1
|
Lubrano C, Parisi F, Cetin I. Impact of Maternal Environment and Inflammation on Fetal Neurodevelopment. Antioxidants (Basel) 2024; 13:453. [PMID: 38671901 PMCID: PMC11047368 DOI: 10.3390/antiox13040453] [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: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
During intrauterine life, external stimuli including maternal nutrition, lifestyle, socioeconomic conditions, anxiety, stress, and air pollution can significantly impact fetal development. The human brain structures begin to form in the early weeks of gestation and continue to grow and mature throughout pregnancy. This review aims to assess, based on the latest research, the impact of environmental factors on fetal and neonatal brain development, showing that oxidative stress and inflammation are implied as a common factor for most of the stressors. Environmental insults can induce a maternal inflammatory state and modify nutrient supply to the fetus, possibly through epigenetic mechanisms, leading to significant consequences for brain morphogenesis and neurological outcomes. These risk factors are often synergic and mutually reinforcing. Fetal growth restriction and preterm birth represent paradigms of intrauterine reduced nutrient supply and inflammation, respectively. These mechanisms can lead to an increase in free radicals and, consequently, oxidative stress, with well-known adverse effects on the offspring's neurodevelopment. Therefore, a healthy intrauterine environment is a critical factor in supporting normal fetal brain development. Hence, healthcare professionals and clinicians should implement effective interventions to prevent and reduce modifiable risk factors associated with an increased inflammatory state and decreased nutrient supply during pregnancy.
Collapse
Affiliation(s)
- Chiara Lubrano
- Nutritional Sciences, Doctoral Programme (PhD), Università degli Studi di Milano, 20157 Milan, Italy;
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Parisi
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy;
| | - Irene Cetin
- Department of Mother, Child and Neonate, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157 Milan, Italy;
| |
Collapse
|
2
|
Yin R, Gao Q, Fu G, Zhao Q. The causal effect of iron status on risk of anxiety disorders: A two-sample Mendelian randomization study. PLoS One 2024; 19:e0300143. [PMID: 38547239 PMCID: PMC10977787 DOI: 10.1371/journal.pone.0300143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/21/2024] [Indexed: 04/02/2024] Open
Abstract
OBJECTIVES Observational studies had investigated the association of iron metabolism with anxiety disorders. The conclusions were inconsistent and not available to reveal the causal or reverse-causal association due to the confounding. In this study we estimated the potential causal effect of iron homeostasis markers on anxiety disorders using two-sample Mendelian randomization (MR) analysis. METHODS Summary data of single nucleotide polymorphisms (SNPs) associated with four iron-related biomarkers were extracted from a recent report about analysis of three genome-wide association study (GWAS), the sample size of which ranged from 131471 to 246139 individuals. The corresponding data for anxiety disorders were from Finngen database (20992 cases and 197800 controls). The analyses were mainly based on inverse variance weighted (IVW) method. In addition, the heterogeneity and pleiotropy of the results were assessed by Cochran's Q test and MR-Egger regression. RESULTS Basing on IVW method, genetically predicted serum iron level, ferritin and transferrin had negative effects on anxiety disorders. The odd ratios (OR) of anxiety disorders per 1 standard deviation (SD) unit increment in iron status biomarkers were 0.922 (95% confidence interval (CI) 0.862-0.986; p = 0.018) for serum iron level, 0.873 (95% CI 0.790-0.964; p = 0.008) for log-transformed ferritin and 0.917 (95% CI 0.867-0.969; p = 0.002) for transferrin saturation. But no statical significance was found in the association of 1 SD unit increased total iron-binding capacity (TIBC) with anxiety disorders (OR 1.080; 95% CI 0.988-1.180; p = 0.091). The analyses were supported by pleiotropy test which suggested no pleiotropic bias. CONCLUSION Our results indicated that genetically determined iron status biomarkers causally linked to the risk of anxiety disorders, providing valuable insights into the genetic research and clinical intervention of anxiety disorders.
Collapse
Affiliation(s)
- Ruiying Yin
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Qi Gao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Guangzhen Fu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Qiang Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
3
|
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] [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
|
4
|
LeVine SM. Examining the Role of a Functional Deficiency of Iron in Lysosomal Storage Disorders with Translational Relevance to Alzheimer's Disease. Cells 2023; 12:2641. [PMID: 37998376 PMCID: PMC10670892 DOI: 10.3390/cells12222641] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
The recently presented Azalea Hypothesis for Alzheimer's disease asserts that iron becomes sequestered, leading to a functional iron deficiency that contributes to neurodegeneration. Iron sequestration can occur by iron being bound to protein aggregates, such as amyloid β and tau, iron-rich structures not undergoing recycling (e.g., due to disrupted ferritinophagy and impaired mitophagy), and diminished delivery of iron from the lysosome to the cytosol. Reduced iron availability for biochemical reactions causes cells to respond to acquire additional iron, resulting in an elevation in the total iron level within affected brain regions. As the amount of unavailable iron increases, the level of available iron decreases until eventually it is unable to meet cellular demands, which leads to a functional iron deficiency. Normally, the lysosome plays an integral role in cellular iron homeostasis by facilitating both the delivery of iron to the cytosol (e.g., after endocytosis of the iron-transferrin-transferrin receptor complex) and the cellular recycling of iron. During a lysosomal storage disorder, an enzyme deficiency causes undigested substrates to accumulate, causing a sequelae of pathogenic events that may include cellular iron dyshomeostasis. Thus, a functional deficiency of iron may be a pathogenic mechanism occurring within several lysosomal storage diseases and Alzheimer's disease.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
Wang L, Xu R, Huang C, Yi G, Li Z, Zhang H, Ye R, Qi S, Huang G, Qu S. Targeting the ferroptosis crosstalk: novel alternative strategies for the treatment of major depressive disorder. Gen Psychiatr 2023; 36:e101072. [PMID: 37901286 PMCID: PMC10603325 DOI: 10.1136/gpsych-2023-101072] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/04/2023] [Indexed: 10/31/2023] Open
Abstract
Depression is a major contributor to poor global health and disability, with a recently increasing incidence. Although drug therapy is commonly used to treat depression, conventional antidepressant drugs have several disadvantages, including slow onset, low response rates and severe adverse effects. Therefore, developing effective therapies for depression remains challenging. Although various aetiological theories of depression exist, the underlying mechanisms of depression are complex, and further research is crucial. Moreover, oxidative stress (OS)-induced lipid peroxidation has been demonstrated to trigger ferroptosis. Both OS and ferroptosis are pivotal mechanisms implicated in the pathogenesis of neurological disorders, and investigation of the mediators involved in these processes has emerged as a prominent and active research direction. One previous study revealed that regulatory proteins involved in ferroptosis are implicated in the pathogenesis of depression, and antidepressant drugs could reverse depressive symptoms by inhibiting ferroptosis in vivo, suggesting an important role of ferroptosis in the pathogenesis of depression. Hence, our current comprehensive review offers an up-to-date perspective on the intricate mechanisms involved, specifically concerning ferroptosis and OS in the context of depression, along with promising prospects for using molecular mediators to target ferroptosis. We delineate the key targets of molecular mediators involved in OS and ferroptosis implicated in depression, most notably reactive oxygen species and iron overload. Considering the pivotal role of OS-induced ferroptosis in the pathogenesis of neurological disorders, delving deeper into the underlying subsequent mechanisms will contribute significantly to the identification of novel therapeutic targets for depression.
Collapse
Affiliation(s)
- Luyao Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Rongyang Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chengying Huang
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huayang Zhang
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Rongxu Ye
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shanqiang Qu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Cubello J, Peterson DR, Wang L, Mayer-Proschel M. Maternal Iron Deficiency and Environmental Lead (Pb) Exposure Alter the Predictive Value of Blood Pb Levels on Brain Pb Burden in the Offspring in a Dietary Mouse Model: An Important Consideration for Cumulative Risk in Development. Nutrients 2023; 15:4101. [PMID: 37836385 PMCID: PMC10574741 DOI: 10.3390/nu15194101] [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: 09/01/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Maternal iron deficiency (ID) and environmental lead (Pb) exposure are co-occurring insults that both affect the neurodevelopment of offspring. Few studies have investigated how ID affects brain-region-specific Pb accumulations using human-relevant Pb concentrations. Furthermore, how these Pb exposures impact blood and brain Fe levels remains unclear. Importantly, we also wanted to determine whether the use of blood Pb levels as a surrogate for the brain Pb burden is affected by underlying iron status. We exposed virgin Swiss Webster female mice to one of six conditions differing by iron diet and Pb water concentration (0 ppm, 19 ppm, or 50 ppm lead acetate) and used Inductively Coupled Plasma Mass Spectrometry to measure the maternal and offspring circulating, stored, and brain Pb levels. We found that maternal ID rendered the offspring iron-deficient anemic and led to a region-specific depletion of brain Fe that was exacerbated by Pb in a dose-specific manner. The postnatal iron deficiency anemia also exacerbated cortical and hippocampal Pb accumulation. Interestingly, BPb levels only correlated with the brain Pb burden in ID pups but not in IN offspring. We conclude that ID significantly increases the brain Pb burden and that BPb levels alone are insufficient as a clinical surrogate to make extrapolations on the brain Pb burden.
Collapse
Affiliation(s)
- Janine Cubello
- Department of Environmental Medicine, University of Rochester, Rochester, NY 14642, USA;
| | - Derick R. Peterson
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY 14642, USA; (D.R.P.); (L.W.)
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY 14642, USA; (D.R.P.); (L.W.)
| | - Margot Mayer-Proschel
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
7
|
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: 10] [Impact Index Per Article: 10.0] [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
|
8
|
Hua M, Shi D, Xu W, Zhu L, Hao X, Zhu B, Shu Q, Lozoff B, Geng F, Shao J. Differentiation between fetal and postnatal iron deficiency in altering brain substrates of cognitive control in pre-adolescence. BMC Med 2023; 21:167. [PMID: 37143078 PMCID: PMC10161450 DOI: 10.1186/s12916-023-02850-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Early iron deficiency (ID) is a common risk factor for poorer neurodevelopment, limiting children's potential and contributing to global burden. However, it is unclear how early ID alters the substrate of brain functions supporting high-order cognitive abilities and whether the timing of early ID matters in terms of long-term brain development. This study aimed to examine the effects of ID during fetal or early postnatal periods on brain activities supporting proactive and reactive cognitive control in pre-adolescent children. METHODS Participants were part of a longitudinal cohort enrolled at birth in southeastern China between December 2008 and November 2011. Between July 2019 and October 2021, 115 children aged 8-11 years were invited to participate in this neuroimaging study. Final analyses included 71 children: 20 with fetal ID, 24 with ID at 9 months (postnatal ID), and 27 iron-sufficient at birth and 9 months. Participants performed a computer-based behavioral task in a Magnetic Resonance Imaging scanner to measure proactive and reactive cognitive control. Outcome measures included accuracy, reaction times, and brain activity. Linear mixed modeling and the 3dlme command in Analysis of Functional NeuroImages (AFNI) were separately used to analyze behavioral performance and neuroimaging data. RESULTS Faster responses in proactive vs. reactive conditions indicated that all groups could use proactive or reactive cognitive control according to contextual demands. However, the fetal ID group was lower in general accuracy than the other 2 groups. Per the demands of cues and targets, the iron-sufficient group showed greater activation of wide brain regions in proactive vs. reactive conditions. In contrast, such condition differences were reversed in the postnatal ID group. Condition differences in brain activation, shown in postnatal ID and iron-sufficient groups, were not found in the fetal ID group. This group specifically showed greater activation of brain regions in the reward pathway in proactive vs. reactive conditions. CONCLUSIONS Early ID was associated with altered brain functions supporting proactive and reactive cognitive control in childhood. Alterations differed between fetal and postnatal ID groups. The findings imply that iron supplement alone is insufficient to prevent persisting brain alterations associated with early ID. Intervention strategies in addition to the iron supplement should consider ID timing.
Collapse
Affiliation(s)
- Mengdi Hua
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Donglin Shi
- Department of Curriculum and Learning Sciences, Zhejiang University, Hangzhou, China
| | - Wenwen Xu
- Department of Curriculum and Learning Sciences, Zhejiang University, Hangzhou, China
| | - Liuyan Zhu
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxin Hao
- Department of Curriculum and Learning Sciences, Zhejiang University, Hangzhou, China
| | - Bingquan Zhu
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Shu
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| | - Betsy Lozoff
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Fengji Geng
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Curriculum and Learning Sciences, Zhejiang University, Hangzhou, China.
- National Clinical Research Center for Child Health, Hangzhou, China.
| | - Jie Shao
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
9
|
Anas M, Diniz WJS, Menezes ACB, Reynolds LP, Caton JS, Dahlen CR, Ward AK. Maternal Mineral Nutrition Regulates Fetal Genomic Programming in Cattle: A Review. Metabolites 2023; 13:metabo13050593. [PMID: 37233634 DOI: 10.3390/metabo13050593] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Maternal mineral nutrition during the critical phases of fetal development may leave lifetime impacts on the productivity of an individual. Most research within the developmental origins of the health and disease (DOHaD) field is focused on the role of macronutrients in the genome function and programming of the developing fetus. On the other hand, there is a paucity of knowledge about the role of micronutrients and, specifically, minerals in regulating the epigenome of livestock species, especially cattle. Therefore, this review will address the effects of the maternal dietary mineral supply on the fetal developmental programming from the embryonic to the postnatal phases in cattle. To this end, we will draw a parallel between findings from our cattle model research with data from model animals, cell lines, and other livestock species. The coordinated role and function of different mineral elements in feto-maternal genomic regulation underlies the establishment of pregnancy and organogenesis and, ultimately, affects the development and functioning of metabolically important tissues, such as the fetal liver, skeletal muscle, and, importantly, the placenta. Through this review, we will delineate the key regulatory pathways involved in fetal programming based on the dietary maternal mineral supply and its crosstalk with epigenomic regulation in cattle.
Collapse
Affiliation(s)
- Muhammad Anas
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 36849, USA
| | | | - Ana Clara B Menezes
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA
| | - Lawrence P Reynolds
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 36849, USA
| | - Joel S Caton
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 36849, USA
| | - Carl R Dahlen
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND 36849, USA
| | - Alison K Ward
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| |
Collapse
|
10
|
Martínez-Iglesias O, Naidoo V, Corzo L, Pego R, Seoane S, Rodríguez S, Alcaraz M, Muñiz A, Cacabelos N, Cacabelos R. DNA Methylation as a Biomarker for Monitoring Disease Outcome in Patients with Hypovitaminosis and Neurological Disorders. Genes (Basel) 2023; 14:genes14020365. [PMID: 36833292 PMCID: PMC9956161 DOI: 10.3390/genes14020365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
DNA methylation remains an under-recognized diagnostic biomarker for several diseases, including neurodegenerative disorders. In this study, we examined differences in global DNA methylation (5mC) levels in serum samples from patients during the initial- and the follow-up visits. Each patient underwent a blood analysis and neuropsychological assessments. The analysis of 5mC levels revealed two categories of patients; Group A who, during the follow-up, had increased 5mC levels, and Group B who had decreased 5mC levels. Patients with low Fe-, folate-, and vitamin B12- levels during the initial visit showed increased levels of 5mC after treatment when assessed during the follow-up. During the follow-up, 5mC levels in Group A patients increased after treatment for hypovitaminosis with the nutraceutical compounds Animon Complex and MineraXin Plus. 5mC levels were maintained during the follow-up in Group A patients treated for neurological disorders with the bioproducts AtreMorine and NeoBrainine. There was a positive correlation between 5mC levels and MMSE scores, and an inverse correlation between 5mC and ADAS-Cog scores. This expected correlation was observed in Group A patients only. Our study appears to indicate that 5mC has a diagnostic value as a biomarker across different pathologies.
Collapse
|
11
|
Early life nutrition and brain development: breakthroughs, challenges and new horizons. Proc Nutr Soc 2022:1-9. [PMID: 36321424 DOI: 10.1017/s0029665122002774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The role of early life nutrition's impact on relevant health outcomes across the lifespan laid the foundation for the field titled the developmental origins of health and disease. Studies in this area initially concentrated on nutrition and the risk of adverse cardio-metabolic and cancer outcomes. More recently the role of nutrition in early brain development and the subsequent influence of later mental health has become more evident. Scientific breakthroughs have elucidated two mechanisms behind long-term nutrient effects on the brain, including the existence of critical periods for certain nutrients during brain development and nutrient-driven epigenetic modifications of chromatin. While multiple nutrients and nutritional conditions have the potential to modify brain development, iron can serve as a paradigm to understand both mechanisms. New horizons in nutritional medicine include leveraging the mechanistic knowledge of nutrient-brain interactions to propose novel nutritional approaches that protect the developing brain through better timing of nutrient delivery and potential reversal of negative epigenetic marks. The main challenge in the field is detecting whether a change in nutritional status truly affects the brain's development and performance in human subjects. To that end, a strong case can be made to develop and utilise bioindicators of a nutrient's effect on the developing brain instead of relying exclusively on biomarkers of the nutrient's status.
Collapse
|
12
|
Abstract
Growing evidence indicates that a suboptimal intrauterine environment confers risk for schizophrenia. The developmental model of schizophrenia posits that aberrant brain growth during early brain development and adolescence may interact to contribute to this psychiatric disease in adulthood. Although a variety of factors may perturb the environment of the developing fetus and predispose for schizophrenia later, a common mechanism has yet to be elucidated. Micronutrient deficiencies during the perinatal period are known to induce potent effects on brain development by altering neurodevelopmental processes. Iron is an important candidate nutrient to consider because of its role in energy metabolism, monoamine synthesis, synaptogenesis, myelination, and the high prevalence of iron deficiency (ID) in the mother-infant dyad. Understanding the current state of science regarding perinatal ID as an early risk factor for schizophrenia is imperative to inform empirical work investigating the etiology of schizophrenia and develop prevention and intervention programs. In this narrative review, we focus on perinatal ID as a common mechanism underlying the fetal programming of schizophrenia. First, we review the neural aberrations associated with perinatal ID that indicate risk for schizophrenia in adulthood, including disruptions in dopaminergic neurotransmission, hippocampal-dependent learning and memory, and sensorimotor gating. Second, we review the pathophysiology of perinatal ID as a function of maternal ID during pregnancy and use epidemiological and cohort studies to link perinatal ID with risk of schizophrenia. Finally, we review potential confounding phenotypes, including nonanemic causes of perinatal brain ID and future risk of schizophrenia.
Collapse
Affiliation(s)
- Andrea M. Maxwell
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN 55455 (USA)
| | - Raghavendra B. Rao
- Department of Pediatrics, Division of Neonatology, University of Minnesota Medical School, Minneapolis, MN 55455 (USA)
- Center for Neurobehavioral Development, University of Minnesota, Minneapolis, MN 55455 (USA)
| |
Collapse
|
13
|
Roth-Walter F. Iron-Deficiency in Atopic Diseases: Innate Immune Priming by Allergens and Siderophores. FRONTIERS IN ALLERGY 2022; 3:859922. [PMID: 35769558 PMCID: PMC9234869 DOI: 10.3389/falgy.2022.859922] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Although iron is one of the most abundant elements on earth, about a third of the world's population are affected by iron deficiency. Main drivers of iron deficiency are beside the chronic lack of dietary iron, a hampered uptake machinery as a result of immune activation. Macrophages are the principal cells distributing iron in the human body with their iron restriction skewing these cells to a more pro-inflammatory state. Consequently, iron deficiency has a pronounced impact on immune cells, favoring Th2-cell survival, immunoglobulin class switching and primes mast cells for degranulation. Iron deficiency during pregnancy increases the risk of atopic diseases in children, while both children and adults with allergy are more likely to have anemia. In contrast, an improved iron status seems to protect against allergy development. Here, the most important interconnections between iron metabolism and allergies, the effect of iron deprivation on distinct immune cell types, as well as the pathophysiology in atopic diseases are summarized. Although the main focus will be humans, we also compare them with innate defense and iron sequestration strategies of microbes, given, particularly, attention to catechol-siderophores. Similarly, the defense and nutritional strategies in plants with their inducible systemic acquired resistance by salicylic acid, which further leads to synthesis of flavonoids as well as pathogenesis-related proteins, will be elaborated as both are very important for understanding the etiology of allergic diseases. Many allergens, such as lipocalins and the pathogenesis-related proteins, are able to bind iron and either deprive or supply iron to immune cells. Thus, a locally induced iron deficiency will result in immune activation and allergic sensitization. However, the same proteins such as the whey protein beta-lactoglobulin can also transport this precious micronutrient to the host immune cells (holoBLG) and hinder their activation, promoting tolerance and protecting against allergy. Since 2019, several clinical trials have also been conducted in allergic subjects using holoBLG as a food for special medical purposes, leading to a reduction in the allergic symptom burden. Supplementation with nutrient-carrying lipocalin proteins can circumvent the mucosal block and nourish selectively immune cells, therefore representing a new dietary and causative approach to compensate for functional iron deficiency in allergy sufferers.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Franziska Roth-Walter ;
| |
Collapse
|
14
|
Taeubert MJ, de Prado-Bert P, Geurtsen ML, Mancano G, Vermeulen MJ, Reiss IKM, Caramaschi D, Sunyer J, Sharp GC, Julvez J, Muckenthaler MU, Felix JF. Maternal iron status in early pregnancy and DNA methylation in offspring: an epigenome-wide meta-analysis. Clin Epigenetics 2022; 14:59. [PMID: 35505416 PMCID: PMC9066980 DOI: 10.1186/s13148-022-01276-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Unbalanced iron homeostasis in pregnancy is associated with an increased risk of adverse birth and childhood health outcomes. DNA methylation has been suggested as a potential underlying mechanism linking environmental exposures such as micronutrient status during pregnancy with offspring health. We performed a meta-analysis on the association of maternal early-pregnancy serum ferritin concentrations, as a marker of body iron stores, and cord blood DNA methylation. We included 1286 mother-newborn pairs from two population-based prospective cohorts. Serum ferritin concentrations were measured in early pregnancy. DNA methylation was measured with the Infinium HumanMethylation450 BeadChip (Illumina). We examined epigenome-wide associations of maternal early-pregnancy serum ferritin and cord blood DNA methylation using robust linear regression analyses, with adjustment for confounders and performed fixed-effects meta-analyses. We additionally examined whether associations of any CpGs identified in cord blood persisted in the peripheral blood of older children and explored associations with other markers of maternal iron status. We also examined whether similar findings were present in the association of cord blood serum ferritin concentrations with cord blood DNA methylation. RESULTS Maternal early-pregnancy serum ferritin concentrations were inversely associated with DNA methylation at two CpGs (cg02806645 and cg06322988) in PRR23A and one CpG (cg04468817) in PRSS22. Associations at two of these CpG sites persisted at each of the follow-up time points in childhood. Cord blood serum ferritin concentrations were not associated with cord blood DNA methylation levels at the three identified CpGs. CONCLUSION Maternal early-pregnancy serum ferritin concentrations were associated with lower cord blood DNA methylation levels at three CpGs and these associations partly persisted in older children. Further studies are needed to uncover the role of these CpGs in the underlying mechanisms of the associations of maternal iron status and offspring health outcomes.
Collapse
Affiliation(s)
- M J Taeubert
- The Generation R Study Group, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Pediatric Oncology, Hematology and Immunology, University Medical Center Heidelberg, Heidelberg, Germany
| | - P de Prado-Bert
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - M L Geurtsen
- The Generation R Study Group, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Pediatrics, Sophia's Children's Hospital, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - G Mancano
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School Population Health Sciences, University of Bristol, Bristol, UK
| | - M J Vermeulen
- Department of Pediatrics, Sophia's Children's Hospital, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - I K M Reiss
- Department of Pediatrics, Sophia's Children's Hospital, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - D Caramaschi
- College of Life and Environmental Sciences, Psychology, University of Exeter, Exeter, UK
| | - J Sunyer
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - G C Sharp
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Bristol Medical School Population Health Sciences, University of Bristol, Bristol, UK
- School of Oral and Dental Sciences, University of Bristol, Bristol, UK
| | - J Julvez
- ISGlobal, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Institut d'Investigació Sanitària Pere Virgili, Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - M U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, University Medical Center Heidelberg, Heidelberg, Germany
| | - J F Felix
- The Generation R Study Group, Erasmus University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Department of Pediatrics, Sophia's Children's Hospital, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
15
|
Porras CA, Rouault TA. Iron Homeostasis in the CNS: An Overview of the Pathological Consequences of Iron Metabolism Disruption. Int J Mol Sci 2022; 23:ijms23094490. [PMID: 35562883 PMCID: PMC9104368 DOI: 10.3390/ijms23094490] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 11/21/2022] Open
Abstract
Iron homeostasis disruption has increasingly been implicated in various neurological disorders. In this review, we present an overview of our current understanding of iron metabolism in the central nervous system. We examine the consequences of both iron accumulation and deficiency in various disease contexts including neurodegenerative, neurodevelopmental, and neuropsychological disorders. The history of animal models of iron metabolism misregulation is also discussed followed by a comparison of three patients with a newly discovered neurodegenerative disorder caused by mutations in iron regulatory protein 2.
Collapse
|
16
|
Lien YC, Pinney SE, Lu XM, Simmons RA. Identification of Novel Regulatory Regions Induced by Intrauterine Growth Restriction in Rat Islets. Endocrinology 2022; 163:6459683. [PMID: 34894232 PMCID: PMC8743043 DOI: 10.1210/endocr/bqab251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Indexed: 01/05/2023]
Abstract
Intrauterine growth restriction (IUGR) leads to the development of type 2 diabetes in adulthood, and the permanent alterations in gene expression implicate an epigenetic mechanism. Using a rat model of IUGR, we performed TrueSeq-HELP Tagging to assess the association of DNA methylation changes and gene dysregulation in islets. We identified 511 differentially methylated regions (DMRs) and 4377 significantly altered single CpG sites. Integrating the methylome and our published transcriptome data sets resulted in the identification of pathways critical for islet function. The identified DMRs were enriched with transcription factor binding motifs, such as Elk1, Etv1, Foxa1, Foxa2, Pax7, Stat3, Hnf1, and AR. In silico analysis of 3-dimensional chromosomal interactions using human pancreas and islet Hi-C data sets identified interactions between 14 highly conserved DMRs and 35 genes with significant expression changes at an early age, many of which persisted in adult islets. In adult islets, there were far more interactions between DMRs and genes with significant expression changes identified with Hi-C, and most of them were critical to islet metabolism and insulin secretion. The methylome was integrated with our published genome-wide histone modification data sets from IUGR islets, resulting in further characterization of important regulatory regions of the genome altered by IUGR containing both significant changes in DNA methylation and specific histone marks. We identified novel regulatory regions in islets after exposure to IUGR, suggesting that epigenetic changes at key transcription factor binding motifs and other gene regulatory regions may contribute to gene dysregulation and an abnormal islet phenotype in IUGR rats.
Collapse
Affiliation(s)
- Yu-Chin Lien
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Sara E Pinney
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Xueqing Maggie Lu
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Correspondence: Rebecca A. Simmons, MD, Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, BRB II/III, 13th Fl, Rm 1308, 421 Curie Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
17
|
Erber L, Liu S, Gong Y, Tran P, Chen Y. Quantitative Proteome and Transcriptome Dynamics Analysis Reveals Iron Deficiency Response Networks and Signature in Neuronal Cells. Molecules 2022; 27:484. [PMID: 35056799 PMCID: PMC8779535 DOI: 10.3390/molecules27020484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 01/17/2023] Open
Abstract
Iron and oxygen deficiencies are common features in pathophysiological conditions, such as ischemia, neurological diseases, and cancer. Cellular adaptive responses to such deficiencies include repression of mitochondrial respiration, promotion of angiogenesis, and cell cycle control. We applied a systematic proteomics analysis to determine the global proteomic changes caused by acute hypoxia and chronic and acute iron deficiency (ID) in hippocampal neuronal cells. Our analysis identified over 8600 proteins, revealing similar and differential effects of each treatment on activation and inhibition of pathways regulating neuronal development. In addition, comparative analysis of ID-induced proteomics changes in cultured cells and transcriptomic changes in the rat hippocampus identified common altered pathways, indicating specific neuronal effects. Transcription factor enrichment and correlation analysis identified key transcription factors that were activated in both cultured cells and tissue by iron deficiency, including those implicated in iron regulation, such as HIF1, NFY, and NRF1. We further identified MEF2 as a novel transcription factor whose activity was induced by ID in both HT22 proteome and rat hippocampal transcriptome, thus linking iron deficiency to MEF2-dependent cellular signaling pathways in neuronal development. Taken together, our study results identified diverse signaling networks that were differentially regulated by hypoxia and ID in neuronal cells.
Collapse
Affiliation(s)
- Luke Erber
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.E.); (Y.G.)
| | - Shirelle Liu
- Department of Pediatrics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA;
| | - Yao Gong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.E.); (Y.G.)
| | - Phu Tran
- Department of Pediatrics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA;
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.E.); (Y.G.)
| |
Collapse
|
18
|
Prenatal Iron Deficiency and Choline Supplementation Interact to Epigenetically Regulate Jarid1b and Bdnf in the Rat Hippocampus into Adulthood. Nutrients 2021; 13:nu13124527. [PMID: 34960080 PMCID: PMC8706459 DOI: 10.3390/nu13124527] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 11/24/2022] Open
Abstract
Early-life iron deficiency (ID) causes long-term neurocognitive impairments and gene dysregulation that can be partially mitigated by prenatal choline supplementation. The long-term gene dysregulation is hypothesized to underlie cognitive dysfunction. However, mechanisms by which iron and choline mediate long-term gene dysregulation remain unknown. In the present study, using a well-established rat model of fetal-neonatal ID, we demonstrated that ID downregulated hippocampal expression of the gene encoding JmjC-ARID domain-containing protein 1B (JARID1B), an iron-dependent histone H3K4 demethylase, associated with a higher histone deacetylase 1 (HDAC1) enrichment and a lower enrichment of acetylated histone H3K9 (H3K9ac) and phosphorylated cAMP response element-binding protein (pCREB). Likewise, ID reduced transcriptional capacity of the gene encoding brain-derived neurotrophic factor (BDNF), a target of JARID1B, associated with repressive histone modifications such as lower H3K9ac and pCREB enrichments at the Bdnf promoters in the adult rat hippocampus. Prenatal choline supplementation did not prevent the ID-induced chromatin modifications at these loci but induced long-lasting repressive chromatin modifications in the iron-sufficient adult rats. Collectively, these findings demonstrated that the iron-dependent epigenetic mechanism mediated by JARID1B accounted for long-term Bdnf dysregulation by early-life ID. Choline supplementation utilized a separate mechanism to rescue the effect of ID on neural gene regulation. The negative epigenetic effects of choline supplementation in the iron-sufficient rat hippocampus necessitate additional investigations prior to its use as an adjunctive therapeutic agent.
Collapse
|
19
|
Early-Life Iron Deficiency Anemia Programs the Hippocampal Epigenomic Landscape. Nutrients 2021; 13:nu13113857. [PMID: 34836113 PMCID: PMC8623089 DOI: 10.3390/nu13113857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023] Open
Abstract
Iron deficiency (ID) anemia is the foremost micronutrient deficiency worldwide, affecting around 40% of pregnant women and young children. ID during the prenatal and early postnatal periods has a pronounced effect on neurodevelopment, resulting in long-term effects such as cognitive impairment and increased risk for neuropsychiatric disorders. Treatment of ID has been complicated as it does not always resolve the long-lasting neurodevelopmental deficits. In animal models, developmental ID results in abnormal hippocampal structure and function associated with dysregulation of genes involved in neurotransmission and synaptic plasticity. Dysregulation of these genes is a likely proximate cause of the life-long deficits that follow developmental ID. However, a direct functional link between iron and gene dysregulation has yet to be elucidated. Iron-dependent epigenetic modifications are one mechanism by which ID could alter gene expression across the lifespan. The jumonji and AT-rich interaction domain-containing (JARID) protein and the Ten-Eleven Translocation (TET) proteins are two families of iron-dependent epigenetic modifiers that play critical roles during neural development by establishing proper gene regulation during critical periods of brain development. Therefore, JARIDs and TETs can contribute to the iron-mediated epigenetic mechanisms by which early-life ID directly causes stable changes in gene regulation across the life span.
Collapse
|
20
|
Shah HE, Bhawnani N, Ethirajulu A, Alkasabera A, Onyali CB, Anim-Koranteng C, Mostafa JA. Iron Deficiency-Induced Changes in the Hippocampus, Corpus Striatum, and Monoamines Levels That Lead to Anxiety, Depression, Sleep Disorders, and Psychotic Disorders. Cureus 2021; 13:e18138. [PMID: 34692346 PMCID: PMC8525689 DOI: 10.7759/cureus.18138] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/20/2021] [Indexed: 01/09/2023] Open
Abstract
Iron deficiency anemia caused by severe iron deficiency in infancy is associated with poor health and severe neurological impairment such as mental, motor, social, emotional, neurophysiological, and neurocognitive dysfunction. The behavioral effects of iron deficiency can present themselves in infancy, but they are also found in adulthood. Some behaviors can start in childhood but persist throughout adulthood. The behaviors that are particularly often seen in infants and children include wariness and hesitance, lack of positive affect, and diminished social engagement. The affected behaviors in adults include anxiety, depression, higher complex cogitative tasks, and other psychological disorders. The mechanisms of how iron deficiency affects behavior include affecting the hippocampus, the corpus striatum, and certain neurotransmitters. The hippocampus is a brain region that is essential for memory, learning, and other purposes. The hippocampus is very sensitive to lack of Iron during early development. The corpus striatum dispatches dopamine-rich projects to the prefrontal cortex, and it is involved in controlling executive activities such as planning, inhibitory control, sustained attention, working memory, regulation of emotion, memory storage and retrieval, motivation, and reward. Iron deficiency has been known to cause changes in behavioral and developmental aspects by affecting neurotransmitters such as serotonin, noradrenaline, and dopamine. Iron deficiency causes behavior changes that can present in infancy and, even if corrected postnatally, it can have long-lasting effects well into adulthood.
Collapse
Affiliation(s)
- Hira E Shah
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nitin Bhawnani
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aarthi Ethirajulu
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Almothana Alkasabera
- General Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Chike B Onyali
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | - Jihan A Mostafa
- Psychiatry, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
21
|
Abstract
Anemia is defined as a low red blood cell count, a low hematocrit, or a low hemoglobin concentration. In pregnancy, a hemoglobin concentration of less than 11.0 g/dL in the first trimester and less than 10.5 or 11.0 g/dL in the second or third trimester (depending on the guideline used) is considered anemia. Anemia is the most common hematologic abnormality in pregnancy. Maternal anemia is associated with adverse fetal, neonatal and childhood outcomes, but causality is not established. Maternal anemia increases the likelihood of transfusion at delivery. Besides hemodilution, iron deficiency is the most common cause of anemia in pregnancy. The American College of Obstetricians and Gynecologists recommends screening for anemia with a complete blood count in the first trimester and again at 24 0/7 to 28 6/7 weeks of gestation. Mild anemia, with a hemoglobin of 10.0 g/dL or higher and a mildly low or normal mean corpuscular volume (MCV) is likely iron deficiency anemia. A trial of oral iron can be both diagnostic and therapeutic. Mild anemia with a very low MCV, macrocytic anemia, moderate anemia (hemoglobin 7.0-9.9 g/dL) or severe anemia (hemoglobin 4.0-6.9 g/dL) requires further investigation. Once a diagnosis of iron deficiency anemia is confirmed, first-line treatment is oral iron. New evidence suggests that intermittent dosing is as effective as daily or twice-daily dosing with fewer side effects. For patients with iron deficiency anemia who cannot tolerate, cannot absorb, or do not respond to oral iron, intravenous iron is preferred. With contemporary formulations, allergic reactions are rare.
Collapse
|
22
|
Erber LN, Luo A, Gong Y, Beeson M, Tu M, Tran P, Chen Y. Iron Deficiency Reprograms Phosphorylation Signaling and Reduces O-GlcNAc Pathways in Neuronal Cells. Nutrients 2021; 13:E179. [PMID: 33430126 PMCID: PMC7826960 DOI: 10.3390/nu13010179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 01/14/2023] Open
Abstract
Micronutrient sensing is critical for cellular growth and differentiation. Deficiencies in essential nutrients such as iron strongly affect neuronal cell development and may lead to defects in neuronal function that cannot be remedied by subsequent iron supplementation. To understand the adaptive intracellular responses to iron deficiency in neuronal cells, we developed and utilized a Stable Isotopic Labeling of Amino acids in Cell culture (SILAC)-based quantitative phosphoproteomics workflow. Our integrated approach was designed to comprehensively elucidate the changes in phosphorylation signaling under both acute and chronic iron-deficient cell models. In addition, we analyzed the differential cellular responses between iron deficiency and hypoxia (oxygen-deprived) in neuronal cells. Our analysis identified nearly 16,000 phosphorylation sites in HT-22 cells, a hippocampal-derived neuronal cell line, more than ten percent of which showed at least 2-fold changes in response to either hypoxia or acute/chronic iron deficiency. Bioinformatic analysis revealed that iron deficiency altered key metabolic and epigenetic pathways including the phosphorylation of proteins involved in iron sequestration, glutamate metabolism, and histone methylation. In particular, iron deficiency increased glutamine-fructose-6-phosphate transaminase (GFPT1) phosphorylation, which is a key enzyme in the glucosamine biosynthesis pathway and a target of 5' AMP-activated protein kinase (AMPK), leading to reduced GFPT1 enzymatic activity and consequently lower global O-GlcNAc modification in neuronal cells. Taken together, our analysis of the phosphoproteome dynamics in response to iron and oxygen deprivation demonstrated an adaptive cellular response by mounting post-translational modifications that are critical for intracellular signaling and epigenetic programming in neuronal cells.
Collapse
Affiliation(s)
- Luke N. Erber
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.N.E.); (A.L.); (Y.G.); (M.T.)
| | - Ang Luo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.N.E.); (A.L.); (Y.G.); (M.T.)
| | - Yao Gong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.N.E.); (A.L.); (Y.G.); (M.T.)
| | - Montana Beeson
- Department of Pediatrics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (M.B.); (P.T.)
| | - Maolin Tu
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.N.E.); (A.L.); (Y.G.); (M.T.)
| | - Phu Tran
- Department of Pediatrics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (M.B.); (P.T.)
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.N.E.); (A.L.); (Y.G.); (M.T.)
| |
Collapse
|
23
|
Paredes DA, Jalloh A, Catlow BJ, Jaishankar A, Seo S, Jimenez DV, Martinowich K, Diaz-Bustamante M, Hoeppner DJ, McKay RDG. Bdnf deficiency in the neonatal hippocampus contributes to global dna hypomethylation and adult behavioral changes. Brain Res 2021; 1754:147254. [PMID: 33422542 DOI: 10.1016/j.brainres.2020.147254] [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: 05/06/2020] [Revised: 10/13/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
Schizophrenia is a neurodevelopmental psychiatric disorder, encompassing genetic and environmental risk factors. For several decades, investigators have been implementing the use of lesions of the neonatal rodent hippocampus to model schizophrenia, resulting in a broad spectrum of adult schizophrenia-related behavioral changes. Despite the extensive use of these proposed animal models of schizophrenia, the mechanisms by which these lesions result in schizophrenia-like behavioral alterations remain unclear. Here we provide in vivo evidence that transient pharmacological inactivation of the hippocampus via tetrodotoxin microinjections or a genetic reduction in brain derived neurotrophic factor (BDNF) protein levels (BDNF+/- rats) lead to global DNA hypomethylation, disrupted maturation of the neuronal nucleus and aberrant acoustic startle response in the adult rat. The similarity between the effects of the two treatments strongly indicate that BDNF signaling is involved in effects obtained after the TTX microinjections. These findings may shed light on the cellular mechanisms underlying the phenotypical features of neonatal transient inhibition of the hippocampus as a preclinical model of schizophrenia and suggest that BDNF signaling represents a target pathway for development of novel treatment therapies.
Collapse
Affiliation(s)
- Daniel A Paredes
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA; Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.
| | - Ahmad Jalloh
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA; Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Briony J Catlow
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA; Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Amritha Jaishankar
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Seungmae Seo
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA; Department of Pediatrics, Columbia University, New York, NY, USA
| | - Dennisse V Jimenez
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Marcelo Diaz-Bustamante
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA; Department of Physiology, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel J Hoeppner
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Ronald D G McKay
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| |
Collapse
|
24
|
Franco B, Cavallaro LAR, Mota DS, Rodrigues NDA, Manchado-Gobatto FDB, Bezerra RMN, Esteves AM. Differences in iron intake during pregnancy influence in trainability response of male rat offspring. EINSTEIN-SAO PAULO 2020; 18:eAO5665. [PMID: 33295427 PMCID: PMC7690933 DOI: 10.31744/einstein_journal/2020ao5665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/29/2020] [Indexed: 11/18/2022] Open
Abstract
Objective: To evaluate if different concentrations of iron in diets during pregnancy would interfere in the aerobic and anaerobic performance of the offspring, observed during 8-week swimming training and measured by lactate minimum test. Methods: Pregnant rats were divided into four groups with different dietary iron concentrations: standard (40mg/kg), supplementation (100mg/kg), restriction since weaning, and restriction only during pregnancy (4mg/kg). After birth, the offspring were assigned to their respective groups (Standard Offspring, Supplementation Offspring, Restriction Offspring or Restriction Offspring 2). The lactate minimum test was performed at three time points: before starting exercise training, after 4 weeks and after 8 weeks of exercise training. Results: The Restriction Offspring Group had a significant reduction in the concentration of lactate minimum and in swimming time to exhaustion, after 4 and 8 weeks of training as compared to before training. Therefore, the results showed the Restriction Offspring Group was not able to maintain regularity during training in lactate minimum tests. Conclusion: Our results suggested the Restriction Offspring Group showed a marked decrease in its performance parameters, which may have occurred due to iron restriction.
Collapse
Affiliation(s)
- Beatriz Franco
- Faculdade de Educação Física, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | | | - Diego Silva Mota
- Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, SP, Brazil
| | | | - Fúlvia de Barros Manchado-Gobatto
- Faculdade de Educação Física, Universidade Estadual de Campinas, Campinas, SP, Brazil.,Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, SP, Brazil
| | | | - Andrea Maculano Esteves
- Faculdade de Educação Física, Universidade Estadual de Campinas, Campinas, SP, Brazil.,Faculdade de Ciências Aplicadas, Universidade Estadual de Campinas, Limeira, SP, Brazil
| |
Collapse
|
25
|
Forgie AJ, Drall KM, Bourque SL, Field CJ, Kozyrskyj AL, Willing BP. The impact of maternal and early life malnutrition on health: a diet-microbe perspective. BMC Med 2020; 18:135. [PMID: 32393275 PMCID: PMC7216331 DOI: 10.1186/s12916-020-01584-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Early-life malnutrition may have long-lasting effects on microbe-host interactions that affect health and disease susceptibility later in life. Diet quality and quantity in conjunction with toxin and pathogen exposure are key contributors to microbe-host physiology and malnutrition. Consequently, it is important to consider both diet- and microbe-induced pathologies as well as their interactions underlying malnutrition. MAIN BODY Gastrointestinal immunity and digestive function are vital to maintain a symbiotic relationship between the host and microbiota. Childhood malnutrition can be impacted by numerous factors including gestational malnutrition, early life antibiotic use, psychological stress, food allergy, hygiene, and exposure to other chemicals and pollutants. These factors can contribute to reoccurring environmental enteropathy, a condition characterized by the expansion of commensal pathobionts and environmental pathogens. Reoccurring intestinal dysfunction, particularly during the critical window of development, may be a consequence of diet-microbe interactions and may lead to life-long immune and metabolic programming and increased disease risk. We provide an overview of the some key factors implicated in the progression of malnutrition (protein, fat, carbohydrate, iron, vitamin D, and vitamin B12) and discuss the microbiota during early life that may contribute health risk later in life. CONCLUSION Identifying key microbe-host interactions, particularly those associated with diet and malnutrition requires well-controlled dietary studies. Furthering our understanding of diet-microbe-host interactions will help to provide better strategies during gestation and early life to promote health later in life.
Collapse
Affiliation(s)
- Andrew J. Forgie
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta Canada
| | - Kelsea M. Drall
- Department of Pediatrics, University of Alberta, Edmonton, Alberta Canada
| | - Stephane L. Bourque
- Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta Canada
| | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta Canada
| | - Anita L. Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, Alberta Canada
| | - Benjamin P. Willing
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta Canada
| |
Collapse
|
26
|
Arija V, Hernández-Martínez C, Tous M, Canals J, Guxens M, Fernández-Barrés S, Ibarluzea J, Babarro I, Soler-Blasco R, Llop S, Vioque J, Sunyer J, Julvez J. Association of Iron Status and Intake During Pregnancy with Neuropsychological Outcomes in Children Aged 7 Years: The Prospective Birth Cohort Infancia y Medio Ambiente (INMA) Study. Nutrients 2019; 11:nu11122999. [PMID: 31817835 PMCID: PMC6949977 DOI: 10.3390/nu11122999] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 01/14/2023] Open
Abstract
Early iron status plays an important role in prenatal neurodevelopment. Iron deficiency and high iron status have been related to alterations in child cognitive development; however, there are no data about iron intake during pregnancy with other environmental factors in relation to long term cognitive functioning of children. The aim of this study is to assess the relationship between maternal iron status and iron intake during pregnancy and child neuropsychological outcomes at 7 years of age. We used data from the INMA Cohort population-based study. Iron status during pregnancy was assessed according to serum ferritin levels, and iron intake was assessed with food frequency questionnaires. Working memory, attention, and executive function were assessed in children at 7 years old with the N-Back task, Attention Network Task, and the Trail Making Test, respectively. The results show that, after controlling for potential confounders, normal maternal serum ferritin levels (from 12 mg/L to 60 mg/L) and iron intake (from 14.5 mg/day to 30.0 mg/day), respectively, were related to better scores in working memory and executive functioning in offspring. Since these functions have been associated with better academic performance and adaptation to the environment, maintaining a good state of maternal iron from the beginning of pregnancy could be a valuable strategy for the community.
Collapse
Affiliation(s)
- Victoria Arija
- Nutrition and Public Health Unit, Research Group on Nutrition and Mental Health (NUTRISAM), Faculty of Medicine and Health Science, Universitat Rovira i Virgili, 43201 Reus, Spain; (V.A.); (C.H.-M.); (M.T.); (J.C.)
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Carmen Hernández-Martínez
- Nutrition and Public Health Unit, Research Group on Nutrition and Mental Health (NUTRISAM), Faculty of Medicine and Health Science, Universitat Rovira i Virgili, 43201 Reus, Spain; (V.A.); (C.H.-M.); (M.T.); (J.C.)
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43003 Tarragona, Spain
- Department of Psychology, Research Center for Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Mónica Tous
- Nutrition and Public Health Unit, Research Group on Nutrition and Mental Health (NUTRISAM), Faculty of Medicine and Health Science, Universitat Rovira i Virgili, 43201 Reus, Spain; (V.A.); (C.H.-M.); (M.T.); (J.C.)
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Josefa Canals
- Nutrition and Public Health Unit, Research Group on Nutrition and Mental Health (NUTRISAM), Faculty of Medicine and Health Science, Universitat Rovira i Virgili, 43201 Reus, Spain; (V.A.); (C.H.-M.); (M.T.); (J.C.)
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43003 Tarragona, Spain
- Department of Psychology, Research Center for Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Mónica Guxens
- ISGlobal- Instituto de Salud Global de Barcelona, 08036 Barcelona, Spain; (M.G.); (S.F.-B.); (J.S.)
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain; (J.I.); (J.V.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706 Santiago de Compostela, Spain
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Centre-Sophia Children’s Hospital, 3000CD Rotterdam, The Netherlands
| | - Silvia Fernández-Barrés
- ISGlobal- Instituto de Salud Global de Barcelona, 08036 Barcelona, Spain; (M.G.); (S.F.-B.); (J.S.)
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain; (J.I.); (J.V.)
| | - Jesús Ibarluzea
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain; (J.I.); (J.V.)
- Department of Health, Public Health Division of Gipuzkoa, 20014 San Sebastian, Spain;
- BIODONOSTIA Health Research Institute, 20014 San Sebastian, Spain
- Faculty of Psychology, University of the Basque Country (UPV/EHU), 20018 San Sebastian, Spain
| | - Izaro Babarro
- Department of Health, Public Health Division of Gipuzkoa, 20014 San Sebastian, Spain;
| | - Raquel Soler-Blasco
- Epidemiology and Environmental Health Joint Research Unit, FISABIO−Universitat Jaume I−Universitat de València, 46010 Valencia, Spain; (R.S.-B.); (S.L.)
| | - Sabrina Llop
- Epidemiology and Environmental Health Joint Research Unit, FISABIO−Universitat Jaume I−Universitat de València, 46010 Valencia, Spain; (R.S.-B.); (S.L.)
| | - Jesús Vioque
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain; (J.I.); (J.V.)
- Unit of Nutritional Epidemiology, Universidad Miguel Hernandez, 03550 Alicante, Spain
| | - Jordi Sunyer
- ISGlobal- Instituto de Salud Global de Barcelona, 08036 Barcelona, Spain; (M.G.); (S.F.-B.); (J.S.)
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain; (J.I.); (J.V.)
| | - Jordi Julvez
- Pere Virgili Institute for Health Research (IISPV), Universitat Rovira i Virgili, 43003 Tarragona, Spain
- ISGlobal- Instituto de Salud Global de Barcelona, 08036 Barcelona, Spain; (M.G.); (S.F.-B.); (J.S.)
- Biomedical Research Centre Network for Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain; (J.I.); (J.V.)
- Correspondence: ; Tel.: +31-932-147-349
| |
Collapse
|