Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Nutr ; 154(3): 875-885, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38072152

RESUMO

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.


Assuntos
Anemia Ferropriva , Encefalopatias , Deficiências de Ferro , Animais , Lactente , Humanos , Criança , Anemia Ferropriva/complicações , Anemia Ferropriva/diagnóstico , Macaca mulatta/metabolismo , Prognóstico , Ferro/metabolismo , Hemoglobinas/metabolismo , Encefalopatias/metabolismo , Biomarcadores , Encéfalo/metabolismo
2.
J Nutr ; 153(1): 148-157, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36913448

RESUMO

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.


Assuntos
Anemia Ferropriva , Anemia , Deficiências de Ferro , Masculino , Feminino , Animais , Reticulócitos/química , Reticulócitos/metabolismo , Anemia/metabolismo , Hemoglobinas/metabolismo , Ferro/metabolismo , Primatas/metabolismo
3.
Data Brief ; 45: 108591, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36164307

RESUMO

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).

4.
Am J Physiol Regul Integr Comp Physiol ; 322(6): R486-R500, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35271351

RESUMO

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.


Assuntos
Anemia Ferropriva , Anemia , Deficiências de Ferro , Anemia Ferropriva/líquido cefalorraquidiano , Animais , Biomarcadores , Humanos , Ferro , Macaca mulatta , Proteômica
5.
Front Hum Neurosci ; 15: 624107, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33716694

RESUMO

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.

6.
Am J Clin Nutr ; 113(4): 915-923, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33740040

RESUMO

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.


Assuntos
Anemia Ferropriva/tratamento farmacológico , Modelos Animais de Doenças , Complexo Ferro-Dextran/uso terapêutico , Macaca mulatta , Metaboloma/efeitos dos fármacos , Animais , Ácidos e Sais Biliares/metabolismo , Ácidos Graxos Essenciais/metabolismo , Injeções Intramusculares , Fígado/metabolismo , Nucleosídeos/metabolismo
7.
Mol Nutr Food Res ; 65(8): e2001018, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33599094

RESUMO

SCOPE: Iron deficiency (ID) compromises the health of infants worldwide. Although readily treated with iron, concerns remain about the persistence of some effects. Metabolic and gut microbial consequences of infantile ID were investigated in juvenile monkeys after natural recovery (pID) from iron deficiency or post-treatment with iron dextran and B vitamins (pID+Fe). METHODS AND RESULTS: Metabolomic profiling of urine and plasma is conducted with 1 H nuclear magnetic resonance (NMR) spectroscopy. Gut microbiota are characterized from rectal swabs by amplicon sequencing of the 16S rRNA gene. Urinary metabolic profiles of pID monkeys significantly differed from pID+Fe and continuously iron-sufficient controls (IS) with higher maltose and lower amounts of microbial-derived metabolites. Persistent differences in energy metabolism are apparent from the plasma metabolic phenotypes with greater reliance on anaerobic glycolysis in pID monkeys. Microbial profiling indicated higher abundances of Methanobrevibacter, Lachnobacterium, and Ruminococcus in pID monkeys and any history of ID resulted in a lower Prevotella abundance compared to the IS controls. CONCLUSIONS: Lingering metabolic and microbial effects are found after natural recovery from ID. These long-term biochemical derangements are not present in the pID+Fe animals emphasizing the importance of the early detection and treatment of early-life ID to ameliorate its chronic metabolic effects.


Assuntos
Anemia Ferropriva/metabolismo , Anemia Ferropriva/microbiologia , Microbioma Gastrointestinal/fisiologia , Complexo Ferro-Dextran/farmacologia , Anemia Ferropriva/tratamento farmacológico , Animais , Animais Recém-Nascidos , Análise Química do Sangue , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Macaca mulatta , Metaboloma , RNA Ribossômico 16S , Urina/química
8.
J Nutr ; 150(4): 685-693, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31722400

RESUMO

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.


Assuntos
Metaboloma/fisiologia , Doenças dos Macacos/sangue , Animais , Ácidos e Sais Biliares/biossíntese , Dieta/veterinária , Ácidos Graxos/biossíntese , Feminino , Deficiências de Ferro , Fígado/fisiopatologia , Macaca mulatta , Masculino , Metabolômica/métodos , Estudos Prospectivos , Uracila/metabolismo
9.
Nutr Neurosci ; 21(1): 40-48, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27499134

RESUMO

OBJECTIVES: Iron deficiency (ID) anemia leads to long-term neurodevelopmental deficits by altering iron-dependent brain metabolism. The objective of the study was to determine if ID induces metabolomic abnormalities in the cerebrospinal fluid (CSF) in the pre-anemic stage and to ascertain the aspects of abnormal brain metabolism affected. METHODS: Standard hematological parameters [hemoglobin (Hgb), mean corpuscular volume (MCV), transferrin (Tf) saturation, and zinc protoporphyrin/heme (ZnPP/H)] were compared at 2, 4, 6, 8, and 12 months in iron-sufficient (IS; n = 7) and iron-deficient (ID; n = 7) infant rhesus monkeys. Five CSF metabolite ratios were determined at 4, 8, and 12 months using 1H NMR spectroscopy at 16.4 T and compared between groups and in relation to hematologic parameters. RESULTS: ID infants developed ID (Tf saturation < 25%) by 4 months of age and all became anemic (Hgb < 110 g/L and MCV < 60 fL) at 6 months. Their heme indices normalized by 12 months. Pyruvate/glutamine and phosphocreatine/creatine (PCr/Cr) ratios in CSF were lower in the ID infants by 4 months (P < 0.05). The PCr/Cr ratio remained lower at 8 months (P = 0.02). ZnPP/H, an established blood marker of pre-anemic ID, was positively correlated with the CSF citrate/glutamine ratio (marginal correlation, 0.34; P < 0.001; family wise error rate = 0.001). DISCUSSION: Metabolomic analysis of the CSF is sensitive for detecting the effects of pre-anemic ID on brain energy metabolism. Persistence of a lower PCr/Cr ratio at 8 months, even as hematological measures demonstrated recovery from anemia, indicate that the restoration of brain energy metabolism is delayed. Metabolomic platforms offer a useful tool for early detection of the impact of ID on brain metabolism in infants.


Assuntos
Anemia Ferropriva/líquido cefalorraquidiano , Encéfalo/metabolismo , Ferro/líquido cefalorraquidiano , Metabolômica , Animais , Animais Recém-Nascidos , Dieta , Carboidratos da Dieta/administração & dosagem , Gorduras na Dieta/administração & dosagem , Fibras na Dieta/administração & dosagem , Proteínas Alimentares/administração & dosagem , Feminino , Hemoglobinas/líquido cefalorraquidiano , Macaca mulatta , Espectroscopia de Ressonância Magnética , Micronutrientes/administração & dosagem , Micronutrientes/líquido cefalorraquidiano , Protoporfirinas/líquido cefalorraquidiano , Manejo de Espécimes , Transferrina/líquido cefalorraquidiano
10.
J Cereb Blood Flow Metab ; 35(1): 48-57, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25315861

RESUMO

Neurologic disorders such as Alzheimer's, Parkinson's disease, and Restless Legs Syndrome involve a loss of brain iron homeostasis. Moreover, iron deficiency is the most prevalent nutritional concern worldwide with many associated cognitive and neural ramifications. Therefore, understanding the mechanisms by which iron enters the brain and how those processes are regulated addresses significant global health issues. The existing paradigm assumes that the endothelial cells (ECs) forming the blood-brain barrier (BBB) serve as a simple conduit for transport of transferrin-bound iron. This concept is a significant oversimplification, at minimum failing to account for the iron needs of the ECs. Using an in vivo model of brain iron deficiency, the Belgrade rat, we show the distribution of transferrin receptors in brain microvasculature is altered in luminal, intracellular, and abluminal membranes dependent on brain iron status. We used a cell culture model of the BBB to show the presence of factors that influence iron release in non-human primate cerebrospinal fluid and conditioned media from astrocytes; specifically apo-transferrin and hepcidin were found to increase and decrease iron release, respectively. These data have been integrated into an interactive model where BBB ECs are central in the regulation of cerebral iron metabolism.


Assuntos
Anemia Ferropriva/metabolismo , Encéfalo/metabolismo , Ferro/metabolismo , Modelos Biológicos , Anemia Ferropriva/líquido cefalorraquidiano , Anemia Ferropriva/genética , Animais , Apoproteínas/metabolismo , Astrócitos/metabolismo , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Bovinos , Células Cultivadas , Células Endoteliais/metabolismo , Feminino , Hepcidinas/metabolismo , Heterozigoto , Homozigoto , Ferro/líquido cefalorraquidiano , Macaca mulatta , Masculino , Microvasos/metabolismo , Ratos Sprague-Dawley , Receptores da Transferrina/metabolismo , Transferrina/metabolismo
11.
Res Vet Sci ; 94(3): 549-54, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23312499

RESUMO

The realization that pregnant and infant monkeys were challenged by high nutritional needs for iron led vendors to markedly increase iron concentrations in commercial diets. Yet, no systematic research was conducted to investigate the consequences of this important dietary change. Hematology and iron panels were determined for 142 infant rhesus monkeys gestated and reared on 3 different diets varying in iron concentration (180, 225 or 380 mg Fe/kg). Anemia was significantly more prevalent in offspring from females fed the 180 and 225 mg Fe/kg diets (32-41% versus 0 for the 380 mg Fe/kg diet, P<0.001). Higher hepcidin levels were protective against iron overload in infants from the 380 mg Fe/kg condition. These findings indicate a highly fortified diet during pregnancy continues to have postnatal benefits for the growing infant. However, for those interested in iron deficiency, lower iron diets provide a reliable way to generate anemic infant monkeys for research.


Assuntos
Anemia Ferropriva/veterinária , Dieta/veterinária , Compostos de Ferro/uso terapêutico , Doenças dos Macacos/prevenção & controle , Anemia Ferropriva/sangue , Anemia Ferropriva/prevenção & controle , Ração Animal , Fenômenos Fisiológicos da Nutrição Animal/efeitos dos fármacos , Animais , Animais Recém-Nascidos/sangue , Relação Dose-Resposta a Droga , Feminino , Alimentos Fortificados , Hepcidinas/sangue , Compostos de Ferro/administração & dosagem , Lactação/efeitos dos fármacos , Macaca mulatta , Masculino , Doenças dos Macacos/sangue , Gravidez
12.
Neurochem Res ; 38(3): 573-80, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23269483

RESUMO

Iron deficiency anemia affects many pregnant women and young infants worldwide. The health impact is significant, given iron's known role in many body functions, including oxidative and lipid metabolism, protein synthesis and brain neurochemistry. The following research determined if (1)H NMR spectroscopy-based metabolomic analysis of cerebrospinal fluid (CSF) could detect the adverse influence of early life iron deficiency on the central nervous system. Using a controlled dietary model in 43 infant primates, distinct differences were found in spectra acquired at 600 MHz from the CSF of anemic monkeys. Three metabolite ratios, citrate/pyruvate, citrate/lactate and pyruvate/glutamine ratios, differed significantly in the iron deficient infant and then normalized following the consumption of dietary iron and improvement of clinical indices of anemia in the heme compartment. This distinctive metabolomic profile associated with anemia in the young infant indicates that CSF can be employed to track the neurological effects of iron deficiency and benefits of iron supplementation.


Assuntos
Anemia Ferropriva/líquido cefalorraquidiano , Encéfalo/metabolismo , Metabolismo Energético , Deficiências de Ferro , Metabolômica , Anemia Ferropriva/sangue , Animais , Ácido Cítrico/líquido cefalorraquidiano , Feminino , Glutamina/líquido cefalorraquidiano , Ácido Láctico/líquido cefalorraquidiano , Macaca mulatta/crescimento & desenvolvimento , Imageamento por Ressonância Magnética , Ácido Pirúvico/líquido cefalorraquidiano
13.
Dev Neurosci ; 34(4): 354-65, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23018452

RESUMO

Iron deficiency affects nearly 2 billion people worldwide, with pregnant women and young children being most severely impacted. Sustained anemia during the first year of life can cause cognitive, attention and motor deficits, which may persist despite iron supplementation. We conducted iTRAQ analyses on cerebrospinal fluid (CSF) from infant monkeys (Macaca mulatta) to identify differential protein expression associated with early iron deficiency. CSF was collected from 5 iron-sufficient and 8 iron-deficient anemic monkeys at weaning age (6-7 months) and again at 12-14 months. Despite consumption of iron-fortified food after weaning, which restored hematological indices into the normal range, expression of 5 proteins in the CSF remained altered. Most of the proteins identified are involved in neurite outgrowth, migration or synapse formation. The results reveal novel ways in which iron deficiency undermines brain growth and results in aberrant neuronal migration and connections. Taken together with gene expression data from rodent models of iron deficiency, we conclude that significant alterations in neuroconnectivity occur in the iron-deficient brain, which may persist even after resolution of the hematological anemia. The compromised brain infrastructure could account for observations of behavioral deficits in children during and after the period of anemia.


Assuntos
Anemia Ferropriva/líquido cefalorraquidiano , Proteínas do Líquido Cefalorraquidiano/análise , Proteômica/métodos , Fatores Etários , Anemia Ferropriva/complicações , Anemia Ferropriva/dietoterapia , Anemia Ferropriva/embriologia , Animais , Dano Encefálico Crônico/líquido cefalorraquidiano , Dano Encefálico Crônico/etiologia , Resinas de Troca de Cátion , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia por Troca Iônica/métodos , Feminino , Compostos Ferrosos/administração & dosagem , Compostos Ferrosos/uso terapêutico , Alimentos Fortificados , Macaca mulatta , Masculino , Desnutrição/fisiopatologia , Modelos Animais , Proteínas do Tecido Nervoso/líquido cefalorraquidiano , Proteínas do Tecido Nervoso/deficiência , Fragmentos de Peptídeos/análise , Gravidez , Complicações na Gravidez/fisiopatologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Desmame
14.
Behav Brain Res ; 219(1): 108-15, 2011 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-21192986

RESUMO

Infections and inflammatory conditions during pregnancy can dysregulate neural development and increase the risk for developing autism and schizophrenia. The following research utilized a nonhuman primate model to investigate the potential impact of a mild endotoxemia during pregnancy on brain maturation and behavioral reactivity as well as the infants' hormone and immune physiology. Nine pregnant female rhesus monkeys (Macaca mulatta) were administered nanogram concentrations of lipopolysaccharide (LPS) on two consecutive days, 6 weeks before term, and their offspring were compared to nine control animals. When tested under arousing challenge conditions, infants from the LPS pregnancies were more behaviorally disturbed, including a failure to show a normal attenuation of startle responses on tests of prepulse inhibition. Examination of their brains at 1 year of age with magnetic resonance imaging (MRI) revealed the unexpected finding of a significant 8.8% increase in global white matter volume distributed across many cortical regions compared to controls. More selective changes in regional gray matter volume and cortical thickness were noted in parietal, medial temporal, and frontal areas. While inhibited neural growth has been described previously after prenatal infection and LPS administration at higher doses in rodents, this low dose endotoxemia in the monkey is the first paradigm to produce a neural phenotype associated with augmented gray and white matter growth.


Assuntos
Comportamento Animal/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Citocinas/metabolismo , Endotoxemia/psicologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Envelhecimento/psicologia , Animais , Ansiedade/psicologia , Atenção/efeitos dos fármacos , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/crescimento & desenvolvimento , Emoções/efeitos dos fármacos , Feminino , Humanos , Hidrocortisona/metabolismo , Processamento de Imagem Assistida por Computador , Sistema Imunitário/efeitos dos fármacos , Injeções Intraventriculares , Interleucina-6/metabolismo , Relações Interpessoais , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/farmacologia , Macaca mulatta , Imageamento por Ressonância Magnética , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/psicologia , Reflexo de Sobressalto/fisiologia
15.
J Med Primatol ; 38(4): 252-6, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19490364

RESUMO

BACKGROUND: Cases of abdominal pregnancy, in the form of intra-abdominal mummified fetuses, have been described in nonhuman primates. Gestational diabetes and pre-eclampsia are common pregnancy complications in women. METHODS: Two timed-bred rhesus monkeys had high-risk pregnancies, an abdominal pregnancy with delivery of a live term infant, and a case of gestational diabetes that later developed pre-eclampsia. RESULTS: The monkey that had abdominal pregnancy later died from septic peritonitis. The monkey had a colonic adenocarcinoma that may have allowed leakage of intestinal contents into the abdomen. Her infant was fostered to another female and survived. The monkey with gestational diabetes and pre-eclampsia was treated with a regimen similar to that used in women, and a live infant was delivered at day 157 of gestation by Caesarian section. CONCLUSION: These cases underscore the value of timed-breeding and the similarities between pregnancy complications in women and in nonhuman primates.


Assuntos
Diabetes Gestacional/veterinária , Macaca mulatta , Pré-Eclâmpsia/veterinária , Gravidez Abdominal/veterinária , Animais , Cesárea/veterinária , Feminino , Gravidez , Gravidez de Alto Risco
16.
Dev Psychobiol ; 51(3): 301-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19194962

RESUMO

Both during and after a period of iron deficiency (ID), iron-dependent neural processes are affected, which raises the potential concern that the anemia commonly experienced by many growing infants could have a protracted effect on the developing brain. To further investigate the effects of ID on the immature brain, 49 infant rhesus monkeys were evaluated across the first year of life. The mothers, and subsequently the infants after weaning, were maintained on a standardized diet containing 180 mg/kg of iron and were not provided other iron-rich foods as treats or supplements. As the infants grew, they were all screened with hematological tests, which documented that 16 (33.3%) became markedly ID between 4 and 8 months of age. During this anemic period and subsequently at 1 year of age, cerebrospinal fluid (CSF) specimens were collected to compare monoamine activity in the ID and iron-sufficient infants. Monoamine neurotransmitters and metabolite levels were normal at 4 and 8 months of age, but by 1 year the formerly anemic monkeys had significantly lower dopamine and significantly higher norepinephrine levels. These findings indicate that ID can affect the developmental trajectory of these two important neurotransmitter systems, which are associated with emotionality and behavioral performance, and further that the impact in the young monkey was most evident during the period of recovery.


Assuntos
Anemia Ferropriva/fisiopatologia , Encéfalo/fisiopatologia , Dopamina/líquido cefalorraquidiano , Norepinefrina/líquido cefalorraquidiano , Ácido 3,4-Di-Hidroxifenilacético/líquido cefalorraquidiano , Fatores Etários , Animais , Emoções/fisiologia , Epinefrina/líquido cefalorraquidiano , Índices de Eritrócitos , Feminino , Hemoglobinometria , Ácido Homovanílico/líquido cefalorraquidiano , Ácido Hidroxi-Indolacético/líquido cefalorraquidiano , Macaca mulatta , Masculino , Gravidez , Valores de Referência , Serotonina/líquido cefalorraquidiano , Fatores Sexuais
17.
J Dev Behav Pediatr ; 29(1): 11-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18300719

RESUMO

OBJECTIVE: While poor nutrition during development is an obvious concern, the magnitude and duration of the neural and cognitive deficits that occur after moderate iron deficiency in infancy have remained controversial. A nonhuman primate model of infancy anemia was refined to investigate the effects on cognitive performance. METHODS: Young rhesus monkeys that experienced a delimited period of iron deficiency were tested on a series of cognitive tasks following normalization of their hematological status. Beginning at 8 to 9 months of age, 2 months after weaning from their mothers and consumption of solid food, the previously iron-deficient (ID) monkeys (n = 17) were compared to age- and gender-matched, iron-sufficient (IS) (n = 27) monkeys on a series of three tests of cognitive performance. Using the Wisconsin General Testing Apparatus, a Black/White Discrimination task was followed by acquisition of Black/White Reversal (BWR). RESULTS: ID monkeys were significantly slower at mastering the BWR task (p < .04), which required reversing and inhibiting the previously learned response. In addition, ID infants were significantly less object oriented (p < .017) and more distractible (p < .018). However, on two subsequent tests, the Concurrent Object Discrimination and Delayed Non-Match-to-Sample, there were no differences in acquisition, performance, or behavioral reactivity. CONCLUSIONS: The initial cognitive and behavioral deficits are similar to those seen in follow-up evaluations of anemic children, but the limited extent of the impairment after this moderate iron deficiency that involved a select nutrient deficiency is encouraging for the benefits attainable through early identification and iron supplementation.


Assuntos
Anemia Ferropriva/complicações , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/etiologia , Convalescença , Fatores Etários , Anemia Ferropriva/tratamento farmacológico , Animais , Encéfalo/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Feminino , Compostos Ferrosos/uso terapêutico , Macaca mulatta , Masculino , Testes Psicológicos , Recompensa , Índice de Gravidade de Doença
18.
J Neurochem ; 105(1): 127-36, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17996030

RESUMO

Iron deficiency (ID) anemia during infancy results in long-term neurological consequences, yet the mediating mechanisms remain unclear. Infant monkeys often become naturally anemic during the first 6 months of life, presenting an opportunity to determine the effect of developmental iron deficiency. After weaning, animals were chosen randomly for supplementation with oral iron or, fed a standard commercial chow diet. The control group was never iron deficient. ID anemia was corrected by 12 months in both groups, as indicated by hematological parameters. CSF was collected for proteomic analysis at 12 months of age to assess the impact of developmental ID on the brain. The CSF proteome for both formerly iron deficient groups was similar and revealed 12 proteins with expression levels altered at least twofold. These proteins were identified by matrix assisted laser desorption ionization time-of-flight spectrometry and included prostaglandin D synthase, olfactory receptors and glial fibrillary acidic protein. Thus the proteomic analysis reveals a persistent effect of ID and provides insights into reports of disturbed sleep, hypomyelination and other behavioral alterations associated with ID. Furthermore, alterations in the CSF proteome despite normal hematologic parameters indicate that there is a hierarchical system that prioritizes repletion of red cell mass at the expense of the brain.


Assuntos
Anemia Ferropriva/líquido cefalorraquidiano , Líquido Cefalorraquidiano/metabolismo , Proteômica/métodos , Fatores Etários , Anemia Ferropriva/metabolismo , Animais , Animais Recém-Nascidos , Peso Corporal/fisiologia , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Proteína Glial Fibrilar Ácida/líquido cefalorraquidiano , Oxirredutases Intramoleculares/líquido cefalorraquidiano , Lipocalinas/líquido cefalorraquidiano , Macaca mulatta , Análise Serial de Proteínas , Distribuição Aleatória , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
19.
Pediatr Res ; 61(5 Pt 1): 520-4, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17413860

RESUMO

The influence of maternal stress during pregnancy on the postpartum iron status and immune maturation of infants was investigated in a nonhuman primate model. Forty infant rhesus monkeys were generated from two types of disturbed pregnancies, early or late gestation stress, and compared with 24 undisturbed controls. Prenatal stress increased the prevalence and magnitude of iron deficiency (ID) as the infants' growth-related demands for iron exceeded dietary intake from breast milk. At 4-6 mo of age, the emergence of ID significantly accentuated an effect of prenatal stress on natural killer cell activity, an important component of innate immunity. These findings indicate that maternal stress, especially early in pregnancy, should be added to the list of risk factors that warrant closer scrutiny of hematological profiles in fast-growing babies.


Assuntos
Imunidade Inata/fisiologia , Deficiências de Ferro , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Estresse Fisiológico , Animais , Dieta , Feminino , Idade Gestacional , Humanos , Ferro/sangue , Células Matadoras Naturais/imunologia , Gravidez/sangue , Gravidez/imunologia , Fatores de Risco , Estresse Fisiológico/sangue , Estresse Fisiológico/imunologia
20.
Brain Behav Immun ; 21(6): 807-15, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17336039

RESUMO

Bacterial infections and proinflammatory cytokines induce behavioral and physiological responses associated with withdrawal and sickness behavior. These effects in rodents are often exacerbated by stressful environmental contexts. To model this synergistic effect of arousal and stress, low doses of lipopolysaccharide (LPS), 4 or 40ng/kg, were administered to rhesus monkeys in different environmental contexts. Activity, emotional and social behaviors, cortisol, interleukin-6 (IL-6), and peripheral leukocyte trafficking were assessed in 4 experiments: an initial 3h time-course in a novel cage (Experiment 1); an extended 24h time-course (Experiment 2); a highly arousing context in which an animal was engaged in the Human Intruder Paradigm (HIP, Experiment 3); and finally in an undisturbed context in their Home Cage (Experiment 4). The moderately arousing novel cage potentiated leukocyte, neutrophilic, IL-6, and cortisol changes as compared to the Home Cage. Unlike the social withdrawal seen in rodents, monkeys engaged in a marked increase in social behavior. IL-6 levels were positively correlated with Proximal Contact, which was induced to a greater degree by the higher dose of LPS. In contrast, the high arousal HIP condition appeared to obviate the effects of LPS. Thus, the rhesus monkey provides an excellent animal model for investigating the behavioral and physiological actions of endotoxemia, which are profoundly influenced by the situational context in which the individual is evaluated.


Assuntos
Nível de Alerta/fisiologia , Interleucina-6/sangue , Leucócitos/imunologia , Comportamento Social , Estresse Psicológico/imunologia , Análise de Variância , Animais , Relação Dose-Resposta a Droga , Meio Ambiente , Comportamento Exploratório/fisiologia , Feminino , Seguimentos , Hidrocortisona/sangue , Interleucina-6/imunologia , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Macaca mulatta , Masculino , Neutrófilos/imunologia , Estresse Psicológico/sangue , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA