Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
País como asunto
Tipo del documento
Publication year range
1.
BMC Vet Res ; 20(1): 64, 2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38389107

RESUMEN

BACKGROUND: The critically low hepatic iron stores of newborn piglets are considered to be a major cause of neonatal iron deficiency in modern breeds of domestic pig (Sus domestica). The main factor believed to contribute to this phenomenon is large litter size, which has been an objective of selective breeding of pigs for decades. As consequence, iron transferred from the pregnant sow has to be distributed among a greater number of fetuses. RESULTS: Here, we investigated whether litter size influences red blood cell (RBC) indices and iron parameters in Polish Large White (PLW) piglets and gilts. Small and large litters were produced by the transfer of different numbers of embryos, derived from the same superovulated donor females, to recipient gilts. Piglets from large litters obtained following routine artificial insemination were also examined. Our results clearly demonstrated that varying the number of piglets in a litter did not affect the RBC and iron status of 1-day-old piglets, with all showing iron deficiency anemia. In contrast, gilts with small litters displayed higher RBC and iron parameters compared to mothers with large litters. A comparative analysis of the RBC status of wild boars (having less than half as many piglets per litter as domestic pigs) and PLW pigs, demonstrated higher RBC count, hemoglobin level and hematocrit value of both wild boar sows and piglets, even compared to small-litter PLW animals. CONCLUSIONS: These findings provide evidence that RBC and iron status in newborn PLW piglets are not primarily determined by litter size, and indicate the need to study the efficiency of iron transport across the placenta in domestic pig and wild boar females.


Asunto(s)
Hierro , Sus scrofa , Embarazo , Porcinos , Animales , Femenino , Tamaño de la Camada , Animales Recién Nacidos , Placenta
2.
Int J Mol Sci ; 25(7)2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38612915

RESUMEN

In pigs, iron deficiency anemia (IDA) is a common disorder that occurs during the early postnatal period, leading to the stunted growth and increased mortality of piglets. The main cause of IDA is low iron stores in the liver of newborn piglets; these stores constitute the main source of iron needed to satisfy the erythropoietic requirements of the piglets in their first weeks of life. Insufficient iron stores in piglets are usually due to the inadequate placental iron transfer from the sow to the fetuses. Therefore, iron supplementation in pregnant sows has been implemented to enhance placental iron transfer and increase iron accumulation in the liver of the fetuses. Over the years, several oral and parenteral approaches have been attempted to supplement sows with various iron preparations, and consequently, to improve piglets' red blood cell indices. However, there is debate with regard to the effectiveness of iron supplementation in pregnant sows for preventing IDA in newborn piglets. Importantly, this procedure should be carried out with caution to avoid iron over-supplementation, which can lead to iron toxicity. This article aims to critically review and evaluate the use of iron supplementation in pregnant sows as a procedure for preventing IDA in piglets.


Asunto(s)
Anemia Ferropénica , Femenino , Embarazo , Animales , Porcinos , Anemia Ferropénica/prevención & control , Anemia Ferropénica/veterinaria , Hierro , Placenta , Hígado , Suplementos Dietéticos
3.
Am J Hematol ; 96(6): 659-670, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33684239

RESUMEN

The demand for iron is high in pregnancy to meet the increased requirements for erythropoiesis. Even pregnant females with initially iron-replete stores develop iron-deficiency anemia, due to inadequate iron absorption. In anemic females, the maternal iron supply is dedicated to maintaining iron metabolism in the fetus and placenta. Here, using a mouse model of iron deficiency in pregnancy, we show that iron recycled from senescent erythrocytes becomes a predominant source of this microelement that can be transferred to the placenta in females with depleted iron stores. Ferroportin is a key protein in the molecular machinery of cellular iron egress. We demonstrate that under iron deficiency in pregnancy, levels of ferroportin are greatly reduced in the duodenum, placenta and fetal liver, but not in maternal liver macrophages and in the spleen. Although low expression of both maternal and fetal hepcidin predicted ferroportin up-regulation in examined locations, its final expression level was very likely correlated with tissue iron status. Our results argue that iron released into the circulation of anemic females is taken up by the placenta, as evidenced by high expression of iron importers on syncytiotrophoblasts. Then, a substantial decrease in levels of ferroportin on the basolateral side of syncytiotrophoblasts, may be responsible for the reduced transfer of iron to the fetus. As attested by the lowest decrease in iron content among analyzed tissues, some part is retained in the placenta. These findings confirm the key role played by ferroportin in tuning iron turnover in iron-deficient pregnant mouse females and their fetuses.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Deficiencias de Hierro , Hierro de la Dieta/administración & dosificación , Hígado/metabolismo , Complicaciones del Embarazo/metabolismo , Bazo/metabolismo , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Citocinas/sangre , Duodeno/metabolismo , Envejecimiento Eritrocítico , Índices de Eritrocitos , Femenino , Feto/metabolismo , Hemoglobinas/metabolismo , Hepcidinas/biosíntesis , Hepcidinas/genética , Hierro/metabolismo , Hígado/embriología , Macrófagos/metabolismo , Intercambio Materno-Fetal , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones de la Cepa 129 , Proteínas Musculares/sangre , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Especificidad de Órganos , Fagocitosis , Placenta/metabolismo , Embarazo , Regulación hacia Arriba
4.
Int J Mol Sci ; 22(18)2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34576090

RESUMEN

Iron deficiency is the most common mammalian nutritional disorder. However, among mammalian species iron deficiency anemia (IDA), occurs regularly only in pigs. To cure IDA, piglets are routinely injected with high amounts of iron dextran (FeDex), which can lead to perturbations in iron homeostasis. Here, we evaluate the therapeutic efficacy of non-invasive supplementation with Sucrosomial iron (SI), a highly bioavailable iron supplement preventing IDA in humans and mice and various iron oxide nanoparticles (IONPs). Analysis of red blood cell indices and plasma iron parameters shows that not all iron preparations used in the study efficiently counteracted IDA comparable to FeDex-based supplementation. We found no signs of iron toxicity of any tested iron compounds, as evaluated based on the measurement of several toxicological markers that could indicate the occurrence of oxidative stress or inflammation. Neither SI nor IONPs increased hepcidin expression with alterations in ferroportin (FPN) protein level. Finally, the analysis of the piglet gut microbiota indicates the individual pattern of bacterial diversity across taxonomic levels, independent of the type of supplementation. In light of our results, SI but not IONPs used in the experiment emerges as a promising nutritional iron supplement, with a high potential to correct IDA in piglets.


Asunto(s)
Anemia Ferropénica/tratamiento farmacológico , Suplementos Dietéticos , Compuestos Férricos/administración & dosificación , Compuestos Férricos/uso terapéutico , Nanopartículas Magnéticas de Óxido de Hierro/administración & dosificación , Nanopartículas Magnéticas de Óxido de Hierro/química , Administración Oral , Anemia Ferropénica/sangre , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Duodeno/metabolismo , Compuestos Férricos/farmacología , Compuestos Ferrosos/uso terapéutico , Hepcidinas/sangre , Hepcidinas/genética , Masculino , Microbiota , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos
5.
Int J Mol Sci ; 21(20)2020 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-33092142

RESUMEN

In most mammals, neonatal intravascular hemolysis is a benign and moderate disorder that usually does not lead to anemia. During the neonatal period, kidneys play a key role in detoxification and recirculation of iron species released from red blood cells (RBC) and filtered out by glomeruli to the primary urine. Activity of heme oxygenase 1 (HO1), a heme-degrading enzyme localized in epithelial cells of proximal tubules, seems to be of critical importance for both processes. We show that, in HO1 knockout mouse newborns, hemolysis was prolonged despite a transient state and exacerbated, which led to temporal deterioration of RBC status. In neonates lacking HO1, functioning of renal molecular machinery responsible for iron reabsorption from the primary urine (megalin/cubilin complex) and its transfer to the blood (ferroportin) was either shifted in time or impaired, respectively. Those abnormalities resulted in iron loss from the body (excreted in urine) and in iron retention in the renal epithelium. We postulate that, as a consequence of these abnormalities, a tight systemic iron balance of HO1 knockout neonates may be temporarily affected.


Asunto(s)
Hemo-Oxigenasa 1/deficiencia , Hemólisis , Hierro/metabolismo , Riñón/metabolismo , Insuficiencia Renal/metabolismo , Anemia/sangre , Anemia/terapia , Animales , Animales Recién Nacidos , Recuento de Eritrocitos , Femenino , Hemo/metabolismo , Hemo-Oxigenasa 1/genética , Hierro/orina , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Insuficiencia Renal/genética , Insuficiencia Renal/terapia
6.
Genes (Basel) ; 12(9)2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34573364

RESUMEN

Iron is an essential nutrient during all stages of mammalian development. Studies carried out over the last 20 years have provided important insights into cellular and systemic iron metabolism in adult organisms and led to the deciphering of many molecular details of its regulation. However, our knowledge of iron handling in prenatal development has remained remarkably under-appreciated, even though it is critical for the health of both the embryo/fetus and its mother, and has a far-reaching impact in postnatal life. Prenatal development requires a continuous, albeit quantitatively matched with the stage of development, supply of iron to support rapid cell division during embryogenesis in order to meet iron needs for erythropoiesis and to build up hepatic iron stores, (which are the major source of this microelement for the neonate). Here, we provide a concise overview of current knowledge of the role of iron metabolism-related genes in the maintenance of iron homeostasis in pre- and post-implantation development based on studies on transgenic (mainly knock-out) mouse models. Most studies on mice with globally deleted genes do not conclude whether underlying in utero iron disorders or lethality is due to defective placental iron transport or iron misregulation in the embryo/fetus proper (or due to both). Therefore, there is a need of animal models with tissue specific targeted deletion of genes to advance the understanding of prenatal iron metabolism.


Asunto(s)
Desarrollo Embrionario/genética , Hierro/metabolismo , Proteínas/genética , Proteínas/metabolismo , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Hemo , Ratones Transgénicos , Embarazo
7.
Cells ; 10(9)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34571908

RESUMEN

The critical function of ferroportin (Fpn) in maintaining iron homeostasis requires complex and multilevel control of its expression. Besides iron-dependent cellular and systemic control of Fpn expression, other metals also seem to be involved in regulating the Fpn gene. Here, we found that copper loading significantly enhanced Fpn transcription in an Nrf2-dependent manner in primary bone-marrow-derived macrophages (BMDMs). However, prolonged copper loading resulted in decreased Fpn protein abundance. Moreover, CuCl2 treatment induced Fpn expression in RAW 264.7 macrophages at both the mRNA and protein level. These data suggest that cell-type-specific regulations have an impact on Fpn protein stability after copper loading. Transcriptional suppression of Fpn after lipopolysaccharide (LPS) treatment contributes to increased iron storage inside macrophages and may result in anemia of inflammation. Here, we observed that in both primary BMDMs and RAW 264.7 macrophages, LPS treatment significantly decreased Fpn mRNA levels, but concomitant CuCl2 stimulation counteracted the transcriptional suppression of Fpn and restored its expression to the control level. Overall, we show that copper loading significantly enhances Fpn transcription in macrophages, while Fpn protein abundance in response to CuCl2 treatment, depending on macrophage type and factors specific to the macrophage population, can influence Fpn regulation in response to copper loading.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis , Hierro/metabolismo , Macrófagos/efectos de los fármacos , Animales , Proteínas de Transporte de Catión/genética , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Ratones
8.
Nutrients ; 13(1)2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-33396831

RESUMEN

In mammals, 2 × 1012 red blood cells (RBCs) are produced every day in the bone marrow to ensure a constant supply of iron to maintain effective erythropoiesis. Impaired iron absorption in the duodenum and inefficient iron reutilization from senescent RBCs by macrophages contribute to the development of anemia. Ferroportin (Fpn), the only known cellular iron exporter, as well as hephaestin (Heph) and ceruloplasmin, two copper-dependent ferroxidases involved in the above-mentioned processes, are key elements of the interaction between copper and iron metabolisms. Crosslinks between these metals have been known for many years, but metabolic effects of one on the other have not been elucidated to date. Neonatal iron deficiency anemia in piglets provides an interesting model for studying this interplay. In duodenal enterocytes of young anemic piglets, we identified iron deposits and demonstrated increased expression of ferritin with a concomitant decline in both Fpn and Heph expression. We postulated that the underlying mechanism involves changes in copper distribution within enterocytes as a result of decreased expression of the copper transporter-Atp7b. Obtained results strongly suggest that regulation of iron absorption within enterocytes is based on the interaction between proteins of copper and iron metabolisms and outcompetes systemic regulation.


Asunto(s)
Anemia Ferropénica/metabolismo , Proteínas de Transporte de Catión/biosíntesis , ATPasas Transportadoras de Cobre/biosíntesis , Cobre/metabolismo , Regulación hacia Abajo , Duodeno/metabolismo , Enterocitos/metabolismo , Enfermedades de los Porcinos/metabolismo , Anemia Ferropénica/veterinaria , Animales , Deficiencias de Hierro , Porcinos
9.
Animals (Basel) ; 10(7)2020 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-32610535

RESUMEN

BACKGROUND: The similarities between swine and humans in physiological and genomic patterns, as well as significant correlation in size and anatomy, make pigs an useful animal model in nutritional studies during pregnancy. In humans and pigs iron needs exponentially increase during the last trimester of pregnancy, mainly due to increased red blood cell mass. Insufficient iron supply during gestation may be responsible for the occurrence of maternal iron deficiency anemia and decreased iron status in neonates. On the other hand, preventive iron supplementation of non-anemic mothers may be of potential risk due to iron toxicity. Several different regimens of iron supplementation have been applied during pregnancy. The majority of oral iron supplementations routinely applied to pregnant sows provide inorganic, non-heme iron compounds, which exhibit low bioavailability and intestinal side effects. The aim of this study was to check, using pig as an animal model, the effect of sucrosomial ferric pyrophosphate (SFP), a new non-heme iron formulation on maternal and neonate iron and hematological status, placental transport and pregnancy outcome; Methods: Fifteen non-anemic pregnant sows were recruited to the experiment at day 80 of pregnancy and randomized into the non-supplemented group (control; n = 5) and two groups receiving oral iron supplementation-sows given sucrosomial ferric pyrophosphate, 60 mg Fe/day (SFP; n = 5) (SiderAL®, Pisa, Italy) and sows given ferrous sulfate 60 mg Fe/day (Gambit, Kutno, Poland) (FeSO4; n = 5) up to delivery (around day 117). Biological samples were collected from maternal and piglet blood, placenta and piglet tissues. In addition, data on pregnancy outcome were recorded.; Results: Results of our study show that both iron supplements do not alter neither systemic iron homeostasis in pregnant sows nor their hematological status at the end of pregnancy. Moreover, we did not detect any changes of iron content in the milk and colostrum of iron supplemented sows in comparison to controls. Neonatal iron status of piglets from iron supplemented sows was not improved compared with the progeny of control females. No statistically significant differences were found in average piglets weight and number of piglets per litter between animals from experimental groups. The placental expression of iron transporters varied depending on the iron supplement.

10.
Biol Trace Elem Res ; 196(2): 472-480, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31701465

RESUMEN

Heme is an efficient dietary iron supplement applied in humans and animals to prevent iron deficiency anemia (IDA). We have recently reported that the use of bovine hemoglobin as a dietary source of heme iron efficiently counteracts the development of IDA in young piglets, which is the common problem in pig industry. Here, we used maternal Polish Large White and terminal sire breed (L990) pigs differing in traits for meat production to evaluate the long-term effect of split supplementation with intramuscularly administered small amount of iron dextran and orally given hemoglobin on hematological indices, iron status, growth performance, slaughter traits, and meat quality at the end of fattening. Results of our study show that in pigs of both breeds split supplementation was effective in maintaining physiological values of RBC and blood plasma iron parameters as well as growth performance, carcass parameters, and meat quality traits. Our results prove the effectiveness of split iron supplementation of piglets in a far-reach perspective.


Asunto(s)
Eritrocitos/efectos de los fármacos , Hemoglobinas/metabolismo , Complejo Hierro-Dextran/farmacología , Hierro/sangre , Carne/análisis , Porcinos , Administración Oral , Animales , Composición Corporal/efectos de los fármacos , Suplementos Dietéticos , Hemoglobinas/administración & dosificación , Complejo Hierro-Dextran/administración & dosificación , Masculino , Polonia , Porcinos/anatomía & histología , Porcinos/sangre , Porcinos/crecimiento & desarrollo , Porcinos/metabolismo , Factores de Tiempo , Aumento de Peso/efectos de los fármacos
11.
Pharmaceuticals (Basel) ; 12(1)2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30699979

RESUMEN

The authors wish to make the following corrections to this paper [¹]: the term "liposomal" should be replaced with the term "sucrosomial" in the following places [...].

12.
Pharmaceuticals (Basel) ; 11(4)2018 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-30467279

RESUMEN

In pigs, iron deficiency anemia (IDA) is the most prevalent deficiency disorder during the early postnatal period, frequently developing into a serious illness. On the other hand, in humans, only low-birth-weight infants, including premature infants, are especially susceptible to developing IDA. In both human and pig neonates, the initial cause of IDA is low birth iron stores. In piglets this shortage of stored iron results mainly from genetic selection over the past few decades for large litter sizes and high birth weights. As a consequence, pregnant sows cannot provide a sufficient amount of iron to the increasing number of developing fetuses. Supplementation with iron is a common practice for the treatment of IDA in piglets. For decades, the preferred procedure for delivering iron supplements during early life stages has been through the intramuscular injection of a large amount of iron dextran. However, this relatively simple therapy, which in general, efficiently corrects IDA, may generate toxic effects, and by inducing hepcidin expression, may decrease bioavailability of supplemental iron. New iron supplements are considered herein with the aim to combine the improvement of hematological status, blunting of hepcidin expression, and minimizing the toxicity of the administered iron. We propose that iron-deficient piglets constitute a convenient animal model for performing pre-clinical studies with iron supplements.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda