Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.069
Filtrar
1.
Metallomics ; 16(5)2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38614957

RESUMEN

Metal ion-catalyzed overproduction of reactive oxygen species (ROS) is believed to contribute significantly to oxidative stress and be involved in several biological processes, from immune defense to development of diseases. Among the essential metal ions, copper is one of the most efficient catalysts in ROS production in the presence of O2 and a physiological reducing agent such as ascorbate. To control this chemistry, Cu ions are tightly coordinated to biomolecules. Free or loosely bound Cu ions are generally avoided to prevent their toxicity. In the present report, we aim to find stable Cu-ligand complexes (Cu-L) that can efficiently catalyze the production of ROS in the presence of ascorbate under aerobic conditions. Thermodynamic stability would be needed to avoid dissociation in the biological environment, and high ROS catalysis is of interest for applications as antimicrobial or anticancer agents. A series of Cu complexes with the well-known tripodal and tetradentate ligands containing a central amine linked to three pyridyl-alkyl arms of different lengths were investigated. Two of them with mixed arm length showed a higher catalytic activity in the oxidation of ascorbate and subsequent ROS production than Cu salts in buffer, which is an unprecedented result. Despite these high catalytic activities, no increased antimicrobial activity toward Escherichia coli or cytotoxicity against eukaryotic AGS cells in culture related to Cu-L-based ROS production could be observed. The potential reasons for discrepancy between in vitro and in cell data are discussed.


Asunto(s)
Cobre , Especies Reactivas de Oxígeno , Cobre/metabolismo , Cobre/química , Especies Reactivas de Oxígeno/metabolismo , Ligandos , Catálisis , Humanos , Escherichia coli/metabolismo , Escherichia coli/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Complejos de Coordinación/farmacología , Ácido Ascórbico/metabolismo , Ácido Ascórbico/química , Oxidación-Reducción
2.
Spectrochim Acta A Mol Biomol Spectrosc ; 315: 124265, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38626674

RESUMEN

In recent years, extensive research has been directed towards understanding the interactions between various zinc complexes with DNA, specifically delving into their intercalation and binding behaviors. The binding of zinc complexes to DNA is particularly intriguing due to their distinctive intercalating capabilities. This study unveils a remarkable phenomenon observed with a specific Zn complex, ([B-Zn-N3], where B is a Schiff base ligand), during DNA intercalation investigations in the popular DMSO-Water binary solvent mixture. An unanticipated observation revealed time-dependent changes in the UV-visible absorption spectroscopic studies, coupled with the existence of an isosbestic point. This observation questions the stability of the intercalating agent itself during the intercalation process. The emergence of a decomposed product during the intercalation study has been confirmed through various analytical techniques, including CHN analysis, MALDI mass, XPS, Raman spectroscopy, and Powder XRD. The change in the chemical species on intercalation is further substantiated by theoretical studies, adding depth to our understanding of the intricate dynamics at play during DNA intercalation with the [B-Zn-N3] complex in the DMSO-Water system.


Asunto(s)
ADN , Dimetilsulfóxido , Sustancias Intercalantes , Agua , Dimetilsulfóxido/química , Sustancias Intercalantes/química , ADN/química , ADN/metabolismo , Agua/química , Espectrometría Raman , Zinc/química , Espectrofotometría Ultravioleta , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Bases de Schiff/química
3.
Biomolecules ; 14(4)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38672503

RESUMEN

The emergence of multidrug-resistant (MDR) microorganisms combined with the ever-draining antibiotic pipeline poses a disturbing and immensely growing public health challenge that requires a multidisciplinary approach and the application of novel therapies aimed at unconventional targets and/or applying innovative drug formulations. Hence, bacterial iron acquisition systems and bacterial Fe2+/3+-containing enzymes have been identified as a plausible target of great potential. The intriguing "Trojan horse" approach deprives microorganisms from the essential iron. Recently, gallium's potential in medicine as an iron mimicry species has attracted vast attention. Different Ga3+ formulations exhibit diverse effects upon entering the cell and thus supposedly have multiple targets. The aim of the current study is to specifically distinguish characteristics of great significance in regard to the initial gallium-based complex, allowing the alien cation to effectively compete with the native ferric ion for binding the siderophores pyochelin and pyoverdine secreted by the bacterium P. aeruginosa. Therefore, three gallium-based formulations were taken into consideration: the first-generation gallium nitrate, Ga(NO3)3, metabolized to Ga3+-hydrated forms, the second-generation gallium maltolate (tris(3-hydroxy-2-methyl-4-pyronato)gallium), and the experimentally proven Ga carrier in the bloodstream-the protein transferrin. We employed a reliable in silico approach based on DFT computations in order to understand the underlying biochemical processes that govern the Ga3+/Fe3+ rivalry for binding the two bacterial siderophores.


Asunto(s)
Antibacterianos , Galio , Hierro , Compuestos Organometálicos , Fenoles , Pseudomonas aeruginosa , Sideróforos , Galio/química , Galio/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Sideróforos/química , Sideróforos/metabolismo , Hierro/metabolismo , Hierro/química , Oligopéptidos/química , Oligopéptidos/metabolismo , Tiazoles/química , Tiazoles/metabolismo , Tiazoles/farmacología , Simulación por Computador , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Pironas/química , Pironas/metabolismo , Pironas/farmacología
4.
Nitric Oxide ; 147: 26-41, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38614230

RESUMEN

Nitric oxide (NO) acts in different physiological processes, such as blood pressure control, antiparasitic activities, neurotransmission, and antitumor action. Among the exogenous NO donors, ruthenium nitrosyl/nitro complexes are potential candidates for prodrugs, due to their physicochemical properties, such as thermal and physiological pH stability. In this work, we proposed the synthesis and physical characterization of the new nitro terpyridine ruthenium (II) complexes of the type [RuII(L)(NO2)(tpy)]PF6 where tpy = 2,2':6',2″-terpyridine; L = 3,4-diaminobenzoic acid (bdq) or o-phenylenediamine (bd) and evaluation of influence of diimine bidentate ligand NH.NHq-R (R = H or COOH) in the HSA/DNA interaction as well as antiviral activity. The interactions between HSA and new nitro complexes [RuII(L)(NO2)(tpy)]+ were evaluated. The Ka values for the HSA-[RuII(bdq)(NO2)(tpy)]+ is 10 times bigger than HSA-[RuII(bd)(NO2)(tpy)]+. The sites of interaction between HSA and the complexes via synchronous fluorescence suppression indicate that the [RuII(bdq)(NO2)(tpy)]+ is found close to the Trp-241 residue, while the [RuII(bd)(NO2)(tpy)]+ complex is close to Tyr residues. The interaction with fish sperm fs-DNA using direct spectrophotometric titration (Kb) and ethidium bromide replacement (KSV and Kapp) showed weak interaction in the system fs-DNA-[RuII(bdq)(NO)(tpy)]+. Furthermore, fs-DNA-[RuII(bd)(NO2)(tpy)]+ and fs-DNA-[RuII(bd)(NO)(tpy)]3+ system showed higher intercalation constant. Circular dichroism spectra for fs-DNA-[RuII(bd)(NO2)(tpy)]+ and fs-DNA-[RuII(bd)(NO)(tpy)]3+, suggest semi-intercalative accompanied by major groove binding interaction modes. The [RuII(bd)(NO2)(tpy)]+ and [RuII(bd)(NO)(tpy)]3+ inhibit replication of Zika and Chikungunya viruses based in the nitric oxide release under S-nitrosylation reaction with cysteine viral.


Asunto(s)
Antivirales , ADN , Rutenio , Humanos , ADN/metabolismo , ADN/química , Rutenio/química , Rutenio/farmacología , Antivirales/farmacología , Antivirales/química , Antivirales/metabolismo , Ligandos , Animales , Albúmina Sérica Humana/química , Albúmina Sérica Humana/metabolismo , Piridinas/química , Piridinas/farmacología , Iminas/química , Iminas/farmacología , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo
5.
Chemistry ; 30(28): e202400268, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38472116

RESUMEN

Modern approaches in metallodrug research focus on compounds that bind protein targets rather than DNA. However, the identification of protein targets and binding sites is challenging. Using intact mass spectrometry and proteomics, we investigated the binding of the antimetastatic agent RAPTA-C to the model proteins ubiquitin, cytochrome c, lysozyme, and myoglobin. Binding to cytochrome c and lysozyme was negligible. However, ubiquitin bound up to three Ru moieties, two of which were localized at Met1 and His68 as [Ru(cym)], and [Ru(cym)] or [Ru(cym)(PTA)] adducts, respectively. Myoglobin bound up to four [Ru(cym)(PTA)] moieties and five sites were identified at His24, His36, His64, His81/82 and His113. Collision-induced unfolding (CIU) studies via ion-mobility mass spectrometry allowed measuring protein folding as a function of collisional activation. CIU of protein-RAPTA-C adducts showed binding of [Ru(cym)] to Met1 caused a significant compaction of ubiquitin, likely from N-terminal S-Ru-N chelation, while binding of [Ru(cym)(PTA)] to His residues of ubiquitin or myoglobin induced a smaller effect. Interestingly, the folded state of ubiquitin formed by His functionalization was more stable than Met1 metalation. The data suggests that selective metalation of amino acids at different positions on the protein impacts the conformation and potentially the biological activity of anticancer compounds.


Asunto(s)
Citocromos c , Muramidasa , Mioglobina , Pliegue de Proteína , Ubiquitina , Ubiquitina/química , Ubiquitina/metabolismo , Mioglobina/química , Mioglobina/metabolismo , Sitios de Unión , Citocromos c/química , Citocromos c/metabolismo , Muramidasa/química , Muramidasa/metabolismo , Unión Proteica , Rutenio/química , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo
6.
Dalton Trans ; 53(14): 6410-6415, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38501501

RESUMEN

An asymmetric bi-nuclear copper(II) complex with both cytotoxic and immunogenic activity towards breast cancer stem cells (CSCs) is reported. The bi-nuclear copper(II) complex comprises of two copper(II) centres bound to flufenamic acid and 3,4,7,8-tetramethyl-1,10-phenanthroline. The bi-nuclear copper(II) complex exhibits sub-micromolar potency towards breast CSCs grown in monolayers and three-dimensional cultures. Remarkably, the bi-nuclear copper(II) complex is up to 25-fold more potent toward breast CSC mammospheres than salinomycin (a gold standard anti-breast CSC agent) and cisplatin (a clinically administered metallodrug). Mechanistic studies showed that the bi-nuclear copper(II) complex readily enters breast CSCs, elevates intracellular reactive oxygen species levels, induces apoptosis, and promotes damage-associated molecular pattern release. The latter triggers phagocytosis of breast CSCs by macrophages. As far as we are aware, this is the first report of a bi-nuclear copper(II) complex to induce engulfment of breast CSCs by immune cells.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Ácido Flufenámico/metabolismo , Cobre/metabolismo , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , Células Madre Neoplásicas
7.
Eur J Med Chem ; 268: 116295, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38437750

RESUMEN

This paper introduces a new ligand, 4,6-dichloro-5-(1H-imidazo [4,5-f]phenanthroline-2-yl)pyrimidin-2-amine (DPPA), and its corresponding new iridium(III) complexes: [Ir(ppy)2(DPPA)](PF6) (2a) (where ppy represents deprotonated 2-phenylpyridine), [Ir(bzq)2(DPPA)](PF6) (2b) (with bzq indicating deprotonated benzo[h]quinoline), and [Ir(piq)2(DPPA)](PF6) (2c) (piq denoting deprotonated 1-phenylisoquinoline). The cytotoxic effects of both DPPA and 2a, 2b, and 2c were evaluated against human lung carcinoma A549, melanoma B16, colorectal cancer HCT116, human hepatocellular carcinoma HepG2 cancer cell lines, as well as the non-cancerous LO2 cell line using the 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. While DPPA exhibited moderate anticancer activity toward A549, B16, HCT116 and HepG2 cells, complexes 2a, 2b, and 2c displayed remarkable efficacy against A549, B16, and HCT116 cells. The cell colonies and wound healing were investigated. Moreover, various aspects of the anticancer mechanisms were explored. The cell cycle analyses revealed that the complexes block cell proliferation of A549 cells during the S phase. Complex 2c induce an early apoptosis, while 2a and 2b cause a late apoptosis. The interaction of 2a, 2b and 2c with endoplasmic reticulum and mitochondria was identified, leading to elevated ROS and Ca2+ amounts. This resulted in a reduced mitochondrial membrane potential, mitochondrial permeability transition pore opening, and an increase of cytochrome c. Also, ferroptosis was investigated through measurements of intracellular glutathione (GSH), malondialdehyde (MDA), and recombinant glutathione peroxidase (GPX4) protein expression. The pyroptosis was explored via cell morphology, release of lactate dehydrogenase (LDH) and expression of pyroptosis-related proteins. RNA sequencing was applied to examine the signaling pathways. Western blot analyses illuminated that the complexes regulate the expression of Bcl-2 family proteins. Additionally, an in vivo antitumor study demonstrated that complex 2c exhibited a remarkable inhibitory rate of 58.58% in restraining tumor growth. In summary, the findings collectively suggest that the iridium(III) complexes induce cell death via ferroptosis, apoptosis by a ROS-mediated mitochondrial dysfunction pathway and GSDMD-mediated pyroptosis.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Ferroptosis , Humanos , Línea Celular Tumoral , Iridio/farmacología , Piroptosis , Especies Reactivas de Oxígeno/metabolismo , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Apoptosis , Proliferación Celular , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , Mitocondrias
8.
Dalton Trans ; 53(13): 5993-6005, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38469684

RESUMEN

Recently, achieving selective cancer therapy with trifling side effects has been a great challenge in the eradication of cancer. Thus, to amplify the cytoselective approach of complexes, herein, we developed a series of Re(I)[2-aryl-1H-imidazo[4,5-f][1,10]phenanthroline] tricarbonyl chloride complexes and screened their potency against HeLa and MCF-7 cell lines together with the evaluation of their toxicity towards a normal kidney cell line (HEK-293). On meticulous investigation, complex [ReI(CO)3Cl(K2-N,N-(2c))] (3c) was found to be the most potent anticancer entity among other complexes. Complex 3c also showed competency to induce apoptosis in MCF-7 cells through G2/M phase cell-cycle arrest in association with the generation of ample reactive oxygen species (ROS), eventually leading to DNA intercalation and internucleosomal cleavage. The order of the cytotoxicity of these complexes depended on their lipophilic character and the electron-withdrawing halogen substitution at the para-position of the phenyl ring in the imidazophenanthroline ligand.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Neoplasias , Humanos , Fenantrolinas/farmacología , Cloruros , Células HEK293 , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , ADN/metabolismo , Daño del ADN , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Apoptosis , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico
9.
Dalton Trans ; 53(11): 5167-5179, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38380977

RESUMEN

Cancer is a perilous life-threatening disease, and attempts are constantly being made to create multinuclear transition metal complexes that could lead to the development of potential anticancer medications and administration procedures. Hence, this work aims to design, synthesize, characterize, and assess the anticancer efficacy of ruthenium p-cymene complexes incorporating N,N'-bis(4-substituted benzoyl)hydrazine ligands. The formation of the new complexes (Ru2H1-Ru2H3) has been thoroughly established by elemental analysis, and FT-IR, UV-vis, NMR, and HR-MS spectral techniques. The solid-state molecular structures of the complexes Ru2H1 and Ru2H3 have been determined using the SC-XRD study, which confirms the N, O, and Cl-legged piano stool pseudo-octahedral geometry of each ruthenium(II) ion. The stability of these complexes in the solution state and their lipophilicity profile have been determined. Furthermore, the title complexes were tested for their in vitro anticancer activity against cancerous H460 (lung cancer cells), SkBr3 (breast cancer cells), HepG2 (liver cancer cells), and HeLa (cervical cancer cells) along with non-cancerous (HEK-293) cells. The IC50 results revealed that complex Ru2H3 exhibits potent activity against the proliferation of all four cancer cells and outscored the effect of the standard metallodrug cisplatin. This may be attributed to the presence of a couple of lipophilic electron-donating methoxy groups in the ligand scaffold and also the ruthenium(II) p-cymene motifs. Advantageously, all the complexes (Ru2H1-Ru2H3) displayed cytotoxic specificity only towards cancerous cells by leaving the off-target non-cancerous cells undamaged. Acridine orange/ethidium bromide (AO/EB) staining, Hoechst 33342, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) staining assays were used to investigate the apoptotic pathway and ROS levels in mitochondria. The results of western blot analysis confirmed that the complexes triggered apoptosis through an intrinsic mitochondrial pathway by upregulating Bax and downregulating Bcl-2 proteins. Finally, the extent of apoptosis triggered by the complex Ru2H3 was quantified with the aid of flow cytometry using the Annexin V-FITC/propidium iodide (PI) double-staining technique.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Cimenos , Rutenio , Humanos , Rutenio/farmacología , Rutenio/química , Especies Reactivas de Oxígeno/metabolismo , Células HEK293 , Espectroscopía Infrarroja por Transformada de Fourier , Apoptosis , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Antineoplásicos/química , Hidrazinas/farmacología , Línea Celular Tumoral
10.
Chemistry ; 30(24): e202304367, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38377169

RESUMEN

Carbonic Anhydrases (CAs) have been a target for de novo protein designers due to the simplicity of the active site and rapid rate of the reaction. The first reported mimic contained a Zn(II) bound to three histidine imidazole nitrogens and an exogenous water molecule, hence closely mimicking the native enzymes' first coordination sphere. Co(II) has served as an alternative metal to interrogate CAs due to its d7 electronic configuration for more detailed solution characterization. We present here the Co(II) substituted [Co(II)(H2O/OH-)]N(TRIL2WL23H)3 n+ that behaves similarly to native Co(II) substituted human-CAs. Like the Zn(II) analogue, the cobalt-derivative at slightly basic pH is incapable of hydrolyzing p-nitrophenylacetate (pNPA); however, as the pH is increased a significant activity develops, which at pH values above 10 eventually yields a catalytic efficiency that exceeds that of the [Zn(II)(OH-)]N(TRIL2WL23H)3 + peptide complex. X-ray absorption analysis is consistent with an octahedral species at pH 7.5 that converts to a 5-coordinate species by pH 11. UV-vis spectroscopy can monitor this transition, giving a pKa for the conversion of 10.3. We assign this conversion to the formation of a 5-coordinate Co(II)(Nimid)3(OH)(H2O) species. The pH dependent kinetic analysis indicates the maximal rate (kcat), and thus the catalytic efficiency (kcat/Km), follow the same pH profile as the spectroscopic conversion to the pentacoordinate species. This correlation suggests that the chemically irreversible ester hydrolysis corresponds to the rate determining process.


Asunto(s)
Anhidrasas Carbónicas , Cobalto , Esterasas , Zinc , Zinc/química , Cobalto/química , Anhidrasas Carbónicas/química , Anhidrasas Carbónicas/metabolismo , Concentración de Iones de Hidrógeno , Humanos , Esterasas/química , Esterasas/metabolismo , Dominio Catalítico , Hidrólisis , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Cinética , Catálisis , Nitrofenoles/química , Nitrofenoles/metabolismo
11.
Dalton Trans ; 53(5): 1977-1988, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38205595

RESUMEN

Most platinum group-based cyclometalated neutral and cationic anticancer complexes with the general formula [(C^N)2Ir(XY)]0/+ (neutral complex: XY = bidentate anionic ligand; cationic complex: XY = bidentate neutral ligand) are notable owing to their intrinsic luminescence properties, good cell permeability, interaction with some biomolecular targets and unique mechanisms of action (MoAs). We herein synthesized a series of neutral and cationic amine-imine cyclometalated iridium(III) complexes using Schiff base ligands with sp2-N/sp3-N N^NH2 chelating donors. The cyclometalated iridium(III) complexes were identified by various techniques. They were stable in aqueous media, displayed moderate fluorescence and exhibited affinity toward bovine serum albumin (BSA). The complexes demonstrated promising cytotoxicity against lung cancer A549 cells, cisplatin-resistant lung cancer A549/DDP cells, cervical carcinoma HeLa cells and human liver carcinoma HepG2 cells, with IC50 values ranging from 9.98 to 19.63 µM. Unfortunately, these complexes had a low selectivity (selectivity index: 1.62-1.98) towards A549 cells and BEAS-2B normal cells. The charge pattern of the metal center (neutral or cationic) and ligand substituents showed little influence on the cytotoxicity and selectivity of these complexes. The study revealed that these complexes could target mitochondria, cause depolarization of the mitochondrial membrane, and trigger the production of intracellular ROS. Additionally, the complexes were observed to induce late apoptosis and perturb the cell cycle in the G2/M or S phase in A549 cells. Based on these results, it appears that the anticancer efficacy of these complexes was predominantly attributed to the redox mechanism.


Asunto(s)
Antineoplásicos , Carcinoma , Complejos de Coordinación , Neoplasias Pulmonares , Humanos , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , Células HeLa , Iridio/farmacología , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Ligandos , Quelantes/farmacología , Mitocondrias/metabolismo , Neoplasias Pulmonares/metabolismo , Carcinoma/metabolismo , Apoptosis , Línea Celular Tumoral
12.
Angew Chem Int Ed Engl ; 63(6): e202317940, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38063406

RESUMEN

The cytotoxic and immunogenic-activating properties of a cobalt(III)-cyclam complex bearing the non-steroidal anti-inflammatory drug, flufenamic acid is reported within the context of anti-cancer stem cell (CSC) drug discovery. The cobalt(III)-cyclam complex 1 displays sub-micromolar potency towards breast CSCs grown in monolayers, 24-fold and 31-fold greater than salinomycin (an established anti-breast CSC agent) and cisplatin (an anticancer metallopharmaceutical), respectively. Strikingly, the cobalt(III)-cyclam complex 1 is 69-fold and 50-fold more potent than salinomycin and cisplatin towards three-dimensionally cultured breast CSC mammospheres. Mechanistic studies reveal that 1 induces DNA damage, inhibits cyclooxygenase-2 expression, and prompts caspase-dependent apoptosis. Breast CSCs treated with 1 exhibit damage-associated molecular patterns characteristic of immunogenic cell death and are phagocytosed by macrophages. As far as we are aware, 1 is the first cobalt complex of any oxidation state or geometry to display both cytotoxic and immunogenic-activating effects on breast CSCs.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Complejos de Coordinación , Compuestos Heterocíclicos , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Cisplatino/farmacología , Ácido Flufenámico/metabolismo , Ácido Flufenámico/farmacología , Ácido Flufenámico/uso terapéutico , Complejos de Coordinación/metabolismo , Cobalto/farmacología , Cobalto/metabolismo , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Células Madre Neoplásicas
13.
Eur J Med Chem ; 264: 115985, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38016298

RESUMEN

The potential use of Ru(II) complexes as photosensitizers (PSs) in photodynamic therapy (PDT) has gained significant attention. In comparison with fluorophores with aggregation-caused quenching (ACQ), fluorophores with aggregation-induced emission (AIE) characteristics exhibit sustained fluorescence and dispersibility in aqueous solutions. PSs with AIE characteristics have received much attention in recent years. Herein, we reported two novel biotin-conjugated Ru(II) polypyridyl complexes (Ru1 and Ru2) with AIE characteristics. When exposed to 460 nm (10 mW cm-2) light, Ru1 and Ru2 exhibited outstanding photostability and photocatalytic activity. Ru1 and Ru2 could efficiently generate singlet oxygen and induce pUC19 DNA photolysis when exposed to 460 nm light. Interestingly, both Ru1 and Ru2 also functioned as catalysts for NADH oxidation when exposed to 460 nm light. The presence of biotin fragments in Ru1 and Ru2 enhanced the specific uptake of these complexes by tumor cells. Both complexes showed minimal toxicity to selected cells in the dark. Nevertheless, the phototoxicity of both complexes significantly increased upon 460 nm light irradiation for 15 min. Further experiments revealed that Ru2 primarily accumulated in mitochondria and might bind to mitochondrial DNA. Under 460 nm light irradiation, Ru2 induced the generation of reactive oxygen species (ROS) and NADH depletion disrupting intracellular redox homeostasis in A549 cells, activating the mitochondrial apoptosis pathway resulting in up-regulation of apoptotic marker caspase-3, effectively damaged A549 cell DNA and arrested A549 cell cycle in the S phase. In vivo anti-tumor experiments were conducted to assess the effects of Ru2 on tumor growth in A549 tumor-bearing mice. The results showed that Ru2 effectively inhibited tumor growth under 460 nm light irradiation conditions. These findings indicate that Ru2 has great potential as a targeted photosensitizer for mitochondrial targeting imaging and photodynamic therapy of tumors.


Asunto(s)
Complejos de Coordinación , Fotoquimioterapia , Rutenio , Animales , Ratones , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/metabolismo , Biotina/farmacología , Biotina/metabolismo , NAD/metabolismo , Fotoquimioterapia/métodos , Mitocondrias/metabolismo , Oxidación-Reducción , ADN/metabolismo , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Rutenio/farmacología
14.
Semin Nucl Med ; 54(1): 163-170, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37923671

RESUMEN

[68Ga]Ga-PentixaFor, a PET agent targeting CXCR4 is emerging as a versatile radiotracer with promising applications in oncology, cardiology and inflammatory disease. Preclinical work in various cancer cell lines have demonstrated high specificity and selectivity. In human investigations of several tumors, the most promising applications may be in multiple myeloma, certain lymphomas and myeloproliferative neoplasms. In the nononcologic setting, [68Ga]Ga-PentixaFor could greatly improve detection for primary aldosteronism and other endocrine abnormalities. Similarly, atherosclerotic disease and other inflammatory conditions could also benefit from enhanced identification by CXCR4 targeting. Rapidly cleared from the body with a favorable imaging and radiation dosimetry profile that has been already studied in over 1000 patients, [68Ga]Ga-PentixaFor is a worthy agent for further clinical exploration with potential for theranostic applications in hematologic malignancies.


Asunto(s)
Complejos de Coordinación , Receptores CXCR4 , Humanos , Receptores CXCR4/metabolismo , Radioisótopos de Galio , Tomografía de Emisión de Positrones/métodos , Péptidos Cíclicos/metabolismo , Complejos de Coordinación/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos
15.
ACS Appl Bio Mater ; 6(12): 5776-5788, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38061031

RESUMEN

Photodynamic therapy (PDT) has emerged as an efficient and noninvasive treatment approach utilizing laser-triggered photosensitizers for combating cancer. Within this rapidly advancing field, iridium-based photosensitizers with their dual functionality as both imaging probes and PDT agents exhibit a potential for precise and targeted therapeutic interventions. However, most reported classes of Ir(III)-based photosensitizers comprise mononuclear iridium(III), with very few examples of dinuclear systems. Exploring the full potential of iridium-based dinuclear systems for PDT applications remains a challenge. Herein, we report a dinuclear Ir(III) complex (IRDI) along with a structurally similar monomer complex (IRMO) having 2-(2,4-difluorophenyl)pyridine and 4'-methyl-2,2'-bipyridine ligands. The comparative investigation of the mononuclear and dinuclear Ir(III) complexes showed similar absorption profiles, but the dinuclear derivative IRDI exhibited a higher photoluminescence quantum yield (Φp) of 0.70 compared to that of IRMO (Φp = 0.47). Further, IRDI showed a higher singlet oxygen generation quantum yield (Φs) of 0.49 compared to IRMO (Φs = 0.28), signifying the enhanced potential of the dinuclear derivative for image-guided photodynamic therapy. In vitro assessments indicate that IRDI shows efficient cellular uptake and significant photocytotoxicity in the triple-negative breast cancer cell line MDA-MB-231. In addition, the presence of a dual positive charge on the dinuclear system facilitates the inherent mitochondria-targeting ability without the need for a specific targeting group. Subcellular singlet oxygen generation by IRDI was confirmed using Si-DMA, and light-activated cellular apoptosis via ROS-mediated PDT was verified through various live-dead assays performed in the presence and absence of the singlet oxygen scavenger NaN3. Further, the mechanism of cell death was elucidated by an annexin V-FITC/PI flow cytometric assay and by investigating the cytochrome c release from mitochondria using Western blot analysis. Thus, the dinuclear complex designed to enhance spin-orbit coupling with minimal excitonic coupling represents a promising strategy for efficient image-guided PDT using iridium complexes.


Asunto(s)
Complejos de Coordinación , Fotoquimioterapia , Neoplasias de la Mama Triple Negativas , Humanos , Fármacos Fotosensibilizantes/metabolismo , Iridio/farmacología , Iridio/metabolismo , Oxígeno Singlete/metabolismo , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Línea Celular Tumoral , Mitocondrias/metabolismo
16.
J Biol Chem ; 299(12): 105435, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37944620

RESUMEN

Copper is essential for all eukaryotic cells but many details of how it is trafficked within the cell and how it is homeostatically regulated remain uncertain. Here, we characterized the copper content of cytosol and mitochondria using liquid chromatography with ICP-MS detection. Chromatograms of cytosol exhibited over two dozen peaks due to copper proteins and coordination complexes. Yeast cells respiring on minimal media did not regulate copper import as media copper concentration increased; rather, they imported copper at increasing rates while simultaneously increasing the expression of metallothionein CUP1 which then sequestered most of the excessive imported copper. Peak intensities due to superoxide dismutase SOD1, other copper proteins, and numerous coordination complexes also increased, but not as drastically. The labile copper pool was unexpectedly diverse and divided into two groups. One group approximately comigrated with copper-glutathione, -cysteine, and -histidine standards; the other developed only at high media copper concentrations and at greater elution volumes. Most cytosolic copper arose from copper-bound proteins, especially CUP1. Cytosol contained an unexpectedly high percentage of apo-copper proteins and apo-coordination complexes. Copper-bound forms of non-CUP1 proteins and complexes coexisted with apo-CUP1 and with the chelator BCS. Both experiments suggest unexpectedly stable-binding copper proteins and coordination complexes in cytosol. COX17Δ cytosol chromatograms were like those of WT cells. Chromatograms of soluble mitochondrial extracts were obtained, and mitoplasting helped distinguish copper species in the intermembrane space versus in the matrix/inner membrane. Issues involving the yeast copperome, copper homeostasis, labile copper pool, and copper trafficking are discussed.


Asunto(s)
Complejos de Coordinación , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Cobre/metabolismo , Complejos de Coordinación/metabolismo , Proteínas Portadoras/metabolismo , Homeostasis , Metalotioneína/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
17.
Dalton Trans ; 52(44): 16376-16387, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37870147

RESUMEN

Breast cancer is the most dangerous type in women and its fatality rate has increased over the past decade. To develop more potent and target-specific breast cancer drugs, six arene ruthenium(II) complexes (1-6) containing naphthoyl benzhydrazine ligands (NL1-NL3) were synthesized and characterized by analytical and spectroscopic (infrared, UV-visible, NMR and HR-MS) methods. The SC-XRD analysis of 1 and 6 demonstrates the bis N^O bidentate binding nature of ligands to ruthenium ions and a pseudo-octahedral geometry around the Ru(II) ion. Solution stability studies using UV-Vis spectroscopy evidenced the instantaneous hydrolysis of the complexes to form monoaquated species in a solution of 1 : 9 (v/v) DMSO/phosphate buffer. All the complexes were screened for their in vitro antiproliferative activities against different human breast cancer cells, including MCF-7, SkBr3, MDA-MB-468, MDA-MB-231, and non-cancerous HEK-293 cells, by an MTT assay, and they displayed good cancer cell growth inhibitory capacity with low IC50 values. Notably, complexes 2 and 5 comprising methoxy and p-cymene groups exhibited excellent cytotoxicity towards SkBr3 cells compared to clinical drug cisplatin. AO-EB and HOECHST-33342 staining assays revealed apoptotic morphological changes in complex-treated cancer cells. Further, reactive oxygen species and mitochondrial membrane potential assays validated that the complexes induce apoptotic cell death via an intrinsic mitochondrial pathway with ROS production. In addition, the apoptotic induction and the quantification of late apoptosis were established with the aid of western blot and flow cytometry analysis, respectively.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Complejos de Coordinación , Rutenio , Humanos , Femenino , Línea Celular Tumoral , Neoplasias de la Mama/tratamiento farmacológico , Rutenio/farmacología , Rutenio/química , Células HEK293 , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Antineoplásicos/química
18.
J Inorg Biochem ; 249: 112397, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37844533

RESUMEN

In this study, we synthesized 4 cyclometalated iridium complexes using N-(1,10-phenanthrolin-5-yl)picolinamide (PPA) as the main ligand, denoted as [Ir(ppy)2PPA]PF6 (ppy = 2-phenylpyridine, Ir1), [Ir(bzq)2PPA]PF6 (bzq = benzo[h]quinoline, Ir2), [Ir(dfppy)2PPA]PF6 (dfppy = 2-(3,5-difluorophenyl)pyridine, Ir3), and [Ir(thpy)2PPA]PF6 (thpy = 2-(thiophene-2-yl)pyridine, Ir4). Compared to cisplatin and oxaliplatin, all four complexes exhibited significant anti-tumor activity. Among them, Ir2 demonstrated higher cytotoxicity against A549 cells, with an IC50 value of 1.6 ± 0.2 µM. The experimental results indicated that Ir2 primarily localized in the mitochondria, inducing a large amount of reactive oxygen species (ROS) generation, that decreased in mitochondrial membrane potential (MMP), reduced ATP production, and further impaired mitochondrial function, leading to cytochrome c release. Additionally, Ir2 caused cell cycle arrest at the S phase and induced apoptosis through the AKT-mediated signaling pathway. Further investigations revealed that Ir2 could simultaneously induce both apoptosis and autophagy in A549 cells, with the latter acting as a non-protective mechanism that promoted cell death. More importantly, Ir2 exhibited low toxicity to both normal LO2 cells in vitro and zebrafish embryos in vivo. Consequently, these newly developed Ir(III) complexes show great potential in the development of novel and low-toxicity anticancer agents.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Animales , Humanos , Células A549 , Iridio/farmacología , Iridio/metabolismo , Pez Cebra , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , Apoptosis , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/metabolismo , Piridinas/farmacología , Autofagia , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Línea Celular Tumoral
19.
Molecules ; 28(19)2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37836742

RESUMEN

Inflammatory mediators constitute a recently coined term in the field of metal-based complexes with antiplatelet activities. Our strategy targets Platelet-Activating Factor (PAF) and its receptor, which is the most potent lipid mediator of inflammation. Thus, the antiplatelet (anti-PAF) potency of any substance could be exerted by inhibiting the PAF-induced aggregation in washed rabbit platelets (WRPs), which internationally is a well-accepted methodology. Herein, a series of mononuclear (mer-[Cr(pqx)Cl3(H2O]) (1), [Co(pqx)Cl2(DMF)] (2) (DMF = N,N'-dimethyl formamide), [Cu(pqx)Cl2(DMSO)] (3) (DMSO = dimethyl sulfoxide), [Zn(pqx)Cl2] (4)) and dinuclear complexes ([Mn(pqx)(H2O)2Cl2]2 (5), [Fe(pqx)Cl2]2 (6) and [Ni(pqx)Cl2]2 (7)) incorporating the 2-(2'-pyridyl)quinoxaline ligand (pqx), were biologically evaluated as inhibitors of the PAF- and thrombin-induced aggregation in washed rabbit platelets (WRPs). The molecular structure of the five-co-ordinate analog (3) has been elucidated by single-crystal X-ray diffraction revealing a trigonal bipyramidal geometry. All complexes are potent inhibitors of the PAF-induced aggregation in WRPs in the micromolar range. Complex (6) displayed a remarkable in vitro dual inhibition against PAF and thrombin, with IC50 values of 1.79 µM and 0.46 µM, respectively. Within the series, complex (5) was less effective (IC50 = 39 µM) while complex (1) was almost 12-fold more potent against PAF, as opposed to thrombin-induced aggregation. The biological behavior of complexes 1, 6 and 7 on PAF's basic metabolic enzymatic pathways reveals that they affect key biosynthetic and catabolic enzymes of PAF underlying the anti-inflammatory properties of the relevant complexes. The in vitro cytotoxic activities of all complexes in HEK293T (human embryonic kidney cells) and HeLa cells (cervical cancer cells) are described via the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The results reveal that complex 3 is the most potent within the series.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Elementos de Transición , Animales , Humanos , Conejos , Agregación Plaquetaria , Factor de Activación Plaquetaria/farmacología , Factor de Activación Plaquetaria/metabolismo , Plaquetas/metabolismo , Trombina/metabolismo , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Ligandos , Mediadores de Inflamación/metabolismo , Dimetilsulfóxido/farmacología , Quinoxalinas/farmacología , Células HEK293 , Células HeLa , Antineoplásicos/farmacología , Elementos de Transición/metabolismo
20.
Environ Res ; 239(Pt 2): 117275, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37827363

RESUMEN

The occurrence of antibiotic residues in diverse water sources has long been acknowledged as a potential health concern due to the emergence and spread of antibiotic-resistant bacteria and genes. However, there have been limited studies into the presence of antibiotic-metal complexes (AMCs) in real-time wastewater matrices, and their impact on wastewater microbial communities. The present work, in this regard, investigated the stability of Imipenem-metal complexes (Me = Mg (II), Ca (II), Fe (II), Cu (II), and Al (III)) with computational studies, stoichiometry with potentiometric measurements, and their antibacterial activity towards wastewater model microorganisms- Bacillus subtilis (B. subtilis) and Escherichia coli (E. Coli) by Colony Forming Unit (CFU) method. The lower energy of Imipenem-metal complexes than the parent antibiotic- Imipenem, during energy optimization using density functional (DFT) methods, revealed that metal interactions of Imipenem stabilize the drug by minimizing its energy. Further, CFU studies indicated that these complexes display higher antimicrobial activity than parent antibiotics. The electron delocalization over the entire chelated system (AMCs) reduces polarity and increases the lipophilicity of the complexes, thereby facilitating stronger interaction between AMCs and the bacterial cell membrane. Results indicate increased antibacterial activity of Imipenem-metal complexes for both E. coli and B. subtilis. The antibacterial activity, was however, more pronounced in B. subtilis, with >97% growth inhibition for metal complexes of Imipenem (at a Minimum Inhibitory Concentration of 20 nM or 6 ppb (i.e., MIC90)), for both the stoichiometric ratios (metal to ligand) ratios (M: L 1: 1 and 2: 1). All around, with increased stability and toxicity, AMCs are emerging as contaminants of concern and demand immediate attention to devise methods for their removal.


Asunto(s)
Complejos de Coordinación , Imipenem , Imipenem/toxicidad , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Complejos de Coordinación/farmacología , Aguas Residuales/toxicidad , Escherichia coli , Antibacterianos/toxicidad , Antibacterianos/química , Metales/química , Bacterias/metabolismo , Pruebas de Sensibilidad Microbiana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...