Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Molecules ; 28(10)2023 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-37241825

RESUMEN

Iron is an essential element involved in a variety of physiological functions. However, excess iron catalyzes the generation of reactive oxygen species (ROS) via the Fenton reaction. Oxidative stress, caused by an increase in intracellular ROS production, can be a contributory factor to metabolic syndromes such as dyslipidemia, hypertension, and type 2 diabetes (T2D). Accordingly, interest has grown recently in the role and use of natural antioxidants to prevent iron-induced oxidative damage. This study investigated the protective effect of the phenolic acids; ferulic acid (FA) and its metabolite ferulic acid 4-O-sulfate disodium salt (FAS) against excess iron-related oxidative stress in murine MIN6 cells and the pancreas of BALB/c mice. Rapid iron overload was induced with 50 µmol/L ferric ammonium citrate (FAC) and 20 µmol/L 8-hydroxyquinoline (8HQ) in MIN6 cells, while iron dextran (ID) was used to facilitate iron overload in mice. Cell viability was determined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide (MTT) assay, ROS levels were determined by dihydrodichlorofluorescein (H2DCF) cell-permeant probe, iron levels were measured by inductively coupled plasma mass spectrometry (ICP-MS), glutathione, SOD (superoxide dismutase) and lipid peroxidation, and mRNA were assayed with commercially available kits. The phenolic acids enhanced cell viability in iron-overloaded MIN6 cells in a dose-dependent manner. Furthermore, MIN6 cells exposed to iron showed elevated levels of ROS, glutathione (GSH) depletion and lipid peroxidation (p < 0.05) compared to cells that were protected by treatment with FA or FAS. The treatment of BALB/c mice with FA or FAS following exposure to ID increased the nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2) gene levels in the pancreas. Consequently, levels of its downstream antioxidant genes, HO-1, NQO1, GCLC and GPX4, increased in the pancreas. In conclusion, this study shows that FA and FAS protect pancreatic cells and liver tissue from iron-induced damage via the Nrf2 antioxidant activation mechanism.


Asunto(s)
Diabetes Mellitus Tipo 2 , Sobrecarga de Hierro , Ratones , Animales , Antioxidantes/farmacología , Antioxidantes/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Hierro/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal , Diabetes Mellitus Tipo 2/metabolismo , Estrés Oxidativo , Glutatión/metabolismo , Sobrecarga de Hierro/metabolismo , Páncreas/metabolismo
2.
Int J Mol Sci ; 23(24)2022 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36555529

RESUMEN

Ferroptosis is a regulated cell death process characterised by the iron-dependent accumulation of oxidised polyunsaturated fatty acid-containing phospholipids. Its initiation is complicated and involves reactive oxygen species (ROS) and a loss of the activity of the lipid repair enzyme glutathione peroxidase 4 (GPX4). These play critical roles in the development of ferroptotic cell damage by lipid peroxidation. Antioxidant therapy is a promising therapeutic strategy to prevent or even reverse the progression of ferroptosis. This study was designed to demonstrate the protective effect of ferulic acid (FA) against oxidative stress and erastin-mediated ferroptosis in murine MIN6 cells. Cells were treated with FA or its metabolite ferulic acid 4-O-sulfate disodium salt (FAS) and 20 µM of erastin. Cell viability was determined by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide (MTT) assay, iron levels were measured by inductively coupled plasma mass spectrometry (ICP-MS), ROS levels were determined by a dihydrodichlorofluorescein (H2DCF) cell-permeant probe, and glutathione and lipid peroxidation were assayed with commercially available kits. The phenolic acids enhanced cell viability in erastin-treated MIN6 cells in a dose-dependent manner. Furthermore, MIN6 cells exposed to erastin alone showed elevated levels of iron and ROS, glutathione (GSH) depletion, and lipid peroxidation (p < 0.05) compared to cells that were protected by co-treatment with FA or FAS. The treatment of MIN6 cells with FA or FAS following exposure to erastin increased the nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2) protein levels. Consequently, levels of its downstream antioxidant proteins, HO-1, NQO1, GCLC, and GPX4, increased. FA and FAS greatly decreased erastin-induced ferroptosis in the presence of the Nrf2 inhibitor, ML385, through the regulation of Nrf2 response genes. In conclusion, these results show that FA and FAS protect MIN6 cells from erastin-induced ferroptosis by the Nrf2 antioxidant protective mechanism.


Asunto(s)
Ferroptosis , Factor 2 Relacionado con NF-E2 , Animales , Ratones , Antioxidantes/farmacología , Glutatión/metabolismo , Hierro/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
3.
Int J Mol Sci ; 20(19)2019 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-31597407

RESUMEN

Iron, the fourth most abundant element in the Earth's crust, is vital in living organisms because of its diverse ligand-binding and electron-transfer properties. This ability of iron in the redox cycle as a ferrous ion enables it to react with H2O2, in the Fenton reaction, to produce a hydroxyl radical (•OH)-one of the reactive oxygen species (ROS) that cause deleterious oxidative damage to DNA, proteins, and membrane lipids. Ferroptosis is a non-apoptotic regulated cell death that is dependent on iron and reactive oxygen species (ROS) and is characterized by lipid peroxidation. It is triggered when the endogenous antioxidant status of the cell is compromised, leading to lipid ROS accumulation that is toxic and damaging to the membrane structure. Consequently, oxidative stress and the antioxidant levels of the cells are important modulators of lipid peroxidation that induce this novel form of cell death. Remedies capable of averting iron-dependent lipid peroxidation, therefore, are lipophilic antioxidants, including vitamin E, ferrostatin-1 (Fer-1), liproxstatin-1 (Lip-1) and possibly potent bioactive polyphenols. Moreover, most of the enzymes and proteins that cascade or interact in the pathway of ferroptosis such as a subunit of the cystine/glutamate transporter xc- (SLC7A11), glutathione peroxidase 4 (GPX4), and the glutamate-cysteine ligase (GCLC) iron metabolism genes transferrin receptor 1 (TfR1) ferroportin, (Fpn) heme oxygenase 1 (HO-1) and ferritin are regulated by the antioxidant response element of the transcription factor, Nrf2. These, as well as other radical trapping antioxidants (RTAs), are discussed in the current review.


Asunto(s)
Apoptosis , Ferroptosis , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores , Ácidos Grasos/metabolismo , Ferroptosis/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/metabolismo , Hierro/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Polifenoles , Especies Reactivas de Oxígeno/metabolismo , Vitamina E/metabolismo
4.
Biochim Biophys Acta Gen Subj ; 1861(8): 1893-1900, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28552631

RESUMEN

Ferroptosis is a form of regulated cell death that is dependent on iron and reactive oxygen species (ROS) and is characterized by lipid peroxidation. It is morphologically and biochemically distinct and disparate from other processes of cell death. As ferroptosis is induced by inhibition of cysteine uptake or inactivation of the lipid repair enzyme glutathione peroxidase 4 (GPX4), the process is favored by chemical or mutational inhibition of the cystine/glutamate antiporter and culminates in the accumulation of reactive oxygen species (ROS) in the form of lipid hydroperoxides. Excessive lipid peroxidation leads to death by ferroptosis and the phenotype is accentuated respectively by the repletion and depletion of iron and glutathione in cells. Furthermore, oxidized phosphatidylethanolamines (PE) harbouring arachidonoyl (AA) and adrenoyl moieties (AdA) have been shown as proximate executioners of ferroptosis. Induction of ferroptosis due to cysteine depletion leads to the degradation of ferritin (i.e. ferritinophagy), which releases iron via the NCOA4-mediated autophagy pathway. Evidence of the manifestation of ferroptosis in vivo in iron overload mice mutants is emerging. Thus, a concerted synchronization of iron availability, ROS generation, glutamate excess and cysteine deficit leads to ferroptosis. A number of questions on the molecular mechanisms of some features of ferroptosis are highlighted as subjects for future investigations.


Asunto(s)
Autofagia , Muerte Celular , Ferritinas/metabolismo , Hierro/metabolismo , Peroxidación de Lípido , Especies Reactivas de Oxígeno/metabolismo , Animales , Cisteína/metabolismo , Glutatión/metabolismo , Humanos
5.
Br J Nutr ; 117(6): 767-774, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28452291

RESUMEN

Strategies for preventing Fe deficiency include Fe supplementation and Fe fortification of foods. The absorption, metabolism and chemical characteristics of Fe multi-amino acid chelate (IMAAC) are not known. Absorption of IMAAC was compared with FeSO4 in Fe-depleted mice and in vitro chemical studies of the Fe supplement was performed in HuTu 80 cells. Hb repletion study was carried out in Fe-deficient CD1 mice that were fed for 10 d a diet supplemented with ferrous IMAAC or FeSO4. A control group of Fe-replete mice was fed a diet with adequate Fe concentrations throughout the study. Tissues were collected from the mice, and the expression of Fe-related genes was determined by quantitative PCR. Ferric reductase and Fe uptake were evaluated in HuTu 80 cells. Supplementation of the diet with FeSO4 or IMAAC significantly increased Hb levels (P<0·001) in Fe-deficient mice from initial 93·9 (SD 10·8) or 116·2 (SD 9·1) to 191 (SD 0·7) or 200 (SD 0·5) g/l, respectively. Initial and final Hb for the Fe-deficient control group were 87·4 (SD 6·7) and 111 (SD 11·7) g/l, respectively. Furthermore, the liver non-haem Fe of both supplement groups increased significantly (P<0·001). IMAAC was more effective at restoring Fe in the spleen compared with FeSO4 (P<0·005). Gene expression showed the IMAAC supplement absorption is regulated by the body's Fe status as it significantly up-regulated hepcidin (P<0·001) and down-regulated duodenal cytochrome b mRNA (P<0·005), similar to the effects seen with FeSO4. A significant proportion of Fe in IMAAC is reduced by ascorbic acid. Fe absorption in mice and cells was similar for both IMAAC and FeSO4 and both compounds induce and regulate Fe metabolism genes similarly in the maintenance of homeostasis in mice.


Asunto(s)
Aminoácidos/farmacología , Anemia Ferropénica/metabolismo , Suplementos Dietéticos , Duodeno/metabolismo , Absorción Intestinal , Quelantes del Hierro/farmacología , Hierro/farmacocinética , Aminoácidos/uso terapéutico , Anemia Ferropénica/tratamiento farmacológico , Animales , Ácido Ascórbico/farmacología , Disponibilidad Biológica , Línea Celular , Dieta , Regulación de la Expresión Génica , Hemoglobinas/metabolismo , Hepcidinas/metabolismo , Humanos , Hierro/metabolismo , Hierro/uso terapéutico , Quelantes del Hierro/uso terapéutico , Deficiencias de Hierro , Hierro de la Dieta/metabolismo , Hierro de la Dieta/uso terapéutico , Hígado/metabolismo , Masculino , Ratones , Estado Nutricional , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Bazo/metabolismo
6.
Biochim Biophys Acta ; 1840(1): 106-12, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23981688

RESUMEN

BACKGROUND: Duodenal cytochrome b (Dcytb) is a mammalian plasma ferric reductase enzyme that catalyses the reduction of ferric to ferrous ion in the process of iron absorption. The current study investigates the relationship between Dcytb, iron, dehydroascorbate (DHA) and Hif-2α in cultured cell lines. METHODS: Dcytb and Hif-2α protein expression was analysed by Western blot technique while gene regulation was determined by quantitative PCR. Functional analyses were carried out by ferric reductase and (59)Fe uptake assays. RESULTS: Iron and dehydroascorbic acid treatment of cells inhibited Dcytb mRNA and protein expression. Desferrioxamine also enhanced Dcytb mRNA level after cells were treated overnight. Dcytb knockdown in HuTu cells resulted in reduced mRNA expression and lowered reductase activity. Preloading cells with DHA (to enhance intracellular ascorbate levels) did not stimulate reductase activity fully in Dcytb-silenced cells, implying a Dcytb-dependence of ascorbate-mediated ferrireduction. Moreover, Hif-2α knockdown in HuTu cells led to a reduction in reductase activity and iron uptake. CONCLUSIONS: Taken together, this study shows the functional regulation of Dcytb reductase activity by DHA and Hif-2α. GENERAL SIGNIFICANCE: Dcytb is a plasma membrane protein that accepts electrons intracellularly from DHA/ascorbic acid for ferrireduction at the apical surface of cultured cells and enterocytes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Grupo Citocromo b/metabolismo , Ácido Deshidroascórbico/farmacología , Neoplasias Duodenales/metabolismo , Regulación de la Expresión Génica , Hierro/farmacología , Riñón/metabolismo , Oxidorreductasas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Células Cultivadas , Grupo Citocromo b/genética , Neoplasias Duodenales/tratamiento farmacológico , Neoplasias Duodenales/patología , FMN Reductasa/metabolismo , Humanos , Riñón/citología , Riñón/efectos de los fármacos , Oxidorreductasas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Br J Haematol ; 164(4): 586-96, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24236640

RESUMEN

ATOH8 has previously been shown to be an iron-regulated transcription factor, however its role in iron metabolism is not known. ATOH8 expression in HEK293 cells resulted in increased endogenous HAMP mRNA levels as well as HAMP promoter activity. Mutation of the E-box or SMAD response elements within the HAMP promoter significantly reduced the effects of ATOH8, indicating that ATOH8 activates HAMP transcription directly as well as through bone morphogenic protein (BMP) signalling. In support of the former, Chromatin immunoprecipitation assays provided evidence that ATOH8 binds to E-box regions within the HAMP promoter while the latter was supported by the finding that ATOH8 expression in HEK293 cells led to increased phosphorylated SMAD1,5,8 levels. Liver Atoh8 levels were reduced in mice under conditions associated with increased erythropoietic activity such as hypoxia, haemolytic anaemia, hypotransferrinaemia and erythropoietin treatment and increased by inhibitors of erythropoiesis. Hepatic Atoh8 mRNA levels increased in mice treated with holo transferrin, suggesting that Atoh8 responds to changes in plasma iron. ATOH8 is therefore a novel transcriptional regulator of HAMP, which is responsive to changes in plasma iron and erythroid activity and could explain how changes in erythroid activity lead to regulation of HAMP.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Eritropoyesis/genética , Eritropoyesis/fisiología , Hepcidinas/genética , Proteínas Smad/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Femenino , Células HEK293 , Hepcidinas/biosíntesis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Proteínas Smad/genética , Transcripción Genética
8.
J Nutr ; 144(12): 1896-902, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25342699

RESUMEN

BACKGROUND: Iron (Fe) deficiency anemia remains the largest nutritional deficiency disorder worldwide. How the gut acquires iron from nano Fe(III), especially at the apical surface, is incompletely understood. OBJECTIVE: We developed a novel Fe supplement consisting of nanoparticulate tartrate-modified Fe(III) poly oxo-hydroxide [here termed nano Fe(III)], which mimics the Fe oxide core of ferritin and effectively treats iron deficiency anemia in rats. METHODS: We determined transfer to the systemic circulation of nano Fe(III) in iron-deficient and iron-sufficient outbread Swiss mouse strain (CD1) mice with use of (59)Fe-labeled material. Iron deficiency was induced before starting the Fe-supplementation period through reduction of Fe concentrations in the rodent diet. A control group of iron-sufficient mice were fed a diet with adequate Fe concentrations throughout the study. Furthermore, we conducted a hemoglobin repletion study in which iron-deficient CD1 mice were fed for 7 d a diet supplemented with ferrous sulfate (FeSO4) or nano Fe(III). Finally, we further probed the mechanism of cellular acquisition of nano Fe(III) by assessing ferritin formation, as a measure of Fe uptake and utilization, in HuTu 80 duodenal cancer cells with targeted inhibition of divalent metal transporter 1 (DMT1) and duodenal cytochrome b (DCYTB) before exposure to the supplemented iron sources. Differences in gene expression were assessed by quantitative polymerase chain reaction. RESULTS: Absorption (means ± SEMs) of nano Fe(III) was significantly increased in iron-deficient mice (58 ± 19%) compared to iron-sufficient mice (18 ± 17%) (P = 0.0001). Supplementation of the diet with nano Fe(III) or FeSO4 significantly increased hemoglobin concentrations in iron-deficient mice (170 ± 20 g/L, P = 0.01 and 180 ± 20 g/L, P = 0.002, respectively). Hepatic hepcidin mRNA expression reflected the nonheme-iron concentrations of the liver and was also comparable for both nano Fe(III)- and FeSO4-supplemented groups, as were iron concentrations in the spleen and duodenum. Silencing of the solute carrier family 11 (proton-coupled divalent metal ion transporter), member 2 (Slc11a2) gene (DMT1) significantly inhibited ferritin formation from FeSO4 (P = 0.005) but had no effect on uptake and utilization of nano Fe(III). Inhibiting DCYTB with an antibody also had no effect on uptake and utilization of nano Fe(III) but significantly inhibited ferritin formation from ferric nitrilotriacetate chelate (Fe-NTA) (P = 0.04). Similarly, cellular ferritin formation from nano Fe(III) was unaffected by the Fe(II) chelator ferrozine, which significantly inhibited uptake and utilization from FeSO4 (P = 0.009) and Fe-NTA (P = 0.005). CONCLUSIONS: Our data strongly support direct nano Fe(III) uptake by enterocytes as an efficient mechanism of dietary iron acquisition, which may complement the known Fe(II)/DMT1 uptake pathway.


Asunto(s)
Duodeno/citología , Duodeno/efectos de los fármacos , Ferritinas/administración & dosificación , Nanopartículas/química , Anemia Ferropénica/tratamiento farmacológico , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Suplementos Dietéticos , Duodeno/metabolismo , Enterocitos/metabolismo , Compuestos Férricos/metabolismo , Ferritinas/farmacocinética , Compuestos Ferrosos/administración & dosificación , Compuestos Ferrosos/farmacocinética , Hemoglobinas , Hepcidinas/genética , Hepcidinas/metabolismo , Hierro de la Dieta/administración & dosificación , Hierro de la Dieta/farmacocinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ácido Nitrilotriacético/análogos & derivados , Ácido Nitrilotriacético/metabolismo , Bazo/efectos de los fármacos , Bazo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Nanomedicine ; 10(7): 1529-38, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24394211

RESUMEN

The 2-5 nm Fe(III) oxo-hydroxide core of ferritin is less ordered and readily bioavailable compared to its pure synthetic analogue, ferrihydrite. We report the facile synthesis of tartrate-modified, nano-disperse ferrihydrite of small primary particle size, but with enlarged or strained lattice structure (~2.7Å for the main Bragg peak versus 2.6Å for synthetic ferrihydrite). Analysis indicated that co-precipitation conditions can be achieved for tartrate inclusion into the developing ferrihydrite particles, retarding both growth and crystallization and favoring stabilization of the cross-linked polymeric structure. In murine models, gastrointestinal uptake was independent of luminal Fe(III) reduction to Fe(II) and, yet, absorption was equivalent to that of ferrous sulphate, efficiently correcting the induced anemia. This process may model dietary Fe(III) absorption and potentially provide a side effect-free form of cheap supplemental iron. From the clinical editor: Small size tartrate-modified, nano-disperse ferrihydrite was used for efficient gastrointestinal delivery of soluble Fe(III) without the risk for free radical generation in murine models. This method may provide a potentially side effect-free form iron supplementation.


Asunto(s)
Anemia/tratamiento farmacológico , Ferritinas/uso terapéutico , Hierro/metabolismo , Nanopartículas , Animales , Ferritinas/administración & dosificación , Masculino , Ratones , Microscopía Electrónica de Transmisión de Rastreo , Oxidación-Reducción
10.
Nutrients ; 16(2)2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38257099

RESUMEN

Iron deficiency anemia (IDA) is a global nutritional disorder affecting large population groups in varying magnitudes in different countries [...].


Asunto(s)
Anemia Ferropénica , Trastornos Nutricionales , Humanos , Estado Nutricional , Anemia Ferropénica/epidemiología , Anemia Ferropénica/prevención & control , Hierro
11.
J Biol Chem ; 287(6): 4099-106, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22144676

RESUMEN

The BMP/SMAD4 pathway has major effects on liver hepcidin levels. Bone morphogenetic protein-binding endothelial cell precursor-derived regulator (Bmper), a known regulator of BMP signaling, was found to be overexpressed at the mRNA and protein levels in liver of genetically hypotransferrinemic mice (Trf(hpx/hpx)). Soluble BMPER peptide inhibited BMP2- and BMP6-dependent hepcidin promoter activity in both HepG2 and HuH7 cells. These effects correlated with reduced cellular levels of pSMAD1/5/8. Addition of BMPER peptide to primary human hepatocytes abolished the BMP2-dependent increase in hepcidin mRNA, whereas injection of Bmper peptide into mice resulted in reduced liver hepcidin and increased serum iron levels. Thus Bmper may play an important role in suppressing hepcidin production in hypotransferrinemic mice.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/sangre , Proteínas Portadoras/metabolismo , Hierro/sangre , Hígado/metabolismo , Transferrina/metabolismo , Regulación hacia Arriba , Animales , Péptidos Catiónicos Antimicrobianos/genética , Proteína Morfogenética Ósea 2/antagonistas & inhibidores , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proteínas Portadoras/genética , Células Hep G2 , Hepcidinas , Humanos , Ratones , Ratones Transgénicos , Péptidos/farmacología , Proteínas Smad/genética , Proteínas Smad/metabolismo , Transferrina/genética
12.
Bioconjug Chem ; 24(9): 1527-32, 2013 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-23888876

RESUMEN

Hepcidin is a peptide hormone that regulates homeostasis in iron metabolism. It binds to the sole known cellular iron exporter ferroportin (Fpn), triggers its internalization, and thereby modulates the efflux of iron from cells. This functional property has been adopted in this study to assess the bioactivity and potency of a range of novel fluorescent hepcidin analogues. Hepcidin was selectively labeled with 6-carboxyfluorescein (CF) and 6-carboxytetramethylrhodamine (TMR) using Fmoc solid phase peptide chemistry. Internalization of Fpn by hepcidin was assessed by high-content microscopic analysis. Both K18- and M21K-labeled hepcidin with TMR and CF exhibited measurable potency when tested in cultured MDCK and T47D cells expressing human ferroportin. The bioactivity of the labeled hepcidin varies with the type of fluorophore and site of attachment of the fluorophores on the hepcidin molecule.


Asunto(s)
Hepcidinas/química , Hepcidinas/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Línea Celular , Perros , Fluoresceínas/química , Colorantes Fluorescentes/química , Hepcidinas/síntesis química , Humanos , Modelos Moleculares , Unión Proteica , Pliegue de Proteína , Rodaminas/química
13.
Eur J Haematol ; 90(1): 10-5, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23078160

RESUMEN

Iron is an important mineral element required for diverse life processes. Its metabolism is almost synonymous to erythrocyte maintenance, erythropoiesis and erythrophagocytosis. Consequently, exercise exertion impacts significantly on red cell haematology. Here, the interactions between exercise and erythropoiesis are explored. Hepcidin, the peptide hormone that regulates systemic iron metabolism, decreases in response to erythropoiesis by facilitating increased iron efflux from ferroportin into circulation. However, during exercise, there is an alarming increase in the expression of hepcidin resulting in a negative iron balance in athletes. In this review, the confounding cause and effect scenarios of exercise, athlete training and haematology and hepcidin interactions are discussed.


Asunto(s)
Atletas , Hierro/metabolismo , Anemia/etiología , Péptidos Catiónicos Antimicrobianos/metabolismo , Eritropoyesis/fisiología , Eritropoyetina/metabolismo , Ejercicio Físico/fisiología , Hemólisis , Hepcidinas , Humanos , Hipoxia/etiología
14.
Eur J Nutr ; 52(1): 135-43, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22241739

RESUMEN

BACKGROUND: Hepcidin, the liver-secreted iron regulatory peptide, maintains systemic iron homeostasis in response to several stimuli including dietary iron levels and body iron status. In addition, iron metabolism is controlled by several local regulatory mechanisms including IRP and Hif-2α activities independently of hepcidin. However, the roles of these mechanisms and their interaction particularly in hepcidin-deficient individuals are not yet fully understood. We, therefore, aimed to explore whether Hamp disruption affects iron homeostatic responses to dietary iron deficiency. METHODS: Hepcidin1 knockout (Hamp (-/-)) mice and heterozygous littermates were fed with control or iron-deficient diet for 2 weeks. The expression of iron-related genes and proteins were determined by quantitative PCR and Western blot, respectively. RESULTS: Two-week iron-deficient diet feeding in Hamp (-/-) mice did not alter serum iron but significantly reduced liver non-heme iron levels. This was also associated with increased ferroportin protein expression in the duodenum and spleen, whereas decreased expression was found in the liver. In addition, significant inductive effects of iron-deficient diet on Dcytb and DMT1 mRNA expression in the duodenum were noted with more pronounced effects in Hamp (-/-) mice compared with controls. CONCLUSIONS: Hamp (-/-) mice exhibited a more dramatic increase in the expression of iron transport machinery, which may be responsible for the unaltered serum iron levels upon iron-deficient diet feeding in these mice. Despite the lack of hepcidin, Hamp (-/-) mice can maintain a degree of iron homeostasis in response to altered dietary iron through several hepcidin-independent mechanisms.


Asunto(s)
Deficiencias de Hierro , Hierro de la Dieta/administración & dosificación , Hierro de la Dieta/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Western Blotting , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Regulación de la Expresión Génica , Hepcidinas , Homeostasis/efectos de los fármacos , Hierro/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Bazo/efectos de los fármacos , Bazo/metabolismo
15.
Biochem Biophys Rep ; 35: 101521, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37560439

RESUMEN

Liver as iron storage organ is particularly susceptible to oxidative stress-induced injury from excess iron. Thus, antioxidant therapies are often used to reverse oxidative damage and protect cells and tissues. This study investigated the protective effects of phenolic acids; ferulic acid (FA) and its metabolite, ferulic acid 4-O-sulfate disodium salt (FAS) against oxidative stress under iron overload conditions in mouse and HepG2 cells. Cells were exposed to FA or FAS and then treated with iron-induced oxidative stress complex of 50 µmol/L FAC and 20 µmol/L of 8-hydroxyquinoline 8HQ (8HQ-FAC). Iron dextran was injected intraperitoneally on alternate days for 10 days to induce the iron overload condition in BALB/c mice. The study revealed that the phenolic acids were protective against ROS production, lipid peroxidation and antioxidant depletion in HepG2 cells and liver tissues of BALB/c mice during iron-induced oxidative stress. The protective function of phenolic acids was achieved by the transcriptional activation of nuclear factor erythroid-2-related factor 2 (Nrf2) to regulate antioxidant genes. In conclusion, the study provides evidence that FA has the potential as a therapeutic agent against iron-related diseases such as T2D.

16.
Food Funct ; 14(8): 3673-3685, 2023 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-36970974

RESUMEN

The detrimental effects of high concentrations of colonic iron have been linked to intestinal inflammation and microbial dysbiosis. Exploiting chelation against this luminal pool of iron may restore intestinal health and have beneficial impacts on microbial communities. This study aimed to explore whether lignin, a heterogenous polyphenolic dietary component, has iron-binding affinity and can sequester iron within the intestine and thus, potentially modulate the microbiome. Within in vitro cell-culture models, the treatment of RKO and Caco-2 cells with lignin almost abolished intracellular iron import (96% and 99% reduction of iron acquisition respectively) with corresponding changes in iron metabolism proteins (ferritin and transferrin receptor-1) and reductions in the labile-iron pool. In a Fe-59 supplemented murine model, intestinal iron absorption was significantly inhibited by 30% when lignin was co-administered compared to the control group with the residual iron lost in the faeces. The supplementation of lignin into a microbial bioreactor colonic model increased the solubilisation and bio-accessibility of iron present by 4.5-fold despite lignin-iron chelation previously restricting intracellular iron absorption in vitro and in vivo. The supplementation of lignin in the model increased the relative abundance of Bacteroides whilst levels of Proteobacteria decreased which could be attributed to the changes in iron bio-accessibility due to iron chelation. In summary, we demonstrate that lignin is an effective luminal iron chelator. Iron chelation leads to the limitation of intracellular iron import whilst, despite increasing iron solubility, favouring the growth of beneficial bacteria.


Asunto(s)
Microbioma Gastrointestinal , Hierro , Humanos , Animales , Ratones , Hierro/metabolismo , Lignina , Radioisótopos de Hierro/farmacología , Células CACO-2 , Intestinos/microbiología , Quelantes del Hierro/farmacología
17.
Proc Nutr Soc ; : 1-7, 2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37395294

RESUMEN

Cereal products provide 50 % of iron and 30 % of zinc in the UK diet. However, despite having high content, the bioavailability of minerals from cereals is low. This review discusses strategies to increase mineral bioavailability from cereal-based foods. Iron and zinc are localised to specific tissue structures within cereals; however, the cell walls of these structures are resistant to digestion in the human gastrointestinal tract and therefore the bioaccessibility of these essential minerals from foods for absorption in the intestine is limited. In addition, minerals are stored in cereals bound to phytate, which is the main dietary inhibitor of mineral absorption. Recent research has focused on ways to enhance mineral bioavailability from cereals. Current strategies include disruption of plant cell walls to increase mineral release (bioaccessibility) during digestion; increasing the mineral:phytate ratio either by increasing the mineral content through conventional breeding and/or agronomic biofortification, or by reducing phytate levels; and genetic biofortification to increase the mineral content in the starchy endosperm, which is used to produce white wheat flour. While much of this work is at an early stage, there is potential for these strategies to lead to the development of cereal-based foods with enhanced nutritional qualities that could address the low mineral status in the UK and globally.

18.
Nutrients ; 15(12)2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37375636

RESUMEN

Increasing numbers of individuals follow plant-based diets. This has sparked interest in the nutritional evaluation of the meat substitute sector. Nutritional understanding of these products is vital as plant-based eating becomes more common. For example, animal products are rich sources of iron and zinc, and plant-based foods could be inadequate in these minerals. The main aim was to analyse the mineral composition and absorption from a range of plant-based meat-free burgers and compare them to a typical beef burger. Total and bioaccessible mineral contents of plant-based burgers and a beef burger were determined using microwave digestion and in vitro simulated gastrointestinal digestion, respectively. Mineral bioavailability was analysed by in vitro simulated gastrointestinal digestion of foods, followed by exposure of Caco-2 cells to the sample digests and assessment of mineral uptake. Mineral quantification for all samples was achieved using inductively coupled ICP-optical emission spectrometry (ICP-OES). The content of minerals varied significantly amongst the burgers. Significantly greater quantities of Fe and Zn were found in the beef burger compared to most meat substitutes. Bioaccessible Fe was significantly higher in the beef compared to most of the plant-based meat alternatives; however, bioavailable Fe of most plant-based burgers was comparable to beef (p > 0.05). Similarly, bioaccessible Zn was significantly (p < 0.001) higher from the beef burger. Moreover, beef was superior regarding bioavailable Zn (p ≤ 0.05-0.0001), with only the mycoprotein burger displaying comparable Zn bioavailability (p > 0.05). Beef is an excellent source of bioaccessible Fe and Zn compared to most plant-based substitutes; however, these plant-based substitutes were superior sources of Ca, Cu, Mg and Mn. The quantity of bioaccessible and absorbable Fe varies dramatically among the meat alternatives. Plant-based burgers have the potential to provide adequate quantities of iron and zinc to those consuming such burgers as part of a varied diet. Thus, guiding consumer choices will depend on the variety of the vegetable constituents and their iron nutritional quality in different burgers.


Asunto(s)
Productos de la Carne , Minerales , Humanos , Animales , Bovinos , Células CACO-2 , Hierro/análisis , Productos de la Carne/análisis , Zinc , Plantas
19.
Blood Cells Mol Dis ; 49(2): 85-91, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22609087

RESUMEN

Hepcidin, an iron regulatory peptide, plays a central role in the maintenance of systemic iron homeostasis by inducing the internalization and degradation of the iron exporter, ferroportin. Hepcidin expression in the liver is regulated in response to several stimuli including iron status, erythropoietic activity, hypoxia and inflammation. Hepcidin expression has been shown to be reduced in phenylhydrazine-treated mice, a mouse model of acute hemolysis. In this mouse model, hepcidin suppression was associated with increased expression of molecules involved in iron transport and recycling. The present study aims to explore whether the response to phenylhydrazine treatment is affected by hepcidin deficiency and/or the subsequently altered iron metabolism. Hepcidin1 knockout (Hamp(-/-)) and wild type mice were treated with phenylhydrazine or saline and parameters of iron homeostasis were determined 3 days after the treatment. In wild type mice, phenylhydrazine administration resulted in significantly reduced serum iron, increased tissue non-heme iron levels and suppressed hepcidin expression. The treatment was also associated with increases in membrane ferroportin protein levels and spleen heme oxygenase 1 mRNA expression. In addition, trends toward increased mRNA expression of duodenal iron transporters were also observed. In contrast, serum iron and tissue non-heme iron levels in Hamp(-/-) mice were unaffected by the treatment. Moreover, the effects of phenylhydrazine on the expression of ferroportin and duodenal iron transporters were not observed in Hamp(-/-) mice. Interestingly, mRNA levels of molecules involved in splenic heme uptake and degradation were significantly induced by Hamp disruption. In summary, our study demonstrates that the response to phenylhydrazine-induced hemolysis differs between wild type and Hamp(-/-) mice. This observation may be caused by the absence of hepcidin per se or the altered iron homeostasis induced by the lack of hepcidin in these mice.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Eritrocitos/citología , Hierro/metabolismo , Fenilhidrazinas/farmacología , Animales , Péptidos Catiónicos Antimicrobianos/genética , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Eritrocitos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hemo/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Hemólisis , Hepcidinas , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , Bazo/efectos de los fármacos , Bazo/metabolismo
20.
J Nutr ; 142(11): 1929-34, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22990466

RESUMEN

Duodenal cytochrome b (Dcytb, Cybrd1) is a ferric reductase localized in the duodenum that is highly upregulated in circumstances of increased iron absorption. To address the contribution of Dcytb to total duodenal ferric reductase activity as well as its wider role in iron metabolism, we first measured duodenal ferric reductase activity in wild-type (WT) and Dcytb knockout (Dcytb(-/-)) mice under 3 conditions known to induce gut ferric reductase: dietary iron deficiency, hypoxia, and pregnancy. Dcytb(-/-) and WT mice were randomly assigned to control (iron deficiency experiment, 48 mg/kg dietary iron; hypoxia experiment, normal atmospheric pressure; pregnancy experiment, nonpregnant animals) or treatment (iron deficiency experiment, 2-3 mg/kg dietary iron; hypoxia experiment, 53.3 kPa pressure; pregnancy experiment, d 20 of pregnancy) groups and duodenal reductase activity measured. We found no induction of ferric reductase activity in Dcytb(-/-) mice under any of these conditions, indicating there are no other inducible ferric reductases present in the duodenum. To test whether Dcytb was required for iron absorption in conditions with increased erythropoietic demand, we also measured tissue nonheme iron levels and hematological indices in WT and Dcytb(-/-) mice exposed to hypoxia. There was no evidence of gross alterations in iron absorption, hemoglobin, or total liver nonheme iron in Dcytb(-/-) mice exposed to hypoxia compared with WT mice. However, spleen nonheme iron was significantly less (6.7 ± 1.0 vs. 12.7 ± 0.9 nmol · mg tissue(-1); P < 0.01, n = 7-8) in hypoxic Dcytb(-/-) compared with hypoxic WT mice and there was evidence of impaired reticulocyte hemoglobinization with a lower reticulocyte mean corpuscular hemoglobin (276 ± 1 vs. 283 ± 2 g · L(-1); P < 0.05, n = 7-8) in normoxic Dcytb(-/-) compared with normoxic WT mice. We therefore conclude that DCYTB is the primary iron-regulated duodenal ferric reductase in the gut and that Dcytb is necessary for optimal iron metabolism.


Asunto(s)
Grupo Citocromo b/metabolismo , Duodeno/enzimología , Eritropoyesis/fisiología , Hipoxia/metabolismo , Hierro/metabolismo , Oxidorreductasas/metabolismo , Bazo/metabolismo , Anemia Ferropénica/metabolismo , Alimentación Animal , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Grupo Citocromo b/genética , Dieta , Eritropoyesis/efectos de los fármacos , Femenino , Regulación Enzimológica de la Expresión Génica/fisiología , Hierro/farmacología , Masculino , Ratones , Ratones Noqueados , Oxidorreductasas/genética , Oxígeno/farmacología , Embarazo , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda