Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
J Med Chem ; 66(22): 15477-15492, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37934858

RESUMEN

Metastases to the brain remain a significant problem in lung cancer, as treatment by most small-molecule targeted therapies is severely limited by efflux transporters at the blood-brain barrier (BBB). Here, we report the discovery of a selective, orally bioavailable, epidermal growth factor receptor (EGFR) inhibitor, 9, that exhibits high brain penetration and potent activity in osimertinib-resistant cell lines bearing L858R/C797S and exon19del/C797S EGFR resistance mutations. In vivo, 9 induced tumor regression in an intracranial patient-derived xenograft (PDX) murine model suggesting it as a potential lead for the treatment of localized and metastatic non-small-cell lung cancer (NSCLC) driven by activating mutant bearing EGFR. Overall, we demonstrate that an underrepresented functional group in medicinal chemistry, the trisubstituted hydroxylamine moiety, can be incorporated into a drug scaffold without the toxicity commonly surmised to accompany these units, all while maintaining potent biological activity and without the molecular weight creep common to drug optimization campaigns.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Humanos , Ratones , Compuestos de Anilina/farmacología , Compuestos de Anilina/uso terapéutico , Barrera Hematoencefálica/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Hidroxilamina/metabolismo , Hidroxilamina/uso terapéutico , Hidroxilaminas/metabolismo , Hidroxilaminas/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/química
2.
Appl Environ Microbiol ; 88(8): e0247021, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35384704

RESUMEN

Ammonia-oxidizing archaea (AOA) and bacteria (AOB) perform key steps in the global nitrogen cycle, the oxidation of ammonia to nitrite. While the ammonia oxidation pathway is well characterized in AOB, many knowledge gaps remain about the metabolism of AOA. Hydroxylamine is an intermediate in both AOB and AOA, but homologues of hydroxylamine dehydrogenase (HAO), catalyzing bacterial hydroxylamine oxidation, are absent in AOA. Hydrazine is a substrate for bacterial HAO, while phenylhydrazine is a suicide inhibitor of HAO. Here, we examine the effect of hydrazines in AOA to gain insights into the archaeal ammonia oxidation pathway. We show that hydrazine is both a substrate and an inhibitor for AOA and that phenylhydrazine irreversibly inhibits archaeal hydroxylamine oxidation. Both hydrazine and phenylhydrazine interfered with ammonia and hydroxylamine oxidation in AOA. Furthermore, the AOA "Candidatus Nitrosocosmicus franklandus" C13 oxidized hydrazine into dinitrogen (N2), coupling this reaction to ATP production and O2 uptake. This study expands the known substrates of AOA and suggests that despite differences in enzymology, the ammonia oxidation pathways of AOB and AOA are functionally surprisingly similar. These results demonstrate that hydrazines are valuable tools for studying the archaeal ammonia oxidation pathway. IMPORTANCE Ammonia-oxidizing archaea (AOA) are among the most numerous living organisms on Earth, and they play a pivotal role in the global biogeochemical nitrogen cycle. Despite this, little is known about the physiology and metabolism of AOA. We demonstrate in this study that hydrazines are inhibitors of AOA. Furthermore, we demonstrate that the model soil AOA "Ca. Nitrosocosmicus franklandus" C13 oxidizes hydrazine to dinitrogen gas, and this reaction yields ATP. This provides an important advance in our understanding of the metabolism of AOA and expands the short list of energy-yielding compounds that AOA can use. This study also provides evidence that hydrazines can be useful tools for studying the metabolism of AOA, as they have been for the bacterial ammonia oxidizers.


Asunto(s)
Amoníaco , Archaea , Adenosina Trifosfato/metabolismo , Amoníaco/metabolismo , Archaea/metabolismo , Bacterias/metabolismo , Humanos , Hidrazinas/metabolismo , Hidrazinas/farmacología , Hidroxilaminas/metabolismo , Nitrificación , Fenilhidrazinas/metabolismo , Microbiología del Suelo
3.
Chem Res Toxicol ; 35(2): 303-314, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-35089032

RESUMEN

Apurinic/apyrimidinic (AP) sites appear in DNA spontaneously and as intermediates of base excision DNA repair. AP sites are noninstructive lesions: they strongly block DNA polymerases, and if bypassed, the nature of the incorporated dNMP is mostly guided by the interactions within the polymerase-DNA active site. Many DNA polymerases follow the "A-rule", preferentially incorporating dAMP opposite to natural AP sites. Methoxyamine (MX), a small molecule, efficiently reacts with the aldehyde moiety of natural AP sites, thereby preventing their cleavage by APEX1, the major human AP endonuclease. MX is currently regarded as a possible sensitizer of cancer cells toward DNA-damaging drugs. To evaluate the mutagenic potential of MX, we have studied the utilization of various dNTPs by five DNA polymerases of different families encountering MX-AP adducts in the template in comparison with the natural aldehydic AP site. The Klenow fragment of Escherichia coli DNA polymerase I strictly followed the A-rule with both natural AP and MX-adducted AP sites. Phage RB69 DNA polymerase, a close relative of human DNA polymerases δ and ε, efficiently incorporated both dAMP and dGMP. DNA polymerase ß mostly incorporated dAMP and dCMP, preferring dCMP opposite to the natural AP site and dAMP opposite to the MX-AP site, while DNA polymerase λ was selective for dGMP, apparently via the primer misalignment mechanism. Finally, translesion DNA polymerase κ also followed the A-rule for MX-AP and additionally incorporated dCMP opposite to a natural AP site. Overall, the MX-AP site, despite structural differences, was similar to the natural AP site in terms of the dNMP misincorporation preference but was bypassed less efficiently by all polymerases except for Pol κ.


Asunto(s)
ADN Polimerasa Dirigida por ADN/metabolismo , Hidroxilaminas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Humanos
4.
Int J Mol Sci ; 22(1)2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-33396638

RESUMEN

The current study was designed to explore the in vitro nephrotoxic potential of four 3,5-dichloroaniline (3,5-DCA) metabolites (3,5-dichloroacetanilide, 3,5-DCAA; 3,5-dichlorophenylhydroxylamine, 3,5-DCPHA; 2-amino-4,6-dichlorophenol, 2-A-4,6-DCP; 3,5-dichloronitrobenzene, 3,5-DCNB) and to determine the renal metabolism of 3,5-DCA in vitro. In cytotoxicity testing, isolated kidney cells (IKC) from male Fischer 344 rats (~4 million/mL, 3 mL) were exposed to a metabolite (0-1.5 mM; up to 90 min) or vehicle. Of these metabolites, 3,5-DCPHA was the most potent nephrotoxicant, with 3,5-DCNB intermediate in nephrotoxic potential. 2-A-4,6-DCP and 3,5-DCAA were not cytotoxic. In separate experiments, 3,5-DCNB cytotoxicity was reduced by pretreating IKC with antioxidants and cytochrome P450, flavin monooxygenase and peroxidase inhibitors, while 3,5-DCPHA cytotoxicity was attenuated by two nucleophilic antioxidants (glutathione and N-acetyl-L-cysteine). Incubation of IKC with 3,5-DCA (0.5-1.0 mM, 90 min) produced only 3,5-DCAA and 3,5-DCNB as detectable metabolites. These data suggest that 3,5-DCNB and 3,5-DCPHA are potential nephrotoxic metabolites and may contribute to 3,5-DCA induced nephrotoxicity in vivo. In addition, the kidney can bioactivate 3,5-DCNB to toxic metabolites, and 3,5-DCPHA appears to generate reactive metabolites to contribute to 3,5-DCA nephrotoxicity. In vitro, N-oxidation of 3,5-DCA appears to be the primary mechanism of bioactivation of 3,5-DCA to nephrotoxic metabolites.


Asunto(s)
Compuestos de Anilina/toxicidad , Hidroxilaminas/toxicidad , Riñón/efectos de los fármacos , Compuestos de Anilina/metabolismo , Animales , Biotransformación , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión/métodos , Hidroxilaminas/metabolismo , Riñón/citología , Riñón/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratas Endogámicas F344
5.
Food Chem Toxicol ; 136: 110964, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31730879

RESUMEN

Processed meats are classified by the International Agency for Research on Cancer (IARC) as carcinogenic to humans. However, information on the responsible agents and the influence of industrial processing on the increased risk of cancer is still lacking. This study aimed to use cultures of Lactobacillus delbrueckii subsp. bulgaricus LB-UFSC 01 to biodegrade harmful C-nitrous, N-nitro, and C-nitro compounds in processed meat matrix. Firstly, positive results for ethylnitrolic acid (ENA) (>5.00 µg kg-1) and 2-methyl-1,4-dinitro-pyrrole (DNMP) (>12.0 µg kg-1) were obtained in mortadellas produced under different experimental conditions employing preservatives and antioxidants. Mortadellas containing nitrite and sorbate in the ratio of 8:1 (w/w) yielded the highest concentrations of mutagens. However, the treatment with the LB-UFSC 01 culture was able to modulate the harmful compounds in the mortadella samples. Several analytical methods employing liquid chromatography coupled to mass spectrometry and statistical models were employed to identify the metabolites and reaction routes during microbial biotransformation. For the first time, relevant information regarding the formation and degradation of ENA and DNMP in a processed meat model simulating real conditions was presented.


Asunto(s)
Hidroxilaminas/metabolismo , Lactobacillus delbrueckii/metabolismo , Productos de la Carne/microbiología , Mutágenos/metabolismo , Nitrilos/metabolismo , Pirroles/metabolismo , Antioxidantes/química , Ácido Ascórbico/química , Microbiología de Alimentos , Conservantes de Alimentos/química , Nitrito de Sodio/química , Ácido Sórbico/química
6.
Eur Rev Med Pharmacol Sci ; 22(4): 950-960, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29509243

RESUMEN

OBJECTIVE: Endometrial cancer is increasingly prevalent in western societies and affects mainly postmenopausal women; notably incidence rates have been rising by 1.9% per year on average since 2005. Although the early-stage endometrial cancer can be effectively managed with surgery, more advanced stages of the disease require multimodality treatment with varying results. In recent years, endometrial cancer has been extensively studied at the molecular level in an attempt to develop effective therapies. Recently, a family of compounds that alter epigenetic expression, namely histone deacetylase inhibitors, have shown promise as possible therapeutic agents in endometrial cancer. The present review aims to discuss the therapeutic potential of these agents. MATERIALS AND METHODS: This literature review was performed using the MEDLINE database; the search terms histone, deacetylase, inhibitors, endometrial, targeted therapies for endometrial cancer were employed to identify relevant studies. We only reviewed English language publications and also considered studies that were not entirely focused on endometrial cancer. Ultimately, sixty-four articles published until January 2018 were incorporated into our review. RESULTS: Studies in cell cultures have demonstrated that histone deacetylase inhibitors exert their antineoplastic activity by promoting expression of p21WAF1 and p27KIP1, cyclin-dependent kinase inhibitors, that have important roles in cell cycle regulation; importantly, the transcription of specific genes (e.g., E-cadherin, PTEN) that are commonly silenced in endometrial cancer is also enhanced. In addition to these abstracts effects, novel compounds with histone deacetylase inhibitor activity (e.g., scriptaid, trichostatin, entinostat) have also demonstrated significant antineoplastic activity both in vitro and in vivo, by liming tumor growth, inducing apoptosis, inhibiting angiogenesis and potentiating the effects of chemotherapy. CONCLUSIONS: The applications of histone deacetylase inhibitors in endometrial cancer appear promising; nonetheless, additional trials are necessary to establish the therapeutic role, clinical utility, and safety of these promising compounds.


Asunto(s)
Antineoplásicos/metabolismo , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/metabolismo , Inhibidores de Histona Desacetilasas/metabolismo , Histona Desacetilasas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/metabolismo , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Hidroxilaminas/metabolismo , Hidroxilaminas/farmacología , Hidroxilaminas/uso terapéutico , Quinolinas/metabolismo , Quinolinas/farmacología , Quinolinas/uso terapéutico
7.
PLoS One ; 10(8): e0131330, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26309022

RESUMEN

DNA damage and repair is a fundamental process that plays an important role in cancer treatment. Base excision repair (BER) is a major repair pathway that often leads to drug resistance in DNA-targeted cancer chemotherapy. In order to measure BER, we have developed a near infrared (NIR) fluorescent probe. This probe binds to a key intermediate, termed apurinic/apyrimidinic (AP) site, in the BER pathway where DNA damage and repair occurs. We have developed an assay to show the efficacy of the probe binding to AP sites and have shown that it can distinguish AP sites in DNA extract from chemotherapy treated cells. This probe has potential application in monitoring patient response to chemotherapy and evaluating new drugs in development.


Asunto(s)
Daño del ADN , Reparación del ADN , Colorantes Fluorescentes/metabolismo , Animales , Emparejamiento Base , Secuencia de Bases , Sitios de Unión , Bovinos , Línea Celular Tumoral , Neoplasias del Colon/patología , ADN/metabolismo , Roturas del ADN de Cadena Simple , Reparación del ADN/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Floxuridina/farmacología , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/química , Humanos , Hidroxilaminas/metabolismo , Metilmetanosulfonato/farmacología , Datos de Secuencia Molecular , Espectrometría de Fluorescencia , Factores de Tiempo
8.
ACS Chem Biol ; 10(4): 1118-27, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25636004

RESUMEN

Oncogenic transcriptional coregulators C-terminal Binding Protein (CtBP) 1 and 2 possess regulatory d-isomer specific 2-hydroxyacid dehydrogenase (D2-HDH) domains that provide an attractive target for small molecule intervention. Findings that the CtBP substrate 4-methylthio 2-oxobutyric acid (MTOB) can interfere with CtBP oncogenic activity in cell culture and in mice confirm that such inhibitors could have therapeutic benefit. Recent crystal structures of CtBP 1 and 2 revealed that MTOB binds in an active site containing a dominant tryptophan and a hydrophilic cavity, neither of which are present in other D2-HDH family members. Here, we demonstrate the effectiveness of exploiting these active site features for the design of high affinity inhibitors. Crystal structures of two such compounds, phenylpyruvate (PPy) and 2-hydroxyimino-3-phenylpropanoic acid (HIPP), show binding with favorable ring stacking against the CtBP active site tryptophan and alternate modes of stabilizing the carboxylic acid moiety. Moreover, ITC experiments show that HIPP binds to CtBP with an affinity greater than 1000-fold over that of MTOB, and enzymatic assays confirm that HIPP substantially inhibits CtBP catalysis. These results, thus, provide an important step, and additional insights, for the development of highly selective antineoplastic CtBP inhibitors.


Asunto(s)
Oxidorreductasas de Alcohol/química , Proteínas de Unión al ADN/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteínas del Tejido Nervioso/química , Oxidorreductasas de Alcohol/antagonistas & inhibidores , Oxidorreductasas de Alcohol/metabolismo , Sitios de Unión , Proteínas Co-Represoras , Cristalografía por Rayos X , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/metabolismo , Humanos , Hidroxilaminas/química , Hidroxilaminas/metabolismo , Hidroxilaminas/farmacología , Ligandos , Modelos Moleculares , NAD/química , NAD/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Fenilpropionatos/química , Fenilpropionatos/metabolismo , Fenilpropionatos/farmacología , Ácidos Fenilpirúvicos/química , Ácidos Fenilpirúvicos/metabolismo , Ácidos Fenilpirúvicos/farmacología , Conformación Proteica , Relación Estructura-Actividad , Termodinámica
9.
Chem Res Toxicol ; 26(6): 993-1004, 2013 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-23682735

RESUMEN

Many aromatic amines and heterocyclic aromatic amines (HAAs) are known carcinogens for animals, and there is also strong evidence of some in human cancer. The activation of these compounds, including some arylamine drugs, involves N-hydroxylation, usually by cytochrome P450 enzymes (P450) in Family 1 (1A2, 1A1, and 1B1). We previously demonstrated that the bioactivation product of the anticancer agent 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203), an N-hydroxylamine, can be reduced by P450 2S1 to its amine precursor under anaerobic conditions and, to a lesser extent, under aerobic conditions [Wang, K., and Guengerich, F. P. (2012) Chem. Res. Toxicol. 25, 1740-1751]. In the study presented here, we tested the hypothesis that P450 2S1 is involved in the reductive biotransformation of known carcinogenic aromatic amines and HAAs. The N-hydroxylamines of 4-aminobiphenyl (4-ABP), 2-naphthylamine (2-NA), and 2-aminofluorene (2-AF) were synthesized and found to be reduced by P450 2S1 under both anaerobic and aerobic conditions. The formation of amines due to P450 2S1 reduction also occurred under aerobic conditions but was less apparent because the competitive disproportionation reactions (of the N-hydroxylamines) also yielded amines. Further, some nitroso and nitro derivatives of the arylamines could also be reduced by P450 2S1. None of the amines tested were oxidized by P450 2S1. These results suggest that P450 2S1 may be involved in the reductive detoxication of several of the activated products of carcinogenic aromatic amines and HAAs.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/metabolismo , Hidroxilaminas/química , Hidroxilaminas/metabolismo , Humanos , Estructura Molecular , Oxidación-Reducción
10.
Free Radic Biol Med ; 57: 49-60, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23261941

RESUMEN

Through free radical-mediated peroxidation, cyclooxygenase (COX) can metabolize dihomo-γ-linolenic acid (DGLA) and arachidonic acid (AA) to form well-known bioactive metabolites, namely, the 1-series of prostaglandins (PGs1) and the 2-series of prostaglandins (PGs2), respectively. Unlike PGs2, which are generally viewed as proinflammatory and procarcinogenic PGs, PGs1 may possess anti-inflammatory and anti-cancer activity. Previous studies using ovine COX along with spin trapping and the LC/ESR/MS technique have shown that certain exclusive free radicals are generated from different free radical reactions in DGLA and AA peroxidation. However, it has been unclear whether the differences were associated with the contrasting bioactivity of DGLA vs AA. The aim of this study was to refine the LC/MS and spin trapping technique to make it possible for the association between free radicals and cancer cell growth to be directly tested. Using a colon cancer cell line, HCA-7 colony 29, and LC/MS along with a solid-phase extraction, we were able to characterize the reduced forms of radical adducts (hydroxylamines) as the free radicals generated from cellular COX-catalyzed peroxidation. For the first time, free radicals formed in the COX-catalyzed peroxidation of AA vs DGLA and their association with cancer cell growth were assessed (cell proliferation via MTS and cell cycle distribution via propidium iodide staining) in the same experimental setting. The exclusive free radicals formed from the COX-catalyzed peroxidation of AA and DGLA were shown to be correlated with the cell growth response. Our results indicate that free radicals generated from the distinct radical reactions in COX-catalyzed peroxidation may represent the novel metabolites of AA and DGLA that correspond to their contrasting bioactivity.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/metabolismo , Ácido Araquidónico/metabolismo , Radicales Libres/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Cromatografía Liquida , Neoplasias del Colon , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Hidroxilaminas/metabolismo , Espectrometría de Masas , Prostaglandinas/metabolismo , Detección de Spin
11.
Toxicol Sci ; 127(2): 348-59, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22403159

RESUMEN

2-Methoxyaniline (o-anisidine) is an industrial and environmental pollutant causing tumors of urinary bladder in rodents. Here, we investigated the formation and persistence of DNA adducts in the Wistar rat. Using the (32)P-postlabeling method, three o-anisidine-derived DNA adducts were found in several organs of rats treated with a total dose of 0.53 mg o-anisidine/kg body wt (0.15, 0.18, and 0.2 mg/kg body wt ip in the first, second, and third day, respectively), of which the urinary bladder had the highest levels. At four posttreatment times (1 day, 13 days, 10 weeks, and 36 weeks), DNA adducts in bladder, liver, kidney, and spleen of rats were analyzed to study their persistence. In all time points, the highest total adduct levels were found in urinary bladder (39 adducts per 10(7) nucleotides after 1 day and 15 adducts per 10(7) nucleotides after 36 weeks) where 39% adducts remained. In contrast to the urinary bladder, no persistence was detected in other organs. All three DNA adducts were identified as deoxyguanosine adducts. When deoxyguanosine was reacted with the oxidative metabolite of o-anisidine, N-(2-methoxyphenyl)hydroxylamine, three adducts could be separated by high-performance liquid chromatography (HPLC) and were identified by mass spectroscopy and/or nuclear magnetic resonance spectrometry. All adducts are products of the nitrenium/carbenium ions, the reactive species generated from N-(2-methoxyphenyl)hydroxylamine. The major adduct was identified to be N-(deoxyguanosin-8-yl)-2-methoxyaniline. Using cochromatography on HPLC, this adduct was found to be identical to the major adduct generated by activation of o-anisidine in vitro and in vivo.


Asunto(s)
Compuestos de Anilina/toxicidad , Aductos de ADN/metabolismo , Contaminantes Ambientales/toxicidad , Compuestos de Anilina/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Desoxiguanosina/metabolismo , Contaminantes Ambientales/metabolismo , Hidroxilaminas/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratas , Ratas Wistar , Espectrometría de Masa por Ionización de Electrospray , Bazo/efectos de los fármacos , Bazo/metabolismo , Factores de Tiempo , Distribución Tisular , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo
12.
Regul Toxicol Pharmacol ; 62(1): 151-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21925229

RESUMEN

The metabolite of several amide anaesthetics, 2,6-xylidine, is a possible human (Group 2B) carcinogen and induced nasal tumours in rats after dietary administration. However, published papers on the genotoxicity of 2,6-xylidine in vitro have given inconsistent results. It has been proposed that the genotoxicity of 2,6-xylidine is dependent on its metabolism to a key metabolite dimethylphenyl N-hydroxylamine (DMHA), which would then be further converted to form a reactive nitrenium ion by phase 2 (mainly acetylation) metabolism. In order to study whether the inconsistent results could be explained by different systems having different potential for DMHA to be formed and to induce genotoxicity in vitro, we have tested 2,6-xylidine in conventional Ames bacteria, and strains engineered to overexpress acetyltransferase, in the presence of different concentrations of induced rat liver and human liver S9. All tests gave consistently negative results. The formation of DMHA by induced rat liver S9 and human S9 was clearly shown to occur, and to be concentration- and time-dependent. The potential inhibitory effects of the solvent DMSO were also studied, but it was clearly not responsible for the negative results with 2,6-xylidine. Thus, whatever is the mode of action of 2,6-xylidine carcinogenicity in rodents, it has proven impossible to detect mutagenic effects in Ames tests with numerous variations of metabolic conditions, or even using acetyltransferase overexpressing strains of bacteria.


Asunto(s)
Compuestos de Anilina/toxicidad , Hidroxilaminas/toxicidad , Compuestos de Anilina/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Humanos , Hidroxilaminas/metabolismo , Hígado/metabolismo , Pruebas de Mutagenicidad , Ratas , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética
13.
Proc Natl Acad Sci U S A ; 108(43): 17649-54, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-21987796

RESUMEN

Glycosyltransferases are useful synthetic catalysts for generating natural products with sugar moieties. Although several natural product glycosyltransferase structures have been reported, design principles of glycosyltransferase engineering for the generation of glycodiversified natural products has fallen short of its promise, partly due to a lack of understanding of the relationship between structure and function. Here, we report structures of all four calicheamicin glycosyltransferases (CalG1, CalG2, CalG3, and CalG4), whose catalytic functions are clearly regiospecific. Comparison of these four structures reveals a conserved sugar donor binding motif and the principles of acceptor binding region reshaping. Among them, CalG2 possesses a unique catalytic motif for glycosylation of hydroxylamine. Multiple glycosyltransferase structures in a single natural product biosynthetic pathway are a valuable resource for understanding regiospecific reactions and substrate selectivities and will help future glycosyltransferase engineering.


Asunto(s)
Aminoglicósidos/biosíntesis , Antibióticos Antineoplásicos/biosíntesis , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Ingeniería de Proteínas/métodos , Dominios y Motivos de Interacción de Proteínas/genética , Carbohidratos/química , Enediinos/química , Hidroxilaminas/metabolismo
14.
Mutat Res ; 726(2): 160-8, 2011 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-21946300

RESUMEN

N-(2-Methoxyphenyl)hydroxylamine is a component in the human metabolism of two industrial and environmental pollutants and bladder carcinogens, viz. 2-methoxyaniline (o-anisidine) and 2-methoxynitrobenzene (o-nitroanisole), and it is responsible for their genotoxicity. Besides its capability to form three deoxyguanosine adducts in DNA, N-(2-methoxyphenyl)-hydroxylamine is also further metabolized by hepatic microsomal enzymes. To investigate its metabolism by human hepatic microsomes and to identify the major microsomal enzymes involved in this process are the aims of this study. N-(2-Methoxyphenyl)hydroxylamine is metabolized by human hepatic microsomes predominantly to o-anisidine, one of the parent carcinogens from which N-(2-methoxyphenyl)hydroxylamine is formed, while o-aminophenol and two N-(2-methoxyphenyl)hydroxylamine metabolites, whose exact structures have not been identified as yet, are minor products. Selective inhibitors of microsomal CYPs, NADPH:CYP reductase and NADH:cytochrome-b(5) reductase were used to characterize human liver microsomal enzymes reducing N-(2-methoxyphenyl)hydroxylamine to o-anisidine. Based on these studies, we attribute the main activity for this metabolic step in human liver to CYP3A4, 2E1 and 2C (more than 90%). The enzymes CYP2D6 and 2A6 also partake in this N-(2-methoxyphenyl)hydroxylamine metabolism in human liver, but only to ∼6%. Among the human recombinant CYP enzymes tested in this study, human CYP2E1, followed by CYP3A4, 1A2, 2B6 and 2D6, were the most efficient enzymes metabolizing N-(2-methoxyphenyl)hydroxylamine to o-anisidine. The results found in this study indicate that genotoxicity of N-(2-methoxyphenyl)hydroxylamine is dictated by its spontaneous decomposition to nitrenium/carbenium ions generating DNA adducts, and by its susceptibility to metabolism by CYP enzymes.


Asunto(s)
Compuestos de Anilina/metabolismo , Anisoles/metabolismo , Carcinógenos/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Contaminantes Ambientales/metabolismo , Hidroxilaminas/metabolismo , Animales , Aductos de ADN/metabolismo , Humanos , Masculino , Microsomas Hepáticos/metabolismo , Ratas , Ratas Wistar
15.
J Exp Bot ; 60(7): 2065-72, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19357430

RESUMEN

Plants are known to produce NO via the reduction of nitrite. Oxidative NO production in plants has been considered only with respect to a nitric oxide synthase (NOS). Here it is shown that tobacco cell suspensions emitted NO when hydroxylamine (HA) or salicylhydroxamate (SHAM), a frequently used AOX inhibitor, was added. N(G)-hydroxy-L-arginine, a putative intermediate in the NOS-reaction, gave no NO emission. Only a minor fraction (< or = 1%) of the added HA or SHAM was emitted as NO. Production of NO was decreased by anoxia or by the addition of catalase, but was increased by conditions inducing reactive oxygen (ROS) or by the addition of hydrogen peroxide. Cell-free enzyme solutions generating superoxide or hydrogen peroxide also led to the formation of NO from HA or (with lower rates) from SHAM, and nitrite was also an oxidation product. Unexpectedly, the addition of superoxide dismutase (SOD) to cell suspensions stimulated NO formation from hydroxylamines, and SOD alone (without cells) also catalysed the production of NO from HA or SHAM. NO production by SOD plus HA was higher in nitrogen than in air, but from SOD plus SHAM it was lower in nitrogen. Thus, SOD-catalysed NO formation from SHAM and from HA may involve different mechanisms. While our data open a new possibility for oxidative NO formation in plants, the existence and role of these reactions under physiological conditions is not yet clear.


Asunto(s)
Hidroxilaminas/metabolismo , Nicotiana/metabolismo , Óxido Nítrico/metabolismo , Células Cultivadas , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Salicilamidas/metabolismo
16.
Cancer Gene Ther ; 15(11): 758-64, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18600257

RESUMEN

Gene-directed enzyme prodrug therapy is a promising approach to the local management of cancer and a number of gene prodrug combinations have entered clinical trials. The antitumor activity of Escherichia coli nitroreductase (NTR) in combination with the prodrug CB1954 relies on the reduction of the nitro groups to reactive N-hydroxylamine intermediates that are toxic in proliferating and nonproliferating cells. We examined whether secondary metabolic activation of the N-hydroxylamines by sulfotransferases or acetyltransferases altered cell responsiveness to the drug. We evaluated the coexpression of NTR with the human cytosolic sulfotransferases SULT1A1, 1A2, 1A3, 1E1 and 2A1, or the human arylamine N-acetyltransferases NAT1 and NAT2 on SKOV3 cell survival. Only NAT2 significantly altered the toxicity of CB1954, decreasing the IC(50) 16-fold from 0.61 to 0.04 microM. These results suggest that one or more of the N-hydroxyl metabolites are a substrate for O-acetylation by NAT2. We also examined the bystander effect of SKOV3 cells expressing NTR or NTR plus NAT2. Addition of the acetyltransferase resulted in a significant decreased bystander effect (P>0.01), possibly due to a lower concentration of reactive metabolites in the culture medium. These results suggest that a combination of bacterial NTR and NAT2 may provide a greater clinical response at therapeutic concentrations of CB1954 provided the reduction in bystander effect is not clinically significant. Moreover, endogenous NAT2, which is localized predominantly in the liver and gut, may be involved in the dose-limiting hepatic toxicity and gastrointestinal side effects seen in patients treated with the higher doses of CB1954.


Asunto(s)
Antineoplásicos/metabolismo , Arilamina N-Acetiltransferasa/metabolismo , Aziridinas/metabolismo , Quimioterapia/métodos , Escherichia coli/enzimología , Nitrorreductasas/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Profármacos/metabolismo , Análisis de Varianza , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Aziridinas/farmacología , Aziridinas/toxicidad , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cartilla de ADN/genética , Femenino , Citometría de Flujo , Humanos , Hidroxilaminas/metabolismo , Concentración 50 Inhibidora , Nitrorreductasas/farmacología , Plásmidos/genética , Profármacos/farmacología , Profármacos/toxicidad , Sulfotransferasas/metabolismo
17.
FASEB J ; 21(14): 4077-86, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17656467

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of severe visual impairment in the elderly in developed countries. AMD patients have elevated levels of iron within the retinal pigment epithelia (RPE), which may lead to oxidative damage to mitochondria, disruption of retinal metabolism, and vision impairment or loss. As a possible model for iron-induced AMD, we investigated the effects of excess iron in cultured human fetal RPE cells on oxidant levels and mitochondrial cytochrome c oxidase (complex IV) function and tested for protection by N-tert-butyl hydroxylamine (NtBHA), a known mitochondrial antioxidant. RPE exposure to ferric ammonium citrate resulted in a time- and dose-dependent increase in intracellular iron, which increased oxidant production and decreased glutathione (GSH) levels and mitochondrial complex IV activity. NtBHA addition to iron-overloaded RPE cells led to a reduction of intracellular iron content, oxidative stress, and partial restoration of complex IV activity and GSH content. NtBHA might be useful in AMD due to its potential to reduce oxidative stress, mitochondrial damage, and age-related iron accumulation, which may damage normal RPE function and lead to loss of vision.


Asunto(s)
Antioxidantes/farmacología , Células Epiteliales/efectos de los fármacos , Hidroxilaminas/farmacología , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/prevención & control , Degeneración Macular/tratamiento farmacológico , Mitocondrias/metabolismo , Epitelio Pigmentado Ocular/efectos de los fármacos , Anciano , Antioxidantes/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Hidroxilaminas/metabolismo , Hierro/metabolismo , Quelantes del Hierro/metabolismo , Quelantes del Hierro/farmacología , Sobrecarga de Hierro/patología , Degeneración Macular/metabolismo , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Epitelio Pigmentado Ocular/metabolismo , Epitelio Pigmentado Ocular/patología , Retina/citología , Retina/efectos de los fármacos , Retina/metabolismo
18.
Mol Aspects Med ; 28(3-4): 375-95, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17560642

RESUMEN

With our growing understanding of the pathways involved in cell proliferation and signaling, targeted therapies, in the treatment of cancer are entering the clinical arena. New and emerging targets are proteins involved in DNA repair pathways. Inhibition of various proteins in the DNA repair pathways sensitizes cancer cells to DNA damaging agents such as chemotherapy and/or radiation. We study the apurinic endonuclease 1/redox factor-1 (Ape1/Ref-1) and believe that its crucial function in DNA repair and reduction-oxidation or redox signaling make it an excellent target for sensitizing tumor cells to chemotherapy. Ape1/Ref-1 is an essential enzyme in the base excision repair (BER) pathway which is responsible for the repair of DNA caused by oxidative and alkylation damage. As importantly, Ape1/Ref-1 also functions as a redox factor maintaining transcription factors in an active reduced state. Ape1/Ref-1 stimulates the DNA binding activity of numerous transcription factors that are involved in cancer promotion and progression such as AP-1 (Fos/Jun), NFkappaB, HIF-1alpha, CREB, p53 and others. We will discuss what is known regarding the pharmacological targeting of the DNA repair activity, as well as the redox activity of Ape1/Ref-1, and explore the budding clinical utility of inhibition of either of these functions in cancer treatment. A brief discussion of the effect of polymorphisms in its DNA sequence is included because of Ape1/Ref-1's importance to maintenance and integrity of the genome. Experimental modification of Ape1/Ref-1 activity changes the response of cells and of organisms to DNA damaging agents, suggesting that Ape1/Ref-1 may also be a productive target of chemoprevention. In this review, we will provide an overview of Ape1/Ref-1's activities and explore the potential of this protein as a target in cancer treatment as well as its role in chemoprevention.


Asunto(s)
Reparación del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Neoplasias , Benzoquinonas/metabolismo , Ácidos Carboxílicos/química , Ácidos Carboxílicos/metabolismo , Daño del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Humanos , Hidroxilaminas/metabolismo , Isoflavonas/metabolismo , Lucantona/metabolismo , Neoplasias/metabolismo , Neoplasias/prevención & control , Neoplasias/terapia , Oxidación-Reducción , Estrés Oxidativo , Propionatos/metabolismo , Resveratrol , Estilbenos/metabolismo
19.
Chem Res Toxicol ; 19(10): 1366-73, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17040106

RESUMEN

Heterocyclic and aromatic amine carcinogens are thought to lead to tumor initiation via the formation of DNA adducts, and bioactivation to arylhydroxylamine metabolites is necessary for reactivity with DNA. Carcinogenic arylhydroxylamine metabolites are cleared by a microsomal, NADH-dependent, oxygen-insensitive reduction pathway in humans, which may be a source of interindividual variability in response to aromatic amine carcinogens. The purpose of this study was to characterize the identity of this reduction pathway in human liver. On the basis of our findings with structurally similar arylhydroxylamine metabolites of therapeutic drugs, we hypothesized that the reductive detoxification of arylhydroxylamine carcinogens was catalyzed by NADH cytochrome b5 reductase (b5R) and cytochrome b5 (cyt b5). We found that reduction of the carcinogenic hydroxylamines of the aromatic amine 4-aminobiphenyl (4-ABP; found in cigarette smoke) and the heterocyclic amine 2-amino-1-methyl-6-phenylimidazo [4,5-b] pyridine (PhIP; found in grilled meats) was indeed catalyzed by a purified system containing only human b5R and cyt b5. Specific activities were 56-346-fold higher in the purified system as compared to human liver microsomes (HLM), with similar Michaelis-Menten constants (K(m) values) in both systems. The stoichiometry for b5R and cyt b5 that yielded the highest activity in the purified system was also similar to that found in native HLM ( approximately 1:8 to 1:10). Polyclonal antisera to either b5R or cyt b5 significantly inhibited N-hydroxy-4-aminobiphenyl (NHOH-4-ABP) reduction by 95 and 89%, respectively, and immunoreactive cyt b5 protein content in individual HLM was significantly correlated with individual reduction of both NHOH-4-ABP and N-hydroxy-PhIP (NHOH-PhIP). Finally, titration of HLM into the purified b5R/cyt b5 system did not enhance the efficiency of reduction activity. We conclude that b5R and cyt b5 are together solely capable of the reduction of arylhydroxylamine carcinogens, and we further hypothesize that this pathway may be a source of individual variability with respect to cancer susceptibility following 4-ABP or PhIP exposure.


Asunto(s)
Carcinógenos/metabolismo , Carcinógenos/toxicidad , Citocromo-B(5) Reductasa/metabolismo , Citocromos b5/metabolismo , Hidroxilaminas/metabolismo , Hidroxilaminas/toxicidad , Citocromo-B(5) Reductasa/aislamiento & purificación , Citocromos b5/aislamiento & purificación , Humanos , Hidroxilamina/química , Hidroxilamina/metabolismo , Cinética , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Microsomas/efectos de los fármacos , Microsomas/enzimología , Microsomas/metabolismo , Oxidación-Reducción , Especificidad por Sustrato
20.
Anal Bioanal Chem ; 386(3): 666-74, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16724218

RESUMEN

Characterizing the biological effects of metabolic transformations (or biotransformation) is one of the key steps in developing safe and effective pharmaceuticals. Sulfate conjugation, one of the major phase II biotransformations, is the focus of this study. While this biotransformation typically facilitates excretion of metabolites by making the compounds more water soluble, sulfation may also lead to bioactivation, producing carcinogenic products. The end result, excretion or bioactivation, depends on the structural features of the sulfation sites, so obtaining the structure of the sulfated metabolites is critically important. We describe herein a very simple, high-throughput procedure for using mass spectrometry to identify the structure-and thus the biological fate-of sulfated metabolites. We have chemically synthesized and analyzed libraries of compounds representing all the biologically relevant types of sulfation products, and using the mass spectral data, the structural features present in these analytes can be reliably determined, with a 97% success rate. This work represents the first example of a high-throughput analysis that can identify the structure of sulfated metabolites and predict their biological effects.


Asunto(s)
Química Farmacéutica/métodos , Preparaciones Farmacéuticas/metabolismo , Sulfatos/química , Sulfatos/metabolismo , Alcoholes/química , Alcoholes/metabolismo , Hidroxilaminas/química , Hidroxilaminas/metabolismo , Fase II de la Desintoxicación Metabólica , Estructura Molecular , Espectrometría de Masa de Ion Secundario , Relación Estructura-Actividad , Sulfatos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA