Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
FASEB J ; 33(12): 13837-13851, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31601121

RESUMO

Elevated proliferation rates in cancer can be visualized with positron emission tomography (PET) using 3'-deoxy-3'-l-[18F]fluorothymidine ([18F]FLT). This study investigates whether [18F]FLT transport proteins are regulated through hypoxia. Expression and function of human equilibrative nucleoside transporter (hENT)-1, hENT2, and thymidine kinase 1 (TK1) were studied under normoxic and hypoxic conditions, and assessed with [18F]FLT-PET in estrogen receptor positive (ER+)-MCF7, triple-negative MDA-MB231 breast cancer (BC) cells, and MCF10A cells (human mammary epithelial cells). Functional involvement of hENT2 [18F]FLT transport was demonstrated in all cell lines. In vitro [18F]FLT uptake was higher in MDA-MB231 than in MCF7: 242 ± 9 vs. 147 ± 18% radioactivity/mg protein after 60 min under normoxia. Hypoxia showed no significant change in radiotracer uptake. Protein analysis revealed increased hENT1 (P < 0.0963) in MDA-MB231. Hypoxia did not change expression of either hENT1, hENT2, or TK1. In vitro inhibition experiments suggested involvement of hENT1, hENT2, and human concentrative nucleoside transporters during [18F]FLT uptake into all cell lines. In vivo PET imaging revealed comparable tumor uptake in MCF7 and MDA-MB231 tumors over 60 min, reaching standardized uptake values of 0.96 ± 0.05 vs. 0.89 ± 0.08 (n = 3). Higher hENT1 expression in MDA-MB231 seems to drive nucleoside transport, whereas TK1 expression in MCF7 seems responsible for comparable [18F]FLT retention in ER+ tumors. Our study demonstrates that hypoxia does not significantly affect nucleoside transport as tested with [18F]FLT in BC.-Krys, D., Hamann, I., Wuest, M., Wuest, F. Effect of hypoxia on human equilibrative nucleoside transporters hENT1 and hENT2 in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Transportador Equilibrativo 2 de Nucleosídeo/metabolismo , Hipóxia/metabolismo , Proteínas de Transporte de Nucleosídeos/metabolismo , Animais , Transporte Biológico/fisiologia , Mama/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons/métodos
2.
FASEB J ; 32(9): 5104-5118, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29913554

RESUMO

Elevated growth in breast cancer (BC) activates hypoxia-inducible factor (HIF1α) and downstream, facilitative glucose transporter 1 (GLUT1), which can be visualized with 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG). GLUT5 (fructose) and GLUT2 (glucose/fructose) might provide alternative targets for BC imaging as to why effects of hypoxia on GLUT1/2/5 levels and function were examined in human BC models. GLUT1/2/5 and HIF1α mRNA was analyzed in BC patient biopsies. In MCF10A, MCF7, and MDA-MB231 cells, [18F]FDG, 6-deoxy-6-[18F]fluoro-d-fructose (6-[18F]FDF) and [18F]-fluoroazomycin arabinoside were used in radiotracer experiments, whereas GLUT1/2/5 mRNA was analyzed with real-time PCR and protein levels determined via Western blot/immunohistochemistry. Positron emission tomography imaging was performed in MCF7 and MDA-MB231 tumor-bearing mice. Glucose/fructose/cytochalasin B reduced cellular 6-[18F]FDF uptake by 50%, indicating functional involvement of GLUT2. With GLUT5 staining lower than GLUT1, 6-[18F]FDF revealed lower uptake than [18F]FDG [standardized uptake value (SUV)6-[18F]FDF, 120 min 0.77 ± 0.06 vs. SUV[18F]FDG, 120 min 1.08 ± 0.07] in MDA-MB231 tumors and was blocked by 20% with cytochalasin B after 10 min. Whereas correspondence between 6-[18F]FDF uptake and GLUT5 protein was low, high GLUT2 levels were detected in all cell lines and tumor models. Besides GLUT1, GLUT5 seems to be regulated under hypoxia on the molecular and functional level. Additionally, results strongly support a functional involvement of GLUT2 in fructose metabolism, possibly by compensating for the weaker expression and function of GLUT5 in BC.-Hamann, I., Krys, D., Glubrecht, D., Bouvet, V., Marshall, A., Vos, L., Mackey, J. R., Wuest, M., Wuest, F. Expression and function of hexose transporters GLUT1, GLUT2, and GLUT5 in breast cancer-effects of hypoxia.


Assuntos
Neoplasias da Mama/metabolismo , Mama/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Transportador de Glucose Tipo 5/metabolismo , Hipóxia/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Animais , Transporte Biológico/fisiologia , Mama/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Fluordesoxiglucose F18/metabolismo , Frutose/metabolismo , Glucose/metabolismo , Humanos , Hipóxia/patologia , Imuno-Histoquímica/métodos , Células MCF-7 , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons/métodos
3.
Mol Pharmacol ; 93(2): 79-89, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29142019

RESUMO

Use of [18F]FDG-positron emission tomography (PET) in clinical breast cancer (BC) imaging is limited mainly by insufficient expression levels of facilitative glucose transporter (GLUT)1 in up to 50% of all patients. Fructose-specific facilitative hexose transporter GLUT5 represents an alternative biomarker for PET imaging of hexose metabolism in BC. The goal of the present study was to compare the uptake characteristics of selected hexose-based PET radiotracers in murine BC model EMT6. Uptake of 1-deoxy-1-[18F]fluoro-d-fructose (1-[18F]FDF), 6-deoxy-6-[18F]fluoro-d-fructose (6-[18F]FDF), 1-deoxy-1-[18F]fluoro-2,5-anhydro-mannitol (1-[18F]FDAM), 2-deoxy-2-[18F]fluoro-d-glucose (2-[18F]FDG), and 6-deoxy-6-[18F]fluoro-d-glucose (6-[18F]FDG) was studied in EMT6 cells, tumors, and muscle and correlated to GLUT1 and GLUT5 expression levels. Fructose-derivative 6-[18F]FDF revealed greater tumor uptake than did structural analog 1-[18F]FDF, whereas 1-[18F]FDAM with locked anomeric configuration showed similar low tumor uptake to that of 1-[18F]FDF. Glucose-derivative 6-[18F]FDG reached maximum tumor uptake at 20 minutes, with no further accumulation over time. Uptake of 2-[18F]FDG was greatest and continuously increasing owing to metabolic trapping through phosphorylation by hexokinase II. In EMT6 tumors, GLUT5 mRNA expression was 20,000-fold lower compared with GLUT1. Whereas the latter was much greater in tumor than in muscle tissue (GLUT1 50:1), the opposite was found for GLUT5 mRNA expression (GLUT5 1:6). GLUT5 protein levels were higher in tumor versus muscle tissue as determined by Western blot and immunohistochemistry. Our data suggest that tumor uptake of fructose metabolism-targeting radiotracers 1-[18F]FDF, 6-[18F]FDF, and 1-[18F]FDAM does not correlate with GLUT5 mRNA levels but is linked to GLUT5 protein levels. In conclusion, our results highlight the importance of detailed biochemical studies on GLUT protein expression levels in combination with PET imaging studies for functional characterization of GLUTs in BC.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Animais , Linhagem Celular Tumoral , Feminino , Radioisótopos de Flúor/metabolismo , Frutose/metabolismo , Expressão Gênica , Proteínas Facilitadoras de Transporte de Glucose/genética , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 5 , Camundongos Endogâmicos BALB C , Músculos/metabolismo , RNA Mensageiro/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Análise Espectral/métodos
4.
Breast Cancer Res ; 17: 107, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26265048

RESUMO

INTRODUCTION: Lysyl oxidase (LOX; ExPASy ENZYME entry: EC 1.4.3.13) and members of the LOX-like family, LOXL1-LOXL4, are copper-dependent enzymes that can modify proteins of the extracellular matrix. Expression of LOX is elevated in many human cancers, including breast cancer. LOX expression correlates with the level of tissue hypoxia, and it is known to play a critical role in breast cancer metastasis. The goal of the present study was to target LOX with (1) molecular probe fluorescent labeling to visualize LOX in vitro and (2) a radiolabeled peptide to target LOX in vivo in three different preclinical models of breast cancer. METHODS: Gene expression of all five members of the LOX family was analyzed at the transcript level via microarray analysis using tissue biopsy samples from 176 patients with breast cancer. An oligopeptide sequence (GGGDPKGGGGG) was selected as a substrate-based, LOX-targeting structure. The peptide was labeled with fluorescein isothiocyanate (FITC) for confocal microscopy experiments with the murine breast cancer cell line EMT-6. In vivo molecular imaging experiments were performed using a C-terminal amidated peptide, GGGDPKGGGGG, labeled with a short-lived positron emitter, fluorine-18 ((18)F), for positron emission tomography (PET) in three different breast cancer models: EMT6, MCF-7 and MDA-MB-231. The PET experiments were carried out in the presence or absence of ß-aminopropionitrile (BAPN), an irreversible inhibitor of LOX. RESULTS: Immunostaining experiments using a LOX-specific antibody on EMT-6 cells cultured under hypoxic conditions confirmed the elevation of LOX expression in these cells. An FITC-labeled oligopeptide, FITC-Ava-GGGDPKGGGGG-NH2, was found to be localized in different cellular compartments under these conditions. After injection of [(18)F]fluorobenzoate-GGGDPKGGGGG-NH2, radioactivity uptake was visible in all three breast cancer models in vivo. Tumor uptake was reduced by predosing the animals with 2 mg of BAPN 4 h or 24 h before injection of the radiotracer. CONCLUSIONS: The present data support further investigation into the development of LOX-binding radiolabeled peptides as molecular probes for molecular imaging of LOX expression in cancer.


Assuntos
Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Imagem Molecular , Proteína-Lisina 6-Oxidase/metabolismo , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Expressão Gênica , Humanos , Hipóxia , Isoenzimas , Camundongos , Microscopia Confocal , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons , Proteína-Lisina 6-Oxidase/genética , RNA Mensageiro/genética
5.
Arch Biochem Biophys ; 558: 42-50, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24933099

RESUMO

Copper ions are known to induce insulin-like effects in various cell lines, stimulating the phosphoinositide 3'-kinase (PI3K)/Akt signaling cascade and leading to the phosphorylation of downstream targets, including FoxO transcription factors. The aim of this work was to study the role of insulin- and IGF1-receptors (IR and IGF1R) in insulin-like signaling induced by copper in HepG2 human hepatoma cells. Cells were exposed to Cu(II) at various concentrations for up to 60 min. While Akt and FoxO1a/FoxO3a were strongly phosphorylated in copper- and insulin-treated cells at all time points studied, only faint tyrosine phosphorylation of IR/IGF1R was detected in cells exposed to Cu(II) by either immunoprecipitation/immunoblot or by immunoblotting using phospho-specific antibodies, whereas insulin triggered strong phosphorylation at these sites. Pharmacological inhibition of IR/IGF1R modestly attenuated Cu-induced Akt and FoxO phosphorylation, whereas no attenuation of Cu-induced Akt activation was achieved by siRNA-mediated IR depletion. Cu(II)-induced FoxO1a nuclear exclusion was only slightly impaired by pharmacological inhibition of IR/IGF1R, whereas insulin-induced effects were blunted. In contrast, genistein, a broad-spectrum tyrosine kinase inhibitor, at concentrations not affecting IR/IGF1R, attenuated Cu(II)-induced Akt phosphorylation, pointing to the requirement of tyrosine kinases other than IR/IGF1R for Cu(II)-induced signaling.


Assuntos
Cobre/farmacologia , Insulina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Receptor IGF Tipo 1/metabolismo
6.
Biometals ; 27(2): 317-32, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24535192

RESUMO

Epidemiological studies have indicated a relationship between the prevalence of diabetes and exposure to arsenic. Mechanisms by which arsenic may cause this diabetogenic effect are largely unknown. The phosphoinositide 3'-kinase (PI3K)/Akt signaling pathway plays an important role in insulin signaling by controlling glucose metabolism, in part through regulating the activity of FoxO transcription factors. The present study aimed at investigating the effect of short and long-term exposure to arsenite on insulin signaling in HepG2 human hepatoma cells, the role of PI3K/Akt signaling therein and the modulation of target genes controlled by insulin. Exposure of cells to arsenite for 24 h rendered cells less responsive toward stimulation of Akt by insulin. At the same time, short-term exposure to arsenite induced a concentration-dependent increase in phosphorylation of Akt at Ser-473, followed by phosphorylation of FoxO proteins at sites known to be phosphorylated by Akt. Phosphorylation of FoxOs was prevented by wortmannin, pointing to the involvement of PI3K. Arsenite exposure resulted in attenuation of FoxO DNA binding and in nuclear exclusion of FoxO1a-EGFP. A 24-h exposure of HepG2 cells to submicromolar concentrations of arsenite resulted in downregulation of glucose 6-phosphatase (G6Pase) and selenoprotein P (SelP) mRNA levels. Curiously, arsenite had a dual effect on SelP protein levels, inducing a small increase in the nanomolar and a distinct decrease in the micromolar concentration range. Interestingly, arsenite-induced long-term effects on G6Pase and SelP mRNA or SelP protein levels were not blocked by the PI3K inhibitor, wortmannin. In conclusion, arsenite perturbs cellular signaling pathways involved in fuel metabolism: it impairs cellular responsiveness toward insulin, while at the same time stimulating insulin-like signaling to attenuate the expression of genes involved in glucose metabolism and the release of the hepatokine SelP, which is known to modulate peripheral insulin sensitivity.


Assuntos
Arsenitos/administração & dosagem , Arsenitos/farmacologia , Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Androstadienos/farmacologia , Células Hep G2 , Humanos , Selectina-P/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Wortmanina
7.
Mutat Res ; 736(1-2): 56-63, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21605570

RESUMO

The tumor suppressor protein p53, often called the guardian of the genome, is involved in important cellular processes, such as cell cycle control, apoptosis and DNA repair. With respect to BER, p53 might physically interact with and affect the transcription of different BER proteins such as hOGG1, APE1 or Polß. In studies in HCT116 p53(-/-) cells previously published, activity and mRNA expression of hOGG1 were found to be significantly decreased, while down-regulation of APE1 mRNA and protein levels in response to genotoxic stress were only described in HCT116 p53(+/+) cells, but not in the isogenic p53 knockout cell line. The predominantly indirect genotoxic carcinogen cadmium inhibits the BER pathway and potentially interferes with zinc binding proteins such as p53. Therefore, this study was accomplished to investigate whether p53 is involved in the cadmium-induced inhibition of BER activity. To address this issue we applied a non-radioactive cleavage test system based on a Cy5-labeled oligonucleotide. We present evidence that p53 is not essential for hOGG1 and APE1 gene expression as well as OGG and APE activity in unstressed HCT116 cells; however, it plays an important role in the cellular response to cadmium treatment. Here, a direct involvement of p53 was only observed with respect to APE1 gene expression contributing to an altered APE activity, while OGG activity was presumably affected indirectly due to a stronger accumulation of cadmium in HCT116 p53(+/+) cells. In summary, p53 indeed affects the BER pathway directly and indirectly in response to cadmium treatment.


Assuntos
Cádmio/toxicidade , DNA Glicosilases/metabolismo , Reparo do DNA/efeitos dos fármacos , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Genes p53 , Células HCT116 , Humanos
8.
Arch Biochem Biophys ; 516(2): 138-45, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22019820

RESUMO

FHRE-Luc is a promoter reporter construct that is widely used to assess the activity of FoxO (forkhead box, class O) transcription factors. We here demonstrate that this promoter construct responds to exposure of HepG2 human hepatoma cells to known agonists of the aryl hydrocarbon receptor (AhR), 3-methylcholanthrene, benzo(a)pyrene, and 6-formylindolo[3,2-b]carbazole. However, FHRE-Luc activation did not coincide with FoxO DNA binding or changes in Akt-induced FoxO phosphorylation after treatment with AhR agonists. Testing FHRE-Luc deletion constructs and using AhR-deficient cells, we found that FHRE-Luc activation by AhR agonists is due to a functional xenobiotic-response element (XRE) spanning the backbone/insert border of the reporter plasmid. In conclusion, care must be taken when using FHRE-Luc to assess FoxO activity in response to stimuli that potentially interfere with xenobiotic signaling.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Receptores de Hidrocarboneto Arílico/agonistas , Elementos de Resposta , Xenobióticos/farmacologia , Benzo(a)pireno/farmacologia , Carbazóis/farmacologia , Genes Reporter , Células Hep G2 , Humanos , Luciferases/genética , Metilcolantreno/farmacologia , Fosforilação , Receptores de Hidrocarboneto Arílico/deficiência , Transdução de Sinais
9.
Mutat Res ; 715(1-2): 32-41, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21782832

RESUMO

Inorganic arsenic is a strong, widespread human carcinogen. How exactly inorganic arsenic exerts carcinogenicity in humans is as yet unclear, but it is thought to be closely related to its metabolism. At exposure-relevant concentrations arsenic is neither directly DNA reactive nor mutagenic. Thus, more likely epigenetic and indirect genotoxic effects, among others a modulation of the cellular DNA damage response and DNA repair, are important molecular mechanisms contributing to its carcinogenicity. In the present study, we investigated the impact of arsenic on several base excision repair (BER) key players in cultured human lung cells. For the first time gene expression, protein level and in case of human 8-oxoguanine DNA glycosylase 1 (hOGG1) protein function was examined in one study, comparing inorganic arsenite and its trivalent and pentavalent mono- and dimethylated metabolites, also taking into account their cellular bioavailability. Our data clearly show that arsenite and its metabolites can affect several cellular endpoints related to DNA repair. Thus, cellular OGG activity was most sensitively affected by dimethylarsinic acid (DMA(V)), DNA ligase IIIα (LIGIIIα) protein level by arsenite and X-ray cross complementing protein 1 (XRCC1 protein) content by monomethylarsonic acid (MMA(V)), with significant effects starting at ≥3.2µM cellular arsenic. With respect to MMA(V), to our knowledge these effects are the most sensitive endpoints, related to DNA damage response, that have been identified so far. In contrast to earlier nucleotide excision repair related studies, the trivalent methylated metabolites exerted strong effects on the investigated BER key players only at cytotoxic concentrations. In summary, our data point out that after mixed arsenic species exposure, a realistic scenario after oral inorganic arsenic intake in humans, DNA repair might be affected by different mechanisms and therefore very effectively, which might facilitate the carcinogenic process of inorganic arsenic.


Assuntos
Arsênio/toxicidade , Reparo do DNA/efeitos dos fármacos , Neoplasias Pulmonares/genética , Mutagênicos/toxicidade , Arsenicais , Arsenitos/toxicidade , Disponibilidade Biológica , Linhagem Celular Tumoral , Sobrevivência Celular , DNA Glicosilases/metabolismo , DNA Ligase Dependente de ATP , DNA Ligases/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Humanos , Proteínas de Ligação a Poli-ADP-Ribose , Proteína 1 Complementadora Cruzada de Reparo de Raio-X , Proteínas de Xenopus
10.
Mutat Res ; 669(1-2): 122-30, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19505484

RESUMO

Oxidative stress is involved in many diseases, and the search for appropriate biomarkers is one major focus in molecular epidemiology. 8-Oxoguanine (8-oxoG), a potentially mutagenic DNA lesion, is considered to be a sensitive biomarker for oxidative stress. Another approach consists in assessing the repair capacity towards 8-oxoG, mediated predominantly by the human 8-oxoguanine DNA glycosylase 1 (hOGG1). With respect to the latter, during the last few years so-called cleavage assays have been described, investigating the incision of (32)P-labelled and 8-oxoG damaged oligonucleotides by cell extracts. Within the present study, a sensitive non-radioactive test system based on a Cy5-labelled oligonucleotide has been established. Sources of incision activity are isolated proteins or extracts prepared from cultured cells and peripheral blood mononuclear cells (PBMC). After comparing different oligonucleotide structures, a hairpin-like structure was selected which was not degraded by cell extracts. Applying this test system the impact of copper on the activity of isolated hOGG1 and on hOGG activity in A549 cells was examined, showing a distinct inhibition of the isolated protein at low copper concentration as compared to a modest inhibition of hOGG activity in cells at beginning cytotoxic concentrations. For investigating PBMC, all reaction conditions, including the amounts of oligonucleotide and cell extract as well as the reaction time have been optimized. The incision activities of PBMC protein extracts obtained from different donors have been investigated, and inter-individual differences have been observed. In summary, the established method is as sensitive and even faster than the radioactive technique, and additionally, offers the advantage of reduced costs and low health risk.


Assuntos
Sulfato de Cobre/farmacologia , Dano ao DNA , DNA Glicosilases/metabolismo , DNA Glicosilases/fisiologia , Reparo do DNA , 8-Hidroxi-2'-Desoxiguanosina , Animais , Ensaio de Unidades Formadoras de Colônias , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Frações Subcelulares , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
11.
Redox Biol ; 20: 19-27, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30261343

RESUMO

Diethyl maleate (DEM), a thiol-reactive α,ß-unsaturated carbonyl compound, depletes glutathione (GSH) in exposed cells and was previously shown by us to elicit a stress response in Caenorhabditis elegans that, at lower concentrations, results in enhanced stress resistance and longer lifespan. This hormetic response was mediated through both the Nrf2 ortholog, SKN-1, and the forkhead box O (FOXO) family transcription factor DAF-16. As FOXO signaling is evolutionarily conserved, we analyzed here the effects of DEM exposure on FOXO in cultured human cells (HepG2, HEK293). DEM elicited nuclear accumulation of GFP-coupled wild-type human FOXO1, as well as of a cysteine-deficient FOXO1 mutant. Despite the nuclear accumulation of FOXO1, neither FOXO1 DNA binding nor FOXO target gene expression were stimulated, suggesting that DEM causes nuclear accumulation but not activation of FOXO1. FOXO1 nuclear exclusion elicited by insulin or xenobiotics such as arsenite or copper ions was attenuated by DEM, suggesting that DEM interfered with nuclear export. In addition, insulin-induced FOXO1 phosphorylation at Thr-24, which is associated with FOXO1 nuclear exclusion, was attenuated upon exposure to DEM. Different from FOXO-dependent expression of genes, Nrf2 target gene mRNAs were elevated upon exposure to DEM. These data suggest that, different from C. elegans, DEM elicits opposing effects on the two stress-responsive transcription factors, Nrf2 and FOXO1, in cultured human cells.


Assuntos
Núcleo Celular/metabolismo , Proteína Forkhead Box O1/metabolismo , Maleatos/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Glutationa , Células HEK293 , Células Hep G2 , Humanos , Espaço Intracelular/metabolismo , Modelos Biológicos , Fosforilação , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Estresse Fisiológico
12.
Nucl Med Biol ; 58: 51-58, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29367096

RESUMO

INTRODUCTION: Receptor tyrosine kinase (RTK) platelet-derived growth factor receptor-alpha (PDGFRα) was recently identified as a molecular switch for dedifferentiation in thyroid cancer that predicts resistance to therapy as well as recurrence of disease in papillary thyroid cancer. Here we describe the radiolabeling and functional characterization of an imaging probe based on a PDGFRα-specific monoclonal antibody (mAb) for immuno-PET imaging of PDGFRα in papillary thyroid cancer. METHODS: Antibody D13C6 (Cell Signaling) was decorated with chelator NOTA using bioconjugation reaction with 2-(p-NCS-Bz)-NOTA. Radiolabeling was carried out using 40 µg of antibody-NOTA conjugate with 143-223 MBq of [64Cu]CuCl2 in 0.25 M NaOAc (pH 5.5) at 30 °C for 1 h. The reaction mixture was purified with size-exclusion chromatography (PD-10 column). PDGFRα and mock transfected B-CPAP thyroid cancer cells lines for validation of 64Cu-labeled immuno-conjugates were generated using LVX-Tet-On technology. PET imaging was performed in NSG mice bearing bilaterally-induced PDGFRα (+/-) B-CPAP tumors. RESULTS: Bioconjugation of NOTA chelator to monoclonal antibody D13C6 resulted in 2.8 ±â€¯1.3 chelator molecules per antibody as determined by radiometric titration with 64Cu. [64Cu]Cu-NOTA-D13C6 was isolated in high radiochemical purity (>98%) and good radiochemical yields (19-61%). The specific activity was 0.9-5.1 MBq/µg. Cellular uptake studies revealed a specific radiotracer uptake in PDGFRα expressing cells compared to control cells. PET imaging resulted in SUVmean values of ~5.5 for PDGFRα (+) and ~2 for PDGFRα (-) tumors, after 48 h p.i.. After 1 h, radiotracer uptake was also observed in the bone marrow (SUVmean ~5) and spleen (SUVmean ~8.5). CONCLUSION: Radiolabeled antibody [64Cu]Cu-NOTA-D13C6 represents a novel and promising radiotracer for immuno-PET imaging of PDGFRα in metastatic papillary thyroid cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: The presented work has the potential to allow physicians to identify papillary thyroid cancer patients at risk of metastases by using the novel immuno-PET imaging assay based on PDGFRα-targeting antibody [64Cu]Cu-NOTA-D13C6.


Assuntos
Tomografia por Emissão de Pósitrons/métodos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Câncer Papilífero da Tireoide/diagnóstico por imagem , Animais , Autorradiografia , Transporte Biológico , Linhagem Celular Tumoral , Radioisótopos de Cobre , Compostos Heterocíclicos/química , Compostos Heterocíclicos com 1 Anel , Humanos , Imunoconjugados/química , Imunoconjugados/metabolismo , Camundongos , Câncer Papilífero da Tireoide/imunologia , Câncer Papilífero da Tireoide/metabolismo
13.
Mol Nutr Food Res ; 51(2): 201-10, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17230584

RESUMO

Copper is an essential trace element involved, among other functions, in enzymatic antioxidative defense systems. However, nonprotein bound copper ions have been shown to generate reactive oxygen species. To gain insight into the discrepancy between the protective properties of copper on the one hand and its toxicity on the other hand, we examined the genotoxic effects of CuSO(4) in cultured human cells. Here we report that copper, at cytotoxic concentrations, induces oxidative DNA base modifications and DNA strand breaks. However, at lower noncytotoxic concentrations, copper inhibits the repair of oxidative DNA damage induced by visible light. As a first mechanistic hint, inhibition of H(2)O(2)-induced poly(ADP-ribosyl)ation was identified in cultured cells and further experiments demonstrated a strong inhibition of the activity of isolated poly(ADP-ribose)polymerase-1 (PARP-1) by copper. Bioavailability studies of copper showed a dose-dependent uptake in cells and pointed out the relevance of the applied concentrations. Taken together, the results indicate that copper, under conditions of either disturbed homeostasis or overload due to high exposure, exerts defined genotoxic effects. Hence, a balance needs to be maintained to ensure sufficient uptake and to prevent overload.


Assuntos
Cobre/farmacologia , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Cobre/metabolismo , Quebras de DNA , Células HeLa , Humanos , Oxirredução , Poli(ADP-Ribose) Polimerase-1
14.
EJNMMI Res ; 6(1): 37, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27112768

RESUMO

BACKGROUND: Cyclooxygenase-2 (COX-2) is the inducible isoform of the cyclooxygenase enzyme family. COX-2 is involved in tumor development and progression, and frequent overexpression of COX-2 in a variety of human cancers has made COX-2 an important drug target for cancer treatment. Non-invasive imaging of COX-2 expression in cancer would be useful for assessing COX-2-mediated effects on chemoprevention and radiosensitization using COX-2 inhibitors as an emerging class of anti-cancer drugs, especially for colorectal cancer. Herein, we describe the radiopharmacological analysis of [(18)F]Pyricoxib, a novel radiolabeled COX-2 inhibitor, for specific PET imaging of COX-2 in colorectal cancer. METHODS: Uptake of [(18)F]Pyricoxib was assessed in human colorectal cancer cell lines HCA-7 (COX-2 positive) and HCT-116 (COX-2 negative). Standard COX-2 inhibitors were used to test for specificity of [(18)F]Pyricoxib for COX-2 binding in vitro and in vivo. PET imaging, biodistribution, and radiometabolite analyses were included into radiopharmacological evaluation of [(18)F]Pyricoxib. RESULTS: Radiotracer uptake in COX-2 positive HCA-7 cells was significantly higher than in COX-2 negative HCT-116 cells (P < 0.05). COX-2 inhibitors, celecoxib, rofecoxib, and SC58125, blocked uptake of [(18)F]Pyricoxib in HCA-7 cells in a concentration-dependent manner. The radiotracer was slowly metabolized in mice, with approximately 60 % of intact compound after 2 h post-injection. Selective COX-2-mediated tumor uptake of [(18)F]Pyricoxib in HCA-7 xenografts was confirmed in vivo. Celecoxib (100 mg/kg) selectively blocked tumor uptake by 16 % (PET image analysis; P < 0.05) and by 51 % (biodistribution studies; P < 0.01). CONCLUSIONS: The novel PET radiotracer [(18)F]Pyricoxib displays a promising radiopharmacological profile to study COX-2 expression in cancer in vivo.

15.
Methods Mol Biol ; 1208: 73-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25323500

RESUMO

The identification of appropriate biomarkers for oxidative stress is one major aim in molecular epidemiology. Besides the quantification of specific DNA lesions such as of 8-oxoguanine (8oxoG), another approach consists in the assessment of the repair capacity towards 8oxoG, mediated predominantly by the human 8-oxoguanine DNA glycosylase 1 (hOGG1); further processing of base excision repair involves AP endonuclease 1 (APE1). Thus, during the last few years the so-called cleavage assays have been described, investigating the incision capacity of cell extracts towards (32)P-labelled and 8oxoG damaged oligonucleotides. Here, we describe a sensitive nonradioactive test system based on Cy5-labelled oligonucleotides with hairpin-like structures, enabling the assessment of activities of the isolated hOGG1 and APE1 as well as their activities in extracts prepared from cultured cells or peripheral blood mononuclear cells (PBMC). This approach allows the sensitive quantification of modulating exposures, such as inhibitory metal compounds, and also the determination of interindividual differences in DNA repair capacities. The method is as sensitive and even faster as compared to the use of radioactively labelled oligonucleotides and additionally offers the advantage of reduced costs and low health risk.


Assuntos
Dano ao DNA , Reparo do DNA , Leucócitos Mononucleares/metabolismo , Biologia Molecular/métodos , Extratos Celulares , Linhagem Celular Tumoral , DNA Glicosilases/isolamento & purificação , DNA Glicosilases/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Humanos , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Oxirredução , Frações Subcelulares/metabolismo
16.
Nucl Med Biol ; 41(8): 660-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24993371

RESUMO

INTRODUCTION: (18)F-labeled amino acids are important PET radiotracers for molecular imaging of cancer. This study describes synthesis and radiopharmacological evaluation of 2-amino-5-(4-[(18)F]fluorophenyl)pent-4-ynoic acid ([(18)F]FPhPA) as a novel amino acid radiotracer for oncologic imaging. METHODS: (18)F]FPhPA was prepared using Pd-mediated Sonogashira cross-coupling reaction between 4-[(18)F]fluoroiodobenzene ([(18)F]FIB) and propargylglycine. The radiopharmacological profile of [(18)F]FPhPA was evaluated in comparison with O-(2-[(18)F]fluoroethyl)-L-tyrosine ([(18)F]FET) using the murine breast cancer cell line EMT6 involving cellular uptake studies, radiotracer uptake competitive inhibition experiments and small animal PET imaging. RESULTS: (18)F]FPhPA was prepared in 42±10% decay-corrected radiochemical yield with high radiochemical purity >95% after semi-preparative HPLC purification. Cellular uptake of L-[(18)F]FPhPA reached a maximum of 58±14 % radioactivity/mg protein at 90 min. Lower uptake was observed for racemic and D-[(18)F]FPhPA. Radiotracer uptake inhibition studies by synthetic and naturally occurring amino acids suggested that Na(+)-dependent system ASC, especially ASCT2, and Na(+)-independent system L are important amino acid transporters for [(18)F]FPhPA uptake into EMT6 cells. Small animal PET studies demonstrated similar high tumor uptake of [(18)F]FPhPA in EMT6 tumor-bearing mice compared to [(18)F]FET reaching a maximum standardized uptake value (SUV) of 1.35 after 60 min p.i.. Muscle uptake of [(18)F]FPhPA was higher (SUV30min=0.65) compared to [(18)F]FET (SUV30min=0.40), whereas [(18)F]FPhPA showed a more rapid uptake and clearance from the brain compared to [(18)F]FET. CONCLUSION: L-[(18)F]FPhPA is the first (18)F-labeled amino acid prepared through Pd-mediated cross-coupling reaction. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: L-[(18)F]FPhPA displayed promising properties as a novel amino acid radiotracer for molecular imaging of system ASC and system L amino acid transporters in cancer.


Assuntos
Aminoácidos Aromáticos/síntese química , Aminoácidos/síntese química , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Sistema ASC de Transporte de Aminoácidos/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Aminoácidos Aromáticos/química , Aminoácidos Aromáticos/metabolismo , Animais , Ligação Competitiva , Transporte Biológico , Linhagem Celular Tumoral , Técnicas de Química Sintética , Regulação Neoplásica da Expressão Gênica , Cinética , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Antígenos de Histocompatibilidade Menor , Radioquímica , Temperatura
17.
Free Radic Biol Med ; 75 Suppl 1: S53, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26461409

RESUMO

Diethyl maleate (DEM) is a thiol-depleting agent frequently employed in cell culture analyses. Here, we investigated the effect of DEM exposure on insulin signaling at the level of FoxO transcription factor activity and its potential consequences for stress resistance and life span. Exposure of HepG2 human hepatoma cells to subcytotoxic concentrations of DEM resulted in nuclear accumulation of overexpressed EGFP-tagged FoxO1a. DEM-induced nuclear accumulation overrode insulin-induced nuclear exclusion of FoxO1a. Despite a slightly enhanced FoxO DNA binding activity in DEM-exposed cells, expression of FoxO-regulated genes (glucose 6-phosphatase, selenoprotein P) was downregulated, indicating that nuclear accumulation does not necessarily coincide with enhanced transcription factor activity. To test for an effect of DEM on organismal stress resistance, we exposed C. elegans roundworms to the thiol depletor. Survival in the presence of the redox cycler paraquat was significantly increased following exposure to DEM, implying that DEM pre-exposure induced cellular resistance against oxidative stress. Furthermore, in DEM-exposed C. elegans populations expressing a GFP-tagged version of the C. elegans FoxO ortholog, DAF-16, numbers of worms with predominantly nuclear DAF-16 increased - in line with the findings from HepG2 cells. In keeping with the known function of DAF-16 in stress resistance, C. elegans life span was elevated upon exposure to DEM in a concentration-dependent manner. A maximum extension of life span and deceleration of aging was achieved at 100 µ? of DEM. In summary, exposure to DEM caused a modulation of FoxO subcellular localization in both HepG2 cells and C. elegans roundworms, followed by a modulation of life span and stress resistance in C. elegans.

18.
Redox Biol ; 1: 104-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24024143

RESUMO

FoxO transcription factors and their regulators in the phosphoinositide 3'-kinase (PI3K)/Akt signaling pathway play an important role in the control of cellular processes involved in carcinogenesis, such as proliferation and apoptosis. We have previously demonstrated that physiologically relevant heavy metal ions, such as copper or zinc ions, can stimulate this pathway, triggering phosphorylation and nuclear export of FoxO transcription factors. The present study aims at investigating the effect of arsenite on FoxO transcription factors and the role of PI3K/Akt signaling therein. Exposure of HaCaT human keratinocytes to arsenite resulted in a distinct decrease of glutathione levels only at cytotoxic concentrations. In contrast, a strong phosphorylation of FoxO1a/FoxO3a and Akt was observed at subcytotoxic concentrations of arsenite in HaCaT human keratinocytes. A time- and concentration-dependent increase in phosphorylation of FoxO1a and FoxO3a at sites known to be phosphorylated by Akt as well as phosphorylation of Akt at Ser-473 was detected. These phosphorylations were blunted in the presence of wortmannin, pointing to the involvement of PI3K.


Assuntos
Arsenitos/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Queratinócitos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Androstadienos/farmacologia , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular , Cobre/farmacologia , Glutationa/metabolismo , Humanos , Queratinócitos/metabolismo , Fosforilação , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA