Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Cell Biochem ; 438(1-2): 199-217, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28795366

RESUMEN

The aim of this work was the synthesis, characterization, and cytotoxicity evaluation of three new Ru(II) complexes with a general formula [Ru(Spy)(bipy)(P-P)]PF6 [Spy = pyridine-6-thiolate; bipy = 2,2'-bipyridine; P-P = 1,2-bis(diphenylphosphine)ethane (1); 1,3-bis(diphenylphosphine) propane (2); and 1,1'-bis(diphenylphosphino)ferrocene] (4). Complex (3) with the 1,4-bis(diphenylphosphine)butane ligand, already known from the literature, was also synthesized, to be better studied here. The cytotoxicities of the complexes toward two kinds of cancerous cells (K562 and S-180 cells) were evaluated and compared to normal cells (L-929 and PBMC) by MTT assay. The complex [Ru(Spy)(bipy)(dppb)]PF6 (3) was selected to study both the cellular and molecular mechanisms underlying its promising anticancer action in S-180 cells. The results obtained from this study indicated that complex (3) induces cell cycle arrest in the G0/G1 phase in S-180 cells associated with a decrease in the number of cells in S phase. After 24 and 48 h of exposure to complex (3), the cell viability decreased when compared to the negative control. Complex (3) does not appear to be involved in the DNA damage, but induced changes in the mitochondrial membrane potential in S-180 cells. Furthermore, there was also an increase in the gene expression of Bax, Caspase 9, and Tp53. According to our results, complex (3) induces cell apoptosis through p53/Bax-dependent intrinsic pathway and suppresses the expression of active antiapoptotic Bcl-2 protein.


Asunto(s)
Apoptosis/efectos de los fármacos , Complejos de Coordinación , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Rutenio , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Humanos , Células K562 , Ratones , Mitocondrias/patología , Rutenio/química , Rutenio/farmacología
2.
Biometals ; 27(3): 459-69, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24781824

RESUMEN

Lung cancer is one of the leading causes of death in the world, and non-small cell lung carcinoma accounts for approximately 75-85 % of all lung cancers. In the present work, we studied the antitumor activity of the compound cis-(dichloro)tetramineruthenium(III) chloride {cis-[RuCl2(NH3)4]Cl} against human lung carcinoma tumor cell line A549. The present study aimed to investigate the relationship between the expression of MDR1 and CYP450 genes in human lung carcinoma cell lines A549 treated with cisCarboPt, cisCRu(III) and cisDRu(III). The ruthenium-based coordinated complexes presented low cytotoxic and antiproliferative activities, with high IC50 values, 196 (±15.49), 472 (±20.29) and 175 (±1.41) for cisCarboPt, cisCRu(III) and cisDRu(III), respectively. The tested compounds induced apoptosis in A549 tumor cells as evidenced by caspase 3 activation, but only at high concentrations. Results also revealed that the amplification of P-gp gene is greater in A549 cells exposed to cisCarboPt and cisCRu(III) than cisDRu(III). Taken together all these results strongly demonstrate that MDR-1 over-expression in A549 cells could be associated to a MDR phenotype of these cells and moreover, it is also contributing to the platinum, and structurally-related compound, resistance in these cells. The identification and characterization of novel mechanisms of drug resistance will enable the development of a new generation of anti-cancer drugs that increase cancer sensitivity and/or represent more effective chemotherapeutic agents.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carboplatino/farmacología , Compuestos Organometálicos/farmacología , Compuestos de Rutenio/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocromo P-450 CYP2C9/genética , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Expresión Génica/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , L-Lactato Deshidrogenasa/metabolismo , Neoplasias Pulmonares
3.
Biomolecules ; 12(5)2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35625568

RESUMEN

TP53 gene mutation is the most common genetic alteration in human malignant tumors and is mainly responsible for Li-Fraumeni syndrome. Among the several cancers related to this syndrome, breast cancer (BC) is the most common. The TP53 p.R337H germline pathogenic variant is highly prevalent in Brazil's South and Southeast regions, accounting for 0.3% of the general population. We investigated the prevalence of TP53 germline pathogenic variants in a cohort of 83 BC patients from the Midwest Brazilian region. All patients met the clinical criteria for hereditary breast and ovarian cancer syndrome (HBOC) and were negative for BRCA1 and BRCA2 mutations. Moreover, 40 index patients fulfilled HBOC and the Li-Fraumeni-like (LFL) syndromes criteria. The samples were tested using next generation sequencing for TP53. Three patients harbored TP53 missense pathogenic variants (p.Arg248Gln, p.Arg337His, and p.Arg337Cys), confirmed by Sanger sequencing. One (1.2%) patient showed a large TP53 deletion (exons 2-11), which was also confirmed. The p.R337H variant was detected in only one patient. In conclusion, four (4.8%) early-onset breast cancer patients fulfilling the HBOC and LFL syndromes presented TP53 pathogenic variants, confirming the relevance of genetic tests in this group of patients. In contrast to other Brazilian regions, TP53 p.R337H variant appeared with low prevalence.


Asunto(s)
Neoplasias de la Mama , Síndrome de Li-Fraumeni , Neoplasias Ováricas , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/genética , Femenino , Mutación de Línea Germinal , Humanos , Síndrome de Li-Fraumeni/epidemiología , Síndrome de Li-Fraumeni/genética , Neoplasias Ováricas/epidemiología , Neoplasias Ováricas/genética , Síndrome , Proteína p53 Supresora de Tumor/genética
4.
Biol Trace Elem Res ; 197(1): 123-131, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31773484

RESUMEN

Ruthenium is attracting considerable interest as the basis for new compounds to treat diseases, and studies have shown that complexes with different structures have significant antineoplastic and antimetastatic potential against several types of tumors, including tumors resistant to cisplatin drugs. We examined the cytotoxic, genotoxic, and pro-apoptotic activities of six ruthenium complexes containing amino acid with general formulation [Ru(AA)(bipy)(dppb)]PF6, where AA = amino acid (alanine, glycine, leucine, lysine, methionine, or tryptophan); bipy = 2,2´-bipyridine; and dppb = [1,4-bis(diphenylphosphine)butane], against A549 (lung carcinoma) and K562 (chronic myelogenous leukemia) cancer cells. The results show that the ruthenium complexes tested were able to induce cytotoxicity in A549 and K562 cancer cells. Complex 1 containing alanine inhibited the cell viability of A549 and K562 tumor cells by inducing apoptosis, as evidenced by an increased number of Annexin V-positive cells and the induction of DNA damage and cell cycle arrest. Complex 1 was able to induce caspase-mediated apoptosis in K562 cells through the mitochondrial dysfunction, the upregulation of apoptotic genes, and the downregulation of Bcl2 anti-apoptotic gene. Besides being cytotoxic to K562 and A549 cells, ruthenium complex containing alanine shows low cytotoxicity and genotoxicity against non-tumor cells. These results suggest that the ruthenium (II) complex is a potential safe and efficient antineoplastic candidate for leukemia treatment.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Leucemia , Rutenio , Aminoácidos , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Humanos , Rutenio/farmacología
5.
Biol Trace Elem Res ; 198(2): 669-680, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32266641

RESUMEN

Ruthenium(II)/benzonitrile complexes have demonstrated promising anticancer properties. Considering that there are no specific therapies for treating sarcoma, we decided to evaluate the cytotoxic, genotoxic, and lethal effects of cis-[RuCl(BzCN)(phen)(dppb)]PF6 (BzCN = benzonitrile; phen = 1,10-phenanthroline; dppb = 1,4-bis-(diphenylphosphino)butane), as well as the mechanism of cell death induction that occurs against murine sarcoma-180 tumor. Thus, MTT assay was applied to assess the ruthenium cytotoxicity, showing that the compound is a more potent inhibitor for the sarcoma-180 tumor cell viability than normal cells (lymphocytes). The comet assay indicated low genotoxic for normal cells. cis-[RuCl(BzCN)(phen)(dppb)]PF6 also showed moderate lethality in Artemia salina. The complex induced cell cycle arrest in the G0/G1 phase in sarcoma-180 cells. In addition, the complex caused S180 cells to die by apoptosis by an increase in Annexin-V-positive cells and morphological changes typical of apoptotic cells. Additionally, cis-[RuCl(BzCN)(phen)(dppb)]PF6 increased the gene expression of Bax, Casp3, and Tp53 in S180 cells. By using a western blot, we observed an increased protein level of TNF-R2, Bax, and p21. In conclusion, cis-[RuCl(BzCN)(phen)(dppb)]PF6 is active and selective for sarcoma-180 cells, leading to cell cycle arrest at the G0/G1 and cell death through a caspases-mediated and Tp53/p21-mediated pathway.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Rutenio , Sarcoma , Animales , Antineoplásicos/farmacología , Apoptosis , Artemia , Caspasas , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ratones , Nitrilos , Rutenio/farmacología , Sarcoma/tratamiento farmacológico , Proteína p53 Supresora de Tumor
6.
Biomed Pharmacother ; 107: 1082-1092, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30257320

RESUMEN

Anticancer potential of ruthenium complexes has been widely investigated, but safety evaluation studies are still scarce. Despite of ruthenium-based anticancer agents are known to cause fewer side effects compared to other metal-based drugs, these compounds are not fully free of toxicity, causing mainly nephrotoxicity. Based on the promising results from antitumor activity of the complexes [Ru(L-Met)(bipy)(dppb)]PF6 (RuMet) and [Ru(L-Trp)(bipy)(dppb)]PF6 (RuTrp), for the first time we investigated the toxicity profile of these complexes in rodent and zebrafish models. The acute oral toxicity was evaluated in Swiss mice. The mutagenic and genotoxic potential was determined by a combination of Micronucleus (MN) and Comet assay protocols, after exposure of Swiss mice to RuMet and RuTrp in therapeutic doses. Zebrafish embryos were exposed to these complexes, and their development observed up to 96 h post-fertilization. RuMet and RuTrp complexes showed low acute oral toxicity. Recorded behavioral changes were not recorded, nor were macroscopic morphological changes or structural modifications in the liver and kidneys. These complexes did not cause genetic toxicity, presenting a lack of micronuclei formation and low DNA damage induction in the cells from Swiss mice. In contradiction, cisplatin treatment exhibited high mutagenicity and genotoxicity. RuMet and RuTrp showed low toxicity in the embryo development of zebrafish. The RuMet and RuTrp complexes demonstrated low toxicity in the two study models, an interesting property in preclinical studies for novel anticancer agents.


Asunto(s)
Antineoplásicos/toxicidad , Daño del ADN/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Compuestos de Rutenio/toxicidad , Administración Oral , Aminoácidos/química , Animales , Antineoplásicos/química , Cisplatino/toxicidad , Ensayo Cometa , Femenino , Masculino , Ratones , Pruebas de Micronúcleos , Compuestos de Rutenio/química , Pruebas de Toxicidad Aguda , Pez Cebra
7.
Arq Bras Oftalmol ; 78(2): 89-93, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25945529

RESUMEN

PURPOSE: To assess the cytotoxicity and genotoxicity of intravitreal adalimumab treatment in an animal experimental model using cytological and molecular techniques. METHODS: Eighteen rabbits were randomly assigned to three groups: control, adalimumab treatment, and placebo. Cytotoxicity on retinal cells was evaluated using flow cytometry assays to determine the level of apoptosis and necrosis. Genotoxicity was evaluated by comet assays to assess DNA damage, and quantitative real-time polymerase chain reaction (qPCR) was used to evaluate expression of apoptosis-inducing caspases (8 and 3). RESULTS: No cytotoxicity or genotoxicity was observed in any of the two treatment groups (adalimumab and placebo) following intravitreal administration compared with the control group. Flow cytometry analysis revealed that more than 90% of the cells were viable, and only a low proportion of retinal cells presented apoptotic (~10%) or necrotic (<1%) activity across all groups. Molecular damage was also low with a maximum of 6.4% DNA degradation observed in the comet assays. In addition, no increase in gene expression of apoptosis-inducing caspases was observed on retinal cells by qPCR in both the adalimumab and placebo groups compared with the control group. CONCLUSION: The use of adalimumab resulted in no detectable cytotoxicity or genotoxicity on retinal cells for up to 60 days upon administration. These results therefore indicate that adalimumab may be a safe option for intravitreal application to treat ocular inflammatory diseases in which TNF-α is involved.


Asunto(s)
Antiinflamatorios/toxicidad , Anticuerpos Monoclonales Humanizados/toxicidad , Inyecciones Intravítreas/métodos , Retina/efectos de los fármacos , Adalimumab , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/análisis , Caspasa 3/efectos de los fármacos , Caspasa 8/análisis , Caspasa 8/efectos de los fármacos , Supervivencia Celular , Ensayo Cometa , Daño del ADN , Citometría de Flujo , Masculino , Modelos Animales , Necrosis , Conejos , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Factor de Necrosis Tumoral alfa/efectos de los fármacos
8.
PLoS One ; 9(10): e105865, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25329644

RESUMEN

Over the past several decades, much attention has been focused on ruthenium complexes in antitumor therapy. Ruthenium is a transition metal that possesses several advantages for rational antitumor drug design and biological applications. In the present study, five ruthenium complexes containing amino acids were studied in vitro to determine their biological activity against sarcoma-180 tumor cells. The cytotoxicity of the complexes was evaluated by an MTT assay, and their mechanism of action was investigated. The results demonstrated that the five complexes inhibited the growth of the S180 tumor cell line, with IC50 values ranging from 22.53 µM to 50.18 µM, and showed low cytotoxicity against normal L929 fibroblast cells. Flow cytometric analysis revealed that the [Ru(gly)(bipy)(dppb)]PF6 complex (2) inhibited the growth of the tumor cells by inducing apoptosis, as evidenced by an increased number of Annexin V-positive cells and G0/G1 phase cell cycle arrest. Further investigation showed that complex 2 caused a loss of mitochondrial membrane potential; activated caspases 3, caspase-8, and caspase-9 and caused a change in the mRNA expression levels of caspase 3, caspase-9 as well as the bax genes. The levels of the pro-apoptotic Bcl-2 family protein Bak were increased. Thus, we demonstrated that ruthenium amino acid complexes are promising drugs against S180 tumor cells, and we recommend further investigations of their role as chemotherapeutic agents for sarcomas.


Asunto(s)
Antineoplásicos/toxicidad , Apoptosis , Compuestos de Rutenio/toxicidad , Sarcoma/metabolismo , Aminoácidos/química , Animales , Antineoplásicos/síntesis química , Línea Celular Tumoral , Ratones , Rutenio/química , Compuestos de Rutenio/síntesis química
9.
Springerplus ; 3: 301, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25019046

RESUMEN

Chemotherapy is a common treatment for leukemia. Ruthenium complexes have shown potential utility in chemotherapy and photodynamic therapy. The identification of new chemotherapeutics agents is critical for further progress in the treatment of leukemia. Ruthenium complexes generally have lower toxicities compared to cisplatin attributed to their specific accumulation in cancer tissues. Based on these evidences, in the present work we studied the cytotoxic activity of the ruthenium(III) compound cis-tetraammine(oxalato)ruthenium(III) dithionate - {cis-[Ru(C2O4)(NH3)4]2(S2O6)} against human chronic myelogenous leukemia cells (K-562) tumor cell line. The tested compound induces cell death in a dose and time dependent manner on K-562 cells. It is found that the effect was improved linearly while prolonging the incubation time. Compared to the cell cycle profiles of untreated cells, flow cytometric analysis indicated the sub-G1 arresting effect of ruthenium compound on K-562 cells. In our study, {cis-[Ru(C2O4)(NH3)4]2(S2O6)} shows a significant increase in tailed cells in any of the concentrations tested compared with negative control. Consequently, the concentration of {cis-[Ru(C2O4)(NH3)4]2(S2O6)} might be associated cytotoxicity with direct effect on K-562 cells DNA. Thus, it can be deducted that ruthenium-based compounds present selectivity to enter both tumor and normal cells. Additional studies are needed to determine the molecular mechanisms of the active components and to evaluate the potential in vivo anticancer activity of the cis-tetraammine(oxalato)ruthenium(III) dithionate.

10.
Biol Trace Elem Res ; 147(1-3): 8-15, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22144016

RESUMEN

Lung cancer is one of the leading causes of death in the world, and non-small cell lung carcinoma (NSCLC) accounts for approximately 75-85% of all lung cancers. In the present work, we studied the cytotoxic activity, cell cycle arrest and induction apoptosis of the compound cis-(dichloro)tetramineruthenium(III) chloride {cis-[RuCl(2)(NH(3))(4)]Cl} in human lung carcinoma tumor cell line A549. The results of MTT and trypan blue assays showed that cis-[RuCl(2)(NH(3))(4)]Cl causes reduction in the viability of A549 cells when treating with 95 and 383 µM of the compound for 48 and 72 h. Lower concentrations of the compound (19, 3.8 and 0.38 µM), however, only slightly affected cell viability. The IC(50) value for the compound was about 383 µM. Survival analysis of the A549 cells after treatment with ruthenium(III) compound using long term clonogenic assay showed that it reduced colony formation ability at concentrations of 0.38 and 3.8 µM, and at concentrations of 95 and 383 µM no colonies were observed. Cell cycle analysis showed that compound ruthenium led to an accumulation of A549 cells in S phase and increased in the sub-G1 peak. In addition, cis-(dichloro)tetramineruthenium(III) chloride treatment induced apoptosis, as observed by the increased numbers of annexin V-positive cells and increased messenger RNA expression of caspase-3.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Compuestos de Rutenio/farmacología , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Fase G1/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Estructura Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Compuestos de Rutenio/química , Fase S/efectos de los fármacos
11.
J Biosci ; 35(3): 371-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20826946

RESUMEN

Ruthenium (III) complexes are increasingly attracting the interest of researchers due to their promising pharmacological properties. Recently, we reported that the cis-(dichloro)tetrammineruthenium (III) chloride compound has cytotoxic effects on murine sarcoma 180 (S-180) cells. In an effort to understand the mechanism responsible for their cytotoxicity, study we investigated the genotoxicity, cell cycle distribution and induction of apoptosis caused by cis- (dichloro) tetrammineruthenium (III) chloride in S-180 tumour cells. cis-(dichloro) tetrammineruthenium (III) chloride treatment induced significant DNA damage in S-180 cells, as detected by the alkaline comet assay. In the cell cycle analysis, cis-(dichloro) tetrammineruthenium (III) chloride caused an increase in the number of cells in G1 phase, accompanied by a decrease in the S and G2 phases after 24 h of treatment. In contrast, the cell cycle distribution of S-180 cells treated with cis-(dichloro) tetrammineruthenium (III) chloride for 48 h showed a concentration-dependent increase in the sub-G1 phase (indicating apoptosis), with a corresponding decrease in cells in the G1, S and G2 phases. In addition, cis-(dichloro) tetrammineruthenium(III) chloride treatment induced apoptosis in a time-dependent manner,as observed by the increased numbers of annexin V-positive cells. Taken together, these findings strongly demonstrate that DNA damage, cell cycle changes and apoptosis may correlate with the cytotoxic effects of cis-(dichloro) tetrammineruthenium (III) chloride on S-180 cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Daño del ADN , Compuestos Organometálicos/farmacología , Rutenio/farmacología , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Ratones , Compuestos de Rutenio/farmacología
12.
Arq. bras. oftalmol ; 78(2): 89-93, Mar-Apr/2015. graf
Artículo en Inglés | LILACS | ID: lil-744287

RESUMEN

Purpose: To assess the cytotoxicity and genotoxicity of intravitreal adalimumab treatment in an animal experimental model using cytological and molecular techniques. Methods: Eighteen rabbits were randomly assigned to three groups: control, adalimumab treatment, and placebo. Cytotoxicity on retinal cells was evaluated using flow cytometry assays to determine the level of apoptosis and necrosis. Genotoxicity was evaluated by comet assays to assess DNA damage, and quantitative real-time polymerase chain reaction (qPCR) was used to evaluate expression of apoptosis-inducing caspases (8 and 3). Results: No cytotoxicity or genotoxicity was observed in any of the two treatment groups (adalimumab and placebo) following intravitreal administration compared with the control group. Flow cytometry analysis revealed that more than 90% of the cells were viable, and only a low proportion of retinal cells presented apoptotic (~10%) or necrotic (<1%) activity across all groups. Molecular damage was also low with a maximum of 6.4% DNA degradation observed in the comet assays. In addition, no increase in gene expression of apoptosis-inducing caspases was observed on retinal cells by qPCR in both the adalimumab and placebo groups compared with the control group. Conclusion: The use of adalimumab resulted in no detectable cytotoxicity or genotoxicity on retinal cells for up to 60 days upon administration. These results therefore indicate that adalimumab may be a safe option for intravitreal application to treat ocular inflammatory diseases in which TNF-α is involved. .


Objetivo: Acessar a citotoxicidade e genotoxicidade do tratamento intravítreo de adalimumabe em um modelo experimental animal utilizando técnicas citológicas e moleculares. Métodos: Dezoito coelhos foram aleatoriamente selecionados em três grupos: controle, tratamento intravítreo com adalimumabe e placebo. Os efeitos tóxicos nas células da retina foram avaliados através de ensaios de citometria de fluxo, para a determinação de atividade apoptótica e necrótica. A genotoxidade foi avaliada através de ensaios cometa para determinar danos ao DNA e através de PCR em tempo real para avaliar a expressão genética de caspases (8 e 3) promotoras de apoptose celular. Resultados: Não foram detectadas citotoxicidade e genotoxidade nos dois grupos de tratamento, adalimumabe e placebo, em comparação com o controle. A citometria de fluxo determinou que mais de 90% das células eram viáveis após o tratamento, e uma pequena quantidade de células da retina apresentaram apoptose (~10%) ou necrose (<1%) em todos os grupos. O dano molecular também foi baixo com uma degradação no DNA de no máximo 6,4% detectados nos ensaios cometa. Adicionalmente, não foram observados aumentos na expressão genética das caspases que induzem a apoptose através dos ensaios de PCR em tempo real. Conclusão: O tratamento intravítreo com adalimumabe não promoveu nenhuma citotoxicidade e genotoxicidade detectável em células da retina por até sessenta dias. Estes resultados, portanto, indicam que o adalimumabe pode ser uma opção segura para o tratamento de doenças oculares inflamatórias em que o TNFα está envolvido. .


Asunto(s)
Humanos , Infecciones Relacionadas con Catéteres/prevención & control , Infección Hospitalaria/prevención & control , Control de Infecciones/métodos , Sistemas de Registros Médicos Computarizados , Vigilancia de la Población/métodos , Infecciones Urinarias/prevención & control , Infecciones Relacionadas con Catéteres/epidemiología , Infección Hospitalaria/epidemiología , Hospitales/estadística & datos numéricos , Distribución de Poisson , Evaluación de Programas y Proyectos de Salud , Pennsylvania/epidemiología , Análisis de Regresión , Estudios Retrospectivos , Infecciones Urinarias/epidemiología , Infecciones Urinarias/etiología
13.
Toxicol In Vitro ; 24(6): 1562-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20600797

RESUMEN

Ruthenium(III) complexes are increasingly attracting the interest of researchers due to their promising pharmacological properties. In the present study, we investigated the ability of cis-(dichloro)tetrammineruthenium(III) chloride to produce lethal effects in human chronic myelogenous leukemia K562 cells. The MTT tetrazolium reduction test and the trypan blue exclusion assay revealed that the IC(50) for the compound after 48 h of incubation with K562 cells was approximately 10.74 and 73.45 microM, respectively. Interestingly, it was observed that this compound exhibits mild cytotoxicity towards MRC-5 human fibroblast cells (IC(50)>383 microM). Flow cytometric analysis revealed that cis-(dichloro)tetrammineruthenium(III) chloride was capable of change cell cycle distribution since the percentage of cells in the G1, S and G2 phases decreased. In addition, treatment with this compound induced apoptotic cell death in K562 cells, demonstrated by increased DNA content in the sub-G1-peak and a significant increase in caspase-3 activity. Assay using cyclosporin A, an inhibitor of the mitochondrial permeability transition pore (MPT) showed that the preincubation of K562 cells with this inhibitor had not effect on cis-(dichloro)tetrammineruthenium(III) chloride induced caspase-3 activation. In summary, cis-(dichloro)tetrammineruthenium(III) chloride displayed a significant cytotoxic effect through cell cycle arrest and apoptotic induction in K562 cells, which suggests that cis-(dichloro)tetrammineruthenium(III) chloride might have therapeutic potential against leukemia.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Compuestos de Rutenio/farmacología , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclosporina/farmacología , Humanos , Células K562 , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo , Azul de Tripano/metabolismo , Ensayo de Tumor de Célula Madre
14.
Biol Trace Elem Res ; 128(3): 258-68, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19020813

RESUMEN

Ruthenium complexes have attracted much attention as possible building blocks for new transition-metal-based antitumor agents. The present study examines the mitotoxic and clastogenic effects induced in the root tips of Allium cepa by cis-tetraammine(oxalato)ruthenium(III) dithionate {cis-[Ru(C(2)O(2))(NH(3))(4)](2)(S(2)O(6))} at different exposure durations and concentrations. Correlation tests were performed to determine the effects of the time of exposure and concentration of ruthenium complex on mitotic index (MI) and mitotic aberration index. A comparison of MI results of cis-[Ru(C(2)O(2))(NH(3))(4)](2)(S(2)O(6)) to those of lead nitrate reveals that the ruthenium complex demonstrates an average mitotic inhibition eightfold higher than lead, with the frequency of cellular abnormalities almost fourfold lower and mitotic aberration threefold lower. A. cepa root cells exposed to a range of ruthenium complex concentrations did not display significant clastogenic effects. Cis-tetraammine(oxalato)ruthenium(III) dithionate therefore exhibits a remarkable capacity to inhibit mitosis, perhaps by inhibiting DNA synthesis or blocking the cell cycle in the G2 phase. Further investigation of the mechanisms of action of this ruthenium complex will be important to define its clinical potential and to contribute to a novel and rational approach to developing a new metal-based drug with antitumor properties complementary to those exhibited by the drugs already in clinical use.


Asunto(s)
Allium/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Aberraciones Cromosómicas/efectos de los fármacos , Meristema/efectos de los fármacos , Compuestos Organometálicos/farmacología , Raíces de Plantas/efectos de los fármacos , Compuestos de Rutenio/farmacología , Allium/citología , Allium/genética , Relación Dosis-Respuesta a Droga , Plomo/farmacología , Meristema/citología , Meristema/genética , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Micronúcleos con Defecto Cromosómico/estadística & datos numéricos , Mitógenos/farmacología , Índice Mitótico , Nitratos/farmacología , Raíces de Plantas/citología , Raíces de Plantas/genética
15.
Biol Trace Elem Res ; 130(3): 249-61, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19214395

RESUMEN

Chemotherapeutic agents play an important role in cancer treatment mostly due their systemic action on human organism allowing access to liquid tumors and even metastases. Among these drugs, ruthenium compounds have been showing promising results to treat tumors and represent an important development of new antitumor therapy. This study presents the evaluation of cis-(dichloro)tetraammineruthenium(III) chloride, cis-[RuCl(2)(NH(3))(4)]Cl, genotoxic effects using human peripheral blood lymphocytes cultured in vitro. Mitotic index (MI), chromosome aberrations (CA), and DNA damage using the comet assay were analyzed. MI in human peripheral blood lymphocyte cultures treated with 1, 10, 100, and 1,000 microg mL(-1) cis-[RuCl(2)(NH(3))(4)]Cl were 5.9%, 4.6%, 3.9%, and 0%, respectively. Doxorubicin chloridate was used as the positive control. CA derived from 1, 10, and 100 microg mL(-1) concentrations were defined as spontaneous when compared with the negative control, and at the concentration of 1,000 microg mL(-1), the cell cycle was inhibited (IM = 0%). Results obtained for the comet assay using cis-[RuCl(2)(NH(3))(4)]Cl suggest that this compound has no genotoxic activity against cultured human peripheral blood lymphocytes.


Asunto(s)
Antineoplásicos/toxicidad , Linfocitos/efectos de los fármacos , Pruebas de Mutagenicidad , Compuestos de Rutenio/toxicidad , Adulto , Antineoplásicos/administración & dosificación , Ciclo Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Aberraciones Cromosómicas/efectos de los fármacos , Ensayo Cometa , Daño del ADN/efectos de los fármacos , Humanos , Linfocitos/citología , Metafase/efectos de los fármacos , Índice Mitótico , Compuestos de Rutenio/administración & dosificación , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA