Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Asia Pac J Clin Nutr ; 33(2): 184-193, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38794978

RESUMEN

BACKGROUND AND OBJECTIVES: This study aimed to assess the associations of maternal iron status and placental iron transport proteins expression with the risk of pre-eclampsia (PE) in Chinese pregnant women. METHODS AND STUDY DESIGN: A total of 94 subjects with PE and 112 healthy pregnant women were enrolled. Fasting blood samples were collected to detect maternal iron status. The placenta samples were collected at delivery to detect the mRNA and protein expression of divalent metal transporter 1 (DMT1) and ferroportin-1 (FPN1). Logistic analysis was used to explore the associations of maternal iron status with PE risk. The associations of placental iron transport proteins with maternal iron status were explored. RESULTS: After adjusting for covariates, dietary total iron, non-heme iron intake and serum hepcidin were negatively associated with PE, with adjusted ORs (95%CIs) were 0.40 (0.17, 0.91), 0.42 (0.18, 0.94) and 0.02 (0.002, 0.13) for the highest versus lowest tertile, respectively. For the highest tertile versus lowest tertile, serum iron (4.08 (1.58, 10.57)) and ferritin (5.61 (2.36, 13.31)) were positively associated with PE. The mRNA expressions and protein levels of DMT1 and FPN1 in placenta were up-regulated in the PE group (p < 0.05). The mRNA expressions of DMT1 and FPN1 in placenta showed a negative correlation with the serum hepcidin (r = -0.71, p < 0.001; r = -0.49, p < 0.05). CONCLUSIONS: In conclusion, the maternal iron status were closely associated with PE risk, placental DMT1 and FPN1 were upregulated in PE which may be a promising target for the prevention of PE.


Asunto(s)
Proteínas de Transporte de Catión , Hierro , Placenta , Preeclampsia , Humanos , Femenino , Embarazo , Preeclampsia/epidemiología , Preeclampsia/sangre , Estudios de Casos y Controles , Adulto , Hierro/sangre , Hierro/metabolismo , Placenta/metabolismo , Proteínas de Transporte de Catión/genética , Hepcidinas/sangre , Factores de Riesgo , China/epidemiología , Estado Nutricional
2.
J Clin Biochem Nutr ; 74(1): 1-8, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38292117

RESUMEN

Mammalian cells contain thousands of metalloproteins and evolved systems to correctly incorporate metal cofactors into their designated sites. Among the transient metals in living cells, iron is the most abundant element that present as an iron sulfur cluster, mono- and dinuclear iron centers or heme for catalytic reactions. Iron homeostasis is tightly regulated by intestinal iron absorption in mammals owing to the lack of an iron excretive transport system, apart from superficial epithelial cell detachment and urinary outflow reabsorptive impairment. In mammals, the central site for iron absorption is in the duodenum, where the divalent metal transporter 1 is essential for iron uptake. The most notable manifestation of mutated divalent metal transporter 1 presents as iron deficiency anemia in humans. In contrast, the mutation of ferroportin, which exports iron, causes iron overload by either gain or loss of function. Furthermore, hepcidin secretion from the liver suppresses iron efflux by internalizing and degrading ferroportin; thus, the hepcidin/ferroportin axis is extensively investigated for its potential as a therapeutic target to treat iron overload. This review focuses on the divalent metal transporter 1-mediated intestinal iron uptake and hepcidin/ferroportin axis that regulate systemic iron homeostasis.

3.
J Biol Chem ; 295(5): 1212-1224, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31882536

RESUMEN

The natural resistance-associated macrophage protein (Nramp) family encompasses transition metal and proton cotransporters that are present in many organisms from bacteria to humans. Recent structures of Deinococcus radiodurans Nramp (DraNramp) in multiple conformations revealed the intramolecular rearrangements required for alternating access of the metal-binding site to the external or cytosolic environment. Here, using recombinant proteins and metal transport and cysteine accessibility assays, we demonstrate that two parallel cytoplasm-accessible networks of conserved hydrophilic residues in DraNramp, one lining the wide intracellular vestibule for metal release and the other forming a narrow proton transport pathway, are essential for metal transport. We further show that mutagenic or posttranslational modifications of transmembrane helix (TM) 6b, which structurally links these two pathways, impede normal conformational cycling and metal transport. TM6b contains two highly conserved histidines, His232 and His237 We found that different mutagenic perturbations of His232, just below the metal-binding site along the proton exit route, differentially affect DraNramp's conformational state, suggesting that His232 serves as a pivot point for conformational changes. In contrast, any replacement of His237, lining the metal exit route, locked the transporter in a transport-inactive outward-closed state. We conclude that these two histidines, and TM6b more broadly, help trigger the bulk rearrangement of DraNramp to the inward-open state upon metal binding and facilitate return of the empty transporter to an outward-open state upon metal release.


Asunto(s)
Proteínas de Transporte de Catión/química , Deinococcus/química , Histidina/química , Metales/metabolismo , Secuencia de Aminoácidos/genética , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Cobalto/química , Cobalto/metabolismo , Deinococcus/genética , Deinococcus/metabolismo , Histidina/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Transporte Iónico , Manganeso/química , Manganeso/metabolismo , Metales/química , Modelos Moleculares , Mutación , Conformación Proteica , Procesamiento Proteico-Postraduccional/genética , Protones
4.
Am J Physiol Cell Physiol ; 318(2): C263-C271, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31721611

RESUMEN

Iron deficiency anemia is a common complication of ulcerative colitis (UC) that can profoundly impact quality of life. Most iron absorption occurs in the duodenum via divalent metal transporter 1 (DMT1)-mediated uptake and ferroportin-1 (FPN1)-mediated export across the apical and basolateral membranes, respectively. However, the colon also contains iron transporters and can participate in iron absorption. Studies have shown increased duodenal DMT1 and FPN1 in patients with UC, but there is conflicting evidence about whether expression is altered in UC colon. We hypothesized that expression of colonic DMT1 and FPN1 will also increase to compensate for iron deficiency. Quantitative RT-PCR and Western blot analyses were performed on duodenal and colonic segmental (right colon, transverse colon, left colon, and rectum) biopsies obtained during colonoscopy. DMT1 mRNA and protein abundances in colonic segments were approximately equal to those in the duodenum, whereas colonic FPN1 mRNA and protein abundances of colonic segments were about one-quarter of those of the duodenum. DMT1 specific mRNA and protein abundances were increased twofold, whereas FPN1 mRNA and protein expressions were increased fivefold in UC distal colon. Immunofluorescence studies revealed enhanced expression of apical membrane- and basolateral membrane-localized DMT1 and FPN1 in UC human colon, respectively. Increased DMT1 expression was associated with enhanced 2-(3-carbamimidoylsulfanylmethyl-benzyl)-isothiourea (CISMBI, DMT1 specific inhibitor)-sensitive 59Fe uptake in UC human colon. We conclude from these results that patients with active UC have increased expression of colonic iron transporters and increased iron absorption, which may be targeted in the treatment of UC-related anemia.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Colitis Ulcerosa/metabolismo , Colon/metabolismo , Absorción Intestinal/fisiología , Hierro/metabolismo , Factores de Transcripción/metabolismo , Animales , Duodeno/metabolismo , Humanos , Transporte Iónico/fisiología , Calidad de Vida , ARN Mensajero/metabolismo
5.
J Neurochem ; 154(2): 177-189, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31693761

RESUMEN

Alzheimer's disease (AD) is characterized by accumulation of amyloid-beta (Aß) senile plaques in patients' brain tissues. Elevated levels of interleukin-1beta (IL-1ß) have been identified in cerebrospinal fluid of living AD patients and in animal models of AD. Increased expression of IL-1ß and iron accumulation have been identified in microglial cells that cluster around amyloid plaques in AD mouse models and post-mortem brain tissues of AD patients. The goals of this study were to determine the effects of Aß on the secretion of IL-1ß by microglial cells and whether iron status influences this pro-inflammatory signaling cue. Immortalized microglial (IMG) cells were incubated with Aß with or without iron. qRT-PCR and western blot analyses showed that Aß induces biosynthesis of IL-1ß by IMG cells. IMG cells secrete the mature form of IL-1ß in a caspase 1-dependent manner. Incubation with iron provoked a greater pro-inflammatory response. Inhibition of the iron transporter divalent metal transporter 1 protected IMG cells against Aß-induced inflammation. Potentiation of Aß-elicited IL-1ß induction by iron was also antagonized by ROS inhibitors, supporting the model that divalent metal transporter 1-mediated iron loading and subsequent increase in ROS contribute to the inflammatory effects of Aß in microglia. Immunoblotting and immunofluorescence microscopy indicate that iron enhances Aß activation of NF-κB signaling to promote IL-1ß synthesis. These results support the hypothesis that Aß stimulates IL-1ß expression by activating NF-κB signaling in microglia cells. Most importantly, iron appears to exacerbate the pro-inflammatory effects of Aß to increase IL-1ß levels.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Interleucina-1beta/biosíntesis , Hierro/farmacología , Microglía/efectos de los fármacos , Microglía/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular , Hierro/metabolismo , Ratones
6.
Adv Exp Med Biol ; 1259: 39-51, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32578170

RESUMEN

Cancer metabolism is a well-known target of cancer therapeutics. Classically, cancer metabolism has been studied in terms of the dependence of cancer cells on crucial metabolites, such as glucose and glutamine. But, the accumulating data show that iron metabolism in tumor microenvironment is also an important factor in preserving the survival of cancer cells. Cancer cells have a distinct phenotype of iron metabolism, which secures the much-needed iron for these metabolically active cells. In order to use this iron efficiently, cancer cells need to increase their iron supply and decrease iron loss. As recent research suggests, this is not only done by modifying the expression of iron-related proteins in cancer cells, but also by interaction of cancer cells with other cells from the tumor milieu. Tumor microenvironment is a dynamic environment characterized with intricate relationship between cancer cells, tumor-associated macrophages, fibroblasts, and other cells. Some of the mechanistic aspects of this relationship have been elucidated, while others are yet to be identified. In any case, identifying the details of the iron phenotype of the cells in tumor microenvironment presents with a new therapeutic opportunity to treat this deadly disease.


Asunto(s)
Hierro/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral , Humanos
7.
Molecules ; 25(7)2020 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-32260496

RESUMEN

The influence of probiotic supplementation on iron metabolism remains poorly investigated. However, a range of studies, especially on Lactobacillus plantarum 299v (Lp229v), have indicated a possible positive impact of probiotics on iron absorption. The aim of the study was to determine the effect of multistrain probiotic supply on iron balance. Thirty Wistar rats were randomized into three groups: placebo (KK group), and multistrain probiotic per os in a daily dose of 2.5 × 109 colony forming units (CFU) (PA group) or 1 × 1010 CFU (PB group). Multistrain probiotic consisted of nine bacterial strains: Bifidobacterium bifidum W23, B. lactis W51, B. lactis W52, Lactobacillus acidophilus W37, L. brevis W63, L. casei W56, L. salivarius W24, Lactococcus lactis W19, and Lc. lactis W58, in equal proportions. After six weeks, blood and organ samples were collected. No differences were found between the three groups in terms of serum concentrations of hepcidin (HEPC), lactoferrin (LTF), homocysteine (HCY), ferritin (Ft), or erythroferrone (ErFe), or in liver content of divalent metal transporter 1 (DMT1), transferrin receptors 1 and 2 (TfR), or ZRT/IRT-like protein 14 (ZIP14) proteins. In the overall sample, positive correlations were noted between the serum concentrations of hepcidin and lactoferrin, and hepcidin and ferritin; serum concentration of hepcidin and DMT1 and TfR1 in the liver; and serum concentration of erythroferrone and TfR2 in the liver. The correlations of serum hepcidin and erythroferrone with liver DMT1 and TfR represent significant mechanisms of Fe homeostasis. Our study has shown that multistrain probiotic supplementation used in the experiment did not disrupt the biochemical and hepatic regulatory processes of Fe balance and did not demonstrate significant influence on selected parameters of Fe metabolism.


Asunto(s)
Hepcidinas/sangre , Hígado/metabolismo , Hormonas Peptídicas/sangre , Animales , Bifidobacterium bifidum/fisiología , Suplementos Dietéticos , Ferritinas/sangre , Homocisteína/sangre , Lactobacillus acidophilus/fisiología , Lactoferrina/sangre , Masculino , Probióticos/uso terapéutico , Ratas , Ratas Wistar
8.
J Biol Chem ; 293(20): 7853-7863, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29610275

RESUMEN

Microglia are immune cells of the central nervous system and are implicated in brain inflammation. However, how brain microglia modulate transport and metabolism of the essential metal iron in response to pro- and anti-inflammatory environmental cues is unclear. Here, we characterized uptake of transferrin (Tf)-bound iron (TBI) and non-Tf-bound iron (NTBI) by immortalized microglial (IMG) cells. We found that these cells preferentially take up NTBI in response to the proinflammatory stimulus lipopolysaccharide (LPS) or ß-amyloid (Aß). In contrast, the anti-inflammatory cytokine interleukin 4 (IL-4) promoted TBI uptake. Concordant with these functional data, levels of the Tf receptor (TfR) in IMG cells were up-regulated in response to IL-4, whereas divalent metal transporter-1 (DMT1) and ferritin levels increased in response to LPS or Aß. Similar changes in expression were confirmed in isolated primary adult mouse microglia treated with pro- or anti-inflammatory inducers. LPS-induced changes in IMG cell iron metabolism were accompanied by notable metabolic changes, including increased glycolysis and decreased oxidative respiration. Under these conditions, the extracellular acidification rate was increased, compatible with changes in the cellular microenvironment that would support the pH-dependent function of DMT1. Moreover, LPS increased heme oxygenase-1 (HO1) expression in IMG cells, and iron released because of HO1 activity increased the intracellular labile free-iron pool. Together, this evidence indicates that brain microglia preferentially acquire iron from Tf or from non-Tf sources, depending on their polarization state; that NTBI uptake is enhanced by the proinflammatory response; and that under these conditions microglia sequester both extra- and intracellular iron.


Asunto(s)
Proteínas de Transporte de Catión/genética , Hierro/metabolismo , Microglía/metabolismo , Receptores de Transferrina/genética , Transferrina/genética , Péptidos beta-Amiloides/farmacología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Transporte de Catión/metabolismo , Línea Celular Transformada , Microambiente Celular , Ferritinas/genética , Ferritinas/metabolismo , Regulación de la Expresión Génica , Glucólisis/efectos de los fármacos , Glucólisis/genética , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Concentración de Iones de Hidrógeno , Inflamación , Transporte Iónico , Lipopolisacáridos/farmacología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Fosforilación Oxidativa/efectos de los fármacos , Cultivo Primario de Células , Receptores de Transferrina/metabolismo , Transducción de Señal , Transferrina/metabolismo
9.
Nanomedicine ; 22: 102091, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31626992

RESUMEN

Iron is a nutrient metal, but excess iron promotes tissue damage. Since iron chelation therapies exhibit multiple off-target toxicities, there is a substantial demand for more specific approaches to decrease iron burden in iron overload. While the divalent metal transporter 1 (DMT1) plays a well-established role in the absorption of dietary iron, up-regulation of intestinal DMT1 is associated with iron overload in both humans and rodents. Hence, we developed a novel pH-sensitive multi-compartmental particulate (MCP) oral delivery system that encapsulates DMT1 siRNA and validated its efficacy in mice. Using the gelatin NPs coated with Eudragit® L100-55, we demonstrated that DMT1 siRNA-loaded MCPs down-regulated DMT1 mRNA levels in the duodenum, which was consistent with decreased intestinal absorption of orally-administered 59Fe. Together, the Eudragit® L100-55-based oral siRNA delivery system could provide an effective strategy to specifically down-regulate duodenal DMT1 and mitigate iron absorption.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Sistemas de Liberación de Medicamentos , Silenciador del Gen , Absorción Intestinal , Intestinos/fisiología , Hierro/metabolismo , Nanopartículas/administración & dosificación , Resinas Acrílicas/química , Administración Oral , Animales , Células CACO-2 , Gelatina/química , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Hierro/administración & dosificación , Masculino , Ratones , Nanopartículas/ultraestructura , Tamaño de la Partícula , ARN Interferente Pequeño/metabolismo
10.
Biochem Biophys Res Commun ; 507(1-4): 128-135, 2018 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-30415773

RESUMEN

BACKGROUND INFORMATION: Divalent metal transporter 1 (DMT1) and transferrin receptor (TfR1) are vital proteins for cellular iron uptake. These proteins have hypoxia-responsive elements (HREs) in their 5'-regulatory region, and they are regulated by hypoxia-inducible factor 1α (HIF-1α) transcriptionally under hypoxic condition. Besides, iron regulatory protein 1 (IRP1) regulates DMT1 and TfR1 by binding to iron-responsive elements (IREs) present in their mRNAs to control cellular iron homeostasis. RESULTS: Here, we explored the effect of acute hypoxia on iron uptake. Ferrous iron uptake was elevated by DMT1(+IRE) and TfR1 under acute hypoxia. The luciferase activity analysis revealed that the functional HREs of DMT1 and TfR1 were increased. However, their IREs-dependent luciferase activities were reduced simultaneously. The mRNA stability of TfR1 and DMT1(+IRE) was suppressed under acute hypoxia. The mRNA levels of TfR1 and DMT1(+IRE) were restrain by silencing IRP1. In sharp contrast, HIF-1α overexpression enhanced the mRNA levels of TfR1 and DMT1(+IRE), which reversed the inhibition of IRP1 on both. HIF-1α konckdown suppressed the hypoxia-induced increase expression of TfR1 and DMT1(+IRE), whereas both proteins had little change when further decreased the IRP1 expression under hypoxia. Hypoxia upregulated the protein expression of Ferrtin-L in a time-dependent manner, yet there was no different when IRP1 silencing or overexperssion under hypoxia. The lactate dehydrogenase (LDH) release induced by hypoxia was increased by TfR1 siRNA silence. CONCLUSIONS: We propose that HIF-1/HRE system might play a principal part in hypoxia induced iron uptake.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína 1 Reguladora de Hierro/metabolismo , Hierro/metabolismo , Proteínas de Transporte de Catión/metabolismo , Hipoxia de la Célula/genética , Ferritinas/metabolismo , Células Hep G2 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteína 1 Reguladora de Hierro/genética , Receptores de Transferrina/metabolismo , Elementos de Respuesta/genética
11.
Br J Nutr ; 120(12): 1338-1348, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30499425

RESUMEN

Studies have shown the positive effects of prebiotics on the intestinal absorption of Ca and Fe. The present study evaluated the effect of fructo-oligosaccharide (FOS) supplementation in soya beverage (SB) on absorption mechanisms of Ca and Fe in recently weaned rats. Male Wistar rats were divided into four groups: lactose-free cows' milk (CM), lactose-free CM with FOS (0·8 g/100 ml) (CMF), SB and soya beverage with FOS (0·8 g/100 ml) (SBF). These rats were euthanised after 1 week of treatment. Organ weight, pH of the caecal content and absorption mechanisms of Ca and Fe were evaluated. The results showed that the weight of the caecal contents increased in the CMF and SBF groups, and the pH of the caecal contents was lower in these groups. The Hb levels of the CMF and SB groups were higher when compared with that of the CM group and lower in relation to the SBF group. The apparent Ca and Fe absorption and apparent Ca retention in the CM group were higher when compared with the SB group, whereas in the CMF group, they were higher in relation to the SBF group. Divalent metal transporter 1 (DMT1) protein expression in the duodenum was higher in the SBF group than in the SB and CMF groups. SB resulted in lower intestinal Ca absorption and higher Hb concentration, despite the lower apparent Fe absorption in relation to CM. Supplementation with FOS provided beneficial effects on Hb and DMT1 protein expression in the duodenum, in addition to improving the absorption process.


Asunto(s)
Bebidas , Calcio de la Dieta/metabolismo , Suplementos Dietéticos , Glycine max , Hierro/metabolismo , Oligosacáridos/administración & dosificación , Alimentación Animal , Animales , Proteínas de Transporte de Catión/metabolismo , Duodeno/metabolismo , Hematócrito , Hemoglobinas/análisis , Concentración de Iones de Hidrógeno , Absorción Intestinal , Intestinos , Hígado/metabolismo , Masculino , Leche , Minerales/metabolismo , Prebióticos/análisis , Ratas , Ratas Wistar , Vitamina D/análogos & derivados , Vitamina D/metabolismo , Destete
12.
Br J Nutr ; 117(7): 923-929, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28534724

RESUMEN

To investigate the effects of maternal dietary protein restriction on offspring Fe metabolism, twenty-four second-parity Landrace×Yorkshire sows were randomly allocated to standard-protein (SP) and low-protein (LP) groups. The SP sows were fed diets containing 15 and 18 % crude protein throughout pregnancy and lactation, respectively, whereas the LP sows were subjected to 50 % dietary protein restriction. Offspring birth weight was not affected, but the body weight at weaning (P=0·06) and average daily gain (P=0·01) of the female piglets were significantly decreased. Serum Fe level in the LP piglets was markedly decreased at weaning, especially in males (P=0·03). Serum ferritin level (P=0·08) tended to be lower, yet serum transferrin was greatly higher (P=0·01) in male weaning piglets of the LP group. Duodenal expression of the divalent metal transporter 1 (DMT1) and ferroportin (FPN) was surprisingly reduced (P<0·05) at the level of protein, but not at the mRNA level, in male weaning piglets of the LP group. Male weaning piglets born to the LP sows exhibited higher hepatic hepcidin levels (P=0·09), lower hepatic expression of transferrin (P<0·01) and transferrin receptor 1 (P<0·05) at the level of mRNA. However, no significant differences were observed for hepatic Fe storage, ferritin, transferrin and transferrin receptor 1 protein expression in male weaning piglets of the two groups. These results indicate that maternal protein restriction during pregnancy and lactation influences growth of female offspring at weaning, reduces duodenal expression of Fe transporters (DMT1 and FPN) and decreases serum Fe level in male weaning piglets.


Asunto(s)
Anemia Ferropénica/veterinaria , Proteínas de Transporte de Catión/metabolismo , Dieta con Restricción de Proteínas/veterinaria , Duodeno/metabolismo , Mucosa Intestinal/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Anemia Ferropénica/sangre , Anemia Ferropénica/etiología , Anemia Ferropénica/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Dieta con Restricción de Proteínas/efectos adversos , Femenino , Hepcidinas/genética , Hepcidinas/metabolismo , Hierro/sangre , Hierro/metabolismo , Lactancia , Hígado/metabolismo , Masculino , Embarazo , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Caracteres Sexuales , Porcinos , Enfermedades de los Porcinos/sangre , Enfermedades de los Porcinos/etiología , Enfermedades de los Porcinos/metabolismo , Transferrina/análisis , Transferrina/genética , Transferrina/metabolismo , Destete , Aumento de Peso
13.
Int J Med Sci ; 14(3): 275-283, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28367088

RESUMEN

Iron overload has recently been associated with the changes in the bone microstructure that occur in osteoporosis. However, the effect of iron overload on osteoblasts is unclear. The purpose of this study was to explore the function of divalent metal transporter 1 (DMT1) in the pathological processes of osteoporosis. Osteoblast hFOB1.19 cells were cultured in medium supplemented with different concentrations (0, 50, 100, 200, 300, 400, 500 µmol/L) of ferric ammonium citrate (FAC) as a donor of ferric ions. We used western blotting and immunofluorescence to determine the levels of DMT1 after treatment with FAC. Apoptosis was evaluated by detecting the levels of cleaved caspase 3, BCL2, and BAX with western blotting. Autophagy was evaluated by detecting the levels of LC3 with western blotting and immunofluorescence. Beclin-1 expression was also assessed with western blotting. The autophagy inhibitor 3-methyladenine was used to determine whether autophagy affects the apoptosis induced by FAC. Our results show that FAC increased the levels of DMT1, upregulated the expression of BCL2, and downregulated the apoptosis-related proteins cleaved caspase 3 and BAX. Both LC3I/LC3II levels and beclin-1 were also increased, indicating that FAC increases the accumulation of autophagosomes in hFOB1.19 cells. FAC-induced autophagy was increased by the apoptosis inhibitor 3-MA but was reduced in DMT1 shRNA hFOB1.19 cells. These results suggest that the increased expression of DMT1 induces iron overload and iron overload induces osteoblast autophagy and apoptosis, thus affecting the pathological processes of osteoporosis. Clarifying the mechanisms underlying the effects of DMT1 will allow the identification of novel targets for the prevention and treatment of osteoporosis.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Osteoblastos/metabolismo , Osteoporosis/genética , Factores de Transcripción/genética , Caspasa 3/biosíntesis , Compuestos Férricos/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hierro/administración & dosificación , Hierro/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Osteoblastos/patología , Osteoporosis/metabolismo , Osteoporosis/patología , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Compuestos de Amonio Cuaternario/administración & dosificación , Proteína X Asociada a bcl-2/biosíntesis
14.
Heart Vessels ; 32(11): 1410-1414, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28669019

RESUMEN

The interaction among heart failure (HF), chronic kidney disease (CKD), and anemia is called cardio-renal anemia syndrome. The mechanism of anemia in cardio-renal anemia syndrome is complex and remains completely unknown. We have previously reported that impaired intestinal iron transporters may contribute to the mechanism of anemia in HF using in vivo HF model rats. In this study, we assessed intestinal iron transporters in CKD model rats to investigate the association of intestinal iron transporters in the mechanism of cardio-renal anemia syndrome. CKD was induced by 5/6 nephrectomy in Sprague-Dawley rats. Sham-operated rats served as a control. After 24-week surgery, CKD rats exhibited normocytic normochromic anemia and normal serum erythropoietin levels despite of anemia. Serum iron levels were decreased in CKD rats compared with the controls. Of interest, intestinal expression of critical iron importers, such as duodenal cytochrome b (Dcyt-b) and divalent metal transporter 1 (DMT-1), was decreased in CKD rats compared with the controls. On the other hand, intestinal expression of ferroportin, an intestinal iron exporter, was not different in the control and CKD groups. Moreover, hepatic expression of hepcidin, a regulator of iron homeostasis, did not differ between the control and CKD groups. These results suggest that impaired intestinal expression of Dcyt-b and DMT-1 might be associated with the reduction of an iron uptake in CKD. Taken together, impaired these intestinal iron transporters may become a novel therapeutic target for cardio-renal anemia syndrome.


Asunto(s)
Anemia/genética , Síndrome Cardiorrenal/genética , Proteínas de Transporte de Catión/genética , Citocromos b/genética , Duodeno/metabolismo , Regulación de la Expresión Génica , ARN/genética , Anemia/metabolismo , Animales , Síndrome Cardiorrenal/metabolismo , Proteínas de Transporte de Catión/biosíntesis , Citocromos b/biosíntesis , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Clin Biochem Nutr ; 61(3): 183-188, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29203959

RESUMEN

Iron is an essential nutrient for life and is involved in many important processes such as oxygen transport and DNA synthesis. However, excess amounts of iron can cause carcinogenesis by producing reactive oxygen species. Thus, the cellular transport of iron must be tightly regulated. In the human body, iron may be present as heme or non-heme iron. The mechanisms governing the cellular transport of these forms have not been clearly elucidated. We previously reported that the expression of an important heme transporter, heme carrier protein 1 was regulated by cancer-specific reactive oxygen species derived from mitochondria. In this study, we have asked if mitochondrial reactive oxygen species may also be related with non-heme iron transport. In order to address this question, we have investigated the relationship between mitochondrial reactive oxygen species and accumulation of cellular non-heme iron in a rat gastric normal, cancer and manganese superoxide dismutase-overexpressing cancer cell line, in which reactive oxygen species from mitochondria are specifically scavenged. We have also analyzed the expression of divalent metal transporter 1 and ferroprotin, involved in the incorporation and excretion of non-heme iron, respectively, as well as a hypoxia-related transcription factor HIF-1α, to elucidate the molecular mechanism of non-heme iron accumulation.

16.
Br J Nutr ; 115(4): 637-43, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26625709

RESUMEN

Ca/vitamin D supplementation maintains bone health and decreases stress fracture risk during initial military training (IMT); however, there is evidence that Ca may negatively affect the absorption of other critical micronutrients, particularly Fe. The objective of this randomised, double-blind, placebo-controlled trial was to determine whether providing 2000 mg/d Ca and 25 µg/d vitamin D in a fortified food product during 9 weeks of military training affects Fe status in young adults. Male (n 98) and female (n 54) volunteers enrolled in US Army basic combat training (BCT) were randomised to receive a snack bar with Ca/vitamin D (n 75) or placebo (snack bar without Ca/vitamin D; n 77) and were instructed to consume 2 snack bars/d between meals throughout the training course. Circulating ionised Ca was higher (P0·05) in markers of Fe status between placebo and Ca/vitamin D groups. Collectively, these data indicate that Ca/vitamin D supplementation through the use of a fortified food product consumed between meals does not affect Fe status during IMT.


Asunto(s)
Anemia Ferropénica/etiología , Calcio de la Dieta/efectos adversos , Alimentos Fortificados/efectos adversos , Hierro de la Dieta/antagonistas & inhibidores , Acondicionamiento Físico Humano/efectos adversos , Bocadillos , Vitamina D/efectos adversos , Adolescente , Adulto , Anemia Ferropénica/sangre , Biomarcadores/sangre , Calcio de la Dieta/uso terapéutico , Método Doble Ciego , Femenino , Fracturas por Estrés/epidemiología , Fracturas por Estrés/prevención & control , Humanos , Hierro de la Dieta/metabolismo , Masculino , Personal Militar/educación , Estado Nutricional , Oklahoma/epidemiología , Factores de Riesgo , Estrés Fisiológico , Vitamina D/uso terapéutico , Adulto Joven
17.
Br J Nutr ; 115(4): 585-93, 2016 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824729

RESUMEN

To determine the effects of dietary Fe concentration on Mn bioavailability in rats fed inorganic or organic Mn sources, fifty-four 22-d-old male rats were randomly assigned and fed a basal diet (2·63 mg Fe/kg) supplemented with 0 (low Fe (L-Fe)), 35 (adequate Fe (A-Fe)) or 175 (high Fe (H-Fe)) mg Fe/kg with 10 mg Mn/kg from MnSO4 or Mn-lysine chelate (MnLys). Tissues were harvested after 21 d of feeding. Serum Mn was greater (P<0·05) in MnLys rats than in MnSO4 rats, and in L-Fe rats than in A-Fe or H-Fe rats. Duodenal divalent metal transporter-1 (DMT1) mRNA was lower (P<0·05) in H-Fe rats than in A-Fe rats for the MnSO4 treatment; however, no significant difference was observed between them for MnLys. Liver DMT1 mRNA abundance was greater (P<0·05) in MnSO4 than in the MnLys group for H-Fe rats. The DMT1 protein in duodenum and liver and ferroportin 1 (FPN1) protein in liver was greater (P<0·05) in the MnSO4 group than in the MnLys group, and in L-Fe rats than in H-Fe rats. Duodenal FPN1 protein was greater (P<0·05) in L-Fe rats than in A-Fe rats for the MnLys treatment, but it was not different between them for the MnSO4 treatment. Results suggest that MnLys increased serum Mn concentration as compared with MnSO4 in rats irrespective of dietary Fe concentration, which was not because of the difference in DMT1 and FPN1 expression in the intestine and liver.


Asunto(s)
Suplementos Dietéticos , Hematínicos/efectos adversos , Absorción Intestinal , Hierro de la Dieta/efectos adversos , Manganeso/administración & dosificación , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Quelantes/administración & dosificación , Complejos de Coordinación/administración & dosificación , Suplementos Dietéticos/efectos adversos , Duodeno/metabolismo , Compuestos Ferrosos/administración & dosificación , Regulación del Desarrollo de la Expresión Génica , Hematínicos/administración & dosificación , Hematínicos/metabolismo , Mucosa Intestinal/metabolismo , Hierro/sangre , Hierro de la Dieta/administración & dosificación , Hierro de la Dieta/metabolismo , Hígado/metabolismo , Lisina/administración & dosificación , Masculino , Manganeso/sangre , Manganeso/química , Manganeso/metabolismo , Compuestos de Manganeso/administración & dosificación , Valor Nutritivo , Distribución Aleatoria , Ratas Sprague-Dawley , Sulfatos/administración & dosificación , Destete
18.
Biochim Biophys Acta ; 1843(12): 2967-75, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25239763

RESUMEN

Iron accumulation is observed in the substantia nigra of patients with Parkinson's disease. However, it is unknown whether neurotrophic factors, brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) participate in the modulation of neuronal iron metabolism. Here, we investigated the effects and underlying mechanisms of BDNF and GDNF on the iron influx process in primary cultured ventral mesencephalic neurons. 6-hydroxydopamine-induced enhanced ferrous iron influx via improper up-regulation of divalent metal transporter 1 with iron responsive element (DMT1+IRE) was consistently relieved by BDNF and GDNF. Both the mRNA and protein levels of DMT1+IRE were down-regulated by BDNF or GDNF treatment alone. We further demonstrated the involvement of iron regulatory protein 1 (IRP1) in BDNF- and GDNF-induced DMT1+IRE expression. Extracellular-regulated kinase 1/2 (ERK1/2) and Akt were activated and participated in these processes. Inhibition of ERK1/2 and Akt phosphorylation abolished the down-regulation of IRP1 and DMT1+IRE induced by BDNF and GDNF. Taken together, these results show that BDNF and GDNF ameliorate iron accumulation via the ERK/Akt pathway, followed by inhibition of IRP1 and DMT1+IRE expression, which may provide new targets for the neuroprotective effects of these neurotrophic factors.

19.
Cell Biol Int ; 39(4): 427-34, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25491917

RESUMEN

Ferristatin II, discovered as an iron transport inhibitor, promotes the internalization and degradation of transferrin receptor 1 (TfR1). DMT1, which mediates iron transport across cell membranes, is located at the plasma membrane of enterocytes and imports dietary iron into the cytosol. TfR1 is not directly engaged in the intestinal absorption of free iron, and iron uptake by DMT1 is attenuated by ferristatin II treatment. In this study, we found another function for ferristatin II in iron uptake. Ferristatin II did not cause degradation of DMT1 but did induce DMT1 internalization from the plasma membrane. Dynasore, a small molecule inhibitor of dynamin, did not inhibit this internalization by ferristatin II, which might occur via a clathrin-independent pathway.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Membrana Celular/metabolismo , Hierro/metabolismo , Sulfonas/farmacología , Factores de Transcripción/metabolismo , Antígenos CD/metabolismo , Línea Celular , Clatrina/metabolismo , Citosol/metabolismo , Dinaminas/antagonistas & inhibidores , Enterocitos/citología , Enterocitos/metabolismo , Humanos , Hidrazonas/farmacología , Microscopía Fluorescente , Receptores de Transferrina/metabolismo
20.
Environ Res ; 137: 8-13, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25483413

RESUMEN

Divalent metal transporter 1 (DMT1), a member of the proton-coupled metal ion transporter family, mediates transport of ferrous iron from the lumen of the intestine into the enterocyte and export of iron from endocytic vesicles. It has an affinity not only for iron but also for other divalent cations including manganese, cobalt, nickel, cadmium, lead, copper, and zinc. DMT1 is encoded by the SLC11a2 gene that is located on chromosome 12q13 in humans and express four major mammalian isoforms (1A/+IRE, 1A/-IRE, 2/+IRE and 2/-IRE). Mutations or polymorphisms of DMT1 gene may have an impact on human health by disturbing metal trafficking. To study the possible association of DMT1 gene with the blood levels of some divalent cations such as iron, lead and cadmium, a single nucleotide polymorphism (SNP) (IVS4+44C/A) in DMT1 gene was investigated in 486 unrelated and healthy individuals in a Turkish population by method of polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The genotype frequencies were found as 49.8% homozygote typical (CC), 38.3% heterozygote (CA) and 11.9% homozygote atypical (AA). Metal levels were analyzed by dual atomic absorption spectrometer system and the average levels of iron, lead and cadmium in the blood samples were 446.01 ± 81.87 ppm, 35.59 ± 17.72 ppb and 1.25 ± 0.87 ppb, respectively. Individuals with the CC genotype had higher blood iron, lead and cadmium levels than those with AA and CA genotypes. Highly statistically significant associations were detected between IVS4+44 C/A polymorphism in the DMT1 gene and iron and lead levels (p=0.001 and p=0.036, respectively), but no association was found with cadmium level (p=0.344). This study suggested that DMT1 IVS4+44 C/A polymorphism is associated with inter-individual variations in blood iron, lead and cadmium levels.


Asunto(s)
Cadmio/sangre , Proteínas de Transporte de Catión/genética , Hierro/sangre , Plomo/sangre , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Anciano , Proteínas de Transporte de Catión/metabolismo , Exposición a Riesgos Ambientales , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Turquía , Adulto Joven
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda