Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 233
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(14): 7174-7183, 2023 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-37351627

RESUMEN

DNA three-way junctions (3WJ) represent one of the simplest supramolecular DNA structures arising as intermediates in homologous recombination in the absence of replication. They are also formed transiently during DNA replication. Here we examine the ability of Fe(II)-based metallohelices to act as DNA 3WJ binders and induce DNA damage in cells. We investigated the interaction of eight pairs of enantiomerically pure Fe(II) metallohelices with four different DNA junctions using biophysical and molecular biology methods. The results show that the metallohelices stabilize all types of tested DNA junctions, with the highest selectivity for the Y-shaped 3WJ and minimal selectivity for the 4WJ. The potential of the best stabilizer of DNA junctions and, at the same time, the most selective 3WJ binder investigated in this work to induce DNA damage was determined in human colon cancer HCT116 cells. These metallohelices proved to be efficient in killing cancer cells and triggering DNA damage that could yield therapeutic benefits.


Asunto(s)
ADN , Neoplasias , Humanos , ADN/química , Daño del ADN , Compuestos Ferrosos , Conformación de Ácido Nucleico , Neoplasias/genética
2.
Nucleic Acids Res ; 50(2): 674-683, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35018455

RESUMEN

The design of efficient and safe gene delivery vehicles remains a major challenge for the application of gene therapy. Of the many reported gene delivery systems, metal complexes with high affinity for nucleic acids are emerging as an attractive option. We have discovered that certain metallohelices-optically pure, self-assembling triple-stranded arrays of fully encapsulated Fe-act as nonviral DNA delivery vectors capable of mediating efficient gene transfection. They induce formation of globular DNA particles which protect the DNA from degradation by various restriction endonucleases, are of suitable size and electrostatic potential for efficient membrane transport and are successfully processed by cells. The activity is highly structure-dependent-compact and shorter metallohelix enantiomers are far less efficient than less compact and longer enantiomers.


Asunto(s)
Cationes/química , ADN/química , Técnicas de Transferencia de Gen , Vectores Genéticos , Nanopartículas del Metal/química , Línea Celular , Supervivencia Celular , ADN/ultraestructura , Compuestos Ferrosos/química , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/ultraestructura , Humanos , Nanopartículas del Metal/ultraestructura , Microscopía de Fuerza Atómica/métodos , Estructura Molecular , Transfección
3.
J Biol Inorg Chem ; 28(7): 669-678, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37624480

RESUMEN

(1R,2R-diaminocyclohexane)(dihydropyrophosphato) platinum(II), also abbreviated as RRD2, belongs to a class of potent antitumor platinum cytostatics called phosphaplatins. Curiously, several published studies have suggested significant mechanistic differences between phosphaplatins and conventional platinum antitumor drugs. Controversial findings have been published regarding the role of RRD2 binding to DNA in the mechanism of its antiproliferative activity in cancer cells. This prompted us to perform detailed studies to confirm or rule out the role of RRD2 binding to DNA in its antiproliferative effect in cancer cells. Here, we show that RRD2 exhibits excellent antiproliferative activity in various cancer cell lines, with IC50 values in the low micromolar or submicromolar range. Moreover, the results of this study demonstrate that DNA lesions caused by RRD2 contribute to killing cancer cells treated with this phosphaplatin derivative. Additionally, our data indicate that RRD2 accumulates in cancer cells but to a lesser extent than cisplatin. On the other hand, the efficiency of cisplatin and RRD2, after they accumulate in cancer cells, in binding to nuclear DNA is similar. Our results also show that RRD2 in the medium, in which the cells were cultured before RRD2 accumulated inside the cells, remained intact. This result is consistent with the view that RRD2 is activated by releasing free pyrophosphate only in the environment of cancer cells, thereby allowing RRD2 to bind to nuclear DNA.


Asunto(s)
Antineoplásicos , Neoplasias , Oxaliplatino/farmacología , Cisplatino/farmacología , Platino (Metal)/farmacología , Difosfatos/farmacología , Compuestos Organoplatinos/farmacología , Compuestos Organoplatinos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , ADN/metabolismo
4.
Inorg Chem ; 62(16): 6474-6487, 2023 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-37040203

RESUMEN

We present the synthesis and characterization of six new heteroleptic osmium(II) complexes of the type [Os(C^N)(N^N)2]OTf (N^N = 2,2'-bipyridine and dipyrido[3,2-d:2',3'-f]quinoxaline; C^N = deprotonated methyl 1-butyl-2aryl-benzimidazolecarboxylate) with varying substituents in the R3 position of the phenyl ring of the cyclometalating C^N ligand. The new compounds are highly kinetically inert and absorb a full-wavelength range of visible light. An investigation of the antiproliferative activity of the new compounds has been performed using a panel of human cancer and noncancerous 2D cell monolayer cultures under dark conditions and green light irradiation. The results demonstrate that the new Os(II) complexes are markedly more potent than conventional cisplatin. The promising antiproliferative activity of selected Os(II) complexes was also confirmed using 3D multicellular tumor spheroids, which have the characteristics of solid tumors and can mimic the tumor tissue microenvironment. The mechanism of antiproliferative action of complexes has also been investigated and revealed that the investigated Os(II) complexes activate the endoplasmic reticulum stress pathway in cancer cells and disrupt calcium homeostasis.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Neoplasias , Humanos , Relación Estructura-Actividad , Osmio/farmacología , Calcio , Línea Celular Tumoral , Bencimidazoles/farmacología , Homeostasis , Antineoplásicos/farmacología , Complejos de Coordinación/farmacología
5.
J Enzyme Inhib Med Chem ; 38(1): 2198678, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37019444

RESUMEN

Some metallo-supramolecular helical assemblies with size, shape, charge and amphipathic architectures similar to short cationic α-helical peptides have been shown to target and stabilise DNA G-quadruplexes (G4s) in vitro and downregulate the expression of G4-regulated genes in human cells. To expand the library of metallohelical structures that can act as efficient DNA G4 binders and downregulate genes containing G4-forming sequences in their promoter regions, we investigated the interaction of the two enantiomeric pairs of asymmetric Fe(II) triplex metallohelices with a series of five different DNA G4s formed by the human telomeric sequence (hTelo) and in the promoter regions of c-MYC, c-KIT, and k-RAS oncogenes. The metallohelices display preferential binding to G4s over duplex DNA in all investigated G4-forming sequences and induced arrest of DNA polymerase on template strands containing G4-forming sequences. Moreover, the investigated metallohelices suppressed the expression of c-MYC and k-RAS genes at mRNA and protein levels in HCT116 human cancer cells, as revealed by RT-qPCR analysis and western blotting.


Asunto(s)
G-Cuádruplex , Neoplasias , Humanos , Oncogenes , Regiones Promotoras Genéticas , ADN/química
6.
Molecules ; 28(13)2023 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-37446835

RESUMEN

The purine derivative fludarabine is part of frontline therapy for chronic lymphocytic leukaemia (CLL). It has shown positive effects on solid tumours such as melanoma, breast, and colon carcinoma in clinical phase I studies. As the treatment of CLL cells with combinations of fludarabine and metal complexes of antitumoural natural products, e.g., illudin M ferrocene, has led to synergistically enhanced apoptosis, in this research study different complexes of fludarabine itself. Four complexes bearing a trans-[Br(PPh3)2]Pt/Pd fragment attached to atom C-8 via formal η1-sigma or η2-carbene bonds were synthesised in two or three steps without protecting polar groups on the arabinose or adenine. The platinum complexes were more cytotoxic than their palladium analogues, with low single-digit micromolar IC50 values against cells of various solid tumour entities, including cisplatin-resistant ones and certain B-cell lymphoma and CLL, presumably due to the ten-fold higher cellular uptake of the platinum complexes. However, the palladium complexes interacted more readily with isolated Calf thymus DNA. Interestingly, the platinum complexes showed vastly greater selectivity for cancer over non-malignant cells when compared with fludarabine.


Asunto(s)
Antineoplásicos , Leucemia Linfocítica Crónica de Células B , Humanos , Platino (Metal)/química , Antimetabolitos/uso terapéutico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Paladio/química , Antineoplásicos/química , Inmunosupresores/uso terapéutico
7.
Angew Chem Int Ed Engl ; 62(42): e202310774, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37646232

RESUMEN

A multitargeting prodrug (2) that releases gemcitabine, oxaliplatin, and doxorubicin in their active form in cancer cells is a potent cytotoxic agent with nM IC50s ; it is highly selective to cancer cells with mean selectivity indices to human (136) and murine (320) cancer cells. It effectively induces release of DAMPs (CALR, ATP & HMGB1) in CT26 cells facilitating more efficient phagocytosis by J774 macrophages than the FDA drugs or their co-administration. The viability of CT26 cells co-cultured with J774 macrophages and treated with 2 was reduced by 32 % compared to the non-treated cells, suggesting a synergistic antiproliferative effect between the chemical and immune reactions. 2 inhibited in vivo tumor growth in two murine models (LLC and CT26) better than the FDA drugs or their co-administration with significantly lower body weight loss. Mice inoculated with CT26 cells treated with 2 showed slightly better tumor free survival than doxorubicin.


Asunto(s)
Antineoplásicos , Neoplasias , Profármacos , Ratones , Humanos , Animales , Oxaliplatino/farmacología , Gemcitabina , Profármacos/farmacología , Profármacos/uso terapéutico , Muerte Celular Inmunogénica , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Línea Celular Tumoral
8.
Chemistry ; 27(45): 11682-11692, 2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34048082

RESUMEN

DNA G-quadruplexes (G4s) have been identified within the promoter regions of many proto-oncogenes. Thus, G4s represent attractive targets for cancer therapy, and the design and development of new drugs as G4 binders is a very active field of medicinal chemistry. Here, molecular biophysics and biology methods were employed to investigate the interaction of chiral metallohelices with a series of four DNA G4s (hTelo, c-myc, c-kit1, c-kit2) that are formed by the human telomeric sequence (hTelo) and in the promoter regions of c-MYC and c-KIT proto-oncogenes. We show that the investigated water-compatible, optically pure metallohelices, which are made by self-assembly of simple nonpeptidic organic components around FeII ions and exhibit bioactivity emulating the natural systems, bind with high affinity to G4 DNA and much lower affinity to duplex DNA. Notably, both enantiomers of a metallohelix containing a m-xylenyl bridge (5 b) were found to effectively inhibit primer elongation catalyzed by Taq DNA polymerase by stabilizing G4 structures formed in the template strands containing c-myc and c-kit2 G4-forming sequences. Moreover, both enantiomers of 5 b downregulated the expression of c-MYC and c-KIT oncogenes in human embryonic kidney cells at mRNA and protein levels. As metallohelices also bind alternative nucleic acid structures, they hold promise as potential multitargeted drugs.


Asunto(s)
G-Cuádruplex , Proteínas Proto-Oncogénicas c-kit , Proteínas Proto-Oncogénicas c-myc , ADN , Compuestos Ferrosos , Humanos , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-myc/genética , Telómero
9.
Chemistry ; 27(33): 8547-8556, 2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-33835526

RESUMEN

A cyclometalated IrIII complex conjugated to a far-red-emitting coumarin, IrIII -COUPY (3), was recently shown as a very promising photosensitizer suitable for photodynamic therapy of cancer. Therefore, the primary goal of this work was to deepen knowledge on the mechanism of its photoactivated antitumor action so that this information could be used to propose a new class of compounds as drug candidates for curing very hardly treatable human tumors, such as androgen resistant prostatic tumors of metastatic origin. Conventional anticancer chemotherapies exhibit several disadvantages, such as limited efficiency to target cancer stem cells (CSCs), which are considered the main reason for chemotherapy resistance, relapse, and metastasis. Herein, we show, using DU145 tumor cells, taken as the model of hormone-refractory and aggressive prostate cancer cells resistant to conventional antineoplastic drugs, that the photoactivated conjugate 3 very efficiently eliminates both prostate bulk (differentiated) and prostate hardly treatable CSCs simultaneously and with a similar efficiency. Notably, the very low toxicity of IrIII -COUPY conjugate in the prostate DU145 cells in the dark and its pronounced selectivity for tumor cells compared with noncancerous cells could result in low side effects and reduced damage of healthy cells during the photoactivated therapy by this agent. Moreover, the experiments performed with the 3D spheroids formed from DU145 CSCs showed that conjugate 3 can penetrate the inner layers of tumor spheres, which might markedly increase its therapeutic effect. Also interestingly, this conjugate induces apoptotic cell death in prostate cancer DU145 cells associated with calcium signaling flux in these cells and autophagy. To the best of our knowledge, this is the first study demonstrating that a photoactivatable metal-based compound is an efficient agent capable of killing even hardly treatable CSCs.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Cumarinas/farmacología , Humanos , Masculino , Células Madre Neoplásicas , Neoplasias de la Próstata/tratamiento farmacológico
10.
Chemistry ; 27(41): 10711-10716, 2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34046954

RESUMEN

Photoactivatable agents offer the prospect of highly selective cancer therapy with low side effects and novel mechanisms of action that can combat current drug resistance. 1,8-Naphthalimides with their extended π system can behave as light-harvesting groups, fluorescent probes and DNA intercalators. We conjugated N-(carboxymethyl)-1,8-naphthalimide (gly-R-Nap) with an R substituent on the naphthyl group to photoactive diazido PtIV complexes to form t,t,t-[Pt(py)2 (N3 )2 (OH)(gly-R-Nap)], R=H (1), 3-NO2 (2) or 4-NMe2 (3). They show enhanced photo-oxidation, cellular accumulation and promising photo-cytotoxicity in human A2780 ovarian, A549 lung and PC3 prostate cancer cells with visible light activation, and low dark cytotoxicity. Complexes 1 and 2 exhibit pre-intercalation into DNA, resulting in enhanced photo-induced DNA crosslinking. Complex 3 has a red-shifted absorption band at 450 nm, allowing photoactivation and photo-cytotoxicity with green light.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Antineoplásicos/farmacología , Línea Celular Tumoral , ADN , Femenino , Humanos , Luz , Compuestos Organoplatinos , Platino (Metal)
11.
Inorg Chem ; 60(13): 9529-9541, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34156246

RESUMEN

Four bipyridine-type ligands variably derivatized with two bioactive groups (taken from ethacrynic acid, flurbiprofen, biotin, and benzylpenicillin) were prepared via sequential esterification steps from commercial 2,2'-bipyridine-4,4'-dicarboxylic acid and subsequently coordinated to ruthenium(II) p-cymene and iridium(III) pentamethylcyclopentadienyl scaffolds. The resulting complexes were isolated as nitrate salts in high yields and fully characterized by analytical and spectroscopic methods. NMR and MS studies in aqueous solution and in cell culture medium highlighted a substantial stability of ligand coordination and a slow release of the bioactive fragments in the latter case. The complexes were assessed for their antiproliferative activity on four cancer cell lines, showing cytotoxicity to the low micromolar level (equipotent with cisplatin). Additional biological experiments revealed a multimodal mechanism of action of the investigated compounds, involving DNA metalation and enzyme inhibition. Synergic effects provided by specific combinations of metal and bioactive fragments were identified, pointing toward an optimal ethacrynic acid/flurbiprofen combination for both Ru(II) and Ir(III) complexes.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Iridio/farmacología , Piridinas/farmacología , Rutenio/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Daño del ADN , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Iridio/química , Ligandos , Estructura Molecular , Piridinas/química , Rutenio/química , Células Tumorales Cultivadas
12.
Int J Mol Sci ; 22(19)2021 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-34639179

RESUMEN

DNA-dependent DNA and RNA polymerases are important modulators of biological functions such as replication, transcription, recombination, or repair. In this work performed in cell-free media, we studied the ability of selected DNA polymerases to overcome a monofunctional adduct of the cytotoxic/antitumor platinum-acridinylthiourea conjugate [PtCl(en)(L)](NO3)2 (en = ethane-1,2-diamine, L = 1-[2-(acridin-9-ylamino)ethyl]-1,3-dimethylthiourea) (ACR) in its favored 5'-CG sequence. We focused on how a single site-specific ACR adduct with intercalation potency affects the processivity and fidelity of DNA-dependent DNA polymerases involved in translesion synthesis (TLS) and repair. The ability of the G(N7) hybrid ACR adduct formed in the 5'-TCGT sequence of a 24-mer DNA template to inhibit the synthesis of a complementary DNA strand by the exonuclease-deficient Klenow fragment of DNA polymerase I (KFexo-) and human polymerases eta, kappa, and iota was supplemented by thermodynamic analysis of the polymerization process. Thermodynamic parameters of a simulated translesion synthesis across the ACR adduct were obtained by using microscale thermophoresis (MST). Our results show a strong inhibitory effect of an ACR adduct on enzymatic TLS: there was only small synthesis of a full-length product (less than 10%) except polymerase eta (~20%). Polymerase eta was able to most efficiently bypass the ACR hybrid adduct. Incorporation of a correct dCMP opposite the modified G residue is preferred by all the four polymerases tested. On the other hand, the frequency of misinsertions increased. The relative efficiency of misinsertions is higher than that of matched cytidine monophosphate but still lower than for the nonmodified control duplex. Thermodynamic inspection of the simulated TLS revealed a significant stabilization of successively extended primer/template duplexes containing an ACR adduct. Moreover, no significant decrease of dissociation enthalpy change behind the position of the modification can contribute to the enzymatic TLS observed with the DNA-dependent, repair-involved polymerases. This TLS could lead to a higher tolerance of cancer cells to the ACR conjugate compared to its enhanced analog, where thiourea is replaced by an amidine group: [PtCl(en)(L)](NO3)2 (complex AMD, en = ethane-1,2-diamine, L = N-[2-(acridin-9-ylamino)ethyl]-N-methylpropionamidine).


Asunto(s)
Aductos de ADN/química , Daño del ADN , Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Sustancias Intercalantes/química , Compuestos Organoplatinos/química , Urea/análogos & derivados , Replicación del ADN , Humanos , Urea/química
13.
Angew Chem Int Ed Engl ; 60(31): 17123-17130, 2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34105220

RESUMEN

Werner's Complex, as a cationic coordination complex (CCC), has hitherto unappreciated biological properties derived from its binding affinity to highly anionic biomolecules such as glycosaminoglycans (GAGs) and nucleic acids. Competitive inhibitor and spectroscopic assays confirm the high affinity to GAGs heparin, heparan sulfate (HS), and its pentasaccharide mimetic Fondaparinux (FPX). Functional consequences of this affinity include inhibition of FPX cleavage by bacterial heparinase and mammalian heparanase enzymes with inhibition of cellular invasion and migration. Werner's Complex is a very efficient condensing agent for DNA and tRNA. In proof-of-principle for translational implications, it is demonstrated to display antiviral activity against human cytomegalovirus (HCMV) at micromolar concentrations with promising selectivity. Exploitation of non-covalent hydrogen-bonding and electrostatic interactions has motivated the unprecedented discovery of these properties, opening new avenues of research for this iconic compound.


Asunto(s)
Antivirales/farmacología , Complejos de Coordinación/farmacología , Citomegalovirus/efectos de los fármacos , Fondaparinux/antagonistas & inhibidores , Glicosaminoglicanos/farmacología , Antivirales/química , Complejos de Coordinación/química , Glicosaminoglicanos/química , Humanos , Pruebas de Sensibilidad Microbiana
14.
J Biol Inorg Chem ; 25(6): 913-924, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32851480

RESUMEN

The search for more effective platinum anticancer drugs has led to the design, synthesis, and preclinical testing of hundreds of new platinum complexes. This search resulted in the recognition and subsequent FDA approval of the third-generation Pt(II) anticancer drug, [Pt(1,2-diaminocyclohexane)(oxalate)], oxaliplatin, as an effective agent in treating colorectal and gastrointestinal cancers. Another promising example of the class of anticancer platinum(II) complexes incorporating the Pt(1,n-diaminocycloalkane) moiety is kiteplatin ([Pt(cis-1,4-DACH)Cl2], DACH = diaminocyclohexane). We report here our progress in evaluating the role of the cycloalkyl moiety in these complexes focusing on the synthesis, characterization, evaluation of the antiproliferative activity in tumor cells and studies of the mechanism of action of new [Pt(cis-1,3-diaminocycloalkane)Cl2] complexes wherein the cis-1,3-diaminocycloalkane group contains the cyclobutyl, cyclopentyl, and cyclohexyl moieties. We demonstrate that [Pt(cis-1,3-DACH)Cl2] destroys cancer cells with greater efficacy than the other two investigated 1,3-diamminocycloalkane derivatives, or cisplatin. Moreover, the investigated [Pt(cis-1,3-diaminocycloalkane)Cl2] complexes show selectivity toward tumor cells relative to non-tumorigenic normal cells. We also performed several mechanistic studies in cell-free media focused on understanding some early steps in the mechanism of antitumor activity of bifunctional platinum(II) complexes. Our data indicate that reactivities of the investigated [Pt(cis-1,3-diaminocycloalkane)Cl2] complexes and cisplatin with glutathione and DNA binding do not correlate with antiproliferative activity of these platinum(II) complexes in cancer cells. In contrast, we show that the higher antiproliferative activity in cancer cells of [Pt(cis-1,3-DACH)Cl2] originates from its highest hydrophobicity and most efficient cellular uptake.


Asunto(s)
Antineoplásicos/síntesis química , Hidrocarburos Cíclicos/síntesis química , Compuestos Organometálicos/síntesis química , Platino (Metal)/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Cisplatino/normas , ADN/química , Ensayos de Selección de Medicamentos Antitumorales , Glutatión/química , Humanos , Compuestos Organometálicos/farmacología
15.
J Biol Inorg Chem ; 25(2): 339-350, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32112290

RESUMEN

The adverse side effects and acquired resistance associated with the clinical application of traditional platinum-based anticancer drugs have forced investigation of alternative transition metal-based compounds and their cytostatic properties. Over the last years, the anticancer potential of cobalt complexes has been extensively studied, and in-depth analyses of their mode of action have been conducted. In this work, we present antiproliferative activity against human cancer cells of the dinuclear Co(III) complexes bearing the quinizarin ligand and tris(2-aminoethyl)amine (tren, compound 1) or tris(2-pyridylmethyl)amine (tpa, compound 2) co-ligands. To contribute the understanding mechanisms of biological action of these compounds, their association with DNA in the cells, DNA binding in cell-free media, and DNA cleavage capability were investigated in detail. The results demonstrate that both complexes interact with DNA in tumor cells. However, their mechanism of antiproliferative action is different, and this difference is mirrored by distinct antiproliferative activity. The antiproliferative effect of 1 is connected with its ability to intercalate into DNA and subsequently to inhibit activities of DNA processing enzymes. In contrast, the total antiproliferative efficiency of 2, thanks to its redox properties, appears to be connected with its ability to form radicals and, consequently, with the ability of 2 to cleave DNA. Hence, the findings presented in this study may significantly contribute to understanding the antitumor potential of cobalt complexes. Dinuclear Co(III) complexes containing the bioactive quinizarin ligand exhibit antiproliferative activity based on distinct mechanism.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Cobalto/farmacología , Complejos de Coordinación/farmacología , ADN/química , Antraquinonas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Sitios de Unión/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cobalto/química , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , División del ADN , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ligandos , Conformación Molecular , Células Tumorales Cultivadas
16.
Chemistry ; 26(38): 8435-8442, 2020 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-32329091

RESUMEN

RNA bulges represent one of the most common motifs in the RNA secondary structure and serve in a variety of biological functions. Compounds stabilizing RNA bulges are important for probing RNA structure and function and for therapy of some diseases. Here, the ability of a series of enantiomeric pairs of optically pure bimetallic metallohelices with different flexible linkers to target various RNA bulges is investigated. The results show that binding affinities of the metallohelices to bulged RNA differ and strongly depend on the size of the bulge and the base composition of the bulge loop. Notably, the shorter, more compact, and less flexible metallohelices bind to RNA bulges most efficiently and selectively. Interestingly, the ability of the metallohelices to bind to RNA bulges correlates with their previously reported antimicrobial activity, which suggests that the selective recognition of bulged regions in RNA by the metallohelices might also contribute to their biological activity.


Asunto(s)
Peptidomiméticos/química , ARN/química , Secuencia de Bases , Conformación de Ácido Nucleico
17.
Chemistry ; 26(69): 16554-16562, 2020 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-33026666

RESUMEN

Bulges are essential structural elements in nucleic acids. The detection and targeting of bulged DNA sequences are highly important. Small molecules capable of targeting DNA bulges have attracted considerable attention because they cannot only be used as reagents for bulge recognition, but also as potential therapeutic drugs. Herein, the interactions of DNA duplexes, containing bulges of various sizes and base compositions, with a series of FeII triplex-forming metallohelices are reported. The results obtained, with the aid of molecular biophysics methods, show that the investigated metallohelices prefer to bind to bulged DNA, rather than double-stranded DNA, and that their binding affinities towards bulges differ among individual metallohelices. Moreover, their binding affinities towards bulges strongly depend on the bulge size and the base composition of the bulge loop. The investigated metallohelices can enter eukaryotic cells and accumulate in the cell nucleus, allowing them to interact with nucleic acids. Hence, it is reasonable to suggest that the interaction of metallohelices with nucleic acid bulges might contribute to the mechanism of their biological activity.


Asunto(s)
Antineoplásicos , ADN , Compuestos Ferrosos/química , Secuencia de Bases
18.
Inorg Chem ; 59(20): 15135-15143, 2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-32988198

RESUMEN

RNA triplexes are significant tertiary structure motifs that are found in many functional RNAs. Hence, small molecules capable of recognition, binding, and stabilization of the triple-helical RNA structures are emerging as attractive potential molecular biology tools and therapeutic agents. Here, we utilize methods of molecular biology and biophysics to study the interactions of a series of antitumor substitution-inert polynuclear platinum complexes (SI-PPCs) with triple-helical RNA structures. We show that SI-PPCs recognize and stabilize RNA triplexes and inhibit reverse transcription preferentially in the RNA template prone to the triplex formation. These so far unexplored properties of SI-PPCs suggest that the targeting of triple-stranded regions in RNA might contribute to the biological effects of SI-PPCs.


Asunto(s)
Antineoplásicos/química , Complejos de Coordinación/química , ARN/química , Transcripción Reversa/efectos de los fármacos , Secuencia de Bases , Conformación de Ácido Nucleico , Platino (Metal)/química
19.
Inorg Chem ; 59(5): 3304-3311, 2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32064865

RESUMEN

The water-compatible optically pure metallohelices made by self-assembly of simple nonpeptidic organic components around Fe(II) ions are now recognized as a distinct subclass of helicates that exhibit similar architecture to some natural cationic antimicrobial peptides. Notably, a new series of metallohelices was recently shown to exhibit biological activity, displaying high, structure-dependent activity against bacteria. It is also important that, thanks to their properties, such metallohelices can exhibit specific interactions with biomacromolecules. Here, following our prior report on the metallohelices that have high, structure-dependent activity against bacteria, we investigated the interactions of the series of iron(II) metallohelices with DNA, which is a potential pharmacological target of this class of coordination compounds. The results obtained with the aid of biophysical and molecular biology methods show that the investigated metallohelices accumulate in eukaryotic cells and that a significant fraction of the metallohelices accumulates in the cell nucleus, allowing them to interact also with nuclear DNA. Additionally, we have demonstrated that some metallohelices have a high affinity to DNA and are able to condense/aggregate DNA molecules more efficiently than conventional DNA-condensing agents, such as polyamines. Moreover, this capability of the metallohelices correlates with their efficiency to inhibit DNA-related enzymatic activities, such as those connected with DNA transcription, catalysis of DNA relaxation by DNA topoisomerase I, and cleavage by restriction enzymes.


Asunto(s)
Núcleo Celular/química , ADN-Topoisomerasas de Tipo I/metabolismo , ADN/antagonistas & inhibidores , Compuestos Ferrosos/farmacología , Inhibidores de Topoisomerasa I/farmacología , Núcleo Celular/metabolismo , ADN/genética , ADN/metabolismo , Compuestos Ferrosos/síntesis química , Compuestos Ferrosos/química , Células HCT116 , Humanos , Estructura Molecular , Fenómenos Ópticos , Inhibidores de Topoisomerasa I/síntesis química , Inhibidores de Topoisomerasa I/química
20.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096927

RESUMEN

Translesion synthesis (TLS) through DNA adducts of antitumor platinum complexes has been an interesting aspect of DNA synthesis in cells treated with these metal-based drugs because of its correlation to drug sensitivity. We utilized model systems employing a DNA lesion derived from a site-specific monofunctional adduct formed by antitumor [PtCl(en)(L)](NO3)2 (complex AMD, en = ethane-1,2-diamine, L = N-[2-(acridin-9-ylamino)ethyl]-N-methylpropionamidine) at a unique G residue. The catalytic efficiency of TLS DNA polymerases, which differ in their processivity and fidelity for the insertion of correct dCTP, with respect to the other incorrect nucleotides, opposite the adduct of AMD, was investigated. For a deeper understanding of the factors that control the bypass of the site-specific adducts of AMD catalyzed by DNA polymerases, we also used microscale thermophoresis (MST) to measure the thermodynamic changes associated with TLS across a single, site-specific adduct formed in DNA by AMD. The relative catalytic efficiency of the investigated DNA polymerases for the insertion of correct dCTP, with respect to the other incorrect nucleotides, opposite the AMD adduct, was reduced. Nevertheless, incorporation of the correct C opposite the G modified by AMD of the template strand was promoted by an increasing thermodynamic stability of the resulting duplex. The reduced relative efficiency of the investigated DNA polymerases may be a consequence of the DNA intercalation of the acridine moiety of AMD and the size of the adduct. The products of the bypass of this monofunctional lesion produced by AMD and DNA polymerases also resulted from the misincorporation of dNTPs opposite the platinated G residues. The MST analysis suggested that thermodynamic factors may contribute to the forces that governed enhanced incorporation of the incorrect dNTPs by DNA polymerases.


Asunto(s)
Aductos de ADN/química , ADN Polimerasa Dirigida por ADN/metabolismo , Compuestos de Platino/farmacología , Acridinas/química , Acridinas/farmacología , Biocatálisis , Catálisis , ADN/biosíntesis , Aductos de ADN/genética , Aductos de ADN/metabolismo , Reparación del ADN , Replicación del ADN , Guanina/metabolismo , Nucleótidos/genética , Nucleótidos/metabolismo , Compuestos de Platino/química , Difusión Térmica , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA