Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Front Cell Dev Biol ; 11: 1272667, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38033861

RESUMEN

Introduction: Detachment from the extracellular matrix (ECM) is the first step of the metastatic cascade. It is a regulated process involving interaction between tumor cells and tumor microenvironment (TME). Iron is a key micronutrient within the TME. Here, we explored the role of iron in the ability of ovarian cancer cells to successfully detach from the ECM. Methods: HEY and PEO1 ovarian cancer cells were grown in 3D conditions. To mimic an iron rich TME, culture media were supplemented with 100 µM Fe3+. Cell mortality was evaluated by cytofluorimetric assay. The invasive potential of tumor spheroids was performed in Matrigel and documented with images and time-lapses. Iron metabolism was assessed by analyzing the expression of CD71 and FtH1, and by quantifying the intracellular labile iron pool (LIP) through Calcein-AM cytofluorimetric assay. Ferroptosis was assessed by quantifying mitochondrial reactive oxygen species (ROS) and lipid peroxidation through MitoSOX and BODIPY-C11 cytofluorimetric assays, respectively. Ferroptosis markers GPX4 and VDAC2 were measured by Western blot. FtH1 knockdown was performed by using siRNA. Results: To generate spheroids, HEY and PEO1 cells prevent LIP accumulation by upregulating FtH1. 3D HEY moderately increases FtH1, and LIP is only slightly reduced. 3D PEO1upregulate FtH1 and LIP results significantly diminished. HEY tumor spheroids prevent iron import downregulating CD71, while PEO1 cells strongly enhance it. Intracellular ROS drop down during the 2D to 3D transition in both cell lines, but more significantly in PEO1 cells. Upon iron supplementation, PEO1 cells continue to enhance CD71 and FtH1 without accumulating the LIP and ROS and do not undergo ferroptosis. HEY, instead, accumulate LIP, undergo ferroptosis and attenuate their sphere-forming ability and invasiveness. FtH1 knockdown significantly reduces the generation of PEO1 tumor spheroids, although without sensitizing them to ferroptosis. Discussion: Iron metabolism reprogramming is a key event in the tumor spheroid generation of ovarian cancer cells. An iron-rich environment impairs the sphere-forming ability and causes cell death only in ferroptosis sensitive cells. A better understanding of ferroptosis sensitivity could be useful to develop effective treatments to kill ECM-detached ovarian cancer cells.

2.
Antioxid Redox Signal ; 28(1): 1-14, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28793778

RESUMEN

AIMS: Iron overload (IO) is a life-threatening complication of chronic hemolytic disorders such as ß-thalassemia. IO results in severe cellular oxidative damage, leading to organ failure. Peroxiredoxin-2 (Prx2), a typical 2-cysteine-(Cys)-peroxiredoxin, is an important component of the cytoprotective system, but its response to IO is still to be fully defined. RESULTS: We studied the effects of IO on Prx2-knockout mice (Prx2-/-). The absence of Prx2 enhanced toxicity due to IO on erythropoiesis. We found that IO failed to induce the typical hepcidin (Hamp) upregulation in Prx2-/- mice due to its failure to activate the signal transducer and activator of transcription-3 (STAT3) with intact Jak2 signaling. In Prx2-/- mice, the loss of Hamp response was also observed after administration of a single dose of oral iron. When lipopolysaccharide (LPS) was used to explore IL6-STAT3 activation in Prx2-/- mice, STAT3 activation and Hamp upregulation were once again defective. Treatment with PEP-fusion-recombinant-Prx2 (PEP Prx2) significantly increased STAT3 activation with upregulation of Hamp expression in both IO- and LPS-exposed Prx2-/- mice. We also confirmed the beneficial effects of PEP Prx2 on Hamp expression through STAT3 activation in ß-thalassemic mice. INNOVATION: We propose that Prx2 plays a key role in responding to cytotoxicity of IO, directly targeting STAT3-transcriptional factor in a Jak2-independent fashion and regulating Hamp in response to canonical stimuli. CONCLUSION: Collectively, our data highlight a novel role of Prx2 in iron homeostasis. Prx2 is a key cytoprotector against IO that is induced either by iron supplementation or due to chronic hemolysis as in ß-thalassemia. Prx2 is required to support STAT3 transcriptional activity and regulation of Hamp expression. Antioxid. Redox Signal. 28, 1-14.


Asunto(s)
Eritropoyesis , Homeostasis , Hierro/metabolismo , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Anemia/tratamiento farmacológico , Anemia/etiología , Anemia/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Citoprotección/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Hepcidinas/genética , Hepcidinas/metabolismo , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/metabolismo , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Modelos Biológicos , Estrés Oxidativo , Peroxirredoxinas/farmacología , Proteínas Recombinantes , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
EMBO Mol Med ; 8(10): 1197-1211, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27516453

RESUMEN

Pantothenate kinase-associated neurodegeneration (PKAN) is an early onset and severely disabling neurodegenerative disease for which no therapy is available. PKAN is caused by mutations in PANK2, which encodes for the mitochondrial enzyme pantothenate kinase 2. Its function is to catalyze the first limiting step of Coenzyme A (CoA) biosynthesis. We generated induced pluripotent stem cells from PKAN patients and showed that their derived neurons exhibited premature death, increased ROS production, mitochondrial dysfunctions-including impairment of mitochondrial iron-dependent biosynthesis-and major membrane excitability defects. CoA supplementation prevented neuronal death and ROS formation by restoring mitochondrial and neuronal functionality. Our findings provide direct evidence that PANK2 malfunctioning is responsible for abnormal phenotypes in human neuronal cells and indicate CoA treatment as a possible therapeutic intervention.


Asunto(s)
Coenzima A/metabolismo , Neuronas/patología , Neurodegeneración Asociada a Pantotenato Quinasa/fisiopatología , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Muerte Celular , Células Cultivadas , Humanos , Mitocondrias/patología , Células Madre Pluripotentes/fisiología , Especies Reactivas de Oxígeno/metabolismo
4.
Hum Mol Genet ; 21(18): 4049-59, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22692681

RESUMEN

Pantothenate kinase-associated neurodegeneration (PKAN) is a neurodegenerative disease belonging to the group of neurodegeneration with brain iron accumulation disorders. It is characterized by progressive impairments in movement, speech and cognition. The disease is inherited in a recessive manner due to mutations in the Pantothenate Kinase-2 (PANK2) gene that encodes a mitochondrial protein involved in Coenzyme A synthesis. To investigate the link between a PANK2 gene defect and iron accumulation, we analyzed primary skin fibroblasts from three PKAN patients and three unaffected subjects. The oxidative status of the cells and their ability to respond to iron were analyzed in both basal and iron supplementation conditions. In basal conditions, PKAN fibroblasts show an increase in carbonylated proteins and altered expression of antioxidant enzymes with respect to the controls. After iron supplementation, the PKAN fibroblasts had a defective response to the additional iron. Under these conditions, ferritins were up-regulated and Transferrin Receptor 1 (TfR1) was down-regulated to a minor extent in patients compared with the controls. Analysis of iron regulatory proteins (IRPs) reveals that, with respect to the controls, PKAN fibroblasts have a reduced amount of membrane-associated mRNA-bound IRP1, which responds imperfectly to iron. This accounts for the defective expression of ferritin and TfR1 in patients' cells. The inaccurate quantity of these proteins produced a higher bioactive labile iron pool and consequently increased iron-dependent reactive oxygen species formation. Our results suggest that Pank2 deficiency promotes an increased oxidative status that is further enhanced by the addition of iron, potentially causing damage in cells.


Asunto(s)
Fibroblastos/metabolismo , Hierro/metabolismo , Neurodegeneración Asociada a Pantotenato Quinasa/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Piel/patología , Estudios de Casos y Controles , Catalasa/metabolismo , Células Cultivadas , Ferritinas/metabolismo , Fibroblastos/enzimología , Humanos , Proteínas Reguladoras del Hierro/metabolismo , Mutación Missense , Oxidación-Reducción , Estrés Oxidativo , Neurodegeneración Asociada a Pantotenato Quinasa/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Unión Proteica , Carbonilación Proteica , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
5.
Blood ; 109(10): 4503-10, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17264300

RESUMEN

Hemojuvelin (HJV) positively modulates the iron regulator hepcidin, and its mutations are the major cause of juvenile hemochromatosis (JH), a recessive disease leading to iron overload. Defective HJV reduces hepcidin up-regulation both in humans and in Hjv-deficient mice. To investigate the JH pathogenesis and the functional properties of human HJV we studied the biosynthesis and maturation of 6 HJV pathogenic mutants in HeLa and HepG2 cells. We show that proteolytic processing is defective in mutants F170S, W191C, and G320V, but not in G99V and C119F. Moreover, we show that mutants G99V and C119F are targeted to the cell surface, while F170S, W191C, G320V, and R326X (lacking the glycosilphosphatidylinositol [GPI] anchor) are mainly retained in the endoplasmic reticulum, although all mutants are released as soluble forms (s-HJV) in a proportion that is modulated by iron supplementation. Membrane HJV (m-HJV) is mainly composed of the cleaved protein, and its level is increased by iron in wild-type (WT) mice but not in the mutants. Altogether, the data demonstrate that the loss of HJV membrane export is central to the pathogenesis of JH, and that HJV cleavage is essential for the export. The results support a dual function for s- and m-HJV in iron deficiency and overload, respectively.


Asunto(s)
Membrana Celular/metabolismo , Hemocromatosis/etiología , Hemocromatosis/metabolismo , Proteínas de la Membrana/metabolismo , Niño , Retículo Endoplásmico/metabolismo , Proteínas Ligadas a GPI , Células HeLa , Proteína de la Hemocromatosis , Humanos , Hierro/fisiología , Proteínas de la Membrana/genética , Proteínas Mutantes/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Células Tumorales Cultivadas
6.
Blood ; 109(8): 3552-9, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17192393

RESUMEN

X-linked sideroblastic anemia with ataxia (XLSA/A) is caused by defects of the transporter ABCB7 and is characterized by mitochondrial iron deposition and excess of protoporphyrin in erythroid cells. We describe ABCB7 silencing in HeLa cells by performing sequential transfections with siRNAs. The phenotype of the ABCB7-deficient cells was characterized by a strong reduction in proliferation rate that was not rescued by iron supplementation, by evident signs of iron deficiency, and by a large approximately 6-fold increase of iron accumulation in the mitochondria that was poorly available to mitochondrial ferritin. The cells showed an increase of protoporphyrin IX, a higher sensitivity to H(2)O(2) toxicity, and a reduced activity of mitochondrial superoxide dismutase 2 (SOD2), while the activity of mitochondrial enzymes, such as citrate synthase or succinate dehydrogenase, and ATP content were not decreased. In contrast, aconitase activity, particularly that of the cytosolic, IRP1 form, was reduced. The results support the hypothesis that ABCB7 is involved in the transfer of iron from mitochondria to cytosol, and in the maturation of cytosolic Fe/S enzymes. In addition, the results indicate that anemia in XLSA/A is caused by the accumulation of iron in a form that is not readily usable for heme synthesis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Anemia Ferropénica/genética , Anemia Sideroblástica/genética , Ataxia/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Sobrecarga de Hierro/genética , Mitocondrias/genética , Interferencia de ARN , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Anemia Ferropénica/metabolismo , Anemia Sideroblástica/metabolismo , Ataxia/metabolismo , Transporte Biológico/genética , Citoplasma/genética , Citoplasma/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Células HeLa , Hemo/biosíntesis , Hemo/genética , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fenotipo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
7.
Br J Haematol ; 127(5): 598-603, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15566364

RESUMEN

Missense mutations in the ferroportin gene (SLC11A3) result in haemochromatosis type 4 [HFE4, Online Mendelian Inheritance in Man (OMIM) reference 606069] or ferroportin disease, an autosomal dominant disorder characterized by predominantly reticuloendothelial iron accumulation. To verify whether HFE4 is caused by defective iron recycling because of loss of functionality of ferroportin, we down-regulated SLC11A gene expression in human macrophages by using small interfering RNAs (siRNAs). Transfection experiments with ferroportin siRNAs resulted in a marked reduction (about two-thirds on average) in ferroportin mRNA levels as detected by quantitative real time polymerase chain reaction. When macrophages were grown in medium supplemented with iron, cells transfected with siRNAs displayed three- to eightfold increases in staining intensities following Perls reaction. These macrophages also showed significant increases in H-ferritin content. The observation that ferroportin mRNA down-regulation to levels compatible with haplo-insufficiency causes increased iron retention and H-ferritin synthesis in cultured macrophages has important implications. First, this indicates that ferroportin levels must be finely regulated in order to maintain cellular iron homeostasis, and that both copies of SLC11A3 must function efficiently to prevent iron accumulation. Second, this observation supports the hypothesis that reticuloendothelial iron overload in patients with ferroportin disease is caused by loss-of-function mutations in the SLC11A3 gene that mainly impair macrophage iron recycling.


Asunto(s)
Proteínas de Transporte de Catión/genética , Ferritinas/biosíntesis , Hierro/metabolismo , Macrófagos/metabolismo , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Silenciador del Gen , Hemostasis , Humanos , ARN Mensajero/análisis , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección/métodos
8.
Blood ; 103(6): 2377-83, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-14615379

RESUMEN

We describe the use of small interfering RNAs (siRNAs) to down-regulate H- and L-ferritin levels in HeLa cells. siRNAs repressed H- and L-ferritin expression to about 20% to 25% of the background level in both stable and transient transfections. HeLa cells transfected with H- and L-ferritin cDNAs were analyzed in parallel to compare the effects of ferritin up- and down-regulation. We found that large modifications of L-ferritin levels did not affect iron availability in HeLa cells but positively affected cell proliferation rate in an iron-independent manner. The transient down-regulation of H-ferritin modified cellular iron availability and resistance to oxidative damage, as expected. In contrast, the stable suppression of H-ferritin in HeLa cell clones transfected with siRNAs did not increase cellular iron availability but made cells less resistant to iron supplementation and chelation. The results indicate that L-ferritin has no direct effects on cellular iron homeostasis in HeLa cells, while it has new, iron-unrelated functions. In addition, they suggest that H-ferritin function is to act as an iron buffer.


Asunto(s)
Compuestos Férricos/toxicidad , Ferritinas/genética , Ferritinas/metabolismo , Compuestos de Amonio Cuaternario/toxicidad , Tampones (Química) , División Celular/efectos de los fármacos , División Celular/fisiología , ADN Complementario , Regulación hacia Abajo , Expresión Génica , Células HeLa , Humanos , Hierro/metabolismo , Estrés Oxidativo/fisiología , ARN Interferente Pequeño , Transfección , Regulación hacia Arriba
9.
FEBS Lett ; 537(1-3): 187-92, 2003 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-12606055

RESUMEN

We found that tumor necrosis factor alpha (TNFalpha)-induced apoptosis in HeLa cells was accompanied by a approximately 2-fold increase in H- and L-ferritin and a decrease in transferrin receptor, two indices of increased iron availability. Iron supplementation and overexpression of H-ferritin or its mutant with an inactivated ferroxidase center reduced by about approximately 50% the number of apoptotic cells after TNFalpha-treatment, while overexpression of L-ferritin was ineffective. The data indicate that H-ferritin has an anti-apoptotic activity unrelated to its ferroxidase activity and to its capacity to modify cellular iron metabolism.


Asunto(s)
Apoptosis/fisiología , Ferritinas/metabolismo , Hierro/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Apoptosis/efectos de los fármacos , Dactinomicina/farmacología , Células HeLa , Humanos , Cinética , Receptores de Transferrina/efectos de los fármacos , Receptores de Transferrina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA