1
|
Wade QW, Connor JR. What Does Iron Mean to an Oligodendrocyte? Glia 2025. [PMID: 40401729 DOI: 10.1002/glia.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025]
Abstract
Iron is essential for life and plays a key role in multiple fundamental cellular functions. The brain has the highest rate of energy consumption, and within the brain, oligodendrocytes have the highest level of oxidative metabolism per volume. Oligodendrocytes also stain the strongest for iron. The high requirement for iron is related to an oligodendrocyte's primary function to produce the myelin sheath, which requires iron as a cofactor. In addition to the high-energy demands that accompany the production of such dense and extensive membranous sheaths, iron is also required for lipid synthesis. Although the involvement of iron in oligodendrocyte functioning is clear, how iron is specifically acquired and utilized by oligodendrocytes is not completely understood. The purpose of this review is to provide a complete and thorough overview of the role of iron in oligodendrocytes. Here, we discuss in detail what is currently known about key iron transport proteins that participate in the balance of iron in oligodendrocytes. Understanding how oligodendrocytes utilize iron is beneficial in understanding dysmyelinating diseases, and the knowledge could be utilized to develop treatment options.
Collapse
Affiliation(s)
- Quinn W Wade
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - James R Connor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
2
|
Yoo JJ, Serafin EK, Baccei ML. Effects of perinatal iron deficiency on spinal dorsal horn circuits. THE JOURNAL OF PAIN 2025; 32:105434. [PMID: 40379071 DOI: 10.1016/j.jpain.2025.105434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/21/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
Clinical association studies have identified early life iron deficiency (ID) as a risk factor for the development of chronic pain. ID during the perinatal period has long-term consequences for the developing nervous system. Mounting evidence from both clinical and preclinical studies suggests that ID alters pain perception. However, nothing is yet known about how perinatal ID impacts nociceptive circuitry. The present study sought to characterize the effects of ID on the spinal superficial dorsal horn (SDH). Using ex vivo patch clamp electrophysiology in a mouse model of perinatal ID, the excitability of inhibitory and putative excitatory interneurons in the SDH was measured. It was found that early life ID did not significantly change the intrinsic excitability of either interneuron cell type in adolescence or adulthood. The investigation of synaptic inputs onto these two populations revealed that ID modulates spontaneous glutamatergic transmission within the SDH, but did not affect the excitatory drive or balance of synaptic excitation and inhibition. Interestingly, while ID altered the pattern of primary afferent inputs onto presumed glutamatergic interneurons in the mature SDH, the overall efficacy of these synapses was not affected by ID. Collectively, these results suggest that spinal nociceptive circuits are resilient to change following perinatal ID. PERSPECTIVE: This study demonstrates that perinatal iron deficiency (ID) elicits few changes to the intrinsic membrane excitability of superficial dorsal horn neurons or the efficacy of their synaptic inputs. These findings represent a critical first step towards elucidating the effects of ID on nociceptive processing in the central nervous system.
Collapse
Affiliation(s)
- Judy J Yoo
- Medical Scientist Training Program and Neuroscience Graduate Program, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Elizabeth K Serafin
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Mark L Baccei
- Medical Scientist Training Program and Neuroscience Graduate Program, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, 231 Albert Sabin Way, Cincinnati, OH 45267, USA.
| |
Collapse
|
3
|
Rugieł M, Setkowicz Z, Czyzycki M, Simon R, Baumbach T, Chwiej J. Element Changes Occurring in Brain Point at the White Matter Abnormalities in Rats Exposed to the Ketogenic Diet During Prenatal Life. ACS Chem Neurosci 2024; 15:3932-3944. [PMID: 39443296 PMCID: PMC11587514 DOI: 10.1021/acschemneuro.4c00283] [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: 05/06/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
A large number of clinical studies demonstrate that the ketogenic diet (KD) may be an effective approach to the reduction of epileptic seizures in children and adults. Such dietary therapy could also help pregnant women with epilepsy, especially since most antiseizure drugs have teratogenic action. However, there is a lack of medical data, considering the safety of using KD during gestation for the progeny. Therefore, we examined the influence of KD used prenatally in rats on the elemental composition of the selected brain regions in their offspring. For this purpose, synchrotron radiation-induced X-ray fluorescence (SR-XRF) microscopy was utilized, and elements such as P, S, K, Ca, Fe, and Zn were determined. Moreover, to verify whether the possible effects of KD are temporary or long-term, different stages of animal postnatal development were taken into account in our experiment. The obtained results confirmed the great applicability of SR-XRF microscopy to track the element changes occurring in the brain during postnatal development as well as those induced by prenatal exposure to the high-fat diet. The topographic analysis of the brains taken from offspring of mothers fed with KD during pregnancy and appropriate control individuals showed a potential influence of such dietary treatment on the brain levels of elements such as P and S. In the oldest progeny, a significant reduction of the surface of brain areas characterized by an increased P and S content, which histologically/morphologically correspond to white matter structures, was noticed. In turn, quantitative elemental analysis showed significantly decreased levels of Fe in the striatum and white matter of 30-day-old rats exposed prenatally to KD. This effect was temporary and was not noticed in adult animals. The observed abnormalities may be related to the changes in the accumulation of sphingomyelin and sulfatides and may testify about disturbances in the structure and integrity of the myelin, present in the white matter.
Collapse
Affiliation(s)
- Marzena Rugieł
- Faculty of
Physics and Applied Computer Science, AGH
University of Krakow, Al. Mickiewicza 30, Krakow 30-059, Poland
| | - Zuzanna Setkowicz
- Institute
of Zoology and Biomedical Research, Jagiellonian
University, Gronostajowa
9, Krakow 30-387, Poland
| | - Mateusz Czyzycki
- Institute
for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen D-76344, Germany
| | - Rolf Simon
- Institute
for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen D-76344, Germany
| | - Tilo Baumbach
- Institute
for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen D-76344, Germany
- Laboratory
for Applications of Synchrotron Radiation, Karlsruhe Institute of Technology, Kaiserstr. 12, Karlsruhe D-76131, Germany
| | - Joanna Chwiej
- Faculty of
Physics and Applied Computer Science, AGH
University of Krakow, Al. Mickiewicza 30, Krakow 30-059, Poland
| |
Collapse
|
4
|
Ipsiroglu OS, Pandher PK, Hill O, McWilliams S, Braschel M, Edwards K, Friedlander R, Keys E, Kuo C, Lewis MS, Richardson A, Wagner AL, Wensley D. Iron Deficiency and Restless Sleep/Wake Behaviors in Neurodevelopmental Disorders and Mental Health Conditions. Nutrients 2024; 16:3064. [PMID: 39339663 PMCID: PMC11435348 DOI: 10.3390/nu16183064] [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: 07/06/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/30/2024] Open
Abstract
Iron deficiency (ID) and restlessness are associated with sleep/wake-disorders (e.g., restless legs syndrome (RLS)) and neurodevelopmental disorders (attention deficit/hyperactivity and autism spectrum disorders (ADHD; ASD)). However, a standardized approach to assessing ID and restlessness is missing. We reviewed iron status and family sleep/ID history data collected at a sleep/wake behavior clinic under a quality improvement/quality assurance project. Restlessness was explored through patient and parental narratives and a 'suggested clinical immobilization test'. Of 199 patients, 94% had ID, with 43% having a family history of ID. ADHD (46%) and ASD (45%) were common conditions, along with chronic insomnia (61%), sleep-disordered breathing (50%), and parasomnias (22%). In unadjusted analysis, a family history of ID increased the odds (95% CI) of familial RLS (OR: 5.98, p = 0.0002, [2.35-15.2]), insomnia/DIMS (OR: 3.44, p = 0.0084, [1.37-8.64]), and RLS (OR: 7.00, p = 0.01, [1.49-32.93]) in patients with ADHD, and of insomnia/DIMS (OR: 4.77, p = 0.0014, [1.82-12.5]), RLS/PLMS (OR: 5.83, p = 0.009, [1.54-22.1]), RLS (OR: 4.05, p = 0.01, [1.33-12.3]), and familial RLS (OR: 2.82, p = 0.02, [1.17-6.81]) in patients with ASD. ID and restlessness were characteristics of ADHD and ASD, and a family history of ID increased the risk of sleep/wake-disorders. These findings highlight the need to integrate comprehensive blood work and family history to capture ID in children and adolescents with restless behaviors.
Collapse
Affiliation(s)
- Osman S Ipsiroglu
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
- Divisions of Developmental Pediatrics, Child and Adolescent Psychiatry, and Respirology, BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Parveer K Pandher
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Olivia Hill
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Scout McWilliams
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Melissa Braschel
- Clinical Research Support Unit, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Katherine Edwards
- Division of Child and Adolescent Psychiatry, BC Children's Hospital, Department of Psychiatry, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Robin Friedlander
- Division of Child and Adolescent Psychiatry, BC Children's Hospital, Department of Psychiatry, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Elizabeth Keys
- School of Nursing, University of British Columbia (Okanagan), Kelowna, BC V1V 1V7, Canada
| | - Calvin Kuo
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marion Suzanne Lewis
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Pacific Autism Family Network, Richmond, BC V7B 1C7, Canada
| | - Anamaria Richardson
- Granville Pediatrics, 205-5678 Granville Street, Vancouver, BC V6M 1X7, Canada
| | - Alexandra L Wagner
- Department of Pediatrics, Division of Neurology, Charité University Hospital Berlin, 10117 Berlin, Germany
| | - David Wensley
- Division of Respirology, BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| |
Collapse
|
5
|
Liu SX, Calixto Mancipe N, Gisslen T, Georgieff MK, Tran PV. Identification of Genes Responding to Iron or Choline Treatment for Early-Life Iron Deficiency in the Male Rat Hippocampal Transcriptomes. J Nutr 2024; 154:1141-1152. [PMID: 38408730 PMCID: PMC11007743 DOI: 10.1016/j.tjnut.2024.02.021] [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: 12/07/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Developmental iron deficiency (ID) is associated with long-term cognitive and affective behavioral impairments in humans. Preclinical studies have shown that developmental ID has short- and long-term effects on gene regulation. Prenatal choline supplementation partially rescues early-life ID-induced cognitive deficits in adult male rats. OBJECTIVES To identify acute and long-term changes in biological processes regulated by developmental ID and modifiable by choline. METHODS This study compares the hippocampal transcriptomes of postnatal day (P) 15 iron-deficient (acute) and P65 formerly ID (persistent) rats with or without prenatal choline treatment. Pregnant rats were fed an ID (4 mg/kg Fe) or iron-sufficient (IS) (200 mg/kg Fe) diet from gestational day (G) 2 to P7 with or without choline supplementation (5 g/kg choline) from G11 to G18. Hippocampi were collected from P15 or P65 offspring and analyzed for gene expression by RNA sequencing. RESULTS Developmental ID-induced changes suggested modified activity of oxidative phosphorylation and fatty acid metabolism. Prenatal choline supplementation induced robust changes in gene expression, particularly in iron-deficient animals, where it partially mitigated the early-life ID-dysregulated genes. Choline supplementation also altered the hippocampal transcriptome in the IS rats, with indications for both beneficial and adverse effects. CONCLUSIONS This study provided global assessments of gene expression regulated by iron and choline. Our new findings highlight genes responding to iron or choline treatments, including a potentially novel choline-regulated transporter (IPO7), with shared effects on neuroinflammation in the male rat hippocampus.
Collapse
Affiliation(s)
- Shirelle X Liu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Natalia Calixto Mancipe
- Research Informatic Solutions, Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, United States
| | - Tate Gisslen
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Michael K Georgieff
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
6
|
Liu SX, Ramakrishnan A, Shen L, Gewirtz JC, Georgieff MK, Tran PV. Chromatin accessibility and H3K9me3 landscapes reveal long-term epigenetic effects of fetal-neonatal iron deficiency in rat hippocampus. BMC Genomics 2024; 25:301. [PMID: 38515015 PMCID: PMC10956188 DOI: 10.1186/s12864-024-10230-4] [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: 08/02/2023] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Iron deficiency (ID) during the fetal-neonatal period results in long-term neurodevelopmental impairments associated with pervasive hippocampal gene dysregulation. Prenatal choline supplementation partially normalizes these effects, suggesting an interaction between iron and choline in hippocampal transcriptome regulation. To understand the regulatory mechanisms, we investigated epigenetic marks of genes with altered chromatin accessibility (ATAC-seq) or poised to be repressed (H3K9me3 ChIP-seq) in iron-repleted adult rats having experienced fetal-neonatal ID exposure with or without prenatal choline supplementation. RESULTS Fetal-neonatal ID was induced by limiting maternal iron intake from gestational day (G) 2 through postnatal day (P) 7. Half of the pregnant dams were given supplemental choline (5.0 g/kg) from G11-18. This resulted in 4 groups at P65 (Iron-sufficient [IS], Formerly Iron-deficient [FID], IS with choline [ISch], and FID with choline [FIDch]). Hippocampi were collected from P65 iron-repleted male offspring and analyzed for chromatin accessibility and H3K9me3 enrichment. 22% and 24% of differentially transcribed genes in FID- and FIDch-groups, respectively, exhibited significant differences in chromatin accessibility, whereas 1.7% and 13% exhibited significant differences in H3K9me3 enrichment. These changes mapped onto gene networks regulating synaptic plasticity, neuroinflammation, and reward circuits. Motif analysis of differentially modified genomic sites revealed significantly stronger choline effects than early-life ID and identified multiple epigenetically modified transcription factor binding sites. CONCLUSIONS This study reveals genome-wide, stable epigenetic changes and epigenetically modifiable gene networks associated with specific chromatin marks in the hippocampus, and lays a foundation to further elucidate iron-dependent epigenetic mechanisms that underlie the long-term effects of fetal-neonatal ID, choline, and their interactions.
Collapse
Affiliation(s)
- Shirelle X Liu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Psychology, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Li Shen
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jonathan C Gewirtz
- Department of Psychology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Michael K Georgieff
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Phu V Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
7
|
Reid B, East P, Blanco E, Doom J, Burrows R, Correa-Burrows P, Lozoff B, Gahagan S. Early-life adversity is associated with poor iron status in infancy. Dev Psychopathol 2023; 35:1856-1867. [PMID: 35678178 PMCID: PMC9732147 DOI: 10.1017/s0954579422000517] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exposure to early-life adversity (ELA) and iron deficiency early in life are known risk factors for suboptimal brain and socioemotional development. Iron deficiency may arise from and co-occur with ELA, which could negatively affect development. In the present study, we investigated whether ELA is associated with iron deficiency in infants receiving no iron supplementation. This study is a secondary analysis of extant data collected in the 1990s; participants were healthy infants from working-class communities in Santiago, Chile (N = 534, 45.5% female). We measured stressful life events, maternal depression, and low home support for child development during infancy and assessed iron status when the infant was 12 months old. Slightly more than half of the infants were iron-deficient (51%), and 25.8% were iron-deficient anemic at 12 months. Results indicated that ELA was associated with lower iron levels and iron deficiency at 12 months. The findings are consistent with animal and human prenatal models of stress and iron status and provide evidence of the association between postnatal ELA and iron status in humans. The findings also highlight a nutritional pathway by which ELA may impact development and present a nutritionally-focused avenue for future research on ELA and psychopathology.
Collapse
Affiliation(s)
- B.M. Reid
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI
- Center for Behavioral and Preventive Medicine, The Miriam Hospital, Providence, RI
| | - P. East
- Department of Pediatrics, University of California, San Diego
| | - E. Blanco
- Department of Public Health, School of Medicine, Pontificia Universidad Católica de Chile
| | - J.R. Doom
- Department of Psychology, University of Denver
| | - R.A. Burrows
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - P. Correa-Burrows
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - B. Lozoff
- Department of Pediatrics, University of Michigan, Ann Arbor
| | - S Gahagan
- Department of Pediatrics, University of California, San Diego
| |
Collapse
|
8
|
Reid BM, Georgieff MK. The Interaction between Psychological Stress and Iron Status on Early-Life Neurodevelopmental Outcomes. Nutrients 2023; 15:3798. [PMID: 37686831 PMCID: PMC10490173 DOI: 10.3390/nu15173798] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
This review presents evidence from animal and human studies demonstrating the possible connection and significant impact of poor iron status and psychological distress on neurocognitive development during pregnancy and the neonatal period, with implications for long-term cognition. Stress and iron deficiency are independently prevalent and thus are frequently comorbid. While iron deficiency and early-life stress independently contribute to long-term neurodevelopmental alterations, their combined effects remain underexplored. Psychological stress responses may engage similar pathways as infectious stress, which alters fundamental iron metabolism processes and cause functional tissue-level iron deficiency. Psychological stress, analogous to but to a lesser degree than infectious stress, activates the hypothalamic-pituitary-adrenocortical (HPA) axis and increases proinflammatory cytokines. Chronic or severe stress is associated with dysregulated HPA axis functioning and a proinflammatory state. This dysregulation may disrupt iron absorption and utilization, likely mediated by the IL-6 activation of hepcidin, a molecule that impedes iron absorption and redistributes total body iron. This narrative review highlights suggestive studies investigating the relationship between psychological stress and iron status and outlines hypothesized mechanistic pathways connecting psychological stress exposure and iron metabolism. We examine findings regarding the overlapping impacts of early stress exposure to iron deficiency and children's neurocognitive development. We propose that studying the influence of psychological stress on iron metabolism is crucial for comprehending neurocognitive development in children exposed to prenatal and early postnatal stressors and for children at risk of early iron insufficiency. We recommend future directions for dual-exposure studies exploring iron as a potential mediating pathway between early stress and offspring neurodevelopment, offering opportunities for targeted interventions.
Collapse
Affiliation(s)
- Brie M. Reid
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Center for Behavioral and Preventive Medicine, The Miriam Hospital, Providence, RI 02906, USA
| | - Michael K. Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| |
Collapse
|
9
|
Larsen B, Baller EB, Boucher AA, Calkins ME, Laney N, Moore TM, Roalf DR, Ruparel K, Gur RC, Gur RE, Georgieff MK, Satterthwaite TD. Development of Iron Status Measures during Youth: Associations with Sex, Neighborhood Socioeconomic Status, Cognitive Performance, and Brain Structure. Am J Clin Nutr 2023; 118:121-131. [PMID: 37146760 PMCID: PMC10375461 DOI: 10.1016/j.ajcnut.2023.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/24/2023] [Accepted: 05/01/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Iron is essential to brain function, and iron deficiency during youth may adversely impact neurodevelopment. Understanding the developmental time course of iron status and its association with neurocognitive functioning is important for identifying windows for intervention. OBJECTIVES This study aimed to characterize developmental change in iron status and understand its association with cognitive performance and brain structure during adolescence using data from a large pediatric health network. METHODS This study included a cross-sectional sample of 4899 participants (2178 males; aged 8-22 y at the time of participation, M [SD] = 14.24 [3.7]) who were recruited from the Children's Hospital of Philadelphia network. Prospectively collected research data were enriched with electronic medical record data that included hematological measures related to iron status, including serum hemoglobin, ferritin, and transferrin (33,015 total samples). At the time of participation, cognitive performance was assessed using the Penn Computerized Neurocognitive Battery, and brain white matter integrity was assessed using diffusion-weighted MRI in a subset of individuals. RESULTS Developmental trajectories were characterized for all metrics and revealed that sex differences emerged after menarche such that females had reduced iron status relative to males [all R2partial > 0.008; all false discovery rates (FDRs) < 0.05]. Higher socioeconomic status was associated with higher hemoglobin concentrations throughout development (R2partial = 0.005; FDR < 0.001), and the association was greatest during adolescence. Higher hemoglobin concentrations were associated with better cognitive performance during adolescence (R2partial = 0.02; FDR < 0.001) and mediated the association between sex and cognition (mediation effect = -0.107; 95% CI: -0.191, -0.02). Higher hemoglobin concentration was also associated with greater brain white matter integrity in the neuroimaging subsample (R2partial = 0.06, FDR = 0.028). CONCLUSIONS Iron status evolves during youth and is lowest in females and individuals of low socioeconomic status during adolescence. Diminished iron status during adolescence has consequences for neurocognition, suggesting that this critical period of neurodevelopment may be an important window for intervention that has the potential to reduce health disparities in at-risk populations.
Collapse
Affiliation(s)
- Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States.
| | - Erica B Baller
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Alexander A Boucher
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Monica E Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Nina Laney
- Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Tyler M Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - David R Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Kosha Ruparel
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Ruben C Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States; Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Raquel E Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States; Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael K Georgieff
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Theodore D Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, United States; Penn/Children's Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
10
|
Gao G, You L, Zhang J, Chang YZ, Yu P. Brain Iron Metabolism, Redox Balance and Neurological Diseases. Antioxidants (Basel) 2023; 12:1289. [PMID: 37372019 DOI: 10.3390/antiox12061289] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The incidence of neurological diseases, such as Parkinson's disease, Alzheimer's disease and stroke, is increasing. An increasing number of studies have correlated these diseases with brain iron overload and the resulting oxidative damage. Brain iron deficiency has also been closely linked to neurodevelopment. These neurological disorders seriously affect the physical and mental health of patients and bring heavy economic burdens to families and society. Therefore, it is important to maintain brain iron homeostasis and to understand the mechanism of brain iron disorders affecting reactive oxygen species (ROS) balance, resulting in neural damage, cell death and, ultimately, leading to the development of disease. Evidence has shown that many therapies targeting brain iron and ROS imbalances have good preventive and therapeutic effects on neurological diseases. This review highlights the molecular mechanisms, pathogenesis and treatment strategies of brain iron metabolism disorders in neurological diseases.
Collapse
Affiliation(s)
- Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Linhao You
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Jianhua Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Peng Yu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| |
Collapse
|
11
|
Liu SX, Fredrickson TK, Calixto Mancipe N, Georgieff MK, Tran PV. Sex-Specific Effects of Early-Life Iron Deficiency and Prenatal Choline Treatment on Adult Rat Hippocampal Transcriptome. Nutrients 2023; 15:nu15061316. [PMID: 36986048 PMCID: PMC10055746 DOI: 10.3390/nu15061316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/04/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Background: Fetal-neonatal iron deficiency (ID) causes long-term neurocognitive and affective dysfunctions. Clinical and preclinical studies have shown that early-life ID produces sex-specific effects. However, little is known about the molecular mechanisms underlying these early-life ID-induced sex-specific effects on neural gene regulation. Objective: To illustrate sex-specific transcriptome alterations in adult rat hippocampus induced by fetal-neonatal ID and prenatal choline treatment. Methods: Pregnant rats were fed an iron-deficient (4 mg/kg Fe) or iron-sufficient (200 mg/kg Fe) diet from gestational day (G) 2 to postnatal day (P) 7 with or without choline supplementation (5 g/kg choline) from G11–18. Hippocampi were collected from P65 offspring of both sexes and analyzed for changes in gene expression. Results: Both early-life ID and choline treatment induced transcriptional changes in adult female and male rat hippocampi. Both sexes showed ID-induced alterations in gene networks leading to enhanced neuroinflammation. In females, ID-induced changes indicated enhanced activity of oxidative phosphorylation and fatty acid metabolism, which were contrary to the ID effects in males. Prenatal choline supplementation induced the most robust changes in gene expression, particularly in iron-deficient animals where it partially rescued ID-induced dysregulation. Choline supplementation also altered hippocampal transcriptome in iron-sufficient rats with indications for both beneficial and adverse effects. Conclusions: This study provided unbiased global assessments of gene expression regulated by iron and choline in a sex-specific manner, with greater effects in female than male rats. Our new findings highlight potential sex-specific gene networks regulated by iron and choline for further investigation.
Collapse
Affiliation(s)
- Shirelle X. Liu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Natalia Calixto Mancipe
- Research Informatic Solutions, Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Phu V. Tran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence: ; Tel.: +01-612-626-7964
| |
Collapse
|
12
|
Azupogo F, Saeed N, Wemakor A, Addae HY, Boah M, Brouwer ID. Moderate-to-severe household food insecurity is associated with depression among adolescent girls in northern Ghana: a cross-sectional analysis. BMJ Nutr Prev Health 2023; 6:56-64. [PMID: 37559964 PMCID: PMC10407409 DOI: 10.1136/bmjnph-2022-000523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/15/2022] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Household food insecurity is positively associated with depression; however, the association among adolescents is not well known. We examined the association between household food insecurity and depression among adolescent girls in Northern Ghana. METHODS We analysed data from the Ten2Twenty-Ghana randomised controlled trial end-line for adolescent girls aged 10-17 years (n=582). The girls were chosen at random from 19 primary schools in the Mion District of Ghana's northern region. The children's depression inventory and the Food Insecurity Experience Scale were used in face-to-face interviews to assess depression and household food insecurity. Hierarchical survey binary logistic regression and linear mixed models were used to examine the association between household food insecurity and depressive symptoms. We took into account a number of potential confounders in the analysis, such as life satisfaction, self-efficacy, self-esteem, health complaints, child's age, menarche status, pubertal development, anaemia, stunting, frequency of consuming fruits and vegetables, frequency of consuming animal-sourced foods, maternal age, household wealth index and size, and the intervention group the girl was assigned to in the trial. RESULTS About 20.1% of adolescent girls were classified as likely depressed, and 70.3% of their households were food insecure, with 22.9% and 18.0% being moderately and severely food insecure, respectively. Compared with girls from food-secure households, those from moderately (adjusted OR (AOR) 2.63, 95% CI (1.35 to 5.12)) and severely (AOR 3.28, 95% CI (1.66 to 6.49)) food insecure households had about three times the odds of being classified as depressed, after controlling for potential confounders. The odds of being likely depressed were about twice for adolescent girls from food-insecure households compared with their peers from food-secure households in both the crude and final adjusted model. CONCLUSION The study discovered high levels of household food insecurity and depression in adolescent girls in Northern Ghana, with a dose-response association between the two.
Collapse
Affiliation(s)
- Fusta Azupogo
- Department of Family and Consumer Sciences, University for Development Studies, Tamale, Ghana
| | - Nawaf Saeed
- Department of Nutritional Sciences, University for Development Studies, Tamale, Ghana
| | - Anthony Wemakor
- Department of Nutritional Sciences, University for Development Studies, Tamale, Ghana
| | - Hammond Yaw Addae
- Nursing, Nursing & Midwifery Training Colege, Kpembe, Salaga, Ghana
- Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Michael Boah
- Department of Epidemiology, Biostatistics, and Disease Control, School of Public Health, University for Development Studies, Tamale, Ghana
| | - Inge D Brouwer
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
13
|
Silvani A, Ghorayeb I, Manconi M, Li Y, Clemens S. Putative Animal Models of Restless Legs Syndrome: A Systematic Review and Evaluation of Their Face and Construct Validity. Neurotherapeutics 2023; 20:154-178. [PMID: 36536233 PMCID: PMC10119375 DOI: 10.1007/s13311-022-01334-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Restless legs syndrome (RLS) is a sensorimotor disorder that severely affects sleep. It is characterized by an urge to move the legs, which is often accompanied by periodic limb movements during sleep. RLS has a high prevalence in the population and is usually a life-long condition. While its origins remain unclear, RLS is initially highly responsive to treatment with dopaminergic agonists that target D2-like receptors, in particular D2 and D3, but the long-term response is often unsatisfactory. Over the years, several putative animal models for RLS have been developed, mainly based on the epidemiological and neurochemical link with iron deficiency, treatment efficacy of D2-like dopaminergic agonists, or genome-wide association studies that identified risk factors in the patient population. Here, we present the first systematic review of putative animal models of RLS, provide information about their face and construct validity, and report their role in deciphering the underlying pathophysiological mechanisms that may cause or contribute to RLS. We propose that identifying the causal links between genetic risk factors, altered organ functions, and changes to molecular pathways in neural circuitry will eventually lead to more effective new treatment options that bypass the side effects of the currently used therapeutics in RLS, especially for long-term therapy.
Collapse
Affiliation(s)
- Alessandro Silvani
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Ravenna Campus, Ravenna, Italy
| | - Imad Ghorayeb
- Département de Neurophysiologie Clinique, Pôle Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, Université de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, CNRS, Bordeaux, France
| | - Mauro Manconi
- Sleep Medicine Unit, Neurocenter of Southern Switzerland, EOC, Ospedale Civico, Lugano, Switzerland
- Department of Neurology, University Hospital, Inselspital, Bern, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Yuqing Li
- Department of Neurology, College of Medicine, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
14
|
Prevalence and Implications of Low Reticulocyte-Hemoglobin Levels among Extreme Preterm Neonates: A Single-Center Retrospective Study. Nutrients 2022; 14:nu14245343. [PMID: 36558502 PMCID: PMC9788547 DOI: 10.3390/nu14245343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
This retrospective cohort study aims to determine the epidemiology of iron deficiency among extreme preterm neonates and the association of iron-deficient status during the NICU stay with neurodevelopmental outcomes at 18−24 months. Neonates ≤29 weeks gestational age (GA) born between June 2016 and December 2019, who received routine iron supplementation were enrolled. Iron deficiency was defined as reticulocyte−hemoglobin (Ret-Hb) levels ≤ 29 pg at 36 weeks corrected age. A subcohort of neonates completed standardized developmental assessment at 18−24 months corrected age. Significant neurodevelopmental impairment (sNDI) was defined as either Bayley Scales of Infant Development score < 70 or cerebral palsy or blindness or hearing aided. Among a cohort of 215 neonates [GA 25.8 (1.7) weeks, birthweight 885 (232) g], prevalence of iron deficiency was 55%, 21%, 26%, and 13%, in neonates <24 weeks, 24−25 + 6 weeks, 26−27 + 6 weeks, and ≥ 28 weeks GA, respectively. Male sex and receipt of corticosteroid therapy were associated with iron-deficiency. In the subcohort analysis (n = 69), there was no statistically significant association between Ret-Hb levels at 36 weeks corrected age and the risk of sNDI [OR 0.99 (95% CI 0.85−1.2)]. Male infants and those who received postnatal corticosteroids are likely to have iron-limited erythropoiesis at corrected term despite routine iron-supplementation; however, low Ret-Hb levels during the neonatal period were not associated with significant neurological disability in early childhood.
Collapse
|
15
|
Yuan Y, Wang Y, Wu S, Zhao MY. Review: Myelin clearance is critical for regeneration after peripheral nerve injury. Front Neurol 2022; 13:908148. [PMID: 36588879 PMCID: PMC9801717 DOI: 10.3389/fneur.2022.908148] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022] Open
Abstract
Traumatic peripheral nerve injury occurs frequently and is a major clinical and public health problem that can lead to functional impairment and permanent disability. Despite the availability of modern diagnostic procedures and advanced microsurgical techniques, active recovery after peripheral nerve repair is often unsatisfactory. Peripheral nerve regeneration involves several critical events, including the recreation of the microenvironment and remyelination. Results from previous studies suggest that the peripheral nervous system (PNS) has a greater capacity for repair than the central nervous system. Thus, it will be important to understand myelin and myelination specifically in the PNS. This review provides an update on myelin biology and myelination in the PNS and discusses the mechanisms that promote myelin clearance after injury. The roles of Schwann cells and macrophages are considered at length, together with the possibility of exogenous intervention.
Collapse
Affiliation(s)
- YiMing Yuan
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan Wang
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China,Department of Rehabilitation, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China,*Correspondence: Yan Wang
| | - ShanHong Wu
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ming Yue Zhao
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China,Department of Rehabilitation, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
16
|
Ferroptosis inhibition by deferiprone, attenuates myelin damage and promotes neuroprotection in demyelinated optic nerve. Sci Rep 2022; 12:19630. [PMID: 36385152 PMCID: PMC9668997 DOI: 10.1038/s41598-022-24152-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease, which leads to focal demyelination in the brain and spinal cord. Studies showed that iron released during the course of myelin breakdown exacerbates tissue damage, which is in agreement with the features of iron-dependent cell death, ferroptosis. Here, we aimed to investigate the possible contribution of ferroptosis in the demyelinated optic nerve, and to explore the effectiveness of ferroptosis inhibitor, deferiprone (DFP), on the extent of demyelination, inflammation and axonal damage. For this purpose, focal demyelination was induced by injection of lysolecithin (LPC), into the optic nerve of male C57BL/6J mice. Afterward, optic nerves were harvested at different time points from as early as 6 h up to 7 days post-LPC injection. Next, to evaluate the effectiveness of DFP two groups of animals received daily intraperitoneal injection of DFP for 3 or 7 continuous days. Vehicle groups received saline. Iron deposition was observed at different time points post-LPC injection from 6 h to 7 days post injection. Examining ferroptosis markers showed a significant reduction in glutathione content along with increased level of malondialdehyde and upregulated ferroptosis marker genes at early time points after injection. Besides, DFP treatment during the inflammatory phase of the model resulted in decreased microgliosis and inflammation. Reduced demyelination, microgliosis and astrogliosis was shown in mice that received DFP for 7 days. Moreover, DFP protected against axonal damage and retinal ganglion cells loss. Our results suggest the possible contribution of ferroptosis pathway in the process of demyelination. The therapeutic strategies targeting iron deposition, e.g. DFP treatment might thus represent a promising therapeutic target for patients with MS.
Collapse
|
17
|
Nampijja M, Mutua AM, Elliott AM, Muriuki JM, Abubakar A, Webb EL, Atkinson SH. Low Hemoglobin Levels Are Associated with Reduced Psychomotor and Language Abilities in Young Ugandan Children. Nutrients 2022; 14:nu14071452. [PMID: 35406065 PMCID: PMC9002834 DOI: 10.3390/nu14071452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/10/2022] Open
Abstract
Children living in Sub-Saharan Africa are vulnerable to developmental delay, particularly in the critical first five years due to various adverse exposures including disease and nutritional deficiencies. Anemia and iron deficiency (ID) are highly prevalent in pregnant mothers and young children and are implicated in abnormal brain development. However, available evidence on the association between anemia, ID and neurodevelopment in sub-Saharan Africa is limited. Using data from the Entebbe Mother and Baby Study prospective birth cohort, we examined the effect of maternal and child hemoglobin (Hb) levels and child iron status on developmental scores in 933 and 530 pre-school Ugandan children respectively. Associations between Hb levels, iron status and developmental scores were assessed using regression analyses adjusting for potential confounders. Lower maternal and child Hb levels were associated with reduced psychomotor scores at 15 months, while only lower Hb levels in infancy were associated with reduced language scores. We found no evidence that anemia or ID was associated with cognitive or motor scores at five years. This study emphasizes the importance of managing anemia in pregnancy and infancy and highlights the need for further studies on the effects of anemia and ID in children living in Sub-Saharan Africa.
Collapse
Affiliation(s)
- Margaret Nampijja
- Maternal and Child Wellbeing (MCW) Unit, African Population and Health Research Center, Nairobi 00100, Kenya;
- Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe P.O. Box 49, Uganda;
| | - Agnes M. Mutua
- Kenya Medical Research Institute (KEMRI) Wellcome Trust Research Programme, KEMRI Centre for Geographic Medicine Research-Coast, Kilifi 230-80108, Kenya; (J.M.M.); (A.A.)
- Correspondence: (A.M.M.); (S.H.A.); Tel.: +254-709983677-76 (A.M.M. & S.H.A.)
| | - Alison M. Elliott
- Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe P.O. Box 49, Uganda;
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - John Muthii Muriuki
- Kenya Medical Research Institute (KEMRI) Wellcome Trust Research Programme, KEMRI Centre for Geographic Medicine Research-Coast, Kilifi 230-80108, Kenya; (J.M.M.); (A.A.)
| | - Amina Abubakar
- Kenya Medical Research Institute (KEMRI) Wellcome Trust Research Programme, KEMRI Centre for Geographic Medicine Research-Coast, Kilifi 230-80108, Kenya; (J.M.M.); (A.A.)
- Department of Public Health, School of Human and Health Sciences, Pwani University, Kilifi 195-80108, Kenya
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
- Institute for Human Development, Aga Khan University, Nairobi 30270-00100, Kenya
| | - Emily L. Webb
- MRC International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK;
| | - Sarah H. Atkinson
- Kenya Medical Research Institute (KEMRI) Wellcome Trust Research Programme, KEMRI Centre for Geographic Medicine Research-Coast, Kilifi 230-80108, Kenya; (J.M.M.); (A.A.)
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
- Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
- Correspondence: (A.M.M.); (S.H.A.); Tel.: +254-709983677-76 (A.M.M. & S.H.A.)
| |
Collapse
|
18
|
Cheli VT, Santiago González DA, Wan Q, Denaroso G, Wan R, Rosenblum SL, Paez PM. H-ferritin expression in astrocytes is necessary for proper oligodendrocyte development and myelination. Glia 2021; 69:2981-2998. [PMID: 34460113 PMCID: PMC10584656 DOI: 10.1002/glia.24083] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/13/2021] [Accepted: 08/21/2021] [Indexed: 12/23/2022]
Abstract
How iron is delivered to the CNS for myelination is poorly understood. Astrocytes are the most abundant glial cells in the brain and are the only cells in close contact with blood vessels. Therefore, they are strategically located to obtain nutrients, such as iron, from circulating blood. To determine the importance of astrocyte iron uptake and storage in myelination and remyelination, we conditionally knocked-out the expression of the divalent metal transporter 1 (DMT1), the transferrin receptor 1 (Tfr1), and the ferritin heavy subunit (Fth) in Glast-1-positive astrocytes. DMT1 or Tfr1 ablation in astrocytes throughout early brain development did not significantly affects oligodendrocyte maturation or iron homeostasis. However, blocking Fth production in astrocytes during the first postnatal week drastically delayed oligodendrocyte development and myelin synthesis. Fth knockout animals presented an important decrease in the number of myelinating oligodendrocytes and a substantial reduction in the percentage of myelinated axons. This postnatal hypomyelination was accompanied by a decline in oligodendrocyte iron uptake and with an increase in brain oxidative stress. We also tested the relevance of astrocytic Fth expression in the cuprizone model of myelin damage and repair. Fth deletion in Glast1-positive astrocytes significantly reduced myelin production and the density of mature myelinating oligodendrocytes throughout the complete remyelination process. These results indicate that Fth iron storage in astrocytes is vital for early oligodendrocyte development as well as for the remyelination of the CNS.
Collapse
Affiliation(s)
- Veronica T Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - Diara A Santiago González
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | - Qiuchen Wan
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York, USA
| | | | | | | | | |
Collapse
|
19
|
German KR, Vu PT, Comstock BA, Ohls RK, Heagerty PJ, Mayock DE, Georgieff M, Rao R, Juul SE, PENUT Consortium. Enteral Iron Supplementation in Infants Born Extremely Preterm and its Positive Correlation with Neurodevelopment; Post Hoc Analysis of the Preterm Erythropoietin Neuroprotection Trial Randomized Controlled Trial. J Pediatr 2021; 238:102-109.e8. [PMID: 34324880 PMCID: PMC8629150 DOI: 10.1016/j.jpeds.2021.07.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/18/2021] [Accepted: 07/02/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To test whether an increased iron dose is associated with improved neurodevelopment as assessed by the Bayley Scales of Infant Development, third edition (BSID-III) among infants enrolled in the Preterm Erythropoietin (Epo) Neuroprotection Trial (PENUT). STUDY DESIGN This is a post hoc analysis of a randomized trial that enrolled infants born at 24-28 completed weeks of gestation. All infants in PENUT who were assessed with BSID-III at 2 years were included in this study. The associations between enteral iron dose at 60 and 90 days and BSID-III component scores were evaluated using generalized estimating equations models adjusted for potential confounders. RESULTS In total, 692 infants were analyzed (355 placebo, 337 Epo). Enteral iron supplementation ranged from 0 to 14.7 mg/kg/d (IQR 2.1-5.8 mg/kg/d) at day 60, with a mean of 3.6 mg/kg/d in infants treated with placebo and 4.8 mg/kg/d in infants treated with Epo. A significant positive association was seen between BSID-III cognitive scores and iron dose at 60 days, with an effect size of 0.77 BSID points per 50 mg/kg increase in cumulative iron dose (P = .03). Greater iron doses were associated with greater motor and language scores but did not reach statistical significance. Results at 90 days were not significant. The effect size in the infants treated with Epo compared with placebo was consistently greater. CONCLUSIONS A positive association was seen between iron dose at 60 days and cognitive outcomes. Our results suggest that increased iron supplementation in infants born preterm, at the doses administered in the PENUT Trial, may have positive neurodevelopmental effects, particularly in infants treated with Epo. TRIAL REGISTRATION Clinicaltrials.gov: NCT01378273.
Collapse
Affiliation(s)
- Kendell R. German
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Phuong T Vu
- Current Affiliation: Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, WA, United States,Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Bryan A. Comstock
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Robin K. Ohls
- Department of Pediatrics, University of Utah, Salt Lake City, UT, United States
| | - Patrick J. Heagerty
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Dennis E. Mayock
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Michael Georgieff
- Department of Pediatrics, University of Minnesota, MN, United States
| | - Raghavendra Rao
- Department of Pediatrics, University of Minnesota, MN, United States
| | - Sandra E. Juul
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | | |
Collapse
|
20
|
Rahi S, Gupta R, Sharma A, Mehan S. Smo-Shh signaling activator purmorphamine ameliorates neurobehavioral, molecular, and morphological alterations in an intracerebroventricular propionic acid-induced experimental model of autism. Hum Exp Toxicol 2021; 40:1880-1898. [PMID: 33906504 DOI: 10.1177/09603271211013456] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disease characterized by cognitive and sensorimotor impairment. Numerous research findings have consistently shown that alteration of Smo-Shh (smoothened-sonic hedgehog) signaling during the developmental process plays a significant role in ASD and triggers neuronal changes by promoting neuroinflammation and apoptotic markers. Purmorphamine (PUR), a small purine-derived agonist of the Smo-Shh pathway, shows resistance to hippocampal neuronal cell oxidation and decreases neuronal cell death. The goal of this study was to investigate the neuroprotective potential of PUR in brain intoxication induced by intracerebroventricular-propionic acid (ICV-PPA) in rats, with a focus on its effect on Smo-Shh regulation in the brain of rats. In addition, we analyze the impact of PUR on myelin basic protein (MBP) and apoptotic markers such as Caspase-3, Bax (pro-apoptotic), and Bcl-2 (anti-apoptotic) in rat brain homogenates. Chronic ICV-PPA infusion was administered consecutively for 11 days to induce autism in rats. In order to investigate behavioral alterations, rats were tested for spatial learning in the Morris Water Maze (MWM), locomotive alterations using actophotometer, and beam crossing task, while Forced Swimming Test (FST) for depressive behavior. PUR treatment with 5 mg/kg and 10 mg/kg (i.p.) was administered from day 12 to 44. Besides cellular, molecular and neuroinflammatory analyses, neurotransmitter levels and oxidative markers have also been studied in brain homogenates. The results of this study have shown that PUR increases the level of Smo-Shh and restores the neurochemical levels, and potentially prevents morphological changes, including demyelination.
Collapse
Affiliation(s)
- S Rahi
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| | - R Gupta
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| | - A Sharma
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| | - S Mehan
- Neuropharmacology Division, Department of Pharmacology, 75126ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
21
|
Pryweller JR, Glass JO, Sabin ND, Laningham FH, Li Y, Jacola LM, Conklin HM, Reddick WE. Characterization of Leukoencephalopathy and Association With Later Neurocognitive Performance in Pediatric Acute Lymphoblastic Leukemia. Invest Radiol 2021; 56:117-126. [PMID: 32769417 PMCID: PMC8059361 DOI: 10.1097/rli.0000000000000715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The most common form of pediatric cancer is acute lymphoblastic leukemia (ALL). Magnetic resonance (MR) neuroimaging studies have revealed leukoencephalopathy (LE) in pediatric ALL, but the impact of LE on long-term neurocognitive performance remains unknown. This study aims to objectively characterize the prevalence, extent, and intensity of LE, and their association with later neurocognitive performance. MATERIALS AND METHODS Pediatric patients (N = 377) treated for ALL without irradiation underwent MR neuroimaging at 4 time points throughout therapy (end of remission induction [MR1], end of consolidation [MR2], and week 31 [MR3] and week 120 [end therapy, MR4] of continuation treatment) and neurocognitive evaluations at the end of therapy and 2 years later. Generalized estimation equation models with logit link were developed to explore the association between LE prevalence and extent with time points throughout therapy, age at diagnosis (≤5 years or >5 years), treatment risk arm (low risk or standard/high risk), and sex. General linear models were also developed to investigate the association between neuroimaging metrics during treatment and neurocognitive performance at 2-year follow-up. RESULTS The prevalence of LE was greatest (22.8%, 74/324) after consolidation therapy. The prevalence of LE increased at MR2 relative to MR1 regardless of treatment risk arm (both P's < 0.001), age group (both P's < 0.001), or sex (male, P < 0.001; female, P = 0.013). The extent of white matter affected also increased at MR2 relative to MR1 regardless of treatment risk arm (standard/high risk, P < 0.001; low risk, P = 0.004), age group (both P's < 0.001), or sex (male, P < 0.001; female, P = 0.001). Quantitative relaxation rates were significantly longer in LE compared with that in normal-appearing white matter in the same examination (T1, P < 0.001; T2, P < 0.001). The LE prevalence early in therapy was associated with increased parent ratings of conduct problems (P = 0.039) and learning difficulties (P = 0.036) at 2-year follow-up compared with that at the end of therapy. A greater extent of LE early in therapy was associated with decreasing performance on a measure of processing speed (P = 0.003) from the end of therapy to 2-year follow-up. A larger extent of LE at the end of therapy was associated with decreased performance in reading (P = 0.004), spelling (P = 0.003), and mathematics (P = 0.019) at 2-year follow-up and increasing problems with attention (omissions, P = 0.045; β, P = 0.015) and memory (list A total recall, P = 0.010) at 2-year follow-up compared with that at the end of therapy. CONCLUSIONS In this large cohort of pediatric patients treated for ALL without irradiation, asymptomatic LE during therapy can be seen in almost a quarter of patients, involves as much as 10% of the white matter volume, and is associated with decreasing neurocognitive performance, increasing parent reports of conduct problems, and learning difficulties in survivors.
Collapse
Affiliation(s)
- Jennifer R. Pryweller
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - John O. Glass
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Noah D. Sabin
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Fred H. Laningham
- Department of Diagnostic Radiology, Children’s Hospital Central California, Madera, California
| | - Yimei Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Lisa M. Jacola
- Department of Psychology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Heather M. Conklin
- Department of Psychology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Wilburn E. Reddick
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee
| |
Collapse
|
22
|
Azupogo F, Abizari AR, Osendarp SJM, Feskens EJ, Brouwer ID. Ten2Twenty-Ghana: Study Design and Methods for an Innovative Randomized Controlled Trial with Multiple-Micronutrient-Fortified Biscuits among Adolescent Girls in Northeastern Ghana. Curr Dev Nutr 2021; 5:nzaa184. [PMID: 33604499 PMCID: PMC7880270 DOI: 10.1093/cdn/nzaa184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 02/04/2023] Open
Abstract
Investing in adolescent girls' nutrition is vital for health and for breaking the intergenerational cycle of malnutrition and deprivation, but limited knowledge on the type, timing, and efficacy of interventions delays progress. We describe the design of a 26-wk randomized placebo-controlled trial with multiple-micronutrient-fortified biscuits (MMBs) among adolescent girls in northeastern Ghana. Apparently healthy, premenarche (n = 312) and postmenarche (n = 309) girls (10-17 y) were randomly assigned to receive the following for 5 d/wk: 1) MMBs (fortified with 11 vitamins and 7 minerals) or 2) unfortified biscuits. Data included plasma micronutrient status, anthropometry, body composition, cognitive function, psychosocial health, fertility, dietary intake, and sociodemographic and socioeconomic covariates, complemented with in-depth interviews (n = 30) and 4 focus group discussions. We hypothesized an increase in plasma ferritin and retinol-binding protein with a resultant increase in hemoglobin, cognition, vertical height, and psychosocial health. Our study seeks to investigate the efficacy and optimal timing of a multiple-micronutrient food intervention program for adolescent girls. The RCT was registered prospectively with the Netherlands Clinical Trials Register (NL7487).
Collapse
Affiliation(s)
- Fusta Azupogo
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
- Department of Family and Consumer Sciences, Faculty of Agriculture, Food, and Consumer Sciences, University for Development Studies, Tamale, Ghana
| | - Abdul-Razak Abizari
- Department of Nutritional Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Saskia J M Osendarp
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
- Micronutrient Forum, Washington, DC, USA
| | - Edith J Feskens
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - Inge D Brouwer
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
23
|
Impaired Postnatal Myelination in a Conditional Knockout Mouse for the Ferritin Heavy Chain in Oligodendroglial Cells. J Neurosci 2020; 40:7609-7624. [PMID: 32868463 DOI: 10.1523/jneurosci.1281-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 01/22/2023] Open
Abstract
To define the importance of iron storage in oligodendrocyte development and function, the ferritin heavy subunit (Fth) was specifically deleted in oligodendroglial cells. Blocking Fth synthesis in Sox10 or NG2-positive oligodendrocytes during the first or the third postnatal week significantly reduces oligodendrocyte iron storage and maturation. The brain of Fth KO animals presented an important decrease in the expression of myelin proteins and a substantial reduction in the percentage of myelinated axons. This hypomyelination was accompanied by a decline in the number of myelinating oligodendrocytes and with a reduction in proliferating oligodendrocyte progenitor cells (OPCs). Importantly, deleting Fth in Sox10-positive oligodendroglial cells after postnatal day 60 has no effect on myelin production and/or oligodendrocyte quantities. We also tested the capacity of Fth-deficient OPCs to remyelinate the adult brain in the cuprizone model of myelin injury and repair. Fth deletion in NG2-positive OPCs significantly reduces the number of mature oligodendrocytes and myelin production throughout the remyelination process. Furthermore, the corpus callosum of Fth KO animals presented a significant decrease in the percentage of remyelinated axons and a substantial reduction in the average myelin thickness. These results indicate that Fth synthesis during the first three postnatal weeks is important for an appropriate oligodendrocyte development, and suggest that Fth iron storage in adult OPCs is also essential for an effective remyelination of the mouse brain.SIGNIFICANCE STATEMENT To define the importance of iron storage in oligodendrocyte function, we have deleted the ferritin heavy chain (Fth) specifically in the oligodendrocyte lineage. Fth ablation in oligodendroglial cells throughout early postnatal development significantly reduces oligodendrocyte maturation and myelination. In contrast, deletion of Fth in oligodendroglial cells after postnatal day 60 has no effect on myelin production and/or oligodendrocyte numbers. We have also tested the consequences of disrupting Fth iron storage in oligodendrocyte progenitor cells (OPCs) after demyelination. We have found that Fth deletion in NG2-positive OPCs significantly delays the remyelination process in the adult brain. Therefore, Fth iron storage is essential for early oligodendrocyte development as well as for OPC maturation in the demyelinated adult brain.
Collapse
|
24
|
Mukherjee C, Kling T, Russo B, Miebach K, Kess E, Schifferer M, Pedro LD, Weikert U, Fard MK, Kannaiyan N, Rossner M, Aicher ML, Goebbels S, Nave KA, Krämer-Albers EM, Schneider A, Simons M. Oligodendrocytes Provide Antioxidant Defense Function for Neurons by Secreting Ferritin Heavy Chain. Cell Metab 2020; 32:259-272.e10. [PMID: 32531201 PMCID: PMC7116799 DOI: 10.1016/j.cmet.2020.05.019] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/25/2020] [Accepted: 05/26/2020] [Indexed: 12/28/2022]
Abstract
An evolutionarily conserved function of glia is to provide metabolic and structural support for neurons. To identify molecules generated by glia and with vital functions for neurons, we used Drosophila melanogaster as a screening tool, and subsequently translated the findings to mice. We found that a cargo receptor operating in the secretory pathway of glia was essential to maintain axonal integrity by regulating iron buffering. Ferritin heavy chain was identified as the critical secretory cargo, required for the protection against iron-mediated ferroptotic axonal damage. In mice, ferritin heavy chain is highly expressed by oligodendrocytes and secreted by employing an unconventional secretion pathway involving extracellular vesicles. Disrupting the release of extracellular vesicles or the expression of ferritin heavy chain in oligodendrocytes causes neuronal loss and oxidative damage in mice. Our data point to a role of oligodendrocytes in providing an antioxidant defense system to support neurons against iron-mediated cytotoxicity.
Collapse
Affiliation(s)
- Chaitali Mukherjee
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Tina Kling
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Belisa Russo
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Kerstin Miebach
- Institute of Developmental Biology and Neurobiology (IDN), University of Mainz, 55128 Mainz, Germany
| | - Eva Kess
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Liliana D Pedro
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Ulrich Weikert
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Maryam K Fard
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Nirmal Kannaiyan
- Department of Psychiatry, Ludwig-Maximillian University, 80336 Munich, Germany
| | - Moritz Rossner
- Department of Psychiatry, Ludwig-Maximillian University, 80336 Munich, Germany
| | - Marie-Louise Aicher
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Eva-Maria Krämer-Albers
- Institute of Developmental Biology and Neurobiology (IDN), University of Mainz, 55128 Mainz, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Bonn, 53127 Bonn, Germany.
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
25
|
Irritability and Perceived Expressed Emotion in Adolescents With Iron Deficiency and Iron Deficiency Anemia: A Case-Control Study. J Pediatr Hematol Oncol 2020; 42:403-409. [PMID: 31725544 DOI: 10.1097/mph.0000000000001663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The objective of this study was to assess the difference in irritability and perceived expressed emotion (EE) between adolescents with iron deficiency (ID) or iron deficiency anemia (IDA) and their healthy peers. In addition, we aimed to investigate the relationship between hemogram parameters, irritability, and perceived EE in adolescents with ID and IDA. The sample of this single-center cross-sectional case-control study consisted of 89 adolescents from 12 to 17 years of age. Of the participants, 19 had been diagnosed with ID, 31 had IDA, and 39 were healthy controls. Significant differences in the self-reported and parent-reported irritability scores were observed between the ID group and the control group and between the IDA group and the control group. There was also a significant difference in the subscale of irritability between the ID group and the control group. The difference between the IDA and control groups in the intrusiveness subscale was found to be significant as well. Adolescents with IDA and ID exhibited significantly perceived irritability compared with the control group. In terms of irritability, adolescents with IDA and ID revealed greater irritability than their healthy peers. The results of this study suggest that irritability and perceived EE should be investigated in cases of ID, whether with or without anemia. As chronic diseases may benefit substantially from psychiatric consultation, psychosocial evaluation and intervention should be considered a complementary treatment option in the management of ID and IDA.
Collapse
|
26
|
Takeuchi H, Taki Y, Nouchi R, Yokoyama R, Kotozaki Y, Nakagawa S, Sekiguchi A, Iizuka K, Yamamoto Y, Hanawa S, Araki T, Miyauchi CM, Sakaki K, Nozawa T, Ikeda S, Yokota S, Daniele M, Sassa Y, Kawashima R. Association of iron levels in hair with brain structures and functions in young adults. J Trace Elem Med Biol 2020; 58:126436. [PMID: 31760327 DOI: 10.1016/j.jtemb.2019.126436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 11/02/2019] [Accepted: 11/12/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND Iron plays a critical role in normal brain functions and development, but it has also been known to have adverse neurological effects. METHODS Here, we investigated the associations of iron levels in hair with regional gray matter volume (rGMV), regional cerebral blood flow (rCBF), fractional anisotropy (FA), mean diffusivity (MD), and cognitive differences in a study cohort of 590 healthy young adults. RESULTS Our findings showed that high iron levels were associated with lower rGMV in areas including the hippocampus, lower rCBF in the anterior and posterior parts of the brain, greater FA in areas including the part of the splenium of the corpus callosum, lower MD in the overlapping area including the splenium of the corpus callosum, as well as greater MD in the left hippocampus and areas including the frontal lobe. CONCLUSION These results are compatible with the notion that iron plays diverse roles in neural mechanisms in healthy young adults.
Collapse
Affiliation(s)
- Hikaru Takeuchi
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| | - Yasuyuki Taki
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Division of Medical Neuroimaging Analysis, Department of Community Medical Supports, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Department of Radiology and Nuclear Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Rui Nouchi
- Creative Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Science, Tohoku University, Sendai, Japan; Human and Social Response Research Division, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan; Department of Advanced Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | - Yuka Kotozaki
- Division of Clinical research, Medical-Industry Translational Research Center, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Seishu Nakagawa
- Department of Human Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Division of Psychiatry, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Atsushi Sekiguchi
- Division of Medical Neuroimaging Analysis, Department of Community Medical Supports, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kunio Iizuka
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuki Yamamoto
- Department of Human Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Sugiko Hanawa
- Department of Human Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | - Carlos Makoto Miyauchi
- Department of Language Sciences, Graduate School of Humanities, Tokyo Metropolitan University, Tokyo, Japan
| | - Kohei Sakaki
- Department of Advanced Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Takayuki Nozawa
- Research Center for the Earth Inclusive Sensing Empathizing with Silent Voices, Tokyo Institute of Technology, Tokyo, Japan
| | - Shigeyuki Ikeda
- Department of Ubiquitous Sensing, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Susumu Yokota
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Magistro Daniele
- Department of Sport Science, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, United Kingdom
| | - Yuko Sassa
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Ryuta Kawashima
- Division of Developmental Cognitive Neuroscience, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Department of Advanced Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Department of Human Brain Science, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
27
|
Iron Metabolism in the Peripheral Nervous System: The Role of DMT1, Ferritin, and Transferrin Receptor in Schwann Cell Maturation and Myelination. J Neurosci 2019; 39:9940-9953. [PMID: 31676601 DOI: 10.1523/jneurosci.1409-19.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/09/2019] [Accepted: 10/29/2019] [Indexed: 12/24/2022] Open
Abstract
Iron is an essential cofactor for many cellular enzymes involved in myelin synthesis, and iron homeostasis unbalance is a central component of peripheral neuropathies. However, iron absorption and management in the PNS are poorly understood. To study iron metabolism in Schwann cells (SCs), we have created 3 inducible conditional KO mice in which three essential proteins implicated in iron uptake and storage, the divalent metal transporter 1 (DMT1), the ferritin heavy chain (Fth), and the transferrin receptor 1 (Tfr1), were postnatally ablated specifically in SCs. Deleting DMT1, Fth, or Tfr1 in vitro significantly reduce SC proliferation, maturation, and the myelination of DRG axons. This was accompanied by an important reduction in iron incorporation and storage. When these proteins were KO in vivo during the first postnatal week, the sciatic nerve of all 3 conditional KO animals displayed a significant reduction in the synthesis of myelin proteins and in the percentage of myelinated axons. Knocking out Fth produced the most severe phenotype, followed by DMT1 and, last, Tfr1. Importantly, DMT1 as well as Fth KO mice showed substantial motor coordination deficits. In contrast, deleting these proteins in mature myelinating SCs results in milder phenotypes characterized by small reductions in the percentage of myelinated axons and minor changes in the g-ratio of myelinated axons. These results indicate that DMT1, Fth, and Tfr1 are critical proteins for early postnatal iron uptake and storage in SCs and, as a consequence, for the normal myelination of the PNS.SIGNIFICANCE STATEMENT To determine the function of the divalent metal transporter 1, the transferrin receptor 1, and the ferritin heavy chain in Schwann cell (SC) maturation and myelination, we created 3 conditional KO mice in which these proteins were postnatally deleted in Sox10-positive SCs. We have established that these proteins are necessary for normal SC iron incorporation and storage, and, as a consequence, for an effective myelination of the PNS. Since iron is indispensable for SC maturation, understanding iron metabolism in SCs is an essential prerequisite for developing therapies for demyelinating diseases in the PNS.
Collapse
|
28
|
McCarthy EK, Kiely ME. The neonatal period: A missed opportunity for the prevention of iron deficiency and its associated neurological consequences? NUTR BULL 2019. [DOI: 10.1111/nbu.12407] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- E. K. McCarthy
- Cork Centre for Vitamin D and Nutrition Research School of Food and Nutritional Sciences University College Cork and INFANT Research Centre Cork Ireland
| | - M. E. Kiely
- Cork Centre for Vitamin D and Nutrition Research School of Food and Nutritional Sciences University College Cork and INFANT Research Centre Cork Ireland
| |
Collapse
|
29
|
Multifactorial Etiology of Anemia in Celiac Disease and Effect of Gluten-Free Diet: A Comprehensive Review. Nutrients 2019; 11:nu11112557. [PMID: 31652803 PMCID: PMC6893537 DOI: 10.3390/nu11112557] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Celiac disease (CD) is a multisystemic disorder with different clinical expressions, from malabsorption with diarrhea, anemia, and nutritional compromise to extraintestinal manifestations. Anemia might be the only clinical expression of the disease, and iron deficiency anemia is considered one of the most frequent extraintestinal clinical manifestations of CD. Therefore, CD should be suspected in the presence of anemia without a known etiology. Assessment of tissue anti-transglutaminase and anti-endomysial antibodies are indicated in these cases and, if positive, digestive endoscopy and intestinal biopsy should be performed. Anemia in CD has a multifactorial pathogenesis and, although it is frequently a consequence of iron deficiency, it can be caused by deficiencies of folate or vitamin B12, or by blood loss or by its association with inflammatory bowel disease (IBD) or other associated diseases. The association between CD and IBD should be considered during anemia treatment in patients with IBD, because the similarity of symptoms could delay the diagnosis. Vitamin B12 deficiency is common in CD and may be responsible for anemia and peripheral myeloneuropathy. Folate deficiency is a well-known cause of anemia in adults, but there is little information in children with CD; it is still unknown if anemia is a symptom of the most typical CD in adult patients either by predisposition due to the fact of age or because biochemical and clinical manifestations take longer to appear.
Collapse
|
30
|
Faissner S, Plemel JR, Gold R, Yong VW. Progressive multiple sclerosis: from pathophysiology to therapeutic strategies. Nat Rev Drug Discov 2019; 18:905-922. [PMID: 31399729 DOI: 10.1038/s41573-019-0035-2] [Citation(s) in RCA: 300] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system that involves demyelination and axonal degeneration. Although substantial progress has been made in drug development for relapsing-remitting MS, treatment of the progressive forms of the disease, which are characterized clinically by the accumulation of disability in the absence of relapses, remains unsatisfactory. This unmet clinical need is related to the complexity of the pathophysiological mechanisms involved in MS progression. Chronic inflammation, which occurs behind a closed blood-brain barrier with activation of microglia and continued involvement of T cells and B cells, is a hallmark pathophysiological feature. Inflammation can enhance mitochondrial damage in neurons, which, consequently, develop an energy deficit, further reducing axonal health. The growth-inhibitory and inflammatory environment of lesions also impairs remyelination, a repair process that might protect axons from degeneration. Moreover, neurodegeneration is accelerated by the altered expression of ion channels on denuded axons. In this Review, we discuss the current understanding of these disease mechanisms and highlight emerging therapeutic strategies based on these insights, including those targeting the neuroinflammatory and degenerative aspects as well as remyelination-promoting approaches.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany. .,Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| | - Jason R Plemel
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
31
|
Guitart ME, Vence M, Correale J, Pasquini JM, Rosato-Siri MV. Ontogenetic oligodendrocyte maturation through gestational iron deprivation: The road not taken. Glia 2019; 67:1760-1774. [PMID: 31162719 DOI: 10.1002/glia.23647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 01/01/2023]
Abstract
Developmental iron deficiency (dID) models facilitate the study of specific oligodendrocyte (OL) requirements for their progression to a mature state and subsequent contribution to myelination. In the current work, we used the dID model in transgenic mice expressing green fluorescence protein under the CNPase promoter allowing the identification of cells belonging to the oligodendroglial lineage, and the visualization of the entire myelin structure and single OL morphology. The present work evaluates dID effects on OL complexity in different brain areas. Control animals showed an increase in OL complexity both during development and along the anterior-posterior axis. In contrast, dID animals exhibited an initial increase in CNPase+ cells with prevalence of immature-OL (i-OL), an effect later compensated during development by selective death of those i-OL. As a consequence, developmental behavior was impaired in terms of body balance, muscle response, and sensorimotor functions. To explore why i-OL fail to mature in dID, expression levels of transcriptional factors involved in the maturation of the OL lineage were studied. In nuclear fractions, dID animals showed an increase in Hes5, which prevents the maturation of i-OL, and a decrease in Sox10, a positive regulator of OL maturation. The cytoplasmic fractions showed a decrease in Olig1, which is critical for precursor cell differentiation into premyelinating OL. Overall, the expression levels of Hes5, Sox10, and Olig1 in dID conditions correlated with an unfavorable OL maturation profile. In sum, the current results provide further evidence of dID impact on myelination, keeping OL away from the maturational path.
Collapse
Affiliation(s)
- María E Guitart
- Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Vence
- Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Juana M Pasquini
- Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María V Rosato-Siri
- Departamento de Química Biológica, IQUIFIB-CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
32
|
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
|
33
|
Rao R, Bora R. Timing of Umbilical Cord Clamping and Infant Brain Development. J Pediatr 2018; 203:8-10. [PMID: 30193752 DOI: 10.1016/j.jpeds.2018.07.080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/24/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Raghavendra Rao
- Division of Neonatology Department of Pediatrics University of Minnesota Minneapolis, Minnesota.
| | - Reeta Bora
- Neonatal Unit Department of Pediatrics Assam Medical College Dibrugarh, Assam, India
| |
Collapse
|
34
|
The Divalent Metal Transporter 1 (DMT1) Is Required for Iron Uptake and Normal Development of Oligodendrocyte Progenitor Cells. J Neurosci 2018; 38:9142-9159. [PMID: 30190412 DOI: 10.1523/jneurosci.1447-18.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/08/2018] [Accepted: 08/27/2018] [Indexed: 01/24/2023] Open
Abstract
The divalent metal transporter 1 (DMT1) is a multimetal transporter with a primary role in iron transport. Although DMT1 has been described previously in the CNS, nothing was known about the role of this metal transporter in oligodendrocyte maturation and myelination. To determine whether DMT1 is required for oligodendrocyte progenitor cell (OPC) maturation, we used siRNAs and the Cre-lox system to knock down/knock out DMT1 expression in vitro as well as in vivo Blocking DMT1 synthesis in primary cultures of OPCs reduced oligodendrocyte iron uptake and significantly delayed OPC development. In vivo, a significant hypomyelination was found in DMT1 conditional knock-out mice in which DMT1 was postnatally deleted in NG2- or Sox10-positive OPCs. The brain of DMT1 knock-out animals presented a decrease in the expression levels of myelin proteins and a substantial reduction in the percentage of myelinated axons. This reduced postnatal myelination was accompanied by a decrease in the number of myelinating oligodendrocytes and a rise in proliferating OPCs. Furthermore, using the cuprizone model of demyelination, we established that DMT1 deletion in NG2-positive OPCs lead to less efficient remyelination of the adult brain. These results indicate that DMT1 is vital for OPC maturation and for the normal myelination of the mouse brain.SIGNIFICANCE STATEMENT To determine whether divalent metal transporter 1 (DMT1), a multimetal transporter with a primary role in iron transport, is essential for oligodendrocyte development, we created two conditional knock-out mice in which DMT1 was postnatally deleted in NG2- or Sox10-positive oligodendrocyte progenitor cells (OPCs). We have established that DMT1 is necessary for normal OPC maturation and is required for an efficient remyelination of the adult brain. Since iron accumulation by OPCs is indispensable for myelination, understanding the iron incorporation mechanism as well as the molecules involved is critical to design new therapeutic approaches to intervene in diseases in which the myelin sheath is damaged or lost.
Collapse
|
35
|
Lumsden AL, Rogers JT, Majd S, Newman M, Sutherland GT, Verdile G, Lardelli M. Dysregulation of Neuronal Iron Homeostasis as an Alternative Unifying Effect of Mutations Causing Familial Alzheimer's Disease. Front Neurosci 2018; 12:533. [PMID: 30150923 PMCID: PMC6099262 DOI: 10.3389/fnins.2018.00533] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
The overwhelming majority of dominant mutations causing early onset familial Alzheimer’s disease (EOfAD) occur in only three genes, PSEN1, PSEN2, and APP. An effect-in-common of these mutations is alteration of production of the APP-derived peptide, amyloid β (Aβ). It is this key fact that underlies the authority of the Amyloid Hypothesis that has informed Alzheimer’s disease research for over two decades. Any challenge to this authority must offer an alternative explanation for the relationship between the PSEN genes and APP. In this paper, we explore one possible alternative relationship – the dysregulation of cellular iron homeostasis as a common effect of EOfAD mutations in these genes. This idea is attractive since it provides clear connections between EOfAD mutations and major characteristics of Alzheimer’s disease such as dysfunctional mitochondria, vascular risk factors/hypoxia, energy metabolism, and inflammation. We combine our ideas with observations by others to describe a “Stress Threshold Change of State” model of Alzheimer’s disease that may begin to explain the existence of both EOfAD and late onset sporadic (LOsAD) forms of the disease. Directing research to investigate the role of dysregulation of iron homeostasis in EOfAD may be a profitable way forward in our struggle to understand this form of dementia.
Collapse
Affiliation(s)
- Amanda L Lumsden
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Jack T Rogers
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, Massachusetts General Hospital (East), Harvard Medical School, Harvard University, Charlestown, MA, United States
| | - Shohreh Majd
- Neuronal Injury and Repair Laboratory, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Morgan Newman
- Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Greg T Sutherland
- Discipline of Pathology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Giuseppe Verdile
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Michael Lardelli
- Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
36
|
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
|
37
|
Oxidative stress and neurodegeneration: the involvement of iron. Biometals 2018; 31:715-735. [PMID: 30014355 DOI: 10.1007/s10534-018-0126-2] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Many evidences indicate that oxidative stress plays a significant role in a variety of human disease states, including neurodegenerative diseases. Iron is an essential metal for almost all living organisms due to its involvement in a large number of iron-containing proteins and enzymes, though it could be also toxic. Actually, free iron excess generates oxidative stress, particularly in brain, where anti-oxidative defences are relatively low. Its accumulation in specific regions is associated with pathogenesis in a variety of neurodegenerative diseases (i.e., Parkinson's disease, Alzheimer's disease, Huntington's chorea, Amyotrophic Lateral Sclerosis and Neurodegeneration with Brain Iron Accumulation). Anyway, the extent of toxicity is dictated, in part, by the localization of the iron complex within the cell (cytosolic, lysosomal and mitochondrial), its biochemical form, i.e., ferritin or hemosiderin, as well as the ability of the cell to prevent the generation and propagation of free radical by the wide range of antioxidants and cytoprotective enzymes in the cell. Particularly, ferrous iron can act as a catalyst in the Fenton reaction that potentiates oxygen toxicity by generating a wide range of free radical species, including hydroxyl radicals (·OH). The observation that patients with neurodegenerative diseases show a dramatic increase in their brain iron content, correlated with the production of reactive oxigen species in these areas of the brain, conceivably suggests that disturbances in brain iron homeostasis may contribute to the pathogenesis of these disorders. The aim of this review is to describe the chemical features of iron in human beings and iron induced toxicity in neurodegenerative diseases. Furthermore, the attention is focused on metal chelating drugs therapeutic strategies.
Collapse
|
38
|
Lynch S, Pfeiffer CM, Georgieff MK, Brittenham G, Fairweather-Tait S, Hurrell RF, McArdle HJ, Raiten DJ. Biomarkers of Nutrition for Development (BOND)-Iron Review. J Nutr 2018; 148:1001S-1067S. [PMID: 29878148 PMCID: PMC6297556 DOI: 10.1093/jn/nxx036] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/27/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022] Open
Abstract
This is the fifth in the series of reviews developed as part of the Biomarkers of Nutrition for Development (BOND) program. The BOND Iron Expert Panel (I-EP) reviewed the extant knowledge regarding iron biology, public health implications, and the relative usefulness of currently available biomarkers of iron status from deficiency to overload. Approaches to assessing intake, including bioavailability, are also covered. The report also covers technical and laboratory considerations for the use of available biomarkers of iron status, and concludes with a description of research priorities along with a brief discussion of new biomarkers with potential for use across the spectrum of activities related to the study of iron in human health.The I-EP concluded that current iron biomarkers are reliable for accurately assessing many aspects of iron nutrition. However, a clear distinction is made between the relative strengths of biomarkers to assess hematological consequences of iron deficiency versus other putative functional outcomes, particularly the relationship between maternal and fetal iron status during pregnancy, birth outcomes, and infant cognitive, motor and emotional development. The I-EP also highlighted the importance of considering the confounding effects of inflammation and infection on the interpretation of iron biomarker results, as well as the impact of life stage. Finally, alternative approaches to the evaluation of the risk for nutritional iron overload at the population level are presented, because the currently designated upper limits for the biomarker generally employed (serum ferritin) may not differentiate between true iron overload and the effects of subclinical inflammation.
Collapse
Affiliation(s)
| | - Christine M Pfeiffer
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN
| | - Gary Brittenham
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY
| | - Susan Fairweather-Tait
- Department of Nutrition, Norwich Medical School, Norwich Research Park, University of East Anglia, Norwich NR4 7JT, UK
| | - Richard F Hurrell
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Harry J McArdle
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, UK
| | - Daniel J Raiten
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH)
| |
Collapse
|
39
|
Robinson SL, Marín C, Oliveros H, Mora-Plazas M, Richards BJ, Lozoff B, Villamor E. Iron Deficiency, Anemia, and Low Vitamin B-12 Serostatus in Middle Childhood Are Associated with Behavior Problems in Adolescent Boys: Results from the Bogotá School Children Cohort. J Nutr 2018; 148:760-770. [PMID: 29897579 DOI: 10.1093/jn/nxy029] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/14/2017] [Accepted: 01/30/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Iron deficiency (ID) in infancy is related to subsequent behavior problems. The effects of micronutrient status in middle childhood are uncertain. OBJECTIVE The aim of the study was to examine the associations of micronutrient status biomarkers in middle childhood with externalizing and internalizing behavior problems in adolescence. METHODS We assessed whether ID (ferritin <15 µg/L), anemia (hemoglobin <12.7 g/dL), or blood concentrations of zinc, vitamins A and B-12, and folate at ages 5-12 y were associated with externalizing or internalizing behavior problems in adolescence in 1042 schoolchildren from Bogotá, Colombia. Behavior problems were assessed with the Youth Self-Report questionnaire after a median 6.2 y of follow-up. Mean problem score differences with 95% CIs were estimated between categories of micronutrient status biomarkers with the use of multivariable linear regression. RESULTS Mean ± SD externalizing and internalizing problems scores were 52.6 ± 9.6 and 53.8 ± 9.9, respectively. Among boys, middle-childhood ID, anemia, and low plasma vitamin B-12 were associated with 5.9 (95% CI: 1.0, 10.7), 6.6 (95% CI: 1.9, 11.3), and 2.7 (95% CI: 0.4, 4.9) units higher mean externalizing problems scores in adolescence, respectively-after adjustment for baseline age, time spent watching television or playing video games, mother's height, and socioeconomic status. Also in boys, ID was related to an adjusted 6.4 (95% CI: 1.2, 11.6) units higher mean internalizing problems score. There were no associations among girls. Other micronutrient status biomarkers were not associated with behavior problems. CONCLUSIONS ID, anemia, and low vitamin B-12 in middle childhood are related to behavior problems in adolescent boys.This study was registered at clinicaltrials.gov as NCT03297970.
Collapse
Affiliation(s)
- Sonia L Robinson
- Department of Epidemiology, School of Public Health, and The Center for Human Growth and Development, University of Michigan, Ann Arbor, MI
| | - Constanza Marín
- Medical School, Department of Medicine, The University of La Sabana, Colombia, Chía.,The Foundation for Research in Nutrition and Health, Colombia, Bogotá
| | - Henry Oliveros
- Medical School, Department of Medicine, The University of La Sabana, Colombia, Chía
| | | | - Blair J Richards
- The Center for Human Growth and Development, University of Michigan, Ann Arbor, MI
| | - Betsy Lozoff
- The Center for Human Growth and Development, University of Michigan, Ann Arbor, MI
| | - Eduardo Villamor
- Department of Epidemiology, School of Public Health, and The Center for Human Growth and Development, University of Michigan, Ann Arbor, MI.,The Center for Human Growth and Development, University of Michigan, Ann Arbor, MI
| |
Collapse
|
40
|
Duck KA, Neely EB, Simpson IA, Connor JR. A role for sex and a common HFE gene variant in brain iron uptake. J Cereb Blood Flow Metab 2018; 38:540-548. [PMID: 28350201 PMCID: PMC5851144 DOI: 10.1177/0271678x17701949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/23/2017] [Accepted: 03/07/2017] [Indexed: 11/16/2022]
Abstract
HFE (high iron) is an essential protein for regulating iron transport into cells. Mutations of the HFE gene result in loss of this regulation causing accumulation of iron within the cell. The mutated protein has been found increasingly in numerous neurodegenerative disorders in which increased levels of iron in the brain are reported. Additionally, evidence that these mutations are associated with elevated brain iron challenges the paradigm that the brain is protected by the blood-brain barrier. While much has been studied regarding the role of HFE in cellular iron uptake, it has remained unclear what role the protein plays in the transport of iron into the brain. We investigated regulation of iron transport into the brain using a mouse model with a mutation in the HFE gene. We demonstrated that the rate of radiolabeled iron (59Fe) uptake was similar between the two genotypes despite higher brain iron concentrations in the mutant. However, there were significant differences in iron uptake between males and females regardless of genotype. These data indicate that brain iron status is consistently maintained and tightly regulated at the level of the blood-brain barrier.
Collapse
Affiliation(s)
- Kari A Duck
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Elizabeth B Neely
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Ian A Simpson
- Department of Neural and Behavioral Sciences, Penn State Hershey Medical Center, Hershey, PA, USA
| | - James R Connor
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
41
|
Mudd AT, Fil JE, Knight LC, Dilger RN. Dietary Iron Repletion following Early-Life Dietary Iron Deficiency Does Not Correct Regional Volumetric or Diffusion Tensor Changes in the Developing Pig Brain. Front Neurol 2018; 8:735. [PMID: 29375469 PMCID: PMC5768607 DOI: 10.3389/fneur.2017.00735] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/20/2017] [Indexed: 11/17/2022] Open
Abstract
Background Iron deficiency is the most common micronutrient deficiency worldwide and children are at an increased risk due to the rapid growth occurring during early life. The developing brain is highly dynamic, requires iron for proper function, and is thus vulnerable to inadequate iron supplies. Iron deficiency early in life results in altered myelination, neurotransmitter synthesis, neuron morphology, and later-life cognitive function. However, it remains unclear if dietary iron repletion after a period of iron deficiency can recover structural deficits in the brain. Method Twenty-eight male pigs were provided either a control diet (CONT; n = 14; 23.5 mg Fe/L milk replacer) or an iron-deficient diet (ID; n = 14; 1.56 mg Fe/L milk replacer) for phase 1 of the study, from postnatal day (PND) 2 until 32. Twenty pigs (n = 10/diet from phase 1) were used in phase 2 of the study from PND 33 to 61, all pigs were provided a common iron sufficient diet, regardless of their early-life dietary iron status. All pigs remaining in the study were subjected to magnetic resonance imaging (MRI) at PND 32 and again at PND 61 using structural imaging sequences and diffusion tensor imaging (DTI) to assess volumetric and microstructural brain development, respectively. Data were analyzed using a two-way ANOVA to assess the main and interactive effects of early-life iron status and time. Results An interactive effect was observed for absolute whole brain volumes, in which whole brain volumes of ID pigs were smaller at PND 32 but were not different than CONT pigs at PND 61. Analysis of brain region volumes relative to total brain volume indicated interactive effects (i.e., diet × day) in the cerebellum, olfactory bulb, and putamen-globus pallidus. Main effects of early-life iron status, regardless of imaging time point, were noted for decreased relative volumes of the left hippocampus, right hippocampus, thalamus, and increased relative white matter volume in ID pigs compared with CONT pigs. DTI indicated interactive effects for fractional anisotropy (FA) in the whole brain, left cortex, and right cortex. Main effects of early-life iron status, regardless of imaging time point, were observed for decreased FA values in the caudate, cerebellum, and internal capsule in ID pigs compared with CONT pigs. All comparisons described above were significant at P < 0.05. Conclusion Results from this study indicate that dietary iron repletion is able to compensate for reduced absolute brain volumes early in life; however, microstructural changes and altered relative brain volumes persist despite iron repletion.
Collapse
Affiliation(s)
- Austin T Mudd
- Piglet Nutrition & Cognition Laboratory, University of Illinois, Urbana, IL, United States.,Neuroscience Program, University of Illinois, Urbana, IL, United States
| | - Joanne E Fil
- Piglet Nutrition & Cognition Laboratory, University of Illinois, Urbana, IL, United States.,Neuroscience Program, University of Illinois, Urbana, IL, United States
| | - Laura C Knight
- Piglet Nutrition & Cognition Laboratory, University of Illinois, Urbana, IL, United States.,Division of Nutrition Sciences, University of Illinois, Urbana, IL, United States
| | - Ryan N Dilger
- Piglet Nutrition & Cognition Laboratory, University of Illinois, Urbana, IL, United States.,Neuroscience Program, University of Illinois, Urbana, IL, United States.,Division of Nutrition Sciences, University of Illinois, Urbana, IL, United States.,Beckman Institute for Advances Science and Technology, University of Illinois, Urbana, IL, United States.,Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| |
Collapse
|
42
|
Otero GA, Fernández T, Pliego-Rivero FB, Mendieta GG. Iron therapy substantially restores qEEG maturational lag among iron-deficient anemic infants. Nutr Neurosci 2017; 22:363-372. [PMID: 29063783 DOI: 10.1080/1028415x.2017.1391529] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To use quantitative electroencephalography (qEEG) to assess the impact of iron-deficiency anemia on central nervous system maturation in the first year of life. METHOD Twenty-five infants (3-12 months old) presenting ferropenic anemia (IDA) and 25 healthy controls (CTL1), matched by age/gender with the former, were studied in two stages. Electroencephalogram during spontaneous sleep was recorded from all participants; the fast Fourier transform was calculated to obtain absolute power (AP) and relative power (RP) qEEG measures. In the first stage, a qEEG comparison between CTL1 and IDA was performed. Second stage consisted in comparing qEEG of the IDA infants before and after supplementation with iron (IDA-IS group), and comparing qEEG of the IDA-IS group with another control age-matched group (CTL2). Non-parametric multivariate permutation tests (NPT) were applied to assess differences between CTL1 and IDA groups, as well as IDA vs. IDA-IS, and IDA-IS vs. CTL2. RESULTS More power in slow frequency bands and less power in fast frequency bands in 64% of IDA babies were observed. NPT evinced higher alpha AP and RP (P < 0.001), less theta AP, and less delta and theta RP in CTL1 than in IDA. After iron-restoration therapy, alpha AP and RP increased while theta AP and theta and delta RP decreased, reaching almost normal values. DISCUSSION This work reveals CNS developmental delay through the study of qEEG (less rapid and more slow frequencies) which recovered significantly with iron supplementation. It is concluded that IDA constitutes a high risk factor for a lag of CNS maturation.
Collapse
Affiliation(s)
- Gloria A Otero
- a Facultad de Medicina , Universidad Autónoma del Estado de México , Toluca , México
| | - Thalía Fernández
- b Instituto de Neurobiología, Universidad Nacional Autónoma de México , Campus Juriquilla, Querétaro , México
| | | | | |
Collapse
|
43
|
Farrelly L, Rosato-Siri MV, Föcking M, Codagnone M, Reines A, Dicker P, Wynne K, Farrell M, Cannon M, Cagney G, Pasquini JM, Cotter DR. The Effects of Prenatal Iron Deficiency and Risperidone Treatment on the Rat Frontal Cortex: A Proteomic Analysis. Proteomics 2017; 17. [PMID: 28762254 DOI: 10.1002/pmic.201600407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 06/12/2017] [Indexed: 11/11/2022]
Abstract
Prenatal iron deficiency (pID) has been described to increase the risk for neurodevelopmental disorders such as autism and schizophrenia; however, the precise molecular mechanisms are still unknown. Here, we utilized high-throughput MS to examine the proteomic effects of pID in adulthood on the rat frontal cortex area (FCA). In addition, the FCA proteome was examined in adulthood following risperidone treatment in adolescence to see if these effects could be prevented. We identified 1501 proteins of which 100 were significantly differentially expressed in the FCA at postnatal day 90. Pathway analysis of proteins affected by pID revealed changes in metabolic processes, including the tricyclic acid cycle, mitochondrial dysfunction, and P13K/Akt signaling. Interestingly, most of these protein changes were not present in the adult pID offspring who received risperidone in adolescence. Considering the link between pID and several neurodevelopmental disorders such as autism and schizophrenia these presented results bring new perspectives to understand the role of iron in metabolic pathways and provide novel biomarkers for future studies of pID.
Collapse
Affiliation(s)
- Lorna Farrelly
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Maria Victoria Rosato-Siri
- Department of Biological Chemistry, IQUIFIB, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Melanie Föcking
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Martin Codagnone
- De Robertis Institute, University of Buenos Aires, Buenos Aires, Argentina
| | - Analia Reines
- De Robertis Institute, University of Buenos Aires, Buenos Aires, Argentina
| | - Patrick Dicker
- Departments of Epidemiology & Public Health, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kieran Wynne
- School of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Ireland
| | - Michael Farrell
- Department of Neuropathology, Beaumont Hospital, Dublin, Ireland
| | - Mary Cannon
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| | - Gerard Cagney
- School of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Ireland
| | - Juana Maria Pasquini
- Department of Biological Chemistry, IQUIFIB, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - David R Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
44
|
Cardoso P, Amaro P, Santos JP, de Assis JT, Carvalho ML. Determination of Nickel and Manganese Contaminants in Pharmaceutical Iron Supplements Using Energy Dispersive X-ray Fluorescence. APPLIED SPECTROSCOPY 2017; 71:432-437. [PMID: 27694429 DOI: 10.1177/0003702816670912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In this study, we investigate the capability of energy dispersive X-ray fluorescence (EDXF) spectrometry in a triaxial geometry apparatus as a fast and nondestructive determination method of both dominant and contaminant elements in pharmaceutical iron supplements. The following iron supplements brands with their respective active ingredients were analyzed: Neutrofer fólico (iron gylcinate), Anemifer (iron(II) sulfate monohydrate), Noripurum (iron(III)-hydroxide polymaltose complex), Sulferbel (iron(II) sulfate monohydrate), and Combiron Fólico (carbonyl iron). Although we observe a good agreement between the iron content obtained by the present method and that indicated in the supplement's prescribed dose, we observe contamination by manganese and nickel of up to 180 μg and 36 μg, respectively. These contents correspond to 7.2% and 14.4% of the permitted daily exposure of manganese and nickel, respectively, for an average adult individual as determined by the European Medicine Agency (EMEA). The method was successfully validated against the concentrations of several certified reference materials of biological light matrices with similar concentrations of contaminants. Moreover, we also validated our method by comparing the concentrations with those obtained with the inductively coupled plasma-atomic emission technique.
Collapse
Affiliation(s)
- Pedro Cardoso
- 1 Laboratório de Instrumentação, Engenharia Biomédica e Física da Radiação (LIBPhys-UNL), Departamento de Física, Faculdade de Ciências e Tecnologia, FCT, Universidade Nova de Lisboa, Caparica, Portugal
| | - Pedro Amaro
- 1 Laboratório de Instrumentação, Engenharia Biomédica e Física da Radiação (LIBPhys-UNL), Departamento de Física, Faculdade de Ciências e Tecnologia, FCT, Universidade Nova de Lisboa, Caparica, Portugal
| | - José Paulo Santos
- 1 Laboratório de Instrumentação, Engenharia Biomédica e Física da Radiação (LIBPhys-UNL), Departamento de Física, Faculdade de Ciências e Tecnologia, FCT, Universidade Nova de Lisboa, Caparica, Portugal
| | - Joaquim T de Assis
- 2 Universidade do Estado do Rio de Janeiro, Instituto Politécnico, Nova Friburgo, RJ, Brazil
| | - Maria Luisa Carvalho
- 1 Laboratório de Instrumentação, Engenharia Biomédica e Física da Radiação (LIBPhys-UNL), Departamento de Física, Faculdade de Ciências e Tecnologia, FCT, Universidade Nova de Lisboa, Caparica, Portugal
| |
Collapse
|
45
|
Horiquini-Barbosa E, Gibb R, Kolb B, Bray D, Lachat JJ. Tactile stimulation partially prevents neurodevelopmental changes in visual tract caused by early iron deficiency. Brain Res 2016; 1657:130-139. [PMID: 27956122 DOI: 10.1016/j.brainres.2016.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/29/2016] [Accepted: 12/02/2016] [Indexed: 01/16/2023]
Abstract
Iron deficiency has a critical impact on maturational mechanisms of the brain and the damage related to neuroanatomical parameters is not satisfactorily reversed after iron replacement. However, emerging evidence suggest that enriched early experience may offer great therapeutic efficacy in cases of nutritional disorders postnatally, since the brain is remarkably responsive to its interaction with the environment. Given the fact that tactile stimulation (TS) treatment has been previously shown to be an effective therapeutic approach and with potential application to humans, here we ask whether exposure to TS treatment, from postnatal day (P) 1 to P32 for 3min/day, could also be employed to prevent neuroanatomical changes in the optic nerve of rats maintained on an iron-deficient diet during brain development. We found that iron deficiency changed astrocyte, oligodendrocyte, damaged fiber, and myelinated fiber density, however, TS reversed the iron-deficiency-induced alteration in oligodendrocyte, damaged fiber and myelinated fiber density, but failed to reverse astrocyte density. Our results suggest that early iron deficiency may act by disrupting the timing of key steps in visual system development thereby modifying the normal progression of optic nerve maturation. However, optic nerve development is sensitive to enriching experiences, and in the current study we show that this sensitivity can be used to prevent damage from postnatal iron deficiency during the critical period.
Collapse
Affiliation(s)
- Everton Horiquini-Barbosa
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil; Laboratory of Neuroanatomy, Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil.
| | - Robbin Gibb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Alberta, Canada
| | - Bryan Kolb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Alberta, Canada
| | - Douglas Bray
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Alberta, Canada
| | - Joao-Jose Lachat
- Laboratory of Neuroanatomy, Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
46
|
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: 53] [Impact Index Per Article: 5.9] [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
|
47
|
Sun MY, Woolley JC, Blohowiak SE, Smith ZR, Siddappa AM, Magness RR, Kling PJ. Dietary-induced gestational iron deficiency inhibits postnatal tissue iron delivery and postpones the cessation of active nephrogenesis in rats. Reprod Fertil Dev 2016; 29:RD15358. [PMID: 26876724 PMCID: PMC5577434 DOI: 10.1071/rd15358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/14/2015] [Indexed: 02/01/2023] Open
Abstract
Gestational iron deficiency (ID) can alter developmental programming through impaired nephron endowment, leading to adult hypertension, but nephrogenesis is unstudied. Iron status and renal development during dietary-induced gestational ID (<6 mg Fe kg-1 diet from Gestational Day 2 to Postnatal Day (PND) 7) were compared with control rats (198 mg Fe kg-1 diet). On PND2-PND10, PND15, PND30 and PND45, blood and tissue iron status were assessed. Nephrogenic zone maturation (PND2-PND10), radial glomerular counts (RGCs), glomerular size density and total planar surface area (PND15 and PND30) were also assessed. Blood pressure (BP) was measured in offspring. ID rats were smaller, exhibiting lower erythrocyte and tissue iron than control rats (PND2-PND10), but these parameters returned to control values by PND30-PND45. Relative kidney iron (µg g-1 wet weight) at PND2-PND10 was directly related to transport iron measures. In ID rats, the maturation of the active nephrogenic zone was later than control. RGCs, glomerular size, glomerular density, and glomerular planar surface area were lower than control at PND15, but returned to control by PND30. After weaning, the kidney weight/rat weight ratio (mg g-1) was heavier in ID than control rats. BP readings at PND45 were lower in ID than control rats. Altered kidney maturation and renal adaptations may contribute to glomerular size, early hyperfiltration and long-term renal function.
Collapse
Affiliation(s)
- Mary Y. Sun
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
- Obstetrics and Gynecology Perinatal Research Laboratories, University of Wisconsin, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Joseph C. Woolley
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Sharon E. Blohowiak
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Zachary R. Smith
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Ashajyothi M. Siddappa
- Department of Pediatrics, Division of Neonatology, Hennepin County Medical Center and University of Minnesota, Minneapolis, MN, USA
- Center for Neurobehavioral Development, 516 Delaware St. SE, Minneapolis, MN 55455, USA
| | - Ronald R. Magness
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
- Obstetrics and Gynecology Perinatal Research Laboratories, University of Wisconsin, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| | - Pamela J. Kling
- Departments of Pediatrics, University of Wisconsin, Neonatology, Meriter UnityPoint Hospital, 202 S. Park St., Madison, WI 53715, USA
| |
Collapse
|
48
|
Ozmen A, Terlemez S, Tunaoglu FS, Soysal S, Pektas A, Cilsal E, Koca U, Kula S, Deniz Oguz A. Evaluation of Neurodevelopment and Factors Affecting it in Children With Acyanotic Congenital Cardiac Disease. IRANIAN JOURNAL OF PEDIATRICS 2016; 26:e3278. [PMID: 26848374 PMCID: PMC4733290 DOI: 10.5812/ijp.3278] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/17/2015] [Accepted: 11/02/2015] [Indexed: 11/29/2022]
Abstract
Background: The rate of congenital heart disease is 0.8% in all live births. The majority of this, however, is acyanotic congenital heart disease. The survival rate of children with cardiac disease has increased with the developments provided in recent years and their lifetime is extended. Objectives: This study aims to evaluate neurodevelopment of children with uncomplicated acyanotic congenital heart disease in preschool period and determine the factors affecting their neurodevelopmental process. Patients and Methods: 132 children with acyanotic congenital heart disease aged 6 - 72 months were involved in the study. Mental development and intelligence levels of patients under 2 years old were assessed by using Bayley Development Scale-III, and Stanford Binet Intelligence test was employed for patients over 2 years old. Denver Developmental Screening Test II was applied to all patients for their personal-social, fine motor, gross motor and language development. Results: The average age of patients (67 girls, 65 boys) included in the study was 35.2 ± 19.6 months. It was determined that there were subnormal mental level in 13 (10%) patients and at least one specific developmental disorder in 33 (25%) patients. Bayley Mental Development Scale score of patients who had received incubator care in perinatal period was found significantly low (88 ± 4.2) compared to those with no incubator care (93.17 ± 8.5) (P = 0.028). Low educational level of father was established to be linked with low mental development scores at the age of 2 and following that age (P < 0.05). Iron deficiency anemia was discovered to be related to low psychometric test scores at every age (P < 0.05). Conclusions: Neurodevelopmental problems in children with acyanotic congenital heart disease were found higher compared to those in society. Mental development and intelligence levels of patients were determined to be closely associated with receiving incubator care, father’s educational level and iron deficiency anemia.
Collapse
Affiliation(s)
- Ayten Ozmen
- Department of Pediatrics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Semiha Terlemez
- Pediatric Cardiology Department, Faculty of Medicine, Gazi University, Ankara, Turkey
- Corresponding author: Semiha Terlemez, Pediatric Cardiology Department, Faculty of Medicine, Gazi University, Ankara, Turkey. Tel: +90-5327217689, Fax: +90-2562562573, E-mail:
| | - Fatma Sedef Tunaoglu
- Pediatric Cardiology Department, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Sebnem Soysal
- Department of Pediatrics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ayhan Pektas
- Pediatric Cardiology Department, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Erman Cilsal
- Pediatric Cardiology Department, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ulker Koca
- Pediatric Hematology Department, Faculty of Medicine Gazi University, Ankara, Turkey
| | - Serdar Kula
- Pediatric Cardiology Department, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ayse Deniz Oguz
- Pediatric Cardiology Department, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
49
|
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
|
50
|
Belke M, Heverhagen JT, Keil B, Rosenow F, Oertel WH, Stiasny-Kolster K, Knake S, Menzler K. DTI and VBM reveal white matter changes without associated gray matter changes in patients with idiopathic restless legs syndrome. Brain Behav 2015; 5:e00327. [PMID: 26442748 PMCID: PMC4589804 DOI: 10.1002/brb3.327] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 01/26/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND PURPOSE We evaluated cerebral white and gray matter changes in patients with iRLS in order to shed light on the pathophysiology of this disease. METHODS Twelve patients with iRLS were compared to 12 age- and sex-matched controls using whole-head diffusion tensor imaging (DTI) and voxel-based morphometry (VBM) techniques. Evaluation of the DTI scans included the voxelwise analysis of the fractional anisotropy (FA), radial diffusivity (RD), and axial diffusivity (AD). RESULTS Diffusion tensor imaging revealed areas of altered FA in subcortical white matter bilaterally, mainly in temporal regions as well as in the right internal capsule, the pons, and the right cerebellum. These changes overlapped with changes in RD. Voxel-based morphometry did not reveal any gray matter alterations. CONCLUSIONS We showed altered diffusion properties in several white matter regions in patients with iRLS. White matter changes could mainly be attributed to changes in RD, a parameter thought to reflect altered myelination. Areas with altered white matter microstructure included areas in the internal capsule which include the corticospinal tract to the lower limbs, thereby supporting studies that suggest changes in sensorimotor pathways associated with RLS.
Collapse
Affiliation(s)
- Marcus Belke
- Department of Neurology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
| | - Johannes T Heverhagen
- Department of Diagnostic Radiology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
| | - Boris Keil
- Department of Diagnostic Radiology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
| | - Felix Rosenow
- Department of Neurology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
- Epilepsy Center Franfurt Rhein-Main, Department of Neurology, Johann Wolfgang Goethe UniversityFrankfurt am Main, Germany
| | - Wolfgang H Oertel
- Department of Neurology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
| | - Karin Stiasny-Kolster
- Department of Neurology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
- Somnomar, Institute for Medical Research and Sleep Medicine MarburgMarburger Strasse 9a, Marburg, 35043, Germany
| | - Susanne Knake
- Department of Neurology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
| | - Katja Menzler
- Department of Neurology, Philipps-University MarburgBaldingerstrasse, Marburg, 35043, Germany
| |
Collapse
|