, despite a low prevalence of iron deficiency [32].
Based on this, accurate assessment of iron status is needed to guide iron supplementation, particularly in neonates who are at risk for both iron deficiency and toxicity, described by Brannon and Taylor as a U-shaped risk curve for iron in this population [33].
Due to the risks attributable to iron deficiency and the increased prevalence of low iron stores in the preterm population, many regulatory bodies recommend iron supplementation in this population, though the amount of iron that optimizes outcomes is not clear and supplementation guidelines are not uniform throughout different organizations. The American Academy of Pediatrics recommends that preterm infants receive 2 mg/kg/day of iron supplementation beginning in infancy and that exclusively breastfed infants receive 1 mg/kg/day of iron supplements beginning at 4 months of age [34,35]. Similarly, the European Society of Pediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN) recommends that breastfed, preterm infants receive 2–3 mg/kg/day of iron supplements through 6 months of life [36]. By contrast, the United States Preventative Services Task Force finds insufficient evidence to recommend routine supplementation of iron [37]. However, emerging research in the field of iron metabolism suggests that 2 mg/kg/day may be insufficient to replenish iron stores in high-risk populations such as preterm neonates and enteral supplementation doses up to 12 mg/kg/day have been used in several prospective studies without noted ill effects [38,39]. In fact, improved developmental scores have been seen with higher doses of supplementation in limited studies [40]. Ongoing research is aimed at evaluating optimal doses of iron supplementation (NCT04691843).

2.3. Monitoring Iron Status

Due to the risks of both iron deficiency and overload, careful regulation of supplementation is likely needed to ensure optimal iron balance. However, due to the normal hematologic changes of infancy, frequent transfusions and inflammatory conditions, accurate assessment of iron status can be challenging in neonatal populations. Table 1 summarizes the iron indices most commonly used in this population, with the advantages and disadvantages of each measure and their relation to neurodevelopment highlighted. Based on currently available evidence, a combined approach is likely needed.

2.4. Iron and Neurodevelopment

The association between iron deficiency and adverse short-, medium- and long-term outcomes has been widely studied in both animal models and in human infants and children. Iron deficiency during the neonatal period has been consistently associated with worse neurodevelopmental outcomes broadly, with specific areas of deficit related to brain regions that appear to be most affected by iron deficiency, such as the hippocampus and myelination [53,54,55]. Unique to infants, brain iron deficiency is induced more quickly in the setting of iron deficiency and disruptions in development caused by iron deficit are not reversible with later repletion [56], which makes ensuring adequate brain iron status particularly critical in the neonatal period.
Animal models have elucidated both biochemical and structural effects of iron deficiency on the brain and demonstrated that susceptibility periods to iron deficiency vary over time and in different areas of the brain [57]. The hippocampus is a highly metabolically active part of the brain that undergoes rapid development in the neonatal and toddler period. Rodent and primate models of iron deficiency demonstrate less complex dendritic arborization [58], alterations in synaptic plasticity and regulation [59], impaired dopamine-mediated neurotransmission [56] and persistent metabolic disruptions [60]. Through the use of knockout mice to prevent iron utilization in the brain without anemia, these models are able to specifically demonstrate that these findings are attributable to iron deficiency rather than anemia, allowing the isolated impact of iron deficiency (versus anemia and impaired oxygen-carrying capacity) to be determined [59].
Neurodevelopmental effects of iron deficiency can be seen early in the neonatal period. In children, one tool for assessing short-term brain changes includes the Auditory Brainstem Response (ABR), a test which correlates with neuronal transmission speed and therefore myelination. Amin et al. showed shorter inter-wave latencies in iron-replete versus -deplete newborns [61]. Similarly, Berglund et al. found that iron supplementation correlated with shortened inter-wave latencies on ABR [62]. Algarin et al. showed that differences in ABR latencies persisted into early childhood, with children with a history of iron deficiency anemia showing longer inter-wave latencies at 4 years of age [63]. Even changes in infant temperament and reflexes can be noted in the neonatal period in association with iron status [64,65].
Medium-term studies have examined neurodevelopmental outcomes of children with perinatal iron deficiency in toddlerhood and early school age. These studies have shown a wide range of neurodevelopmental effects associated with low iron status and/or low iron supplementation including lower language skills [19], higher inattention scores [19], disruptions in sleep patterns [66], impaired recognition memory [67] disrupted motor development [19,68], slower processing speed [68], and social interaction differences [68].
Dr. Lozoff and her colleagues have followed long-term cohorts of children from Chile and Costa Rica, evaluating associations between iron status in infancy and developmental measures from infancy through early adulthood [69,70,71,72,73]. Her group has found that young adults with a history of iron deficiency in infancy have worse executive function skills, recognition memory and cognitive scores. Additionally, iron deficiency in infancy was found to be associated with increased inattention and lower verbal IQ scores in childhood and lower educational attainment in adulthood, which they postulate may be related to the impacts of iron deficiency in childhood [70]. Impacts on self-reported emotional health were also observed [71]. However, studies by this group also show that universal iron supplementation in formula may have detrimental effects highlighting the importance of targeting supplements to need [74,75]. Additional long-term studies include a potential association seen between maternal anemia (felt likely to be secondary to iron deficiency) and increased risk of psychiatric disorders in offspring [76,77], though these studies are observational in nature therefore results must be interpreted with caution. Taken together, there is evidence that iron deficiency in the perinatal period has the potential to effect long-term change into adulthood further emphasizing the importance of this critical window.
Although iron deficiency in the neonatal period is of the most significant importance, later iron deficiency does have cognitive and other neurophysiologic effects. Even in adults who develop iron deficiency, performance differences can be seen [78], though iron overload is associated with neurodegenerative conditions [79], emphasizing the importance of maintaining a normal iron balance throughout an individual’s lifespan.
Conversely, iron overload may also have neurodevelopmental effects, though fewer studies exist in the literature examining the long-term effects of iron overload on preterm infants. This skew may reflect either that iron deficiency versus overload is of greater concern to this population or may reflect a bias in the field [80]. Free iron, through production of hydroxyl radicals via the Fenton reaction, is a pro-oxidant which may lead to direct cellular injury through lipid peroxidation. Preterm infants, who have immature anti-oxidant mechanisms and high transferrin saturation levels, may be at particular risk to free radical-mediated cell injury [81]. Oxidative stress has been implicated in several diseases of prematurity, including retinopathy of prematurity and chronic lung disease. Iron-rich transfusions have been associated with these disease conditions, though these studies suffer from the confounding effects of transfusions’ activation of the inflammatory cascade and higher use in sicker preterm infants which limits our ability to understand the direct role of iron on these conditions [82,83,84].
Free iron released from brain hemorrhage is clearly associated with neonatal brain injury through ferroptosis (i.e., iron-mediated cell death). Hemin, which is produced from hemoglobin breakdown and leads to release of Fe2+ and Fe3+, induces astrocyte injury in mouse models through free radical production leading to lipid peroxidation [85] and protein and DNA injury [81]. This iron-mediated cascade leads to a “second hit” injury in neonates who suffer intraventricular hemorrhage, with an initial insult related to the hematoma itself and a secondary injury caused by iron-mediated ferroptosis. The role of iron in neonatal brain injury is suggested by studies which show protective effects of iron chelators and erythropoietin [86]. However, it is unclear whether non-heme-derived iron has the same detrimental effect [85]. Some studies have shown that elevated iron predates intraventricular hemorrhage, suggesting that elevated iron may play a role in brain injury, though these studies are limited in size, thus making definitive conclusions difficult [87]. Additionally, markers of oxidative injury do not appear to be associated with enteral iron supplementation [88], though studies are mixed [89]. This may suggest that enteral iron does not have the same toxicity risks in comparison with iron released from hemorrhagic injury, though further studies are needed. Any discrepancy may be due to the regulatory effects of hepcidin, which help prevent the absorption of excess iron and appear to be functional in the preterm population [26,28,90] or due to the protective effects of lactoferrin in infants fed human milk, which may protect against iron-mediated lipid peroxidation [91].
Long-term neurodevelopmental effects that are directly attributable to iron overload are unclear [92]. Iron overload is thought to underlie many neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease. Animal studies have suggested a potential risk for these effects based on high-dose supplementation in infancy, but to our knowledge this effect has not been reported in the human setting [93].

3. Recommendations and Future Directions

Despite considerable research focused on the importance of iron status and its impacts on brain development, the optimal iron supplementation dose, monitoring regimen, and target markers are still not clear. Because of this, the most important component of iron regulation in neonates is to avoid the development of iron imbalance through minimizing maternal iron deficiency, optimizing maternal health during pregnancy, and preventing neonatal phlebotomy.
To address ongoing gaps in our knowledge of neonatal iron homeostasis, the evaluation of iron marker set-points that correlate with non-hematologic outcome measures would be of significant benefit [33]. Tamura et al. found that infants with ferritin values in the lowest quartile, <76 microgram/L, were associated with worse neurodevelopmental outcomes [19]. Based on this, we would propose that ferritin values above this range are likely needed to ensure adequate iron is available for brain development. However, optimal cutoffs for ferritin and other iron markers, that maximize outcomes have not been established.
Phlebotomy in critically ill neonates is a primary contributor to iron deficiency. Measuring iron status through serum measures may potentiate these losses. Emerging studies have started to evaluate non-hematologic measures of iron status, such as urine ferritin or urine hepcidin [28,90,94]. Additionally, measures such as reticulocyte hemoglobin equivalent, which is measured in conjunction with a complete blood cell count on many analyzers, may be of benefit, though more robust assessment of goal values that optimize outcomes in critically ill neonates are needed.
Finally, we are beginning to recognize the critical importance of the gut microbiome on health. Of particular relevance to this review, is the association between the gut bacterial population and neurodevelopment, with high levels of Bacteroides species associated with improved developmental outcomes [95]. Although the exact mechanism through which the microbiome affects neurodevelopment is unclear, it has been well documented that dysbiosis is correlated with neurodevelopmental effects. When considering enteral iron supplements, although regulation of iron absorption through hepcidin [96] may protect infants against iron toxicity when supplements are administered enterally, high levels of unabsorbed iron in the gut lumen may have significant impacts on microbial populations and gut inflammation [97,98]. Specifically, potentially pathogenic species, such as Clostridia, E. coli and Pseudomonas are thought to expand in the presence of increased luminal iron [99,100], suggesting that excessive enteral iron supplementation is not without risk, particularly in preterm infants who are already at increased risk for dysbiosis and intestinal inflammation. Further studies are needed to identify ways to balance iron supplementation with microbiome health through alternative formulations and potentially intravenous preparations.

4. Conclusions

In summary, optimizing iron balance is an important component of neurodevelopmentally-focused neonatal critical care as it represents a common yet potentially modifiable intervention. While iron excess should be avoided, there is sufficient evidence to support iron supplementation in infants with iron deficiency in order to prevent anemia and ensure that brain development is not interrupted. However, there is likely no universal supplementation dose that is appropriate for all neonates since infants have variable risk factors both pre and postnatally. By contrast, an individualized supplementation strategy based on iron measures is needed in at-risk infants. Future national and other expert guidelines would benefit from recognition of the need for individualized dosing guidelines in preterm neonates and ideally individualized strategies based on iron measure targets that are based on neurodevelopmental outcomes. Ongoing research in this field has the potential to improve outcomes in both high- and low-resource settings.

Author Contributions

K.R.G. and S.E.J. jointly conceptualized this article. The preliminary draft was written by K.R.G., with extensive reviewing and editing by S.E.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable for this review article.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. De Benoist, B.; Cogswell, M.; Egli, I.; McLean, E. Worldwide Prevalence of Anaemia 1993–2005—WHO Global Database on Anaemia; WHO Press: Geneva, Switzerland, 2008. [Google Scholar]
  2. Auerbach, M.; Abernathy, J.; Juul, S.; Short, V.; Derman, R. Prevalence of iron deficiency in first trimester, nonanemic pregnant women. J. Matern. Neonatal Med. 2021, 34, 1002–1005. [Google Scholar] [CrossRef]
  3. Dee, D.L.; Sharma, A.J.; Cogswell, M.E.; Grummer-Strawn, L.M.; Fein, S.B.; Scanlon, K.S. Sources of Supplemental Iron Among Breastfed Infants During the First Year of Life. Pediatrics 2008, 122, S98–S104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sun, H.; Weaver, C.M. Decreased Iron Intake Parallels Rising Iron Deficiency Anemia and Related Mortality Rates in the US Population. J. Nutr. 2021, 151, 1947–1955. [Google Scholar] [CrossRef]
  5. Youdim, M.B.H.; Green, A.R.; Bloomfield, M.R.; Mitchell, B.D.; Heal, D.J.; Grahame-Smith, D.G. The effects of iron deficiency on brain biogenic monoamine biochemistry and function in rats. Neuropharmacology 1980, 19, 259–267. [Google Scholar] [CrossRef]
  6. Yu, G.S.; Steinkirchner, T.M.; Rao, G.A.; Larkin, E.C. Effect of prenatal iron deficiency on myelination in rat pups. Am. J. Pathol. 1986, 125, 620–624. [Google Scholar] [PubMed]
  7. Zamora, T.G.; Guiang, S.F.; Widness, J.A.; Georgieff, M.K. Iron is prioritized to red blood cells over the brain in phlebotomized anemic newborn lambs. Pediatr. Res. 2016, 79, 922–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Piñero, D.J.; Jones, B.C.; Beard, J.L. Variations in Dietary Iron Alter Behavior in Developing Rats. J. Nutr. 2001, 131, 311–318. [Google Scholar] [CrossRef]
  9. Kwik-Uribe, C.L.; Golub, M.S.; Keen, C.L. Chronic marginal iron intakes during early development in mice alter brain iron concentrations and behavior despite postnatal iron supplementation. J. Nutr. 2000, 130, 2040–2048. [Google Scholar] [CrossRef] [Green Version]
  10. Widdowson, E.M.; Spray, C.M. Chemical Development in utero. Arch. Dis. Child. 1951, 26, 205–214. [Google Scholar] [CrossRef] [Green Version]
  11. Petry, C.D.; Eaton, M.A.; Wobken, J.D.; Mills, M.M.; Johnson, D.E.; Georgieff, M.K. Iron deficiency of liver, heart, and brain in newborn infants of diabetic mothers. J. Pediatr. 1992, 121, 109–114. [Google Scholar] [CrossRef]
  12. MacQueen, B.C.; Christensen, R.D.; Baer, V.L.; Ward, D.M.; Snow, G. Screening umbilical cord blood for congenital Iron deficiency. Blood Cells Mol. Dis. 2019, 77, 95–100. [Google Scholar] [CrossRef] [PubMed]
  13. Ru, Y.; Pressman, E.K.; Guillet, R.; Katzman, P.J.; Bacak, S.J.; O’Brien, K.O. Predictors of anemia and iron status at birth in neonates born to women carrying multiple fetuses. Pediatr. Res. 2018, 84, 199–204. [Google Scholar] [CrossRef] [PubMed]
  14. Rios, E.; Lipschitz, D.A.; Cook, J.D.; Smith, N.J. Relationship of maternal and infant iron stores as assessed by determination of plasma ferritin. Pediatrics 1975, 55, 694–699. [Google Scholar] [PubMed]
  15. Baker, R.D.; Greer, F.R.; The Committee on Nutrition. Diagnosis and Prevention of Iron Deficiency and Iron-Deficiency Anemia in Infants and Young Children (0–3 Years of Age). Pediatrics 2010, 126, 1040–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.; Ganz, T.; Kaplan, J. Hepcidin Regulates Cellular Iron Efflux by Binding to Ferroportin and Inducing Its Internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hentze, M.W.; Muckenthaler, M.U.; Galy, B.; Camaschella, C. Two to Tango: Regulation of Mammalian Iron Metabolism. Cell 2010, 142, 24–38. [Google Scholar] [CrossRef] [Green Version]
  18. Fisher, A.; Nemeth, E. Iron homeostasis during pregnancy. Am. J. Clin. Nutr. 2017, 106, 1567S–1574S. [Google Scholar] [CrossRef] [PubMed]
  19. Tamura, T.; Goldenberg, R.L.; Hou, J.; Johnston, K.E.; Cliver, S.P.; Ramey, S.L.; Nelson, K.G. Cord serum ferritin concentrations and mental and psychomotor development of children at five years of age. J. Pediatr. 2002, 140, 165–170. [Google Scholar] [CrossRef] [PubMed]
  20. Shao, J.; Lou, J.; Rao, R.; Georgieff, M.K.; Kaciroti, N.; Felt, B.T.; Zhao, Z.-Y.; Lozoff, B. Maternal Serum Ferritin Concentration Is Positively Associated with Newborn Iron Stores in Women with Low Ferritin Status in Late Pregnancy. J. Nutr. 2012, 142, 2004–2009. [Google Scholar] [CrossRef]
  21. Dewey, K.G.; Oaks, B.M. U-shaped curve for risk associated with maternal hemoglobin, iron status, or iron supplementation. Am. J. Clin. Nutr. 2017, 106, 1694S–1702S. [Google Scholar] [CrossRef] [Green Version]
  22. Gonzales, G.F.; Steenland, K.; Tapia, V. Maternal hemoglobin level and fetal outcome at low and high altitudes. Am. J. Physiol. Integr. Comp. Physiol. 2009, 297, R1477–R1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Rahmati, S.; Azami, M.; Badfar, G.; Parizad, N.; Sayehmiri, K. The relationship between maternal anemia during pregnancy with preterm birth: A systematic review and meta-analysis. J. Matern. Neonatal Med. 2020, 33, 2679–2689. [Google Scholar] [CrossRef]
  24. Haider, B.A.; Bhutta, Z.A. Multiple-micronutrient supplementation for women during pregnancy. Cochrane Database Syst. Rev. 2017, 4, CD004905. [Google Scholar] [CrossRef] [PubMed]
  25. Wrighting, D.M.; Andrews, N.C. Interleukin-6 induces hepcidin expression through STAT3. Blood 2006, 108, 3204–3209. [Google Scholar] [CrossRef]
  26. Bahr, T.M.; Ward, D.M.; Jia, X.; Ohls, R.K.; German, K.R.; Christensen, R.D. Is the erythropoietin-erythroferrone-hepcidin axis intact in human neonates? Blood Cells Mol. Dis. 2021, 88, 102536. [Google Scholar] [CrossRef] [PubMed]
  27. Berglund, S.; Lönnerdal, B.; Westrup, B.; Domellöf, M. Effects of iron supplementation on serum hepcidin and serum erythropoietin in low-birth-weight infants. Am. J. Clin. Nutr. 2011, 94, 1553–1561. [Google Scholar] [CrossRef] [PubMed]
  28. German, K.R.; Comstock, B.A.; Parikh, P.; Whittington, D.; Mayock, D.E.; Heagerty, P.J.; Bahr, T.M.; Juul, S.E. Do Extremely Low Gestational Age Neonates Regulate Iron Absorption via Hepcidin? J. Pediatr. 2021, SS0022-3476(21). [Google Scholar] [CrossRef]
  29. Sazawal, S.; Black, R.; Ramsan, M.; Chwaya, H.M.; Stoltzfus, R.J.; Dutta, A.; Dhingra, U.; Kabole, I.; Deb, S.; Othman, M.K.; et al. Effects of routine prophylactic supplementation with iron and folic acid on admission to hospital and mortality in preschool children in a high malaria transmission setting: Community-based, randomised, placebo-controlled trial. Lancet 2006, 367, 133–143. [Google Scholar] [CrossRef]
  30. Jonker, F.A.M.; Calis, J.C.J.; Van Hensbroek, M.B.; Phiri, K.; Geskus, R.B.; Brabin, B.; Leenstra, T. Iron Status Predicts Malaria Risk in Malawian Preschool Children. PLoS ONE 2012, 7, e42670. [Google Scholar] [CrossRef] [Green Version]
  31. Lozoff, B.; Castillo, M.; Clark, K.M.; Smith, J.B. Iron-fortified vs. low-iron infant formula: Developmental outcome at 10 years. Arch. Pediatr. Adolesc. Med. 2006, 166, 208–215. [Google Scholar] [CrossRef] [Green Version]
  32. Singhal, A.; Morley, R.; Abbott, R.; Fairweather-Tait, S.; Stephenson, T.; Lucas, A. Clinical safety of iron-fortified formulas. Pediatrics 2000, 105, e38. [Google Scholar] [CrossRef] [Green Version]
  33. Brannon, P.M.; Taylor, C.L. Iron Supplementation during Pregnancy and Infancy: Uncertainties and Implications for Research and Policy. Nutrients 2017, 9, 1327. [Google Scholar] [CrossRef] [Green Version]
  34. American Academy of Pediatrics—Committee on Fetus and Newborn; The American College of Obstetricians and Gynecologists—Committee on Obstetric Practice. Guidelines for Perinatal Care, 7th ed.; American Academy of Pediatrics: Elk Grove Village, IL, USA; The American College of Obstetricians and Gynecologists: Washington, DC, USA, 2012; Available online: http://simponline.it/wp-content/uploads/2014/11/GuidelinesforPerinatalCare.pdf (accessed on 22 October 2021).
  35. American Academy of Pediatrics Committee on Nutrition. Iron. In Pediatric Nutrition: Policy of the American Academy of Pediatrics, 7th ed.; Kleinman, R.E., Greer, F.R., Eds.; American Academy of Pediatrics: Elk Grove Village, IL, USA, 2019. [Google Scholar]
  36. Lapillonne, A.; Bronsky, J.; Campoy, C.; Embleton, N.; Fewtrell, M.; Mis, N.F.; Gerasimidis, K.; Hojsak, I.; Hulst, J.; Indrio, F.; et al. Feeding the Late and Moderately Preterm Infant: A Position Paper of the European Society for Paediatric Gastroenterology, Hepatology and Nutrition Committee on Nutrition. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 259–270. [Google Scholar] [CrossRef]
  37. Cantor, A.G.; Bougatsos, C.; Dana, T.; Blazina, I.; McDonagh, M. Routine Iron Supplementation and Screening for Iron Deficiency Anemia in Pregnancy: A Systematic Review for the U.S. Preventive Services Task Force. Ann. Intern. Med. 2015, 162, 566–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Juul, S.E.; Comstock, B.A.; Wadhawan, R.; Mayock, D.E.; Courtney, S.E.; Robinson, T.; Ahmad, K.A.; Bendel-Stenzel, E.; Baserga, M.; LaGamma, E.F.; et al. A Randomized Trial of Erythropoietin for Neuroprotection in Preterm Infants. N. Engl. J. Med. 2020, 382, 233–243. [Google Scholar] [CrossRef]
  39. German, K.; Vu, P.T.; Grelli, K.N.; Denton, C.; Lee, G.; Juul, S.E. Zinc Protoporphyrin-to-Heme Ratio and Ferritin as Measures of Iron Sufficiency in the Neonatal Intensive Care Unit. J. Pediatr. 2018, 194, 47–53. [Google Scholar] [CrossRef]
  40. German, K.R.; Vu, P.T.; Comstock, B.A.; Ohls, R.K.; Heagerty, P.J.; Mayock, D.E.; Georgieff, M.; Rao, R.; Juul, S.E. Enteral Iron Supplementation in Infants Born Extremely Preterm and its Positive Correlation with Neurodevelopment; Post Hoc Analysis of the Preterm Erythropoietin Neuroprotection Trial Randomized Controlled Trial. J. Pediatr. 2021, S0022-3476(21)00686-7. [Google Scholar] [CrossRef] [PubMed]
  41. Carcillo, J.A.; Sward, K.; Halstead, E.S.; Telford, R.; Jimenez-Bacardi, A.; Shakoory, B.; Simon, D.; Hall, M. A Systemic Inflammation Mortality Risk Assessment Contingency Table for Severe Sepsis. Pediatr. Crit. Care Med. 2017, 18, 143–150. [Google Scholar] [CrossRef]
  42. German, K.R.; Vu, P.T.; Neches, S.; Juul, S.E. Comparison of two markers of iron sufficiency and neurodevelopmental outcomes. Early Hum. Dev. 2021, 158, 105395. [Google Scholar] [CrossRef]
  43. Cheng, C.F.; Zerzan, J.C.; Johnson, D.B.; Juul, S.E. Zinc protoporphyrin-to-heme ratios in high-risk and preterm infants. J. Pediatr. 2012, 161, 81–87. [Google Scholar] [CrossRef] [PubMed]
  44. Yu, K.H. Effectiveness of zinc protoporphyrin/heme ratio for screening iron deficiency in preschool-aged children. Nutr. Res. Pr. 2011, 5, 40–45. [Google Scholar] [CrossRef] [PubMed]
  45. De Waal, C.G.; Uijterschout, L.; Abbink, M.; Boersma, B.; Vos, P.; Rövekamp, W.W.; Hudig, F.; Akkermans, M.D.; Van Goudoever, J.B.; Brus, F. Zinc protoporphyrin/heme ratio as parameter of iron status in moderately preterm infants: Natural course and associations in the first 4 months. J. Perinatol. 2017, 37, 690–694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Rettmer, R.L.; Carlson, T.H.; Origenes, M.L., Jr.; Jack, R.M.; Labbé, R.F. Zinc Protoporphyrin/Heme Ratio for Diagnosis of Preanemic Iron Deficiency. Pediatrics 1999, 104, e37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Miller, S.M. Iron supplementation in premature infants using the zinc protoporphyrin to heme ratio: Short- and long-term outcomes. J. Perinatol. 2013, 33, 712–716. [Google Scholar] [CrossRef] [Green Version]
  48. Chinudomwong, P.; Binyasing, A.; Trongsakul, R.; Paisooksantivatana, K. Diagnostic performance of reticulocyte hemoglobin equivalent in assessing the iron status. J. Clin. Lab. Anal. 2020, 34, e23225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Christensen, R.D.; Henry, E.; Bennett, S.T.; Yaish, H.M. Reference intervals for reticulocyte parameters of infants during their first 90 days after birth. J. Perinatol. 2016, 36, 61–66. [Google Scholar] [CrossRef]
  50. German, K.; Vu, P.T.; Irvine, J.D.; Juul, S.E. Trends in reticulocyte hemoglobin equivalent values in critically ill neonates, stratified by gestational age. J. Perinatol. 2019, 39, 1268–1274. [Google Scholar] [CrossRef]
  51. Franz, A.R.; Engel, C.; Bassler, D.; Rüdiger, M.; Thome, U.H.; Maier, R.F.; Krägeloh-Mann, I.; Kron, M.; Essers, J.; Bührer, C.; et al. Effects of Liberal vs. Restrictive Transfusion Thresholds on Survival and Neurocognitive Outcomes in Extremely Low-Birth-Weight Infants: The ETTNO Randomized Clinical Trial. JAMA 2020, 324, 560–570. [Google Scholar] [CrossRef]
  52. Mireku, M.O.; Davidson, L.L.; Koura, K.G.; Ouédraogo, S.; Boivin, M.J.; Xiong, X.; Accrombessi, M.M.K.; Massougbodji, A.; Cot, M.; Bodeau-Livinec, F. Prenatal Hemoglobin Levels and Early Cognitive and Motor Functions of One-Year-Old Children. Pediatrics 2015, 136, e76–e83. [Google Scholar] [CrossRef] [Green Version]
  53. De Ungria, M.; Rao, R.; Wobken, J.D.; Luciana, M.; Nelson, C.A.; Georgieff, M.K. Perinatal Iron Deficiency Decreases Cytochrome c Oxidase (CytOx) Activity in Selected Regions of Neonatal Rat Brain. Pediatr. Res. 2000, 48, 169–176. [Google Scholar] [CrossRef] [Green Version]
  54. Rao, R.; Tkac, I.; Townsend, E.L.; Gruetter, R.; Georgieff, M.K. Perinatal Iron Deficiency Alters the Neurochemical Profile of the Developing Rat Hippocampus. J. Nutr. 2003, 133, 3215–3221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Jorgenson, L.A.; Wobken, J.D.; Georgieff, M.K. Perinatal iron deficiency alters apical dendritic growth in hippocampall CA1 pyramidal neurons. Dev. Neurosci. 2003, 25, 412–420. [Google Scholar] [CrossRef]
  56. Ben-Shachar, D.; Ashkenazi, R.; Youdim, M.B. Long-term consequence of early iron-deficiency on dopaminergic neurotransmission in rats. Int. J. Dev. Neurosci. 1986, 4, 81–88. [Google Scholar] [CrossRef]
  57. Piñero, D.J.; Li, N.-Q.; Connor, J.R.; Beard, J.L. Variations in dietary iron alter brain iron metabolism in developing rats. J. Nutr. 2000, 130, 254–263. [Google Scholar] [CrossRef] [Green Version]
  58. Brunette, K.E.; Tran, P.V.; Wobken, J.D.; Carlson, E.S.; Georgieff, M.K. Gestational and Neonatal Iron Deficiency Alters Apical Dendrite Structure of CA1 Pyramidal Neurons in Adult Rat Hippocampus. Dev. Neurosci. 2010, 32, 238–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Pisansky, M.T.; Wickham, R.J.; Su, J.; Fretham, S.; Yuan, L.-L.; Sun, M.; Gewirtz, J.C.; Georgieff, M.K. Iron deficiency with or without anemia impairs prepulse inhibition of the startle reflex. Hippocampus 2013, 23, 952–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Sandri, B.J.; Lubach, G.R.; Lock, E.F.; Kling, P.J.; Georgieff, M.K.; Coe, C.L.; Rao, R.B. Correcting iron deficiency anemia with iron dextran alters the serum metabolomic profile of the infant Rhesus Monkey. Am. J. Clin. Nutr. 2021, 113, 915–923. [Google Scholar] [CrossRef]
  61. Amin, S.B.; Orlando, M.; Eddins, A.; MacDonald, M.; Monczynski, C.; Wang, H. In Utero Iron Status and Auditory Neural Maturation in Premature Infants as Evaluated by Auditory Brainstem Response. J. Pediatr. 2010, 156, 377–381. [Google Scholar] [CrossRef] [Green Version]
  62. Berglund, S.K.; Westrup, B.; Haraldsson, E.; Engström, B.; Domellöf, M. Effects of Iron Supplementation on Auditory Brainstem Response in Marginally LBW Infants. Pediatr. Res. 2011, 70, 601–606. [Google Scholar] [CrossRef]
  63. Algarín, C.; Peirano, P.; Garrido, M.; Pizarro, F.; Lozoff, B. Iron Deficiency Anemia in Infancy: Long-Lasting Effects on Auditory and Visual System Functioning. Pediatr. Res. 2003, 53, 217–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wachs, T.D.; Pollitt, E.; Cueto, S.; Jacoby, E.; Creed-Kanashiro, H. Relation of neonatal iron status to individual variability in neonatal temperament. Dev. Psychobiol. 2005, 46, 141–153. [Google Scholar] [CrossRef] [PubMed]
  65. Armony-Sivan, R.; Eidelman, A.I.; Lanir, A.; Sredni, D.; Yehuda, S. Iron Status and Neurobehavioral Development of Premature Infants. J. Perinatol. 2004, 24, 757–762. [Google Scholar] [CrossRef] [Green Version]
  66. Peirano, P.D.; Algarín, C.R.; Garrido, M.I.; Lozoff, B. Iron Deficiency Anemia in Infancy Is Associated with Altered Temporal Organization of Sleep States in Childhood. Pediatr. Res. 2007, 62, 715–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Geng, F.; Mai, X.; Zhan, J.; Xu, L.; Zhao, Z.; Georgieff, M.; Shao, J.; Lozoff, B. Impact of Fetal-Neonatal Iron Deficiency on Recognition Memory at 2 Months of Age. J. Pediatr. 2015, 167, 1226–1232. [Google Scholar] [CrossRef] [Green Version]
  68. Lozoff, B.; De Andraca, I.; Castillo, M.; Smith, J.; Walter, T.; Pino, P. Behavioral and developmental effects of preventing iron-deficiency anemia in healthy full-term infants. Pediatrics 2003, 112, 846–854. [Google Scholar]
  69. Lozoff, B.; Jimenez, E.; Hagen, J.; Mollen, E.; Wolf, A.W. Poorer Behavioral and Developmental Outcome More Than 10 Years After Treatment for Iron Deficiency in Infancy. Pediatrics 2000, 105, e51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. East, P.; Doom, J.R.; Blanco, E.; Burrows, R.; Lozoff, B.; Gahagan, S. Iron deficiency in infancy and neurocognitive and educational outcomes in young adulthood. Dev. Psychol. 2021, 57, 962–975. [Google Scholar] [CrossRef]
  71. Lozoff, B.; Smith, J.; Kaciroti, N.; Clark, K.; Guevara, S.; Jimenez, E. Functional Significance of Early-Life Iron Deficiency: Outcomes at 25 Years. J. Pediatr. 2013, 163, 1260–1266. [Google Scholar] [CrossRef] [Green Version]
  72. Lukowski, A.F.; Koss, M.; Burden, M.J.; Jonides, J.; Nelson, C.A.; Kaciroti, N.; Jimenez, E.; Lozoff, B. Iron deficiency in infancy and neurocognitive functioning at 19 years: Evidence of long-term deficits in executive function and recognition memory. Nutr. Neurosci. 2010, 13, 54–70. [Google Scholar] [CrossRef]
  73. Lozoff, B.; Jimenez, E.; Smith, J.B. Double burden of iron deficiency in infancy and low socioeconomic status: A longitudinal analysis of cognitive test scores to age 19 years. Arch. Pediatrics Adolesc. Med. 2006, 160, 1108–1113. [Google Scholar] [CrossRef] [Green Version]
  74. East, P.; Doom, J.; Blanco, E.; Burrows, R.; Lozoff, B.; Gahagan, S. Young adult outcomes associated with lower cognitive functioning in childhood related to iron-fortified formula in infancy. Nutr. Neurosci. 2020, 2020, 1–10. [Google Scholar] [CrossRef]
  75. Gahagan, S.; Delker, E.; Blanco, E.; Burrows, R.; Lozoff, B. Randomized Controlled Trial of Iron-Fortified versus Low-Iron Infant Formula: Developmental Outcomes at 16 Years. J. Pediatr. 2019, 212, 124–130.e1. [Google Scholar] [CrossRef] [PubMed]
  76. Insel, B.J.; Schaefer, C.A.; McKeague, I.W.; Susser, E.S.; Brown, A.S. Maternal Iron Deficiency and the Risk of Schizophrenia in Offspring. Arch. Gen. Psychiatry 2008, 65, 1136–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Sørensen, H.J.; Nielsen, P.R.; Pedersen, C.B.; Mortensen, P.B. Association Between Prepartum Maternal Iron Deficiency and Offspring Risk of Schizophrenia: Population-Based Cohort Study with Linkage of Danish National Registers. Schizophr. Bull. 2011, 37, 982–987. [Google Scholar] [CrossRef] [PubMed]
  78. Tucker, D.M.; Sandstead, H.H. Spectral electroencephalographic correlates of iron status: Tired blood revisited. Physiol. Behav. 1981, 26, 439–449. [Google Scholar] [CrossRef]
  79. Yan, N.; Zhang, J. Iron Metabolism, Ferroptosis, and the Links with Alzheimer’s Disease. Front. Neurosci. 2020, 13, 1443. [Google Scholar] [CrossRef]
  80. McCarthy, E.K.; Dempsey, E.M.; Kiely, M.E. Iron supplementation in preterm and low-birth-weight infants: A systematic review of intervention studies. Nutr. Rev. 2019, 77, 865–877. [Google Scholar] [CrossRef]
  81. Raffaeli, G.; Manzoni, F.; Cortesi, V.; Cavallaro, G.; Mosca, F.; Ghirardello, S. Iron Homeostasis Disruption and Oxidative Stress in Preterm Newborns. Nutrients 2020, 12, 1554. [Google Scholar] [CrossRef]
  82. Cooke, R.W.I.; Drury, J.A.; Yoxall, C.W.; James, C. Blood transfusion and chronic lung disease in preterm infants. Eur. J. Pediatr. 1997, 156, 47–50. [Google Scholar] [CrossRef] [PubMed]
  83. Hesse, L.; Eberl, W.; Schlaud, M.; Poets, C.F. Blood transfusion. Iron load and retinopathy of prematurity. Eur. J. Pediatr. 1997, 156, 465–470. [Google Scholar] [CrossRef]
  84. Dani, C.; Reali, M.; Bertini, G.; Martelli, E.; Pezzati, M.; Rubaltelli, F. The role of blood transfusions and iron intake on retinopathy of prematurity. Early Hum. Dev. 2001, 62, 57–63. [Google Scholar] [CrossRef]
  85. Owen, J.E.; Bishop, G.M.; Robinson, S.R. Uptake and Toxicity of Hemin and Iron in Cultured Mouse Astrocytes. Neurochem. Res. 2016, 41, 298–306. [Google Scholar] [CrossRef] [PubMed]
  86. Wu, Y.; Song, J.; Wang, Y.; Wang, X.; Culmsee, C.; Zhu, C. The Potential Role of Ferroptosis in Neonatal Brain Injury. Front. Neurosci. 2019, 13, 115. [Google Scholar] [CrossRef] [Green Version]
  87. Lackmann, G.M.; Hesse, L.; Töllner, U. Reduced iron-associated antioxidants in premature newborns suffering intracerebral hemorrhage. Free Radic. Biol. Med. 1996, 20, 407–409. [Google Scholar] [CrossRef]
  88. Braekke, K.; Bechensteen, A.G.; Halvorsen, B.L.; Blomhoff, R.; Haaland, K.; Staff, A.C. Oxidative Stress Markers and Antioxidant Status after Oral Iron Supplementation to Very Low Birth Weight Infants. J. Pediatr. 2007, 151, 23–28. [Google Scholar] [CrossRef] [PubMed]
  89. Friel, J.K.; Andrews, W.L.; Aziz, K.; Kwa, P.G.; Lepage, G.; L’Abbe, M.R. A randomized trial of two levels of iron supplementation and developmental outcome in low birth weight infants. J. Pediatr. 2001, 139, 254–260. [Google Scholar] [CrossRef]
  90. Müller, K.F.; Lorenz, L.; Poets, C.F.; Westerman, M.; Franz, A.R. Hepcidin concentrations in serum and urine correlate with iron homeostasis in preterm infants. J. Pediatr. 2012, 160, 949–953. [Google Scholar] [CrossRef] [PubMed]
  91. Raghuveer, T.S.; McGuire, E.M.; Martin, S.M.; Wagner, B.A.; Rebouché, C.J.; Buettner, G.R.; Widness, J.A. Lactoferrin in the preterm infants’ diet attenuates iron-induced oxidation products. Pediatr. Res. 2002, 52, 964–972. [Google Scholar] [CrossRef]
  92. Amin, S.B.; Myers, G.; Wang, H. Association between neonatal iron overload and early human brain development in premature infants. Early Hum. Dev. 2012, 88, 583–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kaur, D.; Peng, J.; Chinta, S.J.; Rajagopalan, S.; Di Monte, D.A.; Cherny, R.A.; Andersen, J.K. Increased murine neonatal iron intake results in Parkinson-like neurodegeneration with age. Neurobiol. Aging 2007, 28, 907–913. [Google Scholar] [CrossRef]
  94. Gerday, E.; Brereton, J.B.; Bahr, T.M.; Elmont, J.O.; Fullmer, S.; Middleton, B.A.; Ward, D.M.; Ohls, R.K.; Christensen, R.D. Urinary ferritin; A potential noninvasive way to screen NICU patients for iron deficiency. J. Perinatol. 2021, 41, 1419–1425. [Google Scholar] [CrossRef] [PubMed]
  95. Carlson, A.; Xia, K.; Azcarate-Peril, M.A.; Goldman, B.D.; Ahn, M.; Styner, M.A.; Thompson, A.L.; Geng, X.; Gilmore, J.H.; Knickmeyer, R.C. Infant Gut Microbiome Associated with Cognitive Development. Biol. Psychiatry 2018, 83, 148–159. [Google Scholar] [CrossRef] [PubMed]
  96. Henderickx, J.G.E.; Zwittink, R.D.; Van Lingen, R.A.; Knol, J.; Belzer, C. The Preterm Gut Microbiota: An Inconspicuous Challenge in Nutritional Neonatal Care. Front. Cell. Infect. Microbiol. 2019, 9, 85. [Google Scholar] [CrossRef]
  97. Jaeggi, T.; Kortman, G.A.M.; Moretti, D.; Chassard, C.; Holding, P.; Dostal, A.; Boekhorst, J.; Timmerman, H.M.; Swinkels, D.W.; Tjalsma, H.; et al. Iron fortification adversely affects the gut microbiome, increases pathogen abundance and induces intestinal inflammation in Kenyan infants. Gut 2015, 64, 731–742. [Google Scholar] [CrossRef] [PubMed]
  98. Kortman, G.A.M.; Boleij, A.; Swinkels, D.W.; Tjalsma, H. Iron Availability Increases the Pathogenic Potential of Salmonella Typhimurium and Other Enteric Pathogens at the Intestinal Epithelial Interface. PLoS ONE 2012, 7, e29968. [Google Scholar] [CrossRef] [PubMed]
  99. Paganini, D.; Zimmermann, M.B. The effects of iron fortification and supplementation on the gut microbiome and diarrhea in infants and children: A review. Am. J. Clin. Nutr. 2017, 106, 1688S–1693S. [Google Scholar] [CrossRef] [Green Version]
  100. Mahalhal, A.; Williams, J.M.; Johnson, S.; Ellaby, N.; Duckworth, C.A.; Burkitt, M.D.; Liu, X.; Hold, G.L.; Campbell, B.J.; Pritchard, D.M.; et al. Oral iron exacerbates colitis and influences the intestinal microbiome. PLoS ONE 2018, 13, e0202460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Commonly used iron measures in neonates.
Table 1. Commonly used iron measures in neonates.
Iron MeasureDescriptionAdvantagesDisadvantagesAssociation with Neurodevelopment
FerritinStorage form of ironMost widely studied marker of iron status in the neonatal population.
Low ferritin values are a reliable marker of iron status.
Elevated in the setting of inflammation and hepatocellular injury [41].The association between ferritin values and neurodevelopmental outcomes has been studied [19].
Low ferritin values are associated with worse outcomes, but high values may be less reliable due to inflammatory effects [42].
Zinc Protoporphyrin-to-Heme Ratio (ZnPP/H)Zinc or iron can be incorporated into the protoporphyrin ring in the production of heme. When less iron is available, the proportion of zinc incorporated increases, thus raising the ZnPP/H ratio.Less affected by inflammation [39].
Has been evaluated in neonatal and childhood population [43,44,45,46].
ZnPP/H values in transfused adult red blood cells may dilute neonatal values in infants who have received transfusions.
Reflect values in the red blood cell pool as a whole, and therefore acute changes in iron status may not be reflected until older red blood cells are broken down [47].
May be better correlated with neurodevelopmental outcomes than ferritin, though studies are limited [42].
Reticulocyte Hemoglobin Equivalent (Ret HE)The hemoglobin content of reticulocyte cells.As it reflects newly formed cells, it may be more responsive to recent changes in iron status.
Adult studies suggest reliable with inflammation [48].Can be measured in conjunction with a complete blood cell count (CBC) on many CBC analyzers, thus limiting phlebotomy.
Although some studies have begun to establish normative values in the neonatal population, there are limited studies addressing its correlation with long-term outcomes and therefore target values are still unclear [49,50].Limited data examining correlation with neurodevelopmental outcomes.
Hemoglobin/HematocritThe majority of iron in the body is present in red blood cells.Readily available and low cost. As iron is prioritized for erythropoiesis over all other needs, anemia is a late marker of iron deficiency and adverse impacts such as brain iron deficiency may occur in the absence of anemia.Studies examining correlation with outcomes may reflect iron deficiency status or non-iron factors such as transfusion thresholds [51,52].
Transferrin SaturationTransferrin is a protein that transports iron around the body. The transferrin saturation reflects the percentage of transferrin sites that are occupied by iron.Can be calculated from TIBC and serum iron.Normative values not well defined in neonates.Limited data examining correlation with neurodevelopmental outcomes.
Total Iron-Binding Capacity (TIBC)A measure of the level of transferrin in circulation.Commonly used measure in adults.Normative values not well defined in neonates.Limited data examining correlation with neurodevelopmental outcomes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

German, K.R.; Juul, S.E. Iron and Neurodevelopment in Preterm Infants: A Narrative Review. Nutrients 2021, 13, 3737. https://doi.org/10.3390/nu13113737

AMA Style

German KR, Juul SE. Iron and Neurodevelopment in Preterm Infants: A Narrative Review. Nutrients. 2021; 13(11):3737. https://doi.org/10.3390/nu13113737

Chicago/Turabian Style

German, Kendell R., and Sandra E. Juul. 2021. "Iron and Neurodevelopment in Preterm Infants: A Narrative Review" Nutrients 13, no. 11: 3737. https://doi.org/10.3390/nu13113737

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop