Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Toxicol Appl Pharmacol ; 435: 115852, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34973290

RESUMEN

Imatinib Mesylate (IMA) has been widely used to treat with chronic myeloid leukemia (CML). However, cardiotoxicity associated with IMA is included among the therapeutic strategies. The present study was aimed to discover whether ferroptosis, a programmed iron-dependent cell death, is involved in IMA-induced cardiotoxicity. In vivo, mouse model was established after treated with 25 mg/kg, 50 mg/kg and 100 mg/kg IMA. Serum CK, LDH, AST activities were determined. Cardiac tissues were examined by H&E and Oil Red O staining. MDA was measured to assess production of lipid peroxide. Tissue iron and GSH content were measured. In vitro, cell viability, mitochondria membrane potential, generation of reactive oxygen species (ROS) and cellular iron levels were performed to explore the mechanism of IMA. The in vivo results revealed that IMA treatment significantly increased serum CK, LDH and AST. H&E staining showed that IMA caused cardiac structural injuries. The dose-dependent decrease of GSH and increase of tissue iron and MDA were observed in IMA-treated groups. Oil Red O staining suggested obvious cardiac lipid accumulation after treated with IMA. In H9c2 cardiomyocytes, IMA significantly inhibited cell proliferation in a dose-dependent manner. Mitochondria membrane potential assay showed that IMA destroyed the mitochondrial function. Additionally, IMA increased the cellular ROS and iron levels. Furthermore, IMA down-regulated the expression of Nrf2 and up-regulated the expression of P53 and TfR. These results provided compelling evidence that ferroptosis participates in IMA-induced cardiotoxicity. Ferroptosis could be regarded as a target to protect against cardiotoxicity in IMA-exposed patients.


Asunto(s)
Antineoplásicos/toxicidad , Cardiotoxicidad , Ferroptosis/efectos de los fármacos , Mesilato de Imatinib/toxicidad , Factor 2 Relacionado con NF-E2/biosíntesis , Inhibidores de Proteínas Quinasas/toxicidad , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Glutatión/metabolismo , Hierro/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Peróxidos Lipídicos/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Especies Reactivas de Oxígeno/metabolismo
2.
DNA Cell Biol ; 40(8): 1101-1111, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34156283

RESUMEN

Chronic myeloid leukemia (CML) is characterized by the formation of the BCR-ABL fusion gene. The BCR-ABL protein leads to an increased level of reactive oxygen species, which is a major cause of endogenous DNA double-strand breaks (DSBs). CML cells are prone to rely on a highly mutagenic alternative end-joining (Alt-EJ) pathway to cope with enhanced DSBs, which aggravates chromosomal instability. Hence, targeting dysregulated DNA repair proteins provides new insights into cancer treatment. In this study, we discovered the abnormal upregulation of Flap endonuclease 1 (FEN1) in CML, as well as FEN1's participation in the error-prone Alt-EJ repair pathway and its interplay with DNA Ligase1 and proliferating cell nuclear antigen in DSB repair. Knockdown of FEN1 by shRNA not only inhibited the proliferation and induced apoptosis but also enhanced the efficacy of imatinib (IM) in drug-resistant CML cell K562/G01. Moreover, excessive DSB accumulation was detected after FEN1 inhibition. In summary, our results demonstrated that FEN1 is a promising therapeutic target in CML treatment. This work extends the understanding of regulating abnormal DSB repair for cancer treatment.


Asunto(s)
Proliferación Celular , Reparación del ADN por Unión de Extremidades , Endonucleasas de ADN Solapado/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Antineoplásicos/toxicidad , Apoptosis , Células Cultivadas , ADN Ligasa (ATP)/metabolismo , Resistencia a Antineoplásicos , Endonucleasas de ADN Solapado/metabolismo , Células HEK293 , Humanos , Mesilato de Imatinib/toxicidad , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Células THP-1
3.
Biomolecules ; 11(4)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33924068

RESUMEN

Chronic myeloid leukemia (CML) develops due to the presence of the BCR-ABL1 protein, a target of tyrosine kinase inhibitors (TKIs), such as imatinib (IM), used in a CML therapy. CML eradication is a challenge due to developing resistance to TKIs. BCR-ABL1 induces endogenous oxidative stress leading to genomic instability and development of TKI resistance. Model CML cells susceptible or resistant to IM, as well as wild-type, non-cancer cells without the BCR-ABL1 protein were treated with IM, hydrogen peroxide (H2O2) as a model trigger of external oxidative stress, or with IM+H2O2. Accumulation of reactive oxygen species (ROS), DNA damage, activity of selected antioxidant enzymes and glutathione (GSH), and mitochondrial potential (MMP) were assessed. We observed increase in ROS accumulation in BCR-ABL1 positive cells and distinct levels of ROS accumulation in IM-susceptible cells when compared to IM-resistant ones, as well as increased DNA damage caused by IM action in sensitive cells. Depletion of GSH levels and a decreased activity of glutathione peroxidase (GPx) in the presence of IM was higher in the cells susceptible to IM. IM-resistant cells showed an increase of catalase activity and a depletion of MMP. BCR-ABL1 kinase alters ROS metabolism, and IM resistance is accompanied by the changes in activity of GPx, catalase, and alterations in MMP.


Asunto(s)
Antineoplásicos/toxicidad , Resistencia a Antineoplásicos , Mesilato de Imatinib/toxicidad , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Estrés Oxidativo , Animales , Catalasa/metabolismo , Línea Celular Tumoral , Daño del ADN , Proteínas de Fusión bcr-abl/genética , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Potencial de la Membrana Mitocondrial , Ratones
4.
Chemosphere ; 263: 128097, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33297093

RESUMEN

Imatinib mesylate (IM) is an anticancer drug that belongs to tyrosine kinase inhibitors. We report the results of the first investigation of the chronic exposure of zebrafish (Danio rerio) to IM. The exposure to IM (0.01, 1 and 100 µg/L) was initiated in adult fish and continued through hatching and the offspring generation for seven months. In addition to standard toxicological endpoints, induction of genotoxic effects and whole-genome transcriptome of liver samples of offspring generation of zebrafish were analysed. Exposure to IM did not affect the survival and growth of zebrafish, did not cause any histopathological changes, but it induced a marginal increase in the chromosomal damage in blood cells. The whole-genome transcriptome analyses demonstrated dose-dependent increase in the number of differentially expressed genes with a significantly higher number of deregulated genes in female fish compared to male. Differentially expressed genes included genes involved in response to DNA damage, cell cycle control and regulation of circadian rhythm. Based on the low genotoxic activity and the pattern of the changes in DNA damage responsive genes we consider that at current environmental exposure levels, IM represents low risk for genotoxic effects in aquatic organisms. Exposure to IM also induced deregulation of the expression of genes associated with steroidogenesis and hormone metabolism and function, which indicates hormone-disrupting activity of IM that has not been studied so far. The study provide new information on the potential consequences of chronic exposure to the residues of tyrosine kinase inhibitors, which remain to be further explored.


Asunto(s)
Contaminantes Químicos del Agua , Pez Cebra , Animales , Femenino , Mesilato de Imatinib/toxicidad , Estadios del Ciclo de Vida , Masculino , Transcriptoma , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/genética
5.
Hum Exp Toxicol ; 40(4): 634-648, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32990058

RESUMEN

BACKGROUND: The purpose of this study was to examine the effects of exposure to imatinib in the prenatal period on testis development in rats. METHODS: Although all the study groups received intraperitoneal imatinib on prenatal days 1-8, no pregnancy occurred in the Imatinib-80 group. Immunohistochemical analysis, TUNEL, c-kit and PDGF staining revealed no difference between the groups in terms of positivity scoring. RESULTS: A significant decrease was detected in total sperm counts in the Imatinib-20 group compared to the control group, but the sperm count was higher in the Imatinib-60 group than in the Imatinib-20 group. At biochemical measurements, the drug increased oxidative stress in the testis and serum in the Imatinib-20 group, but caused a decrease in tissue in the Imatinib-60 group. Thiol measurements revealed a decrease in the testis and serum in the Imatinib-60 group, while an increase in serum measurements was observed in the Imatinib-40 group. Analysis revealed no difference between the groups in terms of protamine and histone gene expression levels in testis tissue exposed to imatinib. CONCLUSION: Our findings show that prenatal exposure to imatinib can lead to histopathological and biochemical changes in testis tissue, but that no adverse effect occurs in nuclear maturation of germ cells during spermiogenesis.


Asunto(s)
Antineoplásicos/toxicidad , Mesilato de Imatinib/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Testículo/efectos de los fármacos , Animales , Femenino , Masculino , Intercambio Materno-Fetal , Embarazo , Ratas , Recuento de Espermatozoides , Espermatogénesis/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Testículo/crecimiento & desarrollo , Testículo/patología
6.
Fundam Clin Pharmacol ; 34(4): 458-475, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32080901

RESUMEN

There is a great demand to introduce new approaches into cancer treatment field due to incidence of increased breast cancer all over the world. The current study was designed to evaluate the role of imatinib mesylate (IM) and/or hesperidin (HES) nanoparticles alone or in combination in enhancing the anticancer activity and to investigate the ability of nanoencapsulation to reduce cardiotoxicity of IM in solid Ehrlich carcinoma (SEC)-bearing mice. IM and HES were loaded into PLGA (poly(lactic-co-glycolic acid) polymer. SEC was induced in female albino mice as a model for experimentally induced breast cancer. Mice were randomly divided into eight groups (n = 10). On day 28 from tumor inoculation, mice were sacrificed and blood samples were collected in heparinized tubes for hematological studies, biochemical determination of lactate dehydrogenase (LDH), and glutamic oxaloacetic transaminase (SGOT) levels. In addition, tumor and cardiac tissues were utilized for histopathological examination as well as determination of MDR-1 gene expression. Immunohistochemical staining of BAX and BCL-2 was done. Nano IM- and/or Nano HES-treated groups showed a significant reduction in tumor volume, weight, hematological, cardiac markers, and tumor MDR-1 gene downregulation compared to free conventional treated groups. In conclusion, the use of HES as an adjuvant therapy with IM could improve its cytotoxic effects and limit its cardiac toxicity. Furthermore, nanoencapsulation of IM and/or HES with PLGA polymer showed a remarkable anticancer activity.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma de Ehrlich/tratamiento farmacológico , Caspasa 3/metabolismo , Mesilato de Imatinib/farmacología , Indoles/farmacología , Antígeno Ki-67/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/farmacología , Proteína X Asociada a bcl-2/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma de Ehrlich/genética , Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/patología , Cardiotoxicidad , Portadores de Fármacos , Composición de Medicamentos , Femenino , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Humanos , Mesilato de Imatinib/química , Mesilato de Imatinib/toxicidad , Indoles/química , Indoles/toxicidad , Células MCF-7 , Ratones , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Transducción de Señal , Sulfonamidas/química , Sulfonamidas/toxicidad , Carga Tumoral/efectos de los fármacos
7.
Toxicol Appl Pharmacol ; 379: 114645, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31278918

RESUMEN

Imatinib (IM) is a tyrosine kinase (TK) inhibitor (TKI) used to treat chronic myeloid leukemia. Clinical case reports and a few laboratory mammal studies provide inconclusive evidence about its deleterious effects on reproduction. The aim of the current study was to evaluate the potential of zebrafish to characterize IM-induced effects on reproduction and clarify IM effects on reproductive success. To this end, we exposed adult zebrafish to four concentrations of IM for 30 days followed by a 30-day depuration period. IM exposure caused a concentration-dependent, irreversible, suppression of folliculogenesis, reversible decrease in sperm density and motility, decreased fecundity and fertility, but no significant change in atretic follicle abundance. We also observed IM-induced premature hatching, but no significant change in embryo-larvae survivability. However, we found significant IM-induced morphometric malformations. IM decreased expression of vegfaa and igf2a (two reproductive-, angiogenic-, and growth-related genes) in testes and ovaries. The results demonstrate IM can induce significant changes in critical reproductive endpoints and zebrafish as a suitable model organism to show effects of IM on reproduction. The findings suggest that TKI effects on reproductive success should be considered.


Asunto(s)
Gónadas/efectos de los fármacos , Mesilato de Imatinib/toxicidad , Reproducción/efectos de los fármacos , Animales , Embrión no Mamífero/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Femenino , Fertilidad/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Gónadas/crecimiento & desarrollo , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Masculino , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Pez Cebra/fisiología
8.
J Pediatr Hematol Oncol ; 41(5): 376-381, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31033785

RESUMEN

Philadelphia-positive (Ph+) acute lymphoblastic leukemia (ALL) in children had a worse outcome before the use of tyrosine kinase inhibitors. We have evaluated the demographics and outcome of Ph+ ALL patients treated with imatinib without blood marrow transplantation. Of the 206 children with ALL registered for treatment, the demographic data of 15 Ph+ ALL patients were compared with the remaining Ph- patients. Imatinib (340 mg/m) was started on day 5 (D5) of induction in Ph+ patients, and their overall survival was compared with Ph- high-risk patients treated on similar protocols. Statistical analysis was carried out by the Fisher exact test and the t test. The Kaplan-Meier test was used for survival analysis. Philadelphia positivity noted in 15/206 (7.28%) ALL patients was higher than reported earlier. Median initial total leukocyte count and central nervous system positivity were significantly higher in Ph+ patients. Myeloid markers, CD13 and CD33, were also positive in 33.3% Ph+ patients. D15 and D35 marrow showed remissions in a larger proportion of Ph+ ALL, as compared with Ph- patients, but chemotherapy interruptions and neutropenic deaths were significantly higher after starting imatinib, as compared with Philadelphia high-risk patients. Overall survival was similar in Ph+ and Ph- high-risk ALL patients. Ph+ ALL, noted in 7.28%, presented with high initial white blood cell counts, high central nervous system positivity, poor steroid response, and higher induction deaths, as compared with high-risk Ph- ALL, and raised the question about the appropriate dose and time of introduction of imatinib to prevent toxicity.


Asunto(s)
Mesilato de Imatinib/uso terapéutico , Cromosoma Filadelfia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Examen de la Médula Ósea , Neoplasias del Sistema Nervioso Central , Niño , Femenino , Humanos , Mesilato de Imatinib/toxicidad , India , Estimación de Kaplan-Meier , Recuento de Leucocitos , Masculino , Neutropenia/inducido químicamente , Neutropenia/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Inducción de Remisión , Análisis de Supervivencia , Resultado del Tratamiento
9.
Toxicol In Vitro ; 58: 178-186, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30910525

RESUMEN

The anti-cancer receptor tyrosine kinase inhibitors include known cardiotoxins: a component of this toxicity may be mediated by effects on cardiac fibroblasts (CFs). We hypothesised that imatinib mesylate (imatinib) and sunitinib malate (sunitinib) cause significant dysfunction in adult CFs. Following in vitro treatments with imatinib or sunitinib, adult rat CF viability was assessed by fluorescein diacetate assay, proliferation measured by bromodeoxyuridine nuclear incorporation and changes to the expression of CF secretome components determined by real time quantitative RT-PCR. Imatinib and sunitinib significantly reduced cell viability over 48 h, with EC50 values of 11.0 µM (imatinib) and 4.5 µM (sunitinib) respectively. Imatinib reduced CF proliferation from 35.5 ±â€¯3.2% in control to 23.0 ±â€¯5.5% (3 µM; p < 0.001) and to 9.4 ±â€¯2.5% (10 µM; p < 0.001), whereas sunitinib reduced proliferation to 22.9 ±â€¯3.1% (1 µM; p < 0.001) and to 15 ±â€¯1.0% (3 µM; p < 0.001). Further, 10 µM imatinib increased mRNA expression of TGFB1 7-fold, (p < 0.01), IL6 6-fold (p < 0.01), and IL1B 7-fold (p < 0.05) and reduced PDGFD 15-fold (p < 0.01); whereas sunitinib specifically reduced IL1B mRNA expression 17-fold (p < 0.01). Overall, these findings show tyrosine kinase inhibitors cause significant dysfunction in CFs. These data point to an important role for the PDGF pathway in governing CF functions, including survival and proliferation.


Asunto(s)
Fibroblastos/efectos de los fármacos , Mesilato de Imatinib/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Sunitinib/toxicidad , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Fibroblastos/metabolismo , Masculino , Miocardio/citología , Ratas , Ratas Wistar
10.
Sci Total Environ ; 655: 547-556, 2019 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-30476834

RESUMEN

Imatinib (IMA) is a highly potent tyrosine kinase inhibitor used as first-line anti-cancer drug in the treatment of chronic myeloid leukemia. Due to its universal mechanism of action, IMA also has endocrine and mutagenic disrupting effects in vivo and in vitro, which raises the question of its environmental impact. However, to date, very little information is available on its environmental fate and the potential role of its transformation products (TPs) on aquatic organisms. Given the IMA resistance to hydrolysis and direct photolysis according to the literature, we sought to generate TPs through oxidative and radical conditions using the AOPs pathway. Thus, the reactivity of the cytotoxic drug IMA in water in the presence of OH and h+ was investigated for the first time in the present work. In this regard, a non-targeted screening approach was applied in order to reveal its potential TPs. The tentative structural elucidation of the detected TPs was performed by LC-HRMSn. The proposed approach allowed detecting a total of twelve TPs, among which eleven are being described for the first time in this work. Although the structures of these TPs could not be positively confirmed due to lack of standards, their chemical formulas and product ions can be added to databases, which will allow their screening in future monitoring studies. Using the quantitative structure-activity relationship (QSAR) approach and rule-based software, we have shown that the detected TPs possess, like their parent molecule, comparable acute toxicity as well as mutagenic and estrogenic potential. In addition to the in silico studies, we also found that the samples obtained at different exposure times to oxidative conditions, including those where IMA is no longer detected, retained toxicity in vitro. Such results suggest further studies are needed to increase our knowledge of the impact of imatinib on the environment.


Asunto(s)
Organismos Acuáticos/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Mesilato de Imatinib/toxicidad , Mutágenos/toxicidad , Fotólisis , Contaminantes Químicos del Agua/toxicidad , Adsorción , Aliivibrio fischeri/efectos de los fármacos , Organismos Acuáticos/genética , Biodegradación Ambiental , Catálisis , Simulación por Computador , Disruptores Endocrinos/química , Disruptores Endocrinos/efectos de la radiación , Mesilato de Imatinib/química , Mesilato de Imatinib/efectos de la radiación , Estructura Molecular , Mutágenos/química , Mutágenos/efectos de la radiación , Relación Estructura-Actividad Cuantitativa , Titanio/química , Contaminantes Químicos del Agua/química , Contaminantes Químicos del Agua/efectos de la radiación
11.
Biochem Pharmacol ; 155: 162-171, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29983397

RESUMEN

Tyrosine kinase inhibitors (TKIs) have advanced cancer treatment and prognosis but have also resulted in adverse effects such as fatigue, diarrhea, hypothyroidism, and other toxicities. We investigated TKI effects on skeletal muscle as a possible explanation of TKI induced fatigue. Changes in mitochondrial function due to inhibition of oxidative phosphorylation complexes, generation of superoxides, and inhibition of key transporters involved in uptake of glucose and/or nucleosides may result in alteration of energy metabolism and/or mitochondrial function. We investigated effects of imatinib, sorafenib and sunitinib on these processes in cultured C2C12 murine skeletal muscle cells. Imatinib, sorafenib and sunitinib were cytotoxic to C2C12 cells with IC50 values of 20, 8 and 8 µM, respectively. Imatinib stimulated glucose uptake and inhibited complex V activity by 35% at 50 µM. Sorafenib inhibited complex II/III and V with IC50 values of 32 and 28 µM, respectively. Sorafenib caused activation of caspase 3/7 and depolarization of mitochondrial membranes occurred very rapidly with complete loss at 5-10 µM. Sunitinib inhibited Complex I with an IC50 value of 38 µM and caused ATP depletion, caspase 3/7 activation, an increase in reactive oxygen species (ROS), and decreased nucleoside and glucose uptake. In conclusion, imatinib, sunitinib and sorafenib caused changes in mitochondrial complex activities, glucose and nucleoside uptake leading to decreased energy production and mitochondrial function in a skeletal muscle cell model, suggesting that these changes may play a role in fatigue, one of the most common adverse effects of TKIs.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Mesilato de Imatinib/toxicidad , Fibras Musculares Esqueléticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/toxicidad , Sorafenib/toxicidad , Sunitinib/toxicidad , Animales , Línea Celular , Células Cultivadas , Citotoxinas/toxicidad , Relación Dosis-Respuesta a Droga , Metabolismo Energético/fisiología , Ratones , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo
12.
Arch Environ Contam Toxicol ; 74(4): 546-556, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29119204

RESUMEN

Benzalkonium chloride (BAC) is a cationic surfactant commonly used as a disinfectant. Its ubiquitous nature is the result of high usage and frequent discharge into the environment and evidence of interaction with numerous contaminants, such as pharmaceutical active compound residues. Anticancer drugs, among these compounds, are able to exert eco-genotoxic effects at sub ng-µg/L. The purpose of this study was to assess the reproductive toxicity and the genotoxicity of 5-fluorouracil (5-FU), cisplatin (CDDP), etoposide (ET), and imatinib mesylate (IM)-binary mixtures combined with BAC in Ceriodaphnia dubia. The effects of the mixtures were assessed under the assumption of independent action in experiments that applied two effect levels. The type of interaction was not the same over the range of effect sizes. The combined action experiment on reproduction showed an antagonistic effect at higher effect levels for all binary combinations, except for BAC/IM, whereas independent action was observed in all mixtures at a low effect level. The results of binary combinations on genotoxicity showed antagonistic effects for BAC + ET and BAC + CDDP, whereas independence was expressed in BAC + IM and BAC + 5-FU. The antagonistic interactions still led to higher effects than those observed after single exposures at the same doses in most cases. The effects of mixtures of drugs should be taken into account for environmental risk assessment.


Asunto(s)
Antineoplásicos/toxicidad , Compuestos de Benzalconio/toxicidad , Cladóceros/efectos de los fármacos , Pruebas de Toxicidad/métodos , Animales , Cisplatino/toxicidad , Ensayo Cometa , Mezclas Complejas/toxicidad , Daño del ADN/efectos de los fármacos , Ecotoxicología/métodos , Etopósido/toxicidad , Femenino , Fluorouracilo/toxicidad , Agua Dulce , Mesilato de Imatinib/toxicidad , Masculino , Reproducción , Contaminantes Químicos del Agua/toxicidad
13.
Pharmacol Res ; 127: 15-25, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28964914

RESUMEN

Cardiovascular complications are included among the systemic effects of tyrosine kinase inhibitor (TKI)-based therapeutic strategies. To test the hypothesis that inhibition of Kit tyrosine kinase that promotes cardiac progenitor cell (CPC) survival and function may be one of the triggering mechanisms of imatinib mesylate (IM)-related cardiovascular effects, the anatomical, structural and ultrastructural changes in the heart of IM-treated rats were evaluated. Cardiac anatomy in IM-exposed rats showed a dose-dependent, restrictive type of remodeling and depressed hemodynamic performance in the absence of remarkable myocardial fibrosis. The effects of IM on rat and human CPCs were also assessed. IM induced rat CPC depletion, reduced growth and increased cell death. Similar effects were observed in CPCs isolated from human hearts. These results extend the notion that cardiovascular side effects are driven by multiple actions of IM. The identification of cellular mechanisms responsible for cardiovascular complications due to TKIs will enable future strategies aimed at preserving concomitantly cardiac integrity and anti-tumor activity of advanced cancer treatment.


Asunto(s)
Cardiomiopatías/inducido químicamente , Mesilato de Imatinib/toxicidad , Miocardio/patología , Células Madre/efectos de los fármacos , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hemodinámica/efectos de los fármacos , Humanos , Masculino , Miocardio/ultraestructura , Ratas
14.
Chemosphere ; 189: 689-698, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28968575

RESUMEN

Available ecotoxicological data for anti-cancer drugs and their metabolites are incomplete, and only some studies have been accompanied by chemical analysis. Therefore, the main aim of this study was to evaluate the acute toxicity of the six most commonly used cytostatics, namely cyclophosphamide (CF), ifosfamide (IF), 5-fluorouracil (5-FU), imatinib (IMT), tamoxifen (TAM) and methotrexate (MET) and its metabolite - 7-hydroxymethotrexate (7-OH-MET), towards selected aquatic organisms, namely bacteria Vibrio fischeri, algae Raphidocelis subcapitata, crustaceans Daphnia magna and duckweed Lemna minor. All ecotoxicological tests were accompanied by chemical analysis to determine the differences between nominal and actual concentrations of investigated compounds and their stability under test conditions. For unstable compounds, tests were performed in static and semi-static conditions. It was observed that L. minor was the most sensitive organism. The compounds that were most toxic to aquatic organisms were 5-FU (highly toxic to algae, EC50 = 0.075 mg L-1), MET and TAM (very toxic to highly toxic to duckweed depending on the test conditions; EC50MET 0.08-0.16 mg L-1, EC50TAM 0.18-0.23 mg L-1). It is suspected that MET and 5-FU mainly affected algae and plants most probably because the exposure time was long enough for them to cause a specific effect (they inhibit DNA replication and act predominantly on actively dividing cells). Furthermore, the obtained results also suggest that the toxicity of the metabolites/potentially produced degradation products of MET towards duckweed is lower than that of the parent form, whereas the toxicity of TAM degradation products is in the same range as that of TAM.


Asunto(s)
Antineoplásicos/toxicidad , Pruebas de Toxicidad Aguda/métodos , Contaminantes Químicos del Agua/toxicidad , Aliivibrio fischeri/efectos de los fármacos , Animales , Organismos Acuáticos/efectos de los fármacos , Araceae/efectos de los fármacos , Chlorophyta/efectos de los fármacos , Ciclofosfamida/toxicidad , Daphnia/efectos de los fármacos , Ecotoxicología , Fluorouracilo/metabolismo , Fluorouracilo/toxicidad , Mesilato de Imatinib/toxicidad , Metotrexato/análogos & derivados , Metotrexato/toxicidad
15.
PLoS One ; 12(5): e0177871, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28520795

RESUMEN

Baculoviral IAP repeat containing 6 (BIRC6) is a member of the inhibitors of apoptosis proteins (IAPs), a family of functionally and structurally related proteins that inhibit apoptosis. BIRC6 has been implicated in drug resistance in several different human cancers, however mechanisms regulating BIRC6 have not been extensively explored. Our phosphoproteomic analysis of an imatinib-resistant chronic myelogenous leukemia (CML) cell line (MYL-R) identified increased amounts of a BIRC6 peptide phosphorylated at S480, S482, and S486 compared to imatinib-sensitive CML cells (MYL). Thus we investigated the role of BIRC6 in mediating imatinib resistance and compared it to the well-characterized anti-apoptotic protein, Mcl-1. Both BIRC6 and Mcl-1 were elevated in MYL-R compared to MYL cells. Lentiviral shRNA knockdown of BIRC6 in MYL-R cells increased imatinib-stimulated caspase activation and resulted in a ~20-25-fold increase in imatinib sensitivity, without affecting Mcl-1. Treating MYL-R cells with CDK9 inhibitors decreased BIRC6 mRNA, but not BIRC6 protein levels. By contrast, while CDK9 inhibitors reduced Mcl-1 mRNA and protein, they did not affect imatinib sensitivity. Since the Src family kinase Lyn is highly expressed and active in MYL-R cells, we tested the effects of Lyn inhibition on BIRC6 and Mcl-1. RNAi-mediated knockdown or inhibition of Lyn (dasatinib/ponatinib) reduced BIRC6 protein stability and increased caspase activation. Inhibition of Lyn also increased formation of an N-terminal BIRC6 fragment in parallel with reduced amount of the BIRC6 phosphopeptide, suggesting that Lyn may regulate BIRC6 phosphorylation and stability. In summary, our data show that BIRC6 stability is dependent on Lyn, and that BIRC6 mediates imatinib sensitivity independently of Mcl-1 or CDK9. Hence, BIRC6 may be a novel target for the treatment of drug-resistant CML where Mcl-1 or CDK9 inhibitors have failed.


Asunto(s)
Antineoplásicos/toxicidad , Apoptosis , Resistencia a Antineoplásicos/genética , Mesilato de Imatinib/toxicidad , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
16.
Toxicology ; 382: 24-35, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28315715

RESUMEN

Imatinib mesylate is an effective treatment for chronic myelogenous leukemia and gastrointestinal stromal tumors. Although imatinib mesylate is highly tolerable, it has been implicated in severe congestive heart failure in mouse models and patients. A hallmark of imatinib mesylate-induced cardiotoxicity is mitochondrial dysfunction. The mitochondrial scaffold Sab has been implicated in facilitating signaling responsible for mitochondrial dysfunction in a c-Jun N-terminal Kinase (JNK)-dependent manner. We examined the impact of Sab-mediated signaling on imatinib mesylate cardiotoxicity in H9c2 rat cardiomyocyte-like cells. Silencing Sab increased the LD50 of imatinib mesylate 4-fold in H9c2 cells. Disrupting Sab-mediated signaling prevented imatinib mesylate-induced apoptosis as well. Knockdown of Sab or inhibition with a small peptide prevented oxidative stress, which was indicated by decreased reactive oxygen species production, lipid peroxidation, and protein carbonylation. Further, inhibition of Sab-related signaling partially rescued deficits in mitochondrial respiration, ATP production, and membrane potential in imatinib mesylate-treated H9c2 cells. Conversely, over-expression of Sab in H9c2 cells increased the cardiotoxicity of imatinib mesylate in vitro decreasing the LD50 over 4-fold. Sab expression was induced in H9c2 cells following cardiovascular-like stress in an AP-1 dependent manner. These data demonstrate that imatinib mesylate influences mitochondrial signaling leading to mitochondrial dysfunction and cardiotoxicity.


Asunto(s)
Antineoplásicos/toxicidad , Cardiotoxicidad/metabolismo , Mesilato de Imatinib/toxicidad , Proteínas de Microfilamentos/metabolismo , Animales , Línea Celular , Estrés del Retículo Endoplásmico , Proteínas de Microfilamentos/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Ratas , Especies Reactivas de Oxígeno/metabolismo
17.
Cancer Chemother Pharmacol ; 79(4): 711-723, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28283735

RESUMEN

PURPOSE: Pulmonary arterial hypertension (PAH) results from occlusion or vasoconstriction of pulmonary vessels, leading to progressive right ventricular failure. Dasatinib, a BCR-ABL1 tyrosine kinase inhibitor (TKI) approved for the treatment of chronic myelogenous leukemia, has been associated with PAH. In contrast, the BCR-ABL1 TKI imatinib has demonstrated anti-vasoproliferative properties and has been investigated as a potential treatment for PAH. Here we describe studies evaluating the effects of dasatinib and imatinib on cardiovascular and pulmonary functions to understand the reported differential consequences of the two TKIs in a clinical setting. METHODS: The direct effects of dasatinib and imatinib were explored in vivo to investigate possible mechanisms of dasatinib-induced PAH. In addition, effects of dasatinib and imatinib on PAH-related mediators were evaluated in vitro. RESULTS: In rats, both TKIs increased plasma nitric oxide (NO), did not induce PAH-related structural or molecular changes in PA or lungs, and did not alter hemodynamic lung function compared with positive controls. Similarly, in the pulmonary artery endothelial cells and smooth muscle cells co-culture model, imatinib and dasatinib increased NO and decreased endothelin-1 protein and mRNA. CONCLUSIONS: The results of these studies indicated that dasatinib did not induce physiological changes or molecular signatures consistent with PAH when compared to positive controls. Instead, dasatinib induced changes consistent with imatinib. Both dasatinib and imatinib induced biochemical and structural changes consistent with a protective effect for PAH. These data suggest that other factors of unclear etiology contributed to the development of PAH in patients treated with dasatinib.


Asunto(s)
Antineoplásicos/toxicidad , Dasatinib/toxicidad , Hipertensión Pulmonar/inducido químicamente , Mesilato de Imatinib/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Animales , Antineoplásicos/farmacocinética , Dasatinib/farmacocinética , Endotelina-1/sangre , Expresión Génica/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Hipertensión Pulmonar/fisiopatología , Mesilato de Imatinib/farmacocinética , Pulmón/patología , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico/sangre , Inhibidores de Proteínas Quinasas/farmacocinética , Arteria Pulmonar/efectos de los fármacos , Circulación Pulmonar/efectos de los fármacos , ARN Mensajero/sangre , Ratas , Ratas Sprague-Dawley
18.
Artículo en Inglés | MEDLINE | ID: mdl-28137363

RESUMEN

The selective tyrosine kinase inhibitor imatinib mesylate (IM) is a widely used anticancer drug. Recent studies showing that IM can induce DNA and chromosomal damage in crustaceans and higher plants prompted us to re-examine its potential genotoxicity. IM was not mutagenic in the Ames assay (Salmonella typhimurium). Cytotoxicity and genotoxicity were evaluated in vitro in zebrafish (Danio rerio) liver (ZFL), human hepatoma (HepG2), and human peripheral blood lymphocyte (HPBL) cells. Genotoxicity was determined with the comet assay and with the cytokinesis-block micronucleus cytome assay. ZFL and HPBL cells showed comparable sensitivity to IM cytotoxicity, while HepG2 cells were less sensitive. At non-cytotoxic concentrations, IM induced DNA strand breaks in ZFL and HepG2 cells. An increase in the number of micronuclei was observed in ZFL and HPBL cells. In HPBLs, IM also induced an increase in the number of nucleoplasmic bridges and nuclear buds. Based on the data of the consumption of IM in European countries the predicted environmental concentrations (PEC) were calculated to be in the range between 3.3 and 5.0ng/L, which are several orders of magnitude lower from those that caused adverse effects in fish and human derived cells. However, based on the in vitro studies it is not possible to quantitatively predict the hazard for wildlife and humans, therefore further studies are warranted to explore the underlying molecular mechanisms of induced IM genotoxic effects as well as the studies of the occurrence of IM in the aquatic and occupational environment to establish the relevance of these observations for aquatic organisms and occupationally exposed personnel.


Asunto(s)
Mesilato de Imatinib/toxicidad , Proteínas Tirosina Quinasas/toxicidad , Animales , Línea Celular Tumoral , Células Cultivadas , Daño del ADN , Inestabilidad Genómica , Humanos , Pruebas de Mutagenicidad , Pez Cebra
19.
PLoS One ; 11(10): e0164727, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27737004

RESUMEN

Nilotinib and imatinib are tyrosine kinase inhibitors (TKIs) used in the treatment of chronic myeloid leukemia (CML) and gastrointestinal stromal tumors (GIST). In vitro, imatinib and nilotinib inhibit osteoclastogenesis, and in patients they reduce levels of bone resorption. One of the mechanisms that might underlie these effects is an increase in the production of osteoprotegerin (OPG). In the current work we report that platelet-derived growth factor receptor beta (PDGFRß) signaling regulates OPG production in vitro. In addition, we have shown that TKIs have effects on RANKL signaling through inhibition of the PDGFRß and other target receptors. These findings have implications for our understanding of the mechanisms by which TKIs affect osteoclastogenesis, and the role of PDGFRß signaling in regulating osteoclastogenesis. Further studies are indicated to confirm the clinical effects of PDGFRß-inhibitors and to elaborate the intracellular pathways that underpin these effects.


Asunto(s)
Expresión Génica/efectos de los fármacos , Mesilato de Imatinib/toxicidad , Osteoprotegerina/metabolismo , Inhibidores de Proteínas Quinasas/toxicidad , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Becaplermina , Células Cultivadas , Femenino , Masculino , Ratones , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteoprotegerina/genética , Proteínas Proto-Oncogénicas c-sis/farmacología , Ligando RANK/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/efectos de los fármacos
20.
Chemosphere ; 165: 529-538, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27681109

RESUMEN

Antineoplastic drugs are highly cytotoxic chemotherapeutic agents that can often interfere directly or indirectly with the cell's genome. In an environmental or medical setting simultaneous exposure may occur. Such multiple exposures may pose a higher risk than it could be assumed from the studies evaluating the effect of a single substance. Therefore, in the present study we tested the combined cyto/genotoxicity of a mixture of selected antineoplastic drugs with different mechanisms of action (5-fluorouracil, etoposide, and imatinib mesylate) towards human lymphocytes in vitro. The results suggest that the selected antineoplastic drug mixture is potentially cyto/genotoxic and that it can induce cell and genome damage even at low concentrations. Moreover, the changes in the measured oxidative stress parameters suggest the participation of reactive oxygen species in the cyto/genotoxicity of the selected mixture. The obtained results indicate not only that such mixtures may pose a risk to cell and genome integrity, but also that single compound toxicity data are not sufficient for the predicting toxicity in a complex environment. Altogether, the results emphasise the need for further toxicological screening of antineoplastic drug mixtures, especially at low environmentally relevant concentrations, as to avoid any possible adverse effects on the environment and human health.


Asunto(s)
Antineoplásicos/toxicidad , Linfocitos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN , Interacciones Farmacológicas , Etopósido/toxicidad , Fluorouracilo/toxicidad , Humanos , Mesilato de Imatinib/toxicidad , Linfocitos/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA