Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Drug Metab Dispos ; 43(7): 1028-36, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25870102

RESUMEN

V-PYRRO/NO [O(2)-vinyl-1-(pyrrolidin-1-yl)diazen-1-ium-1,2-diolate] and V-PROLI/NO (O2-vinyl-[2-(carboxylato)pyrrolidin-1-yl]diazen-1-ium-1,2-diolate), two structurally similar diazeniumdiolate derivatives, were designed as liver-selective prodrugs that are metabolized by cytochrome P450 isoenzymes, with subsequent release of nitric oxide (NO). Yet, their efficacy in the treatment of nonalcoholic fatty liver disease (NAFLD) and their comparative pharmacokinetic and metabolic profiles have not been characterized. The aim of the present work was to compare the effects of V-PYRRO/NO and V-PROLI/NO on liver steatosis, glucose tolerance, and liver fatty acid composition in C57BL/6J mice fed a high-fat diet, as well as to comprehensively characterize the ADME (absorption, distribution, metabolism and excretion) profiles of both NO donors. Despite their similar structure, V-PYRRO/NO and V-PROLI/NO showed differences in pharmacological efficacy in the murine model of NAFLD. V-PYRRO/NO, but not V-PROLI/NO, attenuated liver steatosis, improved glucose tolerance, and favorably modified fatty acid composition in the liver. Both compounds were characterized by rapid absorption following i.p. administration, rapid elimination from the body, and incomplete bioavailability. However, V-PYRRO/NO was eliminated mainly by the liver, whereas V-PROLI/NO was excreted mostly in unchanged form by the kidney. V-PYRRO/NO was metabolized by CYP2E1, CYP2C9, CYP1A2, and CYP3A4, whereas V-PROLI/NO was metabolized mainly by CYP1A2. Importantly, V-PYRRO/NO was a better NO releaser in vivo and in the isolated, perfused liver than V-PROLI/NO, an effect compatible with the superior antisteatotic activity of V-PYRRO/NO. In conclusion, V-PYRRO/NO displayed a pronounced antisteatotic effect associated with liver-targeted NO release, whereas V-PROLI/NO showed low effectiveness, was not taken up by the liver, and was eliminated mostly in unchanged form by the kidney.


Asunto(s)
Donantes de Óxido Nítrico/farmacocinética , Donantes de Óxido Nítrico/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Pirrolidinas/farmacología , Pirrolidinas/farmacocinética , Pirrolidinas/uso terapéutico , Triazenos/farmacología , Triazenos/uso terapéutico , Animales , Biotransformación , Sistema Enzimático del Citocromo P-450/metabolismo , Dieta Alta en Grasa , Ácidos Grasos/metabolismo , Intolerancia a la Glucosa , Absorción Intestinal , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Distribución Tisular
2.
Bioorg Med Chem ; 23(15): 4980-4988, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26043946

RESUMEN

PABA/NO [O(2)-{2,4-dinitro-5-[4-(N-methylamino)benzoyloxy]phenyl} 1-(N,N-dimethylamino) diazen-1-ium-1,2-diolate] is a nitric oxide (NO)-releasing arylating agent designed to be selectively activated by reaction with glutathione (GSH) on catalysis by glutathione S-transferase P1 (GSTP1), an enzyme frequently overexpressed in cancer cells. PABA/NO has proven active in several cancer models in vitro and in vivo, but its tendency to be metabolized via a variety of pathways, some that generate inactive metabolites and hydrolysis products, limits its potential as a drug. Here we show that a simple replacement of cyano for nitro at the 4 position to give compound 4b ('p-cyano-PABA/NO') has the dual effect of slowing the undesired side reactions while enhancing the proportion of NO release and arylating activity on catalysis by GSTP1. Compound 4b showed increased resistance to hydrolysis and uncatalyzed reaction with GSH, along with a more favorable product distribution in the presence of GSTP1. It also showed significant proapoptotic activity. The data suggest p-cyano-PABA/NO to be a more promising prodrug than PABA/NO, with better selectivity toward cancer cells.


Asunto(s)
Ácido 4-Aminobenzoico/química , Gutatión-S-Transferasa pi/metabolismo , Óxido Nítrico/química , Ácido 4-Aminobenzoico/metabolismo , Ácido 4-Aminobenzoico/farmacología , Biocatálisis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Glutatión/química , Glutatión/metabolismo , Gutatión-S-Transferasa pi/química , Células HL-60 , Humanos , Isomerismo , Cinética , Conformación Molecular , Óxido Nítrico/metabolismo , Óxido Nítrico/toxicidad
3.
J Org Chem ; 79(19): 9389-93, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25210948

RESUMEN

We report the apparently unprecedented direct reaction of nitric oxide (NO) with amides to generate ions of structure R(C═O)NH-N(O)═NO(-), with examples including R = Me (1a) or 3-pyridyl (1b). The sodium salts of both released NO in pH 7.4 buffer, with 37 °C half-lives of 1-3 min. As NO-releasing drug candidates, diazeniumdiolated amides would have the advantage of generating only 1 equiv of base on hydrolyzing exhaustively to NO, in contrast to their amine counterparts, which generate 2 equiv of base.


Asunto(s)
Amidas/química , Compuestos Azo/química , Compuestos Azo/síntesis química , Óxido Nítrico/química , Piridinas/química , Estructura Molecular
4.
J Org Chem ; 79(10): 4512-6, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24766285

RESUMEN

Recent theoretical studies have suggested that the parent diazeniumdiolate ion, H2N-N(O)═NO(-) ("diazeniumdiolated ammonia"), might be stable enough to be isolated and that it could potentially serve as a uniquely advantageous prodrug form of bioactive nitroxyl (HNO). Here, we report on an attempt to isolate its O(2)-benzylated derivative by aminolysis of the C═N bond in PhC(NH2)═N-N(O)═NOBn. The reaction proved remarkably sluggish in comparison to aminolysis of unsubstituted benzamidine, and the desired product could not be isolated, apparently because of base sensitivity of the NH2 group. Consistent with this interpretation, O-benzylhydroxylamine and N2O were recovered from the reaction mixture in high yields, along with N,N'-dibutylbenzamidine. Theoretical calculations rationalize the observed slow aminolysis by demonstrating that the diazeniumdiolate group greatly suppresses the electrophilicity of the adjacent C═N carbon center, rendering attack at that position endothermic. The data provide significant insights into the challenges inherent to the pursuit of diazeniumdiolated ammonia.


Asunto(s)
Amidinas/síntesis química , Amoníaco/química , Compuestos Azo/síntesis química , Benzamidinas/química , Óxidos de Nitrógeno/química , Amidinas/química , Compuestos Azo/química
5.
Nitric Oxide ; 35: 131-6, 2013 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-24126017

RESUMEN

We introduce a strategy for generating mixtures of nitric oxide (NO) and nitroxyl (HNO) at tunable rates in physiological media. The approach involves converting a spontaneously HNO/NO-generating ion to a caged (prodrug) form that is essentially stable in neutral media, but that can be activated for HNO/NO release by adding an enzyme capable of efficiently opening the cage to regenerate the ion. By judiciously choosing the enzyme, substrate, and reaction conditions, unwanted scavenging of the HNO and NO by the protein can be minimised and the catalytic efficiency of the enzyme can be maintained. We illustrate this approach with a proof-of-concept study wherein the prodrug is Gal-IPA/NO, a diazeniumdiolate of structure iPrHN-N(O)NOR, with R=ß-d-galactosyl. Escherichia coli-derived ß-d-galactosidase at concentrations of 1.9-15nM hydrolysed 56µM substrate with half-lives of 140-19min, respectively, producing the IPA/NO anion (iPrHN-N(O)NO(-), half-life ∼3min), which in turn spontaneously hydrolysed to mixtures of HNO with NO. Using saturating substrate concentrations furnished IPA/NO generation rates that were directly proportional to enzyme concentration. Consistent with these data, the enzyme/substrate combination applied to ventricular myocytes isolated from wild-type mouse hearts resulted not only in a significant positive inotropic effect, but also rescued the cells from the negative inotropy, hypercontractions, and occasional cell death seen with the enzyme alone. This mechanism represents an alternate approach for achieving controlled fluxes of NO/HNO to investigate their biological actions.


Asunto(s)
Óxido Nítrico/metabolismo , Óxidos de Nitrógeno/metabolismo , beta-Galactosidasa/metabolismo , Animales , Aniones , Compuestos Azo/metabolismo , Hidrólisis , Ratones , Células Musculares/metabolismo , Miocardio/citología , Miocardio/metabolismo
6.
J Phys Chem A ; 117(30): 6671-7, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23834533

RESUMEN

Amine-based diazeniumdiolates (NONOates) have garnered widespread use as nitric oxide (NO) donors, and their potential for nitroxyl (HNO) release has more recently been realized. While NO release rates can vary significantly with the type of amine, half-lives of seconds to days under physiological conditions, there is as yet no way to determine a priori the NO or HNO production rates of a given species, and no discernible trends have manifested other than that secondary amines produce only NO (i.e., no HNO). As a step to understanding these complex systems, here we describe a procedure for modeling amine-based NONOates in water solvent that provides an excellent correlation (R(2) = 0.94) between experimentally measured dissociation rates of seven secondary amine species and their computed NO release activation energies. The significant difference in behavior of NONOates in the gas and solvent phases is also rigorously demonstrated via explicit additions of quantum mechanical water molecules. The presented results suggest that the as-yet unsynthesized simplest amine-based NONOate, the diazeniumdiolated ammonia anion [H2N-N(O)═NO(-)], could serve as an unperturbed HNO donor. These results provide a step forward toward the accurate modeling of general NO and/or HNO donors as well as for the identification of tailored prodrug candidates.


Asunto(s)
Compuestos Azo/química , Radicales Libres/química , Donantes de Óxido Nítrico/química , Óxido Nítrico/química , Óxidos de Nitrógeno/química , Agua/química , Semivida , Cinética , Teoría Cuántica , Termodinámica
7.
Chem Res Toxicol ; 25(12): 2670-7, 2012 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-23106594

RESUMEN

Attachment of glutathione (GSH) to cysteine residues in proteins (S-glutathionylation) is a reversible post-translational modification that can profoundly alter protein structure and function. Often serving in a protective role, for example, by temporarily saving protein thiols from irreversible oxidation and inactivation, glutathionylation can be identified and semiquantitatively assessed using anti-GSH antibodies, thought to be specific for recognition of the S-glutathionylation modification. Here, we describe an alternate mechanism of protein glutathionylation in which the sulfur atoms of the GSH and the protein's thiol group are covalently bound via a cross-linking agent, rather than through a disulfide bond. This form of thiol cross-linking has been shown to occur and has been confirmed by mass spectrometry at the solution chemistry level, as well as in experiments documenting the potent antiproliferative activity of the bis-diazeniumdiolate Double JS-K in H1703 cells in vitro and in vivo. The modification is recognized by the anti-GSH antibody as if it were authentic S-glutathionylation, requiring mass spectrometry to distinguish between them.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Azo/farmacología , Glutatión/metabolismo , Piperazinas/farmacología , Acetilcisteína/química , Actinas/metabolismo , Animales , Antineoplásicos/química , Compuestos Azo/química , Línea Celular Tumoral , Dinitrofluorobenceno/análogos & derivados , Dinitrofluorobenceno/química , Femenino , Glutatión/química , Humanos , Ratones , Ratones Desnudos , Nanopartículas/química , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Piperazinas/química
8.
J Org Chem ; 77(23): 10804-10, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23137026

RESUMEN

Diazeniumdiolates that have the structure RHN-N(O)═NOR' are of interest as prodrug (caged) forms of the bioeffectors nitric oxide (NO) and nitroxyl (HNO). Previous work has focused on examples possessing α-branched R groups, with isopropylamine (IPA)/NO (R = isopropyl) being the smallest examined to date. To probe the effect of minimizing the alkyl-group size on the chemistry of IPA/NO, we prepared the corresponding methylamine derivative as a sodium salt that was highly unstable but could be trapped in very low overall yield as the stable O(2)-benzyl derivative. To prepare enough for efficient characterization, we devised an alternate synthesis involving a novel N-dealkylation route. CH(3)HN-N(O)═NOBn, synthesized in high yield and crystallized as the Z isomer as determined by X-ray crystallography, was observed to exist as a 11:1 mixture of two isomeric forms in dynamic equilibrium in solution. Similar results were seen for the O(2)-ethyl derivative, whose two equilibrium constituents were partially separated by HPLC to reveal essentially identical UV and mass spectra, indicating them to be Z and E isomers of CH(3)HN-N(O)═NOEt. The results could lead the way to a fuller understanding of the chemistry of the acyclic (E)-diazeniumdiolates.


Asunto(s)
Compuestos Aza/química , Compuestos Aza/síntesis química , Metilaminas/química , Metilaminas/síntesis química , Óxido Nítrico/química , Alquilación , Cristalografía por Rayos X , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Estructura Molecular
9.
Bioorg Med Chem ; 20(9): 3094-9, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22480849

RESUMEN

JS-K, a diazeniumdiolate-based nitric oxide (NO)-releasing prodrug, is currently in late pre-clinical development as an anti-cancer drug candidate. This prodrug was designed to be activated by glutathione (GSH) to release NO. To increase the potency of JS-K, we are investigating the effect of slowing the reaction of the prodrugs with GSH. Herein, we report the effect of replacement of nitro group(s) by other electron-withdrawing group(s) in JS-K and its homo-piperazine analogues on GSH activation and the drugs' biological activity. We show that nitro-to-cyano substitution increases the half-life of the prodrug in the presence of GSH without compromising the compound's in vivo antitumor activity.


Asunto(s)
Antineoplásicos/química , Compuestos Azo/química , Glutatión/metabolismo , Profármacos/química , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Compuestos Azo/farmacología , Estabilidad de Medicamentos , Semivida , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Cinética , Ratones , Neoplasias/tratamiento farmacológico , Óxido Nítrico/metabolismo , Profármacos/farmacología , Transducción de Señal/efectos de los fármacos
10.
J Pharmacol Exp Ther ; 336(2): 313-20, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20962031

RESUMEN

Non-small-cell lung cancer is among the most common and deadly forms of human malignancies. Early detection is unusual, and there are no curative therapies in most cases. Diazeniumdiolate-based nitric oxide (NO)-releasing prodrugs are a growing class of promising NO-based therapeutics. Here, we show that O(2)-(2,4-dinitrophenyl)-1-[(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate (JS-K) is a potent cytotoxic agent against a subset of human non-small-cell lung cancer cell lines both in vitro and as xenografts in mice. JS-K treatment led to 75% reduction in the growth of H1703 lung adenocarcinoma cells in vivo. Differences in sensitivity to JS-K in different lung cancer cell lines seem to be related to their endogenous levels of reactive oxygen species (ROS)/reactive nitrogen species (RNS). Other related factors, levels of peroxiredoxin 1 (PRX1) and 8-oxo-deoxyguanosine glycosylase (OGG1), also correlated with drug sensitivity. Treatment of the lung adenocarcinoma cells with JS-K resulted in oxidative/nitrosative stress in cells with high basal levels of ROS/RNS, which, combined with the arylating properties of the compound, was reflected in glutathione depletion and alteration in cellular redox potential, mitochondrial membrane permeabilization, and cytochrome c release. Inactivation of manganese superoxide dismutase by nitration was associated with increased superoxide and significant DNA damage. Apoptosis followed these events. Taken together, the data suggest that diazeniumdiolate-based NO-releasing prodrugs may have application as a personalized therapy for lung cancers characterized by high levels of ROS/RNS. PRX1 and OGG1 proteins, which can be easily measured, could function as biomarkers for identifying tumors sensitive to the therapy.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Azo/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Piperazinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/efectos de los fármacos , Compuestos Azo/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Daño del ADN , Femenino , Glutatión/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Mitocondrias/metabolismo , Piperazinas/uso terapéutico , Especies de Nitrógeno Reactivo/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
PLoS One ; 11(9): e0163821, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27684484

RESUMEN

Dietary isothiocyanates abundant as glucosinolate precursors in many edible cruciferous vegetables are effective for prevention of cancer in chemically-induced and transgenic rodent models. Some of these agents, including phenethyl isothiocyanate (PEITC), have already advanced to clinical investigations. The primary route of isothiocyanate metabolism is its conjugation with glutathione (GSH), a reaction catalyzed by glutathione S-transferase (GST). The pi class GST of subunit type 1 (hGSTP1) is much more effective than the alpha class GST of subunit type 1 (hGSTA1) in catalyzing the conjugation. Here, we report the crystal structures of hGSTP1 and hGSTA1 each in complex with the GSH adduct of PEITC. We find that PEITC also covalently modifies the cysteine side chains of GST, which irreversibly inhibits enzymatic activity.

12.
Biochem Pharmacol ; 93(3): 389-400, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25534988

RESUMEN

BACKGROUND AND PURPOSE: There is an unmet medical need for novel NAFLD treatments. Here we have examined the effects of liver-selective NO donor (V-PYRRO/NO) as compared with metformin on hepatic steatosis and glucose tolerance in mice fed high fat diet. MATERIAL AND METHODS: Effects of V-PYRRO/NO (5 mgkg(-1)) or metformin (616 mgkg(-1)) were examined in C57BL/6J mice fed high fat diet (HF, 60 kcal% fat). Quantitative determination of steatosis, liver fatty acid composition and western blot analysis of selected proteins involved in mitochondrial biogenesis, fatty acid de novo synthesis and oxidation, triacylglycerols and cholesterol transport from the liver were performed. Liver NOx and nitrate concentration and blood biochemistry were also analyzed. RESULTS: V-PYRRO/NO and metformin reduced liver steatosis with simultaneous reduction of total liver triacylglycerols, diacylglycerols and ceramides fraction and reversed HF-induced decrease in UFA/SFA ratio. V-PYRRO/NO substantially improved postprandial glucose tolerance, while the effect of metformin was modest and more pronounced on HOMA IR index. The anti-steatotic mechanism of V-PYRRO/NO was dependent on NO release, differed from that of metformin and involved improved glucose tolerance and inhibition of de novo fatty acid synthesis by Akt activation and ACC phosphorylation. In turn, major mechanism of metformin action involved increased expression of proteins implicated in mitochondrial biogenesis and metabolism (PGC-1α, PPARα, COX IV, cytochrome c, HADHSC). CONCLUSIONS: V-PYRRO/NO acts as a liver-specific NO donor prodrug affording pronounced anti-steatotic effects and may represent an efficient, mechanistically novel approach to prevent liver steatosis and insulin resistance.


Asunto(s)
Glucemia/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Hígado Graso/prevención & control , Donantes de Óxido Nítrico/uso terapéutico , Periodo Posprandial/efectos de los fármacos , Pirrolidinas/uso terapéutico , Animales , Glucemia/metabolismo , Hígado Graso/sangre , Hígado Graso/etiología , Prueba de Tolerancia a la Glucosa/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Donantes de Óxido Nítrico/farmacología , Periodo Posprandial/fisiología , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Pirrolidinas/farmacología
13.
J Med Chem ; 57(6): 2292-302, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24521039

RESUMEN

We report the antitumor effects of nitric oxide (NO) releasing derivatives of the PARP-1 inhibitor olaparib (1). Compound 5b was prepared by coupling the carboxyl group of 3b and the free amino group of arylated diazeniumdiolated piperazine 4. Analogue 5a has the same structure except that the F is replaced by H. Compound 13 is the same as 5b except that a Me2N-N(O)═NO- group was added para and ortho to the nitro groups of the dinitrophenyl ring. The resulting prodrugs are activated by glutathione in a reaction accelerated by glutathione S-transferase P1 (GSTP1), an enzyme frequently overexpressed in cancers. This metabolism generates NO plus a PARP-1 inhibitor simultaneously, consuming reducing equivalents, leading to DNA damage concomitant with inhibition of DNA repair, and in the case of 13 inducing cross-linking glutathionylation of proteins. Compounds 5b and 13 reduced the growth rates of A549 human lung adenocarcinoma xenografts with no evidence of systemic toxicity.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Gutatión-S-Transferasa pi/metabolismo , Neoplasias/enzimología , Óxido Nítrico/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayo Cometa , Daño del ADN , Diseño de Fármacos , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Humanos , Isoenzimas/efectos de los fármacos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Poli(ADP-Ribosa) Polimerasa-1 , Profármacos/síntesis química , Profármacos/farmacología , Pirazinas/farmacología , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Leuk Res ; 38(3): 377-82, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24461365

RESUMEN

The nitric oxide (NO) prodrug JS-K, a promising anti-cancer agent, consists of a diazeniumdiolate group necessary for the release of NO as well as an arylating ring. In this study, we research the mechanism by which JS-K kills a murine erythroleukemia cell line and determine the roles of NO and arylation in the process. Our studies indicate that JS-K inhibits the PI 3-kinase/Akt and MAP kinase pathways. This correlates with the activation of the tumor suppressor FoxO3a and increased expression of various caspases, leading to apoptosis. The arylating capability of JS-K appears to be sufficient for inducing these biological effects. Overall, these data suggest that JS-K kills tumor cells by arylating and inactivating signaling molecules that block the activation of a tumor suppressor.


Asunto(s)
Compuestos Azo/farmacología , Citotoxinas/farmacología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Piperazinas/farmacología , Profármacos/farmacología , Animales , Caspasas/genética , Caspasas/metabolismo , Línea Celular Tumoral , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/agonistas , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
15.
Redox Biol ; 1: 115-24, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24024144

RESUMEN

JS-K is a nitric oxide (NO)-releasing prodrug of the O (2)-arylated diazeniumdiolate family that has demonstrated pronounced cytotoxicity and antitumor properties in a variety of cancer models both in vitro and in vivo. The current study of the metabolic actions of JS-K was undertaken to investigate mechanisms of its cytotoxicity. Consistent with model chemical reactions, the activating step in the metabolism of JS-K in the cell is the dearylation of the diazeniumdiolate by glutathione (GSH) via a nucleophilic aromatic substitution reaction. The resulting product (CEP/NO anion) spontaneously hydrolyzes, releasing two equivalents of NO. The GSH/GSSG redox couple is considered to be the major redox buffer of the cell, helping maintain a reducing environment under basal conditions. We have quantified the effects of JS-K on cellular GSH content, and show that JS-K markedly depletes GSH, due to JS-K's rapid uptake and cascading release of NO and reactive nitrogen species. The depletion of GSH results in alterations in the redox potential of the cellular environment, initiating MAPK stress signaling pathways, and inducing apoptosis. Microarray analysis confirmed signaling gene changes at the transcriptional level and revealed alteration in the expression of several genes crucial for maintenance of cellular redox homeostasis, as well as cell proliferation and survival, including MYC. Pre-treating cells with the known GSH precursor and nucleophilic reducing agent N-acetylcysteine prevented the signaling events that lead to apoptosis. These data indicate that multiplicative depletion of the reduced glutathione pool and deregulation of intracellular redox balance are important initial steps in the mechanism of JS-K's cytotoxic action.


Asunto(s)
Compuestos Azo/farmacología , Leucemia/metabolismo , Donantes de Óxido Nítrico/farmacología , Piperazinas/farmacología , Profármacos/síntesis química , Acetilcisteína/farmacología , Compuestos Azo/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Humanos , Leucemia/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/síntesis química , Oxidación-Reducción/efectos de los fármacos , Piperazinas/síntesis química , Profármacos/farmacología , Especies de Nitrógeno Reactivo/metabolismo
16.
J Med Chem ; 56(20): 7804-20, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24102516

RESUMEN

Structural modifications of nonsteroidal anti-inflammatory drugs (NSAIDs) have successfully reduced the side effect of gastrointestinal ulceration without affecting anti-inflammatory activity, but they may increase the risk of myocardial infarction with chronic use. The fact that nitroxyl (HNO) reduces platelet aggregation, preconditions against myocardial infarction, and enhances contractility led us to synthesize a diazeniumdiolate-based HNO-releasing aspirin and to compare it to an NO-releasing analogue. Here, the decomposition mechanisms are described for these compounds. In addition to protection against stomach ulceration, these prodrugs exhibited significantly enhanced cytotoxcity compared to either aspirin or the parent diazeniumdiolate toward nonsmall cell lung carcinoma cells (A549), but they were not appreciably toxic toward endothelial cells (HUVECs). The HNO-NSAID prodrug inhibited cylcooxgenase-2 and glyceraldehyde 3-phosphate dehydrogenase activity and triggered significant sarcomere shortening on murine ventricular myocytes compared to control. Together, these anti-inflammatory, antineoplasic, and contractile properties suggest the potential of HNO-NSAIDs in the treatment of inflammation, cancer, or heart failure.


Asunto(s)
Aspirina/síntesis química , Aspirina/farmacología , Compuestos Azo/química , Óxido Nítrico/química , Óxidos de Nitrógeno/química , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Aspirina/química , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Gliceraldehído 3-Fosfato Deshidrogenasa (NADP+)/metabolismo , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Químicos , Estructura Molecular , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Profármacos/síntesis química , Profármacos/química , Profármacos/farmacología , Sarcómeros/efectos de los fármacos , Sarcómeros/metabolismo
17.
For Immunopathol Dis Therap ; 3(2): 91-95, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23585982

RESUMEN

Promising drug candidates of the diazeniumdiolate (NONOate) chemical family include several types of thiol modification among their mechanisms of action: 1) drugs designed to release nitric oxide (NO) on reaction with the thiol group of glutathione (GSH) arylate the GSH, a step that removes reducing equivalents from the cell; (2) a similar reaction of the drug with the thiol group of a protein changes its structure, leading to potentially impaired function and cell death; (3) the NO generated as a byproduct in the above reactions can undergo oxidation, leading to S-nitrosylation and S-glutathionylation; and (4) diazeniumdiolates can also generate nitroxyl, which reacts with thiol groups to form disulfides or sulfinamides.

18.
Org Lett ; 12(19): 4256-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20812718

RESUMEN

The use of Cu(I)-catalyzed "click" reactions of alkyne-substituted diazeniumdiolate prodrugs with bis- and tetrakis-azido compounds is described. The "click" reaction for the bis-azide using CuSO(4)/Na-ascorbate predominantly gave the expected bis-triazole. However, CuI/diisopropylethylamine predominantly gave uncommon triazolo-triazole products as a result of oxidative coupling. Neither set of "click" conditions showed evidence of compromising the integrity of the diazeniumdiolate groups. The chemistry developed has applications in the synthesis of polyvalent and dendritic nitric oxide donors.


Asunto(s)
Compuestos Azo/química , Donantes de Óxido Nítrico/química , Azidas/química , Ciclización , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA