Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Endocrinol ; 28(6): 949-64, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24801505

RESUMO

Androgen receptor (AR) action throughout prostate development and in maintenance of the prostatic epithelium is partly controlled by interactions between AR and forkhead box (FOX) transcription factors, particularly FOXA1. We sought to identity additional FOXA1 binding partners that may mediate prostate-specific gene expression. Here we identify the nuclear factor I (NFI) family of transcription factors as novel FOXA1 binding proteins. All four family members (NFIA, NFIB, NFIC, and NFIX) can interact with FOXA1, and knockdown studies in androgen-dependent LNCaP cells determined that modulating expression of NFI family members results in changes in AR target gene expression. This effect is probably mediated by binding of NFI family members to AR target gene promoters, because chromatin immunoprecipitation (ChIP) studies found that NFIB bound to the prostate-specific antigen enhancer. Förster resonance energy transfer studies revealed that FOXA1 is capable of bringing AR and NFIX into proximity, indicating that FOXA1 facilitates the AR and NFI interaction by bridging the complex. To determine the extent to which NFI family members regulate AR/FOXA1 target genes, motif analysis of publicly available data for ChIP followed by sequencing was undertaken. This analysis revealed that 34.4% of peaks bound by AR and FOXA1 contain NFI binding sites. Validation of 8 of these peaks by ChIP revealed that NFI family members can bind 6 of these predicted genomic elements, and 4 of the 8 associated genes undergo gene expression changes as a result of individual NFI knockdown. These observations suggest that NFI regulation of FOXA1/AR action is a frequent event, with individual family members playing distinct roles in AR target gene expression.


Assuntos
Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fatores de Transcrição NFI/metabolismo , Próstata/metabolismo , Proteína de Ligação a Androgênios/genética , Sequência de Bases , Sítios de Ligação , Sequência Consenso , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Células HeLa , Humanos , Masculino , Especificidade de Órgãos , Regiões Promotoras Genéticas , Ligação Proteica , Mapeamento de Interação de Proteínas , Receptores Androgênicos/metabolismo , Transcrição Gênica
2.
J Mass Spectrom ; 49(2): 178-83, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24677308

RESUMO

Oxidative stress plays an important role in the development of various disease processes and is a putative mechanism in the development of bronchopulmonary dysplasia, the most common complication of extreme preterm birth. Glutathione, a major endogenous antioxidant and redox buffer, also mediates cellular functions through protein thiolation. We sought to determine if post-translational thiol modification of hemoglobin F occurs in neonates by examining erythrocyte samples obtained during the first month of life from premature infants, born at 23 0/7 - 28 6/7 weeks gestational age, who were enrolled at our center in the Prematurity and Respiratory Outcomes Program (PROP). Using liquid chromatography-mass spectrometry (LC-MS), we report the novel finding of in vivo and in vitro glutathionylation of γG and γA subunits of Hgb F. Through tandem mass spectrometry (nanoLC-MS/MS), we confirmed the adduction site as the Cys-γ94 residue and through high-resolution mass spectrometry determined that the modification occurs in both γ subunits. We also identified glutathionylation of the ß subunit of Hgb A in our patient samples; we did not find modified α subunits of Hgb A or F. In conclusion, we are the first to report that glutathionylation of γG and γA of Hgb F occurs in premature infants. Additional studies of this post-translational modification are needed to determine its physiologic impact on Hgb F function and if sG-Hgb is a biomarker for clinical morbidities associated with oxidative stress in premature infants.


Assuntos
Cromatografia Líquida/métodos , Hemoglobina Fetal/análise , Hemoglobina Fetal/química , Glutationa/química , Lactente Extremamente Prematuro/sangue , Espectrometria de Massas em Tandem/métodos , Sequência de Aminoácidos , Humanos , Recém-Nascido , Dados de Sequência Molecular , Espectrometria de Massas por Ionização por Electrospray
3.
Clin Cancer Res ; 17(11): 3794-802, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21346148

RESUMO

PURPOSE: This phase I study assessed the maximum tolerated dose (MTD), safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of SJG-136, a sequence-specific DNA cross-linking agent, in patients with advanced cancer. EXPERIMENTAL DESIGN: In schedule A, seven patients received escalating doses of SJG-136 (6, 12, 24, and 48 µg/m(2)) daily for 5 of 21 days. Blood samples were collected for PK analysis on days 1 and 5 of cycle 1. In schedule B, SJG-136 was given daily for 3 of 21 days (N = 17; doses 20, 25, 30, and 35 µg/m(2)). Blood samples were collected on days 1 and 3 of cycles 1 and 2 for PK and PD analysis. Patients in schedule B received dexamethasone and early diuretic care. RESULTS: Schedule A-dose-limiting toxicities included grade 3 edema, dyspnea, fatigue, and delayed liver toxicity (grade 3-4). PK analysis revealed dose-dependent increases in AUC and C(max). Substantial changes in volume of distribution at steady-state occurred after repeated dosing in some patients prior to the onset of edema. Schedule B-the same toxicities were manageable with steroid premedication and diuretic support. No significant myelosuppression occurred on either schedule. DNA interstrand cross-links correlated with systemic exposure of SJG-136 following the second dose in cycle 1 and were still detectable immediately before cycle 2. CONCLUSIONS: The MTD of SJG-136 in this study was 30 µg/m(2) administered on a daily 3× basis with no myelosuppression effects. Coupled with supportive management, SJG-136 is now advancing to a phase II trial in ovarian cancer.


Assuntos
Benzodiazepinonas/farmacologia , Dose Máxima Tolerável , Neoplasias/tratamento farmacológico , Pirróis/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Benzodiazepinonas/efeitos adversos , Benzodiazepinonas/metabolismo , Benzodiazepinonas/farmacocinética , Dexametasona/administração & dosagem , Dispneia/induzido quimicamente , Edema/induzido quimicamente , Fadiga/induzido quimicamente , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pirróis/efeitos adversos , Pirróis/metabolismo , Pirróis/farmacocinética
4.
Neurobiol Dis ; 39(2): 169-80, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20394819

RESUMO

RNA transcripts encoding the 2C-subtype of serotonin (5HT(2C)) receptor undergo up to five adenosine-to-inosine editing events to encode twenty-four protein isoforms. To examine the effects of altered 5HT(2C) editing in vivo, we generated mutant mice solely expressing the fully-edited (VGV) isoform of the receptor. Mutant animals present phenotypic characteristics of Prader-Willi syndrome (PWS) including a failure to thrive, decreased somatic growth, neonatal muscular hypotonia, and reduced food consumption followed by post-weaning hyperphagia. Though previous studies have identified alterations in both 5HT(2C) receptor expression and 5HT(2C)-mediated behaviors in both PWS patients and mouse models of this disorder, to our knowledge the 5HT(2C) gene is the first locus outside the PWS imprinted region in which mutations can phenocopy numerous aspects of this syndrome. These results not only strengthen the link between the molecular etiology of PWS and altered 5HT(2C) expression, but also demonstrate the importance of normal patterns of 5HT(2C) RNA editing in vivo.


Assuntos
Regulação da Expressão Gênica/genética , Síndrome de Prader-Willi/genética , Edição de RNA/genética , Receptor 5-HT2C de Serotonina/genética , Animais , Animais Recém-Nascidos , Análise Mutacional de DNA , Modelos Animais de Doenças , Comportamento Alimentar/fisiologia , Feminino , Força da Mão/fisiologia , Humanos , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Mutação/genética , Síndrome de Prader-Willi/patologia , Síndrome de Prader-Willi/fisiopatologia , Desempenho Psicomotor/fisiologia , RNA Mensageiro/metabolismo
5.
Biophys J ; 95(3): 1371-81, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18441025

RESUMO

Tissue ablation with mid-infrared irradiation tuned to collagen vibrational modes results in minimal collateral damage. The hypothesis for this effect includes selective scission of protein molecules and excitation of surrounding water molecules, with the scission process currently favored. In this article, we describe the postablation infrared spectral decay kinetics in a model collagen-like peptide (Pro-Pro-Gly)(10). We find that the decay is exponential with different decay times for other, simpler dipeptides. Furthermore, we find that collagen-like polypeptides, such as (Pro-Pro-Gly)(10), show multiple decay times, indicating multiple scission locations and cross-linking to form longer chain molecules. In combination with data from high-resolution mass spectrometry, we interpret these products to result from the generation of reactive intermediates, such as free radicals, cyanate ions, and isocyanic acid, which can form cross-links and protein adducts. Our results lead to a more complete explanation of the reduced collateral damage resulting from infrared laser irradiation through a mechanism involving cross-linking in which collagen-like molecules form a network of cross-linked fibers.


Assuntos
Colágeno/química , Colágeno/efeitos da radiação , Modelos Químicos , Modelos Moleculares , Peptídeos/química , Peptídeos/efeitos da radiação , Colágeno/ultraestrutura , Simulação por Computador , Elétrons , Raios Infravermelhos , Lasers , Doses de Radiação
6.
J Mass Spectrom ; 43(1): 42-52, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17683023

RESUMO

SJG-136 1,1'-[[(propane-1,3-diyl)dioxy]bis[(11aS)-7-methoxy-2-methylidene-1,2,3,11a-tetrahydro-5H-pyr- rolo[2,1-c][1,4]benzodiazepin-5-one]] (NSC 694501), is a bifunctional pyrrolobenzodiazepine (PBD) dimer that forms selective, irreversible, interstrand DNA cross-links via exocyclic N2 atoms of two guanine bases, with a preference for 5'PuGATCPy binding sites. SJG-136 is highly cytotoxic in human tumor cells in vitro and in human tumor xenograft models in vivo at subnanomolar concentrations and is currently in anticancer phase I clinical trials in the United Kingdom and United States. To support correlative pharmacokinetics studies, a highly sensitive HPLC-MS/MS assay was developed and validated for the reliable quantitation of SJG-136 in human plasma, using the structurally similar PBD dimer DSB-120 as an internal standard. Chemical reduction of SJG-136 to its corresponding amine (SJG-136-H(4), [M + H](+)m/z 561) improved HPLC peak resolution and sensitivity by minimizing complications that arose from the reactivity of the labile imine moieties. Plasma samples were processed by protein precipitation and centrifugal membrane dialysis; components were separated by HPLC using an Agilent Rapid Resolution HT 1.8 mm (2.1 mm x 50 mm) analytical column. The total analysis time from injection to injection was 11 min. Electrospray MS/MS detection of SJG-136-H(4) was based on the selected reaction monitoring (SRM) transition [M + H](+)m/z 561 --> 301. The analytical response ratio was linearly proportional to the plasma concentration of SJG-136 over the nominal concentration range of 25 pg/ml to 250 ng/ml, with a coefficient of determination of r > or = 0.999. The intrarun absolute %RE was < or =19.6, 14.2, and 14.0% at 0.056, 2.83, and 56.3 ng/ml, respectively. The corresponding %RSD was < or =14.9%, 9.01, and 4.59%. The interday %RSD was < or =2.72, 3.46, and 5.20%. The lower and upper limits of quantitation were 0.056 and 56 ng/ml, respectively; recovery of SJG-136 from plasma was > or = 62% across the validated concentration range. The sensitivity of the validated assay was sufficient to detect SJG-136 in human subjects for up to 6 h after intravenous administration of 6 microg/m(2), the starting dose of an NCI-sponsored dose escalation study.


Assuntos
Antineoplásicos/sangue , Benzodiazepinonas/sangue , Cromatografia Líquida de Alta Pressão , Ensaios Clínicos Fase I como Assunto , Pirróis/sangue , Espectrometria de Massas por Ionização por Electrospray/métodos , Espectrometria de Massas em Tandem/métodos , Antineoplásicos/farmacocinética , Benzodiazepinonas/farmacocinética , Relação Dose-Resposta a Droga , Humanos , Oxirredução , Pirróis/farmacocinética , Sensibilidade e Especificidade
7.
Cancer Res ; 67(2): 812-7, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234793

RESUMO

The oxidative metabolism of estrogens has been implicated in the development of breast cancer; yet, relatively little is known about the mechanism by which estrogens cause DNA damage and thereby initiate mammary carcinogenesis. To determine how the metabolism of the parent hormone 17beta-estradiol (E2) leads to the formation of DNA adducts, we used the recombinant, purified phase I enzyme, cytochrome P450 1B1 (CYP1B1), which is expressed in breast tissue, to oxidize E2 in the presence of 2'-deoxyguanosine or 2'-deoxyadenosine. We used both gas and liquid chromatography with tandem mass spectrometry to measure E2, the 2- and 4-catechol estrogens (2-OHE2, 4-OHE2), and the depurinating adducts 4-OHE(2)-1(alpha,beta)-N7-guanine (4-OHE2-N7-Gua) and 4-OHE(2)-1(alpha,beta)-N3-adenine (4-OHE2-N3-Ade). CYP1B1 oxidized E2 to the catechol 4-OHE2 and the labile quinone 4-hydroxyestradiol-quinone to produce 4-OHE2-N7-Gua and 4-OHE2-N3-Ade in a time- and concentration-dependent manner. Because the reactive quinones were produced as part of the CYP1B1-mediated oxidation reaction, the adduct formation followed Michaelis-Menten kinetics. Under the conditions of the assay, the 4-OHE2-N7-Gua adduct (Km, 4.6+/-0.7 micromol/L; kcat, 45+/-1.6/h) was produced 1.5 times more efficiently than the 4-OHE2-N3-Ade adduct (Km, 4.6+/-1.0 micromol/L; kcat, 30+/-1.5/h). The production of adducts was two to three orders of magnitude lower than the 4-OHE2 production. The results present direct proof of CYP1B1-mediated, E2-induced adduct formation and provide the experimental basis for future studies of estrogen carcinogenesis.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Desoxiadenosinas/metabolismo , Desoxiguanosina/metabolismo , Estradiol/metabolismo , Citocromo P-450 CYP1B1 , Adutos de DNA/metabolismo , Estrogênios de Catecol/metabolismo , Cinética , Proteínas Recombinantes/metabolismo
8.
Cancer Epidemiol Biomarkers Prev ; 15(9): 1620-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16985022

RESUMO

Oxidative metabolites of estrogens have been implicated in the development of breast cancer, yet relatively little is known about the metabolism of estrogens in the normal breast. We developed a mathematical model of mammary estrogen metabolism based on the conversion of 17beta-estradiol (E(2)) by the enzymes cytochrome P450 (CYP) 1A1 and CYP1B1, catechol-O-methyltransferase (COMT), and glutathione S-transferase P1 into eight metabolites [i.e., two catechol estrogens, 2-hydroxyestradiol (2-OHE(2)) and 4-hydroxyestradiol (4-OHE(2)); three methoxyestrogens, 2-methoxyestradiol, 2-hydroxy-3-methoxyestradiol, and 4-methoxyestradiol; and three glutathione (SG)-estrogen conjugates, 2-OHE(2)-1-SG, 2-OHE(2)-4-SG, and 4-OHE(2)-2-SG]. When used with experimentally determined rate constants with purified enzymes, the model provides for a kinetic analysis of the entire metabolic pathway. The predicted concentration of each metabolite during a 30-minute reaction agreed well with the experimentally derived results. The model also enables simulation for the transient quinones, E(2)-2,3-quinone (E(2)-2,3-Q) and E(2)-3,4-quinone (E(2)-3,4-Q), which are not amenable to direct quantitation. Using experimentally derived rate constants for genetic variants of CYP1A1, CYP1B1, and COMT, we used the model to simulate the kinetic effect of enzyme polymorphisms on the pathway and identified those haplotypes generating the largest amounts of catechols and quinones. Application of the model to a breast cancer case-control population identified a subset of women with an increased risk of breast cancer based on their enzyme haplotypes and consequent E(2)-3,4-Q production. This in silico model integrates both kinetic and genomic data to yield a comprehensive view of estrogen metabolomics in the breast. The model offers the opportunity to combine metabolic, genetic, and lifetime exposure data in assessing estrogens as a breast cancer risk factor.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Neoplasias da Mama/etiologia , Catecol O-Metiltransferase/genética , Citocromo P-450 CYP1A1/genética , Estrogênios/metabolismo , Área Sob a Curva , Neoplasias da Mama/metabolismo , Citocromo P-450 CYP1B1 , Feminino , Haplótipos , Humanos , Modelos Teóricos
9.
J Biol Chem ; 280(51): 42149-55, 2005 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-16251182

RESUMO

The small molecule 3-O-(3',3'-dimethylsuccinyl)-betulinic acid (DSB) potently inhibits human immunodeficiency virus, type 1 (HIV-1) replication by interfering with proteolytic cleavage of the viral Gag protein at a specific site. Here we have demonstrated that the antiviral mechanism involves the association of DSB with Gag at a 1:1 stoichiometry within immature HIV-1 particles. The binding was specific, as mutations in Gag that confer resistance to DSB inhibited the association, which could be competed by DSB but not by the inactive compound betulinic acid. The addition of DSB to purified immature viral cores inhibited the cleavage of Gag at the CA-SP1 junction in vitro, thus reproducing the effect of the drug when present during maturation of HIV-1 particles. Based on these findings, we propose a model in which a trimer of DSB associates with the CA-SP1 junction of adjacent subunits within the Gag polymer. The model may explain the ability of highly similar compounds to specifically target the seemingly unrelated steps of HIV-1 maturation and virus entry.


Assuntos
Produtos do Gene gag/fisiologia , HIV-1/fisiologia , Vírion/fisiologia , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Produtos do Gene gag/efeitos dos fármacos , Humanos
10.
J Lipid Res ; 46(9): 1933-43, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15995174

RESUMO

7-Ketocholesterol (7KC) is a cytotoxic component of oxidized low density lipoproteins (OxLDLs) and induces apoptosis in macrophages by a mechanism involving the activation of cytosolic phospholipase A2 (cPLA2). In the current study, we examined the role of ACAT in 7KC-induced and OxLDL-induced apoptosis in murine macrophages. An ACAT inhibitor, Sandoz 58-035, suppressed 7KC-induced apoptosis in P388D1 cells and both 7KC-induced and OxLDL-induced apoptosis in mouse peritoneal macrophages (MPMs). Furthermore, compared with wild-type MPMs, ACAT-1-deficient MPMs demonstrated significant resistance to both 7KC-induced and OxLDL-induced apoptosis. Macrophages treated with 7KC accumulated ACAT-derived [14C]cholesteryl and [3H]7-ketocholesteryl esters. Tandem LC-MS revealed that the 7KC esters contained primarily saturated and monounsaturated fatty acids. An inhibitor of cPLA2, arachidonyl trifluoromethyl ketone, prevented the accumulation of 7KC esters and inhibited 7KC-induced apoptosis in P388D1 cells. The decrease in 7KC ester accumulation produced by the inhibition of cPLA2 was reversed by supplementing with either oleic or arachidonic acid (AA); however, only AA supplementation restored the induction of apoptosis by 7KC. These results suggest that 7KC not only initiates the apoptosis pathway by activating cPLA2, as we have reported previously, but also participates in the downstream signaling pathway when esterified by ACAT to form 7KC-arachidonate.


Assuntos
Apoptose/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Macrófagos/fisiologia , Esterol O-Aciltransferase/fisiologia , Animais , Ácido Araquidônico/antagonistas & inibidores , Ácido Araquidônico/farmacologia , Radioisótopos de Carbono , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Ésteres do Colesterol/metabolismo , Inibidores Enzimáticos/farmacologia , Esterificação/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Cetocolesteróis/metabolismo , Cetocolesteróis/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipases A/antagonistas & inibidores , Fosfolipases A/metabolismo , Fosfolipases A2 , Esterol O-Aciltransferase/antagonistas & inibidores , Trítio
11.
Phys Med Biol ; 50(8): 1885-99, 2005 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15815102

RESUMO

Previous studies have shown that changing the pulse structure of the free electron laser (FEL) from 1 to 200 ps and thus reducing the peak irradiance of the micropulse by 200 times had little or no effect on both the ablation threshold radiant exposure and the ablated crater depth for a defined radiant exposure. This study focuses on the ablation mechanism at 6.1 and 6.45 microm with an emphasis on the role of the FEL pulse structure. Three different experiments were performed to gain insight into this mechanism. The first was an analysis of the ablation plume dynamics observed for a 1 ps micropulse compared with a 200 ps micropulse as seen through bright-field analysis. Negligible differences are seen in the size, but not the dynamics of ablation, as a result of this imaging. The second experiment was a histological analysis of corneal and dermal tissue to determine whether there is less thermal damage associated with one micropulse duration versus another. No significant difference was seen in the extent of thermal damage on either canine cornea or mouse dermis for the micropulse durations studied at either wavelength. The final set of experiments involved the use of mass spectrometry to determine whether amide bond breakage could occur in the proteins present in tissue as a result of direct absorptions of mid-infrared light into the amide I and amide II absorption bands. This analysis showed that there was no amide bond breakage due to irradiation at 6.45 microm on protein.


Assuntos
Córnea/patologia , Córnea/efeitos da radiação , Terapia a Laser/métodos , Lesões por Radiação/prevenção & controle , Radiometria/métodos , Pele/patologia , Pele/efeitos da radiação , Animais , Córnea/cirurgia , Procedimentos Cirúrgicos Dermatológicos , Fracionamento da Dose de Radiação , Relação Dose-Resposta à Radiação , Elétrons/uso terapêutico , Raios Infravermelhos/uso terapêutico , Luz , Camundongos , Doses de Radiação , Resultado do Tratamento
12.
Clin Cancer Res ; 10(23): 7865-74, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15585619

RESUMO

As molecularly targeted agents reach the clinic, there is a need for assays to detect their presence and effectiveness against target molecules in vivo. Proteinase inhibitors are one example of a class of therapeutic agent for which satisfactory methods of identifying successful target modulation in vivo are lacking. This is of particular importance while these drugs are in clinical trials because standard maximum-tolerated dose-finding studies often are not suitable due to lack of toxicity. Saliva represents a readily accessible bodily fluid that can be repeatedly sampled and used for assaying in vivo effects of systemic drugs. Here we show the development of a simple assay that can be used to measure proteinase activity in saliva and proteinase inhibition after systemic treatment with three different proteinase inhibitors. A variety of gelatinolytic activities present in human and murine saliva have been assayed with a fluorescent dye-labeled substrate and assigned to different proteinase categories by inclusion of specific classes of inhibitors. Treatment of mice with either matrix metalloproteinase inhibitors or a urokinase inhibitor for a period as short as 48 hours results in levels of the drugs that can be detected in saliva by mass spectrometry and concomitant decreases in salivary proteinase activity, thus demonstrating that these inhibitors successfully modulate their targets in vivo.


Assuntos
Biomarcadores/metabolismo , Endopeptidases/metabolismo , Fenilalanina/análogos & derivados , Inibidores de Proteases/farmacologia , Saliva/efeitos dos fármacos , Saliva/enzimologia , Amidinas/farmacologia , Amilases/metabolismo , Animais , Proteínas Sanguíneas/farmacologia , Cromatografia Líquida , Feminino , Humanos , Imidazóis , Masculino , Espectrometria de Massas , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/farmacologia , Camundongos , Compostos Orgânicos/farmacologia , Fenilalanina/farmacologia , Tiofenos/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores
13.
Chem Res Toxicol ; 17(9): 1258-64, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15377160

RESUMO

Estrogens and their oxidative metabolites, the catechol estrogens, have been implicated in the development of breast cancer; yet, relatively little is known about estrogen metabolism in the breast. To determine how the parent hormone, 17 beta-estradiol (E(2)), is metabolized, we used recombinant, purified phase I enzymes, cytochrome P450 (CYP) 1A1 and 1B1, with the phase II enzymes catechol-O-methyltransferase (COMT) and glutathione S-transferase P1 (GSTP1), all of which are expressed in breast tissue. We employed both gas and liquid chromatography with mass spectrometry to measure E(2), the catechol estrogens 2-hydroxyestradiol (2-OHE(2)) and 4-hydroxyestradiol (4-OHE(2)), as well as methoxyestrogens and estrogen-GSH conjugates. The oxidation of E(2) to 2-OHE(2) and 4-OHE(2) was exclusively regulated by CYP1A1 and 1B1, regardless of the presence or concentration of COMT and GSTP1. COMT generated two products, 2-methoxyestradiol and 2-hydroxy-3-methoxyestradiol, from 2-OHE(2) but only one product, 4-methoxyestradiol, from 4-OHE(2). Similarly, GSTP1 yielded two conjugates, 2-OHE(2)-1-SG and 2-OHE(2)-4-SG, from the corresponding quinone 2-hydroxyestradiol-quinone and one conjugate, 4-OHE(2)-2-SG, from 4-hydroxyestradiol-quinone. Using the experimental data, we developed a multicompartment kinetic model for the oxidative metabolism of the parent hormone E(2), which revealed significant differences in rate constants for its C-2 and C-4 metabolites. The results demonstrated a tightly regulated interaction of phase I and phase II enzymes, in which the latter decreased the concentration of catechol estrogens and estrogen quinones, thereby reducing the potential of these oxidative estrogen metabolites to induce DNA damage.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Estradiol/análogos & derivados , Estradiol/metabolismo , Estrogênios de Catecol/metabolismo , Glândulas Mamárias Humanas/enzimologia , Animais , Catecol O-Metiltransferase/metabolismo , Citocromo P-450 CYP1B1 , Glutationa S-Transferase pi , Glutationa Transferase/metabolismo , Humanos , Isoenzimas/metabolismo , Cinética , Glândulas Mamárias Animais/enzimologia , Modelos Químicos , Modelos Moleculares , Oxirredução , Proteínas Recombinantes , Relação Estrutura-Atividade
14.
J Am Chem Soc ; 126(3): 720-1, 2004 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-14733538

RESUMO

Strategies are reported that combine in one step a predictable chemical-based protein digestion with mass spectrometry. Lysine residue amino groups in peptides and proteins are modified by reaction with a peroxycarbonate derived from p-nitrophenol, and tert-butyl hydroperoxide. The peroxycarbonate reacts with lysine residues in peptides and proteins, and the resulting lysine peroxycarbamates undergo homolytic fragmentation under conditions of low-energy collision-induced dissociation (CID). Observed fragmentation of the peptides involves apparent free radical processes including Hofmann-Löffler-type rearrangements that lead to peptide chain fragmentation. Strategies for directed cleavage of peptides by free radical promoted processes are feasible, and such strategies may well simplify schemes for protein analysis.


Assuntos
Carbamatos/química , Lisina/análogos & derivados , Peptídeos/química , Sequência de Aminoácidos , Radicais Livres/química , Lisina/análise , Dados de Sequência Molecular , Mapeamento de Peptídeos/métodos , Peptídeos/análise , Peróxidos/química , Espectrometria de Massas por Ionização por Electrospray/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos
15.
J Biol Chem ; 279(6): 4250-9, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14645247

RESUMO

It has been proposed that the DNA repair protein O6-alkylguanine-DNA alkyltransferase increases the mutagenicity of 1,2-dibromoethane by reacting with it at its cysteine acceptor site to form a highly reactive half-mustard, which can then react with DNA (Liu, L., Pegg, A. E., Williams, K. M., and Guengerich, F. P. (2002) J. Biol. Chem. 277, 37920-37928). Incubation of Escherichia coli-expressed human alkyltransferase with 1,2-dibromoethane and single-stranded oligodeoxyribonucleotides led to the formation of covalent transferaseoligo complexes. The order of reaction determined was Gua>Thy>Cyt>Ade. Mass spectrometry analysis of the tryptic digest of the reaction product indicated that some of the adducts led to depurination with the release of the Gly136-Arg147 peptide cross-linked to a Gua at the N7 position, with the site of reaction being the active site Cys145 as established by chromatographic retention time and the fragmentation pattern determined by tandem mass spectrometry of a synthetic peptide adduct. The alkyltransferase-mediated mutations produced by 1,2-dibromoethane were predominantly Gua to Ade transitions but, in the spectrum of such rifampicin-resistant mutations in the RpoB gene, 20% were Gua to Thy transversions. The latter are likely to have arisen from the apurinic site generated from the Gua-N7 adduct. Support exists for an additional adduct/mutagenic pathway because evidence was obtained for DNA adducts other than at the Gua N7 atom and for mutations other than those attributable to depurination. Thus, chemical and biological evidence supports the existence of at least two alkyltransferase-dependent pathways for 1,2-dibromoethane-induced mutagenicity, one involving Gua N7-alkylation by alkyltransferase-S-CH2CH2Br and depurination, plus another as yet uncharacterized system(s).


Assuntos
Adutos de DNA/metabolismo , Dibrometo de Etileno/metabolismo , Mutagênicos/metabolismo , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Animais , Bovinos , Adutos de DNA/química , Adutos de DNA/toxicidade , Dibrometo de Etileno/química , Dibrometo de Etileno/toxicidade , Humanos , Técnicas In Vitro , Estrutura Molecular , Mutagênicos/química , Mutagênicos/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
16.
Cancer Res ; 63(23): 8492-9, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14679015

RESUMO

The Phase I enzyme cytochrome p450 1B1 (CYP1B1) has been postulated to play a key role in estrogen-induced mammary carcinogenesis by catalyzing the oxidative metabolism of 17beta-estradiol (E(2)) to catechol estrogens (2-OHE(2) and 4-OHE(2)) and highly reactive estrogen quinones (E(2)-2,3-Q and E(2)-3,4-Q). The potential of the quinones to induce mutagenic DNA lesions is expected to be decreased by their conjugation with glutathione (GSH) either nonenzymatically or catalyzed by glutathione S-transferase P1 (GSTP1), a Phase II enzyme. Because the interaction of the Phase I and Phase II enzymes is not well defined in this setting, we prepared recombinant purified CYP1B1 and GSTP1 to examine their individual and combined roles in the oxidative pathway and used gas and liquid chromatography/mass spectrometry to measure the parent hormone E(2), the catechol estrogens, and the GSH conjugates. 2-OHE(2) and 4-OHE(2) did not form conjugates with GSH alone or in the presence of GSTP1. However, incubation of GSH and CYP1B1 with 2-OHE(2) resulted in nearly linear conjugation through C-4 and C-1 (i.e., 2-OHE(2)-4-SG and 2-OHE(2)-1-SG), whereas the reaction of 4-OHE(2) yielded only 4-OHE(2)-2-SG. When CYP1B1 and GSTP1 were added together, the rate of conjugation was accelerated with a hyperbolic pattern of product formation in the order 4-OHE(2)-2-SG > 2-OHE(2)-4-SG >> 2-OHE(2)-1-SG. Incubation of E(2) with CYP1B1 and GSTP1 resulted in the formation of 4-OHE(2), 2-OHE(2), 4-OHE(2)-2-SG, 2-OHE(2)-4-SG, and 2-OHE(2)-1-SG. The production of GSH-estrogen conjugates was dependent on the concentrations of E(2) and GSTP1 but overall yielded only one-tenth of the catechol estrogen production. The concentration gap between catechol estrogens and GSH-estrogen conjugates may result from nonenzymatic reaction of the labile quinones with other nucleophiles besides GSH or may reflect the lower efficiency of GSTP1 compared with CYP1B1. In summary, both reactions are coordinated qualitatively in terms of product formation and substrate utilization, but the quantitative gap would leave room for the accumulation of estrogen quinones and their potential for DNA damage as part of estrogen-induced mammary carcinogenesis.


Assuntos
Hidrocarboneto de Aril Hidroxilases/metabolismo , Estradiol/metabolismo , Estrogênios de Catecol/metabolismo , Glutationa Transferase/metabolismo , Isoenzimas/metabolismo , Glândulas Mamárias Humanas/enzimologia , Citocromo P-450 CYP1B1 , Cromatografia Gasosa-Espectrometria de Massas , Glutationa S-Transferase pi , Humanos , Glândulas Mamárias Humanas/metabolismo , Proteínas Recombinantes/metabolismo
17.
Circulation ; 108(21): 2631-5, 2003 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-14623808

RESUMO

BACKGROUND: Adenosine, a known mediator of preconditioning, has been infused into the coronary circulation to induce therapeutic preconditioning, eg, in preparation for angioplasty. However, results have been disappointing. We tested the hypothesis that endothelial nucleoside transporter acts as a barrier impeding the delivery of intravascular adenosine into the underlying myocardium and that this can be overcome with dipyridamole, a nucleoside transporter blocker. METHODS AND RESULTS: We infused saline or adenosine (0.125 and 0.5 mg/min) into the brachial artery while monitoring forearm blood flow (FBF) and interstitial adenosine levels with microdialysis probes implanted in the flexor digitorum superficialis of the forearm in 7 healthy volunteers during intravenous administration of saline or dipyridamole (loading dose, 0.142 mg/kg per min for 5 minutes followed by 0.004 mg/kg per min). Adenosine produced near maximal forearm vasodilation, increasing FBF from 4.0+/-0.7 to 10.4+/-1.9 and 13.1+/-1.6 mL/100 mL per min for the low and high doses, respectively, but did not increase muscle dialysate adenosine concentration (from 88+/-21 to 65+/-23 and 85+/-26 nmol/L). Intravenous dipyridamole enhanced resting muscle dialysate adenosine (from 77+/-25 to 147+/-50 nmol/L), adenosine-induced increase in FBF (from 4.1+/-0.8 to 12.6+/-3 and 15.1+/-3 mL/100 mL per min for the low and high dose, respectively), and the delivery of adenosine into the interstitium (to 290+/-80 and 299+/-143 nmol/L for the low and high dose, respectively, P=0.04). CONCLUSIONS: Intravascular adenosine is likely ineffective in inducing myocardial preconditioning because of poor interstitial delivery. This can be overcome by blocking the nucleoside transporter with dipyridamole.


Assuntos
Adenosina/administração & dosagem , Adenosina/metabolismo , Líquido Extracelular/metabolismo , Precondicionamento Isquêmico Miocárdico/métodos , Nucleosídeos/metabolismo , Adulto , Transporte Biológico/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Artéria Braquial/fisiologia , Dipiridamol/administração & dosagem , Dipiridamol/farmacologia , Relação Dose-Resposta a Droga , Líquido Extracelular/química , Antebraço/irrigação sanguínea , Frequência Cardíaca/efeitos dos fármacos , Humanos , Infusões Intra-Arteriais , Injeções Intravenosas , Microdiálise , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Proteínas de Transporte de Nucleosídeos/antagonistas & inibidores , Valores de Referência
18.
Biochemistry ; 42(37): 10965-70, 2003 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-12974631

RESUMO

The active site cysteine of human O(6)-alkylguanine-DNA alkyltransferase (hAGT), Cys145, was shown to be highly reactive with model electrophiles unrelated to substrates, including 1-chloro-2,4-dinitrobenzene. The high reactivity suggested that the Cys145 thiolate anion might be stable at neutral pH. The pK(a) was estimated from plots of UV spectra (A(239)) and reactivity toward 4,4'-dithiopyridine vs pH. The estimated pK(a) for hAGT was 4-5, depending upon the method used, and near that of the extensively characterized papain Cys25. Rates of reaction with 4,4'-dithiopyridine were similar for the thiolate forms of hAGT, papain, glutathione, and the bacterial hAGT homologue Ogt (the pK(a) of the latter was 5.4). Bound Zn(2+) has previously been shown to be required for the catalytic activity of hAGT (Rasimas, J. J. et al. (2003) Biochemistry 42, 980-990). Zn(2+) was shown to be required for the low pK(a) of hAGT. The high reactivity of hAGT Cys145 is postulated to be important in normal catalytic function, in cross-linking reactions involving bis-electrophiles, and in inhibition of the DNA repair function of hAGT by electrophiles.


Assuntos
Cisteína/química , Dinitroclorobenzeno/química , O(6)-Metilguanina-DNA Metiltransferase/química , Ânions , Sítios de Ligação , Catálise , Reparo do DNA , Dissulfetos/química , Relação Dose-Resposta a Droga , Escherichia coli/enzimologia , Concentração de Íons de Hidrogênio , Cinética , Piridinas/química , Espectrofotometria , Fatores de Tempo , Raios Ultravioleta , Zinco/química , Zinco/farmacologia
19.
Reprod Toxicol ; 16(1): 29-43, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11934530

RESUMO

The guinea pig exhibits cyclic and luteal similarities to the human, a feature not present in other small experimental animals such as rats, mice, or rabbits. Studies were undertaken to investigate the in vivo effects of three persistent environmental xenobiotics (nonylphenol, 1,1-dichloro-2,2-bis(p-chlorophenyl)ethylene [p,p'-DDE], and pentachlorophenol) on the microanatomy of the adult female guinea pig reproductive system. The effects brought about by these compounds (40 mg/kg/day) were compared to those caused by the synthetic estrogen diethylstilbestrol (DES; 50 microg/kg/day). Adult female guinea pigs, intact and castrated, were treated with 14 daily subcutaneous (s.c.) doses of one of these agents. The 50% decline in the weight of the tract that occurred following castration, was prevented by administration of nonylphenol, p,p'-DDE, and DES, but not of pentachlorophenol. Nonylphenol produced weak estrogenic stimulation of the tract of intact animals and maintained a relatively normal histologic appearance in castrated animals. Focal mucinous metaplasia of the endometrium, however, was observed in both groups. Treatment of intact and castrated animals with p,p'-DDE resulted in cystic hyperplasia and mucinous metaplasia of the endometrium, hyperplasia of the cervical epithelium, estrogenic stimulation of the vagina, and dilation of the rete ovarii. Treatment of intact or castrated animals with DES resulted in effects that were qualitatively similar to those caused by p,p'-DDE. The appearance of the vaginal epithelium, however, was abnormal and the rete ovarii were less dilated. Pentachlorophenol had minimal effect on the histology of the tract of castrated or intact animals. These data support our hypothesis that some environmental toxicants can substitute for estradiol in regulating the microanatomy of the female reproductive tract. They indicate the potential of these compounds to act as endocrine disrupting agents.


Assuntos
Diclorodifenil Dicloroetileno/toxicidade , Moduladores de Receptor Estrogênico/toxicidade , Genitália Feminina/efeitos dos fármacos , Pentaclorofenol/toxicidade , Fenóis/toxicidade , Xenobióticos/toxicidade , Animais , Castração , Colo do Útero/patologia , Diclorodifenil Dicloroetileno/análogos & derivados , Dietilestilbestrol/toxicidade , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Endométrio/patologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Estradiol/metabolismo , Feminino , Doenças dos Genitais Femininos/induzido quimicamente , Doenças dos Genitais Femininos/metabolismo , Doenças dos Genitais Femininos/patologia , Cobaias , Hiperplasia/induzido quimicamente , Metaplasia/induzido quimicamente , Tamanho do Órgão/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Vagina/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA