Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cancer Cell ; 40(12): 1448-1453, 2022 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-36270276

RESUMEN

3D patient tumor avatars (3D-PTAs) hold promise for next-generation precision medicine. Here, we describe the benefits and challenges of 3D-PTA technologies and necessary future steps to realize their potential for clinical decision making. 3D-PTAs require standardization criteria and prospective trials to establish clinical benefits. Innovative trial designs that combine omics and 3D-PTA readouts may lead to more accurate clinical predictors, and an integrated platform that combines diagnostic and therapeutic development will accelerate new treatments for patients with refractory disease.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Neoplasias/diagnóstico , Medicina de Precisión , Estudios Prospectivos , Oncología Médica
2.
Semin Cancer Biol ; 76: 189-194, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33901632

RESUMEN

Iron is an essential metal for cellular metabolism. The reduced form of iron is a cofactor in numerous redox reactions in the cell and is therefore required for many vital physiological functions. Since iron is an oxidatively active metal, its homeostasis is tightly regulated in healthy cell. Most of iron exists in a protein-bound form, in erythrocytes as the heme compound hemoglobin, and in storage proteins such as ferritin, hemosiderin and myoglobin. Iron also is bound to proteins and non-heme enzymes involved in oxidation-reduction reactions and the transfer of electrons. There is no free iron inside the cell, however a small fraction of loosely bound iron is found in the cytoplasm. This poorly defined pool of ferrous iron is called labile iron pool. Under pathological conditions iron homeostasis may be disrupted at different levels including absorption, systemic transportation, and cellular uptake and storage. Cancer cells display dysregulated iron homeostasis and, for reasons yet poorly understood, require more iron for their metabolism and growth. As a result, in cancer cells labile iron pool is increased, and loosely bound iron catalyzes Fenton reaction and perhaps other reactions that generate reactive oxygen species. Oxygen-derived free radicals produce DNA mutations, damage proteins and lipids resulting in either cell death or cell transformation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Hierro/metabolismo , Neoplasias/metabolismo , Animales , Homeostasis/fisiología , Humanos , Neoplasias/patología
3.
Cell ; 171(2): 273-285, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28985560

RESUMEN

Ferroptosis is a form of regulated cell death characterized by the iron-dependent accumulation of lipid hydroperoxides to lethal levels. Emerging evidence suggests that ferroptosis represents an ancient vulnerability caused by the incorporation of polyunsaturated fatty acids into cellular membranes, and cells have developed complex systems that exploit and defend against this vulnerability in different contexts. The sensitivity to ferroptosis is tightly linked to numerous biological processes, including amino acid, iron, and polyunsaturated fatty acid metabolism, and the biosynthesis of glutathione, phospholipids, NADPH, and coenzyme Q10. Ferroptosis has been implicated in the pathological cell death associated with degenerative diseases (i.e., Alzheimer's, Huntington's, and Parkinson's diseases), carcinogenesis, stroke, intracerebral hemorrhage, traumatic brain injury, ischemia-reperfusion injury, and kidney degeneration in mammals and is also implicated in heat stress in plants. Ferroptosis may also have a tumor-suppressor function that could be harnessed for cancer therapy. This Primer reviews the mechanisms underlying ferroptosis, highlights connections to other areas of biology and medicine, and recommends tools and guidelines for studying this emerging form of regulated cell death.


Asunto(s)
Muerte Celular , Animales , Apoptosis , Humanos , Hierro/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
4.
Toxicol Appl Pharmacol ; 257(1): 32-7, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21878346

RESUMEN

The aim of this study was to test a hypothesis that ascorbate depletion could enhance carcinogenicity and acute toxicity of nickel. Homozygous L-gulono--lactone oxidase gene knock-out mice (Gulo-/- mice) unable to produce ascorbate and wild-type C57BL mice (WT mice) were injected intramuscularly with carcinogenic nickel subsulfide (Ni3S2), and observed for the development of injection site tumors for 57 weeks. Small pieces of one of the induced tumors were transplanted subcutaneously into separate groups of Gulo-/- and WT mice and the growth of these tumors was measured for up to 3 months. The two strains of mice differed significantly with regard to (1) Ni3S2 carcinogenesis: Gulo-/- mice were 40% more susceptible than WT mice; and (2) transplanted tumors development: Gulo-/- mice were more receptive to tumor growth than WT mice, but only in terms of a much shorter tumor latency; later in the exponential phase of growth, the growth rates were the same. And, with adequate ascorbate supplementation, the two strains were equally susceptible to acute toxicity of Ni3S2. Statistically significant effects of dietary ascorbate dosing levels were the following: (1) reduction in ascorbate supplementation increased acute toxicity of Ni3S2 in Gulo-/- mice; (2) ascorbate supplementation extended the latency of transplanted tumors in WT mice. In conclusion, the lack of endogenous ascorbate synthesis makes Gulo-/- mice more susceptible to Ni3S2 carcinogenesis. Dietary ascorbate tends to attenuate acute toxicity of Ni3S2 and to extend the latency of transplanted tumors. The latter effects may be of practical importance to humans and thus deserve further studies.


Asunto(s)
Ácido Ascórbico/farmacología , Carcinógenos/toxicidad , L-Gulonolactona Oxidasa/metabolismo , Trasplante de Neoplasias , Neoplasias Experimentales/metabolismo , Níquel/toxicidad , Animales , Ácido Ascórbico/metabolismo , Carcinógenos/administración & dosificación , Interacciones Farmacológicas , Inyecciones Intramusculares , L-Gulonolactona Oxidasa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/patología , Níquel/administración & dosificación
5.
Cancer Res ; 69(2): 616-24, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19147576

RESUMEN

Activation of hypoxia-inducible factors (HIF), responsible for tumor angiogenesis and glycolytic switch, is regulated by reduced oxygen availability. Normally, HIF-alpha proteins are maintained at low levels, controlled by site-specific hydroxylation carried out by HIF prolyl hydroxylases (PHD) and subsequent proteasomal degradation via the von Hippel-Lindau ubiquitin ligase. Using a yeast two-hybrid screen, we identified an interaction between melanoma antigen-11 (MAGE-11) cancer-testis antigen and the major HIF-alpha hydroxylating enzyme PHD2. The interaction was confirmed by a pull-down assay, coimmunoprecipitation, and colocalization in both normoxic and hypoxic conditions. Furthermore, MAGE-9, the closest homologue of MAGE-11, was also found to interact with PHD2. MAGE-11 inhibited PHD activity without affecting protein levels. This inhibition was accompanied by stabilization of ectopic or endogenous HIF-1alpha protein. Knockdown of MAGE-11 by small interfering RNA results in decreased hypoxic induction of HIF-1alpha and its target genes. Inhibition of PHD by MAGE-11, and following activation of HIFs, is a novel tumor-associated HIF regulatory mechanism. This finding provides new insights into the significance of MAGE expression in tumors and may provide valuable tools for therapeutic intervention because of the restricted expression of the MAGE gene family in cancers, but not in normal tissues.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Secuencia de Aminoácidos , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Datos de Secuencia Molecular , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , Transcripción Genética , Transfección , Ubiquitina/metabolismo
6.
Toxicol Sci ; 107(2): 394-403, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074761

RESUMEN

The exposure of cells to several metal ions stabilizes HIF-1 alpha protein. However, the molecular mechanisms are not completely understood. They may involve inhibition of hydroxylation by either substitution of iron by metal ions or by iron oxidation in the hydroxylases. Here we provide evidence supporting the latter mechanism. We show that HIF-1 alpha stabilization in human lung epithelial cells occurred following exposure to various metal and metalloid ions, including those that cannot substitute for iron in the hydroxylases. In each case addition of the reducing agent ascorbic acid (AA)* abolished HIF-1 alpha protein stabilization. To better understand the role of iron oxidation in hydroxylase inhibition and to define the role of AA in the enzyme recovery we applied molecular modeling techniques. Our results indicate that the energy required for iron substitution by Ni(II) in the enzyme is high and unlikely to be achieved in a biological system. Additionally, computer modeling allowed us to identify a tridentate coordination of AA with the enzyme-bound iron, which explains the specific demand for AA as the iron reductant. Thus, the stabilization of HIF-1 alpha by numerous metal ions that cannot substitute for iron in the enzyme, the alleviation of this effect by AA, and our computer modeling data support the hypothesis of iron oxidation in the hydroxylases following exposure to metal ions.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hierro/metabolismo , Metales/toxicidad , Oxigenasas de Función Mixta/metabolismo , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Western Blotting , Línea Celular , Cromatografía Líquida de Alta Presión , Genes Reporteros/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Indicadores y Reactivos , Cinética , Luciferasas/genética , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Molecular , Oxidación-Reducción , Estimulación Química
7.
Carcinogenesis ; 29(8): 1493-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18381358

RESUMEN

Hypoxia-inducible factor (HIF-1) regulates the expression of genes that facilitate tumor cell survival by making them more resistant to therapeutic intervention. Recent evidence suggests that the activation of other transcription factors, in cooperation with HIF-1 or acting alone, is involved in the upregulation of hypoxia-inducible genes. Here we report that high cell density, a condition that might mimic the physiologic situation in growing tumor and most probably representing nutritional starvation, upregulates hypoxia-inducible genes. This upregulation can occur in HIF-independent manner since hypoxia-inducible genes carbonic anhydrase 9 (CA9), lysyloxidase like 2 (LOXL2) and n-myc-down regulated 1 (NDRG1)/calcium activated protein (Cap43) can be upregulated by increased cell density under both normoxic and hypoxic conditions in both HIF-1 alpha-proficient and -deficient mouse fibroblasts. Moreover, cell density upregulates the same genes in 1HAEo- and A549 human lung epithelial cells. Searching for other transcription factors involved in the regulation of hypoxia-inducible genes by cell density, we focused our attention on ETS1. As reported previously, members of v-ets erythroblastosis virus E26 oncogene homolog (ETS) family transcription factors participate in the upregulation of hypoxia-inducible genes. Here, we provide evidence that ETS1 protein is upregulated at high cell density in both human and mouse cells. The involvement of ETS1 in the upregulation of hypoxia-inducible genes was further confirmed in a luciferase reporter assay using cotransfection of ETS1 expression vector with NDRG1/Cap43 promoter construct. The downregulation of ETS1 expression with small interfering RNA (siRNA) inhibited the upregulation of CA9 and NDRG1/Cap43 caused by increased cell density. Collectively, our data indicate the involvement of ETS1 along with HIF-1 in regulating hypoxia-inducible genes.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Factores de Transcripción/metabolismo , Antígenos de Neoplasias/genética , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica , Genes Reporteros , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Pulmón/fisiología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Neoplásico/genética , ARN Interferente Pequeño/genética , Transcripción Genética , Regulación hacia Arriba
8.
Biochem Biophys Res Commun ; 370(4): 536-40, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18359287

RESUMEN

Mutations in the VHL gene are associated with highly vascular tumors of kidney, brain, retina, and adrenal gland. The inability of the mutant VHL protein to destabilize HIF-1 plays a crucial role in malignant angiogenesis. VHL is also associated with ECM assembly but the molecular mechanisms of this activity remain unclear. We used expression arrays and cell lines with different VHL status to identify ECM-associated genes controlled by VHL. One of them, adhesion-associated TGFBI, was repressed by VHL and overexpressed in renal, gastrointestinal, brain, and other tumors. Analyzing the mechanism of TGFBI up-regulation in clear cell carcinoma, we identified a novel VHL target, a Kruppel-like transcriptional factor 10 (KLF10). The TGFBI promoter, which we isolated and studied in Luc-reporter assay, was induced by KLF10 but not hypoxia. These data provide the molecular basis for the observed VHL effect on TGFBI and stimulate further research into the KLF10 and TGFBI roles in cancer.


Asunto(s)
Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Proteínas de la Matriz Extracelular/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias/genética , Factor de Crecimiento Transformador beta/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Antineoplásicos/farmacología , Carcinoma de Células Renales/genética , Adhesión Celular/genética , Humanos , Neoplasias Renales/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
9.
Int J Cancer ; 122(6): 1435-40, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17999410

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) plays an important role in stress-responsive gene expression. Although primarily sensitive to hypoxia, HIF-1 signaling can be regulated by a number of stress factors including metabolic stress, growth factors and molecules present in the extracellular matrix (ECM). Degradation of ECM by metalloproteinases (MMP) is important for tumor progression, invasion and metastasis. ECM is predominantly collagen, and the imino acids (Pro and HyPro) comprise 25% of collagen residues. The final step in collagen degradation is catalyzed by prolidase, the obligate peptidase for imidodipeptides with Pro and HyPro in the carboxyl terminus. Defective wound healing in patients with inherited prolidase deficiency is associated with histologic features of angiopathy suggesting that prolidase may play a role in angiogenesis. Because HIF-1 alpha is central to angiogenesis, we considered that prolidase may modulate this pathway. To test this hypothesis, we made expression constructs of human prolidase and obtained stable transfectants in colorectal cancer cells (RKO). Overexpression of prolidase resulted in increased nuclear hypoxia inducible factor (HIF-1 alpha) levels and elevated expression of HIF-1-dependent gene products, vascular endothelial growth factor (VEGF) and glucose transporter-1 (Glut-1). The activation of HIF-1-dependent transcription was shown by prolidase-dependent activation of hypoxia response element (HRE)-luciferase expression. We used an oxygen-dependent degradation domain (ODD)-luciferase reporter construct as a surrogate for HIF-1 alpha as an in situ prolyl-hydroxylase assay. Since this reporter is degraded by VHL-dependent mechanisms, the increased levels of luciferase observed with prolidase expression reflected the decreased HIF-1 alpha prolyl hydroxylase activity. Additionally, the differential expression of prolidase in 2 breast cancer cell lines showed prolidase-dependent differences in HIF-1 alpha levels. These findings show that metabolism of imidodipeptides by prolidase plays a previously unrecognized role in angiogenic signaling.


Asunto(s)
Dipeptidasas/metabolismo , Matriz Extracelular/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Transducción de Señal , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Humanos , Hidrólisis
10.
Chem Res Toxicol ; 21(1): 28-44, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17970581

RESUMEN

Chronic exposure to nickel(II), chromium(VI), or inorganic arsenic (iAs) has long been known to increase cancer incidence among affected individuals. Recent epidemiological studies have found that carcinogenic risks associated with chromate and iAs exposures were substantially higher than previously thought, which led to major revisions of the federal standards regulating ambient and drinking water levels. Genotoxic effects of Cr(VI) and iAs are strongly influenced by their intracellular metabolism, which creates several reactive intermediates and byproducts. Toxic metals are capable of potent and surprisingly selective activation of stress-signaling pathways, which are known to contribute to the development of human cancers. Depending on the metal, ascorbate (vitamin C) has been found to act either as a strong enhancer or suppressor of toxic responses in human cells. In addition to genetic damage via both oxidative and nonoxidative (DNA adducts) mechanisms, metals can also cause significant changes in DNA methylation and histone modifications, leading to epigenetic silencing or reactivation of gene expression. In vitro genotoxicity experiments and recent animal carcinogenicity studies provided strong support for the idea that metals can act as cocarcinogens in combination with nonmetal carcinogens. Cocarcinogenic and comutagenic effects of metals are likely to stem from their ability to interfere with DNA repair processes. Overall, metal carcinogenesis appears to require the formation of specific metal complexes, chromosomal damage, and activation of signal transduction pathways promoting survival and expansion of genetically/epigenetically altered cells.


Asunto(s)
Arsénico/toxicidad , Carcinógenos/toxicidad , Cromo/toxicidad , Cocarcinogénesis , Metales/toxicidad , Neoplasias/inducido químicamente , Neoplasias/genética , Níquel/toxicidad , Animales , ADN/genética , Humanos
11.
Free Radic Biol Med ; 42(8): 1246-57, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17382205

RESUMEN

Molecular oxygen is involved in hydroxylation and subsequent degradation of HIF-1alpha, a subunit of HIF-1 transcription factor; therefore oxygen shortage (hypoxia) stabilizes this protein. However, HIF-1alpha can also be stabilized by transition metal ions in the presence of oxygen, suggesting that a different mechanism is involved in metal-induced hypoxic stress. Recently, we showed that the depletion of intracellular ascorbate by metals may lead to the inhibition of hydroxylases. Because nickel(II) has similarity to iron(II), an alternative hypothesis suggests that iron substitution for nickel in the enzyme inhibits hydroxylase activity. Here we investigated the induction of HIF-1 by another metal, chromium, which cannot replace iron in the enzyme. We show that chromium(VI), but not chromium(III), can oxidize ascorbate both in cells and in a cell-free system. In agreement with these data chromium(VI) stabilizes HIF-1alpha protein in cells only until it is reduced to chromium(III). In contrast, nickel(II) was found to be a catalyst, which facilitated continuous oxidation of ascorbate by ambient oxygen. These data correlate with extended stabilization of HIF-1alpha after acute exposure to nickel(II). The HIF-1-dependent reporter assays revealed that 20-24 h was required to fully develop the HIF-1 transcriptional response, and the acute exposure to nickel(II), but not chromium(VI), meets this requirement. However, repeated (chronic) exposure to chromium(VI) can also lead to extended stabilization of HIF-1alpha. Thus, the obtained data emphasize the important role of ascorbate in regulation of HIF-1 transcriptional activity in metal-exposed human lung cells.


Asunto(s)
Cromo/farmacología , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Níquel/farmacología , Mucosa Respiratoria/fisiología , Transcripción Genética/efectos de los fármacos , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Transporte Biológico , Línea Celular , Cromo/farmacocinética , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Cinética , Pulmón , Níquel/farmacocinética , Mucosa Respiratoria/efectos de los fármacos
12.
J Toxicol Environ Health A ; 69(10): 935-51, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16728372

RESUMEN

It was hypothesized that relative mass relationships among select constituent metals and iron (Fe3+) govern the pulmonary immunotoxic potential of any PM(2.5) sample, as these determine the extent to which Fe3+ binding by transferrin is affected (resulting in altered alveolar macrophage [AM] Fe status and subsequent antibacterial function). Iron response protein (IRP) binding activity is a useful indirect measurement of changes in Fe status, as reductions in cell Fe levels lead to increases in IRP binding. However, AM IRP activity can be affected by an increased presence of nitric oxide generated by inducible nitric oxide synthase (iNOS). This study sought to determine if any changes in AM IRP activity induced by PM(2.5) constituents V, Mn, or Al were independent from effects of the metals on cell NO formation. NR8383 rat AM were exposed to Fe3+ alone or combined with V, Mn, or Al at metal:Fe ratios representative of those in PM(2.5) collected in New York City, Los Angeles, and Seattle during fall 2001. Cells were then assessed for changes in IRP activity and iNOS expression. Phosphorylated extracellular signal-regulated kinase (ERK) 1 and 2 levels were also measured since activated ERKs are involved in signaling pathways that lead to increased iNOS expression. The results indicate that V and Al, and to a lesser extent Mn, altered IRP activity, though the effects were not consistently concentration dependent. Furthermore, while V and Mn treatments did not induce iNOS expression, Al did. These results confirmed our hypothesis that certain metals associated with PM(2.5) might alter the pulmonary immunocompetence of exposed hosts by affecting the Fe status of AM, a major class of deep lung defense cells.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Hierro/metabolismo , Macrófagos Alveolares/metabolismo , Metales/efectos adversos , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Contaminantes Atmosféricos/efectos adversos , Aluminio/efectos adversos , Animales , Células Cultivadas , Homeostasis , Inmunocompetencia/fisiología , Iones/efectos adversos , Hierro/efectos adversos , Proteínas Reguladoras del Hierro/metabolismo , Macrófagos Alveolares/fisiología , Manganeso/efectos adversos , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Fosforilación , Ratas , Transducción de Señal , Transferrina/metabolismo , Vanadio/efectos adversos
13.
J Phys Chem A ; 110(12): 4223-8, 2006 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-16553373

RESUMEN

Enzymatic hydroxylation reactions carried out by 2-oxoglutarate (2OG) dependent iron-containing oxygenases were recently implicated in oxygen sensing. In addition to oxygen depletion, two metals, cobalt and nickel, are capable of inducing hypoxic stress in cells by inhibiting oxygenase activity. Two possible scenarios have been proposed for the explanation of the hypoxic effects of cobalt and nickel: oxidation of enzyme-bound iron following cobalt or nickel exposure, and substitution of iron by cobalt or nickel. Here, by using density functional theory calculations, we modeled the reaction route from the reaction components to the high-spin metal-oxide intermediate in the activation of oxygen molecule by 2OG-dependent enzymes for three metal ions Fe(II), Ni(II), and Co(II) in the active site. An initial molecular model was constructed based on the crystal structure of iron-containing asparaginyl hydroxylase (FIH-1). Nickel- and cobalt-containing enzymes were modeled by a consequent replacement of the iron in the active center. The energy profiles connecting stationary points on the potential surfaces were computed by using the intrinsic reaction coordinate (IRC) technique from the located transition states. The results of calculations show that the substitution of iron by nickel or cobalt modifies the reaction energy profile; however, qualitatively, the reaction mechanism remains essentially the same. Thus, we would postulate that if the iron ion in the active site were substitutable by nickel and/or cobalt ions enzyme activity would be considerably altered due to high activation barriers.


Asunto(s)
Cobalto/química , Ácidos Cetoglutáricos/química , Modelos Químicos , Níquel/química , Oxigenasas/química , Teoría Cuántica , Sitios de Unión , Cobalto/farmacología , Activación Enzimática/efectos de los fármacos , Hierro/química , Níquel/farmacología , Oxígeno/química , Oxigenasas/antagonistas & inhibidores , Relación Estructura-Actividad
14.
J Cell Biochem ; 97(5): 1025-35, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16288478

RESUMEN

Exposure of human lung cells to carcinogenic nickel compounds in the presence of oxygen up-regulated carbonic anhydrase IX (CA IX) and NDRG1/Cap43, both known as intrinsic hypoxia markers and cancer-associated genes. This suggests that factors other than a shortage of oxygen may be involved in this induction. Both proteins can also be induced in the presence of oxygen by culturing these cells to a high density without medium change. The intracellular ascorbate measurements revealed its rapid depletion in both metal- and density-exposed cells. Nickel exposure caused strong activation of HIF-1alpha and HIF-2alpha proteins, underscoring activation of HIF-1-dependent transcription. In contrast, cell density-dependent transcription was characterized by minor induction of HIF-1alpha or HIF-2alpha. Moreover, the up-regulation of NDRG1/Cap43 in HIF-1alpha deficient fibroblasts suggested the involvement of different transcription factor(s). The repletion of intracellular ascorbate reversed the induction of CA IX and NDRG1/Cap43 caused by cell density or nickel exposure. Thus, the loss of intracellular ascorbate triggered the induction of both tumor markers by two different conditions in the presence of oxygen. Ascorbate is delivered to lung cells via the SVCT2 ascorbate transporter, which was found to be sensitive to nickel or cell density. Collectively these findings establish the importance of intracellular ascorbate levels for the regulation of expression of CA IX and NDRG1/Cap43. We suggest, that, in addition to low oxygenation, insufficient supply of ascorbate or its excessive oxidation in tumors, can contribute to the induction of hypoxia-associated proteins via both HIF-dependent and independent mechanisms.


Asunto(s)
Ácido Ascórbico/farmacología , Recuento de Células , Níquel/farmacología , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Antígenos de Neoplasias/metabolismo , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/metabolismo , Proteínas de Ciclo Celular , Hipoxia de la Célula , Relación Dosis-Respuesta a Droga , Expresión Génica , Regulación de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Hierro/farmacología , Ácido Láctico/farmacología , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Proteínas/metabolismo , Transportadores de Sodio Acoplados a la Vitamina C , Simportadores/metabolismo , Transcripción Genética , Transfección
15.
Environ Health Perspect ; 113(5): 577-84, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15866766

RESUMEN

Nickel compounds are known to cause respiratory cancer in humans and induce tumors in experimental animals. The underlying molecular mechanisms may involve genotoxic effects; however, the data from different research groups are not easy to reconcile. Here, we challenge the common premise that direct genotoxic effects are central to nickel carcinogenesis and probably to that of other metals. Instead, we propose that it is formation of metal complexes with proteins and other molecules that changes cellular homeostasis and provides conditions for selection of cells with transformed phenotype. This is concordant with the major requirement for nickel carcinogenicity, which is prolonged action on the target tissue. If DNA is not the main nickel target, is there another unique molecule that can be attacked with carcinogenic consequences? Our recent observations indicate that ascorbate may be such a molecule. Nickel depletes intracellular ascorbate, which leads to the inhibition of cellular hydroxylases, manifested by the loss of hypoxia-inducible factor (HIF)-1alpha and -2alpha hydroxylation and hypoxia-like stress. Proline hydroxylation is crucial for collagen and extracellular matrix assembly as well as for assembly of other protein molecules that have collagen-like domains, including surfactants and complement. Thus, the depletion of ascorbate by chronic exposure to nickel could be deleterious for lung cells and may lead to lung cancer. Key words: ascorbate, carcinogenesis, collagens, extracellular matrix, hypoxia-inducible transcription factor, metals, nickel, protein hydroxylation.


Asunto(s)
Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Transformación Celular Neoplásica , Neoplasias Pulmonares/inducido químicamente , Níquel/toxicidad , Animales , Daño del ADN , Homeostasis , Humanos , Hidroxilación , Neoplasias Pulmonares/fisiopatología , Níquel/metabolismo , Prolina/metabolismo , Unión Proteica , Ratas
16.
Toxicol Appl Pharmacol ; 205(1): 1-10, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15885260

RESUMEN

B200 cells are Ni(II)-transformed mouse BALB/c-3T3 fibroblasts displaying a malignant phenotype and increased resistance to Ni(II) toxicity. In an attempt to find genes whose expression has been altered by the transformation, the Atlas Mouse Stress/Toxicology cDNA Expression Array (Clontech Laboratories, Inc., Palo Alto, CA) was used to analyze the levels of gene expression in both parental and Ni(II)-transformed cells. Comparison of the results revealed a significant up- or downregulation of the expression of 62 of the 588 genes present in the array (approximately 10.5%) in B200 cells. These genes were assigned to different functional groups, including transcription factors and oncogenes (9/14; fractions in parentheses denote the number of up-regulated versus the total number of genes assigned to this group), stress and DNA damage response genes (11/12), growth factors and hormone receptors (6/9), metabolism (7/7), cell adhesion (2/7), cell cycle (3/6), apoptosis (3/4), and cell proliferation (2/3). Among those genes, overexpression of beta-catenin and its downstream targets c-myc and cyclin D1, together with upregulated cyclin G, points at the malignant character of B200 cells. While the increased expression of glutathione (GSH) synthetase, glutathione-S-transferase A4 (GSTA4), and glutathione-S-transferase theta (GSTT), together with high level of several genes responding to oxidative stress, suggests the enforcement of antioxidant defenses in Ni-transformed cells.


Asunto(s)
Fibroblastos/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Análisis por Micromatrices/métodos , Níquel/efectos adversos , Fenotipo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina G , Ciclina G1 , Ciclinas/efectos de los fármacos , Ciclinas/genética , Ciclinas/metabolismo , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/farmacología , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Fibroblastos/patología , Fibroblastos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Genes bcl-1/efectos de los fármacos , Genes bcl-1/fisiología , Genes cdc/efectos de los fármacos , Genes myc/efectos de los fármacos , Genes myc/fisiología , Glutatión/genética , Glutatión/metabolismo , Glutatión Sintasa/efectos de los fármacos , Glutatión Sintasa/genética , Glutatión Sintasa/metabolismo , Glutatión Transferasa/efectos de los fármacos , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Sustancias de Crecimiento/genética , Sustancias de Crecimiento/metabolismo , Isoenzimas , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Análisis por Micromatrices/tendencias , Oncogenes/efectos de los fármacos , Oncogenes/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Prostaglandina-Endoperóxido Sintasas/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/genética
17.
Lab Invest ; 85(3): 328-41, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15716863

RESUMEN

Invasion into surrounding brain tissue is a fundamental feature of gliomas and the major reason for treatment failure. The process of brain invasion in gliomas is not well understood. Differences in gene expression and/or gene products between invading and noninvading glioma cells may identify potential targets for new therapies. To look for genes associated with glioma invasion, we first employed Affymetrix microarray Genechip technology to identify genes differentially expressed in migrating glioma cells in vitro and in invading glioma cells in vivo using laser capture microdissection. We observed upregulation of a variety of genes, previously reported to be linked to glioma cell migration and invasion. Remarkably, major histocompatiblity complex (MHC) class I and II genes were significantly downregulated in migrating cells in vitro and in invading cells in vivo. Decreased MHC expression was confirmed in migrating glioma cells in vitro using RT-PCR and in invading glioma cells in vivo by immunohistochemical staining of human and murine glioblastomas for beta2 microglobulin, a marker of MHC class I protein expression. To the best of our knowledge, this report is the first to describe the downregulation of MHC class I and II antigens in migrating and invading glioma cells, in vitro and in vivo, respectively. These results suggest that the very process of tumor invasion is associated with decreased expression of MHC antigens allowing glioma cells to invade the surrounding brain in a 'stealth'-like manner.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase I/genética , Animales , Neoplasias Encefálicas/inmunología , Regulación hacia Abajo , Fibronectinas/metabolismo , Perfilación de la Expresión Génica , Glioma/inmunología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Humanos , Ratones , Invasividad Neoplásica , ARN/metabolismo , Células Tumorales Cultivadas
18.
J Biol Chem ; 279(39): 40337-44, 2004 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-15271983

RESUMEN

Exposure of cells to carcinogenic compounds of nickel(II) and cobalt(II) causes activation of the HIF-1 transcription factor and up-regulates a battery of hypoxia-inducible genes. However, the mechanism of HIF-1 activation by these metals is not known. It was shown recently that hydroxylation of prolines in the HIFalpha subunit of HIF-1 is required for its binding with the von Hippel-Lindau tumor suppressor protein and the subsequent proteasomal destruction. Here we show that responsible prolyl hydroxylases are targets for both nickel(II) and cobalt(II) because degradation of a reporter protein containing the oxygen-dependent degradation domain (Pro-402/564) of HIFalpha was abolished in a von Hippel-Lindau-dependent manner in cells exposed to nickel(II) or cobalt(II). The enzymatic activity of prolyl hydroxylases depends on iron as the activating metal, 2-oxoglutarate as a co-substrate, and ascorbic acid as a cofactor. Hydroxylase activity can be impaired by the depletion of any of these factors. We found that exposure of cells to nickel(II) or cobalt(II) did not affect the level of intracellular iron. Instead, nickel(II) or cobalt(II) exposure greatly depleted intracellular ascorbate. Co-exposure of cells to metals and ascorbate resulted in the increase of intracellular ascorbate and reversed both metal-induced stabilization of HIF-1alpha and HIF-1-dependent gene transcription. Because ascorbate is essential for maintaining iron in prolyl hydroxylases in the active iron(II) state, we suggest that the observed depletion of ascorbate by nickel(II) or cobalt(II) favors iron oxidation and thus inactivation of the enzyme.


Asunto(s)
Ácido Ascórbico/metabolismo , Carcinógenos , Cobalto/química , Hipoxia , Níquel/química , Factores de Transcripción , Western Blotting , Línea Celular , Cromatografía Líquida de Alta Presión , Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Genes Reporteros , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Hierro/metabolismo , Ácidos Cetoglutáricos/metabolismo , Modelos Biológicos , Complejos Multienzimáticos/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Oxígeno/metabolismo , Plásmidos/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Prolina/química , Complejo de la Endopetidasa Proteasomal , Estructura Terciaria de Proteína , Factores de Tiempo , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
19.
Toxicol Appl Pharmacol ; 196(2): 258-65, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15081272

RESUMEN

Exposure to ambient air particulate matter (PM) is associated with increased mortality and morbidity in susceptible populations. The epidemiological data also suggest a relationship between PM air pollution and impairment of cardiopulmonary function. The mechanisms that may be responsible for these effects are not fully understood and are likely related to perturbations of cellular and molecular functions. One type of PM, residual oil fly ash (ROFA), is of particular interest. ROFA does not contain much organic material, but does contain relatively high quantities of transition metals, predominantly nickel, vanadium, and iron, as well as black carbon and sulfates. In this study, we investigated the effect of two metals (iron and nickel) on the induction of "hypoxia-like" stress and the production of interleukins (ILs) in minimally transformed human airway epithelial cells (1HAEo(-)). We found that exposure to soluble nickel sulfate results in the induction of hypoxia-inducible genes and IL-8 production by the 1HAEo(-) cells. The simultaneous addition of iron in either ferric or ferrous form and nickel completely inhibited IL-8 production and had no effect on "hypoxia-like" stress caused by nickel, suggesting the existence of two different pathways for the induction "hypoxia-like" stress and IL-8 production. The effect of nickel was not related to the blocking of iron entry into cells since the level of intracellular iron was not affected by co-exposure with nickel. The obtained data indicate that nickel can induce different signaling pathways with or without interference with iron metabolism. Our observations suggest that in some cases the excess of iron in PM can cancel the effects of nickel.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Carbono/toxicidad , Compuestos Férricos/toxicidad , Pulmón/efectos de los fármacos , Níquel/toxicidad , Western Blotting , Proteínas de Ciclo Celular/biosíntesis , Cloruros , Ceniza del Carbón , Deferoxamina/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Compuestos Férricos/farmacocinética , Humanos , Hipoxia/inducido químicamente , Hipoxia/patología , Interleucina-8/antagonistas & inhibidores , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Quelantes del Hierro/farmacología , Pulmón/metabolismo , Material Particulado
20.
Mol Cell Biochem ; 255(1-2): 195-202, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14971660

RESUMEN

Nickel compounds are carcinogenic and induce malignant transformation of cultured cells. Since nickel has low mutagenic potential, it may act predominantly through epigenetic mechanisms, including down-regulation of tumor suppressor genes. FHIT is a tumor suppressor gene whose expression is frequently reduced or lost in tumors and pre-malignant lesions. Previously, we have shown that the phosphohydrolase activity of Fhit protein, associated with its tumor suppressor action, is inhibited by nickel. In cells, such effect would assist in carcinogenesis. The latter could be further enhanced if nickel also lowered cellular levels of Fhit protein itself, e.g. by down-regulation of FHIT gene. To test this possibility, we determined Fhit protein and Fhit-mRNA levels in a nickel-transformed mouse cell line and in nickel-induced murine sarcomas. In B200 cells, derived by nickel treatment of BALB/c-3T3 cells and exhibiting a malignant phenotype, Fhit protein levels were 50% of those in the parental cells, while Fhit-mRNA expression remained unchanged. A decrease of up to > 90% in Fhit protein levels was also observed in 22 local sarcomas (mostly fibrosarcomas) induced by i.m. injection of nickel subsulfide in C57BL/6 and MT+ (C57BL/6 overexpressing metallothionein) mice, as compared with normal muscles. Moreover, Fhit was absent in 3 out of 10 sarcomas from MT+ mice and in 1 of 12 sarcomas from C57BL/6 mice. The lack of Fhit protein coincided with the absence of the Fhit-mRNA transcript in these tumors. However, in the other tumors, the decreased Fhit levels were not always accompanied by reduced expression of Fhit-mRNA. Thus, the observed lowering of Fhit protein levels is mostly associated with changes in mRNA expression and protein translation or turnover rates, and rarely with a full silencing of the gene itself. Overall, the decline of Fhit in cells or tissues malignantly transformed by nickel may indicate possible involvement of this effect in the mechanisms of nickel carcinogenesis.


Asunto(s)
Ácido Anhídrido Hidrolasas/biosíntesis , Transformación Celular Neoplásica/inducido químicamente , Neoplasias de los Músculos/inducido químicamente , Proteínas de Neoplasias/biosíntesis , Níquel/toxicidad , Sarcoma Experimental/inducido químicamente , Ácido Anhídrido Hidrolasas/genética , Animales , Línea Celular Transformada , Transformación Celular Neoplásica/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...