Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Inorg Biochem ; 222: 111497, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34090039

RESUMEN

The use of natural products as potential ligands has been explored as a strategy in the development of metal-based chemotherapy. Since ruthenium complexes are promising alternatives to traditional antitumor agents, this study evaluated the anti-melanoma potential of two ruthenium(II) complexes containing the naphthoquinone ligands lapachol (lap), [Ru(lap)(dppm)2]PF6, and lawsone (law), [Ru(law)(dppm)2]PF6, in addition to the bis(diphenylphosphino)methane (dppm) ligand, referred to as complexes (1) and (2), respectively, using a syngeneic murine melanoma model. Activation of the apoptotic pathway by the treatments was assessed by immunohistochemistry in tumor tissue. Additionally, toxicity of the treatments was evaluated by variation in body and organ weight, quantification of biochemical indicators of renal damage, and genotoxicity in bone marrow and hepatocytes. First, the antiproliferative activity of (1) and (2) was observed in B16F10 cells, with IC50 values of 2.78 and 1.68 µM, respectively. The results obtained in mice showed that, unlike complex (1), (2) possesses significant anti-melanoma activity demonstrated by a reduction in tumor volume and mass (88.42%), as well as in mitosis frequency (83.86%). Additionally, complex (2) increased the levels of cleaved caspase-3, inducing tumor cell apoptosis. When compared to the metallodrug cisplatin, complex (2) exhibited similar anti-melanoma activity and lower toxicity considering all parameters evaluated. In silico studies demonstrated no difference in the binding energy of the naphthoquinone complex between complexes (1) and (2). However, the complex containing the lawsone ligand has a lower molar volume, which may be important for interactions with minor DNA grooves. The present results demonstrate the antitumor efficiency of complex (2) and a significantly lower systemic toxicity compared to cisplatin.


Asunto(s)
Antineoplásicos/uso terapéutico , Complejos de Coordinación/uso terapéutico , Melanoma/tratamiento farmacológico , Naftoquinonas/uso terapéutico , Fosfinas/uso terapéutico , Animales , Antineoplásicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/toxicidad , Ligandos , Masculino , Ratones Endogámicos C57BL , Naftoquinonas/toxicidad , Fosfinas/toxicidad , Rutenio/química , Rutenio/toxicidad
2.
Toxicol Sci ; 182(1): 29-43, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-33822233

RESUMEN

Ruthenium is popular as a metal core for chemotherapeutics, due to versatile molecular coordination. Because new metallodrugs are synthesized at high rates, our studies included assays in zebrafish to expedite the initial evaluation as anticancer agents. Here we evaluated novel metallodrugs (PMC79 and LCR134), and cisplatin, a widely used platinum-based chemotherapeutic. We hypothesized that this model could characterize anticancer properties and recapitulate previous in vitro results in vivo. Our findings suggest anticancer properties of PMC79 and LCR134 were similar with less toxicity than cisplatin. Exposures from 24 to 72 h at or below the LOAELs of PMC79 and LCR134 (3.9 µM and 13.5 µm, respectively), impaired blood vessel development and tailfin regeneration. Blood vessel examination through live imaging of larvae revealed distinct regional antiangiogenic impacts. The significant decrease in gene expression of the VEGF-HIF pathway and beta-actin could explain the morphological effects observed in the whole organism following exposure. Tailfin amputation in larvae exposed to PMC79 or LCR134 inhibited tissue regrowth and cell division, but did not impact normal cell proliferation unlike cisplatin. This suggests Ru drugs may be more selective in targeting cancerous cells than cisplatin. Additionally, in vitro mechanisms were confirmed. PMC79 disrupted cytoskeleton formation in larvae and P-glycoprotein transporters in vivo was inhibited at low doses which could limit off-target effects of chemotherapeutics. Our results demonstrate the value for using the zebrafish in metallodrug research to evaluate mechanisms and off-target effects. In light of the findings reported in this article, future investigation of PMC79 and LCR134 are warranted in higher vertebrate models.


Asunto(s)
Antineoplásicos , Rutenio , Animales , Antineoplásicos/toxicidad , Proliferación Celular , Cisplatino/toxicidad , Rutenio/toxicidad , Pez Cebra
3.
J Biol Inorg Chem ; 26(1): 43-55, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33221954

RESUMEN

Ruthenium complexes have been recently reported as potential chemotherapeutic agents that offer tumor selectivity and low tumor resistance. This study investigates the photochemistry and the effect of four strained photoactivatable polypyridyl ruthenium(II) complexes on non-small-cell lung cancer (A549) and triple negative breast cancer (MDA-MB-231) cells. All four ruthenium(II) complexes, [Ru(bpy)2dmbpy]Cl2 (C1) where (bpy = 2,2'-bipyridine and dmbpy = 6,6'-dimethyl-2,2'-bipyridine), [Ru(phen)2dmbpy]Cl2 (C2) where (phen = 1,10-phenanthroline), [Ru(dpphen)2dmbpy]Cl2 (C3) (where dpphen = 4,7-diphenyl-1,10-phenanthroline) and [Ru(BPS)2dmbpy]Na2 (C4) where (BPS = bathophenanthroline disulfonate) eject the dmbpy ligand upon activation by blue light. Determination of the octanol-water partition coefficient (log P) revealed that C3 was the only lipophilic complex (log P = 0.42). LC-MS/MS studies showed that C3 presented the highest cellular uptake. The cytotoxic effect of the complexes was evaluated with and without blue light activation using WST-1 kit. Data indicated that C3 exhibited the highest cytotoxicity after 72 h (MDA-MB-231, IC50 = 0.73 µM; A549, IC50 = 1.26 µM) of treatment. The phototoxicity indices of C3 were 6.56 and 4.64 for MDA-MB-230 and A549, respectively. Upon light activation, C3 caused significant ROS production and induced apoptosis in MDA-MB-231 cells as shown by flow cytometry. It also significantly increased Bax/Bcl2 ratio and PERK levels without affecting caspase-3 expression. C3 exhibited poor dark toxicity (IC50 = 74 µM) on rat mesenchymal stem cells (MSCs). In conclusion, the physical property of the complexes dictated by the variable ancillary ligands influenced cellular uptake and cytotoxicity. C3 may be considered a promising selective photoactivatable chemotherapeutic agent that induces ROS production and apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Complejos de Coordinación/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Fármacos Fotosensibilizantes/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/efectos de la radiación , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/efectos de la radiación , Complejos de Coordinación/toxicidad , Humanos , Luz , Células Madre Mesenquimatosas/efectos de los fármacos , Fármacos Fotosensibilizantes/efectos de la radiación , Fármacos Fotosensibilizantes/toxicidad , Piridinas/farmacología , Piridinas/efectos de la radiación , Piridinas/toxicidad , Ratas , Especies Reactivas de Oxígeno/metabolismo , Rutenio/química , Rutenio/toxicidad
4.
Chemosphere ; 270: 128650, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33131730

RESUMEN

In the present study, a facile method was used to synthesize the ruthenium and ruthenium oxide (RuO2) nanoparticles (NPs) derived from three different metallosurfactants. Firstly, three metallosurfactants were fabricated i.e. RuCTAC (Bishexadecyltrimethylammonium ruthenium tetrachloride), RuDDA (Bisdodecylamine ruthenium dichloride), and RuHEXA (Bishexadecylamine ruthenium dichloride) and characterized by CHN, FTIR, and 1HNMR. These metallosurfactants were further utilized to fabricate the mixed type of NPs (Ru and RuO2 NPs) using the biocompatible microemulsion technique and NPs were then characterized. Subsequently, the nanotoxicity of mixed NPs (Ru & RuO2) was studied towards Saccharomyces cerevisiae. The detailed study of nanotoxicity against the S. cerevisiae cells was done by employing optical microscopy, FESEM, anti-yeast activity assay, circular dichroism, and gel electrophoresis techniques. FESEM and optical microscopy analyses indicated that RuCTAC nanosuspension (Ns) has the most toxic effect on the S. cerevisiae cells. FESEM analysis confirmed the harmful impact of Ru and RuO2 NPs on the S. cerevisiae cells. From the FESEM analysis, complete alteration in the morphology, cell membrane breakage, and formation of the holes on the cell wall of S. cerevisiae was affirmed in presence of all three types of Ns i.e. RuCTAC, RuDDA, and RuHEXA Ns. Genotoxicity of the NPs was confirmed by circular dichroism and gel electrophoresis and it was found that RuCTAC and RuHEXA Ns have the most damaging influence on the yeast genomic DNA.


Asunto(s)
Nanopartículas , Rutenio , Daño del ADN , Óxidos , Rutenio/toxicidad , Saccharomyces cerevisiae/genética
6.
Metallomics ; 12(6): 876-890, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32329475

RESUMEN

Like platinum-based chemotherapeutics, the anticancer ruthenium complex indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(iii)], or KP1019, damages DNA, induces apoptosis, and causes tumor regression in animal models. Unlike platinum-based drugs, KP1019 showed no dose-limiting toxicity in a phase I clinical trial. Despite these advances, the mechanism(s) and target(s) of KP1019 remain unclear. For example, the drug may damage DNA directly or by causing oxidative stress. Likewise, KP1019 binds cytosolic proteins, suggesting DNA is not the sole target. Here we use the budding yeast Saccharomyces cerevisiae as a model in a proteomic study of the cellular response to KP1019. Mapping protein level changes onto metabolic pathways revealed patterns consistent with elevated synthesis and/or cycling of the antioxidant glutathione, suggesting KP1019 induces oxidative stress. This result was supported by increased fluorescence of the redox-sensitive dye DCFH-DA and increased KP1019 sensitivity of yeast lacking Yap1, a master regulator of the oxidative stress response. In addition to oxidative and DNA stress, bioinformatic analysis revealed drug-dependent increases in proteins involved ribosome biogenesis, translation, and protein (re)folding. Consistent with proteotoxic effects, KP1019 increased expression of a heat-shock element (HSE) lacZ reporter. KP1019 pre-treatment also sensitized yeast to oxaliplatin, paralleling prior research showing that cancer cell lines with elevated levels of translation machinery are hypersensitive to oxaliplatin. Combined, these data suggest that one of KP1019's many targets may be protein metabolism, which opens up intriguing possibilities for combination therapy.


Asunto(s)
Proteómica/métodos , Rutenio/toxicidad , Saccharomyces cerevisiae/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Indazoles/farmacología , Compuestos Organometálicos/farmacología , Oxaliplatino/farmacología , Compuestos de Rutenio/farmacología , Saccharomyces cerevisiae/efectos de los fármacos
7.
J Appl Toxicol ; 39(8): 1173-1180, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30963621

RESUMEN

As novel metallodrugs continue to emerge, they are evaluated using models, including zebrafish, that offer unique sublethal endpoints. Testing metal-based anticancer compounds with high-throughput zebrafish toxicological assays requires analytical methods with the sensitivity to detect these sublethal tissue doses in very small sample masses (e.g., egg mass 100 µg). A robust bioanalytical model, zebrafish embryos coupled with inductively coupled plasma-mass spectrometry (ICPMS) for measurement of delivered dose, creates a very effective means for screening metal-based chemotherapeutic agents. In this study, we used ICPMS quantitation with the zebrafish embryo assays to detect metal equivalents at multiple response endpoints for two compounds, the chemotherapeutic agent cisplatin and ruthenium (Ru)-based prospective metallodrug, PMC79. We hypothesized that cisplatin and PMC79 have different mechanisms for inducing apoptosis and result in similar lesions but different potencies following water-borne exposure. An ICPMS method was developed to detect the metal in waterborne solution and tissue (detection limit: 5 parts per trillion for Ru or platinum [Pt]). The Ru-based compound was more potent (LC50 : 7.8 µm) than cisplatin (LC50 : 158 µm) and induced disparate lesions. Lethality from cisplatin exposure exhibited a threshold (values >15 mg/L) while no threshold was observed for delayed hatching (lowest observed adverse effect level 3.75 mg/L cisplatin; 8.7 Pt (ng)/organism). The Ru organometallic did not have a threshold for lethality. Cisplatin-induced delayed hatching was investigated further by larval-Pt distribution and preferentially distributed to the chorion. We propose that zebrafish embryo-larval assays coupled with ICPMS serve as a powerful platform to evaluate relative potency and toxic effects of metallodrug candidates.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Embrión no Mamífero/efectos de los fármacos , Larva/efectos de los fármacos , Compuestos Organometálicos/toxicidad , Rutenio/toxicidad , Pez Cebra , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Bioensayo , Cisplatino/química , Relación Dosis-Respuesta a Droga , Embrión no Mamífero/patología , Desarrollo Embrionario/efectos de los fármacos , Compuestos Organometálicos/química , Rutenio/química , Espectrofotometría Atómica
8.
ACS Sens ; 4(4): 984-991, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30859818

RESUMEN

Metal complex-based luminescent oxygen nanosensors have been intensively studied for biomedical applications. In terms of monitoring dynamics of intracellular oxygen, however, high-quality nanosensors are still badly needed, because of stringent requirements on stability, biocompatibility and luminescence intensity, aside from oxygen sensitivity. In this paper, we reported a type of highly luminescent and stable oxygen nanosensors prepared from metallopolymer. First, a novel ruthenium(II)-containing metallopolymer was synthesized by chelating the oxygen probe [Ru(bpy)3]2+ with a bipyridine-branched hydrophobic copolymer, which was then doped into polymeric nanoparticles (NPs) by a reprecipitation method, followed by further conjugation to selectively target mitochondria (Mito-NPs). The resultant Mtio-NPs possessed a small hydrodynamic size of ∼85 nm, good biocompatibility and high stability resulting from PEGylation and stable nature of Ru-complex. Because the complexed [Ru(bpy)3]2+ homogeneously resided on particle surface, Mito-NPs exhibited strong luminescence at 608 nm that was free of aggregation-caused-quenching, the utmost oxygen sensitivity of free [Ru(bpy)3]2+ probe ( Q = 75%), and linear Stern-Volmer oxygen luminescence quenching plots. Taking advantage of the mitochondria-specific nanosensors, intracellular oxygenation and deoxygenation processes were real-time monitored for 10 min by confocal luminescence imaging, visualized by the gradual weakening (by more than 90%) and enhancing (by 50%) of the red emission, respectively.


Asunto(s)
Resinas Acrílicas/química , Sustancias Luminiscentes/química , Mitocondrias/metabolismo , Nanopartículas/química , Oxígeno/análisis , Poliestirenos/química , Resinas Acrílicas/síntesis química , Resinas Acrílicas/toxicidad , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/toxicidad , Células Hep G2 , Humanos , Luminiscencia , Sustancias Luminiscentes/síntesis química , Sustancias Luminiscentes/toxicidad , Mediciones Luminiscentes/métodos , Microscopía Confocal/métodos , Nanopartículas/toxicidad , Poliestirenos/síntesis química , Poliestirenos/toxicidad , Prueba de Estudio Conceptual , Rutenio/química , Rutenio/toxicidad
9.
Toxicol In Vitro ; 50: 201-209, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29580987

RESUMEN

Inhaled carbon monoxide (CO) gas is highly toxic, but the human body produces low levels of CO for vasoregulation and other purposes. Given the established protective roles of low concentrations of CO gas against a panel of pathological insults, CO-releasing molecules (CORMs) have been developed and examined in disease models both in vitro and in vivo. Among CORMs, CORM-3 [Ru(CO)3Cl(glycinate)], a ruthenium carbonyl compound, has been extensively studied since it is water-soluble and is suitable for in vivo application. As one of the most prominent features of CO gas is its anti-fibrotic effect, we examined the effects of CORM-3 on mouse embryonic fibroblasts (MEFs). The application of 1 mM CORM-3 to MEFs resulted in the decreased syntheses of collagens I and III within 24 h, confirming an anti-fibrotic effect. To our surprise, CORM-3 caused a rapid (within 1 h) dissociation of cell-associated plasma fibronectin (FN) from the cells, which is associated with formation of a reduction-resistant oligomer of plasma FN. This aberrant oligomerization of plasma FN was reproduced using purified FN in vitro. Furthermore, we showed that RuCl3, but not another water-soluble CORM, CORM-A1 [Na2H3BCO2], also oligomerized plasma FN in vitro. FN depletion from the serum substantially ameliorates cell death by prolonged (72 h) exposure to CORM-3, suggesting a detrimental role of FN oligomerization on cell death. Taken together, we reveal for the first time that FN is a CORM-3-interactive plasma protein, and that the CORM-3-FN interaction is involved in the death of fibroblasts.


Asunto(s)
Monóxido de Carbono/química , Fibroblastos/efectos de los fármacos , Fibronectinas/sangre , Compuestos Organometálicos/toxicidad , Rutenio/toxicidad , Animales , Proteína 5 Relacionada con la Autofagia/genética , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Ratones Noqueados
10.
Metallomics ; 10(1): 194-200, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29296993

RESUMEN

Immature forms of the peptide hormone gastrin have been implicated in the development of colorectal cancer (CRC). The biological activity of glycine-extended gastrin (Ggly) is dependent on the binding of Fe3+ ions in vitro and in vivo. The aim of the present study was to determine the effect of blocking Fe3+ ion binding to Ggly, using Bi3+, In3+ or Ru3+ ions, on the development of intestinal tumors in APCΔ14/+ mice. APCΔ14/+ mice were treated orally with Bi3+, In3+ or Ru3+ ions for up to 60 days, serum trace metals were analyzed by inductively coupled plasma mass spectrometry, and the incidence and size of intestinal tumors were assessed. Bi3+ treatment significantly decreased the number of tumors larger than 3 mm in male mice. In3+ or Ru3+ treatment significantly increased the tumor burden in all animals and In3+ increased the number of tumors larger than 3 mm or 5 mm in male mice alone. The fact that binding of In3+ or Ru3+ ions to Ggly was orders of magnitude stronger than the binding of Bi3+ ions implies that the inhibitory effect of Bi3+ ions is not a consequence of a reduction in Ggly activity. However, further testing of higher doses of Bi3+ ions for longer periods as an oral treatment for intestinal tumors is warranted.


Asunto(s)
Bismuto/farmacología , Indio/toxicidad , Neoplasias Intestinales/inducido químicamente , Neoplasias Intestinales/tratamiento farmacológico , Rutenio/toxicidad , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Bismuto/química , Exones , Pruebas Hematológicas , Indio/química , Neoplasias Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Mutación Puntual , Rutenio/química , Carga Tumoral
11.
PLoS One ; 12(9): e0183275, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28898246

RESUMEN

Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype. The high rate of metastasis associated to the fact that these cells frequently display multidrug resistance, make the treatment of metastatic disease difficult. Development of antitumor metal-based drugs was started with the discovery of cisplatin, however, the severe side effects represent a limitation for its clinical use. Ruthenium (Ru) complexes with different ligands have been successfully studied as prospective antitumor drugs. In this work, we demonstrated the activity of a series of biphosphine bipyridine Ru complexes (1) [Ru(SO4)(dppb)(bipy)], (2) [Ru(CO3)(dppb)(bipy)], (3) [Ru(C2O4)(dppb)(bipy)] and (4) [Ru(CH3CO2)(dppb)(bipy)]PF6 [where dppb = 1,4-bis(diphenylphosphino)butane and bipy = 2,2'-bipyridine], on proliferation of TNBC (MDA-MB-231), estrogen-dependent breast tumor cells (MCF-7) and a non-tumor breast cell line (MCF-10A). Complex (4) was most effective among the complexes and was selected to be further investigated on effects on tumor cell adhesion, migration, invasion and in apoptosis. Moreover, DNA and HSA binding properties of this complex were also investigated. Results show that complex (4) was more efficient inhibiting proliferation of MDA-MB-231 cells over non-tumor cells. In addition, complex (4) was able to inhibit MDA-MB231 cells adhesion, migration and invasion and to induce apoptosis and inhibit MMP-9 secretion in TNBC cells. Complex (4) should be further investigated in vivo in order to stablish its potential to improve breast cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Rutenio/farmacología , Antineoplásicos/química , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/toxicidad , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Molecular , Rutenio/química , Rutenio/toxicidad , Neoplasias de la Mama Triple Negativas , Ensayo de Tumor de Célula Madre
12.
Eur J Med Chem ; 142: 8-31, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-28442170

RESUMEN

Metallodrugs offer potential for unique mechanism of drug action based on the choice of the metal, its oxidation state, the types and number of coordinated ligands and the coordination geometry. This review illustrates notable recent progress in the field of medicinal bioinorganic chemistry as many new approaches to the design of innovative metal-based anticancer drugs are emerging. Current research addressing the problems associated with platinum drugs has focused on other metal-based therapeutics that have different modes of action and on prodrug and targeting strategies in an effort to diminish the side-effects of cisplatin chemotherapy. Examples of metal compounds and chelating agents currently in clinical use, clinical trials or preclinical development are highlighted.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Oro/farmacología , Neoplasias/tratamiento farmacológico , Paladio/farmacología , Platino (Metal)/farmacología , Rutenio/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Complejos de Coordinación/química , Complejos de Coordinación/uso terapéutico , Complejos de Coordinación/toxicidad , Descubrimiento de Drogas , Oro/química , Oro/uso terapéutico , Oro/toxicidad , Humanos , Paladio/química , Paladio/uso terapéutico , Paladio/toxicidad , Platino (Metal)/química , Platino (Metal)/uso terapéutico , Platino (Metal)/toxicidad , Rutenio/química , Rutenio/uso terapéutico , Rutenio/toxicidad
13.
Dalton Trans ; 45(47): 19127-19140, 2016 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-27868117

RESUMEN

The limitations of platinum complexes in cancer treatment have motivated the extensive investigation into other metal complexes such as ruthenium. We herein present the synthesis and characterization of a new family of ruthenium compounds 1a-5a with the general formula [Ru(bipy)2L][CF3SO3]2 (bipy = 2,2'-bipyridine; L = bidentate ligand: N,N; N,P; P,P; P,As) which have been characterized by elemental analysis, ES-MS, 1H and 31P-{1H} NMR, FTIR and conductivity measurements. The molecular structures of four Ru(ii) complexes were determined by single crystal X-ray diffraction. All compounds displayed moderate cytotoxic activity in vitro against human A2780 ovarian, MCF7 breast and HCT116 colorectal tumor cells. Compound 5a was the most cytotoxic compound against A2780 and MCF7 tumor cells with an IC50 of 4.75 ± 2.82 µM and 20.02 ± 1.46 µM, respectively. The compounds showed no cytotoxic effect on normal human primary fibroblasts but rather considerable selectivity for A2780, MCF7 and HCT116 tumor cells. All compounds induce apoptosis and autophagy in A2780 ovarian carcinoma cells and some nuclear DNA fragmentation. All compounds interact with CT-DNA with intrinsic binding constants in the order 1a > 4a > 2a > 3a > 5a. The observed hyperchromic effect may be due to the electrostatic interaction between positively charged cations and the negatively charged phosphate backbone at the periphery of the double helix-CT-DNA. Interestingly, compound 1a shows a concentration dependent DNA double strand cleavage. In addition in vivo toxicity has been evaluated on zebrafish embryos unveiling the differential toxicity between the compounds, with LC50 ranging from 8.67 mg L-1 for compound 1a to 170.30 mg L-1 for compound 2a.


Asunto(s)
2,2'-Dipiridil , Antineoplásicos , Complejos de Coordinación , Rutenio , 2,2'-Dipiridil/química , 2,2'-Dipiridil/farmacología , 2,2'-Dipiridil/toxicidad , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/toxicidad , ADN/química , Fragmentación del ADN , Embrión no Mamífero/efectos de los fármacos , Humanos , Dosificación Letal Mediana , Estructura Molecular , Rutenio/química , Rutenio/farmacología , Rutenio/toxicidad , Pez Cebra
14.
Artículo en Inglés | MEDLINE | ID: mdl-26994489

RESUMEN

Tuberculosis is a top infectious disease killer worldwide, caused by the bacteria Mycobacterium tuberculosis. Increasing incidences of multiple drug-resistance (MDR) strains are emerging as one of the major public health threats. However, the drugs in use are still incapable of controlling the appalling upsurge of MDR. In recent years a marked number of research groups have devoted their attention toward the development of specific and cost-effective antimicrobial agents against targeted MDR-Tuberculosis. In previous studies, ruthenium(II) complexes (SCAR) have shown a promising activity against MDR-Tuberculosis although few studies have indeed considered ruthenium toxicity. Therefore, within the preclinical requirements, we have sought to determine the cyto-genotoxicity of three SCAR complexes in this present study. The treatment with the SCARs induced a concentration-dependent decrease in cell viability in CHO-K1 and HepG2 cells. Based on the clonogenic survival, SCAR 5 was found to be more cytotoxic while SCAR 6 exhibited selectivity action on tumor cells. Although SCAR 4 and 5 did not indicate any mutagenic activity as evidenced by the Ames and Cytokinesis block micronucleus cytome assays, the complex SCAR 6 was found to engender a frameshift mutation detected by Salmonella typhimurium in the presence of S9. Similarly, we observed a chromosomal damage in HepG2 cells with significant increases of micronuclei and nucleoplasmic bridges. These data indicate that SCAR 4 and 5 complexes did not show genotoxicity in our models while SCAR 6 was considered mutagenic. This study presented a comprehensive genotoxic evaluation of SCAR complexes were shown to be genotoxic in vitro. All in all, further studies are required to fully elucidate how the properties can affect human health.


Asunto(s)
Antituberculosos/toxicidad , Complejos de Coordinación/toxicidad , Mutágenos/toxicidad , Rutenio/toxicidad , Animales , Células CHO , Cricetulus , Citocinesis/efectos de los fármacos , Células Hep G2 , Humanos , Pruebas de Micronúcleos , Rutenio/química , Salmonella typhimurium/efectos de los fármacos
15.
J Antimicrob Chemother ; 71(6): 1547-55, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26945708

RESUMEN

OBJECTIVES: The objectives of this study were to: (i) determine the in vitro activities of a series of di-, tri- and tetra-nuclear ruthenium complexes (Rubbn, Rubbn-tri and Rubbn-tetra) against a range of Gram-positive and -negative bacteria and compare the antimicrobial activities with the corresponding toxicities against eukaryotic cells; and (ii) compare MIC values with achievable in vivo serum concentrations for the least toxic ruthenium complex. METHODS: The in vitro activities were determined by MIC assays and time-kill curve experiments, while the toxicities of the ruthenium complexes were determined using the Alamar blue cytotoxicity assay. A preliminary pharmacokinetic study was undertaken to determine the Rubb12 serum concentration in mice as a function of time after administration. RESULTS: Rubb12, Rubb12-tri and Rubb12-tetra are highly active, with MIC values of 1-2 mg/L (0.5-1.5 µM) for a range of Gram-positive strains, but showed variable activities against a panel of Gram-negative bacteria. Time-kill experiments indicated that Rubb12, Rubb12-tri and Rubb12-tetra are bactericidal and kill bacteria within 3-8 h. The di-, tri- and tetra-nuclear complexes were ∼50 times more toxic to Gram-positive bacteria and 25 times more toxic to Gram-negative strains, classified as susceptible, than to liver and kidney cells. Preliminary pharmacokinetic experiments established that serum concentrations higher than MIC values can be obtained for Rubb12 with an administered dose of 32 mg/kg. CONCLUSIONS: The ruthenium complexes, particularly Rubb12, have potential as new antimicrobial agents. The structure of the dinuclear ruthenium complex can be readily further modified in order to increase the selectivity for bacteria over eukaryotic cells.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Células Eucariotas/efectos de los fármacos , Compuestos Organometálicos/farmacología , Rutenio/farmacología , Animales , Antibacterianos/farmacocinética , Antibacterianos/toxicidad , Supervivencia Celular/efectos de los fármacos , Colorimetría/métodos , Femenino , Masculino , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Compuestos Organometálicos/farmacocinética , Compuestos Organometálicos/toxicidad , Oxazinas/análisis , Rutenio/farmacocinética , Rutenio/toxicidad , Suero/química , Xantenos/análisis
16.
Int J Biol Macromol ; 82: 663-70, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26424207

RESUMEN

We report on the synthesis of novel Ru(II) compounds (Ru-1 to Ru-8) bearing R-pdc, 4-Cl-pbinh ligands (where R=4-CF3, 4-F, 4-OH pdc=3-phenyl-5-(1H-pyrrol-2-yl)-4,5-dihydro-1H-pyrazole-1-carbothioamide, pbinh=phenoxybenzylidene isonicotinyl hydrazides) and their in vitro antitumor activity toward the cell lines murine leukemia L1210, human lymphocyte CEM, human epithelial cervical carcinoma HeLa, BEL-7402 and Molt4/C8. Some of the complexes exhibited more potent antiproliferative activity against cell lines than the standard drug cisplatin. Ruthenium complex Ru-2 displayed potent cytotoxicity with than that of cisplatin. DNA-binding, DNA cleavage and protein binding properties of ruthenium complexes with these ligands are reported. Interactions of these ruthenium complexes with DNA revealed an intercalative mode of binding between them. Synchronous fluorescence spectra proved that the interaction of ruthenium complexes with bovine serum albumin (BSA) resulted in a conformational change of the latter.


Asunto(s)
ADN/química , ADN/metabolismo , Rutenio/química , Rutenio/metabolismo , Animales , Línea Celular Tumoral , División del ADN , Humanos , Concentración 50 Inhibidora , Ligandos , Modelos Moleculares , Conformación Molecular , Unión Proteica , Rutenio/toxicidad , Albúmina Sérica Bovina/química
17.
Bioelectrochemistry ; 104: 35-43, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25731757

RESUMEN

A ruthenium complex was prepared and structurally characterized using various techniques. Antibacterial and antifungal activities of ruthenium complex were evaluated. High significant antimicrobial activity against Escherichia coli, Staphylococcus aureus and Candida albicans was recorded. Minor cytotoxicity records were reported at the highest concentration level using MTT assay. The influence of Cu(II), Cr(III), Fe(III) and Ru(III) metal ions of salen Schiff base on the corrosion resistance of Ti-alloy in 0.5M HCl was studied. In vitro corrosion resistance was investigated using electrochemical impedance spectroscopy (EIS) measurements and confirmed by surface examination via scanning electron microscope (SEM) technique. Both impedance and phase angle maximum (θ(max)) values were at maximum in the case of the ruthenium complex with promising antibacterial and antifungal activities. The surface film created by the ruthenium complex was highly resistant against attack or deterioration by bacteria. The EIS study showed high impedance values for the ruthenium complex with increasing exposure time up to 8 days. SEM images showed uniform distribution and adsorption of Ru(III) ions on Ti-alloy surface. The ruthenium complex, as a model of organic-inorganic hybrid complex, offered new prospects with desired properties in industrial and medical applications.


Asunto(s)
Aleaciones/química , Antiinfecciosos/química , Antiinfecciosos/farmacología , Rutenio/química , Rutenio/farmacología , Titanio/química , Animales , Antiinfecciosos/toxicidad , Candida albicans/efectos de los fármacos , Corrosión , Impedancia Eléctrica , Escherichia coli/efectos de los fármacos , Células HEK293 , Humanos , Ratones , Rutenio/toxicidad , Staphylococcus aureus/efectos de los fármacos
18.
J Biomed Opt ; 19(7): 77004, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036215

RESUMEN

Measuring tissue oxygenation in vivo is of interest in fundamental biological as well as medical applications. One minimally invasive approach to assess the oxygen partial pressure in tissue (pO2) is to measure the oxygen-dependent luminescence lifetime of molecular probes. The relation between tissue pO2 and the probes' luminescence lifetime is governed by the Stern-Volmer equation. Unfortunately, virtually all oxygen-sensitive probes based on this principle induce some degree of phototoxicity. For that reason, we studied the oxygen sensitivity and phototoxicity of dichlorotris(1, 10-phenanthroline)-ruthenium(II) hydrate [Ru(Phen)] using a dedicated optical fiber­based, time-resolved spectrometer in the chicken embryo chorioallantoic membrane. We demonstrated that, after intravenous injection, Ru(Phen)'s luminescence lifetime presents an easily detectable pO2 dependence at a low drug dose (1 mg∕kg) and low fluence (120 mJ∕cm2 at 470 nm). The phototoxic threshold was found to be at 10 J∕cm2 with the same wavelength and drug dose, i.e., about two orders of magnitude larger than the fluence necessary to perform a pO2 measurement. Finally, an illustrative application of this pO2 measurement approach in a hypoxic tumor environment is presented.


Asunto(s)
Colorantes Fluorescentes/química , Imagen Óptica/métodos , Oxígeno/análisis , Fenantrolinas/química , Rutenio/química , Animales , Línea Celular Tumoral , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Membrana Corioalantoides/química , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/toxicidad , Humanos , Neoplasias Experimentales , Fibras Ópticas , Imagen Óptica/instrumentación , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacocinética , Compuestos Organometálicos/toxicidad , Presión Parcial , Fenantrolinas/farmacocinética , Fenantrolinas/toxicidad , Rutenio/farmacocinética , Rutenio/toxicidad , Distribución Tisular
19.
Genesis ; 52(4): 309-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24616213

RESUMEN

Photodynamic therapy utilizes light, a photosensitizer, and molecular oxygen as a treatment modality for a variety of cancers. We have recently combined ruthenium(II) polypyridyl groups with a zinc(II) centered porphyrin as a new photosensitizer for the treatment of melanoma. In-vitro studies have indicated that this photosensitizer is toxic to melanoma cells when irradiated with low energy light; however, it is nontoxic to normal cells under similar conditions. To determine the toxicity and cell viability of this compound in-vivo we present, herein, a study using Drosophila melanogaster. In the absence of light, the new photosensitizer shows no discernible effects to fly larvae at various concentrations of compound and stages of larval development. When the larvae were fed the photosensitizer it was observed, by fluorescence microscopy, that the compound passes through the cell membrane and localizes in the cytosol at lower concentrations and the nucleus at slightly higher concentrations indicating that the compound is not immediately metabolized.


Asunto(s)
Metaloporfirinas/toxicidad , Fármacos Fotosensibilizantes/toxicidad , Rutenio/toxicidad , Zinc/toxicidad , Animales , Encéfalo/metabolismo , Drosophila melanogaster , Ensayos de Selección de Medicamentos Antitumorales , Larva/efectos de los fármacos , Larva/metabolismo , Metaloporfirinas/farmacocinética , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacocinética , Rutenio/farmacocinética , Zinc/farmacocinética
20.
Chemistry ; 19(9): 3198-203, 2013 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-23344898

RESUMEN

Hexanuclear thiolato-bridged arene ruthenium metalla-prisms of the general formula [(p-cymene)(6)Ru(6)(SR)(6)(tpt)(2) ](6+) (R=CH(2)Ph, CH(2)C(6)H(4)-p-tBu, CH(2)CH(2)Ph; tpt=2,4,6-tris(4-pyridyl)-1,3,5-triazine), obtained from the dinuclear precursors [(p-cymene)(2)Ru(2)(SR)(2)Cl(2)], AgCF(3)SO(3) and tpt, have been isolated and fully characterised as triflate salts. The metalla-prisms are highly cytotoxic against human ovarian cancer cells, especially towards the cisplatin-resistant cell line A2780cisR (IC(50) <0.25 µM).


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/toxicidad , Cisplatino/química , Cisplatino/toxicidad , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/toxicidad , Neoplasias Ováricas/tratamiento farmacológico , Rutenio/química , Rutenio/toxicidad , Antineoplásicos/química , Línea Celular Tumoral , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Compuestos Organometálicos/química , Triazinas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...