Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Mol Neurobiol ; 56(6): 4566-4581, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30353492

RESUMO

The role of astrocytes is becoming increasingly important to understanding how glioblastoma (GBM) tumor cells diffusely invade the brain. Yet, little is known of the contribution of extracellular vesicle (EV) signaling in GBM/astrocyte interactions. We modeled GBM-EV signaling to normal astrocytes in vitro to assess whether this mode of intercellular communication could support GBM progression. EVs were isolated and characterized from three patient-derived GBM stem cells (NES+/CD133+) and their differentiated (diff) progeny cells (NES-/CD133-). Uptake of GBM-EVs by normal primary astrocytes was confirmed by fluorescence microscopy, and changes in astrocyte podosome formation and gelatin degradation were measured. Quantitative mass spectrometry-based proteomics was performed on GBM-EV stimulated astrocytes. Interaction networks were generated from common, differentially abundant proteins using Ingenuity® (Qiagen Bioinformatics) and predicted upstream regulators were tested by qPCR assays. Podosome formation and Cy3-gelatin degradation were induced in astrocytes following 24-h exposure to GBM-stem and -diff EVs, with EVs released by GBM-stem cells eliciting a greater effect. More than 1700 proteins were quantified, and bioinformatics predicted activations of MYC, NFE2L2, FN1, and TGFß1 and inhibition of TP53 in GBM-EV stimulated astrocytes that were then confirmed by qPCR. Further qPCR studies identified significantly decreased Δ133p53 and increased p53ß in astrocytes exposed to GBM-EVs that might indicate the acquisition of a pro-inflammatory, tumor-promoting senescence-associated secretory phenotype (SASP). Inhibition of TP53 and activation of MYC signaling pathways in normal astrocytes exposed to GBM-EVs may be a mechanism by which GBM manipulates astrocytes to acquire a phenotype that promotes tumor progression.


Assuntos
Astrócitos/metabolismo , Neoplasias Encefálicas/metabolismo , Vesículas Extracelulares/metabolismo , Glioblastoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Idoso , Diferenciação Celular , Linhagem Celular Tumoral , Senescência Celular , Vesículas Extracelulares/ultraestrutura , Gelatina/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Nanopartículas/ultraestrutura , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Tamanho da Partícula , Fenótipo , Podossomos/metabolismo , Isoformas de Proteínas/metabolismo , Proteólise , Proteoma/metabolismo
2.
Oncogenesis ; 6(5): e336, 2017 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-28504693

RESUMO

High-grade glioma (HGG) is an incurable brain cancer. The transcriptomes of cells within HGG tumors are highly heterogeneous. This renders the tumors unresponsive or able to adapt to therapeutics targeted at single pathways, thereby causing treatment failure. To overcome this, we focused on cyclin-dependent kinase 7 (CDK7), a ubiquitously expressed molecule involved in two major drivers of HGG pathogenesis: cell cycle progression and RNA polymerase-II-based transcription. We tested the activity of THZ1, an irreversible CDK7 inhibitor, on patient-derived primary HGG cell lines and ex vivo HGG patient tissue slices, using proliferation assays, microarray analysis, high-resolution respirometry, cell cycle analysis and in vivo tumor orthografts. The cellular processes affected by CDK7 inhibition were analyzed by reverse transcriptase-quantitative PCR, western blot, flow cytometry and immunofluorescence. THZ1 perturbed the transcriptome and disabled CDK activation, leading to cell cycle arrest at G2 and DNA damage. THZ1 halted transcription of the nuclear-encoded mitochondrial ribosomal genes, reducing mitochondrial translation and oxidative respiration. It also inhibited the expression of receptor tyrosine kinases such as epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor-α (PDGFR-α), reducing signaling flux through the AKT, extracellular-signal-regulated kinase 1/2 (ERK1/2) and signal transducer and activator of transcription 3 (STAT3) downstream pathways. Finally, THZ1 disrupted nucleolar, Cajal body and nuclear speckle formation, resulting in reduced cytosolic translation and malfunction of the spliceosome and thus leading to aberrant mRNA processing. These findings indicate that CDK7 is crucial for gliomagenesis, validate CDK7 as a therapeutic target and provide new insight into the cellular processes that are affected by THZ1 and induce antitumor activity.

3.
Br J Cancer ; 114(2): 188-98, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26657652

RESUMO

BACKGROUND: Resistance to temozolomide (TMZ) greatly limits chemotherapeutic effectiveness in glioblastoma (GBM). Here we analysed the ability of the Inhibitor-of-apoptosis-protein (IAP) antagonist birinapant to enhance treatment responses to TMZ in both commercially available and patient-derived GBM cells. METHODS: Responses to TMZ and birinapant were analysed in a panel of commercial and patient-derived GBM cell lines using colorimetric viability assays, flow cytometry, morphological analysis and protein expression profiling of pro- and antiapoptotic proteins. Responses in vivo were analysed in an orthotopic xenograft GBM model. RESULTS: Single-agent treatment experiments categorised GBM cells into TMZ-sensitive cells, birinapant-sensitive cells, and cells that were insensitive to either treatment. Combination treatment allowed sensitisation to therapy in only a subset of resistant GBM cells. Cell death analysis identified three principal response patterns: Type A cells that readily activated caspase-8 and cell death in response to TMZ while addition of birinapant further sensitised the cells to TMZ-induced cell death; Type B cells that readily activated caspase-8 and cell death in response to birinapant but did not show further sensitisation with TMZ; and Type C cells that showed no significant cell death or moderately enhanced cell death in the combined treatment paradigm. Furthermore, in vivo, a Type C patient-derived cell line that was TMZ-insensitive in vitro and showed a strong sensitivity to TMZ and TMZ plus birinapant treatments. CONCLUSIONS: Our results demonstrate remarkable differences in responses of patient-derived GBM cells to birinapant single and combination treatments, and suggest that therapeutic responses in vivo may be greatly affected by the tumour microenvironment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Dacarbazina/análogos & derivados , Dipeptídeos/farmacologia , Glioblastoma/patologia , Indóis/farmacologia , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Animais , Western Blotting , Caspase 8/efeitos dos fármacos , Caspase 8/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Citometria de Fluxo , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia de Contraste de Fase , Transplante de Neoplasias , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cell Death Dis ; 6: e1966, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26539913

RESUMO

Cyclopentenone prostaglandins (CyPGs), such as 15-deoxy-Δ(12,14)-prostaglandin J2 (15dPGJ2), are reactive prostaglandin metabolites exerting a variety of biological effects. CyPGs are produced in ischemic brain and disrupt the ubiquitin-proteasome system (UPS). Ubiquitin-C-terminal hydrolase L1 (UCH-L1) is a brain-specific deubiquitinating enzyme that has been linked to neurodegenerative diseases. Using tandem mass spectrometry (MS) analyses, we found that the C152 site of UCH-L1 is adducted by CyPGs. Mutation of C152 to alanine (C152A) inhibited CyPG modification and conserved recombinant UCH-L1 protein hydrolase activity after 15dPGJ2 treatment. A knock-in (KI) mouse expressing the UCH-L1 C152A mutation was constructed with the bacterial artificial chromosome (BAC) technique. Brain expression and distribution of UCH-L1 in the KI mouse was similar to that of wild type (WT) as determined by western blotting. Primary cortical neurons derived from KI mice were resistant to 15dPGJ2 cytotoxicity compared with neurons from WT mice as detected by the WST-1 cell viability assay and caspase-3 and poly ADP ribose polymerase (PARP) cleavage. This protective effect was accompanied with significantly less ubiquitinated protein accumulation and aggregation as well as less UCH-L1 aggregation in C152A KI primary neurons after 15dPGJ2 treatment. Additionally, 15dPGJ2-induced axonal injury was also significantly attenuated in KI neurons as compared with WT. Taken together, these studies indicate that UCH-L1 function is important in hypoxic neuronal death, and the C152 site of UCH-L1 has a significant role in neuronal survival after hypoxic/ischemic injury.


Assuntos
Isquemia Encefálica/genética , Ciclopentanos/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Mutação Puntual , Prostaglandinas/toxicidade , Ubiquitina Tiolesterase/genética , Animais , Sítios de Ligação , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/enzimologia , Neurônios/metabolismo , Ratos , Ubiquitina Tiolesterase/biossíntese
5.
Br J Cancer ; 111(7): 1255-61, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25144626

RESUMO

The dismal outlook for patients with the most aggressive and common form of adult brain cancer, glioblastoma (GBM), motivates a search for new therapeutic strategies and targets for this aggressive disease. Here we review the findings to date on the role of Eph family receptor tyrosine kinases and their ephrin ligands in brain cancer. Expression of the Eph family of cell surface proteins is generally downregulated to very low levels in normal adult tissues making them particularly attractive for directed therapeutic targeting. Recent Eph targeting studies in pre-clinical models of GBM have been very encouraging and may provide an avenue to treat these highly refractory aggressive tumours.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores da Família Eph/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Ensaios de Seleção de Medicamentos Antitumorais , Efrinas/fisiologia , Glioblastoma/tratamento farmacológico , Humanos , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo , Transdução de Sinais
6.
Toxicol Appl Pharmacol ; 233(3): 411-9, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18929589

RESUMO

Male and female F344 rats but not B6C3F1 mice exposed for 104 weeks to propiverine hydrochloride (1-methylpiperid-4-yl 2,2-diphenyl-2-(1-propoxy)acetate hydrochloride), used for treatment of patients with neurogenic detrusor overactivity (NDO) and overactive bladder (OAB), presented with an accumulation of proteins in the cytosol and nuclei of renal proximal tubule epithelial cells, yet despite this, no increased renal tumor incidence was observed. In order to provide an improved interpretation of these findings and a better basis for human health risk assessment, male and female F344 rats were exposed for 16 weeks to 1000 ppm propiverine in the diet, the accumulating protein was isolated from the kidneys via cytosolic and nuclear preparations or laser-capture microdissection and analyzed using molecular weight determination and mass spectrometry. The accumulating protein was found to be d-amino acid oxidase (DAAO), an enzyme involved in amino and fatty acid metabolism. Subsequent reanalysis of kidney homogenate and nuclear samples as well as tissue sections using western blot and DAAO-immunohistochemistry, confirmed the presence and localization of DAAO in propiverine-treated male and female F344 rats. The accumulation of DAAO only in rats, and the limited similarity of rat DAAO with other species, including humans, suggests a rat-specific mechanism underlying the drug-induced renal DAAO accumulation with little relevance for patients chronically treated with propiverine.


Assuntos
Benzilatos/efeitos adversos , Núcleo Celular/efeitos dos fármacos , Antagonistas Colinérgicos/efeitos adversos , Citosol/efeitos dos fármacos , D-Aminoácido Oxidase , Rim/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Benzilatos/farmacocinética , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Antagonistas Colinérgicos/farmacocinética , Citosol/enzimologia , Citosol/metabolismo , D-Aminoácido Oxidase/isolamento & purificação , D-Aminoácido Oxidase/metabolismo , Eletroforese em Gel de Poliacrilamida , Feminino , Hialina/metabolismo , Imuno-Histoquímica , Rim/enzimologia , Rim/metabolismo , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Dados de Sequência Molecular , Conformação Proteica , Ratos , Ratos Endogâmicos F344 , Fatores Sexuais
7.
J Mol Graph Model ; 19(6): 487-94, 587-90, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11552677

RESUMO

Lipoprotein lipase (LPL) plays a central role in lipid metabolism. The D9N and N291S mutations in the LPL gene are associated with elevated triglyceride and decreased HDL-cholesterol levels. Published in vitro expression studies suggest that these two mutations are associated with reduced LPL enzymatic activity. We sought to gain further insight on the impact of these two mutations on the LPL structure and function by molecular modelling techniques. Homology modelling was used to develop a three-dimensional (3D) structure of LPL from human pancreatic lipase. Two separate LPL models for the D9N and N291S substitutions were constructed and compared with the wild type LPL for differences in hydrophobicity, atomic burial, hydrogen bond pattern, and atomic mobility. In comparison to the wild type model, the 9N model was associated with significantly increased atomic mobility of its neighboring residues, but the catalytic site was not affected. The region near residue 9 in the upper part of the N-domain was considered a candidate site for protein-protein interaction. In the N291S model, alterations in H-bonds and constrained atomic mobility were among conformational changes in the region where the substitution had occurred. These are hypothesized to cause an increase in the rate of dissociation in LPL dimerization, subsequently affecting the LPL enzymatic activity. We also modelled the C-domain of apoCII, the obligatory cofactor of LPL, from 2D NMR data and docked the model with LPL to explore their interaction site. These docking experiments suggest that the C-domain of apoCII interacts with the interface of N- and C-domains of LPL and part of the lid structure that covers the catalytic site. In summary, we provide molecular modelling data on two well-known mutations in the LPL gene to help explain the published in vitro expression findings and propose a possible LPL-apoCII interaction site. Our data indicate that molecular modelling of LPL mutations could provide a valuable tool to understand the effects of a mutation on the structure-function of this important enzyme.


Assuntos
Simulação por Computador , Lipase Lipoproteica/química , Modelos Moleculares , Sequência de Aminoácidos , Apolipoproteína C-II , Apolipoproteínas C/química , Humanos , Lipase Lipoproteica/genética , Dados de Sequência Molecular , Mutagênese , Pâncreas/enzimologia , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
8.
Bioorg Med Chem ; 9(7): 1765-71, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11425578

RESUMO

The epipodophyllotoxin etoposide is a potent and widely used anticancer drug that targets DNA topoisomerase II. The synthesis, photochemical, and biological testing of a photoactivatable aromatic azido analogue of etoposide also containing an iodo group is described. This azido analogue should prove useful for identifying the etoposide interaction site on topoisomerase II. Irradiation of the azido analogue and an aldehyde-containing azido precursor with UV light produced changes in their UV--visible spectra that were consistent with photoactivation. The azido analogue strongly inhibited topoisomerase II and inhibited the growth of Chinese Hamster Ovary cells. Azido analogue-induced topoisomerase II--DNA covalent complexes were significantly increased subsequent to UV irradiation of drug-treated human leukemia K562 cells as compared to etoposide-treated cells. These results suggest that the photoactivated form of etoposide is a more effective topoisomerase II poison either by interacting directly with the enzyme or with DNA subsequent to topoisomerase II-mediated strand cleavage.


Assuntos
Antineoplásicos/química , Etoposídeo/química , Marcadores de Fotoafinidade/síntese química , Marcadores de Fotoafinidade/farmacologia , Animais , Antineoplásicos/farmacologia , Células CHO , Cricetinae , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Humanos , Células K562 , Cinética , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Fotoquímica , Inibidores da Topoisomerase II
9.
Math Biosci ; 170(1): 1-16, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11259801

RESUMO

A mathematical model of cancer cell growth and response to treatment with the experimental antimitotic agent curacin A is presented. Rate parameters for the untreated growth of MCF-7/LY2 breast cancer and A2780 ovarian cell lines are determined from in vitro growth studies. Subsequent growth studies following treatments with 2.5, 25 and 50 nanomolar (nM), concentrations of curacin A are used to determine effects on the cell cycle and cell viability. The model's system of ordinary differential equations yields an approximate analytical solution which predicts the minimum concentration necessary to prevent growth. The model shows that cell growth is arrested when the apoptotic rate is greater than the mitotic rate and that the S-phase transition rate acts to amplify this effect. Analysis of the data suggests that curacin A is rapidly absorbed into both cell lines causing an increase in the S-phase transition and a decrease in the M-phase transition. The model also indicates that the rate of apoptosis remains virtually constant for MCF-7/LY2 while that of A2780 increases 38% at 2.5 nM and 59% at 50 nM as compared to the untreated apoptotic rate.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Ciclopropanos/farmacologia , Modelos Biológicos , Neoplasias Ovarianas/tratamento farmacológico , Tiazóis/farmacologia , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Ciclopropanos/metabolismo , Feminino , Citometria de Fluxo , Humanos , Concentração Inibidora 50 , Cinética , Mitose/efeitos dos fármacos , Neoplasias Ovarianas/patologia , Tiazóis/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
10.
J Pharmacol Exp Ther ; 296(2): 364-71, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160619

RESUMO

The pentacyclic palmarumycins are structurally unique natural products with both antifungal and antibacterial activities but their antineoplastic effects are not well established. We have examined their antiproliferative actions against tumor cells using a temperature-sensitive tsFT210 mouse mammary carcinoma cell line and found that a novel palmarumycin analog, [8-(furan-3-ylmethoxy)-1-oxo-1,4-dihydronaphthalene-4-spiro-2'-naphtho[1",8"-de][1',3'][dioxin] or SR-7, prominently blocked mammalian cell cycle transition in G2/M but not in G1 phase. We found no evidence for inhibition of the critical mitosis-controlling cyclin-dependent kinase Cdk1, or its regulator, the dual specificity phosphatase Cdc25. Moreover, Cdk1 was hypophosphorylated and not directly inhibited by SR-7. SR-7 also failed in vitro to hypernucleate bovine tubulin, did not compete with colchicine for tubulin binding, and only modestly blocked GTP-induced assembly. In addition, SR-7 caused almost equal inhibition of paclitaxel-sensitive and -resistant cell growth. Moreover, unlike benchmark tubulin-disrupting agents, SR-7 did not cause hyperphosphorylation of the antiapoptotic protein Bcl-2. Thus, SR-7 represents a novel chemical structure that can inhibit G2/M transition by a mechanism that appears to be independent of marked tubulin disruption.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Dioxanos/farmacologia , Compostos de Espiro/farmacologia , Animais , Ligação Competitiva/efeitos dos fármacos , Western Blotting , Neoplasias da Mama/metabolismo , Divisão Celular/efeitos dos fármacos , Colchicina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Citometria de Fluxo , Humanos , Camundongos , Naftalenos , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Estrogênio/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
11.
J Biol Chem ; 276(2): 910-4, 2001 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-11036084

RESUMO

Members of the hsc70 family of molecular chaperones are critical players in the folding and quality control of cellular proteins. Because several human diseases arise from defects in protein folding, the activity of hsc70 chaperones is a potential therapeutic target for these disorders. By using a known hsc70 modulator, 15-deoxyspergualin, as a seed, we identified a novel inhibitor of hsc70 activity. This compound, R/1, inhibits the endogenous and DnaJ-stimulated ATPase activity of hsc70 by 48 and 51%, respectively, and blocks the hsc70-mediated translocation of a preprotein into yeast endoplasmic reticulum-derived microsomal vesicles. Biochemical studies demonstrate that R/1 most likely exerts these effects by altering the oligomeric state of hsc70.


Assuntos
Carbamatos/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Pirimidinonas/metabolismo , Antibióticos Antineoplásicos/farmacologia , Guanidinas/farmacologia , Proteínas de Choque Térmico HSC70 , Proteínas de Choque Térmico HSP40 , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico/metabolismo , Humanos , Membranas Intracelulares/fisiologia , Cinética , Microssomos/fisiologia , Transporte Proteico , Células Tumorais Cultivadas
12.
Prostate ; 45(4): 277-88, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11102952

RESUMO

BACKGROUND: Z-1,1-Dichloro-2,3-diphenylcyclopropane (A(II)) has long been known to be active against models of breast carcinoma. Microtubule perturbation and interaction at type II estrogen binding sites mediate its actions. METHODS: Since these targets are potentially useful for treatment of prostate tumors, we studied the drug's effects on androgen-sensitive (LNCaP) and -independent (PC-3) human prostatic carcinoma lines. Effects on cell growth and morphology, prostate-specific antigen (PSA) expression, and cell cycle kinetics were determined by microscopy, antibody-based methods, flow cytometry, and electrophoresis. RESULTS: At 100 microM, A(II) reduced survival of both lines by 50% in 12-24 hr, whereas 10 microM A(II) caused a prolonged block of proliferation in both lines, and parallel and complete block of PSA in LNCaP cells. At 10 microM, A(II) caused no major changes in chromatin, morphology or cell cycle distributions, whereas 100 microM drug caused rapid, large-scale cell detachment, nuclear and internucleosomal DNA fragmentation, and hypodiploidy. These effects were also accompanied by dissolution of cellular microtubule arrays. A more potent tubulin assembly-inhibiting congener of A(II), Z-1, 1-dichloro-2-(4-methoxy-phenyl)-3-phenylcyclopropane, slightly more effectively inhibited cell growth, caused little hypodiploidy, but potently and dose-dependently caused G(2)/M accumulation. CONCLUSIONS: These and previous data suggest that the Z-1, 1-dichloro-2,3-diarylcyclo-propanes may be useful in the treatment of human prostate disease.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Antígeno Prostático Específico/antagonistas & inibidores , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Androgênios/fisiologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Fragmentação do DNA , Relação Dose-Resposta a Droga , Moduladores de Receptor Estrogênico/farmacologia , Inibidores do Crescimento/farmacologia , Humanos , Masculino , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias Hormônio-Dependentes/imunologia , Neoplasias Hormônio-Dependentes/patologia , Antígeno Prostático Específico/biossíntese , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas/efeitos dos fármacos
14.
Chem Biol Interact ; 126(3): 171-83, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10862816

RESUMO

Quantitation of carcinogen-DNA adducts provides an estimate of the biologically effective dose of a chemical carcinogen reaching the target tissue. In order to improve exposure-assessment and cancer risk estimates, we are developing an ultrasensitive procedure for the detection of carcinogen-DNA adducts. The method is based upon postlabeling of carcinogen-DNA adducts by acetylation with 14C-acetic anhydride combined with quantitation of 14C by accelerator mass spectrometry (AMS). For this purpose, adducts of benzo[a]pyrene-r-7,t-8-dihydrodiol-t-9,10-epoxide (BPDE) with DNA and deoxyguanosine (dG) were synthesized. The most promutagenic adduct of BPDE, 7R,8S,9R-trihydroxy-10S-(N(2)-deoxyguanosyl)-7,8,9, 10-tetrahydrobenzo[a]pyrene (BPdG), was HPLC purified and structurally characterized. Postlabeling of the BPdG adduct with acetic anhydride yielded a major product with a greater than 60% yield. The postlabeled adduct was identified by liquid chromatography-mass spectrometry as pentakis(acetyl) BPdG (AcBPdG). Postlabeling of the BPdG adduct with 14C-acetic anhydride yielded a major product coeluting with an AcBPdG standard. Quantitation of the 14C-postlabeled adduct by AMS promises to allow detection of attomolar amounts of adducts. The method is now being optimized and validated for use in human samples.


Assuntos
Anidridos Acéticos/química , Benzo(a)pireno/análise , Carcinógenos Ambientais/análise , Adutos de DNA/análise , Espectrometria de Massas/métodos , Acetilação , Animais , Radioisótopos de Carbono , Bovinos , Cromatografia Líquida de Alta Pressão , Marcação por Isótopo/métodos , Espectrofotometria Atômica
15.
Radiat Res ; 154(1): 73-86, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10856968

RESUMO

Reverse transcription-polymerase chain reaction and immunofluorescence analysis of D2XRII murine bone marrow stromal cells showed that gamma irradiation with doses of 2-50 Gy from (137)Cs stimulated expression of nitric oxide synthase 2 (Nos2, also known as iNos). The activation of Nos2 was accompanied by an increase in the fluorescence of 4,5-diaminofluorescein diacetate, a nitric oxide trap, and accumulation of 3-nitrotyrosine within cellular proteins in a dose-dependent manner. These effects were inhibited by actinomycin D and by N-[3-(aminomethyl)benzyl]acetamidine dihydrochloride, a specific inhibitor of Nos2. The induction of Nos2 expression and Nos2-dependent release of nitric oxide in D2XRII cells was observed within 24 h after irradiation and was similar in magnitude to that observed in cultures incubated with Il1b and Tnf. We conducted (1) confocal fluorescence imaging of 3-nitrotyrosine in bone marrow cells of irradiated C57BL/6J mice and (2) 3-nitrotyrosine fluorescence imaging of FDC-P1JL26 hematopoietic cells that were cocultured with previously irradiated D2XRII bone marrow stromal cells. Exposure to ionizing radiation increased the production of 3-nitrotyrosine in irradiated bone marrow cells in vivo and in nonirradiated FDC-P1JL26 cells cocultured with irradiated D2XRII cells for 1 or 4 h. We suggest that nitrative/oxidative stress to the transplanted multilineage hematopoietic cells due to exposure to nitric oxide released by host bone marrow stromal cells may contribute to the genotoxic events associated with malignant alterations in bone marrow tissue of transplant recipients who are prepared for engraftment by total-body irradiation.


Assuntos
Células da Medula Óssea/efeitos da radiação , Óxido Nítrico Sintase/metabolismo , Tirosina/análogos & derivados , Animais , Células da Medula Óssea/enzimologia , Comunicação Celular , Ativação Enzimática , Imunofluorescência , Células-Tronco Hematopoéticas/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase Tipo II , Doses de Radiação , Radiação Ionizante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/enzimologia , Células Estromais/efeitos da radiação , Tirosina/metabolismo
16.
Chem Res Toxicol ; 12(10): 931-6, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10525268

RESUMO

Diisocyanatotoluene (toluene diisocyanate, TDI), a 4:1 mixture of 2, 4- and 2,6-isomers used in the preparation of polyurethanes, causes occupational asthma by an as yet unknown mechanism. We previously showed that it forms adducts with the apical surface of the bronchoepithelium in vivo, and with ciliary microtubules in cultured human bronchoepithelial (HBE) cells. These results suggested that TDI may not enter HBE cells. In vitro studies, however, showed that TDI avidly forms bis adducts with glutathione (GSH) and that these adducts transfer monoisocyanato-monoglutathionyl-TDI to a sulfhydryl-containing peptide. This study sought to elucidate intracellular reactions of TDI. Using an electron paramagnetic resonance spectrometric (EPR) method, we established that the level of thiol-dependent quenching of phenoxyl radicals of etoposide was decreased >40% in pulmonary tissue of mice that received TDI intrabronchially. Similarly, HBE cells exposed to 100 ppb TDI vapor experienced a >30% reduction in thiol levels as determined with a thiol-specific fluorescent probe (ThioGlo 1). HPLC/UV analysis of lysates from HBE cells exposed to 200 and 500 ppb TDI vapor suggested a dose-related formation of S-glutathionyl adducts. Data from the 500 ppb TDI-treated HBE cells verified the identity of the 2-monoglutathionyl-4-monoisocyanato adduct. The results provide firm evidence that TDI enters pulmonary cells and reacts with GSH. This rapid reaction leading to formation of S-glutathionyl adducts of TDI suggests the importance of cellular thiols in TDI-induced pulmonary disease.


Assuntos
Brônquios/metabolismo , Células Epiteliais/metabolismo , Glutationa/metabolismo , Irritantes/toxicidade , Pulmão/metabolismo , Tolueno 2,4-Di-Isocianato/toxicidade , Animais , Antineoplásicos Fitogênicos/toxicidade , Brônquios/citologia , Brônquios/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Espectroscopia de Ressonância de Spin Eletrônica , Células Epiteliais/efeitos dos fármacos , Etoposídeo/toxicidade , Feminino , Sequestradores de Radicais Livres/farmacologia , Humanos , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas/metabolismo , Espectrofotometria Ultravioleta , Análise Espectral , Compostos de Sulfidrila/farmacologia
17.
Chem Res Toxicol ; 12(1): 28-37, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9894015

RESUMO

Some studies have shown the potential relevance of the oxidation products of 4-hydroxytamoxifen (4OHTAM) in carcinogenesis. Other studies show 4OHTAM has antioxidant properties. We characterized the one-electron oxidative activation reactions of 4OHTAM and three other phenolics, 3-hydroxytamoxifen (3OHTAM), 1-(4-hydroxyphenyl)-1, 2-diphenylethene, and phenol (PhOH), catalyzed by myeloperoxidase (MPx), horseradish peroxidase (HRP), lactoperoxidase, mushroom tyrosinase, and nonenzymatic initiators in vitro under a variety of conditions and in cells. Differences in activation of the phenolics by the enzymes were directly compared using cis-parinaric acid (PnA)-loaded human serum albumin. All phenolics were substrates for the enzymes, but MPx only weakly activated 4OHTAM to its phenoxyl radical. In HL60 cells loaded metabolically with PnA so that effects on phospholipids could be monitored by HPLC with fluorescence detection, PhOH plus H2O2 caused massive oxidation across all phospholipid classes. 4OHTAM dose-dependently protected phosphatidylethanolamine, phosphatidylserine, and phosphatidylcholine against both H2O2-induced and normal metabolic oxidation. This suggested 4OHTAM is a poor substrate for intracellular MPx. In rat aorta smooth muscle cells loaded with PnA, 4OHTAM also protected against AMVN-induced peroxidation of those three phospholipids and sphingomyelin, whereas 3OHTAM did not. Spin trapping of glutathionyl radicals (GS*) with DMPO and quantifying the ESR-silent nitrone form of the GS-DMPO adduct by HPLC showed that neither 3OHTAM plus H2O2 nor 4OHTAM plus H2O2 caused a significant level of GSH oxidation with isolated MPx, nor did the latter in HL60 cells, whereas PhOH plus H2O2 was a potent source of GS* in both systems. Both 4OHTAM and 3OHTAM formed the nitrone adduct under cell-free conditions when activated with HRP. The data show that the substrate specificity of a given (myelo)peroxidase determines if a phenolic exerts pro- (through generation of reactive phenoxyl radicals) or antioxidant (through radical scavenging) properties in intracellular environments.


Assuntos
Antioxidantes/farmacologia , Antagonistas de Estrogênios/farmacologia , Oxidantes/farmacologia , Peroxidases/metabolismo , Tamoxifeno/análogos & derivados , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ácidos Graxos/química , Glutationa/metabolismo , Células HL-60 , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Oxidantes/toxicidade , Oxirredução , Peroxidase/metabolismo , Fenol/farmacologia , Fosfolipídeos/química , Ratos , Albumina Sérica/química , Tamoxifeno/farmacologia , Tamoxifeno/toxicidade
18.
Biochem Pharmacol ; 57(1): 97-110, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9920290

RESUMO

We have shown previously that Z-1,1-dichloro-2,3-diphenylcyclopropane (a.k.a. Analog II, A(II)) inhibits human breast cancer cell proliferation regardless of estrogen receptor status or estrogen sensitivity, and that its cellular targets include microtubules. In the present study, we investigated the apoptosis-inducing effects of A(II). MCF-7, MCF-7/LY2, and MDA-MB-231 cells all showed nuclear fragmentation in response to 100 microM A(II) when stained with Hoechst 33342 and examined by fluorescence microscopy. Pulsed field gel electrophoretic analysis showed that each of the cell lines also developed specific high molecular weight DNA fragments: a low level of 1-2 Mb fragments appeared after 6 hr, while 30-50 kb fragments accumulated subsequently. At 24 hr of drug exposure, the majority of cells became nonadherent, and the 30-50 kb fragments were restricted to detached MCF-7 and MDA-MB-231 cells. Both adherent and detached MCF-7/LY2 cells exhibited these fragments. A previous study by single-color (propidium) flow cytometry demonstrated that A(II) blocks MDA-MB-231 cells in G2/M of the cell cycle. More refined analyses in the present study showed this same result for MDA-MB-231 cells, but MCF-7 and MCF-7/LY2 cells did not reveal apparent drug-induced cell cycle block. A(II) demonstrated growth inhibitory, cell cycle-perturbing, and hypodiploidy-inducing activity against other human breast carcinoma lines, i.e. BT-20, CAMA-1, and SKBR-3, but no such actions in the non-tumorigenic, "normal" human breast epithelial line MCF-10A. Bromodeoxyuridine labeling and two-color flow cytometric analysis, however, suggested that A(II) caused stimulation into S phase, and that G2/M was the phase of the cell cycle from which cells apoptosed. A(II) caused cell rounding, detachment from the growth matrix, and nuclear shrinkage and fragmentation in parallel with biochemical changes. Cycloheximide inhibited A(II)-induced cell death, indicating that its toxicity requires de novo protein synthesis.


Assuntos
Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Tamoxifeno/análogos & derivados , Benzimidazóis , Neoplasias da Mama , Ciclo Celular/fisiologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Fragmentação do DNA , DNA de Neoplasias/efeitos dos fármacos , Eletroforese em Gel de Campo Pulsado , Feminino , Fase G2 , Humanos , Mitose , Estrutura Molecular , Tamoxifeno/toxicidade , Células Tumorais Cultivadas
19.
Brain Res Mol Brain Res ; 60(2): 270-81, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9757062

RESUMO

Incubation of mock-transfected PC12 rat pheochromocytoma cells (PC12) for 2 h with increasing concentrations of glutamate caused progressive loss of viability (e.g., 67% with 15 mM glutamate). In contrast, the viability of bcl-2-transfected cells (PC12/bcl-2) was unaffected by glutamate. Neither PC12 nor PC12/bcl-2 cells showed a significant incidence of apoptosis in response to glutamate. Conventional phospholipid analysis by high-performance TLC and phosphorous determination showed no significant changes in the phospholipid composition of either cell line incubated with 5 mM glutamate. The peroxyl radical initiator 2,2'-azobis(2,4-dimethylvaleronitrile) caused a pronounced loss of all major phospholipid classes in PC12 cells, but no loss of cell viability. No phospholipid peroxidation was detected in PC12/bcl-2 cells incubated with

Assuntos
Sobrevivência Celular/fisiologia , Genes bcl-2 , Ácido Glutâmico/toxicidade , Glutationa/metabolismo , Fosfolipídeos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Neoplasias das Glândulas Suprarrenais , Animais , Compostos Azo/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cinética , Proteínas de Neoplasias/metabolismo , Nitrilas/farmacologia , Oxirredução , Células PC12 , Feocromocitoma , Proteínas Proto-Oncogênicas c-bcl-2/genética , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Compostos de Sulfidrila/metabolismo , Transfecção
20.
DNA Cell Biol ; 17(6): 541-9, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9655247

RESUMO

In vitro DNA replication by exonuclease-deficient T7 DNA polymerase (Sequenase) and an exonuclease deficient T4 DNA polymerase was examined on a 244-nucleotide DNA template treated with three electrophilic polycyclic aromatic hydrocarbon (PAH) metabolites: racemic trans-7,8-dihydroxy-anti-9,10-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene (BaPDE), trans-2,3-dihydroxy-anti-1,10b-epoxy-10b,1,2,3-tetrahydrofluoranthene (FADE), or 3,4-epoxy-3,4-dihydrocyclopenta[cd]pyrene (CPPE). The DNA replication terminated opposite template guanines and, to a lesser extent, at template adenines, as expected, as purines were modified preferentially by the chemical treatments. Analysis of the products synthesized on the damaged templates indicated that bypass replication by Sequenase proceeded in three steps: (1) replication first terminated one base 3' to each adduct; (2) a nucleotide was then incorporated opposite the PAH-modified base; and (3) replication continued at some sites to give full bypass of the lesions. The rate of lesion bypass was affected by the type of chemical adduct, the sequence context of the adduct, and the concentration of deoxynucleoside triphosphates. Short DNA repeats appeared to facilitate translesion replication.


Assuntos
Adutos de DNA , Replicação do DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/metabolismo , Exonucleases/metabolismo , Hidrocarbonetos Policíclicos Aromáticos/farmacologia , Proteínas Virais/metabolismo , Bacteriófago T4/enzimologia , Bacteriófago T7/enzimologia , Sequência de Bases , Dano ao DNA , Primers do DNA , Hidrocarbonetos Policíclicos Aromáticos/farmacocinética , Sequências Repetitivas de Ácido Nucleico , Moldes Genéticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...