Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 171(2): 273-285, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28985560

RESUMO

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.


Assuntos
Morte Celular , Animais , Apoptose , Humanos , Ferro/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
2.
Semin Cancer Biol ; 76: 189-194, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33901632

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/metabolismo , Ferro/metabolismo , Neoplasias/metabolismo , Animais , Homeostase/fisiologia , Humanos , Neoplasias/patologia
3.
Cancer Cell ; 40(12): 1448-1453, 2022 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-36270276

RESUMO

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.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/terapia , Neoplasias/diagnóstico , Medicina de Precisão , Estudos Prospectivos , Oncologia
4.
Toxicol Appl Pharmacol ; 257(1): 32-7, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21878346

RESUMO

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.


Assuntos
Ácido Ascórbico/farmacologia , Carcinógenos/toxicidade , L-Gulonolactona Oxidase/metabolismo , Transplante de Neoplasias , Neoplasias Experimentais/metabolismo , Níquel/toxicidade , Animais , Ácido Ascórbico/metabolismo , Carcinógenos/administração & dosagem , Interações Medicamentosas , Injeções Intramusculares , L-Gulonolactona Oxidase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/patologia , Níquel/administração & dosagem
5.
Carcinogenesis ; 29(8): 1493-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18381358

RESUMO

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.


Assuntos
Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteína Proto-Oncogênica c-ets-1/metabolismo , Fatores de Transcrição/metabolismo , Antígenos de Neoplasias/genética , Anidrase Carbônica IX , Anidrases Carbônicas/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Genes Reporter , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Pulmão/fisiologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Neoplásico/genética , RNA Interferente Pequeno/genética , Transcrição Gênica , Regulação para Cima
6.
Int J Cancer ; 122(6): 1435-40, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17999410

RESUMO

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.


Assuntos
Dipeptidases/metabolismo , Matriz Extracelular/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Transdução de Sinais , Western Blotting , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos , Hidrólise
7.
Biochem Biophys Res Commun ; 370(4): 536-40, 2008 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-18359287

RESUMO

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.


Assuntos
Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Proteínas da Matriz Extracelular/genética , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/genética , Neoplasias/genética , Fator de Crescimento Transformador beta/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Antineoplásicos/farmacologia , Carcinoma de Células Renais/genética , Adesão Celular/genética , Humanos , Neoplasias Renais/genética , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima
8.
Free Radic Biol Med ; 42(8): 1246-57, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17382205

RESUMO

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.


Assuntos
Cromo/farmacologia , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Níquel/farmacologia , Mucosa Respiratória/fisiologia , Transcrição Gênica/efeitos dos fármacos , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Transporte Biológico , Linhagem Celular , Cromo/farmacocinética , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Cinética , Pulmão , Níquel/farmacocinética , Mucosa Respiratória/efeitos dos fármacos
9.
Mol Cell Biol ; 22(6): 1734-41, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11865053

RESUMO

Hypoxia causes the accumulation of the transcription factor hypoxia-inducible factor 1 (HIF-1), culminating in the expression of hypoxia-inducible genes such as those for vascular endothelial growth factor (VEGF) and NDRG-1/Cap43. Previously, we have demonstrated that intracellular calcium (Ca(2+)) is required for the expression of hypoxia-inducible genes. Here we found that, unlike with hypoxia or hypoxia-mimicking conditions, the elevation of intracellular Ca(2+) neither induced the HIF-1alpha protein nor stimulated HIF-1-dependent transcription. Furthermore, the elevation of intracellular Ca(2+) induced NDRG-1/Cap43 mRNA in HIF-1alpha-deficient cells. It also increased levels of c-Jun protein, causing its phosphorylation. The protein kinase inhibitor K252a abolished c-Jun induction and activator protein 1 (AP-1)-dependent reporter expression caused by Ca(2+) ionophore or hypoxia. K252a also significantly decreased hypoxia-induced VEGF and NDRG-1/Cap43 gene expression in both human and mouse cells. Using a set of deletion VEGF-Luc promoter constructs, we found that both HIF-1 and two AP-1 sites contribute to hypoxia-mediated induction of transcription. In contrast, only AP-1 sites contributed to Ca(2+)-mediated VEGF-Luc induction. A dominant-negative AP-1 prevented Ca(2+)-dependent transcription and partially impaired hypoxia-mediated transcription. In addition, dominant-negative AP-1 diminished the expression of the NDRG-1/Cap43 gene following hypoxia. We conclude that during hypoxia, an increase in intracellular Ca(2+) activates a HIF-1-independent signaling pathway that involves AP-1-dependent transcription. Cooperation between the HIF-1 and AP-1 pathways allows fine regulation of gene expression during hypoxia.


Assuntos
Cálcio/metabolismo , Hipóxia Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Cálcio/farmacologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Fatores de Crescimento Endotelial/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Dominantes , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Líquido Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Ionóforos/farmacologia , Linfocinas/genética , Camundongos , Fatores de Transcrição NFATC , Regiões Promotoras Genéticas/fisiologia , Fator de Transcrição AP-1/genética , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
10.
J Toxicol Environ Health A ; 69(10): 935-51, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16728372

RESUMO

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.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/biossíntese , Ferro/metabolismo , Macrófagos Alveolares/metabolismo , Metais/efeitos adversos , Óxido Nítrico Sintase Tipo II/biossíntese , Poluentes Atmosféricos/efeitos adversos , Alumínio/efeitos adversos , Animais , Células Cultivadas , Homeostase , Imunocompetência/fisiologia , Íons/efeitos adversos , Ferro/efeitos adversos , Proteínas Reguladoras de Ferro/metabolismo , Macrófagos Alveolares/fisiologia , Manganês/efeitos adversos , Proteína Quinase 1 Ativada por Mitógeno/biossíntese , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Fosforilação , Ratos , Transdução de Sinais , Transferrina/metabolismo , Vanádio/efeitos adversos
11.
Cancer Res ; 63(13): 3524-30, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12839937

RESUMO

Nickel is a potent environmental pollutant in industrial countries. Because nickel compounds are carcinogenic, exposure to nickel represents a serious hazard to human health. The understanding of how nickel exerts its toxic and carcinogenic effects at a molecular level may be important in risk assessment, as well as in the treatment and prevention of occupational diseases. Previously, using human and rodent cells in vitro, we showed that hypoxia-inducible signaling pathway was activated by carcinogenic nickel compounds. Acute exposure to nickel resulted in the accumulation of hypoxia-inducible transcription factor (HIF)-1, which strongly activated hypoxia-inducible genes, including the recently discovered tumor marker NDRG1 (Cap43). To further identify HIF-1-dependent nickel-inducible genes and to understand the role of the HIF-dependent signaling pathway in nickel-induced transformation, we used the Affymetrix GeneChip to compare the gene expression profiles in wild-type cells or in cells from HIF-1 alpha knockout mouse embryos exposed to nickel chloride. As expected, when we examined 12,000 genes for expression changes, we found that genes coding for glycolytic enzymes and glucose transporters, known to be regulated by HIF-1 transcription factor, were induced by nickel only in HIF-1 alpha-proficient cells. In addition, we found a number of other hypoxia-inducible genes up-regulated by nickel in a HIF-dependent manner including BCL-2-binding protein Nip3, EGLN1, hypoxia-inducible gene 1 (HIG1), and prolyl 4-hydroxylase. Additionally, we found a number of genes induced by nickel in a HIF-independent manner, suggesting that Ni activated other signaling pathways besides HIF-1. Finally, we found that in HIF-1 alpha knockout cells, nickel strongly induced the expression of the whole group of genes that were not expressed in the presence of HIF-1. Because the majority of modulated genes were induced or suppressed by nickel in a HIF-1-dependent manner, we elucidated the role of HIF-1 transcription factor in cell transformation. In HIF-1 alpha-proficient cells, nickel exposure increased soft agar growth, whereas it decreased soft agar growth in HIF-1 alpha-deficient cells. We hypothesize that the induction of HIF-1 transcription factor by nickel may be important during the nickel-induced carcinogenic process.


Assuntos
Carcinógenos/toxicidade , Proteínas de Ligação a DNA/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Níquel/toxicidade , Proteínas Nucleares/fisiologia , Fatores de Transcrição , Animais , Linhagem Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Sequências Hélice-Alça-Hélice , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos
12.
Environ Health Perspect ; 113(5): 577-84, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15866766

RESUMO

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.


Assuntos
Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Transformação Celular Neoplásica , Neoplasias Pulmonares/induzido quimicamente , Níquel/toxicidade , Animais , Dano ao DNA , Homeostase , Humanos , Hidroxilação , Neoplasias Pulmonares/fisiopatologia , Níquel/metabolismo , Prolina/metabolismo , Ligação Proteica , Ratos
13.
Clin Cancer Res ; 10(1 Pt 1): 222-7, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14734473

RESUMO

PURPOSE: The protein encoded by N-myc downstream-regulated gene 1 (NDRG1) is a recently discovered protein whose transcription is induced by androgens and hypoxia. We hypothesized that NDRG1 expression patterns might reveal a biological basis for the disparity of clinical outcome of prostate cancer patients with different ethnic backgrounds. EXPERIMENTAL DESIGN: Patients who underwent radical prostatectomy between 1990 and 2000 at Veterans Administration Medical Center of New York were examined. We studied 223 cases, including 157 African Americans and 66 Caucasians (T2, n = 144; >/=T3, n = 79; Gleason <7, n = 122; >/=7, n = 101). Three patterns of NDRG1 expression were identified in prostate cancer: (a) intense, predominately membranous staining similar to benign prostatic epithelium; (b) intense, nucleocytoplasmic localization; and (c) low or undetectable expression. We then examined the correlations between patients' clinicopathological parameters and different NDRG1 expression patterns. RESULTS: In this study of patients with equal access to care, African-American ethnic origin was an independent predictor of prostate-specific antigen recurrence (P < 0.05). We also observed a significant correlation between different patterns of NDRG1 expression and ethnic origin. Pattern 2 was less frequent in African Americans (21% versus 38%), whereas the reverse was observed for pattern 3 (60% in African Americans versus 44% in Caucasians; P = 0.03). This association remained significant after controlling for both grade and stage simultaneously (P = 0.02). CONCLUSIONS: Our data suggest that different NDRG1 expression patterns reflect differences in the response of prostatic epithelium to hypoxia and androgens in African-American compared with Caucasian patients. Further studies are needed to determine the contribution of NDRG1 to the disparity in clinical outcome observed between the two groups.


Assuntos
Negro ou Afro-Americano/etnologia , Proteínas de Ciclo Celular/metabolismo , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo , População Branca/etnologia , Idoso , Idoso de 80 Anos ou mais , Membrana Celular/metabolismo , Membrana Celular/patologia , Humanos , Técnicas Imunoenzimáticas , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Antígeno Prostático Específico/metabolismo , Prostatectomia , Hiperplasia Prostática/etnologia , Neoplasias da Próstata/etnologia , Estudos Retrospectivos
14.
Cancer Biol Ther ; 3(1): 29-35, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14726713

RESUMO

Normal development and function of metazoan organisms depend on oxygen availability. The level of oxygen can be sensed by individual cells, which respond to reduced oxygenation (hypoxia) largely through activation of hypoxia-inducible factor-1 (HIF-1). At the organism level the response to hypoxia involves an increase in red blood cell production. Within tissues, HIF activation increases the blood supply and blood vessel growth. At the individual cell level it is manifested as an increase in anaerobic metabolism in order to sustain basic cellular functions. Iron is central to the oxygen sensing mechanism, and sensitivity to other metals, namely cobalt and nickel, is a distinctive feature of the HIF system; in fact, this is often used as an initial way of implicating HIF-1 in a biological response. Historically, the fact that nickel or cobalt mimicked hypoxia provided an important clue as to the nature of the oxygen sensing mechanism. It also raises the possibility that nickel or cobalt exposure may have important toxic and pathological effects mediated by HIF activation. Here we review the implications of the metal sensitivity of the HIF-1 system, and examine the hypothesis that HIF-1 activation may play an important role in metal induced carcinogenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Neoplasias/fisiopatologia , Proteínas Nucleares/metabolismo , Consumo de Oxigênio , Fatores de Transcrição/metabolismo , Animais , Carcinógenos , Hipóxia Celular , Transformação Celular Neoplásica , Cobalto/toxicidade , Enzimas/metabolismo , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias/genética , Níquel/toxicidade , Transcrição Gênica
15.
Environ Health Perspect ; 110 Suppl 5: 831-4, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12426141

RESUMO

Using human and rodent cells in vitro, we characterized a hypoxia-inducible signaling pathway as one of the pathways affected by carcinogenic nickel compounds. Acute exposure to nickel activates hypoxia-inducible transcription factor-1 (HIF-1), which strongly induces hypoxia-inducible genes, including the recently discovered tumor marker Cap43. This gene has been cloned based on its nickel inducibility and was found to be highly inducible by hypoxia. To identify other HIF-1-dependent/independent nickel-inducible genes, we used cells obtained from HIF-1 alpha null mouse embryos and analyzed gene expression changes using the microarray technique. We found that genes coding for glycolytic enzymes, known to be regulated by HIF-1, were also induced in nickel-exposed cells. In addition, we identified a number of new genes highly induced by nickel in an HIF-dependent manner. Elevated HIF-1 activity after acute nickel exposure might be selectively advantageous because nickel-transformed rodent and human cells possess increased HIF-1 transcriptional activity. Hypoxia plays an important role in tumor progression. It selects for cells with enhanced glycolytic activity, causing production of large amounts of lactic acid, one of the most common features of tumor cells (Warburg effect). Here, we hypothesize that exposure to nickel activates the hypoxia-inducible pathway and facilitates selection of cells with increased transcriptional activity of hypoxia-inducible genes, which may be important in the nickel-induced carcinogenic process.


Assuntos
Transformação Celular Neoplásica , Proteínas de Ligação a DNA/biossíntese , Níquel/toxicidade , Proteínas Nucleares/biossíntese , Proteínas , Transdução de Sinais/fisiologia , Fatores de Transcrição , Animais , Proteínas de Ciclo Celular , Hipóxia Celular , Proteínas de Ligação a DNA/genética , Progressão da Doença , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Peptídeos e Proteínas de Sinalização Intracelular , Ácido Láctico/metabolismo , Níquel/farmacologia , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Biossíntese de Proteínas , Transcrição Gênica/efeitos dos fármacos
16.
Environ Health Perspect ; 110 Suppl 5: 719-23, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12426119

RESUMO

Ni(II) compounds are well known as human carcinogens, though the molecular events which are responsible for this are not yet fully understood. It has been proposed that the binding of N(II) ions within the cell nucleus is a crucial element in the mechanism of carcinogenesis. The most abundant proteins in the cell nucleus are histones, and this makes them the prime candidates for this role. This article is a review of our recent studies of histone H4 models of Ni(II) binding. We analyzed the sequence of the N-terminal tail of the histone H4, Ac-SGRGKGGKGLGKGGAKRH(18)RKVL-Am, for Ni(II) binding. This site has been proposed mainly because of the potent inhibitory effect of Ni(II) on the acetylation of lysine residues near the histidine H(18), and also because of the accessibility of the H4 tail in the histone octamer. Combined potentiometric and spectroscopic studies showed that the histidine 18 acted as an anchoring binding site for metal ions in the peptide investigated. Comparison with the results for Cu(II) binding are also reported. The results allowed us to propose that the binding of Ni(II) is able to promote a secondary structure with organized side-chain orientation on the N-terminal tail of histone H4.


Assuntos
Transformação Celular Neoplásica , Histonas/química , Modelos Químicos , Níquel/efeitos adversos , Sequência de Aminoácidos , Sítios de Ligação/fisiologia , Cobre/efeitos adversos , Cobre/farmacocinética , Humanos , Dados de Sequência Molecular , Níquel/farmacocinética , Análise Espectral , Relação Estrutura-Atividade
17.
Environ Health Perspect ; 110 Suppl 5: 783-8, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12429530

RESUMO

Cap43 is a protein whose RNA is induced under conditions of severe hypoxia or prolonged elevations of intracellular calcium. Additionally, Ni and Co also induce Cap43 because they produce a state of hypoxia in cells. Cap43 protein is expressed at low levels in normal tissues; however, in a variety of cancers, including lung, brain, melanoma, liver, prostate, breast, and renal cancers, Cap43 protein is overexpressed in cancer cells. The low level of expression of Cap43 in some normal tissues compared with their cancerous counterparts, combined with the high stability of Cap43 protein and mRNA, makes the Cap43 gene a new, important cancer marker. We hypothesize that the mechanism of Cap43 overexpression in cancer cells involves a state of hypoxia characteristic of cancer cells where the Cap43 protein becomes a signature for this hypoxic state.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias do Colo/genética , Regulação da Expressão Gênica , Neoplasias Pulmonares/genética , Biossíntese de Proteínas , Proteínas , Fatores de Transcrição , Northern Blotting , Proteínas de Ciclo Celular , Hipóxia Celular , Neoplasias do Colo/fisiopatologia , Proteínas de Ligação a DNA/farmacologia , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Pulmonares/fisiopatologia , Proteínas Nucleares/farmacologia , RNA Mensageiro/análise , Células Tumorais Cultivadas
18.
Mutat Res ; 533(1-2): 67-97, 2003 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-14643413

RESUMO

Human exposure to highly nickel-polluted environments, such as those associated with nickel refining, electroplating, and welding, has the potential to produce a variety of pathologic effects. Among them are skin allergies, lung fibrosis, and cancer of the respiratory tract. The exact mechanisms of nickel-induced carcinogenesis are not known and have been the subject of numerous epidemiologic and experimental investigations. These mechanisms are likely to involve genetic and epigenetic routes. The present review provides evidence for the genotoxic and mutagenic activity of Ni(II) particularly at high doses. Such doses are best delivered into the cells by phagocytosis of sparingly soluble nickel-containing dust particles. Ni(II) genotoxicity may be aggravated through the generation of DNA-damaging reactive oxygen species (ROS) and the inhibition of DNA repair by this metal. Broad spectrum of epigenetic effects of nickel includes alteration in gene expression resulting from DNA hypermethylation and histone hypoacetylation, as well as activation or silencing of certain genes and transcription factors, especially those involved in cellular response to hypoxia. The investigations of the pathogenic effects of nickel greatly benefit from the understanding of the chemical basis of Ni(II) interactions with intracellular targets/ligands and oxidants. Many pathogenic effects of nickel are due to the interference with the metabolism of essential metals such as Fe(II), Mn(II), Ca(II), Zn(II), or Mg(II). Research in this field allows for identification of putative Ni(II) targets relevant to carcinogenesis and prediction of pathogenic effects caused by exposure to nickel. Ultimately, the investigations of nickel carcinogenesis should be aimed at the development of treatments that would inhibit or prevent Ni(II) interactions with critical target molecules and ions, Fe(II) in particular, and thus avert the respiratory tract cancer and other adverse health effects in nickel workers.


Assuntos
Carcinógenos/toxicidade , Mutagênicos/toxicidade , Neoplasias/induzido quimicamente , Níquel/toxicidade , Poluentes Atmosféricos , Animais , Transformação Celular Neoplásica , Células Cultivadas , Dano ao DNA , Humanos , Espécies Reativas de Oxigênio
19.
Biol Trace Elem Res ; 86(1): 11-22, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12002656

RESUMO

We are trying to understand individual differences in susceptibility to chromate toxicity by comparing three different lymphoblastic cell lines derived from three different individuals. We have compared the uptake of CrO4(2-), the release of LDH from cells, the proliferation ability of the cells, and the DNA-protein crosslinks in these lymphoblastic cell lines exposed to chromate. We report here that one lymphoblastic cell line, GM0922B, appears to be considerably less sensitive than the other two cells lines to the cytotoxic effects of hexavalent chromium. The diminished sensitivity is almost twofold and can be accounted for by the decreased uptake of hexavalent chromium, which results in less lactate dehydrogenase release, and greater tolerance to chromate inhibition of cell proliferation and less DNA-protein crosslinking. This lower uptake of chromate combined with interindividual differences in extracellular Cr(VI) reducing capacity are probably the two most important determinants of genetic susceptibility to chromate toxicity.


Assuntos
Cromatos/toxicidade , DNA/metabolismo , Compostos de Potássio/toxicidade , Proteínas/metabolismo , Adulto , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Criança , Cromatos/farmacocinética , Feminino , Predisposição Genética para Doença , Humanos , L-Lactato Desidrogenase/metabolismo , Linfócitos/efeitos dos fármacos , Compostos de Potássio/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA