Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Nutr ; 154(3): 875-885, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38072152

ABSTRACT

BACKGROUND: The current pediatric practice of monitoring for infantile iron deficiency (ID) via hemoglobin (Hgb) screening at one y of age does not identify preanemic ID nor protect against later neurocognitive deficits. OBJECTIVES: To identify biomarkers of iron-related metabolic alterations in the serum and brain and determine the sensitivity of conventional iron and heme indices for predicting risk of brain metabolic dysfunction using a nonhuman primate model of infantile ID. METHODS: Simultaneous serum iron and RBC indices, and serum and cerebrospinal fluid (CSF) metabolomic profiles were determined in 20 rhesus infants, comparing iron sufficient (IS; N = 10) and ID (N = 10) infants at 2 and 4 mo of age. RESULTS: Reticulocyte hemoglobin (RET-He) was lower at 2 wk in the ID group. Significant IS compared with ID differences in serum iron indices were present at 2 mo, but Hgb and RBC indices differed only at 4 mo (P < 0.05). Serum and CSF metabolomic profiles of the ID and IS groups differed at 2 and 4 mo (P < 0.05). Key metabolites, including homostachydrine and stachydrine (4-5-fold lower at 4 mo in ID group, P < 0.05), were altered in both serum and CSF. Iron indices and RET-He at 2 mo, but not Hgb or other RBC indices, were correlated with altered CSF metabolic profile at 4 mo and had comparable predictive accuracy (area under the receiver operating characteristic curve scores, 0.75-0.80). CONCLUSIONS: Preanemic ID at 2 mo was associated with metabolic alterations in serum and CSF in infant monkeys. Among the RBC indices, only RET-He predicted the future risk of abnormal CSF metabolic profile with a predictive accuracy comparable to serum iron indices. The concordance of homostachydrine and stachydrine changes in serum and CSF indicates their potential use as early biomarkers of brain metabolic dysfunction in infantile ID.


Subject(s)
Anemia, Iron-Deficiency , Brain Diseases , Iron Deficiencies , Animals , Infant , Humans , Child , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/diagnosis , Macaca mulatta/metabolism , Prognosis , Iron/metabolism , Hemoglobins/metabolism , Brain Diseases/metabolism , Biomarkers , Brain/metabolism
2.
WMJ ; 122(4): 250-256, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37768764

ABSTRACT

INTRODUCTION: To promote scientific inquiry, medical schools encourage medical students to participate in scholarly concentration programs (SCP). Manuscript publishing, a proxy of productivity, enhances medical student understanding of scientific inquiry. To evaluate an elective medical SCP offered between the first two years of medical school, the pediatrician authors' primary aim was to study the publishing productivity of the program participants in the University of Wisconsin (UW) School of Medicine and Public Health Department of Pediatrics compared to other departments. Secondary aims were to study whether productivity was influenced by the following predictors: (1) self-identified medical student gender, (2) working with a frequent mentor, (3) mentor degree, (4) funding source, and (5) area of research. METHODS: PubMed joint publications from 2002 through 2017 were searched using both medical student and mentor names through 2 years post-graduation. RESULTS: From all UW School of Medicine Public Health departments, 1108 medical students self-selected projects and mentors. One hundred two (9.2%) students chose the Department of Pediatrics. The majority of these students were female (61%) compared to female medical student participation (42%) in other departments (P = 0.0004). The majority of projects were clinical (53%), with basic science (26%) and public/global health (21%) following, though with more public/global health projects chosen in the Department of Pediatrics (P = 0.002) versus other departments. Overall, frequent mentors improved publication rates (P =0.0008), though frequent mentors (P = 0.45) and publication rates (P = 0.60) did not differ between pediatrics and other departments. CONCLUSIONS: Medical students' SCP manuscript productivity benefitted from working with frequent mentors, but productivity in the Department of Pediatrics did not differ from other departments.


Subject(s)
Biomedical Research , Medicine , Students, Medical , Humans , Male , Female , Child , Schools, Medical , Mentors
3.
J Nutr ; 153(1): 148-157, 2023 01.
Article in English | MEDLINE | ID: mdl-36913448

ABSTRACT

BACKGROUND: Infantile iron deficiency (ID) causes anemia and compromises neurodevelopment. Current screening relies on hemoglobin (Hgb) determination at 1 year of age, which lacks sensitivity and specificity for timely detection of infantile ID. Low reticulocyte Hgb equivalent (RET-He) indicates ID, but its predictive accuracy relative to conventional serum iron indices is unknown. OBJECTIVES: The objective was to compare diagnostic accuracies of iron indices, red blood cell (RBC) indices, and RET-He for predicting the risk of ID and IDA in a nonhuman primate model of infantile ID. METHODS: Serum iron, total iron binding capacity, unsaturated iron binding capacity, transferrin saturation (TSAT), Hgb, RET-He, and other RBC indices were determined at 2 wk and 2, 4, and 6 mo in breastfed male and female rhesus infants (N = 54). The diagnostic accuracies of RET-He, iron, and RBC indices for predicting the development of ID (TSAT < 20%) and IDA (Hgb < 10 g/dL + TSAT < 20%) were determined using t tests, area under the receiver operating characteristic curve (AUC) analysis, and multiple regression models. RESULTS: Twenty-three (42.6%) infants developed ID and 16 (29.6%) progressed to IDA. All 4 iron indices and RET-He, but not Hgb or RBC indices, predicted future risk of ID and IDA (P < 0.001). The predictive accuracy of RET-He (AUC = 0.78, SE = 0.07; P = 0.003) for IDA was comparable to that of the iron indices (AUC = 0.77-0.83, SE = 0.07; P ≤ 0.002). A RET-He threshold of 25.5 pg strongly correlated with TSAT < 20% and correctly predicted IDA in 10 of 16 infants (sensitivity: 62.5%) and falsely predicted possibility of IDA in only 4 of 38 unaffected infants (specificity: 89.5%). CONCLUSIONS: RET-He is a biomarker of impending ID/IDA in rhesus infants and can be used as a hematological parameter to screen for infantile ID.


Subject(s)
Anemia, Iron-Deficiency , Anemia , Iron Deficiencies , Male , Female , Animals , Reticulocytes/chemistry , Reticulocytes/metabolism , Anemia/metabolism , Hemoglobins/metabolism , Iron/metabolism , Primates/metabolism
4.
Data Brief ; 45: 108591, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36164307

ABSTRACT

The effects of early-life iron deficiency anemia (IDA) extend past the blood and include both short- and long-term adverse effects on many tissues including the brain. Prior to IDA, iron deficiency (ID) can cause similar tissue effects, but a sensitive biomarker of iron-dependent brain health is lacking. To determine serum and CSF biomarkers of ID-induced metabolic dysfunction we performed proteomic and metabolomic analysis of serum and CSF at 4- and 6- months from a nonhuman primate model of infantile IDA. LC/MS/MS analyses identified a total of 227 metabolites and 205 proteins in serum. In CSF, we measured 210 metabolites and 1,560 proteins. Data were either processed from a Q-Exactive (Thermo Scientific, Waltham, MA) through Progenesis QI with accurate mass and retention time comparisons to a proprietary small molecule database and Metlin or with raw files imported directly from a Fusion Orbitrap (Thermo Scientific, Waltham, MA) through Sequest in Proteome Discoverer 2.4.0.305 (Thermo Scientific, Waltham, MA) with peptide matches through the latest Rhesus Macaque HMDB database. Metabolite and protein identifiers, p-values, and q-values were utilized for molecular pathway analysis with Ingenuity Pathways Analysis (IPA). We applied multiway distance weighted discrimination (DWD) to identify a weighted sum of the features (proteins or metabolites) that distinguish ID from IS at 4-months (pre-anemic period) and 6-months of age (anemic).

5.
Am J Physiol Regul Integr Comp Physiol ; 322(6): R486-R500, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35271351

ABSTRACT

The effects of iron deficiency (ID) during infancy extend beyond the hematologic compartment and include short- and long-term adverse effects on many tissues including the brain. However, sensitive biomarkers of iron-dependent brain health are lacking in humans. To determine whether serum and cerebrospinal fluid (CSF) biomarkers of ID-induced metabolic dysfunction are concordant in the pre/early anemic stage of ID before anemia in a nonhuman primate model of infantile iron deficiency anemia (IDA). ID (n = 7), rhesus infants at 4 mo (pre-anemic period) and 6 mo of age (anemic) were examined. Hematological, metabolomic, and proteomic profiles were generated via HPLC/MS at both time points to discriminate serum biomarkers of ID-induced brain metabolic dysfunction. We identified 227 metabolites and 205 proteins in serum. Abnormalities indicating altered liver function, lipid dysregulation, and increased acute phase reactants were present in ID. In CSF, we measured 210 metabolites and 1,560 proteins with changes in ID infants indicative of metabolomic and proteomic differences indexing disrupted synaptogenesis. Systemic and CSF proteomic and metabolomic changes were present and concurrent in the pre-anemic and anemic periods. Multiomic serum and CSF profiling uncovered pathways disrupted by ID in both the pre-anemic and anemic stages of infantile IDA, including evidence for hepatic dysfunction and activation of acute phase response. Parallel changes observed in serum and CSF potentially provide measurable serum biomarkers of ID that reflect at-risk brain processes prior to progression to clinical anemia.


Subject(s)
Anemia, Iron-Deficiency , Anemia , Iron Deficiencies , Anemia, Iron-Deficiency/cerebrospinal fluid , Animals , Biomarkers , Humans , Iron , Macaca mulatta , Proteomics
6.
J Perinatol ; 42(8): 1103-1109, 2022 08.
Article in English | MEDLINE | ID: mdl-35132153

ABSTRACT

OBJECTIVE: Examine interactions between perinatal risk factors for congenital iron deficiency (ID) using two cohorts. STUDY DESIGN: Iron status in a composite 767-member cord blood cohort and a NICU cohort of 257 infants < 33 weeks of gestation or small for gestational age (SGA). Risks for ID were examined. Cord ferritin levels < 84 µg/L defined congenital ID. Serum ferritin < 70 µg/L defined infantile ID at one-month. RESULTS: 31% of the cord cohort had congenital ID; risks summative (p < 0.0015). 16% of the NICU cohort had infantile ID; risks not summative. However, 32% had ID if the ferritin threshold was 100 µg/L. Being both preterm (p < 0.0001) and SGA (p < 0.05) negatively impacted cord iron status. Maternal hypertension was a novel predictor of iron status (p = 0.023 in preterm cord; p < 0.0025 in NICU). CONCLUSION: Summing risks in term and understanding compounding risks in preterm infants can improve screening and management of ID in at-risk infants.


Subject(s)
Anemia, Iron-Deficiency , Iron Deficiencies , Anemia, Iron-Deficiency/epidemiology , Female , Ferritins , Humans , Infant , Infant, Newborn , Infant, Premature , Iron , Pregnancy , Risk Factors
7.
J Pediatr ; 245: 217-221, 2022 06.
Article in English | MEDLINE | ID: mdl-35114287

ABSTRACT

The American Academy of Pediatrics recommends universal hemoglobin screening for iron deficiency anemia using hemoglobin <110 g/L at the 1-year-old well child visit. Our retrospective study suggests the need for combined hemoglobin and serum ferritin iron deficiency screening and raising the diagnostic serum ferritin threshold to 24-25 µg/L.


Subject(s)
Anemia, Iron-Deficiency , Iron Deficiencies , Anemia, Iron-Deficiency/diagnosis , Child , Ferritins , Hemoglobins/analysis , Humans , Infant , Retrospective Studies
9.
J Pediatr ; 238: 181-186.e3, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34214586

ABSTRACT

OBJECTIVE: To test the hypothesis that term-born small for gestational age (SGA) neonates have elevated thyroid-stimulating hormone (TSH) concentrations and an increased incidence of congenital hypothyroidism compared with non-SGA term neonates. STUDY DESIGN: This retrospective cohort study included all term neonates screened in Wisconsin in 2015 and 2016. The cohort was divided based on SGA status, defined as birth weight <10th percentile as calculated from the World Health Organization's sex-specific growth charts for age 0-2 years. TSH concentration on first newborn screening performed between birth and 96 hours of life and incidence of congenital hypothyroidism were compared between the SGA and non-SGA groups. RESULTS: A total of 115 466 term neonates, including 11 498 (9.96%) SGA neonates, were included in the study. TSH concentration and incidence of congenital hypothyroidism was significantly higher in the SGA group, but only TSH concentration remained significant when adjusted for potential confounding variables. CONCLUSIONS: Our data do not support a higher incidence of congenital hypothyroidism in term SGA neonates after adjusting for potential confounders. However, TSH concentrations were higher in term SGA neonates compared with term non-SGA neonates. The effects of mild thyroid hormone dysfunction on neurodevelopmental outcomes and development of chronic medical conditions merit long-term study.


Subject(s)
Congenital Hypothyroidism/epidemiology , Infant, Small for Gestational Age/blood , Congenital Hypothyroidism/blood , Female , Gestational Age , Humans , Infant, Newborn , Male , Neonatal Screening , Retrospective Studies , Thyrotropin/blood , Wisconsin
10.
J Nutr ; 151(9): 2509-2510, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34320201

Subject(s)
Iron
11.
Sci Rep ; 11(1): 12181, 2021 06 09.
Article in English | MEDLINE | ID: mdl-34108589

ABSTRACT

Maternal and environmental factors influence brain networks and architecture via both physiological pathways and epigenetic modifications. In particular, prenatal maternal depression and anxiety symptoms appear to impact infant white matter (WM) microstructure, leading us to investigate whether epigenetic modifications (i.e., DNA methylation) contribute to these WM differences. To determine if infants of women with depression and anxiety symptoms exhibit epigenetic modifications linked to neurodevelopmental changes, 52 umbilical cord bloods (CBs) were profiled. We observed 219 differentially methylated genomic positions (DMPs; FDR p < 0.05) in CB that were associated with magnetic resonance imaging measures of WM microstructure at 1 month of age and in regions previously described to be related to maternal depression and anxiety symptoms. Genomic characterization of these associated DMPs revealed 143 unique genes with significant relationships to processes involved in neurodevelopment, GTPase activity, or the canonical Wnt signaling pathway. Separate regression models for female (n = 24) and male (n = 28) infants found 142 associated DMPs in females and 116 associated DMPs in males (nominal p value < 0.001, R > 0.5), which were annotated to 98 and 81 genes, respectively. Together, these findings suggest that umbilical CB DNA methylation levels at birth are associated with 1-month WM microstructure.


Subject(s)
Anxiety Disorders/physiopathology , Brain/pathology , DNA Methylation , Depressive Disorder/physiopathology , Fetal Blood/chemistry , Prenatal Exposure Delayed Effects/pathology , White Matter/pathology , Adolescent , Adult , Brain/metabolism , Epigenesis, Genetic , Female , Humans , Infant, Newborn , Male , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/metabolism , White Matter/metabolism , Young Adult
12.
Front Hum Neurosci ; 15: 624107, 2021.
Article in English | MEDLINE | ID: mdl-33716694

ABSTRACT

A high percent of oxidative energy metabolism is needed to support brain growth during infancy. Unhealthy diets and limited nutrition, as well as other environmental insults, can compromise these essential developmental processes. In particular, iron deficiency anemia (IDA) has been found to undermine both normal brain growth and neurobehavioral development. Even moderate ID may affect neural maturation because when iron is limited, it is prioritized first to red blood cells over the brain. A primate model was used to investigate the neural effects of a transient ID and if deficits would persist after iron treatment. The large size and postnatal growth of the monkey brain makes the findings relevant to the metabolic and iron needs of human infants, and initiating treatment upon diagnosis of anemia reflects clinical practice. Specifically, this analysis determined whether brain maturation would still be compromised at 1 year of age if an anemic infant was treated promptly once diagnosed. The hematology and iron status of 41 infant rhesus monkeys was screened at 2-month intervals. Fifteen became ID; 12 met clinical criteria for anemia and were administered iron dextran and B vitamins for 1-2 months. MRI scans were acquired at 1 year. The volumetric and diffusion tensor imaging (DTI) measures from the ID infants were compared with monkeys who remained continuously iron sufficient (IS). A prior history of ID was associated with smaller total brain volumes, driven primarily by significantly less total gray matter (GM) and smaller GM volumes in several cortical regions. At the macrostructual level, the effect on white matter volumes (WM) was not as overt. However, DTI analyses of WM microstructure indicated two later-maturating anterior tracts were negatively affected. The findings reaffirm the importance of iron for normal brain development. Given that brain differences were still evident even after iron treatment and following recovery of iron-dependent hematological indices, the results highlight the importance of early detection and preemptive supplementation to limit the neural consequences of ID.

13.
Am J Clin Nutr ; 113(4): 915-923, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33740040

ABSTRACT

BACKGROUND: The effects of infantile iron deficiency anemia (IDA) extend beyond hematological indices and include short- and long-term adverse effects on multiple cells and tissues. IDA is associated with an abnormal serum metabolomic profile, characterized by altered hepatic metabolism, lowered NAD flux, increased nucleoside levels, and a reduction in circulating dopamine levels. OBJECTIVES: The objective of this study was to determine whether the serum metabolomic profile is normalized after rapid correction of IDA using iron dextran injections. METHODS: Blood was collected from iron-sufficient (IS; n = 10) and IDA (n = 12) rhesus infants at 6 months of age. IDA infants were then administered iron dextran and vitamin B via intramuscular injections at weekly intervals for 2 to 8 weeks. Their hematological and metabolomic statuses were evaluated following treatment and compared with baseline and a separate group of age-matched IS infants (n = 5). RESULTS: Serum metabolomic profiles assessed at baseline and after treatment via HPLC/MS using isobaric standards identified 654 quantifiable metabolites. At baseline, 53 metabolites differed between IS and IDA infants. Iron treatment restored traditional hematological indices, including hemoglobin and mean corpuscular volume, into the normal range, but the metabolite profile in the IDA group after iron treatment was markedly altered, with 323 metabolites differentially expressed when compared with an infant's own baseline profile. CONCLUSIONS: Rapid correction of IDA with iron dextran resulted in extensive metabolic changes across biochemical pathways indexing the liver function, bile acid release, essential fatty acid production, nucleoside release, and several neurologically important metabolites. The results highlight the importance of a cautious approach when developing a route and regimen of iron repletion to treat infantile IDA.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Disease Models, Animal , Iron-Dextran Complex/therapeutic use , Macaca mulatta , Metabolome/drug effects , Animals , Bile Acids and Salts/metabolism , Fatty Acids, Essential/metabolism , Injections, Intramuscular , Liver/metabolism , Nucleosides/metabolism
15.
Clin Pediatr (Phila) ; 60(3): 178-183, 2021 03.
Article in English | MEDLINE | ID: mdl-33148015

ABSTRACT

Children obese at the age of 5 years are at greater risk of lifelong obesity. Because certain risks of obesity can be identified in early infancy, a tool for obesity risk prediction in early life would be clinically useful. We investigated predictors of obesity risk in a novel, prospectively collected healthy birth cohort recruited for demographic risks to develop iron deficiency at 1 year, a cohort leveraged because risk factors for iron deficiency and obesity overlap. Obesity at the age of 5 years was defined as age- and sex-specific body mass index Z-score (zBMI) >2SD. For each child, obesity risk factors were summed. Of 10 total risk factors, the following 4 key risks were identified: maternal obesity, maternal diabetes, large for gestational age, or breastfeeding <6 months. Childhood obesity was predicted by either ≥3 total number of risks (P < .033), any key risk (P < .002), or summing key risks (P < .0001). In clinical practice, summing early life risk factors may be a useful strategy for preemptive counseling.


Subject(s)
Body Mass Index , Breast Feeding/statistics & numerical data , Diabetes Mellitus/epidemiology , Gestational Weight Gain , Mothers/statistics & numerical data , Pediatric Obesity/diagnosis , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Male , Obesity/epidemiology , Pediatric Obesity/epidemiology , Prospective Studies , Risk Factors
16.
Neoreviews ; 21(2): e80-e88, 2020 02.
Article in English | MEDLINE | ID: mdl-32005718

ABSTRACT

Prematurity, maternal diabetes, maternal smoking, being medically underserved, and small size for gestational age are common characteristics of neonates in the NICU and can predispose them to develop congenital iron deficiency. Iron is critical for organ development. In the fetus and newborn, iron is prioritized for red blood cell production, sometimes at the expense of other tissues, including the brain. It is critical to optimize iron levels in newborns to support erythropoiesis, growth, and brain development. Available studies support improved neurodevelopmental outcomes with either iron supplementation or delayed umbilical cord clamping at birth. Erythropoietic doses of erythropoietin/erythrocyte-stimulating agents may also improve neurocognitive outcomes. However, the literature on the effect of liberal red blood cell transfusions on long-term neurodevelopment is mixed. Understanding age-specific normal values and monitoring of iron indices can help individualize and optimize the iron status of patients in the NICU.


Subject(s)
Anemia, Neonatal , Child Development/physiology , Deficiency Diseases , Erythrocyte Transfusion , Erythrocytes/physiology , Erythropoiesis/physiology , Erythropoietin/therapeutic use , Hematinics/therapeutic use , Intensive Care Units, Neonatal , Iron/physiology , Anemia, Neonatal/ethnology , Anemia, Neonatal/therapy , Child Development/drug effects , Deficiency Diseases/congenital , Deficiency Diseases/drug therapy , Deficiency Diseases/ethnology , Erythrocytes/drug effects , Erythropoiesis/drug effects , Humans , Infant, Newborn , Iron Deficiencies
17.
J Nutr ; 150(4): 685-693, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31722400

ABSTRACT

BACKGROUND: Iron deficiency is the most common nutrient deficiency in human infants aged 6 to 24 mo, and negatively affects many cellular metabolic processes, including energy production, electron transport, and oxidative degradation of toxins. There can be persistent influences on long-term metabolic health beyond its acute effects. OBJECTIVES: The objective was to determine how iron deficiency in infancy alters the serum metabolomic profile and to test whether these effects persist after the resolution of iron deficiency in a nonhuman primate model of spontaneous iron deficiency. METHODS: Blood was collected from naturally iron-sufficient (IS; n = 10) and iron-deficient (ID; n = 10) male and female infant rhesus monkeys (Macaca mulatta) at 6 mo of age. Iron deficiency resolved without intervention upon feeding of solid foods, and iron status was re-evaluated at 12 mo of age from the IS and formerly ID monkeys using hematological and other indices; sera were metabolically profiled using HPLC/MS and GC/MS with isobaric standards for identification and quantification at both time points. RESULTS: A total of 413 metabolites were measured, with differences in 40 metabolites identified between IS and ID monkeys at 6 mo (P$\le $ 0.05). At 12 mo, iron-related hematological parameters had returned to normal, but the formerly ID infants remained metabolically distinct from the age-matched IS infants, with 48 metabolites differentially expressed between the groups. Metabolomic profiling indicated altered liver metabolites, differential fatty acid production, increased serum uridine release, and atypical bile acid production in the ID monkeys. CONCLUSIONS: Pathway analyses of serum metabolites provided evidence of a hypometabolic state, altered liver function, differential essential fatty acid production, irregular uracil metabolism, and atypical bile acid production in ID infants. Many metabolites remained altered after the resolution of ID, suggesting long-term effects on metabolic health.


Subject(s)
Metabolome/physiology , Monkey Diseases/blood , Animals , Bile Acids and Salts/biosynthesis , Diet/veterinary , Fatty Acids/biosynthesis , Female , Iron Deficiencies , Liver/physiopathology , Macaca mulatta , Male , Metabolomics/methods , Prospective Studies , Uracil/metabolism
18.
Alcohol ; 84: 57-66, 2020 05.
Article in English | MEDLINE | ID: mdl-31734307

ABSTRACT

Prenatal alcohol exposure (PAE) causes developmental abnormalities known as fetal alcohol spectrum disorder (FASD). Maternal iron status modulates the severity of these defects in the offspring. Because the placenta is central in supporting fetal development, we investigated whether maternal iron status similarly modulates alcohol's effects in the placenta. We hypothesized that PAE causes placental insufficiency by decreasing placental weight and efficiency, and we hypothesized that these are worsened by maternal iron deficiency (ID) and alleviated by dietary iron fortification (IF). We also determined whether altered placental iron flux and inflammatory balance contribute to placental insufficiency. Pregnant Long-Evans rats consumed an iron-deficient (ID; 2-6 ppm), iron-sufficient (IS; 100 ppm), or iron-fortified (IF; 500 ppm) diet. Alcohol (5 g/kg body weight) or isocaloric maltodextrin (MD) was gavaged daily from gestational day (GD) 13.5-19.5. Placental outcomes were evaluated on GD20.5. PAE reduced fetal weight (p < 0.0001), placental weight (p = 0.0324), and placental efficiency (p = 0.0043). PAE downregulated placental transferrin receptor (p = 0.0032); it also altered placental Il1b and Tnf expression and the Il6:Il10 ratio (p = 0.0337, 0.0300, and 0.0034, respectively) to generate a response favoring inflammation. ID-PAE further reduced fetal growth and placental efficiency and induced a heightened pro-inflammatory placental profile. IF did not rescue the alcohol-reduced fetal weight, but it normalized placental efficiency and decreased placental inflammation. These placental cytokines correlated with fetal and placental growth, and explained 45% of the variability in fetal weight and 20% of the variability in placental efficiency. In summary, alcohol induces placental insufficiency and is associated with a pro-inflammatory cytokine profile exacerbated by maternal ID and mitigated by maternal IF. Because the placenta is closely linked to intrauterine growth, the placental insufficiency reported here may correlate with the lower birth weights in a subgroup of individuals who experienced PAE.


Subject(s)
Cytokines/metabolism , Ethanol/administration & dosage , Fetal Alcohol Spectrum Disorders , Iron Deficiencies , Iron, Dietary/administration & dosage , Maternal Nutritional Physiological Phenomena , Placentation/drug effects , Animals , Disease Models, Animal , Female , Inflammation , Pregnancy , Rats , Rats, Long-Evans
19.
Nutrients ; 11(10)2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31623079

ABSTRACT

Maternal iron deficiency anemia, obesity, and diabetes are prevalent during pregnancy. All are associated with neonatal brain iron deficiency (ID) and neurodevelopmental impairment. Exosomes are extracellular vesicles involved in cell-cell communication. Contactin-2 (CNTN2), a neural-specific glycoprotein, and brain-derived neurotrophic factor (BDNF) are important in neurodevelopment and found in exosomes. We hypothesized that exosomal CNTN2 and BDNF identify infants at risk for brain ID. Umbilical cord blood samples were measured for iron status. Maternal anemia, diabetes, and body mass index (BMI) were recorded. Cord blood exosomes were isolated and validated for the exosomal marker CD81 and the neural-specific exosomal marker CNTN2. Exosomal CNTN2 and BDNF levels were quantified by ELISA. Analysis of CNTN2 and BDNF levels as predictors of cord blood iron indices showed a direct correlation between CNTN2 and ferritin in all neonates (n = 79, ß = 1.75, p = 0.02). In contrast, BDNF levels inversely correlated with ferritin (ß = -1.20, p = 0.03), with stronger association in female neonates (n = 37, ß = -1.35, p = 0.06), although there is no evidence of a sex-specific effect. Analysis of maternal risk factors for neonatal brain ID as predictors of exosomal CNTN2 and BDNF levels showed sex-specific relationships between infants of diabetic mothers (IDMs) and CNTN2 levels (Interaction p = 0.0005). While male IDMs exhibited a negative correlation (n = 42, ß = -0.69, p = 0.02), female IDMs showed a positive correlation (n = 37, ß = 0.92, p = 0.01) with CNTN2. A negative correlation between BNDF and maternal BMI was found with stronger association in female neonates (per 10 units BMI, ß = -0.60, p = 0.04). These findings suggest CNTN2 and BNDF are respective molecular markers for male and female neonates at risk for brain ID. This study supports the potential of exosomal markers to assess neonatal brain status in at-risk infants.


Subject(s)
Anemia, Iron-Deficiency/diagnosis , Brain-Derived Neurotrophic Factor/blood , Brain/metabolism , Contactin 2/blood , Exosomes/metabolism , Fetal Blood/metabolism , Adolescent , Adult , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/physiopathology , Biomarkers/blood , Brain/growth & development , Female , Ferritins/blood , Humans , Infant, Newborn , Male , Pregnancy , Risk Factors , Sex Factors , Young Adult
20.
Alcohol Clin Exp Res ; 43(11): 2332-2343, 2019 11.
Article in English | MEDLINE | ID: mdl-31524964

ABSTRACT

BACKGROUND: Prenatal alcohol exposure (PAE) causes long-term growth and neurodevelopmental deficits that are worsened by maternal iron deficiency (ID). In our preclinical rat model, PAE causes fetal anemia, brain ID, and elevated hepatic iron via increased maternal and fetal hepcidin synthesis. These changes are normalized by a prenatal iron-fortified (IF) diet. Here, we hypothesize that iron status and PAE dysregulate the major upstream pathways that govern hepcidin production-EPO/BMP6/SMAD and IL-6/JAK2/STAT3. METHODS: Pregnant, Long Evans rat dams consumed ID (2 to 6 ppm iron), iron-sufficient (IS, 100 ppm iron), or IF (500 ppm iron) diets and received alcohol (5 g/kg) or isocaloric maltodextrin daily from gestational days (GD) 13.5 to 19.5. Protein and gene expression were quantified in the 6 experimental groups at GD 20.5. RESULTS: PAE did not affect Epo or Bmp6 expression, but reduced p-SMAD1/5/8/SMAD1/5/8 protein ratios in both IS and ID maternal and fetal liver (all p's < 0.01). In contrast, PAE stimulated maternal hepatic expression of Il-6 (p = 0.03) and elevated p-STAT3/STAT3 protein ratios in both IS and ID maternal and fetal liver (all p's < 0.02). PAE modestly elevated maternal Il-1ß, Tnf-α, and Ifn-γ. Fetal cytokine responses to PAE were muted compared with dams, and PAE did not affect hepatic Il-6 (p = 0.78) in IS and ID fetuses. Dietary iron fortification sharply attenuated Il-6 expression in response to PAE, with IF driving a 150-fold decrease (p < 0.001) in maternal liver and a 10-fold decrease (p < 0.01) in fetal liver. The IF diet also normalized p-STAT3/STAT3 ratios in both maternal and fetal liver. CONCLUSIONS: These findings suggest that alcohol-driven stimulation of the IL-6/JAK2/STAT3 pathway mediates the elevated hepcidin observed in the PAE dam and fetus. Normalization of these signals by IF suggests that dysregulated hepcidin is driven by alcohol's disruption of the IL-6/JAK2/STAT3 pathway. Prenatal dietary IF represents a potential therapeutic approach for PAE that warrants further investigation.


Subject(s)
Anemia, Iron-Deficiency/complications , Ethanol/adverse effects , Fetus/drug effects , Interleukin-6/blood , Prenatal Exposure Delayed Effects/blood , STAT3 Transcription Factor/blood , Animals , Disease Models, Animal , Female , Fetus/metabolism , Interleukin-6/metabolism , Iron, Dietary , Pregnancy , Rats , Rats, Long-Evans , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...