Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 820 word(s) 820 2021-11-02 04:12:51 |
2 format correct Meta information modification 820 2021-12-21 02:08:03 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tran, P.V. Iron Deficiency Anemia. Encyclopedia. Available online: https://encyclopedia.pub/entry/17352 (accessed on 26 July 2024).
Tran PV. Iron Deficiency Anemia. Encyclopedia. Available at: https://encyclopedia.pub/entry/17352. Accessed July 26, 2024.
Tran, Phu V. "Iron Deficiency Anemia" Encyclopedia, https://encyclopedia.pub/entry/17352 (accessed July 26, 2024).
Tran, P.V. (2021, December 20). Iron Deficiency Anemia. In Encyclopedia. https://encyclopedia.pub/entry/17352
Tran, Phu V. "Iron Deficiency Anemia." Encyclopedia. Web. 20 December, 2021.
Iron Deficiency Anemia
Edit

Iron deficiency (ID) anemia is the foremost micronutrient deficiency worldwide, affecting around 40% of pregnant women and young children. ID during the prenatal and early postnatal periods has a pronounced effect on neurodevelopment, resulting in long-term effects such as cognitive impairment and increased risk for neuropsychiatric disorders. Treatment of ID has been complicated as it does not always resolve the long-lasting neurodevelopmental deficits. A better understanding of the underlying molecular mechanisms is needed in order to develop more effective treatments.

perinatal iron deficiency cognition neuroscience epigenetics micronutrients

1. Introduction

Iron deficiency (ID), which affects 40–50% of pregnant women and preschool-aged children and is the most common micronutrient deficiency worldwide [1][2]. Given the prevalence of this early-life nutritional exposure, it is important to understand its effect on long-term health outcomes, and the biological basis underlying these effects. ID during the fetal and early childhood periods has a significant effect on neurodevelopment, resulting in cognitive, socio-emotional, and learning and memory deficits that last into early adulthood [3][4]. ID also carries long-term health risks, including increased risk for neuropsychiatric disorders such as autism and schizophrenia [5][6]. Parallel studies in pre-clinical models have shown that early-life ID results in abnormal hippocampal structure, function, and gene expression acutely during the period of rapid neurodevelopment and persistently into adulthood despite prompt iron therapy after diagnosis [7][8][9][10][11][12][13][14][15][16]. The persistent gene dysregulation likely drives the adult neurobehavioral abnormalities of early-life or developmental ID. However, specific iron-dependent mechanisms by which early-life ID alters gene expression across the lifespan are unknown.

2. Early-Life Iron Deficiency Modifies Gene Regulation and Epigenetic Landscape in the Adult Rat Hippocampus

Understanding the biology behind the association of maternal and early postnatal ID with neurocognitive impairments and psychopathology risks is a prerequisite for developing effective prevention and treatment strategies. Based on a series of observational studies, two hypotheses are formulated to explicate the poor long-term neurodevelopmental outcomes. The structural defects hypothesis posits that developmental ID causes abnormalities ranging from gross structures (e.g., brain and white matter volumes) to fine ultrastructures (e.g., dendrite branching and synaptic spines) that persist despite later iron reconstitution [11][13][17]. The gene dysregulation hypothesis postulates that early-life ID reprograms gene regulation, which in turn contributes to subsequent phenotypic changes [16][18][19][20][21]. These two hypotheses are not mutually exclusive, and their interactions likely drive abnormal structure during neurodevelopment and function throughout the lifetime, accounting for the risk of psychopathology in later life. The structural hypothesis is discussed elsewhere [22]. This review focuses on the gene dysregulation hypothesis and the potential underlying mechanisms.

3. Early-Life Iron Deficiency Reprograms Gene Regulation

Extensive gene dysregulation has been demonstrated in both rodent and porcine models of fetal-neonatal ID acutely during ID [8][23] and persistently in adulthood following iron repletion [16][21]. The dysregulated genes implicate abnormal neurodevelopment and increased propensity of neuropsychiatric disorders [8][16][21][23]. These widespread and lasting effects implicate global and stable changes in gene regulatory mechanisms such as epigenetic regulation. Epigenetic regulation refers to covalent modifications of DNA and histones to alter gene transcriptional activity and phenotype without changes in the genetic code. Importantly, DNA and histone modifications can be altered depending on environmental exposures such as stress [24][25], toxicants [26][27][28], and nutrients [29][18][19][20][30][31]. Thus, epigenetic regulation can be a mechanism by which ID alters gene regulation during critical windows of the nervous system development, contributing to poor long-term neuropsychiatric outcomes.

4. Prenatal Choline Supplementation and Iron Deficiency Interact to Regulate the Rat Hippocampal Epigenomic Landscape

Given the beneficial effects of prenatal choline supplementation in reversing the repression and epigenetic modifications of the hippocampal Bdnf gene, an important mediator of neuronal differentiation and plasticity, by early-life ID [29], it is enticing to use choline as a prevention (prenatal period) or treatment (children diagnosed with ID anemia) in clinical studies [32]. However, there are several caveats to the use of such a potential epigenetic modulator in the treatment of ID. First, given multiple epigenetic modifications by ID, the therapeutic efficacy of modulating any single modification is unclear. Our recent genome-wide analysis reveals that prenatal choline supplementation produces a distinct epigenomic effect from early-life ID, where choline modifies specifically histone H3K9me3 landscape. Second, the efficacy of epigenetic regulators has to be considered in the context of critical periods of neurodevelopment. For example, once the process of dendrite outgrowth is complete, altering gene expression by treatment with an epigenetic modulator cannot reverse the structural damage caused by ID since the critical period is closed. Therefore, appropriate timing of choline use in the context of neurodevelopment is crucial for its efficacy. Since the brain is not developmentally homogeneous, certain brain regions may be affected more or less than others depending on whether they are or are not in a rapid developmental period. Finally, potential adverse effects of inappropriate use of epigenetic modulators such as choline remain possible. We found little and even negative effects in our control iron-sufficient rats with prenatal supplementation from gestational day 11 through 18 [16][33].  These observations indicate the need to continue to study appropriate choline dosing, timing, and duration of supplementation to avoid potential long-term adverse consequences.

References

  1. McLean, E.; Cogswell, M.; Egli, I.; Wojdyla, D.; De Benoist, B. Worldwide prevalence of anaemia, WHO Vitamin and Mineral Nutrition Information System, 1993-2005. Public Health Nutr. 2009, 12, 444–454.
  2. Stevens, G.A.; Finucane, M.M.; De-Regil, L.M.; Paciorek, C.J.; Flaxman, S.R.; Branca, F.; Peña-Rosas, J.P.; Bhutta, Z.A.; Ezzati, M. Global, regional, and national trends in haemoglobin concentration and prevalence of total and severe anaemia in children and pregnant and non-pregnant women for 1995-2011: A systematic analysis of population-representative data. Lancet Glob. Health 2013, 1, e16–e25.
  3. Lozoff, B.; Smith, J.B.; Kaciroti, N.; Clark, K.M.; Guevara, S.; Jimenez, E. Functional significance of early-life iron deficiency: Outcomes at 25 years. J. Pediatr. 2013, 163, 1260–1266.
  4. 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.
  5. 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.
  6. Schmidt, R.J.; Tancredi, D.J.; Krakowiak, P.; Hansen, R.L.; Ozonoff, S. Maternal intake of supplemental iron and risk of autism spectrum disorder. Am. J. Epidemiol. 2014, 180, 890–900.
  7. 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.
  8. Carlson, E.S.; Stead, J.D.H.; Neal, C.R.; Petryk, A.; Georgieff, M.K. Perinatal iron deficiency results in altered developmental expression of genes mediating energy metabolism and neuronal morphogenesis in hippocampus. Hippocampus 2007, 17, 679–691.
  9. Carlson, E.S.; Tkac, I.; Magid, R.; O’Connor, M.B.; Andrews, N.C.; Schallert, T.; Gunshin, H.; Georgieff, M.K.; Petryk, A. Iron Is Essential for Neuron Development and Memory Function in Mouse Hippocampus. J. Nutr. 2009, 139, 672–679.
  10. Fretham, S.J.B.; Carlson, E.S.; Georgieff, M.K. Neuronal-specific iron deficiency dysregulates mammalian target of rapamycin signaling during hippocampal development in nonanemic genetic mouse Models1,2. J. Nutr. 2013, 143, 260–266.
  11. Fretham, S.J.B.; Carlson, E.S.; Wobken, J.; Tran, P.V.; Petryk, A.; Georgieff, M.K. Temporal manipulation of transferrin-receptor-1-dependent iron uptake identifies a sensitive period in mouse hippocampal neurodevelopment. Hippocampus 2012, 22, 1691–1702.
  12. Jorgenson, L.A.; Sun, M.; O’Connor, M.; Georgieff, M.K. Fetal iron deficiency disrupts the maturation of synaptic function and efficacy in area CA1 of the developing rat hippocampus. Hippocampus 2005, 15, 1094–1102.
  13. Jorgenson, L.A.; Wobken, J.D.; Georgieff, M.K. Perinatal Iron Deficiency Alters Apical Dendritic Growth in Hippocampal CA1 Pyramidal Neurons. Dev. Neurosci. 2003, 25, 412–420.
  14. Tran, P.V.; Carlson, E.S.; Fretham, S.J.B.; Georgieff, M.K. Early-Life Iron Deficiency Anemia Alters Neurotrophic Factor Expression and Hippocampal Neuron Differentiation in Male Rats. J. Nutr. 2008, 138, 2495.
  15. Tran, P.V.; Fretham, S.J.B.; Carlson, E.S.; Georgieff, M.K. Long-term reduction of hippocampal brain-derived neurotrophic factor activity after fetal-neonatal iron deficiency in adult rats. Pediatr. Res. 2009, 65, 493–498.
  16. Tran, P.V.; Kennedy, B.C.; Pisansky, M.T.; Won, K.-J.; Gewirtz, J.C.; Simmons, R.A.; Georgieff, M.K. Prenatal Choline Supplementation Diminishes Early-Life Iron Deficiency–Induced Reprogramming of Molecular Networks Associated with Behavioral Abnormalities in the Adult Rat Hippocampus. J. Nutr. 2016, 146, 484–493.
  17. Hensch, T.K. Critical period regulation. Annu. Rev. Neurosci. 2004, 27, 549–579.
  18. Tyagi, E.; Zhuang, Y.; Agrawal, R.; Ying, Z.; Gomez-Pinilla, F. Interactive actions of Bdnf methylation and cell metabolism for building neural resilience under the influence of diet. Neurobiol. Dis. 2015, 73, 307–318.
  19. Zeisel, S. Choline, Other Methyl-Donors and Epigenetics. Nutrients 2017, 9, 445.
  20. Ly, A.; Ishiguro, L.; Kim, D.; Im, D.; Kim, S.E.; Sohn, K.J.; Croxford, R.; Kim, Y.I. Maternal folic acid supplementation modulates DNA methylation and gene expression in the rat offspring in a gestation period-dependent and organ-specific manner. J. Nutr. Biochem. 2016, 33, 103–110.
  21. Barks, A.; Fretham, S.J.B.; Georgieff, M.K.; Tran, P.V. Early-Life Neuronal-Specific Iron Deficiency Alters the Adult Mouse Hippocampal Transcriptome. J. Nutr. 2018, 148, 1521–1528.
  22. Barks, A.; Hall, A.M.; Tran, P.V.; Georgieff, M.K. Iron as a model nutrient for understanding the nutritional origins of neuropsychiatric disease. Pediatr. Res. 2019, 85, 176–182.
  23. Schachtschneider, K.M.; Liu, Y.; Rund, L.A.; Madsen, O.; Johnson, R.W.; Groenen, M.A.M.; Schook, L.B. Impact of neonatal iron deficiency on hippocampal DNA methylation and gene transcription in a porcine biomedical model of cognitive development. BMC Genomics 2016, 17, 1–14.
  24. Lubin, F.D.; Roth, T.L.; Sweatt, J.D. Epigenetic Regulation of bdnf Gene Transcription in the Consolidation of Fear Memory. J. Neurosci. 2008, 28, 10576–10586.
  25. McGowan, P.O.; Suderman, M.; Sasaki, A.; Huang, T.C.T.; Hallett, M.; Meaney, M.J.; Szyf, M. Broad Epigenetic Signature of Maternal Care in the Brain of Adult Rats. PLoS ONE 2011, 6, e14739.
  26. Anderson, O.S.; Nahar, M.S.; Faulk, C.; Jones, T.R.; Liao, C.; Kannan, K.; Weinhouse, C.; Rozek, L.S.; Dolinoy, D.C. Epigenetic responses following maternal dietary exposure to physiologically relevant levels of bisphenol A. Environ. Mol. Mutagen. 2012, 53, 334–342.
  27. McBirney, M.; King, S.E.; Pappalardo, M.; Houser, E.; Unkefer, M.; Nilsson, E.; Sadler-Riggleman, I.; Beck, D.; Winchester, P.; Skinner, M.K. Atrazine induced epigenetic transgenerational inheritance of disease, lean phenotype and sperm epimutation pathology biomarkers. PLoS ONE 2017, 12, e0184306.
  28. Perkins, A.; Lehmann, C.; Lawrence, R.C.; Kelly, S.J. Alcohol exposure during development: Impact on the epigenome. Int. J. Dev. Neurosci. 2013, 31, 391–397.
  29. Tran, P.V.; Kennedy, B.C.; Lien, Y.-C.; Simmons, R.A.; Georgieff, M.K. Fetal iron deficiency induces chromatin remodeling at the Bdnf locus in adult rat hippocampus. Am. J. Physiol. Integr. Comp. Physiol. 2015, 308, R276–R282.
  30. Ke, X.; Xing, B.; Yu, B.; Yu, X.; Majnik, A.; Cohen, S.; Lane, R.; Joss-Moore, L. IUGR disrupts the PPARγ-Setd8-H4K20me1 and Wnt signaling pathways in the juvenile rat hippocampus. Int. J. Dev. Neurosci. 2014, 38, 59–67.
  31. Waterland, R.A.; Jirtle, R.L. Transposable Elements: Targets for Early Nutritional Effects on Epigenetic Gene Regulation. Mol. Cell. Biol. 2003, 23, 5293–5300.
  32. Georgieff, M.K.; Ramel, S.E.; Cusick, S.E. Nutritional influences on brain development. Acta Paediatr. 2018, 107, 1310–1321.
  33. Kennedy, B.C.; Dimova, J.G.; Siddappa, A.J.M.; Tran, P.V.; Gewirtz, J.C.; Georgieff, M.K. Prenatal choline supplementation ameliorates the long-term neurobehavioral effects of fetal-neonatal iron deficiency in rats. J. Nutr. 2014, 144, 1858–1865.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 450
Entry Collection: Environmental Sciences
Revisions: 2 times (View History)
Update Date: 21 Dec 2021
1000/1000
Video Production Service