Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
PLoS Pathog ; 15(9): e1007651, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31513674

RESUMEN

Bacterial type IV secretion systems (T4SS) are a highly diversified but evolutionarily related family of macromolecule transporters that can secrete proteins and DNA into the extracellular medium or into target cells. It was recently shown that a subtype of T4SS harboured by the plant pathogen Xanthomonas citri transfers toxins into target cells. Here, we show that a similar T4SS from the multi-drug-resistant opportunistic pathogen Stenotrophomonas maltophilia is proficient in killing competitor bacterial species. T4SS-dependent duelling between S. maltophilia and X. citri was observed by time-lapse fluorescence microscopy. A bioinformatic search of the S. maltophilia K279a genome for proteins containing a C-terminal domain conserved in X. citri T4SS effectors (XVIPCD) identified twelve putative effectors and their cognate immunity proteins. We selected a putative S. maltophilia effector with unknown function (Smlt3024) for further characterization and confirmed that it is indeed secreted in a T4SS-dependent manner. Expression of Smlt3024 in the periplasm of E. coli or its contact-dependent delivery via T4SS into E. coli by X. citri resulted in reduced growth rates, which could be counteracted by expression of its cognate inhibitor Smlt3025 in the target cell. Furthermore, expression of the VirD4 coupling protein of X. citri can restore the function of S. maltophilia ΔvirD4, demonstrating that effectors from one species can be recognized for transfer by T4SSs from another species. Interestingly, Smlt3024 is homologous to the N-terminal domain of large Ca2+-binding RTX proteins and the crystal structure of Smlt3025 revealed a topology similar to the iron-regulated protein FrpD from Neisseria meningitidis which has been shown to interact with the RTX protein FrpC. This work expands our current knowledge about the function of bacteria-killing T4SSs and increases the panel of effectors known to be involved in T4SS-mediated interbacterial competition, which possibly contribute to the establishment of S. maltophilia in clinical and environmental settings.


Asunto(s)
Proteínas Bacterianas/fisiología , Stenotrophomonas maltophilia/fisiología , Stenotrophomonas maltophilia/patogenicidad , Sistemas de Secreción Tipo IV/fisiología , Secuencia de Aminoácidos , Antibiosis/genética , Antibiosis/fisiología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Secuencia Conservada , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Genes Bacterianos , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Proteínas Reguladoras del Hierro/química , Proteínas Reguladoras del Hierro/genética , Proteínas Reguladoras del Hierro/fisiología , Modelos Moleculares , Infecciones Oportunistas/microbiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad de la Especie , Stenotrophomonas maltophilia/genética , Sistemas de Secreción Tipo IV/química , Sistemas de Secreción Tipo IV/genética , Xanthomonas/genética , Xanthomonas/crecimiento & desarrollo
3.
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
4.
J Biol Chem ; 292(31): 12725-12726, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28615455

RESUMEN

In this tenth Thematic Series in Metals in Biology, six Minireviews deal with aspects of iron metabolism. A number of important proteins control iron homeostasis, including hepcidin and ferroportin, in various cells. Other aspects of iron dealt with here include biogenesis of iron-sulfur proteins and chaperones that deliver iron cofactors in cells. Additionally, an iron-regulated metastasis suppressor interacts with the epidermal growth factor receptor and mediates its downstream signaling activity.


Asunto(s)
Homeostasis , Hierro/fisiología , Animales , Transporte Biológico , Humanos , Proteínas Reguladoras del Hierro/fisiología , Proteínas Hierro-Azufre/fisiología
5.
Nefrologia ; 37(6): 587-591, 2017.
Artículo en Inglés, Español | MEDLINE | ID: mdl-28610806

RESUMEN

Chronic kidney disease and anaemia are common in heart failure (HF) and are associated with a worse prognosis in these patients. Iron deficiency is also common in patients with HF and increases the risk of morbidity and mortality, regardless of the presence or absence of anaemia. While the treatment of anaemia with erythropoiesis-stimulating agents in patients with HF have failed to show a benefit in terms of morbidity and mortality, treatment with IV iron in patients with HF and reduced ejection fraction and iron deficiency is associated with clinical improvement. In a posthoc analysis of a clinical trial, iron therapy improved kidney function in patients with HF and iron deficiency. In fact, the European Society of Cardiology's recent clinical guidelines on HF suggest that in symptomatic patients with reduced ejection fraction and iron deficiency, treatment with IV ferric carboxymaltose should be considered to improve symptoms, the ability to exercise and quality of life. Iron plays a key role in oxygen storage (myoglobin) and in energy metabolism, and there are pathophysiological bases that explain the beneficial effect of IV iron therapy in patients with HF. All these aspects are reviewed in this article.


Asunto(s)
Compuestos Férricos/uso terapéutico , Insuficiencia Cardíaca/etiología , Deficiencias de Hierro , Maltosa/análogos & derivados , Anemia Ferropénica/tratamiento farmacológico , Anemia Ferropénica/etiología , Anemia Ferropénica/fisiopatología , Animales , Modelos Animales de Enfermedad , Compuestos Férricos/administración & dosificación , Tasa de Filtración Glomerular , Insuficiencia Cardíaca/tratamiento farmacológico , Hematínicos/uso terapéutico , Humanos , Infusiones Intravenosas , Proteínas Reguladoras del Hierro/deficiencia , Proteínas Reguladoras del Hierro/fisiología , Maltosa/administración & dosificación , Maltosa/uso terapéutico , Metaanálisis como Asunto , Ratones , Estudios Multicéntricos como Asunto , Guías de Práctica Clínica como Asunto , Ensayos Clínicos Controlados Aleatorios como Asunto , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/fisiopatología , Volumen Sistólico/efectos de los fármacos
6.
Eur Heart J ; 38(5): 362-372, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27545647

RESUMEN

Aims: Iron deficiency (ID) is associated with adverse outcomes in heart failure (HF) but the underlying mechanisms are incompletely understood. Intracellular iron availability is secured by two mRNA-binding iron-regulatory proteins (IRPs), IRP1 and IRP2. We generated mice with a cardiomyocyte-targeted deletion of Irp1 and Irp2 to explore the functional implications of ID in the heart independent of systemic ID and anaemia. Methods and results: Iron content in cardiomyocytes was reduced in Irp-targeted mice. The animals were not anaemic and did not show a phenotype under baseline conditions. Irp-targeted mice, however, were unable to increase left ventricular (LV) systolic function in response to an acute dobutamine challenge. After myocardial infarction, Irp-targeted mice developed more severe LV dysfunction with increased HF mortality. Mechanistically, the activity of the iron-sulphur cluster-containing complex I of the mitochondrial electron transport chain was reduced in left ventricles from Irp-targeted mice. As demonstrated by extracellular flux analysis in vitro, mitochondrial respiration was preserved at baseline but failed to increase in response to dobutamine in Irp-targeted cardiomyocytes. As shown by 31P-magnetic resonance spectroscopy in vivo, LV phosphocreatine/ATP ratio declined during dobutamine stress in Irp-targeted mice but remained stable in control mice. Intravenous injection of ferric carboxymaltose replenished cardiac iron stores, restored mitochondrial respiratory capacity and inotropic reserve, and attenuated adverse remodelling after myocardial infarction in Irp-targeted mice but not in control mice. As shown by electrophoretic mobility shift assays, IRP activity was significantly reduced in LV tissue samples from patients with advanced HF and reduced LV tissue iron content. Conclusions: ID in cardiomyocytes impairs mitochondrial respiration and adaptation to acute and chronic increases in workload. Iron supplementation restores cardiac energy reserve and function in iron-deficient hearts.


Asunto(s)
Insuficiencia Cardíaca/prevención & control , Deficiencias de Hierro , Proteínas Reguladoras del Hierro/fisiología , Miocitos Cardíacos/metabolismo , Animales , Cardiotónicos/farmacología , Dopamina/farmacología , Compuestos Férricos/farmacología , Ferritinas/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Hierro/metabolismo , Proteínas Reguladoras del Hierro/deficiencia , Angiografía por Resonancia Magnética , Maltosa/análogos & derivados , Maltosa/farmacología , Mitocondrias Cardíacas/fisiología , Fenotipo , ARN Mensajero/fisiología , Función Ventricular Izquierda/fisiología
7.
J Biol Chem ; 290(43): 25876-90, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26342079

RESUMEN

Biogenesis of the iron-sulfur (Fe-S) cluster is an indispensable process in living cells. In mammalian mitochondria, the initial step of the Fe-S cluster assembly process is assisted by the NFS1-ISD11 complex, which delivers sulfur to scaffold protein ISCU during Fe-S cluster synthesis. Although ISD11 is an essential protein, its cellular role in Fe-S cluster biogenesis is still not defined. Our study maps the important ISD11 amino acid residues belonging to putative helix 1 (Phe-40), helix 3 (Leu-63, Arg-68, Gln-69, Ile-72, Tyr-76), and C-terminal segment (Leu-81, Glu-84) are critical for in vivo Fe-S cluster biogenesis. Importantly, mutation of these conserved ISD11 residues into alanine leads to its compromised interaction with NFS1, resulting in reduced stability and enhanced aggregation of NFS1 in the mitochondria. Due to altered interaction with ISD11 mutants, the levels of NFS1 and Isu1 were significantly depleted, which affects Fe-S cluster biosynthesis, leading to reduced electron transport chain complex (ETC) activity and mitochondrial respiration. In humans, a clinically relevant ISD11 mutation (R68L) has been associated in the development of a mitochondrial genetic disorder, COXPD19. Our findings highlight that the ISD11 R68A/R68L mutation display reduced affinity to form a stable subcomplex with NFS1, and thereby fails to prevent NFS1 aggregation resulting in impairment of the Fe-S cluster biogenesis. The prime affected machinery is the ETC complex, which showed compromised redox properties, causing diminished mitochondrial respiration. Furthermore, the R68L ISD11 mutant displayed accumulation of mitochondrial iron and reactive oxygen species, leading to mitochondrial dysfunction, which correlates with the phenotype observed in COXPD19 patients.


Asunto(s)
Liasas de Carbono-Azufre/fisiología , Proteínas Reguladoras del Hierro/fisiología , Enfermedades Mitocondriales/fisiopatología , Secuencia de Aminoácidos , Liasas de Carbono-Azufre/química , Liasas de Carbono-Azufre/metabolismo , Progresión de la Enfermedad , Células HeLa , Humanos , Proteínas Reguladoras del Hierro/química , Proteínas Reguladoras del Hierro/metabolismo , Proteínas Hierro-Azufre/metabolismo , Enfermedades Mitocondriales/metabolismo , Datos de Secuencia Molecular , Unión Proteica , Estabilidad Proteica , Homología de Secuencia de Aminoácido
8.
J Biol Chem ; 290(12): 7634-46, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25572399

RESUMEN

Adequate availability of iron is important for cellular energy metabolism. Catecholamines such as epinephrine and norepinephrine promote energy expenditure to adapt to conditions that arose due to stress. To restore the energy balance, epinephrine/norepinephrine-exposed cells may face higher iron demand. So far, no direct role of epinephrine/norepinephrine in cellular iron homeostasis has been reported. Here we show that epinephrine/norepinephrine regulates iron homeostasis components such as transferrin receptor-1 and ferritin-H in hepatic and skeletal muscle cells by promoting the binding of iron regulatory proteins to iron-responsive elements present in the UTRs of transferrin receptor-1 and ferritin-H transcripts. Increased transferrin receptor-1, decreased ferritin-H, and increased iron-responsive element-iron regulatory protein interaction are also observed in liver and muscle tissues of epinephrine/norepinephrine-injected mice. We demonstrate the role of epinephrine/norepinephrine-induced generation of reactive oxygen species in converting cytosolic aconitase (ACO1) into iron regulatory protein-1 to bind iron-responsive elements present in UTRs of transferrin receptor-1 and ferritin-H. Our study further reveals that mitochondrial iron content and mitochondrial aconitase (ACO2) activity are elevated by epinephrine/norepinephrine that are blocked by the antioxidant N-acetyl cysteine and iron regulatory protein-1 siRNA, suggesting involvement of reactive oxygen species and iron regulatory protein-1 in this mechanism. This study reveals epinephrine and norepinephrine as novel regulators of cellular iron homeostasis.


Asunto(s)
Catecolaminas/fisiología , Metabolismo Energético , Homeostasis , Proteínas Reguladoras del Hierro/fisiología , Hierro/metabolismo , Procesamiento Postranscripcional del ARN , Animales , Línea Celular Tumoral , Cartilla de ADN , Humanos , Hígado/citología , Hígado/metabolismo , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo
9.
Neurobiol Aging ; 36(2): 1183-93, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25467637

RESUMEN

Elevated iron levels and increased expression of divalent metal transporter 1 (DMT1) in the substantia nigra of Parkinson's disease (PD) have been reported. Nedd4 family-interacting protein 1 (Ndfip1), an adaptor protein for the Nedd4 family of ubiquitin ligases, played an essential role in regulating DMT1 and iron homeostasis in human cortical neurons. In this study, we demonstrated that the expression of Ndfip1 decreased in 6-hydroxydopamine (6-OHDA)-induced PD rats and 6-OHDA-treated MES23.5 dopaminergic cells. Further study showed that the decrease of Ndfip1 occurred earlier than the increase of DMT1 with iron-responsive element (DMT1 + IRE) in 6-OHDA-treated MES23.5 cells, indicating that the decrease of Ndfip1 might be involved in the increase of DMT1 + IRE. In addition, we demonstrated that overexpression of Ndfip1 caused DMT1 + IRE downregulation, resulting in the decreased iron influx and iron-induced neurotoxicity. Although Ndfip1 knockdown led to decreased protein levels of DMT1 + IRE, partially aggravated iron-induced neurotoxicity. Further experiments showed that 6-OHDA-induced decrease in Ndfip1 levels might be related to proteasomal and lysosomal activations and oxidative stress caused by 6-OHDA. These data suggest that decreased Ndfip1 expression might contribute to the pathogenesis of 6-OHDA-induced iron accumulation and Ndfip1 could attenuate 6-OHDA-induced iron accumulation via regulating the degradation of DMT1.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/fisiología , Hierro/metabolismo , Proteínas de la Membrana/fisiología , Oxidopamina/efectos adversos , Enfermedad de Parkinson/genética , Proteolisis , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Neuronas Dopaminérgicas/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular , Hierro/toxicidad , Proteínas Reguladoras del Hierro/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Enfermedad de Parkinson/metabolismo , Ratas , Sustancia Negra/metabolismo
10.
Insect Sci ; 20(5): 601-19, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23956079

RESUMEN

Secreted ferritin is the major iron storage and transport protein in insects. Here, we characterize the message and protein expression profiles of yellow fever mosquito (Aedes aegypti) ferritin heavy chain homologue (HCH) and light chain homologue (LCH) subunits in response to iron and bacterial challenge. In vivo experiments demonstrated tissue-specific regulation of HCH and LCH expression over time post-blood meal (PBM). Transcriptional regulation of HCH and LCH was treatment specific, with differences in regulation for naïve versus mosquitoes challenged with heat-killed bacteria (HKB). Translational regulation by iron regulatory protein (IRP) binding activity for the iron-responsive element (IRE) was tissue-specific and time-dependent PBM. However, mosquitoes challenged with HKB showed little change in IRP/IRE binding activity compared to naïve animals. The changes in ferritin regulation and expression in vivo were confirmed with in vitro studies. We challenged mosquitoes with HKB followed by a blood meal to determine the effects on ferritin expression, and demonstrate a synergistic, time-dependent regulation of expression for HCH and LCH.


Asunto(s)
Aedes/metabolismo , Aedes/microbiología , Bacillus subtilis/fisiología , Escherichia coli/fisiología , Ferritinas/metabolismo , Aedes/genética , Animales , Línea Celular , Supervivencia Celular , Cuerpo Adiposo/citología , Cuerpo Adiposo/metabolismo , Femenino , Compuestos Ferrosos , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/metabolismo , Regulación de la Expresión Génica/fisiología , Calor , Hierro , Proteínas Reguladoras del Hierro/fisiología , Larva/citología , Larva/microbiología , Maleimidas , Metalocenos , Ovario/citología , Ovario/metabolismo , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Mol Med (Berl) ; 90(10): 1209-21, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22527885

RESUMEN

Siderophores are best known as small iron binding molecules that facilitate microbial iron transport. In our previous study we identified a siderophore-like molecule in mammalian cells and found that its biogenesis is evolutionarily conserved. A member of the short chain dehydrogenase family of reductases, 3-hydroxy butyrate dehydrogenase (BDH2) catalyzes a rate-limiting step in the biogenesis of the mammalian siderophore. We have shown that depletion of the mammalian siderophore by inhibiting expression of bdh2 results in abnormal accumulation of cellular iron and mitochondrial iron deficiency. These observations suggest that the mammalian siderophore is a critical regulator of cellular iron homeostasis and facilitates mitochondrial iron import. By utilizing bioinformatics, we identified an iron-responsive element (IRE; a stem-loop structure that regulates genes expression post-transcriptionally upon binding to iron regulatory proteins or IRPs) in the 3'-untranslated region of the human BDH2 (hBDH2) gene. In cultured cells as well as in patient samples we now demonstrate that the IRE confers iron-dependent regulation on hBDH2 and binds IRPs in RNA electrophoretic mobility shift assays. In addition, we show that the hBDH2 IRE associates with IRPs in cells and that abrogation of IRPs by RNAi eliminates the iron-dependent regulation of hBDH2 mRNA. The key physiologic implication is that iron-mediated post-transcriptional regulation of hBDH2 controls mitochondrial iron homeostasis in human cells. These observations provide a new and an unanticipated mechanism by which iron regulates its intracellular trafficking.


Asunto(s)
Hidroxibutirato Deshidrogenasa/genética , Proteínas Reguladoras del Hierro/metabolismo , Hierro/metabolismo , Sideróforos/metabolismo , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Transporte Biológico , Células Cultivadas , Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Hemocromatosis/metabolismo , Hemocromatosis/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hidroxibutirato Deshidrogenasa/metabolismo , Secuencias Invertidas Repetidas , Proteínas Reguladoras del Hierro/fisiología , Leontopithecus , Hígado/metabolismo , Hígado/patología , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Mitocondrias/metabolismo , Pan troglodytes , Unión Proteica , Elementos de Respuesta , Análisis de Secuencia de ADN , Sideróforos/fisiología
13.
Blood ; 118(22): e168-79, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21940823

RESUMEN

Iron regulatory proteins (IRPs) 1 and 2 are RNA-binding proteins that control cellular iron metabolism by binding to conserved RNA motifs called iron-responsive elements (IREs). The currently known IRP-binding mRNAs encode proteins involved in iron uptake, storage, and release as well as heme synthesis. To systematically define the IRE/IRP regulatory network on a transcriptome-wide scale, IRP1/IRE and IRP2/IRE messenger ribonucleoprotein complexes were immunoselected, and the mRNA composition was determined using microarrays. We identify 35 novel mRNAs that bind both IRP1 and IRP2, and we also report for the first time cellular mRNAs with exclusive specificity for IRP1 or IRP2. To further explore cellular iron metabolism at a system-wide level, we undertook proteomic analysis by pulsed stable isotope labeling by amino acids in cell culture in an iron-modulated mouse hepatic cell line and in bone marrow-derived macrophages from IRP1- and IRP2-deficient mice. This work investigates cellular iron metabolism in unprecedented depth and defines a wide network of mRNAs and proteins with iron-dependent regulation, IRP-dependent regulation, or both.


Asunto(s)
Proteína 1 Reguladora de Hierro/genética , Proteína 1 Reguladora de Hierro/metabolismo , Proteína 2 Reguladora de Hierro/genética , Proteína 2 Reguladora de Hierro/metabolismo , Proteínas Reguladoras del Hierro/fisiología , Proteoma/metabolismo , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Reguladoras del Hierro/genética , Proteínas Reguladoras del Hierro/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Proteoma/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcriptoma/fisiología
14.
Biochem J ; 434(3): 365-81, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21348856

RESUMEN

Iron is an essential but potentially hazardous biometal. Mammalian cells require sufficient amounts of iron to satisfy metabolic needs or to accomplish specialized functions. Iron is delivered to tissues by circulating transferrin, a transporter that captures iron released into the plasma mainly from intestinal enterocytes or reticuloendothelial macrophages. The binding of iron-laden transferrin to the cell-surface transferrin receptor 1 results in endocytosis and uptake of the metal cargo. Internalized iron is transported to mitochondria for the synthesis of haem or iron-sulfur clusters, which are integral parts of several metalloproteins, and excess iron is stored and detoxified in cytosolic ferritin. Iron metabolism is controlled at different levels and by diverse mechanisms. The present review summarizes basic concepts of iron transport, use and storage and focuses on the IRE (iron-responsive element)/IRP (iron-regulatory protein) system, a well known post-transcriptional regulatory circuit that not only maintains iron homoeostasis in various cell types, but also contributes to systemic iron balance.


Asunto(s)
Proteínas Reguladoras del Hierro/fisiología , Hierro/metabolismo , Animales , Transporte Biológico , Ferritinas/metabolismo , Humanos , Proteínas Reguladoras del Hierro/genética , Mitocondrias/metabolismo , Neoplasias/metabolismo , Oxidación-Reducción , ARN Mensajero/genética , ARN Mensajero/fisiología , Elementos de Respuesta , Transferrina/metabolismo
15.
Rinsho Byori ; 58(12): 1211-8, 2010 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-21348241

RESUMEN

Iron is an essential metal not only in oxygen delivery, but also in cell proliferation and drug metabolism, while it is a very toxic metal producing reactive oxygen species(ROS). In order to avoid the toxicity and shortage of iron, the level of iron is strictly regulated in the body and cells. The central player regulating the amount of iron in the body is hepcidin. Hepcidin inhibits the release of iron from enterocytes and macrophages by accelerating the degradation of ferroportin, which is an exporter of iron. The amount of cellular iron is regulated by the IRE (iron responsive element) and IRP (iron regulatory protein) system. IRP1 and 2, whose activities depend on the concentration of cellular iron, bind to IRE, and regulate the translation of iron-related genes, which have IRE in 5' or 3'-UTR to balance iron uptake and utilization. Iron is utilized for the generation of heme and the iron-sulfur (Fe-S) cluster in mitochondoria. Mutations of genes involved in heme biosynthesis, iron-sulfur (Fe-S) cluster biogenesis, or Fe-S cluster transport cause an accumulation of iron in mitochondoria, leading to the onset of inherited sideroblastic anemia. The most common inherited sideroblastic anemia is X-linked sideroblastic anemia (XLSA) caused by mutations of the erythroid-specific delta-aminolevulinate synthase gene (ALAS2), which is the first enzyme involved in heme biosynthesis in erythroid cells. However, there are still significant numbers of cases with genetically undefined, inherited sideroblastic anemia. Molecular analysis of these cases will contribute to the understanding of mitochondrial iron metabolism.


Asunto(s)
Anemia Sideroblástica/etiología , Hemo/biosíntesis , Hierro/metabolismo , 5-Aminolevulinato Sintetasa/genética , Anemia Ferropénica/etiología , Anemia Sideroblástica/genética , Péptidos Catiónicos Antimicrobianos/fisiología , Proteínas de Transporte de Catión/fisiología , Glutarredoxinas/genética , Hepcidinas , Humanos , Proteínas Reguladoras del Hierro/fisiología , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Mutación
16.
Antioxid Redox Signal ; 12(4): 445-58, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19650690

RESUMEN

The ability of iron to accept or donate electrons, coupled with the ability of oxygen to act as an electron acceptor, renders both elements essential to normal cellular biology. However, these same chemical properties allow free iron in solution to generate toxic free radicals, particularly in combination with oxygen. Thus, closely interwoven homeostatic mechanisms have evolved to regulate both iron and oxygen concentrations at the systemic and the cellular levels. Systemically, iron levels are regulated through hepcidin-mediated uptake of iron in the duodenum, whereas intracellular free-iron levels are controlled through iron-regulatory proteins (IRPs). Cardiorespiratory changes increase systemic oxygen delivery, whereas at a cellular level, many responses to altered oxygen levels are coordinated by hypoxia-inducible factor (HIF). However, the mechanisms of iron homeostasis also are regulated by oxygen availability, with alterations in both hepcidin and IRP activity. In addition, many genes involved in iron homeostasis are direct targets of HIF. Furthermore, HIF activation is modulated by intracellular iron, through regulation of hydroxylase activity, which requires iron as a cofactor. In addition, HIF-2alpha translation is controlled by IRP activity, providing another level of interdependence between iron and oxygen homeostasis.


Asunto(s)
Homeostasis/fisiología , Hierro/fisiología , Procolágeno-Prolina Dioxigenasa/fisiología , Animales , Péptidos Catiónicos Antimicrobianos/fisiología , Hepcidinas , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Hierro/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Proteínas Reguladoras del Hierro/fisiología , Ratones , Oxígeno/metabolismo , Oxígeno/fisiología , Biosíntesis de Proteínas/fisiología , Ratas
18.
Biochim Biophys Acta ; 1790(7): 702-17, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18485918

RESUMEN

Cancer contributes to 50% of deaths worldwide and new anti-tumour therapeutics with novel mechanisms of actions are essential to develop. Metabolic inhibitors represent an important class of anti-tumour agents and for many years, agents targeting the nutrient folate were developed for the treatment of cancer. This is because of the critical need of this factor for DNA synthesis. Similarly to folate, Fe is an essential cellular nutrient that is critical for DNA synthesis. However, in contrast to folate, there has been limited effort applied to specifically design and develop Fe chelators for the treatment of cancer. Recently, investigations have led to the generation of novel di-2-pyridylketone thiosemicarbazone (DpT) and 2-benzoylpyridine thiosemicarbazone (BpT) group of ligands that demonstrate marked and selective anti-tumour activity in vitro and also in vivo against a wide spectrum of tumours. Indeed, administration of these compounds to mice did not induce whole body Fe-depletion or disturbances in haematological or biochemical indices due to the very low doses required. The mechanism of action of these ligands includes alterations in expression of molecules involved in cell cycle control and metastasis suppression, as well as the generation of redox-active Fe complexes. This review examines the alterations in Fe metabolism in tumour cells and the systematic development of novel aroylhydrazone and thiosemicarbazone Fe chelators for cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Quelantes del Hierro/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias/fisiología , Péptidos Catiónicos Antimicrobianos/fisiología , Proteínas de Transporte de Catión , Ciclo Celular/efectos de los fármacos , FMN Reductasa/metabolismo , Hepcidinas , Humanos , Absorción Intestinal , Proteínas Reguladoras del Hierro/fisiología , Antígenos Específicos del Melanoma , Metástasis de la Neoplasia/fisiopatología , Proteínas de Neoplasias/fisiología , Neovascularización Patológica/fisiopatología , Receptores de Transferrina/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...