Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Dev Orig Health Dis ; 11(3): 264-272, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31543090

RESUMO

Iron deficiency is common in pregnant and lactating women and is associated with reduced cognitive development of the offspring. Since iron affects lipid metabolism, the availability of fatty acids, particularly the polyunsaturated fatty acids required for early neural development, was investigated in the offspring of female rats fed iron-deficient diets during gestation and lactation. Subsequent to the dams giving birth, one group of iron-deficient dams was recuperated by feeding an iron-replete diet. Dams and neonates were killed on postnatal days 1, 3 and 10, and the fatty acid composition of brain and stomach contents was assessed by gas chromatography. Changes in the fatty acid profile on day 3 became more pronounced on day 10 with a decrease in the proportion of saturated fatty acids and a compensatory increase in monounsaturated fatty acids. Long-chain polyunsaturated fatty acids in the n-6 family were reduced, but there was no change in the n-3 family. The fatty acid profiles of neonatal brain and stomach contents were similar, suggesting that the change in milk composition may be related to the changes in the neonatal brain. When the dams were fed an iron-sufficient diet at birth, the effects of iron deficiency on the fatty acid composition of lipids in both dam's milk and neonates' brains were reduced. This study showed an interaction between maternal iron status and fatty acid composition of the offspring's brain and suggests that these effects can be reduced by iron repletion of the dam's diet at birth.


Assuntos
Anemia Ferropriva/complicações , Encéfalo/crescimento & desenvolvimento , Metabolismo dos Lipídeos/fisiologia , Complicações Hematológicas na Gravidez/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Anemia Ferropriva/fisiopatologia , Animais , Animais Recém-Nascidos/metabolismo , Animais Lactentes/metabolismo , Encéfalo/patologia , Química Encefálica/fisiologia , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/análise , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-6/análise , Ácidos Graxos Ômega-6/metabolismo , Feminino , Humanos , Ferro/sangue , Deficiências de Ferro , Lactação/fisiologia , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos
2.
Br J Nutr ; 121(2): 121-129, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30482256

RESUMO

Fe deficiency is relatively common in pregnancy and has both short- and long-term consequences. However, little is known about the effect on the metabolism of other micronutrients. A total of fifty-four female rats were fed control (50 mg Fe/kg) or Fe-deficient diets (7·5 mg/kg) before and during pregnancy. Maternal liver, placenta and fetal liver were collected at day 21 of pregnancy for Cu and Zn analysis and to measure expression of the major genes of Cu and Zn metabolism. Cu levels increased in the maternal liver (P=0·002) and placenta (P=0·018) of Fe-deficient rats. Zn increased (P<0·0001) and Cu decreased (P=0·006) in the fetal liver. Hepatic expression of the Cu chaperones antioxidant 1 Cu chaperone (P=0·042) and cytochrome c oxidase Cu chaperone (COX17, P=0·020) decreased in the Fe-deficient dams, while the expression of the genes of Zn metabolism was unaltered. In the placenta, Fe deficiency reduced the expression of the chaperone for superoxide dismutase 1, Cu chaperone for superoxide dismutase (P=0·030), ceruloplasmin (P=0·042) and Zn transport genes, ZRT/IRT-like protein 4 (ZIP4, P=0·047) and Zn transporter 1 (ZnT1, P=0·012). In fetal liver, Fe deficiency increased COX17 (P=0·020), ZRT/IRT-like protein 14 (P=0·036) and ZnT1 (P=0·0003) and decreased ZIP4 (P=0·004). The results demonstrate that Fe deficiency during pregnancy has opposite effects on Cu and Zn levels in the fetal liver. This may, in turn, alter metabolism of these nutrients, with consequences for development in the fetus and the neonate.


Assuntos
Cobre/metabolismo , Deficiências de Ferro , Zinco/metabolismo , Animais , Proteínas de Transporte de Cátions , Ceruloplasmina , Cobre/análise , Complexo IV da Cadeia de Transporte de Elétrons/genética , Feminino , Feto/metabolismo , Expressão Gênica/fisiologia , Fígado/química , Fígado/embriologia , Fígado/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Chaperonas Moleculares , Placenta/química , Placenta/metabolismo , Gravidez , Ratos , Zinco/análise
3.
J Nutr Biochem ; 32: 55-63, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27142737

RESUMO

Iron deficiency impairs vitamin A (VA) metabolism in the rat but the mechanisms involved are unknown and the effect during development has not been investigated. We investigated the effect of pregnancy and maternal iron deficiency on VA metabolism in the mother and fetus. 54 rats were fed either a control or iron deficient diet for 2weeks prior to mating and throughout pregnancy. Another 15 female rats followed the same diet and were used as non-pregnant controls. Maternal liver, placenta and fetal liver were collected at d21 for total VA, retinol and retinyl ester (RE) measurement and VA metabolic gene expression analysis. Iron deficiency increased maternal hepatic RE (P<.05) and total VA (P<.0001), fetal liver RE (P<.05), and decreased placenta total VA (P<.05). Pregnancy increased Cellular Retinol Binding Protein (CRBP)-II gene expression by 7 fold (P=.001), decreased VA levels (P=.0004) and VA metabolic gene expression (P<.0001) in the liver. Iron deficiency increased hepatic CRBPII expression by a further 2 fold (P=.044) and RBP4 by~20% (P=.005), increased RBPR2 and decreased CRBPII, LRAT, and TTR in fetal liver, while it had no effect on VA metabolic gene expression in the placenta. Hepatic CRBPII expression is increased by pregnancy and further increased by iron deficiency, which may play an important role in VA metabolism and homeostasis. Maternal iron deficiency also alters VA metabolism in the fetus, which is likely to have consequences for development.


Assuntos
Anemia Ferropriva/fisiopatologia , Dieta/efeitos adversos , Desenvolvimento Fetal , Regulação da Expressão Gênica no Desenvolvimento , Fígado/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Proteínas Celulares de Ligação ao Retinol/metabolismo , Anemia Ferropriva/embriologia , Anemia Ferropriva/etiologia , Anemia Ferropriva/metabolismo , Animais , Diterpenos , Esterificação , Feminino , Ferro/sangue , Ferro/metabolismo , Deficiências de Ferro , Fígado/embriologia , Fígado/patologia , Tamanho do Órgão , Placenta/metabolismo , Placenta/patologia , Gravidez , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos Endogâmicos , Proteínas Celulares de Ligação ao Retinol/genética , Ésteres de Retinil , Vitamina A/administração & dosagem , Vitamina A/análogos & derivados , Vitamina A/sangue , Vitamina A/metabolismo , Desmame
4.
J Membr Biol ; 248(6): 1199-206, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26439821

RESUMO

Iron deficiency during pregnancy has many effects on both the mother and her developing foetus. These can be both short and long term. One effect is an alteration in fatty acid metabolism and we hypothesised that these changes may result in alterations in membrane function and structure. In order to test this hypothesis, we measured osmotic sensitivity in red blood cells isolated from neonates and their mothers at different times following birth. We fed female rats control or iron-deficient diets for 4 weeks prior to mating and kept them on the same diet until term. At that time, we returned one group of deficient dams to the control diet. The others were kept on the same diet. We showed that iron deficiency results in a decrease in osmotic sensitivity in the mothers but not in their neonates. Returning the dams to the control diet resulted in a return of their red cell osmotic sensitivity to control levels. In the neonates, there was no recovery in haematocrit or in any other parameter, though they did not get any worse, in contrast to the pups being suckled by deficient mothers. The data show two things. The first is that following birth, the mother restores her own iron stores at the expense of the pups, and secondly, there are differences in properties and sensitivities between red cells from mothers and their neonates. This latter observation cannot be explained by differences in the membrane fatty acid profiles, which were not significantly different.


Assuntos
Eritrócitos/metabolismo , Deficiências de Ferro , Fragilidade Osmótica , Animais , Animais Recém-Nascidos , Peso Corporal , Índices de Eritrócitos , Membrana Eritrocítica/metabolismo , Ácidos Graxos/metabolismo , Feminino , Ferro/metabolismo , Lipídeos de Membrana/metabolismo , Gravidez , Ratos
5.
Br J Nutr ; 112(12): 2018-27, 2014 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-25342229

RESUMO

Maternal nutritional status during pregnancy has been reported to be associated with childhood asthma and atopic disease. The Avon Longitudinal Study of Parents and Children has reported associations between reduced umbilical cord Fe status and childhood wheeze and eczema; however, follow-up was short and lung function was not measured. In the present study, the associations between maternal Fe status during pregnancy and childhood outcomes in the first 10 years of life were investigated in a subgroup of 157 mother-child pairs from a birth cohort with complete maternal, fetal ultrasound, blood and child follow-up data. Maternal Fe intake was assessed using FFQ at 32 weeks of gestation and Hb concentrations and serum Fe status (ferritin, soluble transferrin receptor and TfR-F (transferrin receptor:ferritin) index) were measured at 11 weeks of gestation and at delivery. Maternal Fe intake, Hb concentrations and serum Fe status were found to be not associated with fetal or birth measurements. Unit increases in first-trimester maternal serum TfR concentrations (OR 1.44, 95% CI 1.05, 1.99) and TfR-F index (OR 1.42, 95% CI 1.10, 1.82) (i.e. decreasing Fe status) were found to be associated with an increased risk of wheeze, while unit increases in serum ferritin concentrations (i.e., increasing Fe status) were found to be associated with increases in standardised mean peak expiratory flow (PEF) (ß 0.25, 95% CI 0.09, 0.42) and forced expiratory volume in the first second (FEV1) (ß 0.20, 95% CI 0.08, 0.32) up to 10 years of age. Increasing maternal serum TfR-F index at delivery was found to be associated with an increased risk of atopic sensitisation (OR 1.35, 95% CI 1.02, 1.79). The results of the present study suggest that reduced maternal Fe status during pregnancy is adversely associated with childhood wheeze, lung function and atopic sensitisation, justifying further studies on maternal Fe status and childhood asthma and atopic disease.


Assuntos
Anemia Ferropriva/complicações , Asma/etiologia , Ferritinas/sangue , Hipersensibilidade Imediata/etiologia , Ferro/sangue , Efeitos Tardios da Exposição Pré-Natal , Receptores da Transferrina/sangue , Adulto , Anemia Ferropriva/sangue , Asma/fisiopatologia , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Volume Expiratório Forçado , Hemoglobinas/metabolismo , Humanos , Lactente , Recém-Nascido , Ferro/administração & dosagem , Deficiências de Ferro , Ferro da Dieta/administração & dosagem , Ferro da Dieta/sangue , Pulmão/fisiopatologia , Masculino , Estado Nutricional , Razão de Chances , Pico do Fluxo Expiratório , Gravidez , Complicações na Gravidez/sangue , Primeiro Trimestre da Gravidez , Sons Respiratórios/etiologia , Sons Respiratórios/fisiopatologia , Inquéritos e Questionários
6.
Proc Nutr Soc ; 73(1): 9-15, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24176079

RESUMO

This review examines the importance of the placenta in iron metabolism during development and the effect of iron deficiency on maternal and fetal physiology. Iron is an essential micronutrient, required for a wide variety of biological processes. During pregnancy, the mother has to deplete her iron stores in order to provide the baby with adequate amounts. Trans-placental iron transfer involves binding transferrin (Tf)-bound iron to the Tf receptor, uptake into an endosome, acidification, release of iron through divalent metal transporter 1, efflux across the basolateral membrane through ferroportin and oxidation of Fe(II) by zyklopen. An additional haem transport system has been hypothesised, which may explain why certain gene knockouts are not lethal for the developing fetus. Iron deficiency is a common phenomenon during pregnancy, and the placenta adapts by up-regulating its transfer systems, maintaining iron at the expense of the mother. Despite these adaptations, deficiency cannot be completely prevented, and the offspring suffers both short- and long-term consequences. Some of these, at least, may arise from decreased expression of genes involved in the cell cycle and altered expression of transcription factors, such as c-myc, which in turn can produce, for example, kidneys with reduced numbers of nephrons. The mechanism whereby these changes are induced is not certain, but may simply be as a result of the reduced availability of iron resulting in decreased enzyme activity. Since these changes are so significant, and because some of the changes are irreversible, we believe that iron prophylaxis should be considered in all pregnancies.


Assuntos
Anemia Ferropriva/complicações , Compostos Ferrosos/sangue , Transtornos da Nutrição Fetal , Feto , Deficiências de Ferro , Placenta/metabolismo , Complicações na Gravidez/sangue , Anemia Ferropriva/sangue , Proteínas de Transporte de Cátions/sangue , Feminino , Transtornos da Nutrição Fetal/etiologia , Transtornos da Nutrição Fetal/prevenção & controle , Heme/metabolismo , Humanos , Ferro/sangue , Gravidez , Receptores da Transferrina/sangue , Transferrina/metabolismo
7.
Reprod Biol Endocrinol ; 11: 32, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23635304

RESUMO

BACKGROUND: Iron deficiency anaemia during pregnancy is a global problem, with short and long term consequences for maternal and child health. Animal models have demonstrated that the developing fetus is vulnerable to maternal iron restriction, impacting on postnatal metabolic and blood pressure regulation. Whilst long-term outcomes are similar across different models, the commonality in mechanistic events across models is unknown. This study examined the impact of iron deficiency on maternal and fetal iron homeostasis in two strains of rat. METHODS: Wistar (n=20) and Rowett Hooded Lister (RHL, n=19) rats were fed a control or low iron diet for 4 weeks prior to and during pregnancy. Tissues were collected at day 21 of gestation for analysis of iron content and mRNA/protein expression of regulatory proteins and transporters. RESULTS: A reduction in maternal liver iron content in response to the low iron diet was associated with upregulation of transferrin receptor expression and a reduction in hepcidin expression in the liver of both strains, which would be expected to promote increased iron absorption across the gut and increased turnover of iron in the liver. Placental expression of transferrin and DMT1+IRE were also upregulated, indicating adaptive responses to ensure availability of iron to the fetus. There were considerable differences in hepatic maternal and fetal iron content between strains. The higher quantity of iron present in livers from Wistar rats was not explained by differences in expression of intestinal iron transporters, and may instead reflect greater materno-fetal transfer in RHL rats as indicated by increased expression of placental iron transporters in this strain. CONCLUSIONS: Our findings demonstrate substantial differences in iron homeostasis between two strains of rat during pregnancy, with variable impact of iron deficiency on the fetus. Whilst common developmental processes and pathways have been observed across different models of nutrient restriction during pregnancy, this study demonstrates differences in maternal adaptation which may impact on the trajectory of the programmed response.


Assuntos
Dieta , Feto/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Ferro/farmacologia , Fenômenos Fisiológicos da Nutrição Materna/efeitos dos fármacos , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Criança , Feminino , Peso Fetal/genética , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Idade Gestacional , Humanos , Ferro/administração & dosagem , Ferro/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Ratos , Ratos Wistar , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie
8.
PLoS One ; 7(10): e48133, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23110188

RESUMO

Poor iron status is a global health issue, affecting two thirds of the world population to some degree. It is a particular problem among pregnant women, in both developed and developing countries. Feeding pregnant rats a diet deficient in iron is associated with both hypertension and reduced nephron endowment in adult male offspring. However, the mechanistic pathway leading from iron deficiency to fetal kidney development remains elusive. This study aimed to establish the underlying processes associated with iron deficiency by assessing gene and protein expression changes in the rat embryo, focussing on the responses occurring at the time of the nutritional insult. Analysis of microarray data showed that iron deficiency in utero resulted in the significant up-regulation of 979 genes and down-regulation of 1545 genes in male rat embryos (d13). Affected processes associated with these genes included the initiation of mitosis, BAD-mediated apoptosis, the assembly of RNA polymerase II preinitiation complexes and WNT signalling. Proteomic analyses highlighted 7 proteins demonstrating significant up-regulation with iron deficiency and the down-regulation of 11 proteins. The main functions of these key proteins included cell proliferation, protein transport and folding, cytoskeletal remodelling and the proteasome complex. In line with our recent work, which identified the perturbation of the proteasome complex as a generalised response to in utero malnutrition, we propose that iron deficiency alone leads to a more specific failure in correct protein folding and transport. Such an imbalance in this delicate quality-control system can lead to cellular dysfunction and apoptosis. Therefore these findings offer an insight into the underlying mechanisms associated with the development of the embryo during conditions of poor iron status, and its health in adult life.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Embrião de Mamíferos/metabolismo , Perfilação da Expressão Gênica/métodos , Deficiências de Ferro , Proteômica/métodos , Animais , Peso Corporal/genética , Peso Corporal/fisiologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Nutr Rev ; 69 Suppl 1: S17-22, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22043877

RESUMO

During pregnancy, the developing fetus is entirely dependent on its mother for nutrition and waste disposal, which take place almost exclusively across the placenta. The placenta is, however, not just a passive mediator; it can also regulate the rates and amounts of nutrient transported. This is particularly true for iron transport. This review examines how the fetus, and not the mother, regulates iron uptake and transfer across the placenta, how these processes interact, and how the fetus can modulate iron metabolism in the mother.


Assuntos
Feto/metabolismo , Ferro da Dieta/farmacocinética , Troca Materno-Fetal , Placenta/metabolismo , Circulação Placentária , Gravidez , Transporte Biológico , Feminino , Desenvolvimento Fetal , Humanos , Ferro da Dieta/administração & dosagem
10.
Semin Cell Dev Biol ; 22(6): 637-44, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21893209

RESUMO

Copper and iron are both essential micronutrients. Because they can both accept and donate electrons, they are central to many energy dependent chemical reactions. For example, copper is a critical part of ferroxidase enzymes ceruloplasmin, hephaestin and zyklopen, as well as enzymes such as dopamine-ß-monoxygenase, while iron is part of the catalytic site of many cytochromes and enzymes involved in fatty acid desaturation. Unsurprisingly, therefore, copper and iron deficiency, especially during pregnancy, when cell proliferation and differentiation are very active, sub-optimal nutrient status can lead to serious consequences. These problems can persist into adulthood, with an increased risk of mental problems such as schizophrenia and, in animal models at least, hypertension and obesity. In this review, we consider what these problems are and how they may arise. We examine the role of copper and iron deficiencies separately during fetal development, in terms of birth outcome and then how problems with status in utero can have long term sequelae for the offspring. We examine several possible mechanisms of action, both direct and indirect. Direct causes include, for example, reduced enzyme activity, while indirect ones may result from changes in cytokine activity, reductions in cell number or increased apoptosis, to name but a few. We examine a very important area of nutrition-interactions between the micronutrients and conclude that, while we have made significant advances in understanding the relationship between micronutrient status and pregnancy outcome, there is still much to be learned.


Assuntos
Cobre/deficiência , Biologia do Desenvolvimento , Desenvolvimento Fetal/fisiologia , Deficiências de Ferro , Neurônios/metabolismo , Estado Nutricional/fisiologia , Adulto , Animais , Ceruloplasmina/metabolismo , Citocromos/metabolismo , Embrião de Mamíferos , Feminino , Feto , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Recém-Nascido , Ferro da Dieta , Camundongos , Neurônios/citologia , Obesidade/metabolismo , Obesidade/fisiopatologia , Oxirredução , Gravidez , Resultado da Gravidez , Transtornos Psicóticos/metabolismo , Transtornos Psicóticos/fisiopatologia
11.
PLoS One ; 6(8): e23189, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21858025

RESUMO

Many mechanisms purport to explain how nutritional signals during early development are manifested as disease in the adult offspring. While these describe processes leading from nutritional insult to development of the actual pathology, the initial underlying cause of the programming effect remains elusive. To establish the primary drivers of programming, this study aimed to capture embryonic gene and protein changes in the whole embryo at the time of nutritional insult rather than downstream phenotypic effects. By using a cross-over design of two well established models of maternal protein and iron restriction we aimed to identify putative common "gatekeepers" which may drive nutritional programming.Both protein and iron deficiency in utero reduced the nephron complement in adult male Wistar and Rowett Hooded Lister rats (P<0.05). This occurred in the absence of damage to the glomerular ultrastructure. Microarray, proteomic and pathway analyses identified diet-specific and strain-specific gatekeeper genes, proteins and processes which shared a common association with the regulation of the cell cycle, especially the G1/S and G2/M checkpoints, and cytoskeletal remodelling. A cell cycle-specific PCR array confirmed the down-regulation of cyclins with protein restriction and the up-regulation of apoptotic genes with iron deficiency.The timing and experimental design of this study have been carefully controlled to isolate the common molecular mechanisms which may initiate the sequelae of events involved in nutritional programming of embryonic development. We propose that despite differences in the individual genes and proteins affected in each strain and with each diet, the general response to nutrient deficiency in utero is perturbation of the cell cycle, at the level of interaction with the cytoskeleton and the mitotic checkpoints, thereby diminishing control over the integrity of DNA which is allowed to replicate. These findings offer novel insight into the primary causes and mechanisms leading to the pathologies which have been identified by previous programming studies.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal/fisiologia , Ciclo Celular/fisiologia , Citoesqueleto/metabolismo , Desenvolvimento Embrionário/fisiologia , Fenômenos Fisiológicos da Nutrição Animal/genética , Animais , Animais Recém-Nascidos , Ciclo Celular/genética , Estudos Cross-Over , Dieta , Proteínas Alimentares/administração & dosagem , Eletroforese em Gel Bidimensional , Desenvolvimento Embrionário/genética , Feminino , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Ferro/administração & dosagem , Deficiências de Ferro , Rim/embriologia , Rim/crescimento & desenvolvimento , Rim/metabolismo , Masculino , Espectrometria de Massas , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Proteômica/métodos , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Am J Clin Nutr ; 94(6 Suppl): 1903S-1907S, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21543532

RESUMO

During pregnancy, iron is transported from mother to fetus across the placenta. Iron is essential for many biological processes, including the transfer of oxygen in blood, but it can also be toxic. Elaborate and elegant mechanisms have evolved to make sure that the potential for oxidative damage is minimized. In this article, we examine how iron is transferred from the maternal liver to the placenta, taken up, and transferred to the fetal liver. We consider the molecular mechanisms and how they are regulated. In addition, we use data from previously published articles to examine how the processes are regulated and what adaptations can occur to ameliorate the consequences of iron deficiency--an all too common problem in pregnancy, even in the developed world. Finally, we examine some of the many questions that remain about the transfer process and consider how nutrients interact and what the consequences of these interactions may be for the mother and her developing infant.


Assuntos
Feto/metabolismo , Ferro da Dieta/farmacocinética , Troca Materno-Fetal/fisiologia , Circulação Placentária , Animais , Feminino , Humanos , Transporte de Íons , Fígado/metabolismo , Placenta/metabolismo , Gravidez/fisiologia
13.
Reprod Fertil Dev ; 22(8): 1230-6, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20883648

RESUMO

Maternal and fetal liver iron concentrations and associated haematology parameters were determined in an adolescent sheep paradigm characterised by rapid maternal growth, premature delivery and feto-placental growth restriction. Singleton-bearing dams were offered a control or high dietary intake to induce normal or growth-restricted pregnancies, respectively. Pregnancies were terminated on Day 90 or 130 of gestation or progressed to term. Relative blood volume increased (P < 0.05) and liver iron concentration decreased (P < 0.003) from mid to late gestation in control, but not in high-intake dams. At 90 and 130 days gestation, liver iron concentrations were reduced (P < 0.001) in high-intake dams but fetal liver iron was independent of dam nutrition. High intakes leading to poor pregnancy outcome at term were characterised by increased maternal haematocrit, haemoglobin, total plasma protein, albumin (all P < 0.001) and serum iron (P < 0.05), and by reduced oestradiol 17ß (P < 0.001) at Day 130. Thus, high dietary intakes that promote rapid maternal growth and adiposity are associated with early depletion of maternal liver iron stores and a relative failure of normal blood volume expansion, which may, in turn, underlie the reduction in uteroplacental blood flows and fetal nutrient delivery previously established for this paradigm.


Assuntos
Retardo do Crescimento Fetal/veterinária , Ferro/metabolismo , Fígado/metabolismo , Resultado da Gravidez/veterinária , Técnicas de Reprodução Assistida/veterinária , Transferência de Embrião Único/veterinária , Adiposidade , Fenômenos Fisiológicos da Nutrição Animal , Animais , Biomarcadores/sangue , Proteínas Sanguíneas/metabolismo , Volume Sanguíneo , Ingestão de Alimentos , Estradiol/sangue , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/fisiopatologia , Idade Gestacional , Hemoglobinas/metabolismo , Ferro/sangue , Fígado/embriologia , Idade Materna , Fenômenos Fisiológicos da Nutrição Materna , Circulação Placentária , Gravidez , Nascimento Prematuro , Ovinos
14.
J Nutr ; 140(10): 1728-35, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20685892

RESUMO

We previously detected a membrane-bound, copper-containing oxidase that may be involved in iron efflux in BeWo cells, a human placental cell line. We have now identified a gene encoding a predicted multicopper ferroxidase (MCF) with a putative C-terminal membrane-spanning sequence and high sequence identity to hephaestin (Heph) and ceruloplasmin (Cp), the other known vertebrate MCF. Molecular modeling revealed conservation of all type I, II, and III copper-binding sites as well as a putative iron-binding site. Protein expression was observed in multiple diverse mouse tissues, including placenta and mammary gland, and the expression pattern was distinct from that of Cp and Heph. The protein possessed ferroxidase activity, and protein levels decreased in cellular copper deficiency. Knockdown with small interfering RNA in BeWo cells indicates that this gene represents the previously detected oxidase. We propose calling this new member of the MCF family "zyklopen."


Assuntos
Ceruloplasmina/química , Ceruloplasmina/genética , Cobre/análise , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Ceruloplasmina/análise , Cobre/metabolismo , Feminino , Expressão Gênica , Humanos , Ferro/metabolismo , Glândulas Mamárias Animais/enzimologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Modelos Moleculares , Especificidade de Órgãos , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Fragmentos de Peptídeos/química , Placenta/enzimologia , Gravidez , RNA Interferente Pequeno/farmacologia , Ratos , Homologia de Sequência
15.
J Toxicol Environ Health A ; 73(2): 101-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20077281

RESUMO

This review considers the development of the regulatory process for metals in the European Union (Regulation for Registration, Evaluation, Authorisation and Restriction of Chemicals, REACH). The manner in which the REACH process was developed, its history, and the relative involvement of government and industry are examined. The problems involved in setting up the system and the steps in optimisation are considered. One of the major difficulties in the development of the REACH mechanism was incorporating the fact that many metals are essential elements, which is not a factor that most toxicological examinations consider. How the REACH process evolved to deal with these problems and how the mechanisms were put in place to overcome them is examined. Looking specifically at examples of industry working with academia, one needs to consider how a risk assessment can be developed and submitted in a timely and successful fashion.


Assuntos
Poluentes Ambientais/toxicidade , União Europeia , Metais/toxicidade , Participação da Comunidade , Exposição Ambiental/prevenção & controle , Monitoramento Ambiental/métodos , Poluentes Ambientais/química , Governo , Humanos , Indústrias , Cooperação Internacional , Metais/química , Política Pública , Medição de Risco , Fatores de Risco
16.
J Inorg Biochem ; 103(5): 709-16, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19232737

RESUMO

This paper investigates the extent to which Cu loading influences Fe levels in HepG2 cells and the effect on proteins regulated by Fe status. Cu supplementation increased Cu content 3-fold, concomitant with a decrease in cellular Fe levels. Intracellular levels of both transferrin (Tf) and ceruloplasmin (Cp) protein rose in parallel with increased secretion into the culture media. There was no increase in mRNA levels for either protein. Rather, our data suggested increased translation of the mRNA. The increase was not reflected in total protein synthesis, which actually decreased. The effect was not a generalised stress or cell damage response, since heat shock protein 70 levels and lactate dehydrogenase secretion were not significantly altered. To test whether the Cu effect could be acting though the decrease in Fe levels, we measured transferrin receptor (TfR) levels using (125)I labeled Tf and mRNA analysis. Neither protein nor mRNA levels were changed. Neither was the level of ferroportin mRNA. As a positive control, Fe chelation increased Tf and Cp secretion significantly, and TfR mRNA levels rose 2-fold. We excluded the possibility that the increased Cp or Tf could provide the required substrate to stimulate Fe efflux, and instead demonstrate that Cu can substitute for Fe in the iron regulatory protein - iron responsive element regulation mechanism.


Assuntos
Cobre/farmacologia , Expressão Gênica/efeitos dos fármacos , Proteínas Reguladoras de Ferro/metabolismo , Ferro/metabolismo , Northern Blotting , Western Blotting , Linhagem Celular Tumoral , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Cobre/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Expressão Gênica/genética , Humanos , Proteínas Reguladoras de Ferro/genética , Ligação Proteica/efeitos dos fármacos , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Transferrina/genética , Transferrina/metabolismo
17.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R1063-70, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19176888

RESUMO

Iron metabolism during pregnancy is biased toward maintaining the fetal supply, even at the cost of anemia in the mother. The mechanisms regulating this are not well understood. Here, we examine iron deficiency and supplementation on the hierarchy of iron supply and the gene expression of proteins that regulate iron metabolism in the rat. Dams were fed iron-deficient diets for 4 wk, mated, and either continued on the deficient diet or an iron-supplemented diet during either the first half or the second half of their pregnancy. A control group was maintained on normal iron throughout. They were killed at 0.5, 12.5, or 21.5 days of gestation, and tissues and blood samples were collected. Deficiency and supplementation had differential effects on maternal and fetal hematocrit and liver iron levels. From early in pregnancy, a hierarchy of iron supply is established benefiting the fetus to the detriment of the mother. Transferrin receptor, transferrin receptor 2, and hepcidin mRNA expression were regulated by both iron deficiency and supplementation. Expression patterns showed both organ and supplementation protocol dependence. Further analysis indicated that iron levels in the fetal, and not maternal, liver regulate the expression of liver transferrin receptor and hepcidin expression in the mother.


Assuntos
Anemia Ferropriva/metabolismo , Suplementos Nutricionais , Feto/metabolismo , Ferro/metabolismo , Fígado/metabolismo , Troca Materno-Fetal , Anemia Ferropriva/sangue , Anemia Ferropriva/prevenção & controle , Fenômenos Fisiológicos da Nutrição Animal , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Modelos Animais de Doenças , Feminino , Sangue Fetal/metabolismo , Feto/efeitos dos fármacos , Regulação da Expressão Gênica , Idade Gestacional , Hematócrito , Hepcidinas , Ferro/sangue , Ferro/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/embriologia , Fenômenos Fisiológicos da Nutrição Materna , Troca Materno-Fetal/efeitos dos fármacos , Placenta/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo
18.
Biochem Soc Trans ; 36(Pt 6): 1258-61, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19021536

RESUMO

During development, the fetus is entirely dependent on the mother for its nutrient requirements. Subsequently, it is a period when both are vulnerable to changes in dietary supply, especially of those nutrients that are marginal under normal circumstances. In developed countries, this applies mainly to micronutrients. Even now, iron deficiency is a common disorder, especially in pregnancy. Similarly, copper intake in the U.K. population is rarely above adequate levels. It is now becoming clear that nutrient deficiencies during pregnancy can result in problems for the offspring, in both the short- and long-term. Early studies showed that lambs born to mothers on copper-deficient pastures developed 'swayback', with neurological and muscular symptoms that could not be reversed by postnatal supplementation. Our own findings have shown that prenatal iron deficiency results in increased postnatal blood pressure, even though the offspring have normal dietary iron levels from birth. These observations emphasize the importance of iron and copper in growth and development. Complicating the situation further is the fact that copper and iron are known to interact with each other in many ways, including absorption and intracellular transport. However, their interactions during the pregnancy appear to be more complex than during the non-pregnant state. In the present review, we examine the importance of these metals and their interactions, the consequences, both short- and long-term, of deficiency and consider some possible mechanisms whereby these effects may be generated.


Assuntos
Cobre/metabolismo , Desenvolvimento Embrionário , Ferro/metabolismo , Animais , Ceruloplasmina/metabolismo , Feminino , Humanos , Absorção Intestinal , Gravidez
19.
Br J Nutr ; 97(2): 239-46, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17298691

RESUMO

Cu and Fe metabolism are known to be linked, but the interactions during pregnancy are less well studied. In the present study we used rats to examine the effect of Cu deficiency during pregnancy on Fe and Cu levels in maternal and fetal tissue and on the gene expression profile of proteins involved in Cu and Fe metabolism in the placenta. Rats were fed diets with different Cu contents before and during pregnancy. Samples were collected on day 21 of gestation. Cu levels, ceruloplasmin activity and serum Fe all decreased in maternal serum of Cu-deficient animals. Maternal liver Fe inversely correlated with liver Cu. Placental Cu levels decreased with no change in Fe. Fe and Cu levels both decreased in the fetal liver. The drop in maternal liver Cu was significantly correlated with a decrease in organ weight of fetal liver, lung and kidney. No changes were observed in mRNA expression of Cu transporter 1, Menkes P-type Cu-ATPase 7A, Wilson P-type Cu-ATPase 7B, cytochrome-c oxidase, and Cu chaperone Atox1 in the placenta of Cu-deficient dams. Transferrin receptor 1 and the Fe-responsive element (IRE)-regulated divalent metal transporter 1 (DMT1) were up regulated; while ferroportin and non-IRE1-regulated DMT1 levels did not change. These data show that Cu deficiency during pregnancy not only has a direct effect on Fe levels but also regulates the expression of Fe transporters. The pattern closely mirrors that seen in Fe deficiency, suggesting that the changes are a consequence of the decrease in serum Fe, implying that the developing fetus not only suffers from Cu, but also from Fe deficiency.


Assuntos
Cobre/deficiência , Dieta , Ferro/metabolismo , Gravidez/metabolismo , Animais , Proteínas de Transporte de Cátions/análise , Ceruloplasmina/análise , Cobre/administração & dosagem , Cobre/metabolismo , Feminino , Desenvolvimento Fetal/fisiologia , Perfilação da Expressão Gênica/métodos , Fígado/metabolismo , Tamanho do Órgão , Placenta/metabolismo , RNA Mensageiro/análise , Ratos , Receptores da Transferrina/análise
20.
J Nutr ; 136(5): 1171-7, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16614400

RESUMO

Imbalances in nutrition during pregnancy can lead to long-, as well as short-term consequences, a phenomenon known as fetal programming. However, there is little information about when the fetus is most sensitive to its environment during gestation. We hypothesize that different fetal systems are most vulnerable to nutritional stress during periods of maximal growth and differentiation. We used iron (Fe) deficiency, which causes hypertension in the offspring, to test this hypothesis. We examined development between embryonic day (E) 10.5 and 12.5, when cardiovascular development is maximal, using whole embryo culture. Female rats were fed Fe-deficient or control diet for 4 wk before mating and up to E10.5. The embryos were cultured for 48 h in 95% rat serum collected from males fed either a control or Fe-deficient diet. Growth was impaired and heart size increased in embryos taken from Fe-deficient mothers and cultured in deficient serum compared with control embryos cultured in control serum. To test whether restoring normal Fe levels could reverse these effects, we cultured embryos from control and deficient dams in either control or deficient medium. The yolk sac circulation of embryos from dams fed either diet cultured in deficient medium was less developed, with a thinner and less branched network than that in all embryos cultured in control serum. The heart was enlarged in embryos of deficient dams cultured in deficient serum compared with the heart size of those cultured in control serum. Culturing embryos in control serum reversed these changes. We conclude, therefore, that this period of cardiovascular organogenesis is one of the sensitive windows during which optimal Fe status is critical for normal development.


Assuntos
Anemia Ferropriva/fisiopatologia , Sistema Cardiovascular/embriologia , Implantação do Embrião/fisiologia , Desenvolvimento Embrionário/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Encéfalo/embriologia , Modelos Animais de Doenças , Feminino , Hipertensão/epidemiologia , Gravidez , Ratos , Saco Vitelino/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...