Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38673999

RESUMO

E. coli nitroreductase A (NfsA) is a candidate for gene-directed prodrug cancer therapy using bioreductively activated nitroaromatic compounds (ArNO2). In this work, we determined the standard redox potential of FMN of NfsA to be -215 ± 5 mV at pH 7.0. FMN semiquinone was not formed during 5-deazaflavin-sensitized NfsA photoreduction. This determines the two-electron character of the reduction of ArNO2 and quinones (Q). In parallel, we characterized the oxidant specificity of NfsA with an emphasis on its structure. Except for negative outliers nitracrine and SN-36506, the reactivity of ArNO2 increases with their electron affinity (single-electron reduction potential, E17) and is unaffected by their lipophilicity and Van der Waals volume up to 386 Å. The reactivity of quinoidal oxidants is not clearly dependent on E17, but 2-hydroxy-1,4-naphthoquinones were identified as positive outliers and a number of compounds with diverse structures as negative outliers. 2-Hydroxy-1,4-naphthoquinones are characterized by the most positive reaction activation entropy and the negative outlier tetramethyl-1,4-benzoquinone by the most negative. Computer modelling data showed that the formation of H bonds with Arg15, Arg133, and Ser40, plays a major role in the binding of oxidants to reduced NfsA, while the role of the π-π interaction of their aromatic structures is less significant. Typically, the calculated hydride-transfer distances during ArNO2 reduction are smallwer than for Q. This explains the lower reactivity of quinones. Another factor that slows down the reduction is the presence of positively charged aliphatic substituents.


Assuntos
Proteínas de Escherichia coli , Escherichia coli , Nitrorredutases , Oxirredução , Pró-Fármacos , Nitrorredutases/metabolismo , Nitrorredutases/química , Nitrorredutases/genética , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Especificidade por Substrato , Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Potenciometria , Catálise , Simulação de Acoplamento Molecular
2.
Int J Mol Sci ; 24(15)2023 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-37569833

RESUMO

NADPH:thioredoxin reductase (TrxR) is considered a potential target for anticancer agents. Several nitroheterocyclic sulfones, such as Stattic and Tri-1, irreversibly inhibit TrxR, which presumably accounts for their antitumor activity. However, it is necessary to distinguish the roles of enzymatic redox cycling, an inherent property of nitroaromatics (ArNO2), and the inhibition of TrxR in their cytotoxicity. In this study, we calculated the previously unavailable values of single-electron reduction potentials of known inhibitors of TrxR (Stattic, Tri-1, and 1-chloro-2,4-dinitrobenzene (CDNB)) and inhibitors identified (nitrofuran NSC697923 and nitrobenzene BTB06584). These calculations were according to the rates of their enzymatic single-electron reduction (PMID: 34098820). This enabled us to compare their cytotoxicity with that of model redox cycling ArNO2. In MH22a and HCT-116 cells, Tri-1, Stattic, CDNB, and NSC697023 possessed at least 10-fold greater cytotoxicity than can be expected from their redox cycling activity. This may be related to TrxR inhibition. The absence of enhanced cytotoxicity in BTB06548 may be attributed to its instability. Another known inhibitor of TrxR, tetryl, also did not possess enhanced cytotoxicity, probably because of its detoxification by DT-diaphorase (NQO1). Apart from the reactions with NQO1, the additional mechanisms influencing the cytotoxicity of the examined inhibitors of TrxR are their reactions with cytochromes P-450. Furthermore, some inhibitors, such as Stattic and NSC697923, may also inhibit glutathione reductase. We suggest that these data may be instrumental in the search for TrxR inhibitors with enhanced cytotoxic/anticancer activity.


Assuntos
Antineoplásicos , Tiorredoxina Dissulfeto Redutase , Animais , Tiorredoxina Dissulfeto Redutase/metabolismo , Antineoplásicos/farmacologia , Oxirredução , Mamíferos/metabolismo
3.
Int J Mol Sci ; 24(7)2023 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-37047836

RESUMO

3-Amino-1,2,4-benzotriazine-1,4-dioxide (tirapazamine, TPZ) and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities. Their action is attributed to the enzymatic single-electron reduction to free radicals that initiate the prooxidant processes. In order to clarify the mechanisms of aerobic mammalian cytotoxicity of ArN→O, we derived a TPZ-resistant subline of murine hepatoma MH22a cells (resistance index, 5.64). The quantitative proteomic of wild-type and TPZ-resistant cells revealed 5818 proteins, of which 237 were up- and 184 down-regulated. The expression of the antioxidant enzymes aldehyde- and alcohol dehydrogenases, carbonyl reductases, catalase, and glutathione reductase was increased 1.6-5.2 times, whereas the changes in the expression of glutathione peroxidase, superoxide dismutase, thioredoxin reductase, and peroxiredoxins were less pronounced. The expression of xenobiotics conjugating glutathione-S-transferases was increased by 1.6-2.6 times. On the other hand, the expression of NADPH:cytochrome P450 reductase was responsible for the single-electron reduction in TPZ and for the 2.1-fold decrease. These data support the fact that the main mechanism of action of TPZ under aerobic conditions is oxidative stress. The unchanged expression of intranuclear antioxidant proteins peroxiredoxin, glutaredoxin, and glutathione peroxidase, and a modest increase in the expression of DNA damage repair proteins, tend to support non-site-specific but not intranuclear oxidative stress as a main factor of TPZ aerobic cytotoxicity.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Tirapazamina/farmacologia , Triazinas/farmacologia , Antineoplásicos/farmacologia , Antioxidantes , Proteômica , Oxirredução , Glutationa Peroxidase , Mamíferos
4.
Free Radic Res ; 55(3): 246-254, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34098820

RESUMO

The therapeutic action of nitroaromatic antiandrogens nilutamide and flutamide may be complicated by their cytotoxicity, whose mechanisms are still incomprehensively understood. In particular this concerns the enzymatic redox cycling of flutamide and its metabolites, and its impact on their cytotoxicity. In this work, we examined the single-electron reduction of nilutamide, flutamide, its metabolites 2-hydroxyflutamide and 4-nitro-3-trifluorormethyl-phenylamine, and a topical antiandrogen (3-amino-2-hydroxy-2-methyl-N-(4-nitro-3-trifluoromethyl)-phenyl) propanamide by NADPH:cytochrome P-450 reductase and adrenodoxin reductase/adrenodoxin. The obtained steady-state bimolecular rate constants of oxidant reduction (kcat/Km) enabled to establish single-electron reduction midpoint potentials (E17) of compounds, -0.377 - -0.413 V, which were in line with enthalpies of formation of their free radicals, obtained by quantum mechanical calculations. Using murine hepatoma MH22a cells, the obtained cytotoxicity vs. E17 correlation based on the data of model nitroaromatic compounds shows that redox cycling and oxidative stress could be the main factor of cytotoxicity of nitroaromatic antiandrogens. Other minor cytotoxicity factors could be their redox metabolism involving NAD(P)H:quinone oxidoreductase (NQO1) and cytochromes P-450.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Oxirredução/efeitos dos fármacos , Antagonistas de Androgênios/farmacologia , Animais , Catálise , Elétrons , Humanos , Ratos
5.
Int J Mol Sci ; 21(22)2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33228195

RESUMO

Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit tumoricidal, antibacterial, and antiprotozoal activities, which are typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the role of NAD(P)H:quinone oxidoreductase (NQO1) in ArN→O aerobic cytotoxicity. We synthesized 9 representatives of ArN→O with uncharacterized redox properties and examined their single-electron reduction by rat NADPH:cytochrome P-450 reductase (P-450R) and Plasmodium falciparum ferredoxin:NADP+ oxidoreductase (PfFNR), and by rat NQO1. NQO1 catalyzed both redox cycling and the formation of stable reduction products of ArN→O. The reactivity of ArN→O in NQO1-catalyzed reactions did not correlate with the geometric average of their activity towards P-450R- and PfFNR, which was taken for the parameter of their redox cycling efficacy. The cytotoxicity of compounds in murine hepatoma MH22a cells was decreased by antioxidants and the inhibitor of NQO1, dicoumarol. The multiparameter regression analysis of the data of this and a previous study (DOI: 10.3390/ijms20184602) shows that the cytotoxicity of ArN→O (n = 18) in MH22a and human colon carcinoma HCT-116 cells increases with the geometric average of their reactivity towards P-450R and PfFNR, and with their reactivity towards NQO1. These data demonstrate that NQO1 is a potentially important target of action of heteroaromatic N-oxides.


Assuntos
Antibacterianos/farmacologia , Antioxidantes/farmacologia , Antiprotozoários/farmacologia , Óxidos N-Cíclicos/farmacologia , Ferredoxina-NADP Redutase/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , NADPH-Ferri-Hemoproteína Redutase/antagonistas & inibidores , Aerobiose , Animais , Antibacterianos/síntese química , Antioxidantes/síntese química , Antiprotozoários/síntese química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Óxidos N-Cíclicos/síntese química , Dicumarol/farmacologia , Ensaios Enzimáticos , Inibidores Enzimáticos/farmacologia , Ferredoxina-NADP Redutase/química , Ferredoxina-NADP Redutase/metabolismo , Células HCT116 , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Hepatócitos/patologia , Humanos , Cinética , Camundongos , NAD(P)H Desidrogenase (Quinona)/química , NAD(P)H Desidrogenase (Quinona)/metabolismo , NADPH-Ferri-Hemoproteína Redutase/química , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Oxirredução , Plasmodium falciparum/química , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Ratos , Tirapazamina/química , Tirapazamina/farmacologia
6.
Int J Mol Sci ; 20(18)2019 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-31533349

RESUMO

Derivatives of tirapazamine and other heteroaromatic N-oxides (ArN→O) exhibit promising antibacterial, antiprotozoal, and tumoricidal activities. Their action is typically attributed to bioreductive activation and free radical generation. In this work, we aimed to clarify the mechanism(s) of aerobic mammalian cell cytotoxicity of ArN→O performing the parallel studies of their reactions with NADPH:cytochrome P-450 reductase (P-450R), adrenodoxin reductase/adrenodoxin (ADR/ADX), and NAD(P)H:quinone oxidoreductase (NQO1); we found that in P-450R and ADR/ADX-catalyzed single-electron reduction, the reactivity of ArN→O (n = 9) increased with their single-electron reduction midpoint potential (E17), and correlated with the reactivity of quinones. NQO1 reduced ArN→O at low rates with concomitant superoxide production. The cytotoxicity of ArN→O in murine hepatoma MH22a and human colon adenocarcinoma HCT-116 cells increased with their E17, being systematically higher than that of quinones. The cytotoxicity of both groups of compounds was prooxidant. Inhibitor of NQO1, dicoumarol, and inhibitors of cytochromes P-450 α-naphthoflavone, isoniazid and miconazole statistically significantly (p < 0.02) decreased the toxicity of ArN→O, and potentiated the cytotoxicity of quinones. One may conclude that in spite of similar enzymatic redox cycling rates, the cytotoxicity of ArN→O is higher than that of quinones. This is partly attributed to ArN→O activation by NQO1 and cytochromes P-450. A possible additional factor in the aerobic cytotoxicity of ArN→O is their reductive activation in oxygen-poor cell compartments, leading to the formation of DNA-damaging species similar to those forming under hypoxia.


Assuntos
Antineoplásicos/farmacologia , Oxidantes/farmacologia , Tirapazamina/farmacologia , Antineoplásicos/química , Biomarcadores , Humanos , Estrutura Molecular , NAD(P)H Desidrogenase (Quinona)/metabolismo , NADP/metabolismo , Oxidantes/química , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio , Tirapazamina/análogos & derivados , Tirapazamina/química
7.
EXCLI J ; 16: 663-678, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28694766

RESUMO

Nitrogen-based tetracyclic ortho-quinones (naphtho[1'2':4.5]imidazo[1,2-a]pyridine-5,6-diones, NPDOs) and their nitro-substituted derivatives (nitro-(P)NPDOs) were obtained by condensation of substituted 2,3-dichloro-1,4-naphthoquinones with 2-amino-pyridine and -pyrimidine and nitration at an elevated temperature. The structural features of the compounds as well as their global and regional electrophilic potency were characterized by means of DFT computation. The compounds were highly reactive substrates of single- and two-electron (hydride) - transferring P-450R (CPR; EC 1.6.2.4) and NQO-1 (DTD; EC 1.6.99.2), respectively, concomitantly producing reactive oxygen species. Their catalytic efficiency defined in terms of the apparent second-order rate constant (kcat/KM (Q)) values in P-450R- and NQO-1-mediated reactions varied in the range of 3-6 × 107 M-1 s-1 and 1.6-7.4 × 108 M-1 s-1, respectively. The cytotoxic activities of the compounds on tumor cell lines followed the concentration-dependent manner exhibiting relatively high cytotoxic potency against breast cancer MCF-7, with CL50 values of 0.08-2.02 µM L-1 and lower potency against lung cancer A-549 (CL50 = 0.28-7.66 µM L-1). 3-nitro-pyrimidino-NPDO quinone was the most active compound against MCF-7 with CL50 of 0.08 ± 0.01 µM L-1 (0.02 µg mL-1)) which was followed by 3-nitro-NPDO with CL50 of 0.12 ± 0.03 µM L-1 (0.035 µg mL-1)) and 0.28 ± 0.08 µM L-1 (0.08 µg mL-1) on A-549 and MCF-7 cells, respectively, while 1- and 4-nitro-quinoidals produced the least cytotoxic effects. Tumor cells quantified by AO/EB staining showed that the cell death induced by the compounds occurs primarily through apoptosis.

8.
EXCLI J ; 16: 151-159, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28435434

RESUMO

A number of quinones have been shown to be efficient anticancer agents. However, some mechanisms of their action, in particular cell signaling are not well understood. The aim of this study was to partly fill this gap by characterizing the mode of cytotoxicity of 2,5-diaziridinyl-3,6-dimethyl-1,4-benzoquinone (MeDZQ) in malignant mouse hepatoma cells (MH-22A) with regard to the expression and activation of main molecules in MAPK cell signaling pathway. The study revealed unequal roles of MAP kinases in MeDZQ-induced cell death: the compound did not induce significant changes in ERK expression or its phosphorylation; JNK appeared to be responsible for cell survival, however, p38 kinase was shown to be involved in cell death. In order to assess the enzymatic activation mechanisms responsible for the action of MeDZQ, we have also found that the antioxidant N,N'-diphenyl-p-phenylene diamine, the iron-chelating agent desferrioxamine, and DT-diaphorase inhibitor, dicoumarol, partly protected the cells from MeDZQ cytotoxicity. It points to parallel oxidative stress and bioreductive alkylation modes of the cytotoxicity of MeDZQ.

9.
Spectrochim Acta A Mol Biomol Spectrosc ; 178: 136-141, 2017 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-28182983

RESUMO

The detailed structure, chemical and spectroscopic properties of the derivatives of the selected 2,5-bis(1-aziridinyl)-benzo-1,4-quinone conformers were studied by applying quantum chemical and experimental methods. The relationship between the structure and chemical activity of the selected 3 bifunctional bioreductive quinonic anticancer agents - aziridinyl benzoquinones (AzBQ compounds) was obtained. The results obtained showed that the position of aziridine rings influenced by the chemical activity of the investigated compound were more significant than the substitutions of the benzene ring of the AzBQ compounds. The solvents influencing this activity were obtained, too.


Assuntos
Aziridinas/química , Modelos Moleculares , Quinonas/química , Cristalografia por Raios X , Dureza , Conformação Molecular , Fenômenos Ópticos , Quinonas/síntese química , Espectrofotometria Ultravioleta , Termodinâmica , Difração de Raios X
10.
Arch Biochem Biophys ; 614: 14-22, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27986535

RESUMO

NfsA, a major FMN-associated nitroreductase of E. coli, reduces nitroaromatic compounds via consecutive two-electron transfers. NfsA has potential applications in the biodegradation of nitroaromatic environment pollutants, e.g. explosives, and is also of interest for the anticancer strategy gene-directed enzyme prodrug therapy. However, the catalytic mechanism of NfsA is poorly characterized. Here we examined the NADPH-dependent reduction of quinones (n = 16) and nitroaromatic compounds (n = 12) by NfsA. We confirmed a general "ping-pong" reaction scheme, and preliminary rapid reaction studies of the enzyme reduction by NADPH showed that this step is much faster than the steady-state turnover number, i.e., the enzyme turnover is limited by the oxidative half-reaction. The reactivity of nitroaromatic compounds (log kcat/Km) followed a linear dependence on their single-electron reduction potential (E17), indicating a limited role for compound structure or active site flexibility in their reactivity. The reactivity of quinones was lower than that of nitroaromatics having similar E17 values, except for the significantly enhanced reactivity of 2-OH-1,4-naphthoquinones, consistent with observations previously made for the group B nitroreductase of Enterobacter cloacae. We present evidence that the reduction of quinones by NfsA is most consistent with a single-step (H-) hydride transfer mechanism.


Assuntos
Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Nitrorredutases/química , Quinonas/química , Catálise , Domínio Catalítico , Transporte de Elétrons , Elétrons , Concentração de Íons de Hidrogênio , Cinética , NADP/química , Nitrogênio/química , Oxirredução , Oxigênio/química , Ligação Proteica , Especificidade por Substrato , Temperatura
11.
Biochim Biophys Acta ; 1864(2): 219-32, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26596252

RESUMO

Acquired resistance of tumor cells to the therapeutic treatment is a major challenge in virtually any chemotherapy. A novel anticancer agent 2,5-diaziridinyl-3-(hydroxymethyl)-6-methyl-1,4-benzoquinone (RH1) is designed to be activated by NAD(P)H: quinone oxidoreductase, an enzyme expressed at high levels in many types of tumors. Here we investigated the potential mechanisms of acquired RH1 drug resistance in cancer cells by applying high-throughput differential quantitative proteomic analysis of the newly established RH1-resistant hepatoma cell lines. Over 400 proteins display significantly altered levels between drug-sensitive and drug-resistant cell lines. Differentially expressed proteins were clustered into more than 14 groups according to their functional annotation and protein-protein interactions. Bioinformatic analysis highlights the biological processes that might be responsible for acquired resistance to RH1. The level of several xenobiotic metabolism enzymes (total n=17) involved in RH1 activation and detoxification is decreased (Nqo1, catalase, Gst, Gsr), corresponding with the decrease in their catalytic activity. The altered biological processes also include the decrease of cell cycle positive regulators (n=15) and the increase of DNA repair proteins (n=5) as well as annexin family members (n=5) in the RH1-resistant cells. Drug-resistant hepatoma cell proteomes are also distinguished by the altered level of proteins involved in energy production and metabolism (n=55). Our data provide the basis for in-depth study of molecular mechanisms of tumor cell resistance to the promising anticancer drug RH1 enabling the further validation of protein biomarkers for the drug insusceptibility and of potential secondary pharmacological targets of RH1 resistant cells.


Assuntos
Aziridinas/administração & dosagem , Benzoquinonas/administração & dosagem , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Proteínas de Neoplasias/biossíntese , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/genética , Proteínas de Neoplasias/genética , Proteoma/efeitos dos fármacos , Proteoma/genética , Proteômica
12.
Chem Res Toxicol ; 29(1): 26-39, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26630137

RESUMO

Aziridinylquinone RH-1 (2,5-diaziridinyl-3-hydroxymethyl-6-methyl-cyclohexa-2,5-diene-1,4-dione) is a potential anticancer agent. RH-1 action is associated with NAD(P)H: quinone oxidoreductase (NQO1) which reduces this diaziridinylbenzoquinone into DNA-alkylating hydroquinone and is overexpressed in many tumors. Another suggested mechanism of RH-1 toxicity is the formation of reactive oxygen species (ROS) arising from its redox cycling. In order to improve anticancer action of this and similar antitumor quinones, we investigated the involvement of different signaling molecules in cytotoxicity induced by RH-1 by using wild-type tumor suppressor p53 bearing nonsmall cell lung carcinoma A549 cells as a model. Gradual and prolonged increase of mitogen-activated protein kinases (MAPK) ERK, P38, and JNK phosphorylation was observed during 24-h RH-1 treatment. In parallel, activation of DNA damage-sensing ATM kinase, upregulation, and phosphorylation of TP53 (human p53) took place. Inhibition studies revealed that RH-1-induced A549 apoptosis involved the NQO1-ATM-p53 signaling pathway and ROS generation. TP53 participated in ROS- and DNA damage-induced cell death differently. Moreover, MAP kinase JNK was another TP53 activator and death inducer in A549 cells. At the same time, rapid and prolonged activation of AKT kinase during RH-1 treatment was found, and it proved to be antiapoptotic kinase in our model system. Therefore, we identified that different and opposite cell death regulating signaling pathways, which may counteract one another, are induced in cancer cells during chemotherapeutic RH-1 treatment.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Aziridinas/farmacologia , Cicloexenos/farmacologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Aziridinas/química , Aziridinas/metabolismo , Linhagem Celular Tumoral , Cicloexenos/química , Cicloexenos/metabolismo , Dano ao DNA , Humanos , Espécies Reativas de Oxigênio/metabolismo
13.
Bioorg Med Chem Lett ; 26(2): 512-517, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26681508

RESUMO

Naphtho[1',2':4,5]imidazo[1,2-a]pyridine-5,6-diones (NPDOs), a new type of N-heterocycle-fused o-quinones, have been synthesized. They have been found to be efficient electron-accepting substrates of NADPH-dependent single-electron-transferring P-450R and two-electron transferring NQO1, generating reactive oxygen species (ROS) with a concomitant decrease in NADPH, which is consistent with redox-cycling. The reactivity of NPDOs toward P-450R (in terms of kcat/Km) varied in the range of 10(6)-10(7)M(-1)s(-1), while their reduction by NQO1 proceeded much faster, approaching the diffusion control limit (kcat/Km∼10(8)-10(9)M(-1)s(-1)). NPDOs exhibited relatively high cytotoxic activity against human lung carcinoma (A-549) and breast tumor (MCF-7) cell lines (LC50=0.1-8.3µM), while promyelocytic leukemia cells (HL-60) were less sensitive to NPDOs (LC50⩾10µM). 3-Nitro-substituted NPDO (11) revealed the highest potency against both A-549 and MCF-7 cell lines, with LC50 of 0.12±0.03µM and 0.28±0.08µM, respectively. Dicoumarol partly suppressed the activity of the compounds against A-594 and MCF-7 cell lines, suggesting that their cytotoxic action might be partially influenced by NQO1-mediated bioreductive activation.


Assuntos
Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Naftoquinonas/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Sistema Enzimático do Citocromo P-450/metabolismo , Dicumarol/farmacologia , Escherichia coli/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/síntese química , Compostos Heterocíclicos de 4 ou mais Anéis/química , Humanos , Imidazóis/síntese química , Imidazóis/química , Imidazóis/farmacologia , Naftoquinonas/síntese química , Naftoquinonas/química , Oxirredução , Piridinas/síntese química , Piridinas/química , Piridinas/farmacologia , Salmonella enterica/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos
14.
Acta Biochim Pol ; 61(4): 833-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25566548

RESUMO

The anticancer activity of aziridinyl-quinones is mainly attributed to their NAD(P)H: quinone oxidoreductase 1 (NQO1)-catalyzed two-electron reduction into DNA-alkylating products. However, little is known about their cytotoxicity in primary cells, which may be important in understanding their side effects. We found that the cytotoxicity of aziridinyl-unsubstituted quinones (n = 12) in mice splenocytes with a low amount of NQO1, 4 nmol × mg(-1) × min(-1), was caused mainly by the oxidative stress. Aziridinyl-benzoquinones (n = 6) including a novel anticancer agent RH1 were more cytotoxic than aziridinyl-unsubstituted ones with the similar redox properties, and their cytotoxicity was not decreased by an inhibitor of NQO1, dicumarol. The possible reasons for their enhanced cytotoxicity are discussed.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/química , Benzoquinonas/efeitos adversos , Benzoquinonas/química , Baço/metabolismo , Animais , Células Cultivadas , Feminino , Masculino , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Baço/citologia
15.
Acta Biochim Pol ; 60(2): 217-22, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23748219

RESUMO

We examined the kinetics of single-electron reduction of a large number of structurally diverse quinones and nitroaromatic compounds, including a number of antitumour and antiparasitic drugs, and nitroaromatic explosives by recombinant rat neuronal nitric oxide synthase (nNOS, EC 1.14.13.39), aiming to characterize the role of nNOS in the oxidative stress-type cytotoxicity of the above compounds. The steady-state second-order rate constants (kcat/Km) of reduction of the quinones and nitroaromatics varied from 10² M⁻¹s⁻¹ to 106 M⁻¹s⁻¹, and increased with an increase in their single-electron reduction potentials (E¹7). The presence of Ca²âº/calmodulin enhanced the reactivity of nNOS. These reactions were consistent with an 'outer sphere' electron-transfer mechanism, considering the FMNH∙/FMNH2 couple of nNOS as the most reactive reduced enzyme form. An analysis of the reactions of nNOS within the 'outer sphere' electron-transfer mechanism gave the approximate values of the distance of electron transfer, 0.39-0.47 nm, which are consistent with the crystal structure of the reductase domain of nNOS. On the other hand, at low oxygen concentrations ([O2] = 40-50 µM), nNOS performs a net two-electron reduction of quinones and nitroaromatics. This implies that NOS may in part be responsible for the bioreductive alkylation by two-electron reduced forms of antitumour aziridinyl-substituted quinones under a modest hypoxia.


Assuntos
Aziridinas/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Quinonas/metabolismo , Xenobióticos/metabolismo , Animais , Benzoquinonas/metabolismo , Cálcio/metabolismo , Calmodulina/metabolismo , Transporte de Elétrons , Cinética , Nitrocompostos/metabolismo , Oxirredução , Relação Quantitativa Estrutura-Atividade , Ratos , Proteínas Recombinantes/metabolismo
16.
Acta Biochim Pol ; 60(2): 227-31, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23757451

RESUMO

In order to characterize the possible mechanism(s) of cytotoxicity of a neuroleptic agent 6,7-dinitrodihydroquinoxaline-2,3-dione (DNQX) we examined the redox properties of DNQX, and its mononitro- (NQX) and denitro- (QX) derivatives. The irreversible electrochemical reduction of the nitro groups of DNQX was characterized by the reduction peak potentials (Ep,7) of -0.43 V and -0.72 V vs. Ag/AgCl at pH 7.0, whereas NQX was reduced at Ep,7 = -0.67 V. The reactivities of DNQX and NQX towards the single-electron transferring enzymes NADPH:cytochrome P-450 reductase and NADPH:adrenodoxin reductase/adrenodoxin complex were similar to those of model nitrobenzenes with the single-electron reduction potential (E¹7) values of -0.29 V - -0.42 V. DNQX and NQX also acted as substrates for two-electron transferring mammalian NAD(P)H:quinone oxidoreductase (DT-diaphorase). The cytotoxicity of DNQX in bovine leukemia virus-transformed lamb kidney fibroblasts (line FLK) was prevented by antioxidants and an inhibitor of NQO1, dicoumarol, and was enhanced by the prooxidant alkylating agent 1,3-bis(2-chloromethyl)-1-nitrosourea. A comparison with model nitrobenzene compounds shows that the cytotoxicity of DNQX and NQX reasonably agrees with the ease of their electrochemical reduction, and/or their reactivities towards the used enzymatic single-electron reducing systems. Thus, our data imply that the cytotoxicity of DNQX in FLK cells is exerted mainly through oxidative stress.


Assuntos
Quinoxalinas/química , Animais , Antioxidantes/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Viral , Dicumarol/farmacologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , NADPH-Ferri-Hemoproteína Redutase/efeitos dos fármacos , Nitrobenzenos/farmacologia , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Quinoxalinas/farmacologia
17.
Acta Biochim Pol ; 59(2): 299-305, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22696302

RESUMO

Flavonoids exhibit prooxidant cytotoxicity in mammalian cells due to the formation of free radicals and oxidation products possessing quinone or quinomethide structure. However, it is unclear how the cytotoxicity of flavonoids depends on the ease of their single-electron oxidation in aqueous medium, i.e., the redox potential of the phenoxyl radical/phenol couple. We verified the previously calculated redox potentials for several flavonoids according to their rates of reduction of cytochrome c and ferricyanide, and proposed experimentally-based values of redox potentials for myricetin, fisetin, morin, kaempferol, galangin, and naringenin. We found that the cytotoxicity of flavonoids (n=10) in bovine leukemia virus-transformed lamb kidney fibroblasts (line FLK) and murine hepatoma (line MH-22a) increases with a decrease in their redox potential of the phenoxyl radical/phenol couple and an increase in their lipophilicity. Their cytotoxicity was decreased by antioxidants and inhibitors of cytochromes P-450, α-naphthoflavone and isoniazide, and increased by an inhibitor of catechol-O-methyltransferase, 3,5-dinitrocatechol. It shows that although the prooxidant action of flavonoids may be the main factor in their cytotoxicity, the hydroxylation and oxidative demethylation by cytochromes P-450 and O-methylation by catechol-O-methyltransferase can significantly modulate the cytotoxicity of the parent compounds.


Assuntos
Flavonoides/toxicidade , Oxidantes/toxicidade , Fenóis/química , Animais , Antioxidantes/farmacologia , Catecol O-Metiltransferase/metabolismo , Inibidores de Catecol O-Metiltransferase , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Citocromos c/química , Inibidores Enzimáticos/farmacologia , Ferricianetos/química , Flavonoides/química , Flavonoides/metabolismo , Hidroxilação , Cinética , Camundongos , Oxidantes/química , Oxidantes/metabolismo , Oxirredução , Relação Quantitativa Estrutura-Atividade , Ovinos , Termodinâmica
18.
Arch Biochem Biophys ; 512(2): 183-9, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21664341

RESUMO

Mitochondrial apoptosis-inducing factor (AIF) is a FAD-containing protein that under certain conditions translocates to the nucleus and causes a programmed cell death, apoptosis. The apoptogenic action of AIF is redox controlled as the NADH-reduced AIF dimer has lower affinity for DNA than the oxidized monomer. To gain further insights into the mechanism of AIF, we investigated its interaction with a series of quinone oxidants, including a number of anticancer quinones. Our data indicate that the NADH:quinone oxidoreduction catalyzed by AIF follows a "ping-pong" scheme, with the reductive half-reaction being rate-limiting and the FADH(-)-NAD(+) charge-transfer complex serving as an electron donor. AIF is equally reactive toward benzo- and naphthoquinones, but may discriminate structures with a higher number of aromatic rings. The reactivity of quinones is mainly defined by their one-electron reduction potential, whereas the size and nature of the substituents play a minor role. AIF is unlikely to significantly contribute to bioreductive activation of low-potential quinoidal anticancer quinones. However, high-potential quinones, e.g. a toxic natural compound naphthazarin, maintain AIF in the oxidized state when a significant excess of NADH is present. Thus, these compounds may prevent the accumulation of the reduced form of AIF in vivo, and enhance AIF-mediated apoptosis.


Assuntos
Fator de Indução de Apoptose/metabolismo , Animais , Apoptose/fisiologia , Fator de Indução de Apoptose/química , Fator de Indução de Apoptose/genética , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Técnicas In Vitro , Cinética , Camundongos , Modelos Biológicos , Oxidantes/metabolismo , Oxirredução , Quinonas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenobióticos/metabolismo
19.
Acta Biochim Pol ; 57(2): 157-63, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20454706

RESUMO

Strong radical-scavenging activity of Geranium macrorrhizum extracts isolated by using various solvent systems has been reported previously. This study aimed at expanding the knowledge on the bioactivities of antioxidatively active G. macrorrhizum butanol fraction, which was isolated from ethanolic extract (EB), and water fraction, which was isolated from water extract (WW) by measuring their singlet oxygen scavenging properties, as well as preliminary assessment of cytotoxicity and genotoxicity toward mammalian cells. The cytotoxicity (necrosis induction) of the extracts in bovine leukemia virus-transformed lamb kidney fibroblasts (line FLK) was partly prevented by antioxidants and stimulated by the prooxidant BCNU (N,N'-bis(2-chloroethyl)-N-nitrosourea). This indicates that the cytotoxicity of G. macrorrhizum extracts is at least partly attributed to their prooxidant action, presumably due to the formation of quinoidal products of their (auto)oxidation. The latter was evidenced by the nature of the peroxidase-catalyzed oxidation products, which supported DT-diaphorase-catalyzed oxidation of NADPH and participated in conjugation reactions with reduced glutathione. The genotoxic properties were studied using chromosome aberration (CA) and sister chromatid exchange (SCE) tests in human lymphocytes in vitro and Drosophila melanogaster somatic mutation and recombination test (SMART) in vivo. In the CA test, only the highest doses of both fractions significantly increased chromosome aberration frequency. In the SCE test, both fractions induced SCEs in a clear dose-dependent manner. G. macrorrhizum extracts were not genotoxic in the SMART test in vivo. Our data indicate that in spite of the possible beneficial (antioxidant) effects of Geranium extracts, the possibilities of their use as ingredients of functional foods and/or food supplements should be further examined due to their cyto- and genotoxic effects resulting mainly from the action of quercetin-derived components abundant in the extracts.


Assuntos
Eritrócitos/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Sequestradores de Radicais Livres/antagonistas & inibidores , Geranium/química , Linfócitos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Oxigênio Singlete/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Aberrações Cromossômicas/efeitos dos fármacos , Drosophila/efeitos dos fármacos , Humanos , Fígado/enzimologia , Oxigênio Singlete/antagonistas & inibidores
20.
Arch Biochem Biophys ; 494(1): 32-9, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19919822

RESUMO

Although quinones have been the subject of great interest as possible antimalarial agents, the mechanism of their antimalarial activity is poorly understood. Flavoenzyme electrontransferase-catalyzed redox cycling of quinones, and their inhibition of the antioxidant flavoenzyme glutathione reductase (GR, EC 1.8.1.7) have been proposed as possible mechanisms. Here, we have examined the activity of a number of quinones, including the novel antitumor agent RH1, against the malaria parasite Plasmodium falciparum strain FcB1 in vitro, their single-electron reduction rates by P. falciparum ferredoxin:NADP(+) reductase (PfFNR, EC 1.18.1.2), and their ability to inhibit P. falciparum GR. The multiparameter statistical analysis of our data implies, that the antiplasmodial activity of fully-substituted quinones (n=15) is relatively independent from their one-electron reduction potential (E(7)(1)). The presence of aziridinyl groups in quinone ring increased their antiplasmodial activity. Since aziridinyl-substituted quinones do not possess enhanced redox cycling activity towards PfFNR, we propose that they could act as as DNA-alkylating agents after their net two-electron reduction into aziridinyl-hydroquinones. We found that under the partial anaerobiosis, i.e., at the oxygen concentration below 40-50 microM, this reaction may be carried out by single-electron transferring flavoenzymes present in P. falciparum, like PfFNR. Another parameter increasing the antiplasmodial activity of fully-substituted quinones is an increase in their potency as P. falciparum GR inhibitors, which was revealed using multiparameter regression analysis. To our knowledge, this is the first quantitative demonstration of a link between the antiplasmodial activity of compounds and GR inhibition.


Assuntos
Aziridinas/química , Inibidores Enzimáticos/farmacologia , Glutationa Redutase/antagonistas & inibidores , Plasmodium falciparum/enzimologia , Quinonas/farmacologia , Animais , Inibidores Enzimáticos/química , Eritrócitos/enzimologia , Glutationa Redutase/sangue , Humanos , Cinética , Plasmodium falciparum/efeitos dos fármacos , Quinonas/química , Análise de Regressão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA