Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Intervalo de año de publicación
1.
Chem Res Toxicol ; 36(1): 66-82, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36548215

RESUMEN

Cancer incidence is increasing, and the drugs are not very selective. These drugs cause adverse effects, and the cells become resistant. Therefore, new drugs are needed. Here, we evaluated the effects of ZIM, a candidate for chemotherapy, and 4-AA alone and in association with commercial chemotherapeutic agents. Subsequently, the results of ZIM and 4-AA were compared. Male Swiss mice were treated with doses of 12, 24, or 48 mg/kg ZIM or 4-AA alone or in association with cisplatin (6 mg/kg), doxorubicin (16 mg/kg), and cyclophosphamide (100 mg/kg). Biometric parameters, DNA damage (comet and micronuclei), cell death, and splenic phagocytosis were evaluated. DNA docking was also performed to confirm the possible interactions of ZIM and 4-AA with DNA. 4-AA has been shown to have low genotoxic potential, increase the frequency of cell death, and activate phagocytosis. ZIM causes genomic and chromosomal damage in addition to causing cell death and activating phagocytosis. In association with chemotherapeutical agents, both 4-AA and ZIM have a chemopreventive effect and, therefore, reduce the frequency of DNA damage, cell death, and splenic phagocytosis. The association of 4-AA and ZIM with commercial chemotherapeutic agents increased the frequency of lymphocytes compared to chemotherapeutic agents alone. Molecular docking demonstrated that ZIM has more affinity for DNA than 4-AA and its precursors (1 and 2). This was confirmed by the lower interaction energy of the complex (-119.83 kcal/mol). ZIM can break the DNA molecule and, therefore, its chemotherapeutic effect can be related to DNA damage. It is considered that ZIM has chemotherapeutic potential. However, it should not be used in combination with cisplatin, doxorubicin, and cyclophosphamide as it reduces the effects of these drugs.


Asunto(s)
Antineoplásicos , Cisplatino , Ratones , Animales , Masculino , Cisplatino/toxicidad , Ampirona/farmacología , Simulación del Acoplamiento Molecular , Muerte Celular , Ciclofosfamida/farmacología , Doxorrubicina/farmacología , Daño del ADN , ADN , Norbornanos/farmacología , Antineoplásicos/toxicidad
2.
Genet Mol Biol ; 42(2): 399-410, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31259359

RESUMEN

Despite rapid advances in both the early detection and treatment of cancer, the mortality from this disease remains high, which justifies the development of new products that are more selective and effective and have fewer side effects. Accordingly, a novel ester was synthesized that contains two pharmacophores with important biological activities: (I) 4-aminoantipyrine, which has anti-inflammatory and antioxidant effects, and (II) the pharmacophore 1,4-dioxo-butenyl, which has cytotoxic activity. When administered alone, this compound is non-genotoxic, and it does not cause an increasing in splenic phagocytosis. Nevertheless, it can induce cell death. When administered in combination with commercial chemotherapeutic agents, such as doxorubicin, cisplatin, and cyclophosphamide, the ester shows antigenotoxic activity and decreases phagocytosis and reduces the potential to cause cell death. These results indicate that the compound should not be used in combination with chemotherapeutic agents that exert their effect through DNA damage, an important feature of antitumor drugs.

3.
Genet. mol. biol ; 41(1): 154-166, Jan.-Mar. 2018. tab, graf
Artículo en Inglés | LILACS | ID: biblio-892473

RESUMEN

Abstract The increased incidence of cancer and its high treatment costs have encouraged the search for new compounds to be used in adjuvant therapies for this disease. This study discloses the synthesis of (Z)-4-((1,5-dimethyl-3-oxo-2-phenyl-2,3dihydro-1H-pyrazol-4-yl) amino)-4-oxobut-2-enoic acid (IR-01) and evaluates not only the action of this compound on genetic integrity, increase in splenic phagocytosis and induction of cell death but also its effects in combination with the commercial chemotherapeutic agents doxorubicin, cisplatin and cyclophosphamide. IR-01 was designed and synthesized based on two multifunctionalyzed structural fragments: 4-aminoantipyrine, an active dipyrone metabolite, described as an antioxidant and anti-inflammatory agent; and the pharmacophore fragment 1,4-dioxo-2-butenyl, a cytotoxic agent. The results indicated that IR-01 is an effective chemoprotector because it can prevent clastogenic and/or aneugenic damage, has good potential to prevent genomic damage, can increase splenic phagocytosis and lymphocyte frequency and induces cell death. However, its use as an adjuvant in combination with chemotherapy is discouraged since IR-01 interferes in the effectiveness of the tested chemotherapeutic agents. This is a pioneer study as it demonstrates the chemopreventive effects of IR-01, which may be associated with the higher antioxidant activity of the precursor structure of 4-aminoantipyrine over the effects of the 1,4-dioxo-2-butenyl fragment.

4.
Genet Mol Biol ; 41(1): 154-166, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29473933

RESUMEN

The increased incidence of cancer and its high treatment costs have encouraged the search for new compounds to be used in adjuvant therapies for this disease. This study discloses the synthesis of (Z)-4-((1,5-dimethyl-3-oxo-2-phenyl-2,3dihydro-1H-pyrazol-4-yl) amino)-4-oxobut-2-enoic acid (IR-01) and evaluates not only the action of this compound on genetic integrity, increase in splenic phagocytosis and induction of cell death but also its effects in combination with the commercial chemotherapeutic agents doxorubicin, cisplatin and cyclophosphamide. IR-01 was designed and synthesized based on two multifunctionalyzed structural fragments: 4-aminoantipyrine, an active dipyrone metabolite, described as an antioxidant and anti-inflammatory agent; and the pharmacophore fragment 1,4-dioxo-2-butenyl, a cytotoxic agent. The results indicated that IR-01 is an effective chemoprotector because it can prevent clastogenic and/or aneugenic damage, has good potential to prevent genomic damage, can increase splenic phagocytosis and lymphocyte frequency and induces cell death. However, its use as an adjuvant in combination with chemotherapy is discouraged since IR-01 interferes in the effectiveness of the tested chemotherapeutic agents. This is a pioneer study as it demonstrates the chemopreventive effects of IR-01, which may be associated with the higher antioxidant activity of the precursor structure of 4-aminoantipyrine over the effects of the 1,4-dioxo-2-butenyl fragment.

5.
Regul Toxicol Pharmacol ; 92: 239-244, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29233772

RESUMEN

In the present study, mice were subjected to prolonged treatment with ethanolic extract of Salvia lachnostachys Benth leaves (SLEE), and the inflammatory and arthritic parameters were evaluated using the Complete Freund's Adjuvant (CFA) model. The genotoxicity of SLEE were also assayed using genetic toxicological tests. For the CFA model, 28 male C57BL/6 mice were distributed randomly into four groups (control, 50 mg/kg of SLEE, 100 mg/kg of SLEE and dexamethasone) for the evaluation of hyperalgesia and paw edema for 21 days after injection of CFA into the paw. To conduct the toxicogenetic assessments (comet assay and micronuclei assay), apoptosis and splenic phagocytosis were evaluated in male Swiss mice after the administration of saline (control group), cyclophosphamide (positive control group) and SLEE (10, 100 and 1000 mg/kg). SLEE significantly reduced the mechanical hyperalgesia and edema caused by CFA injection. The results of the toxicogenetic assessment revealed no toxicogenetic potential in the mice, and the evaluation of apoptosis showed an increase in apoptotic cells in the spleen after 72 h of treatment with SLEE (1000 mg/kg). SLEE exhibited anti-arthritic activity with no toxicogenetic damage. These toxicogenic results support the safety of SLEE.


Asunto(s)
Artritis/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Hojas de la Planta/química , Salvia/química , Animales , Apoptosis/efectos de los fármacos , Artritis/inducido químicamente , Canfanos , Edema/inducido químicamente , Edema/tratamiento farmacológico , Etanol/química , Adyuvante de Freund/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Panax notoginseng , Fitoterapia/métodos , Salvia miltiorrhiza , Toxicogenética/métodos
6.
Regul Toxicol Pharmacol ; 82: 32-38, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27793745

RESUMEN

This study evaluates the toxicological, genotoxic, mutagenic and apoptotic potential of an in vivo assay from Echinodorus macrophyllus extract (EEM). The acute toxicity test used 02 groups (n = 5) of female Wistar rats: negative control group (saline) and experimental group (2000 mg/kg b.w. EEM), both orally administered (gavage) at single doses and monitored for 14 days. To assess the genotoxic, mutagenic and apoptotic potential, 50 male Swiss mice were divided into 5 groups (n = 10): Group I: negative control (saline solution 0.1 ml/10 g b.w.); Group II: positive control (cyclophosphamide 100 mg/kg b.w.) intraperitoneally administered; groups III-V received EEM at 500, 1000 and 2000 mg/kg b.w., respectively. Groups I, III-V received oral administrations (gavage). The results showed that there was no acute lethality or any signs of acute toxicity, indicating that LD50 is greater than 2000 mg/kg b.w. The groups treated with EEM showed no genotoxic or mutagenic activity and did not induce apoptosis in the liver and kidney. Therefore, EEM showed no acute toxicity and at doses of 500, 1000 and 2000 mg/kg b.w. absence of genotoxicity, mutagenicity and no apoptotic events were observed.


Asunto(s)
Alismataceae/toxicidad , Apoptosis/efectos de los fármacos , Etanol/química , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Extractos Vegetales/toxicidad , Hojas de la Planta/toxicidad , Solventes/química , Toxicocinética , Administración Oral , Alismataceae/química , Animales , Cromatografía Líquida de Alta Presión , Ensayo Cometa , Femenino , Inyecciones Intraperitoneales , Riñón/patología , Dosificación Letal Mediana , Hígado/patología , Masculino , Ratones , Pruebas de Micronúcleos , Fitoterapia , Extractos Vegetales/administración & dosificación , Extractos Vegetales/aislamiento & purificación , Hojas de la Planta/química , Plantas Medicinales , Ratas Wistar , Medición de Riesgo , Factores de Tiempo , Pruebas de Toxicidad Aguda
7.
Artículo en Inglés | MEDLINE | ID: mdl-27402479

RESUMEN

The analgesic drug dipyrone is used to treat side effects (including pain and fever) of cancer chemotherapeutic agents. Dipyrone is metabolized to 4-aminoantipyrine (4-AA), a PGE2-dependent blocker and inhibitor of cyclooxygenase (COX). We evaluated the genotoxic, mutagenic, apoptotic, and immunomodulatory activities of 4-AA in vivo and the effects of its combination with the antineoplastic drugs doxorubicin, cisplatin, and cyclophosphamide. 4-AA did not cause genotoxic/mutagenic damage, splenic phagocytosis, or leukocyte alterations. However, when combined with the antineoplastic agents, 4-AA decreased their genotoxic, mutagenic, apoptotic, and phagocytic effects. These results suggest that 4-AA might interfere with DNA damage-mediated chemotherapy.


Asunto(s)
Ampirona/farmacología , Cisplatino/toxicidad , Ciclofosfamida/toxicidad , Doxorrubicina/toxicidad , Animales , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Ensayo Cometa , Daño del ADN/efectos de los fármacos , Masculino , Ratones , Pruebas de Micronúcleos , Fagocitosis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...