Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 10935, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32616918

RESUMEN

Cystic fibrosis (CF) is a genetic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in chronic bacterial lung infections and tissue damage. CF macrophages exhibit reduced bacterial killing and increased inflammatory signaling. Iron is elevated in the CF lung and is a critical nutrient for bacteria, including the common CF pathogen Pseudomonas aeruginosa (Pa). While macrophages are a key regulatory component of extracellular iron, iron metabolism has yet to be characterized in human CF macrophages. Secreted and total protein levels were analyzed in non-CF and F508del/F508del CF monocyte derived macrophages (MDMs) with and without clinically approved CFTR modulators ivacaftor/lumacaftor. CF macrophage transferrin receptor 1 (TfR1) was reduced with ivacaftor/lumacaftor treatment. When activated with LPS, CF macrophage expressed reduced ferroportin (Fpn). After the addition of exogenous iron, total iron was elevated in conditioned media from CF MDMs and reduced in conditioned media from ivacaftor/lumacaftor treated CF MDMs. Pa biofilm formation and viability were elevated in conditioned media from CF MDMs and biofilm formation was reduced in the presence of conditioned media from ivacaftor/lumacaftor treated CF MDMs. Defects in iron metabolism observed in this study may inform host-pathogen interactions between CF macrophages and Pa.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Fibrosis Quística/metabolismo , Hierro/metabolismo , Macrófagos/metabolismo , Pseudomonas aeruginosa/fisiología , Adolescente , Adulto , Aminofenoles/farmacología , Aminopiridinas/farmacología , Benzodioxoles/farmacología , Biopelículas/efectos de los fármacos , Niño , Medios de Cultivo Condicionados/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Combinación de Medicamentos , Femenino , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/genética , Masculino , Persona de Mediana Edad , Quinolonas/farmacología , Esputo/microbiología
2.
J Biol Chem ; 292(31): 12744-12753, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28615439

RESUMEN

Fe-S cofactors are composed of iron and inorganic sulfur in various stoichiometries. A complex assembly pathway conducts their initial synthesis and subsequent binding to recipient proteins. In this minireview, we discuss how discovery of the role of the mammalian cytosolic aconitase, known as iron regulatory protein 1 (IRP1), led to the characterization of the function of its Fe-S cluster in sensing and regulating cellular iron homeostasis. Moreover, we present an overview of recent studies that have provided insights into the mechanism of Fe-S cluster transfer to recipient Fe-S proteins.


Asunto(s)
Homeostasis , Proteína 1 Reguladora de Hierro/fisiología , Hierro/fisiología , Modelos Moleculares , Animales , Apoenzimas/química , Apoenzimas/metabolismo , Liasas de Carbono-Azufre/biosíntesis , Liasas de Carbono-Azufre/química , Liasas de Carbono-Azufre/fisiología , Transporte de Electrón , Regulación Enzimológica de la Expresión Génica , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/fisiología , Humanos , Proteína 1 Reguladora de Hierro/biosíntesis , Proteína 1 Reguladora de Hierro/química , Proteínas de Unión a Hierro/biosíntesis , Proteínas de Unión a Hierro/química , Proteínas de Unión a Hierro/fisiología , Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/química , Proteínas Reguladoras del Hierro/fisiología , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/fisiología , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/química , Proteínas Mitocondriales/fisiología , Chaperonas Moleculares/biosíntesis , Chaperonas Moleculares/química , Chaperonas Moleculares/fisiología , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Elementos de Respuesta , Succinato Deshidrogenasa/biosíntesis , Succinato Deshidrogenasa/química , Succinato Deshidrogenasa/fisiología , Frataxina
3.
Anesthesiology ; 122(4): 908-20, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25501899

RESUMEN

BACKGROUND: Hyperalgesia is one of the negative consequences following intraoperative analgesia with remifentanil. Peroxynitrite is a critical determinant in nociceptive process. Peroxynitrite inactivates iron-sulfur cluster that results in mitochondrial dysfunction and the release of iron, leading to mitochondrial iron accumulation. Iron accumulation mediated by divalent metal transporter 1 (DMT1) plays a key role in N-methyl-D-aspartate neurotoxicity. This study aims to determine whether peroxynitrite contributes to remifentanil-induced postoperative hyperalgesia via DMT1-mediated iron accumulation. METHODS: Behavior testing was performed in rat model at different time points. Three-nitrotyrosine, nitrated manganese superoxide dismutase, and DMT1 with/without iron-responsive element [DMT1(+)IRE and DMT1(-)IRE] in spinal cord were detected by Western blot and immunohistochemistry. Spinal iron concentration was measured using the Perl stain and atomic absorption spectrophotometer. Hydrogen-rich saline imparting selectivity for peroxynitrite decomposition and iron chelator was applied in mechanistic study on the roles of peroxynitrite and iron, as well as the prevention of hyperalgesia. RESULTS: Remifentanil induced thermal and mechanical hyperalgesia at postoperative 48 h. Compared with control, there were higher levels of 3-nitrotyrosine (mean ± SD, hyperalgesia vs. control, 1.22 ± 0.18 vs. 0.25 ± 0.05, n = 4), nitrated manganese superoxide dismutase (1.01 ± 0.1 vs. 0.19 ± 0.03, n = 4), DMT1(-)IRE (1.42 ± 0.19 vs. 0.33 ± 0.06, n = 4), and iron concentration (12.87 ± 1.14 vs. 5.26 ± 0.61 µg/g, n = 6) in remifentanil-induced postoperative hyperalgesia, while DMT1(+)IRE was unaffected. Eliminating peroxynitrite with hydrogen-rich saline protected against hyperalgesia and attenuated DMT1(-)IRE overexpression and iron accumulation. Iron chelator prevented hyperalgesia in a dose-dependent manner. CONCLUSIONS: Our study identifies that spinal peroxynitrite activates DMT1(-)IRE, leading to abnormal iron accumulation in remifentanil-induced postoperative hyperalgesia, while providing the rationale for the development of molecular hydrogen and "iron-targeted" therapies.


Asunto(s)
Proteínas de Transporte de Catión/biosíntesis , Hiperalgesia/metabolismo , Proteínas Reguladoras del Hierro/biosíntesis , Ácido Peroxinitroso/biosíntesis , Piperidinas/toxicidad , Médula Espinal/metabolismo , Animales , Hiperalgesia/inducido químicamente , Hierro/metabolismo , Masculino , Dolor Postoperatorio/metabolismo , Ratas , Ratas Sprague-Dawley , Remifentanilo
4.
Exp Hematol ; 42(12): 1059-67, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25220979

RESUMEN

Iron cardiomyopathy is the leading cause of death in iron overload. Men have twice the mortality rate of women, though the cause is unknown. In hemojuvelin-knockout mice, a model of the disease, males load more cardiac iron than females. We postulated that sex differences in cardiac iron import cause differences in cardiac iron concentration. Reverse transcription polymerase chain reaction was used to measure mRNA of cardiac iron transporters in hemojuvelin-knockout mice. No sex differences were discovered among putative importers of nontransferrin-bound iron (L-type and T-type calcium channels, ZRT/IRT-like protein 14 zinc channels). Transferrin-bound iron transporters were also analyzed; these are controlled by the iron regulatory element/iron regulatory protein (IRE/IRP) system. There was a positive relationship between cardiac iron and ferroportin mRNA in both sexes, but it was significantly steeper in females (p < 0.05). Transferrin receptor 1 and divalent metal transporter 1 were more highly expressed in females than males (p < 0.01 and p < 0.0001, respectively), consistent with their lower cardiac iron levels, as predicted by IRE/IRP regulatory pathways. Light-chain ferritin showed a positive correlation with cardiac iron that was nearly identical in males and females (R(2) = 0.41, p < 0.01; R(2) = 0.56, p < 0.05, respectively), whereas heavy-chain ferritin was constitutively expressed in both sexes. This represents the first report of IRE/IRP regulatory pathways in the heart. Transcriptional regulation of ferroportin was suggested in both sexes, creating a potential mechanism for differential set points for iron export. Constitutive heavy-chain-ferritin expression suggests a logical limit to cardiac iron buffering capacity at levels known to produce heart failure in humans.


Asunto(s)
Apoferritinas/genética , Regulación de la Expresión Génica , Sobrecarga de Hierro/genética , Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/genética , Hierro/metabolismo , Miocardio/metabolismo , ARN Mensajero/biosíntesis , Transcripción Genética , Animales , Apoferritinas/biosíntesis , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI , Hormonas Esteroides Gonadales/administración & dosificación , Hemocromatosis/genética , Hemocromatosis/metabolismo , Proteína de la Hemocromatosis , Hepcidinas/biosíntesis , Hepcidinas/genética , Terapia de Reemplazo de Hormonas , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Hierro de la Dieta/toxicidad , Hígado/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Orquiectomía , Tamaño de los Órganos , Ovariectomía , Subunidades de Proteína , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Caracteres Sexuales
5.
Int J Hematol ; 96(6): 701-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23179904

RESUMEN

Hepcidin is the central regulator of body iron homeostasis, and dysregulation of hepcidin expression causes various clinical disorders, such as anemia and hemochromatosis. Various stimuli, including iron load and interleukin-6, are involved in the regulation of hepcidin expression. We previously reported that serum hepcidin levels were high in patients with end-stage renal disease, compared with healthy subjects. Since metabolic acidosis is commonly observed in these patients, we hypothesized that acidic milieu might augment hepcidin expression. In this study, we investigated the effect of changes in the pH of the microenvironment on hepcidin expression in human hepatoma and leukemia cell lines. We found that hepcidin expression in these cells was augmented by the acidic milieu created with lactic acid, hydrochloric acid and excess carbon dioxide. Acidic milieu did not clearly enhance hepcidin promoter activity, but rather stabilized hepcidin transcript in the hepatoma cells. We speculate that metabolic acidosis may contribute in part to the elevation of serum hepcidin levels in patients with end-stage chronic kidney disease. Further studies are needed to elucidate the association between acidosis and hepcidin expression in various clinical settings.


Asunto(s)
Ácidos/farmacología , Péptidos Catiónicos Antimicrobianos/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hierro/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Péptidos Catiónicos Antimicrobianos/genética , Dióxido de Carbono/farmacología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Microambiente Celular , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Hepcidinas , Homeostasis/efectos de los fármacos , Humanos , Ácido Clorhídrico/farmacología , Concentración de Iones de Hidrógeno , Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/genética , Células K562/efectos de los fármacos , Células K562/metabolismo , Ácido Láctico/farmacología , Neoplasias Hepáticas/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis
6.
Cell Tissue Res ; 344(2): 299-312, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21437659

RESUMEN

The "acute phase" is clinically characterized by homeostatic alterations such as somnolence, adinamia, fever, muscular weakness, and leukocytosis. Dramatic changes in iron metabolism are observed under acute-phase conditions. Rats were administered turpentine oil (TO) intramuscularly to induce a sterile abscess and killed at various time points. Tissue iron content in the liver and brain increased progressively after TO administration. Immunohistology revealed an abundant expression of transferrin receptor-1 (TfR1) in the membrane and cytoplasm of the liver cells, in contrast to almost only nuclear expression of TfR1 in brain tissue. The expression of TfR1 increased at the protein and RNA levels in both organs. Gene expression of hepcidin, ferritin-H, iron-regulatory protein-1, and heme oxygenase-1 was also upregulated, whereas that of hemojuvelin, ferroportin-1, and the hemochromatosis gene was significantly downregulated at the same time points in both the brain and the liver at the RNA level. However, in contrast to observations in the liver, gene expression of the main acute-phase cytokine (interleukin-6) in the brain was significantly upregulated. In vitro experiments revealed TfR1 membranous protein expression in the liver cells, whereas nuclear and cytoplasmic TfR1 protein was detectable in brain cells. During the non-bacterial acute phase, iron content in the liver and brain increased together with the expression of TfR1. The iron metabolism proteins were regulated in a way similar to that observed in the liver, possibly by locally produced acute-phase cytokines. The significance of the presence of TfR1 in the nucleus of the brain cells has to be clarified.


Asunto(s)
Reacción de Fase Aguda/metabolismo , Encéfalo/metabolismo , Proteínas Reguladoras del Hierro/biosíntesis , Hierro/metabolismo , Hígado/metabolismo , Receptores de Transferrina/biosíntesis , Reacción de Fase Aguda/genética , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Línea Celular Tumoral , Proteínas Ligadas a GPI , Regulación de la Expresión Génica , Proteína de la Hemocromatosis , Hepatocitos/metabolismo , Hepcidinas , Humanos , Inmunohistoquímica , Proteínas Reguladoras del Hierro/genética , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Receptores de Transferrina/sangre , Receptores de Transferrina/genética
7.
FEBS J ; 275(15): 3793-803, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18557934

RESUMEN

Hepcidin is a liver produced cysteine-rich peptide hormone that acts as the central regulator of body iron metabolism. Hepcidin is synthesized under the form of a precursor, prohepcidin, which is processed to produce the biologically active mature 25 amino acid peptide. This peptide is secreted and acts by controlling the concentration of the membrane iron exporter ferroportin on intestinal enterocytes and macrophages. Hepcidin binds to ferroportin, inducing its internalization and degradation, thus regulating the export of iron from cells to plasma. The aim of the present study was to develop a novel method to produce human and mouse recombinant hepcidins, and to compare their biological activity towards their natural receptor ferroportin. Hepcidins were expressed in Escherichia coli as thioredoxin fusion proteins. The corresponding peptides, purified after cleavage from thioredoxin, were properly folded and contained the expected four-disulfide bridges without the need of any renaturation or oxidation steps. Human and mouse hepcidins were found to be biologically active, promoting ferroportin degradation in macrophages. Importantly, biologically inactive aggregated forms of hepcidin were observed depending on purification and storage conditions, but such forms were unrelated to disulfide bridge formation.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/biosíntesis , Proteínas Reguladoras del Hierro/biosíntesis , Animales , Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Péptidos Catiónicos Antimicrobianos/fisiología , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Cartilla de ADN , Electroforesis en Gel de Poliacrilamida , Hepcidinas , Humanos , Proteínas Reguladoras del Hierro/aislamiento & purificación , Proteínas Reguladoras del Hierro/fisiología , Espectrometría de Masas/métodos , Ratones , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
8.
J Cell Biochem ; 104(1): 213-23, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17990282

RESUMEN

Bovine herpesvirus 1 (BHV-1), a dsDNA animal virus, is an economically important pathogen of cattle and the aetiological agent of many types of disease. The efficient replication of a DNA virus is strictly dependent on iron since this metal plays a crucial role in the catalytic center of viral ribonucleotide reductase. Consequently, iron metabolism is an important area for virus/host interaction and a large body of evidence suggests that viral infection is potentially influenced by the iron status of the host. The aim of the present study was to address the effects of BHV-1 on iron metabolism in Madin-Darby bovine kidney (MDBK) cells at different times of post-infection. For this purpose, cell viability, iron regulatory proteins (IRPs) activity and levels, transferrin receptor 1 (TfR-1), ferritin expression and LIP were evaluated. Our data demonstrate that a productive BHV-1 infection in MDBK cells determines an overall decrease of IRPs RNA-binding activity without affecting their expression. As consequence of this modulation, an increased ferritin mRNA translation and a decreased TfR-1 mRNA translation were also observed. Moreover, the LIP level was decreased following viral infection. These results are consistent with the hypothesis that by reducing the iron up-take and by enhancing the sequestration of free iron, animal cells will limit the iron availability for virus proliferation. Therefore, the results presented herein support the view that iron metabolism could be critical for the interaction between DNA viruses, such as BHV-1, and mammalian cells. Delineation of the interplay among pathogen and host may provide new antimicrobial agents.


Asunto(s)
Infecciones por Herpesviridae/metabolismo , Herpesvirus Bovino 1/fisiología , Proteínas Reguladoras del Hierro/biosíntesis , Hierro/metabolismo , Replicación Viral , Animales , Bovinos , Línea Celular , Ferritinas/biosíntesis , Riñón/patología , Transferrina/biosíntesis
9.
Liver Int ; 26(8): 976-85, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16953838

RESUMEN

BACKGROUND/AIMS: The altered iron metabolism in hepatocellular carcinomas (HCCs), characterized by the iron-deficient phenotype, is suggested to be of importance for tumour growth. However, the underlying molecular mechanisms remain poorly understood. We asked whether these iron perturbations would involve altered expression of genes controlling iron homeostasis. METHODS: HCCs were induced in rats by the Solt and Farber protocol of chemical hepatocarcinogenesis, and to evaluate the effects of iron loading, one group of animals were supplemented with dietary iron during tumour progression. Tissue iron contents were determined, labelling indices of S-phase nuclei were calculated, and mRNA levels of iron-regulatory genes were quantitated. Protein levels of ferroportin1 were determined with Western blot. RESULTS: HCCs displayed reduced amount of tissue iron and lack of histologically stainable iron. HCCs expressed significantly higher mRNA levels of genes involved in iron uptake (transferrin receptor-1, divalent metal ion transporter-1), ferroxidase activity (Ferritin-H), and iron extrusion (ferroportin1). The protein levels of ferroportin1 in iron-deficient HCCs were similar as in control livers, and did not increase in HCCs exposed to iron. Hepcidin mRNA levels were decreased in iron-deficient HCCs, rose in response to iron loading and correlated to the tissue iron content. CONCLUSIONS: Taken together, the altered expressions of iron-regulatory genes in HCCs possibly reflect an increased demand for bioavailable iron and a high iron turnover in neoplastic cells.


Asunto(s)
Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/genética , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Apoferritinas/biosíntesis , Apoferritinas/genética , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Expresión Génica , Hepcidinas , Neoplasias Hepáticas Experimentales/patología , Masculino , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Receptores de Transferrina/biosíntesis , Receptores de Transferrina/genética
10.
Blood Cells Mol Dis ; 37(2): 95-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16904349

RESUMEN

We tested the effect of iron deprivation on the uptake of iron from ferric citrate by human erythroleukemia K562 cells. The iron uptake after 24-h preincubation in defined iron-free medium was approximately 2-3x higher than after the preincubation in control transferrin-containing medium. The preincubation of K562 cells in iron-free medium together with the inhibitor of protein synthesis cycloheximide completely abrogated the stimulation of the iron uptake. The preincubation in iron-free medium resulted in a slight decrease (20%) of DMT1 mRNA level. The level of Dcytb, ferroportin and hephaestin mRNA did not exert any significant change. We also did not find any significant effect on the protein level of DMT1, Dcytb, ferroportin and hephaestin. We conclude that iron deprivation stimulates the uptake of non-transferrin iron in K562 cells and that this stimulation depends on protein synthesis. It seems that the expression of an unknown or seemingly unrelated protein(s) is involved.


Asunto(s)
Hierro/metabolismo , Transferrina/metabolismo , Cicloheximida/farmacología , Humanos , Hierro/farmacocinética , Proteínas Reguladoras del Hierro/antagonistas & inhibidores , Proteínas Reguladoras del Hierro/biosíntesis , Células K562 , Relación Estructura-Actividad , Factores de Tiempo , Células Tumorales Cultivadas
12.
J Anim Sci ; 83(9): 2137-45, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16100069

RESUMEN

Two experiments were conducted to evaluate the effects of supplemental Fe on the binding activity of iron regulatory proteins (IRP) and the subsequent effect on growth performance and indices of hematological and mineral status of young pigs. In Exp. 1, male pigs (n = 10; 1.8 kg; age = 14 +/- 1 h) were allotted by BW to two treatments (five pigs per treatment). Treatments administered by i.m. injection were as follows: 1) 1 mL of sterile saline solution (Sal); and 2) 1 mL of 200 mg Fe as Fe-dextran (Fe). Pigs were bled (d 0 and 13) to determine hemoglobin (Hb), hematocrit (Hct), transferrin (Tf), and plasma Fe (PFe), and then killed (d 13) to determine spontaneous and 2-mercaptoethanol (2-ME)-inducible IRP RNA binding activity in liver and liver and whole-body mineral concentrations. Contemporary pigs (n = 5; 2.2 kg; age = 14 +/- 2 h) were killed at d 0 to establish baseline (BL1) measurements. In Exp. 2, pigs (six pigs per treatment; 6.5 kg; age = 19 +/- 3 d) were fed a basal diet (Phase 1 = d 0 to 7; Phase 2 = d 7 to 21; Phase 3 = d 21 to 35) supplemented with 0 or 150 mg/kg of Fe as ferrous sulfate and killed at d 35 (18.3 kg; age = 54 +/- 3 d). In addition, pigs (n = 5; 5.9 kg; age = 19 +/- 3 d) were killed at the start of Exp. 2 to establish baseline (BL2) measurements, and liver samples were collected and analyzed for IRP RNA binding activity. In Exp. 1, no difference (P = 0.482) was observed in ADG. On d 13, Fe-treated pigs had greater (P = 0.001) Hb, Hct, and PFe and less (P = 0.002) Tf than Sal-treated pigs. Whole-body Fe concentration was greater (P = 0.002) in Fe- vs. Sal-treated pigs. Treated pigs (Fe or Sal) had greater (P = 0.006) whole-body Cu and less (P = 0.002) whole-body Ca, Mg, Mn, P, and Zn concentrations than BL1. Liver Fe concentration was greater (P = 0.001) in Fe- vs. Sal-treated pigs, but liver Fe concentration of Sal-treated pigs was less (P = 0.001) than that of BL1 pigs. Sal-treated pigs had greater (P = 0.004) spontaneous IRP binding activity than Fe-treated pigs. In Exp. 2, spontaneous and 2-ME inducible IRP binding activities were greater (P = 0.013 and 0.005, respectively) in pigs fed diets containing 0 vs. 150 mg of added Fe/kg of diet. Moreover, pigs fed either treatment for 35 d had greater (P = 0.001) 2-ME inducible IRP binding activity than BL2 pigs. Results indicate that IRP binding activity is influenced by Fe supplementation. Subsequently, other indicators of Fe status are affected via the role of IRP in posttranscriptional expression of Fe storage and transport proteins.


Asunto(s)
Hierro de la Dieta/farmacología , Proteínas Reguladoras del Hierro/metabolismo , Porcinos/fisiología , Animales , Autorradiografía/veterinaria , Proteínas Sanguíneas/efectos de los fármacos , Western Blotting/veterinaria , Suplementos Dietéticos , Crecimiento/efectos de los fármacos , Hematócrito/veterinaria , Hierro/sangre , Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/efectos de los fármacos , Hígado/química , Hígado/efectos de los fármacos , Masculino , Minerales/análisis , Unión Proteica/efectos de los fármacos , Distribución Aleatoria , Porcinos/sangre , Porcinos/crecimiento & desarrollo
13.
Mol Cell Biochem ; 265(1-2): 37-45, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15543932

RESUMEN

Iron may populate distinct hepatocellular iron pools that differentially regulate expression of proteins such as ferritin and transferrin receptor (TfR) through iron-regulatory mRNA-binding proteins (IRPs), and may additionally regulate uptake and accumulation of non-transferrin-bound iron (NTBI). We examined iron-regulatory protein (IRP) binding activity and ferritin/TfR expression in human hepatoma (HepG2) cells exposed to iron at different levels for different periods. Several iron-dependent RNA-binding activities were identified, but only IRP increased with beta-mercaptoethanol. With exposures between 0 and 20 microg/ml iron, decreases in IRP binding accompanied large changes in TfR and ferritin expression, while chelation of residual iron with deferoxamine (DFO) caused a large increase in IRP binding with little additional effect on TfR or ferritin expression. Cellular iron content increased beyond 4 days of exposure to iron at 20 microg/ml, when IRP binding, TfR, and ferritin had all reached stable levels. However, iron content of the cells plateaued by 7 days, or decreased with 24 h exposure to very high concentrations (>50 microg/ml) of iron. These results indicate that iron-replete HepG2 cells exhibit a narrow range of maximal responsiveness of the IRP-regulatory mechanism, whose functional response is blunted both by excessive iron exposure and by removal of iron from a chelatable pool. HepG2 cells are able to limit iron accumulation upon higher or prolonged exposure to NTBI, apparently independent of the IRP mechanism.


Asunto(s)
Proteínas Reguladoras del Hierro/biosíntesis , Hierro/metabolismo , Western Blotting , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Citoplasma/metabolismo , Relación Dosis-Respuesta a Droga , Ácido Edético/farmacología , Ferritinas/química , Ferritinas/metabolismo , Hepatocitos/metabolismo , Humanos , Hierro/química , ARN/química , Receptores de Transferrina/biosíntesis , Proteínas Recombinantes/química , Factores de Tiempo
14.
Am J Physiol Regul Integr Comp Physiol ; 287(4): R894-901, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15178542

RESUMEN

Placental iron transport during the last trimester of pregnancy determines the iron endowment of the neonate. Iron transport is a function of the major iron transport proteins: transferrin receptor-1 (TfR-1) and ferroportin-1 (FPN-1). The mRNAs for TfR-1 and, potentially, FPN-1 are posttranscriptionally regulated by iron regulatory protein (IRP)-1 and IRP-2. We assessed the effect of gestational age and fetal iron status on IRP-1- and IRP-2-binding activity and on the localization and protein expression of TfR-1 and FPN-1 protein at 24-40 wk of gestation in 21 placentas obtained from iron-sufficient nonanemic mothers. Gestational age had no effect on cord serum ferritin concentration, IRP-2 RNA-binding activity, transporter protein location, and TfR-1 or FPN-1 protein expression. IRP-1 activity remained constant until full term, when it decreased (P = 0.01). Placental ferritin (r = 0.76, P < 0.001) and FPN-1 (r = 0.44, P < 0.05) expression increased with gestational age. Fetal iron status, as indexed by cord serum ferritin concentration, was inversely related to placental IRP-1 (r = -0.66, P < 0.001) and IRP-2 (r = -0.42, P = 0.05) activities. Placental ferritin protein expression correlated better with IRP-1 (r = -0.45, P = 0.04) than with IRP-2 (r = -0.35, P = 0.10) activity. Placental TfR-1 and FPN-1 protein expression was independent of fetal or placental iron status and IRP activities. Iron status had no effect on transport protein localization. We conclude that, toward the end of the third trimester of iron-sufficient human pregnancy, the placenta accumulates ferritin and potentially increases placental-fetal iron delivery through increased FPN-1 expression. IRP-1 may have a more dominant role than IRP-2 activity in regulating ferritin expression.


Asunto(s)
Proteínas Portadoras/biosíntesis , Feto/metabolismo , Edad Gestacional , Proteínas Reguladoras del Hierro/biosíntesis , Hierro/metabolismo , Placenta/metabolismo , Adulto , Antígenos CD , Western Blotting , Femenino , Ferritinas/metabolismo , Sangre Fetal/metabolismo , Humanos , Inmunohistoquímica , Estado Nutricional , Embarazo , Tercer Trimestre del Embarazo , Unión Proteica , Receptores de Transferrina/metabolismo
16.
Mol Biochem Parasitol ; 126(2): 231-8, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12615322

RESUMEN

Plasmodium falciparum iron regulatory-like protein (PfIRPa, accession AJ012289) has homology to a family of iron-responsive element (IRE)-binding proteins (IRPs) found in different species. We have previously demonstrated that erythrocyte P. falciparum PfIRPa binds a mammalian consensus IRE and that the binding activity is regulated by iron status. In the work we now report, we have cloned a C-terminus histidine-tagged PfIRPa and overexpressed it in a bacterial expression system in soluble form capable of binding IREs. To overexpress PfIRPa, we used the T7 promoter-driven vector, pET28a(+), in conjunction with the Rosetta(DE3)pLysS strain of E. coli, which carries extra copies of tRNA genes usually found in organisms such as P. falciparum whose genome is (A+T)-rich. The histidine-tagged recombinant protein (rPfIRPa) in soluble form was partially purified using His-bind resin. We searched the plasmodial database, plasmoDB, to identify sequences capable of forming IRE loops using a specially developed algorithm, and found three plasmodial sequences matching the search criteria. In gel retardation assays, rPfIRPa bound three 32P-labeled putative plasmodial IREs with affinity exceeding the affinity for the mammalian consensus IRE. The binding was concentration-dependent and was not inhibited by heparin, an inhibitor of non-specific binding. Immunodepletion of rPfIRPa resulted in substantial inhibition of the signal intensity in the gel retardation assays and in Western blot-determinations of rPfIRPa protein levels. Endogenous PfIRPa retained all three putative 32P-IREs at the same position on the gel as the recombinant PfIRPa.


Asunto(s)
Proteínas Reguladoras del Hierro/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas de Pez Cebra , Animales , Secuencia de Bases , Sitios de Unión , Cartilla de ADN , Humanos , Proteínas Reguladoras del Hierro/biosíntesis , Proteínas Reguladoras del Hierro/genética , Células Jurkat , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Unión Proteica , Proteínas Tirosina Quinasas/metabolismo , Proteínas Protozoarias/biosíntesis , Proteínas Protozoarias/genética , ARN Protozoario/química , ARN Protozoario/genética , Proteínas Recombinantes/metabolismo , Proteínas Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...