1
|
Duarte-Silva E, Maes M, Alves Peixoto C. Iron metabolism dysfunction in neuropsychiatric disorders: Implications for therapeutic intervention. Behav Brain Res 2025; 479:115343. [PMID: 39557130 DOI: 10.1016/j.bbr.2024.115343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Iron is a trace metal that takes part in the maintenance of body homeostasis by, for instance, aiding in energy production and immunity. A body of evidence now demonstrates that dysfunction in iron metabolism can have detrimental effects and is intricately associated with the development of neuropsychiatric disorders, including Major Depressive Disorder (MDD), anxiety, and schizophrenia. For instance, changes in serum and central nervous system (CNS) levels of iron and in proteins mediating iron metabolism have been documented in patients grappling with the aforementioned diseases. By contrast, targeting iron metabolism by using iron chelators, for instance, has proven to be effective in alleviating disease burden. Therefore, here we review the state-of-the-art regarding the role of iron metabolism and its dysfunction in the context of neuropsychiatric disorders. Furthermore, we discuss how targeting iron metabolism can be an effective therapeutic option to tackle this class of diseases. Finally, we discuss the mechanisms linking this dysfunction to behavioral changes in these disorders. Harnessing the knowledge of iron metabolism is not only key to the characterization of novel molecular targets and disease biomarkers but also crucial to drug repurposing and drug design.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, Department of Pharmacology, University of São Paulo, São Paulo, Brazil; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Ribeirão Preto, SP, Brazil.
| | - Michael Maes
- Mental Health Center, University of Electronic Science and Technology of China, Chengdu 611731, China; Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia; Research Institute, Medical University of Plovdiv, Plovdiv 4002, Bulgaria; Department of Psychiatry, Medical University of Plovdiv, Plovdiv 4002, Bulgaria; Cognitive Impairment and Dementia Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), PE, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Burkhart A, Johnsen KB, Skjørringe T, Nielsen AH, Routhe LJ, Hertz S, Møller LB, Thomsen LL, Moos T. Normalization of Fetal Cerebral and Hepatic Iron by Parental Iron Therapy to Pregnant Rats with Systemic Iron Deficiency without Anemia. Nutrients 2024; 16:3264. [PMID: 39408231 PMCID: PMC11479134 DOI: 10.3390/nu16193264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Iron (Fe) is a co-factor for enzymes of the developing brain necessitating sufficient supply. We investigated the effects of administering ferric derisomaltose/Fe isomaltoside (FDI) subcutaneously to Fe-deficient (ID) pregnant rats on cerebral and hepatic concentrations of essential metals and the expression of iron-relevant genes. METHODS Pregnant rats subjected to ID were injected with FDI on the day of mating (E0), 14 days into pregnancy (E14), or the day of birth (postnatal (P0)). The efficacy was evaluated by determination of cerebral and hepatic Fe, copper (Cu), and zinc (Zn) and gene expression of ferroportin, hepcidin, and ferritin H + L in pups on P0 and as adults on P70. RESULTS Females fed an ID diet (5.2 mg/kg Fe) had offspring with significantly lower cerebral and hepatic Fe compared to female controls fed a standard diet (158 mg/kg Fe). Cerebral Cu increased irrespective of supplying a standard diet or administering FDI combined with the standard diet. Hepatic hepcidin mRNA was significantly lower following ID. Cerebral hepcidin mRNA was hardly detectable irrespective of iron status. CONCLUSIONS In conclusion, administering FDI subcutaneously to ID pregnant rats on E0 normalizes fetal cerebral and hepatic Fe. When applied at later gestational ages, supplementation with additional Fe to the offspring is needed to normalize cerebral and hepatic Fe.
Collapse
Affiliation(s)
- Annette Burkhart
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (A.B.); (T.S.); (L.J.R.)
| | - Kasper Bendix Johnsen
- Section of Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark;
| | - Tina Skjørringe
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (A.B.); (T.S.); (L.J.R.)
| | - Asbjørn Haaning Nielsen
- Division of Water and Soil, Department of the Built Environment, Aalborg University, 9220 Aalborg, Denmark;
| | - Lisa Juul Routhe
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (A.B.); (T.S.); (L.J.R.)
| | - Sandra Hertz
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (A.B.); (T.S.); (L.J.R.)
| | - Lisbeth Birk Møller
- Center for Applied Human Genetics, Kennedy Center, Copenhagen University Hospital, 2600 Glostrup, Denmark;
| | | | - Torben Moos
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (A.B.); (T.S.); (L.J.R.)
| |
Collapse
|
3
|
Rudy MJ, Salois G, Cubello J, Newell R, Mayer-Proschel M. Gestational iron deficiency affects the ratio between interneuron subtypes in the postnatal cerebral cortex in mice. Development 2023; 150:dev201068. [PMID: 36805633 PMCID: PMC10110419 DOI: 10.1242/dev.201068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023]
Abstract
Gestational iron deficiency (gID) is highly prevalent and associated with an increased risk of intellectual and developmental disabilities in affected individuals that are often defined by a disrupted balance of excitation and inhibition (E/I) in the brain. Using a nutritional mouse model of gID, we previously demonstrated a shift in the E/I balance towards increased inhibition in the brains of gID offspring that was refractory to postnatal iron supplementation. We thus tested whether gID affects embryonic progenitor cells that are fated towards inhibitory interneurons. We quantified relevant cell populations during embryonic inhibitory neuron specification and found an increase in the proliferation of Nkx2.1+ interneuron progenitors in the embryonic medial ganglionic eminence at E14 that was associated with increased Shh signaling in gID animals at E12. When we quantified the number of mature inhibitory interneurons that are known to originate from the MGE, we found a persistent disruption of differentiated interneuron subtypes in early adulthood. Our data identify a cellular target that links gID with a disruption of cortical interneurons which play a major role in the establishment of the E/I balance.
Collapse
Affiliation(s)
- Michael J. Rudy
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
- Department of Neurology, University of Colorado Denver – Anschutz Medical Campus, 13001 East 17th Place, Aurora, CO 80045, USA
| | - Garrick Salois
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Janine Cubello
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Robert Newell
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Margot Mayer-Proschel
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| |
Collapse
|
4
|
Sandri BJ, Ennis-Czerniak K, Kanajam P, Frey WH, Lock EF, Rao RB. Intranasal insulin treatment partially corrects the altered gene expression profile in the hippocampus of developing rats with perinatal iron deficiency. Am J Physiol Regul Integr Comp Physiol 2023; 325:R423-R432. [PMID: 37602386 PMCID: PMC10639019 DOI: 10.1152/ajpregu.00311.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/17/2023] [Accepted: 08/09/2023] [Indexed: 08/22/2023]
Abstract
Perinatal iron deficiency (FeD) targets the hippocampus and leads to long-term cognitive deficits. Intranasal insulin administration improves cognitive deficits in adult humans with Alzheimer's disease and type 2 diabetes and could provide benefits in FeD-induced hippocampal dysfunction. To objective was to assess the effects of intranasal insulin administration intranasal insulin administration on the hippocampal transcriptome in a developing rat model of perinatal FeD. Perinatal FeD was induced using low-iron diet from gestational day 3 until postnatal day (P) 7, followed by an iron sufficient (FeS) diet through P21. Intranasal insulin was administered at a dose of 0.3 IU twice daily from P8 to P21. Hippocampi were removed on P21 from FeS control, FeD control, FeS insulin, and FeD insulin groups. Total RNA was isolated and profiled using next-generation sequencing. Gene expression profiles were characterized using custom workflows and expression patterns examined using ingenuity pathways analysis (n = 7-9 per group). Select RNAseq results were confirmed via qPCR. Transcriptomic profiling revealed that mitochondrial biogenesis and flux, oxidative phosphorylation, quantity of neurons, CREB signaling in neurons, and RICTOR-based mTOR signaling were disrupted with FeD and positively affected by intranasal insulin treatment with the most benefit observed in the FeD insulin group. Both perinatal FeD and intranasal insulin administration altered gene expression profile in the developing hippocampus. Intranasal insulin treatment reversed the adverse effects of FeD on many molecular pathways and could be explored as an adjunct therapy in perinatal FeD.
Collapse
Affiliation(s)
- Brian J Sandri
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, Minnesota, United States
| | - Kathleen Ennis-Czerniak
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States
| | - Priya Kanajam
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States
| | - William H Frey
- HealthPartners Center for Memory and Aging, HealthPartners Neurosciences, St. Paul, Minnesota, United States
| | - Eric F Lock
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Raghavendra B Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
5
|
Satrom KM, Rao RB, Tkáč I. Neonatal hyperbilirubinemia differentially alters the neurochemical profiles of the developing cerebellum and hippocampus in a preterm Gunn rat model. NMR IN BIOMEDICINE 2023; 36:e4946. [PMID: 37009906 PMCID: PMC11809466 DOI: 10.1002/nbm.4946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 06/19/2023]
Abstract
Neonatal hyperbilirubinemia (NHB) can lead to brain injury in newborn infants by affecting specific regions including the cerebellum and hippocampus. Extremely preterm infants are more vulnerable to bilirubin neurotoxicity, but the mechanism and extent of injury is not well understood. A preterm version of the Gunn rat model was utilized to investigate severe preterm NHB. Homozygous/jaundiced Gunn rat pups were injected (i.p.) on postnatal day (P) 5 with sulfadimethoxine, which increases serum free bilirubin capable of crossing the blood-brain barrier and causing brain injury. The neurochemical profiles of the cerebellum and hippocampus were determined using in vivo 1 H MRS at 9.4 T on P30 and compared with those of heterozygous/non-jaundiced control rats. Transcript expression of related genes was determined by real-time quantitative PCR. MRI revealed significant morphological changes in the cerebellum of jaundiced rats. The concentrations of myo-inositol (+54%), glucose (+51%), N-acetylaspartylglutamate (+21%), and the sum of glycerophosphocholine and phosphocholine (+17%) were significantly higher in the cerebellum of the jaundiced group compared with the control group. Despite the lack of morphologic changes in the hippocampus, the concentration of myo-inositol (+9%) was higher and the concentrations of creatine (-8%) and of total creatine (-3%) were lower in the jaundiced group. In the hippocampus, expression of calcium/calmodulin dependent protein kinase II alpha (Camk2a), glucose transporter 1 (Glut1), and Glut3 transcripts were downregulated in the jaundiced group. In the cerebellum, glial fibrillary acidic protein (Gfap), myelin basic protein (Mbp), and Glut1 transcript expression was upregulated in the jaundiced group. These results indicate osmotic imbalance, gliosis, and changes in energy utilization and myelination, and demonstrate that preterm NHB critically affects brain development in a region-specific manner, with the cerebellum more severely impacted than the hippocampus.
Collapse
Affiliation(s)
- Katherine M Satrom
- Department of Pediatrics, Division of Neonatology, University of Minnesota, Minneapolis, MN, USA
| | - Raghavendra B Rao
- Department of Pediatrics, Division of Neonatology, University of Minnesota, Minneapolis, MN, USA
| | - Ivan Tkáč
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
6
|
Abstract
Growing evidence indicates that a suboptimal intrauterine environment confers risk for schizophrenia. The developmental model of schizophrenia posits that aberrant brain growth during early brain development and adolescence may interact to contribute to this psychiatric disease in adulthood. Although a variety of factors may perturb the environment of the developing fetus and predispose for schizophrenia later, a common mechanism has yet to be elucidated. Micronutrient deficiencies during the perinatal period are known to induce potent effects on brain development by altering neurodevelopmental processes. Iron is an important candidate nutrient to consider because of its role in energy metabolism, monoamine synthesis, synaptogenesis, myelination, and the high prevalence of iron deficiency (ID) in the mother-infant dyad. Understanding the current state of science regarding perinatal ID as an early risk factor for schizophrenia is imperative to inform empirical work investigating the etiology of schizophrenia and develop prevention and intervention programs. In this narrative review, we focus on perinatal ID as a common mechanism underlying the fetal programming of schizophrenia. First, we review the neural aberrations associated with perinatal ID that indicate risk for schizophrenia in adulthood, including disruptions in dopaminergic neurotransmission, hippocampal-dependent learning and memory, and sensorimotor gating. Second, we review the pathophysiology of perinatal ID as a function of maternal ID during pregnancy and use epidemiological and cohort studies to link perinatal ID with risk of schizophrenia. Finally, we review potential confounding phenotypes, including nonanemic causes of perinatal brain ID and future risk of schizophrenia.
Collapse
Affiliation(s)
- Andrea M. Maxwell
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN 55455 (USA)
| | - Raghavendra B. Rao
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55455 (USA)
- Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN 55455 (USA)
| |
Collapse
|
7
|
McWilliams S, Singh I, Leung W, Stockler S, Ipsiroglu OS. Iron deficiency and common neurodevelopmental disorders-A scoping review. PLoS One 2022; 17:e0273819. [PMID: 36173945 PMCID: PMC9522276 DOI: 10.1371/journal.pone.0273819] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/16/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND A wealth of human and experimental studies document a causal and aggravating role of iron deficiency in neurodevelopmental disorders. While pre-, peri-, and early postnatal iron deficiency sets the stage for the risk of developing neurodevelopmental disorders, iron deficiency acquired at later ages aggravates pre-existing neurodevelopmental disorders. Yet, the association of iron deficiency and neurodevelopmental disorders in childhood and adolescence has not yet been explored comprehensively. In this scoping review, we investigate 1) the association of iron deficiency in children and adolescents with the most frequent neurodevelopmental disorders, ADHD, ASD, and FASD, and 2) whether iron supplementation improves outcomes in these disorders. METHOD Scoping review of studies published between 1994 and 2021 using "iron deficiency / iron deficiency anemia" AND "ADHD" OR "autism" OR "FASD" in four biomedical databases. The main inclusion criterion was that articles needed to have quantitative determination of iron status at any postnatal age with primary iron markers such as serum ferritin being reported in association with ADHD, ASD, or FASD. RESULTS For ADHD, 22/30 studies and 4/4 systematic reviews showed an association of ADHD occurrence or severity with iron deficiency; 6/6 treatment studies including 2 randomized controlled trials demonstrated positive effects of iron supplementation. For ASD, 3/6 studies showed an association with iron deficiency, while 3/6 and 1/1 systematic literature review did not; 4 studies showed a variety of prevalence rates of iron deficiency in ASD populations; 1 randomized controlled trial found no positive effect of iron supplementation on behavioural symptoms of ASD. For FASD, 2/2 studies showed an association of iron deficiency with growth retardation in infants and children with prenatal alcohol exposure. CONCLUSION Evidence in favor of screening for iron deficiency and using iron supplementation for pediatric neurodevelopmental disorders comes primarily from ADHD studies and needs to be further investigated for ASD and FASD. Further analysis of study methodologies employed and populations investigated is needed to compare studies against each other and further substantiate the evidence created.
Collapse
Affiliation(s)
- Scout McWilliams
- H-Behaviours Research Lab (previously Sleep/Wake-Behaviour Research Lab), BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Ishmeet Singh
- H-Behaviours Research Lab (previously Sleep/Wake-Behaviour Research Lab), BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Wayne Leung
- H-Behaviours Research Lab (previously Sleep/Wake-Behaviour Research Lab), BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Sylvia Stockler
- H-Behaviours Research Lab (previously Sleep/Wake-Behaviour Research Lab), BC Children’s Hospital Research Institute, Vancouver, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- Division of Biochemical Diseases, Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Osman S. Ipsiroglu
- H-Behaviours Research Lab (previously Sleep/Wake-Behaviour Research Lab), BC Children’s Hospital Research Institute, Vancouver, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- Divisions of Child & Adolescent Psychiatry, Developmental Pediatrics and Respirology, Department of Pediatrics, Sleep/Wake-Behaviour Clinic at Sleep Program, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
8
|
Sandri BJ, Kim J, Lubach GR, Lock EF, Guerrero C, Higgins L, Markowski TW, Kling PJ, Georgieff MK, Coe CL, Rao RB. Multiomic profiling of iron-deficient infant monkeys reveals alterations in neurologically important biochemicals in serum and cerebrospinal fluid before the onset of anemia. Am J Physiol Regul Integr Comp Physiol 2022; 322:R486-R500. [PMID: 35271351 PMCID: PMC9054343 DOI: 10.1152/ajpregu.00235.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/17/2022]
Abstract
The effects of iron deficiency (ID) during infancy extend beyond the hematologic compartment and include short- and long-term adverse effects on many tissues including the brain. However, sensitive biomarkers of iron-dependent brain health are lacking in humans. To determine whether serum and cerebrospinal fluid (CSF) biomarkers of ID-induced metabolic dysfunction are concordant in the pre/early anemic stage of ID before anemia in a nonhuman primate model of infantile iron deficiency anemia (IDA). ID (n = 7), rhesus infants at 4 mo (pre-anemic period) and 6 mo of age (anemic) were examined. Hematological, metabolomic, and proteomic profiles were generated via HPLC/MS at both time points to discriminate serum biomarkers of ID-induced brain metabolic dysfunction. We identified 227 metabolites and 205 proteins in serum. Abnormalities indicating altered liver function, lipid dysregulation, and increased acute phase reactants were present in ID. In CSF, we measured 210 metabolites and 1,560 proteins with changes in ID infants indicative of metabolomic and proteomic differences indexing disrupted synaptogenesis. Systemic and CSF proteomic and metabolomic changes were present and concurrent in the pre-anemic and anemic periods. Multiomic serum and CSF profiling uncovered pathways disrupted by ID in both the pre-anemic and anemic stages of infantile IDA, including evidence for hepatic dysfunction and activation of acute phase response. Parallel changes observed in serum and CSF potentially provide measurable serum biomarkers of ID that reflect at-risk brain processes prior to progression to clinical anemia.
Collapse
Affiliation(s)
- Brian J Sandri
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, Minnesota
| | - Jonathan Kim
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Gabriele R Lubach
- Harlow Center for Biological Psychology, University of Wisconsin, Madison, Wisconsin
| | - Eric F Lock
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Candace Guerrero
- Department of Biochemistry, Molecular Biology, & Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology, & Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Todd W Markowski
- Department of Biochemistry, Molecular Biology, & Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Pamela J Kling
- Division of Neonatology, Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, Minnesota
| | - Christopher L Coe
- Harlow Center for Biological Psychology, University of Wisconsin, Madison, Wisconsin
| | - Raghavendra B Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
- Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
9
|
Sandri BJ, Lubach GR, Lock EF, Kling PJ, Georgieff MK, Coe CL, Rao RB. 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. [PMID: 33740040 PMCID: PMC8023818 DOI: 10.1093/ajcn/nqaa393] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The effects of infantile iron deficiency anemia (IDA) extend beyond hematological indices and include short- and long-term adverse effects on multiple cells and tissues. IDA is associated with an abnormal serum metabolomic profile, characterized by altered hepatic metabolism, lowered NAD flux, increased nucleoside levels, and a reduction in circulating dopamine levels. OBJECTIVES The objective of this study was to determine whether the serum metabolomic profile is normalized after rapid correction of IDA using iron dextran injections. METHODS Blood was collected from iron-sufficient (IS; n = 10) and IDA (n = 12) rhesus infants at 6 months of age. IDA infants were then administered iron dextran and vitamin B via intramuscular injections at weekly intervals for 2 to 8 weeks. Their hematological and metabolomic statuses were evaluated following treatment and compared with baseline and a separate group of age-matched IS infants (n = 5). RESULTS Serum metabolomic profiles assessed at baseline and after treatment via HPLC/MS using isobaric standards identified 654 quantifiable metabolites. At baseline, 53 metabolites differed between IS and IDA infants. Iron treatment restored traditional hematological indices, including hemoglobin and mean corpuscular volume, into the normal range, but the metabolite profile in the IDA group after iron treatment was markedly altered, with 323 metabolites differentially expressed when compared with an infant's own baseline profile. CONCLUSIONS Rapid correction of IDA with iron dextran resulted in extensive metabolic changes across biochemical pathways indexing the liver function, bile acid release, essential fatty acid production, nucleoside release, and several neurologically important metabolites. The results highlight the importance of a cautious approach when developing a route and regimen of iron repletion to treat infantile IDA.
Collapse
Affiliation(s)
- Brian J Sandri
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA,Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN, USA
| | - Gabriele R Lubach
- Harlow Center for Biological Psychology, University of Wisconsin, Madison, WI, USA
| | - Eric F Lock
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamela J Kling
- Division of Neonatology, Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA,Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN, USA
| | - Christopher L Coe
- Harlow Center for Biological Psychology, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
10
|
Hsieh HY, Chen YC, Hsu MH, Yu HR, Su CH, Tain YL, Huang LT, Sheen JM. Maternal Iron Deficiency Programs Offspring Cognition and Its Relationship with Gastrointestinal Microbiota and Metabolites. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E6070. [PMID: 32825437 PMCID: PMC7504367 DOI: 10.3390/ijerph17176070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Iron is an essential micronutrient for the brain development of the fetus. Altered intestinal microbiota might affect behavior and cognition through the so-called microbiota-gut-brain axis. We used a Sprague-Dawley rat model of a maternal low-iron diet to explore the changes in cognition, dorsal hippocampal brain-derived neurotrophic factor (BDNF) and related pathways, gut microbiota, and related metabolites in adult male offspring. We established maternal iron-deficient rats by feeding them a low-iron diet (2.9 mg/kg), while the control rats were fed a standard diet (52.3 mg/kg). We used a Morris water maze test to assess spatial learning and long-term memory. Western blot (WB) assays and a quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were used to detect the BDNF concentration and related signaling pathways. We collected fecal samples for microbiota profiling and measured the concentrations of plasma short-chain fatty acids. The adult male offspring of maternal rats fed low-iron diets before pregnancy, during pregnancy and throughout the lactation period had (1) spatial deficits, (2) a decreased BDNF mRNA expression and protein concentrations, accompanied by a decreased TrkB protein abundance, (3) a decreased plasma acetate concentration, and (4) an enrichment of the Bacteroidaceae genus Bacteroides and Lachnospiraceae genus Marvinbryantia. Maternal iron deficiency leads to an offspring spatial deficit and is associated with alternations in gastrointestinal microbiota and metabolites.
Collapse
Affiliation(s)
- Hsin-Yi Hsieh
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-Y.H.); (Y.-C.C.); (M.-H.H.); (H.-R.Y.); (Y.-L.T.)
| | - Yu-Chieh Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-Y.H.); (Y.-C.C.); (M.-H.H.); (H.-R.Y.); (Y.-L.T.)
| | - Mei-Hsin Hsu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-Y.H.); (Y.-C.C.); (M.-H.H.); (H.-R.Y.); (Y.-L.T.)
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-Y.H.); (Y.-C.C.); (M.-H.H.); (H.-R.Y.); (Y.-L.T.)
| | - Chung-Hao Su
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Chiayi 613, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-Y.H.); (Y.-C.C.); (M.-H.H.); (H.-R.Y.); (Y.-L.T.)
| | - Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-Y.H.); (Y.-C.C.); (M.-H.H.); (H.-R.Y.); (Y.-L.T.)
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-Y.H.); (Y.-C.C.); (M.-H.H.); (H.-R.Y.); (Y.-L.T.)
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Chiayi 613, Taiwan;
| |
Collapse
|
11
|
Bastian TW, Rao R, Tran PV, Georgieff MK. The Effects of Early-Life Iron Deficiency on Brain Energy Metabolism. Neurosci Insights 2020; 15:2633105520935104. [PMID: 32637938 PMCID: PMC7324901 DOI: 10.1177/2633105520935104] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Iron deficiency (ID) is one of the most prevalent nutritional deficiencies in the world. Iron deficiency in the late fetal and newborn period causes abnormal cognitive performance and emotional regulation, which can persist into adulthood despite iron repletion. Potential mechanisms contributing to these impairments include deficits in brain energy metabolism, neurotransmission, and myelination. Here, we comprehensively review the existing data that demonstrate diminished brain energetic capacity as a mechanistic driver of impaired neurobehavioral development due to early-life (fetal-neonatal) ID. We further discuss a novel hypothesis that permanent metabolic reprogramming, which occurs during the period of ID, leads to chronically impaired neuronal energetics and mitochondrial capacity in adulthood, thus limiting adult neuroplasticity and neurobehavioral function. We conclude that early-life ID impairs energy metabolism in a brain region- and age-dependent manner, with particularly strong evidence for hippocampal neurons. Additional studies, focusing on other brain regions and cell types, are needed.
Collapse
Affiliation(s)
- Thomas W Bastian
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Raghavendra Rao
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Phu V Tran
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Michael K Georgieff
- Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
12
|
Schachtschneider KM, Welge ME, Auvil LS, Chaki S, Rund LA, Madsen O, Elmore MR, Johnson RW, Groenen MA, Schook LB. Altered Hippocampal Epigenetic Regulation Underlying Reduced Cognitive Development in Response to Early Life Environmental Insults. Genes (Basel) 2020; 11:genes11020162. [PMID: 32033187 PMCID: PMC7074491 DOI: 10.3390/genes11020162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is involved in learning and memory and undergoes significant growth and maturation during the neonatal period. Environmental insults during this developmental timeframe can have lasting effects on brain structure and function. This study assessed hippocampal DNA methylation and gene transcription from two independent studies reporting reduced cognitive development stemming from early life environmental insults (iron deficiency and porcine reproductive and respiratory syndrome virus (PRRSv) infection) using porcine biomedical models. In total, 420 differentially expressed genes (DEGs) were identified between the reduced cognition and control groups, including genes involved in neurodevelopment and function. Gene ontology (GO) terms enriched for DEGs were associated with immune responses, angiogenesis, and cellular development. In addition, 116 differentially methylated regions (DMRs) were identified, which overlapped 125 genes. While no GO terms were enriched for genes overlapping DMRs, many of these genes are known to be involved in neurodevelopment and function, angiogenesis, and immunity. The observed altered methylation and expression of genes involved in neurological function suggest reduced cognition in response to early life environmental insults is due to altered cholinergic signaling and calcium regulation. Finally, two DMRs overlapped with two DEGs, VWF and LRRC32, which are associated with blood brain barrier permeability and regulatory T-cell activation, respectively. These results support the role of altered hippocampal DNA methylation and gene expression in early life environmentally-induced reductions in cognitive development across independent studies.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Michael E. Welge
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Loretta S. Auvil
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Sulalita Chaki
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Laurie A. Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Monica R.P. Elmore
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Rodney W. Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Martien A.M. Groenen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
- Correspondence:
| |
Collapse
|
13
|
Xing Y, Zhang W, Zhao H, Shen Z, Liang W, Zhou J, Shi L, Chen J, Zhong X, Tang S. Multi‑organ assessment via a 9.4‑Tesla MRS evaluation of metabolites during the embryonic development of cleft palate induced by dexamethasone. Mol Med Rep 2019; 20:3326-3336. [PMID: 31432193 PMCID: PMC6755240 DOI: 10.3892/mmr.2019.10558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 06/19/2019] [Indexed: 02/05/2023] Open
Abstract
The aim of the present study was to determine the association between maternal metabolism and development of the fetal palate, and to suggest a potential non‑invasive prenatal diagnostic method for fetal cleft palate (CP). Dexamethasone (DXM) was used to create a CP mouse model. A 9.4‑Tesla (T) magnetic resonance spectroscopy (MRS) imager was used to measure an array of metabolites in the maternal serum, placental tissue, amniotic fluid and fetal palates. Multivariate statistical analysis was performed using SIMCA‑P 14.1 software. Following DXM treatment, variations were detected in multiple metabolites in the female mice and their fetuses based on 9.4T MRS. It was indicated that in the experimental group during CP formation, leucine, valine, creatine, acetate and citrate levels in the palatal tissue were lower, whereas lactate, alanine, proline/inositol and glutamate‑containing metabolite levels were higher, compared with the levels in the control group. In placental tissue and amniotic fluid, succinate and choline levels were lower in the experimental group. The relative concentrations of cholesterol and lipids in palatal tissues from mice treated with DXM were higher compared with the concentrations in tissues from mice in the control group, with the exception of (CH2)n lipids. In the placental tissue, the alteration in cholesterol level exhibited the opposite trend. Lipid levels for the different lipid forms varied and most of them were unsaturated lipids.
Collapse
Affiliation(s)
- Yue Xing
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Wancong Zhang
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Hanxing Zhao
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Zhiwei Shen
- Department of Medical Imaging, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Weijie Liang
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, Central South University Third Xiangya Hospital, Changsha, Hunan 410013, P.R. China
| | - Lungang Shi
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Jiasheng Chen
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiaoping Zhong
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Dr Shijie Tang, Department of Burns and Plastic Surgery, and Cleft Lip and Palate Treatment Center, The Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia Road, Shantou, Guangdong 515041, P.R. China, E-mail: ;
| |
Collapse
|
14
|
Ennis K, Felt B, Georgieff MK, Rao R. Early-Life Iron Deficiency Alters Glucose Transporter-1 Expression in the Adult Rodent Hippocampus. J Nutr 2019; 149:1660-1666. [PMID: 31162576 PMCID: PMC6736205 DOI: 10.1093/jn/nxz100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/26/2018] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Early-life iron deficiency (ID) impairs hippocampal energy production. Whether there are changes in glucose transporter (GLUT) expression is not known. OBJECTIVE The aim of this study was to investigate whether early-life ID and the treatment iron dose alter brain regional GLUT expression in adult rats and mice. METHODS In Study 1, ID was induced in male and female Sprague Dawley rat pups by feeding dams a 3-mg/kg iron diet during gestation and the first postnatal week, followed by treatment using low-iron [3-10 mg/kg; formerly iron-deficient (FID)-10 group], standard-iron (40-mg/kg; FID-40 group), or high-iron (400-mg/kg; FID-400 group) diets until weaning. The control group received the 40 mg/kg iron diet. GLUT1, GLUT3, hypoxia-inducible factor (HIF)-1α, and prolyl-hydroxylase-2 (PHD2) mRNA and protein expression in the cerebral cortex, hippocampus, striatum, cerebellum, and hypothalamus were determined at adulthood. In Study 2, the role of hippocampal ID in GLUT expression was examined by comparing the Glut1, Glut3, Hif1α, and Phd2 mRNA expression in adult male and female wild-type (WT) and nonanemic hippocampal iron-deficient and iron-replete dominant negative transferrin receptor 1 (DNTfR1-/-) transgenic mice. RESULTS In Study 1, Glut1, Glut3, and Hif1α mRNA, and GLUT1 55-kDa protein expression was upregulated 20-33% in the hippocampus of the FID-10 group but not the FID-40 group, relative to the control group. Hippocampal Glut1 mRNA (-39%) and GLUT1 protein (-30%) expression was suppressed in the FID-400 group, relative to the control group. Glut1 and Glut3 mRNA expression was not altered in the other brain regions in the 3 FID groups. In Study 2, hippocampal Glut1 (+14%) and Hif1α (+147%) expression was upregulated in the iron-deficient DNTfR1-/- mice, but not in the iron-replete DNTfR1-/- mice, relative to the WT mice (P < 0.05, all). CONCLUSIONS Early-life ID is associated with altered hippocampal GLUT1 expression in adult rodents. The mouse study suggests that tissue ID is potentially responsible.
Collapse
Affiliation(s)
- Kathleen Ennis
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Barbara Felt
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA,Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN, USA
| | - Raghavendra Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA,Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN, USA,Address correspondence to RR (e-mail: )
| |
Collapse
|
15
|
Markova V, Holm C, Pinborg AB, Thomsen LL, Moos T. Impairment of the Developing Human Brain in Iron Deficiency: Correlations to Findings in Experimental Animals and Prospects for Early Intervention Therapy. Pharmaceuticals (Basel) 2019; 12:ph12030120. [PMID: 31416268 PMCID: PMC6789712 DOI: 10.3390/ph12030120] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
Due to the necessity of iron for a variety of cellular functions, the developing mammalian organism is vulnerable to iron deficiency, hence causing structural abnormalities and physiological malfunctioning in organs, which are particularly dependent on adequate iron stores, such as the brain. In early embryonic life, iron is already needed for proper development of the brain with the proliferation, migration, and differentiation of neuro-progenitor cells. This is underpinned by the widespread expression of transferrin receptors in the developing brain, which, in later life, is restricted to cells of the blood–brain and blood–cerebrospinal fluid barriers and neuronal cells, hence ensuring a sustained iron supply to the brain, even in the fully developed brain. In embryonic human life, iron deficiency is thought to result in a lower brain weight, with the impaired formation of myelin. Studies of fully developed infants that have experienced iron deficiency during development reveal the chronic and irreversible impairment of cognitive, memory, and motor skills, indicating widespread effects on the human brain. This review highlights the major findings of recent decades on the effects of gestational and lactational iron deficiency on the developing human brain. The findings are correlated to findings of experimental animals ranging from rodents to domestic pigs and non-human primates. The results point towards significant effects of iron deficiency on the developing brain. Evidence would be stronger with more studies addressing the human brain in real-time and the development of blood biomarkers of cerebral disturbance in iron deficiency. Cerebral iron deficiency is expected to be curable with iron substitution therapy, as the brain, privileged by the cerebral vascular transferrin receptor expression, is expected to facilitate iron extraction from the circulation and enable transport further into the brain.
Collapse
Affiliation(s)
- Veronika Markova
- Department of Obstetrics and Gynaecology, Hvidovre Hospital, Copenhagen University Hospital, 2650 Hvidovre, Denmark
- Pharmacosmos A/S, 4300 Holbæk, Denmark
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Charlotte Holm
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Anja Bisgaard Pinborg
- Fertility Clinic, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Lars Lykke Thomsen
- Pharmacosmos A/S, 4300 Holbæk, Denmark
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark
| | - Torben Moos
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark.
| |
Collapse
|
16
|
Roles of taurine in cognitive function of physiology, pathologies and toxication. Life Sci 2019; 231:116584. [DOI: 10.1016/j.lfs.2019.116584] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/09/2019] [Accepted: 06/17/2019] [Indexed: 11/23/2022]
|
17
|
Lien YC, Condon DE, Georgieff MK, Simmons RA, Tran PV. Dysregulation of Neuronal Genes by Fetal-Neonatal Iron Deficiency Anemia Is Associated with Altered DNA Methylation in the Rat Hippocampus. Nutrients 2019; 11:nu11051191. [PMID: 31137889 PMCID: PMC6566599 DOI: 10.3390/nu11051191] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023] Open
Abstract
Early-life iron deficiency results in long-term abnormalities in cognitive function and affective behavior in adulthood. In preclinical models, these effects have been associated with long-term dysregulation of key neuronal genes. While limited evidence suggests histone methylation as an epigenetic mechanism underlying gene dysregulation, the role of DNA methylation remains unknown. To determine whether DNA methylation is a potential mechanism by which early-life iron deficiency induces gene dysregulation, we performed whole genome bisulfite sequencing to identify loci with altered DNA methylation in the postnatal day (P) 15 iron-deficient (ID) rat hippocampus, a time point at which the highest level of hippocampal iron deficiency is concurrent with peak iron demand for axonal and dendritic growth. We identified 229 differentially methylated loci and they were mapped within 108 genes. Among them, 63 and 45 genes showed significantly increased and decreased DNA methylation in the P15 ID hippocampus, respectively. To establish a correlation between differentially methylated loci and gene dysregulation, the methylome data were compared to our published P15 hippocampal transcriptome. Both datasets showed alteration of similar functional networks regulating nervous system development and cell-to-cell signaling that are critical for learning and behavior. Collectively, the present findings support a role for DNA methylation in neural gene dysregulation following early-life iron deficiency.
Collapse
Affiliation(s)
- Yu-Chin Lien
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - David E Condon
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Michael K Georgieff
- Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA.
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA.
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA.
| |
Collapse
|
18
|
Iron as a model nutrient for understanding the nutritional origins of neuropsychiatric disease. Pediatr Res 2019; 85:176-182. [PMID: 30341413 PMCID: PMC6353667 DOI: 10.1038/s41390-018-0204-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/04/2018] [Accepted: 10/04/2018] [Indexed: 12/19/2022]
Abstract
Adequate nutrition during the pre- and early-postnatal periods plays a critical role in programming early neurodevelopment. Disruption of neurodevelopment by nutritional deficiencies can result not only in lasting functional deficits, but increased risk of neuropsychiatric disease in adulthood. Historical periods of famine such as the Dutch Hunger Winter and the Chinese Famine have provided foundational evidence for the long-term effects of developmental malnutrition on neuropsychiatric outcomes. Because neurodevelopment is a complex process that consists of many nutrient- and brain-region-specific critical periods, subsequent clinical and pre-clinical studies have aimed to elucidate the specific roles of individual macro- and micronutrient deficiencies in neurodevelopment and neuropsychiatric pathologies. This review will discuss developmental iron deficiency (ID), the most common micronutrient deficiency worldwide, as a paradigm for understanding the role of early-life nutrition in neurodevelopment and risk of neuropsychiatric disease. We will review the epidemiologic data linking ID to neuropsychiatric dysfunction, as well as the underlying structural, cellular, and molecular mechanisms that are thought to underlie these lasting effects. Understanding the mechanisms driving lasting dysfunction and disease risk is critical for development and implementation of nutritional policies aimed at preventing nutritional deficiencies and their long-term sequelae.
Collapse
|
19
|
Abstract
Appraising success in meeting the world's nutritional needs has largely focused on infant mortality and anthropometric measurements with an emphasis on the first 1,000 days (conception to approximately age 2 years). This ignores the unique nutritional needs of the human brain. Although the intrauterine environment and the early postnatal years are important, equally critical periods follow during which the brain's intricate wiring is established for a lifetime of experience-driven remodeling. At the peak of this process during childhood, the human brain may account for 50% of the body's basal nutritional requirement. Thus, the consequences of proper nutritional management of the brain play out over a lifetime. Our motivation in preparing this review was to move the human brain into a more central position in the planning of nutritional programs. Here we review the macro- and micronutrient requirements of the human brain and how they are delivered, from conception to adulthood.
Collapse
Affiliation(s)
- Manu S. Goyal
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| | - Lora L. Iannotti
- Brown School, Institute for Public Health, Washington University, St. Louis, Missouri 63130, USA
| | - Marcus E. Raichle
- Mallinckrodt Institute of Radiology and Department of Neurology, Washington University School of Medicine, Washington University, St. Louis, Missouri 63130, USA
| |
Collapse
|
20
|
Georgieff MK, Tran PV, Carlson ES. Atypical fetal development: Fetal alcohol syndrome, nutritional deprivation, teratogens, and risk for neurodevelopmental disorders and psychopathology. Dev Psychopathol 2018; 30:1063-1086. [PMID: 30068419 PMCID: PMC6074054 DOI: 10.1017/s0954579418000500] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Accumulating evidence indicates that the fetal environment plays an important role in brain development and sets the brain on a trajectory across the life span. An abnormal fetal environment results when factors that should be present during a critical period of development are absent or when factors that should not be in the developing brain are present. While these factors may acutely disrupt brain function, the real cost to society resides in the long-term effects, which include important mental health issues. We review the effects of three factors, fetal alcohol exposure, teratogen exposure, and nutrient deficiencies, on the developing brain and the consequent risk for developmental psychopathology. Each is reviewed with respect to the evidence found in epidemiological and clinical studies in humans as well as preclinical molecular and cellular studies that explicate mechanisms of action.
Collapse
Affiliation(s)
| | - Phu V Tran
- University of Minnesota School of Medicine
| | | |
Collapse
|
21
|
Rao R, Nashawaty M, Fatima S, Ennis K, Tkac I. Neonatal hyperglycemia alters the neurochemical profile, dendritic arborization and gene expression in the developing rat hippocampus. NMR IN BIOMEDICINE 2018; 31. [PMID: 29532970 PMCID: PMC6489495 DOI: 10.1002/nbm.3910] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Hyperglycemia (blood glucose concentration >150 mg/dL) is common in extremely low gestational age newborns (ELGANs; birth at <28 week gestation). Hyperglycemia increases the risk of brain injury in the neonatal period. The long-term effects are not well understood. In adult rats, hyperglycemia alters hippocampal energy metabolism. The effects of hyperglycemia on the developing hippocampus were studied in rat pups. In Experiment 1, recurrent hyperglycemia of graded severity (moderate hyperglycemia (moderate-HG), mean blood glucose 214.6 ± 11.6 mg/dL; severe hyperglycemia (severe-HG), 338.9 ± 21.7 mg/dL; control, 137.7 ± 2.6 mg/dL) was induced from postnatal day (P) 3 to P12. On P30, the hippocampal neurochemical profile was determined using in vivo 1 H MR spectroscopy. Dendritic arborization in the hippocampal CA1 region was determined using microtubule-associated protein (MAP)-2 immunohistochemistry. In Experiment 2, continuous hyperglycemia (mean blood glucose 275.3 ± 25.8 mg/dL; control, 142.3 ± 2.6 mg/dL) was induced from P2 to P6 by injecting streptozotocin (STZ) on P2. The mRNA expression of glycogen synthase 1 (Gys1), lactate dehydrogenase (Ldh), glucose transporters 1 (Glut1) and 3 (Glut3) and monocarboxylate transporters 1 (Mct1), 2 (Mct2) and 4 (Mct4) in the hippocampus was determined on P6. In Experiment 1, MRS demonstrated lower lactate concentration and glutamate/glutamine (Glu/Gln) ratio in the severe-HG group, compared with the control group (p < 0.05). Phosphocreatine/creatine ratio was higher in both hyperglycemia groups (p < 0.05). MAP-2 histochemistry demonstrated longer apical segment length, indicating abnormal synaptic efficacy in both hyperglycemia groups (p < 0.05). Experiment 2 showed lower Glut1, Gys1 and Mct4 expression and higher Mct1 expression in the hyperglycemia group, relative to the control group (p < 0.05). These results suggest that hyperglycemia alters substrate transport, lactate homeostasis, dendritogenesis and Glu-Gln cycling in the developing hippocampus. Abnormal neurochemical profile and dendritic structure due to hyperglycemia may partially explain the long-term hippocampus-mediated cognitive deficits in human ELGANs.
Collapse
Affiliation(s)
- Raghavendra Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota
- Centre for Neurobehavioral Development, University of Minnesota
- Address for correspondence: Mayo Mail Code 39, 420 Delaware Street, SE, Minneapolis, MN 55455 (USA). Phone: 612-625-3260; Fax: 612-624-8176;
| | - Motaz Nashawaty
- Division of Neonatology, Department of Pediatrics, University of Minnesota
| | - Saher Fatima
- Division of Neonatology, Department of Pediatrics, University of Minnesota
| | - Kathleen Ennis
- Division of Neonatology, Department of Pediatrics, University of Minnesota
| | - Ivan Tkac
- Center for Magnetic Resonance Research, University of Minnesota
| |
Collapse
|
22
|
Metabolic phenotyping of malnutrition during the first 1000 days of life. Eur J Nutr 2018; 58:909-930. [PMID: 29644395 PMCID: PMC6499750 DOI: 10.1007/s00394-018-1679-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/26/2018] [Indexed: 02/07/2023]
Abstract
Nutritional restrictions during the first 1000 days of life can impair or delay the physical and cognitive development of the individual and have long-term consequences for their health. Metabolic phenotyping (metabolomics/metabonomics) simultaneously measures a diverse range of low molecular weight metabolites in a sample providing a comprehensive assessment of the individual's biochemical status. There are a growing number of studies applying such approaches to characterize the metabolic derangements induced by various forms of early-life malnutrition. This includes acute and chronic undernutrition and specific micronutrient deficiencies. Collectively, these studies highlight the diverse and dynamic metabolic disruptions resulting from various forms of nutritional deficiencies. Perturbations were observed in many pathways including those involved in energy, amino acid, and bile acid metabolism, the metabolic interactions between the gut microbiota and the host, and changes in metabolites associated with gut health. The information gleaned from such studies provides novel insights into the mechanisms linking malnutrition with developmental impairments and assists in the elucidation of candidate biomarkers to identify individuals at risk of developmental shortfalls. As the metabolic profile represents a snapshot of the biochemical status of an individual at a given time, there is great potential to use this information to tailor interventional strategies specifically to the metabolic needs of the individual.
Collapse
|
23
|
Larson LM, Martorell R, Bauer PJ. A Path Analysis of Nutrition, Stimulation, and Child Development Among Young Children in Bihar, India. Child Dev 2018. [PMID: 29529358 PMCID: PMC6174960 DOI: 10.1111/cdev.13057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nutrition plays an important role in the development of a child, particularly in low‐ and middle‐income countries where malnutrition is often widespread. The relation between diet, hemoglobin, nutritional status, motor development, stimulation and mental development was examined in a cross‐sectional sample of 1,079 children 12–18 months of age living in rural Bihar, India. Path analysis revealed associations between (a) length‐for‐age z‐scores and motor development, standardized β (β) = .285, p < .001, and (b) motor and all mental development outcomes (language: β = .422; personal‐social: β = .490; memory: β = .139; and executive function: β = .072, all p < .001). Additionally, stimulation was significantly associated with language scores and hemoglobin concentration with memory. These findings inform interventions aimed at improving child development in Northern India.
Collapse
|
24
|
Basu S, Kumar D, Anupurba S, Verma A, Kumar A. Effect of maternal iron deficiency anemia on fetal neural development. J Perinatol 2018; 38:233-239. [PMID: 29234149 DOI: 10.1038/s41372-017-0023-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/03/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Perinatal iron deficiency may have deleterious consequences on fetal neural development. The present study was conducted to determine the effect of maternal iron deficiency anemia (IDA) on fetal hippocampal morphogenesis and production of brain-derived neurotrophic factor (BDNF). STUDY DESIGN Seventy term, singleton neonates born to mothers with IDA (hemoglobin <110g/L and serum ferritin <12 μg/L) formed the study group. Twenty gestational age-matched neonates born to healthy mothers without IDA (hemoglobin ≥110 g/L and serum ferritin >12 μg/L) served as controls. Maternal and fetal inflammatory conditions, infections and neonates with perinatal asphyxia were excluded. Cord blood BDNF concentrations were estimated by enzyme-linked immunosorbent assay. Volumetric analysis of hippocampus (right, left and combined, corrected for total intracranial volume) was done by cranial magnetic resonance imaging on days 3-5 of life. RESULTS In the study group, 24 mothers had mild (hemoglobin 100.0-109.0 g/L), 24 had moderate (hemoglobin 70.0-99.0 g/L), and 22 had severe (hemoglobin <70.0 g/L) anemia. Both hippocampal volumes and serum BDNF concentrations of neonates born to iron-deficient mothers were significantly reduced compared to controls. A progressive decline in hippocampal volumes and BDNF concentrations was observed with increasing severity of maternal anemia. Pearson correlation showed significant correlations among maternal and cord blood hemoglobin, iron indices, hippocampal volumes and BDNF concentrations. CONCLUSIONS Maternal IDA adversely affects hippocampal morphogenesis and fetal production of BDNF. The degree of affection is proportional to the severity of maternal anemia.
Collapse
Affiliation(s)
- Sriparna Basu
- Division of Neonatology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| | - Dinesh Kumar
- Division of Neonatology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shampa Anupurba
- Department of Microbiology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashish Verma
- Department of Radiodiagnosis, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ashok Kumar
- Division of Neonatology, Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
25
|
Li Y, Yan J, Zhu X, Zhu Y, Qin J, Zhang N, Ju S. Increased hippocampal fissure width is a sensitive indicator of rat hippocampal atrophy. Brain Res Bull 2017; 137:91-97. [PMID: 29174731 DOI: 10.1016/j.brainresbull.2017.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 11/08/2017] [Accepted: 11/22/2017] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Volume loss within the hippocampus is known as the most replicated finding of structural brain imaging studies of neuropsychiatric diseases. Although voxel-based auto or semi-auto volumetric measurements are widely used in the determination of the human hippocampus, the detection of hippocampal atrophy in rats is still a dilemma as it relies on a relatively primitive and complex approach. In this study, we aimed to develop a convenient way to measure the atrophy of the hippocampus in rats. METHODS Twenty-four male Wistar rats were exposed to chronic unpredictable mild stress (CUMS) and a wheel running test (WRT) to simulate the conditions of hippocampal volume atrophy and improvement. The hippocampal volume and hippocampal fissure (HiF) width were dynamically measured using 7 T structural magnetic resonance imaging (MRI) with the grayscale method at week 0, 2, 4, and 8. The changes in the hippocampal volume and HiF width in rats were compared. In addition, hematoxylin-eosin (HE) staining of the HiF was used to verify the MRI findings. RESULTS The hippocampal volume and the HiF width presented opposite trends based on the MRI findings and the histology data. The atrophy of the hippocampal subfields was closely related to the corresponding increase in the HiF width. CONCLUSION Determination of the HiF width may serve as a sensitive and convenient indicator of rat hippocampal atrophy.
Collapse
Affiliation(s)
- Yuefeng Li
- Department of Radiology, Zhongda Affiliated Hospital of Southeast University, Nanjing, China; Department of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jinchuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaolan Zhu
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Yan Zhu
- Department of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jiasheng Qin
- Department of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ningning Zhang
- Department of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shenghong Ju
- Department of Radiology, Zhongda Affiliated Hospital of Southeast University, Nanjing, China.
| |
Collapse
|
26
|
Murray-Kolb LE, Wenger MJ, Scott SP, Rhoten SE, Lung’aho MG, Haas JD. Consumption of Iron-Biofortified Beans Positively Affects Cognitive Performance in 18- to 27-Year-Old Rwandan Female College Students in an 18-Week Randomized Controlled Efficacy Trial. J Nutr 2017; 147:2109-2117. [PMID: 28954841 PMCID: PMC5657139 DOI: 10.3945/jn.117.255356] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/22/2017] [Accepted: 08/29/2017] [Indexed: 12/31/2022] Open
Abstract
Background: Evidence shows that iron deficiency in adulthood may affect cognitive performance, possibly by disrupting neurotransmitter regulation or brain energy metabolism. Women of reproductive age (WRA) are among those who are most vulnerable to iron deficiency; however, they have been largely ignored in the literature relating iron status to cognition.Objective: Our aim was to determine the efficacy of iron-biofortified beans in improving cognition in WRA compared with control beans.Methods: A double-blind, randomized intervention study was conducted in 150 women aged 18-27 y with low iron status (ferritin <20 μg/L). Women were randomly assigned to consume iron-biofortified beans (86.1 ppm iron) or control beans (50.1 ppm iron) daily for 18 wk. Iron status was assessed based on hemoglobin, ferritin, transferrin receptor, and body iron values and on cognitive performance on 5 computerized tasks at baseline and endline.Results: Groups did not differ on any variables at baseline. Per protocol analyses revealed that consumption of the biofortified beans resulted in a 17% larger improvement in the speed of spatial selective attention; a nearly 7-fold larger improvement in the speed, a 68% greater improvement in the efficiency, and a >2-fold greater improvement in the specificity of memory retrieval; and a >2-fold larger improvement in the speed and a >3-fold larger improvement in the efficiency of memory search-all of which are relative to consumption of the control beans (P < 0.01 for all comparisons).Conclusions: Cognitive performance is sensitive to iron status, and consumption of iron-biofortified beans for 18 wk improved cognitive performance, especially the efficiency of search and the speed of retrieval on memory tasks, in young adult women. This trial was registered at clinicaltrials.gov as NCT01594359.
Collapse
Affiliation(s)
- Laura E Murray-Kolb
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA;
| | - Michael J Wenger
- Department of Psychology and Cellular and Behavioral Neurobiology, University of Oklahoma, Norman, OK;,Division of Nutritional Sciences, Cornell University, Ithaca, NY; and
| | - Samuel P Scott
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Stephanie E Rhoten
- Department of Psychology and Cellular and Behavioral Neurobiology, University of Oklahoma, Norman, OK
| | | | - Jere D Haas
- Division of Nutritional Sciences, Cornell University, Ithaca, NY; and
| |
Collapse
|
27
|
Bastian TW, von Hohenberg WC, Mickelson DJ, Lanier LM, Georgieff MK. Iron Deficiency Impairs Developing Hippocampal Neuron Gene Expression, Energy Metabolism, and Dendrite Complexity. Dev Neurosci 2016; 38:264-276. [PMID: 27669335 DOI: 10.1159/000448514] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/05/2016] [Indexed: 12/28/2022] Open
Abstract
Iron deficiency (ID), with and without anemia, affects an estimated 2 billion people worldwide. ID is particularly deleterious during early-life brain development, leading to long-term neurological impairments including deficits in hippocampus-mediated learning and memory. Neonatal rats with fetal/neonatal ID anemia (IDA) have shorter hippocampal CA1 apical dendrites with disorganized branching. ID-induced dendritic structural abnormalities persist into adulthood despite normalization of the iron status. However, the specific developmental effects of neuronal iron loss on hippocampal neuron dendrite growth and branching are unknown. Embryonic hippocampal neuron cultures were chronically treated with deferoxamine (DFO, an iron chelator) beginning at 3 days in vitro (DIV). Levels of mRNA for Tfr1 and Slc11a2, iron-responsive genes involved in iron uptake, were significantly elevated in DFO-treated cultures at 11DIV and 18DIV, indicating a degree of neuronal ID similar to that seen in rodent ID models. DFO treatment decreased mRNA levels for genes indexing dendritic and synaptic development (i.e. BdnfVI,Camk2a,Vamp1,Psd95,Cfl1, Pfn1,Pfn2, and Gda) and mitochondrial function (i.e. Ucp2,Pink1, and Cox6a1). At 18DIV, DFO reduced key aspects of energy metabolism including basal respiration, maximal respiration, spare respiratory capacity, ATP production, and glycolytic rate, capacity, and reserve. Sholl analysis revealed a significant decrease in distal dendritic complexity in DFO-treated neurons at both 11DIV and 18DIV. At 11DIV, the length of primary dendrites and the number and length of branches in DFO-treated neurons were reduced. By 18DIV, partial recovery of the dendritic branch number in DFO-treated neurons was counteracted by a significant reduction in the number and length of primary dendrites and the length of branches. Our findings suggest that early neuronal iron loss, at least partially driven through altered mitochondrial function and neuronal energy metabolism, is responsible for the effects of fetal/neonatal ID and IDA on hippocampal neuron dendritic and synaptic maturation. Impairments in these neurodevelopmental processes likely underlie the negative impact of early life ID and IDA on hippocampus-mediated learning and memory.
Collapse
Affiliation(s)
- Thomas W Bastian
- Department of Pediatrics and Center for Neurobehavioral Development, School of Medicine, Minneapolis, Minn., USA
| | | | | | | | | |
Collapse
|
28
|
Alwan NA, Hamamy H. Maternal Iron Status in Pregnancy and Long-Term Health Outcomes in the Offspring. J Pediatr Genet 2016; 4:111-23. [PMID: 27617121 DOI: 10.1055/s-0035-1556742] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Iron is an essential micronutrient and is important not only in carrying oxygen but also to the catalytic activity of a variety of enzymes. In the fetus, it is vital to the synthesis of hemoglobin and in brain development. Iron deficiency (ID) anemia in pregnancy is a common problem, even in high-income country settings. Around 50% of pregnant women worldwide are anemic, with at least half of this burden due to ID. Iron supplements are widely recommended and used during pregnancy globally. However, the evidence on the extent of benefit they contribute to the offspring's health is not well established, and their routine use has its side effects and drawbacks. Dietary iron intake is difficult to assess accurately and it is unlikely to be sufficient to meet the demands of pregnancy if women start with inadequate body iron stores at conception. Evidence from experimental animal models suggests that maternal ID during pregnancy is associated with fetal growth restriction, as well as offspring obesity and high blood pressure later in life. The possible biological mechanisms for this observed association may be due to ID-induced changes in placental structure and function, enzyme expression, nutrient absorption, and fetal organ development. However, such evidence is limited in human studies. Prenatal ID in experimental animal models also adversely affected the developing brain structures, neurotransmitter systems, and myelination resulting in acute brain dysfunction during the period of deficiency and persistence of various postnatal neurobehavioral abnormalities as well as persistent dysregulation of some genes into adult life after iron repletion pointing to the possibility of gene expression changes. The evidence from human population studies is limited and heterogeneous and more research is needed in the future, investigating the effects of ID in pregnancy on future offspring health outcomes.
Collapse
Affiliation(s)
- Nisreen A Alwan
- Academic Unit of Primary Care and Population Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Hanan Hamamy
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| |
Collapse
|
29
|
Rao R, Ennis K, Lubach GR, Lock EF, Georgieff MK, Coe CL. Metabolomic analysis of CSF indicates brain metabolic impairment precedes hematological indices of anemia in the iron-deficient infant monkey. Nutr Neurosci 2016; 21:40-48. [PMID: 27499134 DOI: 10.1080/1028415x.2016.1217119] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Iron deficiency (ID) anemia leads to long-term neurodevelopmental deficits by altering iron-dependent brain metabolism. The objective of the study was to determine if ID induces metabolomic abnormalities in the cerebrospinal fluid (CSF) in the pre-anemic stage and to ascertain the aspects of abnormal brain metabolism affected. METHODS Standard hematological parameters [hemoglobin (Hgb), mean corpuscular volume (MCV), transferrin (Tf) saturation, and zinc protoporphyrin/heme (ZnPP/H)] were compared at 2, 4, 6, 8, and 12 months in iron-sufficient (IS; n = 7) and iron-deficient (ID; n = 7) infant rhesus monkeys. Five CSF metabolite ratios were determined at 4, 8, and 12 months using 1H NMR spectroscopy at 16.4 T and compared between groups and in relation to hematologic parameters. RESULTS ID infants developed ID (Tf saturation < 25%) by 4 months of age and all became anemic (Hgb < 110 g/L and MCV < 60 fL) at 6 months. Their heme indices normalized by 12 months. Pyruvate/glutamine and phosphocreatine/creatine (PCr/Cr) ratios in CSF were lower in the ID infants by 4 months (P < 0.05). The PCr/Cr ratio remained lower at 8 months (P = 0.02). ZnPP/H, an established blood marker of pre-anemic ID, was positively correlated with the CSF citrate/glutamine ratio (marginal correlation, 0.34; P < 0.001; family wise error rate = 0.001). DISCUSSION Metabolomic analysis of the CSF is sensitive for detecting the effects of pre-anemic ID on brain energy metabolism. Persistence of a lower PCr/Cr ratio at 8 months, even as hematological measures demonstrated recovery from anemia, indicate that the restoration of brain energy metabolism is delayed. Metabolomic platforms offer a useful tool for early detection of the impact of ID on brain metabolism in infants.
Collapse
Affiliation(s)
- Raghavendra Rao
- a Department of Pediatrics, Division of Neonatology , University of Minnesota , Minneapolis , USA.,b Center for Neurobehavioral Development , University of Minnesota , Minneapolis , USA
| | - Kathleen Ennis
- a Department of Pediatrics, Division of Neonatology , University of Minnesota , Minneapolis , USA
| | - Gabriele R Lubach
- c Harlow Center for Biological Psychology , University of Wisconsin-Madison , USA
| | - Eric F Lock
- d Division of Biostatistics , School of Public Health, University of Minnesota , Minneapolis , USA
| | - Michael K Georgieff
- a Department of Pediatrics, Division of Neonatology , University of Minnesota , Minneapolis , USA.,b Center for Neurobehavioral Development , University of Minnesota , Minneapolis , USA
| | - Christopher L Coe
- c Harlow Center for Biological Psychology , University of Wisconsin-Madison , USA
| |
Collapse
|
30
|
Tran PV, Kennedy BC, Pisansky MT, Won KJ, Gewirtz JC, Simmons RA, Georgieff MK. 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; 146:484-93. [PMID: 26865644 PMCID: PMC4763487 DOI: 10.3945/jn.115.227561] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Early-life iron deficiency is a common nutrient deficiency worldwide. Maternal iron deficiency increases the risk of schizophrenia and autism in the offspring. Postnatal iron deficiency in young children results in cognitive and socioemotional abnormalities in adulthood despite iron treatment. The rat model of diet-induced fetal-neonatal iron deficiency recapitulates the observed neurobehavioral deficits. OBJECTIVES We sought to establish molecular underpinnings for the persistent psychopathologic effects of early-life iron deficiency by determining whether it permanently reprograms the hippocampal transcriptome. We also assessed the effects of maternal dietary choline supplementation on the offspring's hippocampal transcriptome to identify pathways through which choline mitigates the emergence of long-term cognitive deficits. METHODS Male rat pups were made iron deficient (ID) by providing pregnant and nursing dams an ID diet (4 g Fe/kg) from gestational day (G) 2 through postnatal day (PND) 7 and an iron-sufficient (IS) diet (200 g Fe/kg) thereafter. Control pups were provided IS diet throughout. Choline (5 g/kg) was given to half the pregnant dams in each group from G11 to G18. PND65 hippocampal transcriptomes were assayed by next generation sequencing (NGS) and analyzed with the use of knowledge-based Ingenuity Pathway Analysis. Real-time polymerase chain reaction was performed to validate a subset of altered genes. RESULTS Formerly ID rats had altered hippocampal expression of 619 from >10,000 gene loci sequenced by NGS, many of which map onto molecular networks implicated in psychological disorders, including anxiety, autism, and schizophrenia. There were significant interactions between iron status and prenatal choline treatment in influencing gene expression. Choline supplementation reduced the effects of iron deficiency, including those on gene networks associated with autism and schizophrenia. CONCLUSIONS Fetal-neonatal iron deficiency reprograms molecular networks associated with the pathogenesis of neurologic and psychological disorders in adult rats. The positive response to prenatal choline represents a potential adjunctive therapeutic supplement to the high-risk group.
Collapse
Affiliation(s)
| | | | | | - Kyoung-Jae Won
- Institute for Diabetes, Obesity and Metabolism, Department of Genetics, and
| | - Jonathan C Gewirtz
- Graduate Program in Neuroscience, and Department of Psychology, University of Minnesota, Minneapolis, MN
| | - Rebecca A Simmons
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
31
|
Scott SP, Murray-Kolb LE. Iron Status Is Associated with Performance on Executive Functioning Tasks in Nonanemic Young Women. J Nutr 2016; 146:30-7. [PMID: 26661838 DOI: 10.3945/jn.115.223586] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Iron deficiency (ID) is prevalent, particularly among women of reproductive age (WRA). How mild ID without anemia relates to cognition is poorly understood. Executive functioning (EF) has emerged as potentially being affected by mild ID in WRA. OBJECTIVE We sought to examine how iron markers relate to performance on EF tasks in nonanemic WRA. METHODS Participants included 127 females aged 18-35 y. Hematological indicators included hemoglobin, RBC distribution width, transferrin saturation (TSAT), ferritin, transferrin receptor (TfR), and total body iron (TBI). EF was assessed using 5 tasks. Associations between EF outcomes and iron status were examined using continuous iron predictors and group comparisons. RESULTS Better iron status was associated with better attention [faster reaction time (RT) with lower TfR (P = 0.028) and higher TSAT (P = 0.013)], inhibitory control [lower RT variability with higher TSAT (P = 0.042) and planning ability (faster planning time and a smaller planning time increase with increasing difficulty with higher ferritin; P = 0.010)]. No associations with iron status were found for several EF outcomes, possibly due to performance ceilings. Paradoxically, worse performance on a working memory task was related to better iron status, which may reflect hippocampal-frontal interference [lower capacity with lower TfR (P = 0.034) and higher TBI (P = 0.043) and a larger accuracy change with increasing difficulty with higher TBI (P = 0.016)]. Longer RTs on a working memory task were observed among those with positive TBI (iron surplus; P = 0.021) and <2 abnormal iron markers (P = 0.013) compared with those with negative TBI (iron deficit) and ≥2 abnormal markers, respectively. CONCLUSIONS These findings suggest cognitive ramifications of mild ID in otherwise healthy WRA and have implications for daily well-being. Future investigators should explore how brain system interactions change according to iron availability.
Collapse
Affiliation(s)
- Samuel P Scott
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| | - Laura E Murray-Kolb
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA
| |
Collapse
|
32
|
Bakoyiannis I, Gkioka E, Daskalopoulou A, Korou LM, Perrea D, Pergialiotis V. An explanation of the pathophysiology of adverse neurodevelopmental outcomes in iron deficiency. Rev Neurosci 2015; 26:479-88. [PMID: 25951130 DOI: 10.1515/revneuro-2015-0012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/01/2015] [Indexed: 12/19/2022]
Abstract
Iron deficiency (ID) is a major public health problem worldwide among children aged 0-12 months. Several factors seem to contribute to the iron-deficient state in infancy, including insufficient antenatal and neonatal iron supplementation, exclusive breastfeeding, and early umbilical cord clamping after birth. The most concerning complications of ID, except for anemia, are related to altered long-term neurodevelopment. Clinical studies have shown a negative impact of ID anemia on fetal and neonatal behavior including impairments of motor maturity, autonomic response, memory/learning, and mood. ID-induced defects during infancy seem to persist later in life, even after ID treatment. The underlying mechanisms involve dysfunctional myelination, neurotransmission alterations, and altered synaptogenesis and/or dendritogenesis. The purpose of the present review is to summarize these mechanisms and to provide recommendations for future clinical research in the field.
Collapse
|
33
|
Choudhury V, Amin SB, Agarwal A, Srivastava LM, Soni A, Saluja S. Latent iron deficiency at birth influences auditory neural maturation in late preterm and term infants. Am J Clin Nutr 2015; 102:1030-4. [PMID: 26310540 DOI: 10.3945/ajcn.115.113084] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/03/2015] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND In utero latent iron deficiency has been associated with abnormal neurodevelopmental outcomes during childhood. Its concomitant effect on auditory neural maturation has not been well studied in late preterm and term infants. OBJECTIVE The objective was to determine whether in utero iron status is associated with auditory neural maturation in late preterm and term infants. DESIGN This prospective cohort study was performed at Sir Ganga Ram Hospital, New Delhi, India. Infants with a gestational age ≥34 wk were eligible unless they met the exclusion criteria: craniofacial anomalies, chromosomal disorders, hemolytic disease, multiple gestation, third-trimester maternal infection, chorioamnionitis, toxoplasmosis, other infections, rubella, cytomegalovirus infection, and herpes simplex virus infections (TORCH), Apgar score <5 at 5 min, sepsis, cord blood not collected, or auditory evaluation unable to be performed. Sixty consecutive infants with risk factors for iron deficiency, such as small for gestational age and maternal diabetes, and 30 without risk factors for iron deficiency were enrolled. Absolute wave latencies and interpeak latencies, evaluated by auditory brainstem response within 48 h after birth, were measured and compared between infants with latent iron deficiency (serum ferritin ≤75 ng/mL) and infants with normal iron status (serum ferritin >75 ng/mL) at birth. RESULTS Twenty-three infants had latent iron deficiency. Infants with latent iron deficiency had significantly prolonged wave V latencies (7.10 ± 0.68 compared with 6.60 ± 0.66), III-V interpeak latencies (2.37 ± 0.64 compared with 2.07 ± 0.33), and I-V interpeak latencies (5.10 ± 0.57 compared with 4.72 ± 0.56) compared with infants with normal iron status (P < 0.05). This difference remained significant on regression analyses after control for confounders. No difference was noted between latencies I and III and interpeak latencies I-III. CONCLUSION Latent iron deficiency is associated with abnormal auditory neural maturation in infants at ≥34 wk gestational age. This trial was registered at clinicaltrials.gov as NCT02503397.
Collapse
Affiliation(s)
| | - Sanjiv B Amin
- Department of Pediatrics, Division of Neonatology, University of Rochester, Rochester, NY
| | - Asha Agarwal
- Department of Otorhinology and Cochlear Implant Unit, and
| | - L M Srivastava
- Department of Biochemistry, Sir Ganga Ram Hospital, New Delhi, India; and
| | | | | |
Collapse
|
34
|
Huebner SM, Tran TD, Rufer ES, Crump PM, Smith SM. Maternal iron deficiency worsens the associative learning deficits and hippocampal and cerebellar losses in a rat model of fetal alcohol spectrum disorders. Alcohol Clin Exp Res 2015; 39:2097-107. [PMID: 26399568 DOI: 10.1111/acer.12876] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 08/09/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Gestational alcohol exposure causes lifelong physical and neurocognitive deficits collectively referred to as fetal alcohol spectrum disorders (FASDs). Micronutrient deficiencies are common in pregnancies of alcohol-abusing women. Here we show the most common micronutrient deficiency of pregnancy-iron deficiency without anemia-significantly worsens neurocognitive outcomes following perinatal alcohol exposure. METHODS Pregnant rats were fed iron-deficient (ID) or iron-sufficient diets from gestational day 13 to postnatal day (P) 7. Pups received alcohol (0, 3.5, 5.0 g/kg) from P 4 to P 9, targeting the brain growth spurt. At P 32, learning was assessed using delay or trace eyeblink classical conditioning (ECC). Cerebellar interpositus nucleus (IPN) and hippocampal CA1 cellularity was quantified using unbiased stereology. RESULTS Global analysis of variance revealed that ID and alcohol separately and significantly reduced ECC learning with respect to amplitude (ps ≤ 0.001) and conditioned response [CR] percentage (ps ≤ 0.001). Iron and alcohol interacted to reduce CR percentage in the trace ECC task (p = 0.013). Both ID and alcohol significantly reduced IPN (ps < 0.001) and CA1 cellularity (ps < 0.005). CR amplitude correlated with IPN cellularity (delay: r = 0.871, trace: r = 0.703, ps < 0.001) and CA1 cellularity (delay: r = 0.792, trace: r = 0.846, ps < 0.001) across both tasks. The learning impairments persisted even though the offsprings' iron status had normalized. CONCLUSIONS Supporting our previous work, gestational ID exacerbates the associative learning deficits in this rat model of FASD. This is strongly associated with cellular reductions within the ECC neurocircuitry. Significant learning impairments in FASD could be the consequence, in part, of pregnancies in which the mother was also iron inadequate.
Collapse
Affiliation(s)
- Shane M Huebner
- Departments of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Tuan D Tran
- Department of Psychology, Multidisciplinary Studies Program in Neuroscience, East Carolina University, Greenville, North Carolina
| | - Echoleah S Rufer
- Departments of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Peter M Crump
- Computing and Biometry, University of Wisconsin-Madison, Madison, Wisconsin
| | - Susan M Smith
- Departments of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
35
|
Abstract
The human brain undergoes a remarkable transformation during fetal life and the first postnatal years from a relatively undifferentiated but pluripotent organ to a highly specified and organized one. The outcome of this developmental maturation is highly dependent on a sequence of environmental exposures that can have either positive or negative influences on the ultimate plasticity of the adult brain. Many environmental exposures are beyond the control of the individual, but nutrition is not. An ever-increasing amount of research demonstrates not only that nutrition shapes the brain and affects its function during development but also that several nutrients early in life have profound and long-lasting effects on the brain. Nutrients have been shown to alter opening and closing of critical and sensitive periods of particular brain regions. This paper discusses the roles that various nutrients play in shaping the developing brain, concentrating specifically on recently explicated biological mechanisms by which particularly salient nutrients influence childhood and adult neural plasticity.
Collapse
|
36
|
Wattez JS, Delmont A, Bouvet M, Beseme O, Goers S, Delahaye F, Laborie C, Lesage J, Foligné B, Breton C, Metges CC, Vieau D, Pinet F. Maternal perinatal undernutrition modifies lactose and serotranferrin in milk: relevance to the programming of metabolic diseases? Am J Physiol Endocrinol Metab 2015; 308:E393-401. [PMID: 25550282 DOI: 10.1152/ajpendo.00452.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A close link between intrauterine growth restriction and development of chronic adult diseases such as obesity, diabetes, and hypertension has been established both in humans and animals. Modification of growth velocity during the early postnatal period (i.e., lactation) may also sensitize to the development of metabolic syndrome in adulthood. This suggests that milk composition may have long-lasting programming/deprogramming metabolic effects in the offspring. We therefore assess the effects of maternal perinatal denutrition on breast milk composition in a food-restricted 50% (FR50) rat model. Monosaccharides and fatty acids were characterized by gas chromatography, and proteins were profiled by surface-enhanced laser desorption/ionization-time-of-flight analysis in milk samples from FR50 and control rat dams. Milk analysis of FR50 rats demonstrated that maternal undernutrition decreases lactose concentration and modulates lipid profile at postnatal day 10 by increasing the unsaturated fatty acids/saturated fatty acids and diminishes serotransferrin levels at postnatal day 21. Our data indicate that maternal perinatal undernutrition modifies milk composition both quantitatively and qualitatively. These modifications by maternal nutrition open new perspectives to identify molecules that could be used in artificial milk to protect from the subsequent development of metabolic diseases.
Collapse
Affiliation(s)
- J S Wattez
- Environnement Périnatal et Croissance (EA4489), Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille, Villeneuve d'Ascq, France
| | - A Delmont
- Unité de Glycobiologie Structurale et Fonctionnelle (UMR8576), Université de Lille, Villeneuve d'Ascq, France
| | - M Bouvet
- Inserm U744, Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| | - O Beseme
- Inserm U744, Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| | - S Goers
- Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany; and
| | - F Delahaye
- Environnement Périnatal et Croissance (EA4489), Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille, Villeneuve d'Ascq, France
| | - C Laborie
- Environnement Périnatal et Croissance (EA4489), Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille, Villeneuve d'Ascq, France
| | - J Lesage
- Environnement Périnatal et Croissance (EA4489), Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille, Villeneuve d'Ascq, France
| | - B Foligné
- Lactic Acid Bacteria & Mucosal Immunity (U1019-UMR8204), Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
| | - C Breton
- Environnement Périnatal et Croissance (EA4489), Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille, Villeneuve d'Ascq, France
| | - C C Metges
- Institute of Nutritional Physiology "Oskar Kellner," Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany; and
| | - D Vieau
- Environnement Périnatal et Croissance (EA4489), Université Lille-Nord de France, Equipe Dénutritions Maternelles Périnatales, Université de Lille, Villeneuve d'Ascq, France;
| | - F Pinet
- Inserm U744, Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
| |
Collapse
|
37
|
Kennedy BC, Dimova JG, Siddappa AJM, Tran PV, Gewirtz JC, Georgieff MK. Prenatal choline supplementation ameliorates the long-term neurobehavioral effects of fetal-neonatal iron deficiency in rats. J Nutr 2014; 144:1858-65. [PMID: 25332485 PMCID: PMC4195423 DOI: 10.3945/jn.114.198739] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 07/12/2014] [Accepted: 08/21/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Gestational iron deficiency in humans and rodents produces long-term deficits in cognitive and socioemotional function and alters expression of plasticity genes in the hippocampus that persist despite iron treatment. Prenatal choline supplementation improves cognitive function in other rodent models of developmental insults. OBJECTIVE The objective of this study was to determine whether prenatal choline supplementation prevents the long-term effects of fetal-neonatal iron deficiency on cognitive and social behaviors and hippocampal gene expression. METHODS Pregnant rat dams were administered an iron-deficient (2-6 g/kg iron) or iron-sufficient (IS) (200 g/kg iron) diet from embryonic day (E) 3 to postnatal day (P) 7 with or without choline supplementation (5 g/kg choline chloride, E11-18). Novel object recognition (NOR) in the test vs. acquisition phase, social approach (SA), and hippocampal mRNA expression were compared at P65 in 4 male adult offspring groups: formerly iron deficient (FID), FID with choline supplementation (FID-C), IS, and IS with choline supplementation. RESULTS Relative to the intact NOR in IS rats (acquisition: 47.9%, test: 60.2%, P < 0.005), FID adult rats had impaired recognition memory at the 6-h delay (acquisition: 51.4%, test: 55.1%, NS), accompanied by a 15% reduction in hippocampal expression of brain-derived neurotrophic factor (Bdnf) (P < 0.05) and myelin basic protein (Mbp) (P < 0.05). Prenatal choline supplementation in FID rats restored NOR (acquisition: 48.8%, test: 64.4%, P < 0.0005) and increased hippocampal gene expression (FID-C vs. FID group: Bdnf, Mbp, P < 0.01). SA was also reduced in FID rats (P < 0.05 vs. IS rats) but was only marginally improved by prenatal choline supplementation. CONCLUSIONS Deficits in recognition memory, but not social behavior, resulting from gestational iron deficiency are attenuated by prenatal choline supplementation, potentially through preservation of hippocampal Bdnf and Mbp expression. Prenatal choline supplementation may be a promising adjunct treatment for fetal-neonatal iron deficiency.
Collapse
Affiliation(s)
- Bruce C Kennedy
- Graduate Program in Neuroscience, Center for Neurobehavioral Development,
| | | | - Asha J M Siddappa
- Center for Neurobehavioral Development, Department of Pediatrics, and
| | - Phu V Tran
- Center for Neurobehavioral Development, Department of Pediatrics, and
| | - Jonathan C Gewirtz
- Graduate Program in Neuroscience, Center for Neurobehavioral Development, Department of Psychology
| | - Michael K Georgieff
- Graduate Program in Neuroscience, Center for Neurobehavioral Development, Department of Pediatrics, and Institute of Child Development, University of Minnesota, Minneapolis, MN
| |
Collapse
|
38
|
Harvey L, Boksa P. Additive effects of maternal iron deficiency and prenatal immune activation on adult behaviors in rat offspring. Brain Behav Immun 2014; 40:27-37. [PMID: 24930842 DOI: 10.1016/j.bbi.2014.06.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/16/2014] [Accepted: 06/05/2014] [Indexed: 01/11/2023] Open
Abstract
Both iron deficiency (ID) and infection are common during pregnancy and studies have described altered brain development in offspring as a result of these individual maternal exposures. Given their high global incidence, these two insults may occur simultaneously during pregnancy. We recently described a rat model which pairs dietary ID during pregnancy and prenatal immune activation. Pregnant rats were placed on iron sufficient (IS) or ID diets from embryonic day 2 (E2) until postnatal day 7, and administered the bacterial endotoxin, lipopolysaccharide (LPS) or saline on E15/16. In this model, LPS administration on E15 caused greater induction of the pro-inflammatory cytokines, interleukin-6 and tumor necrosis factor-α, in ID dams compared to IS dams. This suggested that the combination of prenatal immune activation on a background of maternal ID might have more adverse neurodevelopmental consequences for the offspring than exposure to either insult alone. In this study we used this model to determine whether combined exposure to maternal ID and prenatal immune activation interact to affect juvenile and adult behaviors in the offspring. We assessed behaviors relevant to deficits in humans or animals that have been associated with exposure to either maternal ID or prenatal immune activation alone. Adult offspring from ID dams displayed significant deficits in pre-pulse inhibition of acoustic startle and in passive avoidance learning, together with increases in cytochrome oxidase immunohistochemistry, a marker of metabolic activity, in the ventral hippocampus immediately after passive avoidance testing. Offspring from LPS treated dams showed a significant increase in social behavior with unfamiliar rats, and subtle locomotor changes during exploration in an open field and in response to amphetamine. Surprisingly, there was no interaction between effects of the two insults on the behaviors assessed, and few observed alterations in juvenile behavior. Our findings show that long-term effects of maternal ID and prenatal LPS were additive, such that offspring exposed to both insults displayed more adult behavioral abnormalities than offspring exposed to one alone.
Collapse
Affiliation(s)
- Louise Harvey
- Department of Psychiatry, McGill University, Douglas Mental Health University Institute, 6875 La Salle Blvd, Verdun H4H 1R3, Quebec, Canada
| | - Patricia Boksa
- Department of Psychiatry, McGill University, Douglas Mental Health University Institute, 6875 La Salle Blvd, Verdun H4H 1R3, Quebec, Canada.
| |
Collapse
|
39
|
Duarte JM, Do KQ, Gruetter R. Longitudinal neurochemical modifications in the aging mouse brain measured in vivo by 1H magnetic resonance spectroscopy. Neurobiol Aging 2014; 35:1660-8. [DOI: 10.1016/j.neurobiolaging.2014.01.135] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 11/19/2013] [Accepted: 01/27/2014] [Indexed: 12/29/2022]
|
40
|
Greminger AR, Lee DL, Shrager P, Mayer-Pröschel M. Gestational iron deficiency differentially alters the structure and function of white and gray matter brain regions of developing rats. J Nutr 2014; 144:1058-66. [PMID: 24744313 PMCID: PMC4056646 DOI: 10.3945/jn.113.187732] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/12/2013] [Accepted: 03/21/2014] [Indexed: 11/14/2022] Open
Abstract
Gestational iron deficiency (ID) has been associated with a wide variety of central nervous system (CNS) impairments in developing offspring. However, a focus on singular regions has impeded an understanding of the CNS-wide effects of this micronutrient deficiency. Because the developing brain requires iron during specific phases of growth in a region-specific manner, we hypothesized that maternal iron deprivation would lead to region-specific impairments in the CNS of offspring. Female rats were fed an iron control (Fe+) or iron-deficient (Fe-) diet containing 240 or 6 μg/g iron during gestation and lactation. The corpus callosum (CC), hippocampus, and cortex of the offspring were analyzed at postnatal day 21 (P21) and/or P40 using structural and functional measures. In the CC at P40, ID was associated with reduced peak amplitudes of compound action potentials specific to myelinated axons, in which diameters were reduced by ∼20% compared with Fe+ controls. In the hippocampus, ID was associated with a 25% reduction in basal dendritic length of pyramidal neurons at P21, whereas branching complexity was unaffected. We also identified a shift toward increased proximal branching of apical dendrites in ID without an effect on overall length compared with Fe+ controls. ID also affected cortical neurons, but unlike the hippocampus, both apical and basal dendrites displayed a uniform decrease in branching complexity, with no significant effect on overall length. These deficits culminated in significantly poorer performance of P40 Fe- offspring in the novel object recognition task. Collectively, these results demonstrate that non-anemic gestational ID has a significant and region-specific impact on neuronal development and may provide a framework for understanding and recognizing the presentation of clinical symptoms of ID.
Collapse
Affiliation(s)
| | - Dawn L Lee
- Biomedical Genetics Pathology and Laboratory Medicine, and
| | - Peter Shrager
- Neurobiology and Anatomy, University of Rochester, Rochester, NY
| | | |
Collapse
|
41
|
Affiliation(s)
- Elizabeth L Prado
- Department of Nutrition; University of California at Davis; Davis CA USA
- SUMMIT Institute of Development; Mataram Nusa Tenggara Barat Indonesia
| | - Kathryn G Dewey
- Department of Nutrition; University of California at Davis; Davis CA USA
- SUMMIT Institute of Development; Mataram Nusa Tenggara Barat Indonesia
| |
Collapse
|
42
|
Harvey L, Boksa P. Do prenatal immune activation and maternal iron deficiency interact to affect neurodevelopment and early behavior in rat offspring? Brain Behav Immun 2014; 35:144-54. [PMID: 24064370 DOI: 10.1016/j.bbi.2013.09.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/27/2013] [Accepted: 09/12/2013] [Indexed: 11/17/2022] Open
Abstract
Infection and iron deficiency are common during pregnancy and studies have described altered brain development in the offspring as a result of these individual maternal exposures. Both exposures have been identified as risk factors for schizophrenia yet they have never been modeled simultaneously. We developed a rat model of prenatal immune activation on a background of maternal iron deficiency to determine whether these factors interact to affect neurodevelopment and early behavior in offspring. Pregnant rats were placed on iron sufficient (IS) or iron deficient (ID) diets from E2 to P7, and administered LPS or saline on E15/16. Iron was reduced in liver, spleen, serum and placenta from ID dams by E15. LPS administration on E15 caused greater induction of serum interleukin-6 and tumor necrosis factor-α in ID dams compared to IS dams. Offspring (P0, P7) from ID dams had reduced iron in spleen, liver and brain compared to IS, which normalized by P21. Pups from ID dams showed differences in forelimb grasp and acoustic startle, whilst pups from LPS dams displayed differences in grip ability, geotaxis reflex, cliff avoidance and acoustic startle. Offspring from LPS dams displayed reduced locomotor activity at P7 and P60; offspring from ID dams showed no change. Our findings show effects of prenatal LPS and maternal iron deficiency were additive, such that offspring exposed to both insults displayed more neurodevelopmental abnormalities than offspring exposed to one alone. Yet surprisingly there was no interaction between factors, suggesting independent mechanisms of action.
Collapse
Affiliation(s)
- Louise Harvey
- Department of Psychiatry, McGill University, Douglas Mental Health University Institute, 6875 La Salle Blvd, Verdun, H4H 1R3 Quebec, Canada
| | - Patricia Boksa
- Department of Psychiatry, McGill University, Douglas Mental Health University Institute, 6875 La Salle Blvd, Verdun, H4H 1R3 Quebec, Canada.
| |
Collapse
|
43
|
Tran PV, Dakoji S, Reise KH, Storey KK, Georgieff MK. Fetal iron deficiency alters the proteome of adult rat hippocampal synaptosomes. Am J Physiol Regul Integr Comp Physiol 2013; 305:R1297-306. [PMID: 24089371 DOI: 10.1152/ajpregu.00292.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fetal and neonatal iron deficiency results in cognitive impairments in adulthood despite prompt postnatal iron replenishment. To systematically determine whether abnormal expression and localization of proteins that regulate adult synaptic efficacy are involved, we used a quantitative proteomic approach (isobaric tags for relative and absolute quantitation, iTRAQ) and pathway analysis to identify dysregulated proteins in hippocampal synapses of fetal iron deficiency model. Rat pups were made iron deficient (ID) from gestational day 2 through postnatal day (P) 7 by providing pregnant and nursing dams an ID diet (4 ppm Fe) after which they were rescued with an iron-sufficient diet (200 ppm Fe). This paradigm resulted in a 40% loss of brain iron at P15 with complete recovery by P56. Synaptosomes were prepared from hippocampi of the formerly iron-deficient (FID) and always iron-sufficient controls rats at P65 using a sucrose gradient method. Six replicates per group that underwent iTRAQ labeling and LC-MS/MS analysis for protein identification and comparison elucidated 331 differentially expressed proteins. Western analysis was used to confirm findings for selected proteins in the glutamate receptor signaling pathway, which regulates hippocampal synaptic plasticity, a cellular process critical for learning and memory. Bioinformatics were performed using knowledge-based Interactive Pathway Analysis. FID synaptosomes show altered expression of synaptic proteins-mediated cellular signalings, supporting persistent impacts of fetal iron deficiency on synaptic efficacy, which likely cause the cognitive dysfunction and neurobehavioral abnormalities. Importantly, the findings uncover previously unsuspected pathways, including neuronal nitric oxide synthase signaling, identifying additional mechanisms that may contribute to the long-term biobehavioral deficits.
Collapse
Affiliation(s)
- Phu V Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | |
Collapse
|
44
|
Blegen MB, Kennedy BC, Thibert KA, Gewirtz JC, Tran PV, Georgieff MK. Multigenerational effects of fetal-neonatal iron deficiency on hippocampal BDNF signaling. Physiol Rep 2013; 1:e00096. [PMID: 24303168 PMCID: PMC3841032 DOI: 10.1002/phy2.96] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 08/29/2013] [Indexed: 01/15/2023] Open
Abstract
Fetal-neonatal iron deficiency induces adult learning impairments concomitant with changes in expression of key genes underlying hippocampal learning and memory in spite of neonatal iron replenishment. Notably, expression of brain-derived neurotrophic factor (BDNF), a gene critical for neuronal maturation and synaptic plasticity, is lowered both acutely and in adulthood following early-life iron deficiency. Although the mechanism behind its long-term downregulation remains unclear, epigenetic modification in BDNF, as seen in other models of early-life adversity, may play a role. Given that early iron deficiency occurs during critical periods in both hippocampal and gonadal development, we hypothesized that the iron-sufficient offspring (F2 IS) of formerly iron-deficient (F1 FID) rats would show a similar suppression of the BDNF gene as their parents. We compared hippocampal mRNA levels of BDNF and functionally related genes among F1 IS, F1 ID, and F2 IS male rats at postnatal day (P) 15 and P65 using RT-qPCR. As expected, the F1 ID group showed a downregulation of BDNF and associated genes acutely at P15 and chronically at P65. However, the F2 IS group showed an upregulation of these genes at P15, returning to control levels at P65. These results demonstrate that adverse effects of early iron deficiency on hippocampal gene expression observed in the F1 are not present in the F2 generation, suggesting differential effects of nutritionally induced epigenetic programing during the critical periods of hippocampal and gonadal development.
Collapse
Affiliation(s)
- Mariah B Blegen
- Department of Pediatrics, University of Minnesota Minneapolis, 55455, Minnesota
| | | | | | | | | | | |
Collapse
|
45
|
Pisansky MT, Wickham RJ, Su J, Fretham S, Yuan LL, Sun M, Gewirtz JC, Georgieff MK. Iron deficiency with or without anemia impairs prepulse inhibition of the startle reflex. Hippocampus 2013; 23:952-62. [PMID: 23733517 DOI: 10.1002/hipo.22151] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2013] [Indexed: 12/29/2022]
Abstract
Iron deficiency (ID) during early life causes long-lasting detrimental cognitive sequelae, many of which are linked to alterations in hippocampus function, dopamine synthesis, and the modulation of dopaminergic circuitry by the hippocampus. These same features have been implicated in the origins of schizophrenia, a neuropsychiatric disorder with significant cognitive impairments. Deficits in sensorimotor gating represent a reliable endophenotype of schizophrenia that can be measured by prepulse inhibition (PPI) of the acoustic startle reflex. Using two rodent model systems, we investigated the influence of early-life ID on PPI in adulthood. To isolate the role of hippocampal iron in PPI, our mouse model utilized a timed (embryonic day 18.5), hippocampus-specific knockout of Slc11a2, a gene coding an important regulator of cellular iron uptake, the divalent metal transport type 1 protein (DMT-1). Our second model used a classic rat dietary-based global ID during gestation, a condition that closely mimics human gestational ID anemia (IDA). Both models exhibited impaired PPI in adulthood. Furthermore, our DMT-1 knockout model displayed reduced long-term potentiation (LTP) and elevated paired-pulse facilitation (PPF), electrophysiological results consistent with previous findings in the IDA rat model. These results, in combination with previous findings demonstrating impaired hippocampus functioning and altered dopaminergic and glutamatergic neurotransmission, suggest that iron availability within the hippocampus is critical for the neurodevelopmental processes underlying sensorimotor gating. Ultimately, evidence of reduced PPI in both of our models may offer insights into the roles of fetal ID and the hippocampus in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Marc T Pisansky
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Ranade SC, Nawaz S, Chakrabarti A, Gressens P, Mani S. Spatial memory deficits in maternal iron deficiency paradigms are associated with altered glucocorticoid levels. Horm Behav 2013; 64:26-36. [PMID: 23631927 DOI: 10.1016/j.yhbeh.2013.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 11/22/2022]
Abstract
"The goal of this study was to examine the effect of maternal iron deficiency on the developing hippocampus in order to define a developmental window for this effect, and to see whether iron deficiency causes changes in glucocorticoid levels. The study was carried out using pre-natal, post-natal, and pre+post-natal iron deficiency paradigm. Iron deficient pregnant dams and their pups displayed elevated corticosterone which, in turn, differentially affected glucocorticoid receptor (GR) expression in the CA1 and the dentate gyrus. Brain Derived Neurotrophic Factor (BDNF) was reduced in the hippocampi of pups following elevated corticosterone levels. Reduced neurogenesis at P7 was seen in pups born to iron deficient mothers, and these pups had reduced numbers of hippocampal pyramidal and granule cells as adults. Hippocampal subdivision volumes also were altered. The structural and molecular defects in the pups were correlated with radial arm maze performance; reference memory function was especially affected. Pups from dams that were iron deficient throughout pregnancy and lactation displayed the complete spectrum of defects, while pups from dams that were iron deficient only during pregnancy or during lactation displayed subsets of defects. These findings show that maternal iron deficiency is associated with altered levels of corticosterone and GR expression, and with spatial memory deficits in their pups."
Collapse
Affiliation(s)
- Sayali C Ranade
- National Brain Research Centre, NH-8, Manesar, Haryana-122050, India.
| | | | | | | | | |
Collapse
|
47
|
Rao R, Tkac I, Unger EL, Ennis K, Hurst A, Schallert T, Connor J, Felt B, Georgieff MK. Iron supplementation dose for perinatal iron deficiency differentially alters the neurochemistry of the frontal cortex and hippocampus in adult rats. Pediatr Res 2013; 73:31-7. [PMID: 23095980 PMCID: PMC3563322 DOI: 10.1038/pr.2012.143] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Long-term prefrontal cortex (PFC)- and hippocampus-based cognitive deficits are the sequelae of perinatal iron deficiency, despite iron supplementation starting in the newborn period. Whether high-dose iron supplementation prevents these deficits is yet to be determined. METHODS Perinatal iron deficiency was induced in rat pups using a low-iron (3 mg/kg diet) diet during gestation until postnatal day (P)8. Iron was supplemented using a standard (40 mg/kg diet) or a 10-fold higher (400 mg/kg diet) iron-containing diet until P21. PFC and hippocampal neurochemistry was determined using in vivo (1)H nuclear magnetic resonance (NMR) spectroscopy at 9.4 Tesla on P90. RESULTS Both standard and 10-fold higher iron supplementation doses corrected anemia and brain iron deficiency by P21. The neurochemical profile of the PFC in both supplementation groups was comparable with the control group. In the hippocampus, standard-dose iron supplementation resulted in lower concentrations of N-acetylaspartate (NAA) and phosphoethanolamine (PE) and higher concentrations of N-acetylaspartylglutamate (NAAG) and glycerophosphocholine + phosphocholine (GPC + PC). High-dose iron supplementation resulted in lower PE and higher GPC + PC concentrations. CONCLUSION The iron supplementation dose for perinatal iron deficiency differentially alters the neurochemical profile of the PFC and hippocampus in adults. The neurochemical changes suggest altered glutamatergic neurotransmission, hypomyelination, and abnormal phospholipid metabolism in the formerly iron-deficient (FID) hippocampus.
Collapse
Affiliation(s)
- Raghavendra Rao
- Department of Pediatrics, Neonatology Division, University of Minnesota, Minneapolis, MN, USA.
| | - Ivan Tkac
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Erica L. Unger
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
| | - Kathleen Ennis
- Department of Pediatrics, Neonatology Division, University of Minnesota, Minneapolis, MN, USA
| | - Amy Hurst
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - Timothy Schallert
- Department of Psychology, University of Texas-Austin, Austin, TX, USA
| | - James Connor
- Department of Neurosurgery, Pennsylvania State University, Hershey, PA, USA
| | - Barbara Felt
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
| | - Michael K. Georgieff
- Department of Pediatrics, Neonatology Division, University of Minnesota, Minneapolis, MN, USA,Center for Neurobehavioral Development, University of Minnesota, Minneapolis MN, USA,Institute of Child Development, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
48
|
Metabolomic analysis of cerebrospinal fluid indicates iron deficiency compromises cerebral energy metabolism in the infant monkey. Neurochem Res 2012; 38:573-80. [PMID: 23269483 DOI: 10.1007/s11064-012-0950-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 12/14/2012] [Accepted: 12/17/2012] [Indexed: 01/05/2023]
Abstract
Iron deficiency anemia affects many pregnant women and young infants worldwide. The health impact is significant, given iron's known role in many body functions, including oxidative and lipid metabolism, protein synthesis and brain neurochemistry. The following research determined if (1)H NMR spectroscopy-based metabolomic analysis of cerebrospinal fluid (CSF) could detect the adverse influence of early life iron deficiency on the central nervous system. Using a controlled dietary model in 43 infant primates, distinct differences were found in spectra acquired at 600 MHz from the CSF of anemic monkeys. Three metabolite ratios, citrate/pyruvate, citrate/lactate and pyruvate/glutamine ratios, differed significantly in the iron deficient infant and then normalized following the consumption of dietary iron and improvement of clinical indices of anemia in the heme compartment. This distinctive metabolomic profile associated with anemia in the young infant indicates that CSF can be employed to track the neurological effects of iron deficiency and benefits of iron supplementation.
Collapse
|
49
|
Rytych JL, Elmore MRP, Burton MD, Conrad MS, Donovan SM, Dilger RN, Johnson RW. Early life iron deficiency impairs spatial cognition in neonatal piglets. J Nutr 2012; 142:2050-6. [PMID: 23014488 DOI: 10.3945/jn.112.165522] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Iron deficiency is common throughout the world and has been linked to cognitive impairments. Using neonatal piglets to model human infants, we assessed the impact of iron deficiency on spatial learning and memory. Artificially reared piglets were fed 1 of 3 liquid diets with varying concentrations of iron: control (CON), mildly deficient (MID), or severely deficient (SID; 100, 25.0, or 10.0 mg iron/kg milk solids, respectively) for 4 wk. Relative to CON, SID and MID piglets had reduced hemoglobin (P < 0.05) as well as magenta skin color (P < 0.001), which correlated with hematocrit (R(2) = 0.76; P < 0.001). SID and MID hemoglobin differed at wk 3 and 4 (P < 0.05). In a hippocampal-dependent, spatial, T-maze task, SID piglets were unable to acquire the task (post hoc contrast: first vs. last day of acquisition), while MID piglets demonstrated deficits in reversal learning (P = 0.032). Iron concentrations in the liver (P < 0.001), serum (P = 0.003), and hippocampus (P = 0.004), but not prefrontal cortex, were lower in MID and SID compared with CON piglets. The level of the transferrin receptor mRNA (TFR) was greater in the prefrontal cortex of CON piglets than in MID and SID piglets (P = 0.001) but not the hippocampus. Gene expression of several neurotrophic factors and proinflammatory cytokines, as well as whole-brain and hippocampal volume, were not affected by dietary treatment. In conclusion, neonatal iron deficiency leads to cognitive impairment, which may be due in part to a reduced iron concentration in the hippocampus.
Collapse
Affiliation(s)
- Jennifer L Rytych
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Patton SM, Coe CL, Lubach GR, Connor JR. Quantitative proteomic analyses of cerebrospinal fluid using iTRAQ in a primate model of iron deficiency anemia. Dev Neurosci 2012; 34:354-65. [PMID: 23018452 DOI: 10.1159/000341919] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 07/16/2012] [Indexed: 12/17/2022] Open
Abstract
Iron deficiency affects nearly 2 billion people worldwide, with pregnant women and young children being most severely impacted. Sustained anemia during the first year of life can cause cognitive, attention and motor deficits, which may persist despite iron supplementation. We conducted iTRAQ analyses on cerebrospinal fluid (CSF) from infant monkeys (Macaca mulatta) to identify differential protein expression associated with early iron deficiency. CSF was collected from 5 iron-sufficient and 8 iron-deficient anemic monkeys at weaning age (6-7 months) and again at 12-14 months. Despite consumption of iron-fortified food after weaning, which restored hematological indices into the normal range, expression of 5 proteins in the CSF remained altered. Most of the proteins identified are involved in neurite outgrowth, migration or synapse formation. The results reveal novel ways in which iron deficiency undermines brain growth and results in aberrant neuronal migration and connections. Taken together with gene expression data from rodent models of iron deficiency, we conclude that significant alterations in neuroconnectivity occur in the iron-deficient brain, which may persist even after resolution of the hematological anemia. The compromised brain infrastructure could account for observations of behavioral deficits in children during and after the period of anemia.
Collapse
|