Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(10): e1011679, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37812650

RESUMEN

Malaria and iron deficiency are major global health problems with extensive epidemiological overlap. Iron deficiency-induced anaemia can protect the host from malaria by limiting parasite growth. On the other hand, iron deficiency can significantly disrupt immune cell function. However, the impact of host cell iron scarcity beyond anaemia remains elusive in malaria. To address this, we employed a transgenic mouse model carrying a mutation in the transferrin receptor (TfrcY20H/Y20H), which limits the ability of cells to internalise iron from plasma. At homeostasis TfrcY20H/Y20H mice appear healthy and are not anaemic. However, TfrcY20H/Y20H mice infected with Plasmodium chabaudi chabaudi AS showed significantly higher peak parasitaemia and body weight loss. We found that TfrcY20H/Y20H mice displayed a similar trajectory of malaria-induced anaemia as wild-type mice, and elevated circulating iron did not increase peak parasitaemia. Instead, P. chabaudi infected TfrcY20H/Y20H mice had an impaired innate and adaptive immune response, marked by decreased cell proliferation and cytokine production. Moreover, we demonstrated that these immune cell impairments were cell-intrinsic, as ex vivo iron supplementation fully recovered CD4+ T cell and B cell function. Despite the inhibited immune response and increased parasitaemia, TfrcY20H/Y20H mice displayed mitigated liver damage, characterised by decreased parasite sequestration in the liver and an attenuated hepatic immune response. Together, these results show that host cell iron scarcity inhibits the immune response but prevents excessive hepatic tissue damage during malaria infection. These divergent effects shed light on the role of iron in the complex balance between protection and pathology in malaria.


Asunto(s)
Anemia , Deficiencias de Hierro , Malaria , Plasmodium chabaudi , Animales , Ratones , Hierro , Malaria/parasitología , Inmunidad , Plasmodium chabaudi/fisiología
2.
J Hepatol ; 60(2): 354-61, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24021424

RESUMEN

BACKGROUND & AIMS: The liver, being the major site of iron storage, is particularly exposed to the toxic effects of iron. Transcription factor NRF2 is critical for protecting the liver against disease by activating the transcription of genes encoding detoxification/antioxidant enzymes. We aimed to determine if the NRF2 pathway plays a significant role in the protection against hepatic iron overload. METHODS: Wild-type and Nrf2(-/-) mouse primary hepatocytes were incubated with ferric ammonium citrate. Wild-type and Nrf2(-/-) mice were fed standard rodent chow or iron-rich diet for 2weeks, with or without daily injection of the antioxidant mito-TEMPOL. RESULTS: In mouse hepatocytes, iron induced the nuclear translocation of NRF2 and the expression of cytoprotective genes in an NRF2-dependent manner. Moreover, Nrf2(-/-) hepatocytes were highly susceptible to iron-induced cell death. Wild-type and Nrf2(-/-) mice fed iron-rich diet accumulated similar amounts of iron in the liver and were equally able to increase the expression of hepatic hepcidin and ferritin. Nevertheless, in Nrf2-null mice the iron loading resulted in progressive liver injury, ranging from mild confluent necrosis to severe necroinflammatory lesions. Hepatocytic cell death was associated with gross ultrastructural damage to the mitochondria. Notably, liver injury was prevented in iron-fed animals that received mito-TEMPOL. CONCLUSIONS: NRF2 protects the mouse liver against the toxicity of dietary iron overload by preventing hepatocytic cell death. We identify NRF2 as a potential modifier of liver disease in iron overload pathology and show the beneficial effect of the antioxidant mito-TEMPOL in a mouse model of dietary iron-induced liver injury.


Asunto(s)
Hepatocitos/metabolismo , Hierro de la Dieta/toxicidad , Hígado/lesiones , Hígado/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Antioxidantes/farmacología , Óxidos N-Cíclicos/farmacología , Modelos Animales de Enfermedad , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Marcadores de Spin
3.
Leukemia ; 38(1): 96-108, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37857886

RESUMEN

Iron overload (IOL) is hypothesized to contribute to dysplastic erythropoiesis. Several conditions, including myelodysplastic syndrome, thalassemia and sickle cell anemia, are characterized by ineffective erythropoiesis and IOL. Iron is pro-oxidant and may participate in the pathophysiology of these conditions by increasing genomic instability and altering the microenvironment. There is, however, lack of in vivo evidence demonstrating a role of IOL and oxidative damage in dysplastic erythropoiesis. NRF2 transcription factor is the master regulator of antioxidant defenses, playing a crucial role in the cellular response to IOL in the liver. Here, we crossed Nrf2-/- with hemochromatosis (Hfe-/-) or hepcidin-null (Hamp1-/-) mice. Double-knockout mice developed features of ineffective erythropoiesis and myelodysplasia including macrocytic anemia, splenomegaly, and accumulation of immature dysplastic bone marrow (BM) cells. BM cells from Nrf2/Hamp1-/- mice showed increased in vitro clonogenic potential and, upon serial transplantation, recipients disclosed cytopenias, despite normal engraftment, suggesting defective differentiation. Unstimulated karyotype analysis showed increased chromosome instability and aneuploidy in Nrf2/Hamp1-/- BM cells. In HFE-related hemochromatosis patients, NRF2 promoter SNP rs35652124 genotype TT (predicted to decrease NRF2 expression) associated with increased MCV, consistent with erythroid dysplasia. Our results suggest that IOL induces ineffective erythropoiesis and dysplastic hematologic features through oxidative damage in Nrf2-deficient cells.


Asunto(s)
Anemia , Hemocromatosis , Sobrecarga de Hierro , Síndromes Mielodisplásicos , Animales , Humanos , Ratones , Anemia/metabolismo , Eritropoyesis/genética , Hemocromatosis/genética , Hemocromatosis/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Ratones Noqueados , Síndromes Mielodisplásicos/genética , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo
4.
Cell Oncol (Dordr) ; 46(6): 1545-1558, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37273145

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the second deadliest malignancy worldwide. Current dietary habits are associated with increased levels of iron and heme, both of which increase the risk of developing CRC. The harmful effects of iron overload are related to the induction of iron-mediated pro-tumorigenic pathways, including carcinogenesis and hyperproliferation. On the other hand, iron deficiency may also promote CRC development and progression by contributing to genome instability, therapy resistance, and diminished immune responses. In addition to the relevance of systemic iron levels, iron-regulatory mechanisms in the tumor microenvironment are also believed to play a significant role in CRC and to influence disease outcome. Furthermore, CRC cells are more prone to escape iron-dependent cell death (ferroptosis) than non-malignant cells due to the constitutive activation of antioxidant genes expression. There is wide evidence that inhibition of ferroptosis may contribute to the resistance of CRC to established chemotherapeutic regimens. As such, ferroptosis inducers represent promising therapeutic drugs for CRC. CONCLUSIONS AND PERSPECTIVES: This review addresses the complex role of iron in CRC, particularly in what concerns the consequences of iron excess or deprivation in tumor development and progression. We also dissect the regulation of cellular iron metabolism in the CRC microenvironment and emphasize the role of hypoxia and of oxidative stress (e.g. ferroptosis) in CRC. Finally, we underline some iron-related players as potential therapeutic targets against CRC malignancy.


Asunto(s)
Carcinogénesis , Neoplasias Colorrectales , Humanos , Carcinogénesis/metabolismo , Muerte Celular , Hierro/metabolismo , Neoplasias Colorrectales/metabolismo , Microambiente Tumoral
5.
J Vis Exp ; (179)2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35156663

RESUMEN

Iron is an essential micronutrient. Both iron overload and deficiency are highly detrimental to humans, and tissue iron levels are finely regulated. The use of experimental animal models of iron overload or deficiency has been instrumental to advance knowledge of the mechanisms involved in the systemic and cellular regulation of iron homeostasis. The measurement of total iron levels in animal tissues is commonly performed with atomic absorption spectroscopy or with a colorimetric assay based on the reaction of non-heme iron with a bathophenanthroline reagent. For many years, the colorimetric assay has been used for the measurement of the non-heme iron content in a wide range of animal tissues. Unlike atomic absorption spectroscopy, it excludes the contribution of heme iron derived from hemoglobin contained in red blood cells. Moreover, it does not require sophisticated analytical skills or highly expensive equipment, and can thus be easily implemented in most laboratories. Finally, the colorimetric assay can be either cuvette-based or adapted to a microplate format, allowing higher sample throughput. The present work provides a well-established protocol that is suited for the detection of alterations in tissue iron levels in a variety of experimental animal models of iron overload or iron deficiency.


Asunto(s)
Colorimetría , Sobrecarga de Hierro , Animales , Hierro/química , Fenantrolinas
6.
Antioxid Redox Signal ; 35(6): 433-452, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32791852

RESUMEN

Significance: Oxygen metabolism and iron homeostasis are closely linked. Iron facilitates the oxygen-carrying capacity of blood, and its deficiency causes anemia. Conversely, excess free iron is detrimental for stimulating the formation of reactive oxygen species, causing tissue damage. The amount and distribution of iron thus need to be tightly regulated by the liver-expressed hormone hepcidin. This review analyzes the roles of key oxygen-sensing pathways in cellular and systemic regulation of iron homeostasis; specifically, the prolyl hydroxylase domain (PHD)/hypoxia-inducible factor (HIF) and the Kelch-like ECH-associated protein 1/NF-E2 p45-related factor 2 (KEAP1/NRF2) pathways, which mediate tissue adaptation to low and high oxygen, respectively. Recent Advances: In macrophages, NRF2 regulates genes involved in hemoglobin catabolism, iron storage, and iron export. NRF2 was recently identified as the molecular sensor of iron-induced oxidative stress and is responsible for BMP6 expression by liver sinusoidal endothelial cells, which in turn activates hepcidin synthesis by hepatocytes to restore systemic iron levels. Moreover, NRF2 orchestrates the activation of antioxidant defenses that are crucial to protect against iron toxicity. On the contrary, low iron/hypoxia stabilizes renal HIF2a via inactivation of iron-dependent PHD dioxygenases, causing an erythropoietic stimulus that represses hepcidin via an inhibitory effect of erythroferrone on bone morphogenetic proteins. Intestinal HIF2a is also stabilized, increasing the expression of genes involved in dietary iron absorption. Critical Issues: An intimate crosstalk between oxygen-sensing pathways and iron regulatory mechanisms ensures that fluctuations in systemic iron levels are promptly detected and restored. Future Directions: The realization that redox-sensitive transcription factors regulate systemic iron levels suggests novel therapeutic approaches. Antioxid. Redox Signal. 35, 433-452.


Asunto(s)
Homeostasis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia , Hierro/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Humanos , Oxidación-Reducción
7.
Blood Adv ; 5(16): 3102-3112, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34402883

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease with poor prognosis and limited treatment strategies. Determining the role of cell-extrinsic regulators of leukemic cells is vital to gain clinical insights into the biology of AML. Iron is a key extrinsic regulator of cancer, but its systemic regulation remains poorly explored in AML. To address this question, we studied iron metabolism in patients with AML at diagnosis and explored the mechanisms involved using the syngeneic MLL-AF9-induced AML mouse model. We found that AML is a disorder with a unique iron profile, not associated with inflammation or transfusion, characterized by high ferritin, low transferrin, high transferrin saturation (TSAT), and high hepcidin. The increased TSAT in particular, contrasts with observations in other cancer types and in anemia of inflammation. Using the MLL-AF9 mouse model of AML, we demonstrated that the AML-induced loss of erythroblasts is responsible for iron redistribution and increased TSAT. We also show that AML progression is delayed in mouse models of systemic iron overload and that elevated TSAT at diagnosis is independently associated with increased overall survival in AML. We suggest that TSAT may be a relevant prognostic marker in AML.


Asunto(s)
Anemia , Leucemia Mieloide Aguda , Animales , Eritroblastos , Humanos , Hierro , Ratones , Transferrina
8.
Med ; 2(2): 164-179.e12, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33665641

RESUMEN

BACKGROUND: How specific nutrients influence adaptive immunity is of broad interest. Iron deficiency is the most common micronutrient deficiency worldwide and imparts a significant burden of global disease; however, its effects on immunity remain unclear. METHODS: We used a hepcidin mimetic and several genetic models to examine the effect of low iron availability on T cells in vitro and on immune responses to vaccines and viral infection in mice. We examined humoral immunity in human patients with raised hepcidin and low serum iron caused by mutant TMPRSS6. We tested the effect of iron supplementation on vaccination-induced humoral immunity in piglets, a natural model of iron deficiency. FINDINGS: We show that low serum iron (hypoferremia), caused by increased hepcidin, severely impairs effector and memory responses to immunizations. The intensified metabolism of activated lymphocytes requires the support of enhanced iron acquisition, which is facilitated by IRP1/2 and TFRC. Accordingly, providing extra iron improved the response to vaccination in hypoferremic mice and piglets, while conversely, hypoferremic humans with chronically increased hepcidin have reduced concentrations of antibodies specific for certain pathogens. Imposing hypoferremia blunted the T cell, B cell, and neutralizing antibody responses to influenza virus infection in mice, allowing the virus to persist and exacerbating lung inflammation and morbidity. CONCLUSIONS: Hypoferremia, a well-conserved physiological innate response to infection, can counteract the development of adaptive immunity. This nutrient trade-off is relevant for understanding and improving immune responses to infections and vaccines in the globally common contexts of iron deficiency and inflammatory disorders. FUNDING: Medical Research Council, UK.


Asunto(s)
Deficiencias de Hierro , Trastornos del Metabolismo del Hierro , Animales , Hepcidinas/genética , Humanos , Inmunidad Humoral , Hierro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Porcinos , Vacunación
9.
DNA Repair (Amst) ; 7(12): 1982-9, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18793759

RESUMEN

Thiopurine antimetabolites, such as azathioprine (Aza) and 6-thioguanine (6-TG), are widely used in the treatment of cancer, inflammatory conditions and organ transplantation patients. Recent work has shown that cells treated with 6-TG and UVA generate ROS, with implied oxidatively generated modification of DNA. In a study of urinary 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) in renal transplant patients, we provided the first in vivo evidence linking Aza and oxidatively damaged DNA. Using the hOGG1 comet assay, we herein demonstrate high levels of 8-oxodG and alkali-labile sites (ALS) in cells treated with biologically relevant doses of 6-TG, or Aza, plus UVA. This damage was induced dose-dependently. Surprisingly, given the involvement of 6-TG incorporation into DNA in its therapeutic effect, significant amounts of 8-oxodG and ALS were induced in quiescent cells, although less than in proliferating cells. We speculate that some activity of hOGG1 towards unirradiated, 6-TG treated cells, implies possible recognition of 6-TG or derivatives thereof. This is the first report to conclusively demonstrate oxidatively damaged DNA in cells treated with thiopurines and UVA. These data indicate that Aza-derived oxidative stress will occur in the skin of patients on Aza, following even low level UVA exposure. This is a probable contributor to the increased risk of non-melanoma skin cancer in these patients. However, as oxidative stress is unlikely to be involved in the therapeutic effects of Aza, intercepting ROS production in the skin could be a viable route by which this side effect may be minimised.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azatioprina/farmacología , Desoxiguanosina/análogos & derivados , Especies Reactivas de Oxígeno/metabolismo , Rayos Ultravioleta , 8-Hidroxi-2'-Desoxicoguanosina , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Ensayo Cometa , ADN/efectos de los fármacos , ADN/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , ADN Glicosilasas/metabolismo , Desoxiguanosina/metabolismo , Relación Dosis-Respuesta en la Radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Humanos , Estrés Oxidativo , Tioguanina/farmacología
10.
Toxicol Lett ; 185(1): 23-31, 2009 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-19095052

RESUMEN

Polybrominated diphenyl ethers (PBDEs) are commonly used as flame retardants in a variety of commercial and household products. They have been detected in the environment and accumulate in mammalian tissues and fluids. PBDE toxicity is thought to be associated with endocrine disruption, developmental neurotoxicity and changes in fetal development. Although humans are exposed to PBDEs, our knowledge of the effects of PBDE metabolites on human cells with respect to health risk is insufficient. Two hydroxylated PBDEs (OH-PBDEs), 2-OH-BDE47 and 2-OH-BDE85, were investigated for their effects on cell viability/proliferation, DNA damage, cell cycle distribution and gene expression profiling in H295R adrenocortical carcinoma cells. We show that the two agents are cytotoxic in a dose-dependent manner only at micromolar concentrations, with 2-OH-BDE85 being more toxic than 2-OH-BDE47. However, no DNA damage was observed for either chemical, suggesting that the biological effects of OH-PBDEs occur primarily via non-genotoxic routes. Furthermore, no evidence of aryl hydrocarbon receptor (AHR)-mediated, dioxin-like toxicity was observed. Instead, we report that a micromolar concentration of OH-PBDEs induces transcriptional changes associated with endoplasmic reticulum stress and the unfolded protein response. We discuss whether OH-PBDE bioaccumulation could result in impairment of the adrenocortical secretory function.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/patología , Perfilación de la Expresión Génica , Éteres Difenilos Halogenados/toxicidad , Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/efectos de los fármacos , Éteres Difenilos Halogenados/metabolismo , Humanos
11.
Mutat Res ; 669(1-2): 112-21, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19505483

RESUMEN

A number of recent observations have suggested a potential role for membrane-bound gamma-glutamyltransferase (GGT) in tumor progression and appearance of more aggressive and resistant phenotypes, through redox interactions leading to production of reactive oxygen species. The present study was aimed to evaluate whether such pro-oxidant activity of GGT can promote oxidative DNA damage, thus contributing to cancer genomic instability. Human GGT-transfected melanoma cells were studied, and DNA damage was measured by using the alkaline comet assay. Our results indicate that higher levels of GGT activity are associated with higher levels of background DNA damage and oxidized bases. This association cannot be explained by differences in cell cycle distribution or apoptotic rates. GGT-over-expressing cells also presented with a markedly higher glucose uptake, a phenomenon potentially leading to higher metabolic rate and oxidative DNA damage. Anyway, when GGT-over-expressing cells were incubated in the presence of GGT substrates and a source of catalytic iron, increased levels of DNA damage and oxidized bases were observed, an effect completely prevented in the presence of GGT inhibitors or various antioxidants.The findings reported indicate that GGT activity is able to promote iron-dependent DNA oxidative damage, thus potentially representing an important mechanism in initiation/progression of neoplastic transformation.


Asunto(s)
Membrana Celular/enzimología , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Compuestos Férricos/farmacología , Melanoma/enzimología , Melanoma/genética , gamma-Glutamiltransferasa/metabolismo , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cloruros , Ensayo Cometa , Fibrinolíticos/farmacología , Humanos , Melanoma/patología , Oxidación-Reducción , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
12.
Nat Metab ; 1(5): 519-531, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31276102

RESUMEN

Iron is critical for life but toxic in excess because of iron-catalysed formation of pro-oxidants that cause tissue damage in a range of disorders. The Nrf2 transcription factor orchestrates cell-intrinsic protective antioxidant responses, and the peptide hormone hepcidin maintains systemic iron homeostasis, but is pathophysiologically decreased in haemochromatosis and beta-thalassaemia. Here, we show that Nrf2 is activated by iron-induced, mitochondria-derived pro-oxidants and drives Bmp6 expression in liver sinusoid endothelial cells, which in turn increases hepcidin synthesis by neighbouring hepatocytes. In Nrf2 knockout mice, the Bmp6-hepcidin response to oral and parenteral iron is impaired and iron accumulation and hepatic damage are increased. Pharmacological activation of Nrf2 stimulates the Bmp6-hepcidin axis, improving iron homeostasis in haemochromatosis and counteracting the inhibition of Bmp6 by erythroferrone in beta-thalassaemia. We propose that Nrf2 links cellular sensing of excess toxic iron to control of systemic iron homeostasis and antioxidant responses, and may be a therapeutic target for iron-associated disorders.


Asunto(s)
Proteína Morfogenética Ósea 6/fisiología , Hepcidinas/fisiología , Homeostasis/fisiología , Hierro/metabolismo , Factor 2 Relacionado con NF-E2/fisiología , Talasemia beta/fisiopatología , Humanos
13.
Int J Cancer ; 122(8): 1810-9, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18074354

RESUMEN

Systemic chemotherapy is extensively used in cancer therapy, however, for many treatments' response rates are limited. Furthermore, certain regimens are frequently associated with significant morbidity and occasional mortality. Consequently, when alternative options exist, it is desirable to reserve a particular chemotherapy for those patients whose tumours will respond. Therefore, attention is turning to the development of techniques that could provide predictive information regarding a tumour's particular chemosensitivity, as a means of enhancing patient selection for that specific treatment. One approach has been to focus on measures of DNA damage formation and repair as being potentially predictive of cancer cell chemosensitivity, the premise being that higher levels of induced DNA damage (resulting from the chemotherapeutic agents) and/or deficiencies in DNA damage repair are indicative of greater sensitivity. In the present study we have investigated the Comet assay response of a panel of non-small cell lung cancer cell lines towards cisplatin and found an inverse correlation between sensitivity and damage formation resulting from this agent. Moreover, an inverse correlation was found between resistance and extent of damage repair. Further analysis of multiple alternate cellular end-points (including cell cycle analysis, apoptosis and gene expression changes) revealed cisplatin damage tolerance to be a chemoresistance mechanism in this model system. This study highlights damage tolerance mechanisms as potentially confounding factors in attempts to develop predictive tests based on measures of genotoxicity. To address this we would argue that a range of multiple end-points should be analysed to ascertain the "complete predictive picture".


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/farmacología , Ensayo Cometa , Reactivos de Enlaces Cruzados/farmacología , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/genética , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cruzamientos Genéticos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Valor Predictivo de las Pruebas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
14.
DNA Repair (Amst) ; 5(2): 219-25, 2006 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-16253571

RESUMEN

Oxaliplatin is frequently used in the therapy of cancer. In DNA, oxaliplatin induces, like cisplatin, the formation of crosslinks, which are commonly accepted as being responsible for the cytotoxicity of platinum agents. The detection of oxaliplatin-induced DNA crosslink formation and repair could be a good measure of assessing how a patient is responding to the agent. In this study, we used a validated modification of the alkaline comet assay for detecting the presence of these crosslinks in vitro and in cancer patients. The H460 tumour cell line was treated in vitro with a range of oxaliplatin and cisplatin doses, and the subsequent crosslink formation and repair compared between the two agents. In addition, lymphocytes from cancer patients undergoing oxaliplatin-based chemotherapy were assayed for the formation and repair of oxaliplatin-induced crosslinks. A dose-response was observed in the in vitro samples, with cisplatin producing more crosslinks than oxaliplatin at equimolar concentrations and lesions induced by both agents showing different repair efficiencies. Furthermore, evidence of crosslink formation and repair was observed in the peripheral blood lymphocytes of all cancer patients studied, along with the detection of interindividual variability in crosslink formation and repair efficiencies. To the best of our knowledge, this is the first time that oxaliplatin DNA crosslinks have been detected either in vitro or in patient samples using the alkaline comet assay. Due to its sensitivity, rapidity, small cell sample and low cost, the alkaline comet assay is a good method for the detection of oxaliplatin-induced crosslinks and their subsequent repair and, in future clinical studies, could prove to be a valuable tool in assessing/predicting a patient's response to chemotherapy.


Asunto(s)
Carcinoma de Células Grandes/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Reactivos de Enlaces Cruzados/farmacología , ADN/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias/genética , Compuestos Organoplatinos/farmacología , Anciano , Línea Celular Tumoral , Cisplatino/farmacología , Ensayo Cometa/métodos , ADN/metabolismo , Daño del ADN , Reparación del ADN , ADN Polimerasa Dirigida por ADN/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Masculino , Persona de Mediana Edad , Modelos Químicos , Modelos Estadísticos , Metástasis de la Neoplasia , Oxaliplatino , Sensibilidad y Especificidad , Factores de Tiempo , ADN Polimerasa iota
15.
Free Radic Biol Med ; 43(8): 1165-75, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17854712

RESUMEN

Vitamin C (ascorbic acid, AA) is an important antioxidant in human plasma. It is clear, however, that AA has other important, nonantioxidant roles in cells. Of particular interest is its involvement in iron metabolism, since AA enhances dietary iron absorption, increases the activity of Fe(2+)-dependent cellular enzymes, promotes Fenton reactions in vitro, and was reported to have deleterious effects in individuals with iron overload. Nevertheless, the ability of AA to modulate iron metabolism and enhance iron-dependent damage in cells, tissues, and organisms has not been fully elucidated. Here we investigated the effect of AA on iron-mediated oxidative stress in normal human fibroblasts. Incubation with physiologically relevant concentrations of AA was not harmful but sensitised cells toward H(2)O(2)-induced, iron-dependent DNA strand breakage and cell death. We also report that AA increased the levels of intracellular catalytic iron and concomitantly modulated the expression of two well-established iron-regulated genes, ferritin and transferrin receptor. In summary, we present evidence of a novel, nonantioxidant role of AA in human cells, where it increases iron availability and enhances ROS-mediated, iron-dependent damage. We suggest that AA may exacerbate the deleterious effects of metals in vivo and promote normal tissue injury in situations associated with elevated ROS production.


Asunto(s)
Ácido Ascórbico/farmacología , Homeostasis/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Hierro/metabolismo , Ácido Ascórbico/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Regulación hacia Abajo , Ferritinas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Transferrina/metabolismo
16.
Toxicol Lett ; 170(1): 57-65, 2007 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-17382497

RESUMEN

In the presence of oxygen, ascorbic acid (AA) is unstable in aqueous media and oxidises to dehydroascorbate (DHA), generating reactive intermediates such as ascorbate free radical and H2O2. It is proposed that the cytotoxicity of AA is due to the extracellular production of H2O2 and that this is mediated by transition metal ions present in cell media. Here we investigate the role of extracellular H2O2 and metal ions in the genotoxicity of AA in cell culture models. Our preliminary results confirmed that physiological concentrations of AA were not toxic to confluent human fibroblasts, although they inhibited the proliferation of cells at low density. No inhibition was observed with ascorbic acid 2-phosphate (AA2P), a vitamin C derivative that remains stable in culture media. Furthermore, high concentrations of AA induced DNA strand breakage in a dose-dependent manner, whereas DHA and AA2P were not genotoxic. The genotoxic effect of AA was transient, required the formation of extracellular H2O2 and the presence of intracellular iron, but not of extracellular transition metal ions. These observations further clarify the pro-oxidant effect of AA solutions in cell culture models. The possibility that intravenous administration of high-dose AA may cause a similar genotoxic effect in vivo is discussed.


Asunto(s)
Ácido Ascórbico/toxicidad , Daño del ADN , Espacio Extracelular/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Metales/metabolismo , Mutágenos/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Espacio Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Elementos de Transición/metabolismo
17.
Redox Biol ; 11: 157-169, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27936457

RESUMEN

BACKGROUND AND AIMS: In hereditary hemochromatosis, iron deposition in the liver parenchyma may lead to fibrosis, cirrhosis and hepatocellular carcinoma. Most cases are ascribed to a common mutation in the HFE gene, but the extent of clinical expression is greatly influenced by the combined action of yet unidentified genetic and/or environmental modifying factors. In mice, transcription factor NRF2 is a critical determinant of hepatocyte viability during exposure to acute dietary iron overload. We evaluated if the genetic disruption of Nrf2 would prompt the development of liver damage in Hfe-/- mice (an established model of human HFE-hemochromatosis). METHODS: Wild-type, Nrf2-/-, Hfe-/- and double knockout (Hfe/Nrf2-/-) female mice on C57BL/6 genetic background were sacrificed at the age of 6 (young), 12-18 (middle-aged) or 24 months (old) for evaluation of liver pathology. RESULTS: Despite the parenchymal iron accumulation, Hfe-/- mice presented no liver injury. The combination of iron overload (Hfe-/-) and defective antioxidant defences (Nrf2-/-) increased the number of iron-related necroinflammatory lesions (sideronecrosis), possibly due to the accumulation of toxic oxidation products such as 4-hydroxy-2-nonenal-protein adducts. The engulfment of dead hepatocytes led to a gradual accumulation of iron within macrophages, featuring large aggregates. Myofibroblasts recruited towards the injury areas produced substantial amounts of collagen fibers involving the liver parenchyma of double-knockout animals with increased hepatic fibrosis in an age-dependent manner. CONCLUSIONS: The genetic disruption of Nrf2 promotes the transition from iron accumulation (siderosis) to liver injury in Hfe-/- mice, representing the first demonstration of spontaneous hepatic fibrosis in the long term in a mouse model of hereditary hemochromatosis displaying mildly elevated liver iron.


Asunto(s)
Proteína de la Hemocromatosis/genética , Hemocromatosis/genética , Cirrosis Hepática/genética , Factor 2 Relacionado con NF-E2/genética , Animales , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Hemocromatosis/metabolismo , Hemocromatosis/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hierro/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/patología , Ratones , Ratones Noqueados , Mutación
19.
Nat Commun ; 8(1): 403, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28864822

RESUMEN

Hepcidin regulates systemic iron homeostasis. Suppression of hepcidin expression occurs physiologically in iron deficiency and increased erythropoiesis but is pathologic in thalassemia and hemochromatosis. Here we show that epigenetic events govern hepcidin expression. Erythropoiesis and iron deficiency suppress hepcidin via erythroferrone-dependent and -independent mechanisms, respectively, in vivo, but both involve reversible loss of H3K9ac and H3K4me3 at the hepcidin locus. In vitro, pan-histone deacetylase inhibition elevates hepcidin expression, and in vivo maintains H3K9ac at hepcidin-associated chromatin and abrogates hepcidin suppression by erythropoietin, iron deficiency, thalassemia, and hemochromatosis. Histone deacetylase 3 and its cofactor NCOR1 regulate hepcidin; histone deacetylase 3 binds chromatin at the hepcidin locus, and histone deacetylase 3 knockdown counteracts hepcidin suppression induced either by erythroferrone or by inhibiting bone morphogenetic protein signaling. In iron deficient mice, the histone deacetylase 3 inhibitor RGFP966 increases hepcidin, and RNA sequencing confirms hepcidin is one of the genes most differentially regulated by this drug in vivo. We conclude that suppression of hepcidin expression involves epigenetic regulation by histone deacetylase 3.Hepcidin controls systemic iron levels by inhibiting intestinal iron absorption and iron recycling. Here, Pasricha et al. demonstrate that the hepcidin-chromatin locus displays HDAC3-mediated reversible epigenetic modifications during both erythropoiesis and iron deficiency.


Asunto(s)
Regulación de la Expresión Génica , Hepcidinas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Acetilación , Secuencias de Aminoácidos , Animales , Epigénesis Genética , Eritropoyetina/genética , Eritropoyetina/metabolismo , Hepcidinas/metabolismo , Histona Desacetilasas/genética , Histonas/química , Humanos , Deficiencias de Hierro , Masculino , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas
20.
Free Radic Biol Med ; 41(12): 1829-36, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17157185

RESUMEN

While ELISA is a frequently used means of assessing 8-oxo-7,8-dihydro-2-deoxyguanosine (8-oxodG) in biological fluids, differences in baseline urinary 8-oxodG levels, compared to chromatographic techniques, have raised questions regarding the specificity of immunoassays. Recently, ELISA of salivary 8-oxodG has been used to report on periodontal disease. We compared salivary 8-oxodG levels, determined by two commercial ELISA kits, to liquid chromatography-tandem mass spectrometry (LC-MS/MS) with prior purification using solid-phase extraction. While values were obtained with both ELISA kits, salivary 8-oxodG values were below or around the limit of detection of our LC-MS/MS assay. As the limit of detection for the LC-MS/MS procedure is much lower than ELISA, we concluded that the assessment of salivary 8-oxodG by ELISA is not accurate. In contrast to previous studies, ELISA levels of urinary 8-oxodG (1.67 +/- 0.53 pmol/mumol creatinine) were within the range reported previously only for chromatographic assays, although still significantly different than LC-MS/MS (0.41 +/- 0.39 pmol/mumol creatinine; p = 0.002). Furthermore, no correlation with LC-MS/MS was seen. These results question the ability of ELISA approaches, at present, to specifically determine absolute levels of 8-oxodG in saliva and urine. Ongoing investigation in our laboratories aims to identify the basis of the discrepancy between ELISA and LC-MS/MS.


Asunto(s)
Cromatografía Liquida/métodos , Desoxiguanosina/análogos & derivados , Ensayo de Inmunoadsorción Enzimática/métodos , Espectrometría de Masas/métodos , Saliva/química , 8-Hidroxi-2'-Desoxicoguanosina , Adulto , Desoxiguanosina/análisis , Desoxiguanosina/orina , Femenino , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA