Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Naunyn Schmiedebergs Arch Pharmacol ; 397(1): 507-520, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37477660

RESUMO

Epirubicin (EPI) is an effective chemotherapeutic against breast cancer, though EPI-related cardiotoxicity limits its usage. Endogenously derived 3-indolepropionic acid (3-IPA) from tryptophan metabolism is of interest due to its antioxidant capabilities which may have cardioprotective effects. Supplementation with 3-IPA may abate EPI's cardiotoxicity, and herein we studied the possibility of lessening EPI-induced cardiotoxicity in Wistar rats. Experimental rats (n = 30; BW 180-200 g) were randomly distributed in five cohorts (A-E; n = 6 each). Group A (control), Group B (EPI 2.5 mg/mL), and group C (3-IPA 40 mg/kg) while Groups D and E were co-treated with EPI (2.5 mg/mL) together with 3-IPA (D: 20 and E: 40 mg/kg). Following sacrifice, oxidative status, lipid profile, transaminases relevant to cardiac function, and inflammatory biomarkers were analysed. Also, 8-hydroxyl-2'-deoxyguanosine (8-OHdG) and cardiac troponin T (cTnT) levels were assessed using an enzyme-linked immunosorbent assay (ELISA). EPI-initiated increases in cardiotoxicity biomarkers were significantly (p < 0.05) reduced by 3-IPA supplementation. Decreased antioxidant and increases in reactive oxygen and nitrogen species (RONS), 8-OHdG and lipid peroxidation were lessened (p < 0.05) in rat hearts co-treated with 3-IPA. EPI-induced increases in nitric oxide and myeloperoxidase were reduced (p < 0.05) by 3-IPA co-treatment. In addition, 3-IPA reversed EPI-mediated alterations in alanine aminotransferase (ALT), aspartate amino transaminases (AST), lactate dehydrogenase (LDH), cardiac troponin T (cTnT), and serum lipid profile including total cholesterol and triglycerides. Microscopic examination of the cardiac tissues showed that histopathological lesions severity induced by EPI was lesser in 3-IPA co-treated rats. Our findings demonstrate that supplementing endogenously derived 3-IPA can enhance antioxidant protection in the cardiac tissue susceptible to EPI toxicity in female rats. These findings may benefit breast cancer patients undergoing chemotherapy by further validating these experimental data.


Assuntos
Neoplasias da Mama , Cardiotoxicidade , Humanos , Ratos , Feminino , Animais , Epirubicina/toxicidade , Epirubicina/metabolismo , Cardiotoxicidade/tratamento farmacológico , Miócitos Cardíacos/metabolismo , Ratos Wistar , Antioxidantes/uso terapêutico , Troponina T , Propionatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias da Mama/metabolismo , Biomarcadores/metabolismo , Estresse Oxidativo
2.
Chem Biol Interact ; 374: 110414, 2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-36822302

RESUMO

The "anthracycline, Epirubicin (EPI)," in managing breast cancer, is highly cytotoxic. Tryptophan-derived 3-indolepropionic acid (3-IPA) decreases oxidative damage, and its prospect of alleviating EPI-induced cytotoxicity was examined in rats' hypothalamus-ovary-uterus axis. Female rats: Control, EPI (2.5 mg/kg), 3-IPA alone (40 mg/kg), EPI+3-IPA (2.5 mg/kg + 20 mg/kg), EPI + 3-IPA2 (2.5 mg/kg + 40 mg/kg) were treated for 28 days. Subsequently, reproductive hormones, oxidative and inflammatory stress biomarkers, and tissue histology were examined. 3-IPA prevented EPI-induced decreases in the follicle-stimulating hormone, estradiol, progesterone and prolactin levels. EPI-mediated reduction in antioxidant enzymes, reduced glutathione and total sulfhydryl groups were partially counteracted by 3-IPA co-treatment. Increased oxidative and inflammatory stress biomarkers caused by treatment with EPI alone were lessened by 3-IPA co-treatment. Also, 3-IPA reduced histological damage in the examined tissues. Conclusively, 3-IPA ameliorated biochemical markers and tissue injury caused by EPI treatment alone via an antioxidative and anti-inflammatory mechanism while stabilising serum hormone dynamics.


Assuntos
Antioxidantes , Ovário , Feminino , Ratos , Animais , Ovário/patologia , Epirubicina/toxicidade , Epirubicina/metabolismo , Ratos Wistar , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Estresse Oxidativo , Hormônio Foliculoestimulante , Suplementos Nutricionais , Útero/metabolismo , Biomarcadores/metabolismo
3.
Front Biosci (Landmark Ed) ; 27(6): 173, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35748249

RESUMO

BACKGROUND: Epirubicin (EPI) is an important anticancer drug that is well-known for its cardiotoxic side effect. Studying epigenetic modification such as DNA methylation can help to understand the EPI-related toxic mechanisms in cardiac tissue. In this study, we analyzed the DNA methylation profile in a relevant human cell model and inspected the expression of differentially methylated genes at the transcriptome level to understand how changes in DNA methylation could affect gene expression in relation to EPI-induced cardiotoxicity. METHODS: Human cardiac microtissues were exposed to either therapeutic or toxic (IC20) EPI doses during 2 weeks. The DNA and RNA were collected from microtissues in triplicates at 2, 8, 24, 72, 168, 240, and 336 hours of exposure. Methylated DNA immunoprecipitation-sequencing (MeDIP-seq) analysis was used to detect DNA methylation levels in EPI-treated and control samples. The MeDIP-seq data were analyzed and processed using the QSEA package with a recently published workflow. RNA sequencing (RNA-seq) was used to measure global gene expression in the same samples. RESULTS: After processing the MeDIP-seq data, we detected 35, 37, 15 candidate genes which show strong methylated alterations between all EPI-treated, EPI therapeutic and EPI toxic dose-treated samples compared to control, respectively. For several genes, gene expressions changed compatibly reflecting the DNA methylation regulation. CONCLUSIONS: The observed DNA methylation modifications provide further insights into the EPI-induced cardiotoxicity. Multiple differentially methylated genes under EPI treatment, such as SMARCA4, PKN1, RGS12, DPP9, NCOR2, SDHA, POLR2A, and AGPAT3, have been implicated in different cardiac dysfunction mechanisms. Together with other differentially methylated genes, these genes can be candidates for further investigations of EPI-related toxic mechanisms. Data Repository: The data has been generated by the HeCaToS project (http://www.ebi.ac.uk/biostudies) under accession numbers S-HECA433 and S-HECA434 for the MeDIP-seq data and S-HECA11 for the RNA-seq data. The R code is available on Github (https://github.com/NhanNguyen000/MeDIP).


Assuntos
Cardiotoxicidade , Metilação de DNA , Cardiotoxicidade/genética , DNA , DNA Helicases , Epirubicina/toxicidade , Humanos , Proteínas Nucleares , Análise de Sequência de DNA , Fatores de Transcrição
4.
Toxicol In Vitro ; 68: 104964, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32800948

RESUMO

Epirubicin is a cytotoxic drug used in the treatment of different types of cancer and increasing evidence suggests that its target is cell membranes. In order to gain insight on its toxic effects, intact red blood cells (RBC), human erythrocyte membranes and molecular models were used. The latter consisted in bilayers of dimyristoylphosphatidylcholine (DMPC) and dimyristoylphosphatidylethanolamine (DMPE), phospholipid classes found mainly in the outer and inner monolayers of the human erythrocyte membrane, respectively. The results obtained by X-ray diffraction displayed that epirubicin induced structural perturbations in multilayers of DMPC. Differential scanning calorimetry (DSC) showed that epirubicin disturbed the thermotropic behavior of both DMPC and DMPE vesicles, whereas fluorescence spectroscopy demonstrated alterations in the fluidity of DMPC vesicles and the erythrocyte membrane. Scanning electron microscopy (SEM) revealed that epirubicin changed the normal discoid form of RBC to echinocytes and stomatocytes. Electron paramagnetic resonance (EPR) disclosed that this drug induced conformational changes in the erythrocyte membrane proteins. These findings demonstrate that epirubicin interacts with lipids and proteins of the human erythrocyte membrane, effects that might compromise the integrity and function of cell membranes. This is the first time that its toxic effects on the human erythrocyte membrane have been described.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Epirubicina/toxicidade , Eritrócitos/efeitos dos fármacos , Varredura Diferencial de Calorimetria , Células Cultivadas , Dimiristoilfosfatidilcolina , Eritrócitos/patologia , Eritrócitos/ultraestrutura , Humanos , Lipossomos , Microscopia Eletrônica de Varredura , Fosfatidiletanolaminas , Difração de Raios X
5.
Cardiovasc Res ; 116(5): 970-982, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31346605

RESUMO

AIMS: The clinical application of doxorubicin (DOX) is severely compromised by its cardiotoxic effects, which limit the therapeutic index and the cumulative dose. Liposomal encapsulation of DOX (Myocet®) provides a certain protective effect against cardiotoxicity by reducing myocardial drug accumulation. We aimed to evaluate transcriptomic responses to anthracyclines with different cardiotoxicity profiles in a translational large animal model for identifying potential alleviation strategies. METHODS AND RESULTS: We treated domestic pigs with either DOX, epirubicin (EPI), or liposomal DOX and compared the cardiac, laboratory, and haemodynamic effects with saline-treated animals. Cardiotoxicity was encountered in all groups, reflected by an increase of plasma markers N-terminal pro-brain-natriuretic peptide and Troponin I and an impact on body weight. High morbidity of EPI-treated animals impeded further evaluation. Cardiac magnetic resonance imaging with gadolinium late enhancement and transthoracic echocardiography showed stronger reduction of the left and right ventricular systolic function and stronger myocardial fibrosis in DOX-treated animals than in those treated with the liposomal formulation. Gene expression profiles of the left and right ventricles were analysed by RNA-sequencing and validated by qPCR. Interferon-stimulated genes (ISGs), linked to DNA damage repair and cell survival, were downregulated by DOX, but upregulated by liposomal DOX in both the left and right ventricle. The expression of cardioprotective translocator protein (TSPO) was inhibited by DOX, but not its liposomal formulation. Cardiac fibrosis with activation of collagen was found in all treatment groups. CONCLUSIONS: All anthracycline-derivatives resulted in transcriptional activation of collagen synthesis and processing. Liposomal packaging of DOX-induced ISGs in association with lower cardiotoxicity, which is of high clinical importance in anticancer treatment. Our study identified potential mechanisms for rational development of strategies to mitigate anthracycline-induced cardiomyopathy.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/prevenção & controle , Dano ao DNA , Doxorrubicina/análogos & derivados , Fatores Reguladores de Interferon/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacocinética , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Cardiotoxicidade , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Relação Dose-Resposta a Droga , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidade , Composição de Medicamentos , Epirubicina/toxicidade , Feminino , Fibrose , Humanos , Fatores Reguladores de Interferon/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Polietilenoglicóis/farmacocinética , Polietilenoglicóis/toxicidade , Sus scrofa , Transcriptoma/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Direita/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
6.
J Reprod Dev ; 66(1): 19-27, 2020 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-31735743

RESUMO

One major side effect of chemotherapy that young women with cancer suffer from is ovarian damage. Therefore, it is necessary to study the pathogenesis of chemotherapeutic drugs in order to develop pharmaceutical agents to preserve fertility. Epirubicin is one of the commonly used chemotherapy drugs for breast cancer patients. This research explored the side effects of epirubicin in mice. We found that epirubicin significantly reduced the body weight, the weight of the ovaries and uteri, and the pups' number, while melatonin, which is extremely resistant to oxidation, significantly reduced these damages. Moreover, co-treatment with melatonin prevented epirubicin-induced decrease in E2 and progesterone, and the loss of follicles. Mechanism study showed that melatonin significantly reduced the levels of proapoptotic genes p53, Caspase3, and Caspase9 while it upregulated antiapoptotic factors Bcl-2 and Bcl2l1, and antioxidant genes superoxide dismutase 1 and catalase compared with the epirubicin group. In addition, melatonin markedly reduced reactive oxygen species (ROS) and the transcription of Caspase12 and Chop, which is vital in endoplasmic reticulum stress (ERS)-mediated apoptosis. These results indicate melatonin protects against epirubicin-induced ovarian damage by reducing ROS-induced ERS. Therefore, melatonin has a therapeutic potential for the protection of ovarian function and preservation of fertility during chemotherapy.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Epirubicina/toxicidade , Melatonina/farmacologia , Ovário/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Peso Corporal/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estradiol/sangue , Feminino , Camundongos , Camundongos Endogâmicos ICR , Ovário/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Progesterona/sangue , Espécies Reativas de Oxigênio/metabolismo , Útero/efeitos dos fármacos , Útero/metabolismo
7.
Aging (Albany NY) ; 11(22): 10532-10556, 2019 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-31711044

RESUMO

In vitro culture models were used to characterize the effects of chemotherapeutic drugs and of LH on somatic cells from prepuberal mouse ovaries. All cell types (pre- and granulosa cells, pre-thecal and OSE cells) underwent apoptosis following Epirubicin (0.5µM) exposure for 24hrs (about 60%) and 48hrs (>80%). Cisplatin (10µM) and the Cyclophosphamide active metabolite, Phosphoramide Mustard (10µM), didn't cause apoptosis in 90% of pre-thecal and pre-granulosa cells up to 72hrs of exposure, although they suffered extensive DNA damage and cell cycle arrest, and acquired stress induced premature senescence (SIPS) features. Cultured granulosa cells didn't show evident DNA damage and remained viable without acquiring SIPS features; OSE cells were resistant to apoptosis and SIPS but not to DNA damage. These latter, like pre-thecal and pre-granulosa cells, were able of efficient DNA repair involving MLH1-dependent MMR pathways. SIPS features were also observed in ovary after in vivo treatment with Cisplatin. LH (200mIU/mL) didn't significantly influence apoptosis, SIPS and DNA damage but favoured DNA repair. These results show that somatic cells of prepuberal ovary response to drugs in different ways, either undergoing apoptosis or SIPS, either showing resistance to Cisplatin and Phosphoramide Mustard. Moreover, a new role of LH in promoting DNA repair was shown.


Assuntos
Antineoplásicos/toxicidade , Cisplatino/toxicidade , Ciclofosfamida/toxicidade , Epirubicina/toxicidade , Células da Granulosa/efeitos dos fármacos , Células Tecais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Feminino , Camundongos
8.
Ecotoxicol Environ Saf ; 165: 174-181, 2018 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-30195999

RESUMO

INTRODUCTION: Cytotoxic drug exposure of hospital staff preparing intravenous chemotherapy is a major issue and related mutagenic risks should be more explored. The aim of this study was to assess the mutagenicity of several cytotoxic mixtures prepared at fixed concentrations, and the mutagenicity of environmental samples collected in a hospital centralized reconstitution unit. In parallel cytotoxic exposure in environmental samples was quantified. METHODS: Environmental samples were performed by wiping method using swabs in five critical production unit areas. Mutagenicity was assessed with a liquid microplate AMES test using two salmonella typhimurium strains (TA98 and TA100), in prepared cytotoxic mixtures containing 14 cytotoxic drugs (cyclophosphamide, cytarabine, dacarbazine, docetaxel, doxorubicin, epirubicin, etoposide, 5-fluorouracil, gemcitabine, ifosfamide, irinotecan, methotrexate, paclitaxel and pemetrexed) according a dichotomous strategy and in environmental samples. Cytotoxic drugs were quantified in samples using liquid chromatography coupled to mass tandem spectrometry. RESULTS: Mutagenesis was observed for the mix of 14 cytotoxic drugs with TA98 strain ±â€¯S9 fraction but not TA100 strain. After dichotomous approach, only doxorubicin and epirubicin exposure were associated to mutagenesis. The mutagenesis observed was expressed at lower concentrations with the mix of the 14 drugs than with anthracyclins alone, assuming a synergistic effect. Despite measurable level of cytotoxic contamination in environmental samples, no mutagenesis was highlighted in Ames tests performed on these environmental samples. CONCLUSIONS: The analyses carried out show the conservation of the mutagenicity of cytotoxic drugs found in very low quantities in the environment. The traces of cytotoxic drugs found in our unit regularly exceed the limits given by some authors. This approach may be considered as a new tool to monitor environmental contamination by cytotoxic drugs.


Assuntos
Antraciclinas/toxicidade , Antineoplásicos/toxicidade , Contaminação de Equipamentos , Hospitais , Testes de Mutagenicidade , Cromatografia Líquida , Ciclofosfamida/toxicidade , Doxorrubicina/toxicidade , Monitoramento Ambiental , Poluição Ambiental , Epirubicina/toxicidade , Etoposídeo/toxicidade , Irinotecano/toxicidade , Salmonella typhimurium/efeitos dos fármacos , Espectrometria de Massas em Tandem
9.
Cell Death Differ ; 24(4): 731-746, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28211872

RESUMO

How metabolic pathways required for epidermal tissue growth and remodeling influence the ability of keratinocytes to survive stressful conditions is still largely unknown. The mechanistic target of rapamycin complex 2 (mTORC2) regulates growth and metabolism of several tissues, but its functions in epidermal cells are poorly defined. Rictor is an adaptor protein essential for mTORC2 activity. To explore the roles of mTORC2 in the epidermis, we have conditionally deleted rictor in mice via K14-Cre-mediated homologous recombination and found that its deficiency causes moderate tissue hypoplasia, reduced keratinocyte proliferation and attenuated hyperplastic response to TPA. Noteworthy, rictor-deficient keratinocytes displayed increased lifespan, protection from senescence, and enhanced tolerance to cellular stressors such as growth factors deprivation, epirubicin and X-ray in vitro and radioresistance in vivo. Rictor-deficient keratinocytes exhibited changes in global gene expression profiles consistent with metabolic alterations and enhanced stress tolerance, a shift in cell catabolic processes from glycids and lipids to glutamine consumption and increased production of mitochondrial reactive oxygen species (ROS). Mechanistically, the resiliency of rictor-deficient epidermal cells relies on these ROS increases, indicating stress resistance via mitohormesis. Thus, our findings reveal a new link between metabolic changes and stress adaptation of keratinocytes centered on mTORC2 activity, with potential implications in skin aging and therapeutic resistance of epithelial tumors.


Assuntos
Mitocôndrias/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Acetilcisteína/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Senescência Celular/efeitos da radiação , Epirubicina/toxicidade , Ácido Glutâmico/metabolismo , Hiperplasia , Queratina-14/genética , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Tolerância a Radiação/efeitos da radiação , Proteína Companheira de mTOR Insensível à Rapamicina/antagonistas & inibidores , Proteína Companheira de mTOR Insensível à Rapamicina/deficiência , Espécies Reativas de Oxigênio/metabolismo , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Transcriptoma/efeitos dos fármacos , Transcriptoma/efeitos da radiação , Raios X
10.
Eur J Pharmacol ; 795: 84-93, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27940053

RESUMO

Renal injury is a dose-dependent side effect of epirubicin that limits its clinical application in the field of tumor chemotherapy. Paeonol is an active ingredient with a variety of biological activities, including the prevention of multiple antineoplastic-induced toxicities. In the present study, we assessed the renoprotective effect of paeonol on epirubicin-induced nephrotoxicity and investigated the underlying mechanism. Renal function, kidney histology, oxidative stress, nitrative stress, inflammation, apoptotic proteins and the effects on signaling pathways were investigated. Paeonol lowered the levels of biomarkers of renal injury, relieved histopathological alterations, alleviated oxidative stress and nitrative stress, and ameliorated inflammation. Moreover, paeonol inhibited epirubicin-induced apoptosis by suppressing the activation of caspase-9 and caspase-3, the Bax/Bcl-2 imbalance and cytochrome c release. Further studies suggest that paeonol up-regulates the Nrf2/HO-1 pathway by increasing the expression of Nrf2 and HO-1 and down-regulates the NF-κB pathway by reducing IκBα degradation and blocking the p-NF-κB nuclear translocation. In conclusion, paeonol alleviates epirubicin-induced renal injury in mice by regulating the Nrf2 and NF-κB signaling pathways.


Assuntos
Acetofenonas/farmacologia , Citoproteção/efeitos dos fármacos , Epirubicina/toxicidade , Rim/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/citologia , Rim/lesões , Rim/metabolismo , Camundongos
11.
Mutagenesis ; 31(2): 137-45, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26399706

RESUMO

Dexrazoxane is the only clinically approved cardioprotectant against anthracyclines-induced cardiotoxicity. Thus, detailed evaluation of the genotoxic potential of dexrazoxane and anthracyclines combination is essential to provide more insights into genotoxic and anti-genotoxic alterations that may play a role in the development of the secondary malignancies after treatment with anthracyclines. Thus, our aim was to determine whether non-genotoxic doses of dexrazoxane in combination with the anthracycline, epirubicin can modulate epirubicin-induced genotoxicity and apoptosis in somatic cells. Bone marrow micronucleus test complemented with fluorescence in situ hybridization assay and comet assay were performed to assess the genotoxicity of dexrazoxane and/or epirubicin. Apoptosis was analysed by using the annexin V assay and the occurrence of the hypodiploid DNA content. Generation of reactive oxygen species was also assessed in bone marrow by using the oxidant-sensing fluorescent probe 2',7'-dichlorodihydrofluorescein diacetate. Dexrazoxane was neither genotoxic nor apoptogenic in mice at a single dose of 75 or 150mg/kg. Moreover, it has been shown that dexrazoxane affords significant protection against epirubicin-induced genotoxicity and apoptosis in the bone marrow cells in a dose-dependent manner. Epirubicin induced marked generation of intracellular reactive oxygen species and prior administration of dexrazoxane ahead of epirubicin challenge ameliorated accumulation of these free radicals. It is thus concluded that dexrazoxane can be safely combined with epirubicin and that pre-treatment with dexrazoxane attenuates epirubicin-induced generation of reactive oxygen species and subsequent genotoxicity and apoptosis. Thus, epirubicin-induced genotoxicity can be effectively mitigated by using dexrazoxane.


Assuntos
Antineoplásicos/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Cardiotônicos/farmacologia , Dexrazoxano/farmacologia , Epirubicina/toxicidade , Testes de Mutagenicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinógenos/toxicidade , Ensaio Cometa , Quebras de DNA/efeitos dos fármacos , Masculino , Camundongos , Micronúcleos com Defeito Cromossômico , Testes para Micronúcleos , Testes de Mutagenicidade/métodos , Espécies Reativas de Oxigênio/metabolismo
12.
J Cardiovasc Pharmacol ; 66(4): 371-5, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26080056

RESUMO

OBJECTIVE: To investigate the clinical value of heart rate deceleration capacity (DC) in predicting the risk of epirubicin-induced cardiotoxicity. METHODS: The CK-MB and cTnI levels and DC values of 86 patients were examined before chemotherapy and again after 2 and 4 cycles of chemotherapy. Patients were divided into low-risk group (LRG) (40 cases), medium-risk group (26 cases), and high-risk group (HRG) (20 cases) based on the calculated DC values. RESULTS: After 4 cycles of chemotherapy, HRG showed a significantly greater increase in serum CK-MB (17.1 ± 4.9 vs. 14.6 ± 3.7) and cTnI (1.28 ± 0.38 vs. 1.0 ± 0.29) concentrations over the prechemotherapy levels when compared with LRG. After 2 and 4 cycles of chemotherapy, HRG exhibited a significantly greater increase in mean heart rate (2 cycles: 79.6 ± 6.0 vs. 77.6 ± 6.7; 4 cycles: 88.2 ± 10.2 vs. 82.4 ± 6.2) and the supraventricular (2 cycles: 68.9 ± 19.3 vs. 57.2 ± 17.6; 4 cycles: 131.1 ± 29.5 vs. 91.7 ± 16.5) and ventricular arrhythmia counts (2 cycles: 179.0 ± 20.5 vs. 162.3 ± 16.3; 4 cycles: 228.6 ± 44.8 vs. 187.4 ± 22.6) over the prechemotherapy values compared with LRG. When the supraventricular and ventricular arrhythmia counts measured after 4 cycles of chemotherapy were compared with those obtained before chemotherapy, HRG (131.1 ± 29.5 and 228.6 ± 44.8, respectively) showed the largest differences, followed by medium-risk group (107.4 ± 31.9 and 202.0 ± 29.8, respectively) and then LRG (91.7 ± 16.5 and 187.4 ± 22.6, respectively) (P < 0.01). After 4 cycles of chemotherapy, the incidence rates of ventricular arrhythmia greater than Lown's grade 3 (30% vs. 2.5%), QTc (20% vs. 0) elongation, and ST-T (40% vs. 5%) changes in HRG were significantly higher than those observed in LRG (P < 0.05). CONCLUSIONS: DC test was shown to be an effective predictor of the risk of epirubicin-induced cardiotoxicity.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/diagnóstico , Epirubicina/toxicidade , Frequência Cardíaca/efeitos dos fármacos , Adulto , Idoso , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/uso terapêutico , Cardiotoxicidade , Creatina Quinase Forma MB/sangue , Desaceleração , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Eletrocardiografia , Epirubicina/administração & dosagem , Epirubicina/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Troponina I/sangue , Adulto Jovem
13.
Drug Chem Toxicol ; 38(4): 442-51, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25609004

RESUMO

Mucositis is a serious disorder of the gastrointestinal tract that results from cancer chemotherapy. We investigated the protective effects of silymarin on epirubicin-induced mucosal barrier injury in CD-1 mice. Immunohistochemical activity of both pro-apoptotic Bax and anti-apoptotic Bcl-2 markers, together with p53, cyt-P450 expression and DNA damage analysis on stomach, small intestine and colon were evaluated. Our results indicated stronger expression for cyt P450 in all analyzed gastrointestinal tissues of Epi group, which demonstrate intense drug detoxification. Bax immunopositivity was intense in the absorptive enterocytes and lamina connective cells of the small intestine, surface epithelial cells of the stomach and also in the colonic epithelium and lamina concomitant with a decreased Bcl-2 expression in all analyzed tissues. Epirubicin-induced gastrointestinal damage was verified by a goblet cell count and morphology analysis on histopathological sections stained for mucins. In all analyzed tissues, Bax immunopositivity has been withdrawn by highest dose of silymarin concomitant with reversal of Bcl-2 intensity at a level comparable with control. p53 expression was found in all analyzed tissues and decreased by high dose of silymarin. Also, DNA internucleosomal fragmentation was observed in the Epi groups for all analyzed tissues was almost suppressed at 100 mg/kg Sy co-treatment. Histological aspect and goblet cell count were restored at a highest dose of Sy for both small and large intestine. In conclusion, our findings suggest that silymarin may prevent cellular damage of epirubicin-induced toxicity and was effective in reducing the severity indicators of gastrointestinal mucositis in mice.


Assuntos
Apoptose/efeitos dos fármacos , Epirubicina/toxicidade , Mucosite/prevenção & controle , Silimarina/farmacologia , Animais , Antibióticos Antineoplásicos/toxicidade , Sistema Enzimático do Citocromo P-450/metabolismo , Dano ao DNA/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/patologia , Gastroenteropatias/prevenção & controle , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Camundongos , Mucosite/induzido quimicamente , Mucosite/patologia , Substâncias Protetoras/administração & dosagem , Substâncias Protetoras/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Índice de Gravidade de Doença , Silimarina/administração & dosagem , Proteína X Associada a bcl-2/metabolismo
14.
Chemistry ; 21(6): 2680-5, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25504676

RESUMO

A pH-responsive free-blockage release system was achieved through controlling the hydrophobic/hydrophilic conversion of mesoporous silica nanopores. This system further presented pulsatile release with changing pH values between 4.0 and 7.0 for several cycles. This free-blockage release system could also release antitumor agents to induce cell death after infecting tumor cells and could have the ability of continuous infection to tumor cells with high drug-delivery efficiency and few side effects.


Assuntos
Portadores de Fármacos/química , Nanoporos , Dióxido de Silício/química , Antineoplásicos/química , Antineoplásicos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada , Epirubicina/química , Epirubicina/toxicidade , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Nanopartículas/química , Porosidade
15.
PLoS One ; 8(11): e82478, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24303084

RESUMO

Multidrug resistance (MDR), an unfavorable factor compromising the treatment efficacy of anticancer drugs, involves the upregulation of ATP binding cassette (ABC) transporters and induction of galectin-3 signaling. Galectin-3 plays an anti-apoptotic role in many cancer cells and regulates various pathways to activate MDR. Thus, the inhibition of galectin-3 has the potential to enhance the efficacy of the anticancer drug epirubicin. In this study, we examined the effects and mechanisms of silencing galectin-3 via RNA interference (RNAi) on the ß-catenin/GSK-3ß pathway in human colon adenocarcinoma Caco-2 cells. Galectin-3 knockdown increased the intracellular accumulation of epirubicin in Caco-2 cells; suppressed the mRNA expression of galectin-3, ß-catenin, cyclin D1, c-myc, P-glycoprotein (P-gp), MDR-associated protein (MRP) 1, and MRP2; and downregulated the protein expression of P-gp, cyclin D1, galectin-3, ß-catenin, c-Myc, and Bcl-2. Moreover, galectin-3 RNAi treatment significantly increased the mRNA level of GSK-3ß, Bax, caspase-3, and caspase-9; remarkably increased the Bax-to-Bcl-2 ratio; and upregulated the GSK-3ß and Bax protein expressions. Apoptosis was induced by galectin-3 RNAi and/or epirubicin as demonstrated by chromatin condensation, a higher sub-G1 phase proportion, and increased caspase-3 and caspase-9 activity, indicating an intrinsic/mitochondrial apoptosis pathway. Epirubicin-mediated resistance was effectively inhibited via galectin-3 RNAi treatment. However, these phenomena could be rescued after galectin-3 overexpression. We show for the first time that the silencing of galectin-3 sensitizes MDR cells to epirubicin by inhibiting ABC transporters and activating the mitochondrial pathway of apoptosis through modulation of the ß-catenin/GSK-3ß pathway in human colon cancer cells.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Galectina 3/genética , Inativação Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Células CACO-2 , Caspases/genética , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Epirubicina/farmacologia , Epirubicina/toxicidade , Galectina 3/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo , beta Catenina/genética
16.
Toxicol Appl Pharmacol ; 269(2): 121-31, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23531514

RESUMO

Widely used chemotherapeutic breast cancer drugs such as Tamoxifen citrate (TC), Capecitabine (CP) and Epirubicin (EP) are known to cause various cardiovascular side-effects among long term cancer survivors. Vascular modulation warrants nitric oxide (NO) signal transduction, which targets the vascular endothelium. We hypothesize that TC, CP and EP interference with the nitric oxide downstream signaling specifically, could lead to cardiovascular dysfunctions. The results demonstrate that while all three drugs attenuate NO and cyclic guanosine mono-phosphate (cGMP) production in endothelial cells, they caused elevated levels of NO in the plasma and RBC. However, PBMC and platelets did not show any significant changes under treatment. This implies that the drug effects are specific to the endothelium. Altered eNOS and phosphorylated eNOS (Ser-1177) localization patterns in endothelial cells were observed following drug treatments. Similarly, the expression of phosphorylated eNOS (Ser-1177) protein was decreased under the treatment of drugs. Altered actin polymerization was also observed following drug treatment, while addition of SpNO and 8Br-cGMP reversed this effect. Incubation with the drugs decreased endothelial cell migration whereas addition of YC-1, SC and 8Br-cGMP recovered the effect. Additionally molecular docking studies showed that all three drugs exhibited a strong binding affinity with the catalytic domain of human sGC. In conclusion, results indicate that TC, CP and EP cause endothelial dysfunctions via the NO-sGC-cGMP pathway and these effects could be recovered using pharmaceutical agonists of NO signaling pathway. Further, the study proposes a combination therapy of chemotherapeutic drugs and cGMP analogs, which would confer protection against chemotherapy mediated vascular dysfunctions in cancer patients.


Assuntos
Desoxicitidina/análogos & derivados , Células Endoteliais/efeitos dos fármacos , Epirubicina/toxicidade , Fluoruracila/análogos & derivados , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/toxicidade , Animais , Antineoplásicos/toxicidade , Capecitabina , Bovinos , Células Cultivadas , Desoxicitidina/toxicidade , Relação Dose-Resposta a Droga , Fluoruracila/toxicidade , Regulação Enzimológica da Expressão Gênica , Humanos , Modelos Moleculares , Estrutura Molecular , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Conformação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais/fisiologia
17.
Ann Cardiol Angeiol (Paris) ; 62(1): 28-33, 2013 Feb.
Artigo em Francês | MEDLINE | ID: mdl-22513244

RESUMO

OBJECTIVE: Evaluate the early anthracyclines cardiotoxicity. METHODS: It is a prospective study made on 10 months of period from October 2008 to July 2009 and on patients who contracted a solid canny tumor hospitalized or followed in their movement and who would receive chemotherapy with an anthracycline molecule. On this effect, we have used tissue Doppler of mitral ring to detect clinic infratoxicity. RESULT: Forty-five patients (43 women and 2 men) who contracted the solid cancers were included in the study. The patients were 48 of age in average ± 10.12. All our patients did not show any cardiovascular symptoms at the time of the study. Cardiothoracic and electrocardiograms were not significantly modified by the chemotherapy. The cardioecography with the use of tissue Doppler revealed as followed: (a) significant low of the ejection fraction and the pick of systolic myocardia wave (Sa) on four patients (8.8%). These concerned patients were considered as having anthracycline cardio toxicity. The factor causing this cardiotoxicity was the nature of the anthracycline, which was used: the doxorubicin. The quantity accumulated threshold of the doxorubicin that shod (where toxicity appeared was 150 mg/m(2)); (b) a low of Sa pick without that of left ventricular fraction ejection observed on five patients (11.11%). These concerned patients were considered as having potential risks to develop anthracyclines cardiotoxicity; (c) the left ventricular ejection fraction was not a good indicator the check up of the patients under chemotherapy made up with anthracyclines. CONCLUSION: The tissue Doppler not only enables to make diagnostics of early myocardia dysfunctions but it mainly allows to identify people with risks of a cardiotoxicity due to a going on chemotherapy.


Assuntos
Antraciclinas/toxicidade , Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/diagnóstico por imagem , Cardiotoxinas/toxicidade , Países em Desenvolvimento , Ecocardiografia Doppler/métodos , Interpretação de Imagem Assistida por Computador/métodos , Neoplasias/tratamento farmacológico , Adulto , Idoso , Antraciclinas/uso terapêutico , Antineoplásicos/uso terapêutico , Cardiotoxinas/uso terapêutico , Relação Dose-Resposta a Droga , Doxorrubicina/uso terapêutico , Doxorrubicina/toxicidade , Epirubicina/uso terapêutico , Epirubicina/toxicidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valva Mitral/diagnóstico por imagem , Valva Mitral/efeitos dos fármacos , Volume Sistólico/efeitos dos fármacos , Sístole/efeitos dos fármacos , Togo , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/diagnóstico por imagem
18.
Folia Med (Plovdiv) ; 54(3): 42-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23270206

RESUMO

AIM: To evaluate the effect of esculin, a plant coumarin glucoside, on free radicals and against epirubicin-induced toxicity on bone marrow cells. MATERIALS AND METHODS: Antioxidant activity was assessed by a luminol-dependent chemiluminescence method or NBT test in a xanthine-xanthine oxidase system, and two iron-dependent lipid peroxidation systems. In vivo experiments were carried out in epirubicin-treated mice, alone or in a combination with esculin. Genotoxicity of the anthracycline drug was assessed by cytogenetic analysis and an autoradiographic assay. RESULTS: Esculin inactivated superoxide anion radicals in both systems we used. It exerted SOD-mimetic effect and reduced the level of superoxide radicals generated in a xanthine-xanthine oxidase system by 30%. Esculin also showed an antioxidant effect in a model of Fe2+-induced lipid peroxidation. Cytogenetic analysis showed that epirubicin had a marked influence on the structure of metaphase chromosomes of normal bone marrow cells. Inclusion of esculin in the treatment protocol failed to ameliorate the epirubicin-induced antiproliferative effects and genotoxicity in bone marrow cells. CONCLUSION: In this study the ability of the coumarin glucoside esculin to scavenge superoxide radicals and to decrease Fe-induced lipid peroxidation was documented. However, despite the registered antioxidant effects the tested compound failed to exert cytoprotection in models of anthracycline-induced genotoxicity in bone marrow cells. The results of this study warrant for more precise further evaluation of esculin, employing different test systems and end-points and a wider range of doses to more precisely appraise its potential role as a chemoprotective/resque agent.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Antioxidantes/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Epirubicina/toxicidade , Esculina/farmacologia , Radicais Livres/antagonistas & inibidores , Animais , Masculino , Camundongos , Testes de Mutagenicidade , Superóxidos/metabolismo
20.
Mol Med Rep ; 6(5): 1133-9, 2012 11.
Artigo em Inglês | MEDLINE | ID: mdl-22895628

RESUMO

Clusterin (CLU) is a glycoprotein that is over-expressed in a number of malignant tumors and has been proven to correlate closely with the chemoresistance of several cancer cells to chemotherapeutic agents. However, the effect of CLU expression on the chemoresistance of bladder cancer to epirubicin remains unknown. In the present study, we aimed to elucidate the role of CLU in the chemoresistance of bladder cancer cells to epirubicin. Lentivirus-mediated RNA interference was applied to knock down CLU in EJ bladder cancer cells. The efficiency was examined by RT-PCR and western blot analysis. After stable CLU silencing, an EJ cell line was established and cells were treated with or without epirubicin. Cell viability, migration, invasiveness, clone formation and cell cycle progression were assessed by MTT assay, wound healing assay, Matrigel invasion assay, plate clone formation assay and flow cytometry, respectively. The results indicated that lentivirus-mediated RNA interference effectively silenced CLU at the RNA and protein levels. CLU knockdown increased the cytotoxicity of epirubicin to EJ bladder cancer cells. Combined treatment with lentivirus-mediated shRNA targeting CLU and epirubicin had maximum effects in bladder cancer cells on cell viability, migration, invasiveness and clone-forming ability. Furthermore, cell cycle analysis indicated that CLU knockdown reinforced the efficacy of epirubicin on G0/G1 cell cycle arrest. Taken together, our results suggest that CLU silencing enhances chemosensitivity of EJ bladder cancer cells to epirubicin. Lentivirus-mediated shRNA targeting CLU may be an alternative approach in the treatment of bladder cancer.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Clusterina/antagonistas & inibidores , Epirubicina/toxicidade , Lentivirus/genética , Interferência de RNA/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Clusterina/genética , Clusterina/metabolismo , Resistencia a Medicamentos Antineoplásicos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , RNA Interferente Pequeno/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...