Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Inorg Biochem ; 261: 112706, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39197384

RESUMEN

In this study, the ligand EIPP (5-ethoxy-2-(1H-imidazo[4,5-f] [1,10] phenanthrolin-2-yl)phenol) and [Ir(ppy)2(EIPP)](PF6)] (5a, ppy = 2-phenylpyridine) and [Ir(piq)2(EIPP)](PF6)] (5b, piq = 1-phenylisoquinoline) were synthesized and they were entrapped into liposomes to produce 5alipo and 5blipo. 5a and 5b were characterized via HRMS, NMR, UV-vis and IR. The cytotoxicity of 5a, 5b, 5alipo and 5blipo on cancer and non-cancer cells was estimated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). MTT assay demonstrated that 5a and 5b did not show any significant cellular activity but their liposome-encapsulated 5alipo and 5blipo had significant toxic effects. The mechanism of 5alipo, 5blipo-inducing apoptosis was explored by studying cellular uptake, mitochondrial localization, mitochondrial membrane potential, cytochrome C, glutathione (GSH), malondialdehyde (MDA) and protein immunoblotting. The results demonstrated that 5alipo and 5blipo caused a release of cytochrome C, downregulated the expression of Bcl-2, upregulated the expression of BAX, activated caspase 3, and downregulated PARP expression. It was shown that 5alipo and 5blipo could inhibit cancer cell proliferation in G2/M phase by regulating p53 and p21 proteins. Additionally, 5alipo and 5blipo induced autophagy through an adjustment from LC3-I to LC3-II and caused ferroptosis. The in vivo antitumor activity of 5alipo was examined in detail.


Asunto(s)
Antineoplásicos , Apoptosis , Complejos de Coordinación , Iridio , Liposomas , Humanos , Iridio/química , Iridio/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Animales , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Ratones , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos
2.
Molecules ; 29(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38999026

RESUMEN

To investigate the impact of the electron-donating morpholinyl (morph) group on the ground- and excited-state properties of two different types of Ir(III) complexes, [IrCl3(R-C6H4-terpy-κ3N)] and [Ir(R-C6H4-terpy-κ3N)2](PF6)3, the compounds [IrCl3(morph-C6H4-terpy-κ3N)] (1A), 4[Ir(morph-C6H4-terpy-κ3N)2](PF6)3 (2A), [IrCl3(Ph-terpy-κ3N)] (1B) and [Ir(Ph-terpy-κ3N)2](PF6)3 (2B) were obtained. Their photophysical properties were comprehensively investigated with the aid of static and time-resolved spectroscopic methods accompanied by theoretical DFT/TD-DFT calculations. In the case of bis-terpyridyl iridium(III) complexes, the attachment of the morpholinyl group induced dramatic changes in the absorption and emission characteristics, manifested by the appearance of a new, very strong visible absorption tailing up to 600 nm, and a significant bathochromic shift in the emission of 2A relative to the model chromophore. The emission features of 2A and 2B were found to originate from the triplet excited states of different natures: intraligand charge transfer (3ILCT) for 2A and intraligand with a small admixture of metal-to-ligand charge transfer (3IL-3MLCT) for 2B. The optical properties of the mono-terpyridyl iridium(III) complexes were less significantly impacted by the morpholinyl substituent. Based on UV-Vis absorption spectra, emission wavelengths and lifetimes in different environments, transient absorption studies, and theoretical calculations, it was demonstrated that the visible absorption and emission features of 1A are governed by singlet and triplet excited states of a mixed MLLCT-ILCT nature, with a dominant contribution of the first component, that is, metal-ligand-to-ligand charge transfer (MLLCT). The involvement of ILCT transitions was reflected by an enhancement of the molar extinction coefficients of the absorption bands of 1A in the range of 350-550 nm, and a small red shift in its emission relative to the model chromophore.

3.
ACS Appl Mater Interfaces ; 16(30): 38916-38930, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39041453

RESUMEN

Despite the potential of photodynamic therapy (PDT) in cancer treatment, the development of efficient and photostable photosensitizing molecules that operate at long wavelengths of light has become a major hurdle. Here, we report for the first time an Ir(III)-phthalocyanine conjugate (Ir-ZnPc) as a novel photosensitizer for high-efficiency synergistic PDT treatment that takes advantage of the long-wavelength excitation and near infrared (NIR) emission of the phthalocyanine scaffold and the known photostability and high phototoxicity of cyclometalated Ir(III) complexes. In order to increase water solubility and cell membrane permeability, the conjugate and parent zinc phthalocyanine (ZnPc) were encapsulated in amphoteric redox-responsive polyurethane-polyurea hybrid nanocapsules (Ir-ZnPc-NCs and ZnPc-NCs, respectively). Photobiological evaluations revealed that the encapsulated Ir-ZnPc conjugate achieved high photocytotoxicity in both normoxic and hypoxic conditions under 630 nm light irradiation, which can be attributed to dual Type I and Type II reactive oxygen species (ROS) photogeneration. Interestingly, PDT treatments with Ir-ZnPc-NCs and ZnPc-NCs significantly inhibited the growth of three-dimensional (3D) multicellular tumor spheroids. Overall, the nanoencapsulation of Zn phthalocyanines conjugated to cyclometalated Ir(III) complexes provides a new strategy for obtaining photostable and biocompatible red-light-activated nano-PDT agents with efficient performance under challenging hypoxic environments, thus offering new therapeutic opportunities for cancer treatment.


Asunto(s)
Antineoplásicos , Indoles , Isoindoles , Fotoquimioterapia , Fármacos Fotosensibilizantes , Humanos , Indoles/química , Indoles/farmacología , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Iridio/química , Iridio/farmacología , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Compuestos de Zinc/química , Especies Reactivas de Oxígeno/metabolismo , Nanocápsulas/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos
4.
Eur J Med Chem ; 276: 116648, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-38968786

RESUMEN

Medical applications of iridium (III) complexes include their use as state-of-the-art theranostic agents - molecules that combine therapeutic and diagnostic functions into a single entity. These complexes offer a promising avenue in medical diagnostics, precision imaging at single-cell resolution and targeted anticancer therapy due to their unique properties. In this review we report a short summary of their application in medical diagnostics, imaging at single-cell level and targeted anticancer therapy. The exceptional photophysical properties of Iridium (III) complexes, including their brightness and photostability, make them excellent candidates for bioimaging. They can be used to image cellular processes and the microenvironment within single cells with unprecedented clarity, aiding in the understanding of disease mechanisms at the molecular level. Moreover the iridium (III) complexes can be designed to selectively target cancer cells,. Upon targeting, these complexes can act as photosensitizers for photodynamic therapy (PDT), generating reactive oxygen species (ROS) upon light activation to induce cell death. The integration of diagnostic and therapeutic capabilities in Iridium (III) complexes offers the potential for a holistic approach to cancer treatment, enabling not only the precise eradication of cancer cells but also the real-time monitoring of treatment efficacy and disease progression. This aligns with the goals of personalized medicine, offering hope for more effective and less invasive cancer treatment strategies.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Iridio , Neoplasias , Humanos , Iridio/química , Iridio/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/síntesis química , Neoplasias/tratamiento farmacológico , Neoplasias/diagnóstico por imagen , Neoplasias/patología , Neoplasias/diagnóstico , Fotoquimioterapia , Nanomedicina Teranóstica , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/síntesis química , Animales , Medicina de Precisión , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/síntesis química , Análisis de la Célula Individual
5.
J Inorg Biochem ; 259: 112652, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38945112

RESUMEN

Anticancer activity has been extensively studies. In this article, three ligands 2-(6-bromobenzo[d][1,3]dioxol-5-yl)-1H-imidazo[4,5-f][1,10]phenanthroline (BDIP), 2-(7-methoxybenzo[d][1,3]dioxol-5-yl)-1H-imidazo[4,5-f][1,10]phenanthroline (MDIP), 2-(6-nitrobenzo[d][1,3]dioxol-5-yl)-1H-imidazo[4,5-f][1,10]phenanthroline (NDIP) and their iridium(III) complexes: [Ir(ppy)2(BDIP)](PF6) (ppy = deprotonated 2-phenylpyridine, 3a), [Ir(ppy)2(MDIP)](PF6) (3b) and [Ir(ppy)2(NDIP)](PF6) (3c) were synthesized. The cytotoxicity of 3a, 3b, 3c against Huh7, A549, BEL-7402, HepG2, HeLa, and non-cancer NIH3T3 was tested using 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method. The results obtained from the MTT test stated clearly that these complexes demonstrated moderate or non-cytotoxicity toward Huh7, BEL-7402, HepG2 and HeLa except A549 cells. To improve the anticancer efficacy, we used white light to irradiate the mixture of cells and complexes for 30 min, the anticancer activity of the complexes was greatly enhanced. Particularly, 3a and 3b exhibited heightened capability to inhibit A549 cells proliferation with IC50 (half maximal inhibitory concentration) values of 0.7 ± 0.3 µM and 1.8 ± 0.1 µM, respectively. Cellular uptake has shown that 3a and 3b can be accumulated in the cytoplasm. Wound healing and colony forming showed that 3a and 3b significantly hinder the cell migration and growth in the S phase. The complexes open mitochondrial permeability transition pore (MPTP) channel and cause the decrease of membrane potential, release of cytochrome C, activation of caspase 3, and finally lead to apoptosis. In addition, 3a and 3b cause autophagy, increase the lipid peroxidation and lead to ferroptosis. Also, 3a and 3b increase the expression of calreticulin (CRT), high mobility group box 1 (HMGB1), heat shock protein 70 (HSP70), thereby inducing immunogenic cell death.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Iridio , Neoplasias Pulmonares , Iridio/química , Iridio/farmacología , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Células A549 , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Apoptosis/efectos de los fármacos , Luz , Animales , Ratones , Proliferación Celular/efectos de los fármacos , Células 3T3 NIH
6.
J Inorg Biochem ; 257: 112596, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38759264

RESUMEN

The design and synthesis of a series of metal complexes formed by non-steroidal anti-inflammatory drugs (NSAIDs) ibuprofen (IBP) and iridium(III), with the molecular formula [Ir(C^N)2bpy(4-CH2OIBP-4'-CH2OIBP)](PF6) (Ir-IBP-1, Ir-IBP-2) (C^N = 2-phenylpyridine (ppy, Ir-IBP-1), 2-(2-thienyl)pyridine (thpy, Ir-IBP-2)) was introduced in this article. Firstly, it was found that the anti-proliferative activity of these complexes was more effective than that of cisplatin. Further research showed that Ir-IBP-1 and Ir-IBP-2 can accumulate in intracellular mitochondria, thereby disrupting mitochondrial membrane potential (MMP), increasing intracellular reactive oxygen species (ROS), blocking the G2/M phase of the cell cycle, and inducing cell apoptosis. In terms of protein expression, the expression of COX-2, MMP-9, NLRP3 and Caspase-1 proteins can be downregulated, indicating their ability to anti-inflammatory and overcome immune evasion. Furthermore, Ir-IBP-1 and Ir-IBP-2 can induce immunogenic cell death (ICD) by triggering the release of cell surface calreticulin (CRT), high mobility group box 1 (HMGB1) and adenosine triphosphate (ATP). Overall, iridium(III)-IBP conjugates exhibit various anti-tumor mechanisms, including mitochondrial damage, cell cycle arrest, inflammatory suppression, and induction of ICD.


Asunto(s)
Antineoplásicos , Apoptosis , Complejos de Coordinación , Ibuprofeno , Iridio , Iridio/química , Iridio/farmacología , Humanos , Ibuprofeno/farmacología , Ibuprofeno/química , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Apoptosis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/síntesis química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos
7.
J Inorg Biochem ; 256: 112549, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38579631

RESUMEN

Herein, we synthesized and characterized two novel iridium (III) complexes: [Ir(bzq)2(PPD)](PF6) (4a, with bzq = deprotonated benzo[h]quinoline and PPD = pteridino[6,7-f][1,10]phenanthroline-11,13-diamine) and [Ir(piq)2(PPD)](PF6) (4b, with piq = deprotonated 1-phenylisoquinoline). The anticancer efficacy of these complexes, 4a and 4b, was investigated using 3-(4,5-dimethylthiazole)-2,5-diphenltetraazolium bromide (MTT). Complex 4a exhibited no cytotoxic activity, while 4b demonstrated moderate efficacy against SGC-7901, A549, and HepG2 cancer cells. To enhance their anticancer potential, we explored two strategies: (I) light irradiation and (II) encapsulation of the complexes in liposomes, resulting in the formation of 4alip and 4blip. Both strategies significantly increased the ability of 4a, 4b to kill cancer cells. The cellular studies indicated that both the free complexes 4a, 4b and their liposomal forms 4alip and 4blip effectively inhibited cell proliferation. The cell cycle arrest analysis uncovered 4alip and 4blip arresting cell growth in the S period. Additionally, we investigated apoptosis and ferroptosis pathways, observing an increase in malondialdehyde (MDA) levels, a reduction of glutathione (GSH), a down-regulation of GPX4 (glutathione peroxidase) expression, and lipid peroxidation. The effects on mitochondrial membrane potential and intracellular Ca2+ concentrations were also examined, revealing that both light-activated and liposomal forms of 4alip and 4blip caused a decline in mitochondrial membrane potential and an enhancement in intracellular Ca2+ levels. In conclusion, these complexes and them encapsulated liposomes induce cell death through apoptosis and ferroptosis.


Asunto(s)
Antineoplásicos , Apoptosis , Complejos de Coordinación , Iridio , Liposomas , Humanos , Iridio/química , Iridio/farmacología , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Potencial de la Membrana Mitocondrial/efectos de los fármacos
8.
Anal Chim Acta ; 1304: 342470, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38637058

RESUMEN

BACKGROUND: Iridium(III) complexes, exhibiting high luminescence quantum yields and a wide range of emission colours, are promising alternatives to tris(2,2'-bipyridine)ruthenium(II) for chemiluminescence (CL) and electrochemiluminescence (ECL) detection. This emerging class of reagent, however, is limited by the poor solubility of many iridium(III) complexes in aqueous solution, and lack of understanding of their remarkably variable selectivities towards different analytes. RESULTS: Seven [Ir(C^N)2(pt-TEG)]+ complexes, exhibiting a wide range of reduction potentials and emission energies, were examined with six model analytes. For CL, cerium(IV) was used as the oxidant. The alkylamine analytes generally produced greater CL and ECL with the more readily oxidised Ir(III) complexes (C^N = piq, bt, ppy), predominantly through the 'direct' pathway requiring oxidation of both metal complex and analyte. Aniline derivatives that did not also contain secondary or tertiary alkylamines elicited CL from the less readily oxidised complexes (C^N = df-ppy-CF3, df-ppy) via energy transfer. The most difficult to oxidise complexes (C^N = df(CF3)-ppy-Me, df(CN)-ppy) gave poor responses due to the limited potential window of the solvent and inefficiency of energy transfer to their high energy excited states. Greater CL and/or ECL intensities were generally obtained for each analyte with at least one Ir(III) complex than with [Ru(bpy)3]2+; superior limits of detection for two analytes were demonstrated. SIGNIFICANCE: This exploration of CL/ECL in which the properties of luminophore, analyte and oxidant are all varied provides a new understanding of the influence of the metal-complex potentials and excited state energy on the light-producing and quenching pathways, and consequently, their distinct selectivity towards different analytes. These findings will guide the development of water-soluble Ir(III) complexes as CL and ECL reagents.

9.
Luminescence ; 39(3): e4700, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38506122

RESUMEN

Three new neutral and ionic phosphorescent iridium(III) complexes were successfully prepared using 1-(6-methoxynaphthalen-2-yl)isoquinoline as the main ligand, while the auxiliary ligand was 2-(2-1H-imidazolyl)pyridine. Three complexes (Ir1, Ir2, Ir3) showed red emission, peaking at 610, 609, and 615 nm, respectively, and they exhibited good solubility and excellent photophysical properties in different solvents, which is suitable to prepare organic light-emitting diodes (OLEDs) by solution method. Among the three OLEDs prepared by iridium(III) complexes using the solution method, the device based on Ir2 possessed better electroluminescent properties, and its maximum brightness, current efficiency (CE), power efficiency (PE), and the maximum external quantum efficiency (EQE) were 507.2 cd m-2 , 0.14 cd A-1 , 0.06 lm W-1 , and 0.14%. respectively, proving that the three complexes have a certain of potential for OLEDs applications and are expected to expand the applications of iridium(III) complexes for OLEDs.


Asunto(s)
Iridio , Ligandos , Iones , Solubilidad , Solventes
10.
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
11.
Eur J Med Chem ; 265: 116112, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38183779

RESUMEN

This paper unveils a novel perspective on synthesis and characterization of the ligand 5-bromo-2-amino-2'-(phenyl-1H-imidazo[4,5-f][1,10]phenanthroline) (BAPIP), and its iridium(III) complexes [Ir(PPY-)2(BAPIP)](PF6) (1a, with PPY- as deprotonated 2-phenylpyridine), [Ir(PIQ-)2(BAPIP)](PF6) (1b, piq- denoting deprotonated 1-phenylisoquinoline), and [Ir(BZQ-)2(BAPIP)](PF6) (1c, bzq- signifying deprotonated benzo[h]quinoline). Systematic evaluation of the cytotoxicity of 1a, 1b, and 1c across diverse cell lines encompassing B16, HCT116, HepG2, A549, HeLa, and LO2 using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. Unexpectedly, compounds 1b and 1c demonstrated no cytotoxicity against the above cell lines. Motivated by the pursuit of heightened anti-proliferative potential, a strategic encapsulation approach yielded liposomes 1alip, 1blip, and 1clip. As expectation, 1alip, 1blip, and 1clip displayed remarkable anti-proliferative efficacy, particularly noteworthy in A549 cells, exhibiting IC50 values of 4.9 ± 1.0, 5.9 ± 0.1, and 7.6 ± 0.2 µM, respectively. Moreover, our investigation illuminated the mitochondrial accumulation of these liposomal entities, 1alip, 1blip, and 1clip, evoking apoptosis through the mitochondrial dysfunction mediated by reactive oxygen species (ROS). The ferroptosis was confirmed by decrease in glutathione (GSH) concentrations, the downregulation of glutathione peroxidase 4 (GPX4), increase of high mobility group protein 1 (HMGB1), and lipid peroxidation. Simultaneously, pyroptosis as another mode of cell death was undertaken. RNA-sequencing was employed to investigate intricate signalling pathways. In vivo examination provided tangible evidence of 1alip in effectively curbing tumor growth. Collectively, this study provides a multifaceted mode of cellular demise orchestrated by 1a, 1alip, 1blip, and 1clip, involving pathways encompassing apoptosis, ferroptosis, and pyroptosis.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Ferroptosis , Humanos , Liposomas , Línea Celular Tumoral , Iridio/farmacología , Gasderminas , Piroptosis , Proliferación Celular , Apoptosis , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Chembiochem ; 25(4): e202300798, 2024 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-38169080

RESUMEN

Site-specific modification of proteins with synthetic fluorescent tag effectively improves the resolution of imaging, and such a labeling method with negligible three-dimensional structural perturbations and minimal impact on the biological functions of proteins is of high interest to dissect the high-resolution activities of biomolecules in complex systems. To this end, several non-emissive iridium(III) complexes [Ir(C-N)2 (H2 O)2 ]+ OTF- (C-N denotes various cyclometalated ligands) were designed and synthesized. These complexes were tested for attaching a protein by coordinating to H/X (HisMet, HisHis, and HisCys) that are separated by i and i+4 in α-helix. Replacement of the two labile water ligands in the iridium(III) complex by a protein HisHis pair increases the luminescent intensity up to over 100 folds. This labeling approach has been demonstrated in a highly specific and efficient manner in a number of proteins, and it is also feasible for labeling target proteins in cell lysates.


Asunto(s)
Iridio , Luminiscencia , Iridio/química
13.
Molecules ; 29(1)2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38202839

RESUMEN

Iridium(III) complexes are emerging as a promising tool in the area of detection and therapy due to their prominent photophysical properties, including higher photostability, tunable phosphorescence emission, long-lasting phosphorescence, and high quantum yields. In recent years, much effort has been devoted to develop novel near-infrared (NIR) iridium(III) complexes to improve signal-to-noise ratio and enhance tissue penetration. In this review, we summarize different classes of organometallic NIR iridium(III) complexes for detection and therapy, including cyclometalated ligand-enabled NIR iridium(III) complexes and NIR-dye-conjugated iridium(III) complexes. Moreover, the prospects and challenges for organometallic NIR iridium(III) complexes for targeted detection and therapy are discussed.


Asunto(s)
Iridio , Relación Señal-Ruido
14.
Small ; 20(12): e2307500, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37940631

RESUMEN

The promising cyclometalated iridium (III) complexes have been proved to possess great potential in vacuum-deposited organic light-emitting diodes (OLEDs) applications for full-color displays and white solid-state lighting sources. Herein, based on the unique bidentate ligand of dibenzo[a,c]phenazine (dbpz) group with strong conjugated effect of aromatic rings for red emission, four novel [3+2+1] coordinated iridium (III) emissive materials have been rationally designed and synthesized. The monodentate ligands of -CN and -OCN have been effectively employed to tune the deep-red emission of 628-675 nm with high photoluminescence quantum yields up to 98%. Moreover, all devices displayed deep-red color coordinates ranging from (0.675, 0.325) to (0.716, 0.284), which is close to the standard-red color coordinates of (0.708, 0.292), as recommended by International Telecommunication Union Radiocommunication (ITU-R) BT.2020. The device based on nBuIr(dbpz)CN with an exciplex cohost has exhibited maximum external quantum efficiencies of 20.7% and good stability. With nBuIr(dbpz)CN as an effective sensitizer, the nBuIr(dbpz)OCN based phosphorescent OLED devices have successfully demonstrated cascading energy transfer processes, contributing to pure red emission with maximum luminance as high as 6471 cd m-2. Therefore, this work has been successfully demonstrated rational molecular design strategy of [3+2+1] iridium complexes to obtain highly efficient deep-red electrophosphorescent emission.

15.
Front Cell Infect Microbiol ; 13: 1200747, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37545853

RESUMEN

Candida albicans (C. albicans) is a ubiquitous clinical fungal pathogen. In recent years, combination therapy, a potential treatment method to overcome C. albicans resistance, has gained traction. In this study, we synthesized a series of cyclometalated iridium(III) complexes with the formula [Ir(C-N)2(tpphz)](PF6) (C-N = 2-phenylpyridine (ppy, in Ir1), 2-(2-thienyl)pyridine (thpy, in Ir2), 2-(2,4-difluorophenyl) pyridine (dfppy, in Ir3), tpphz = tetrapyrido[3,2-a:2',3'-c:3'',2''-h:2''',3'''-j]phenazine) and polypyridyl ruthenium(II) complexes with the formula [Ru(N-N)2(tpphz)](PF6)2 (N-N = 2,2'-bipyridine (bpy, in Ru1), 1,10-phenanthroline (phen, in Ru2), 4,7-diphenyl-1,10-phenanthroline (DIP, in Ru3)), and investigated their antifungal activities against drug-resistant C. albicans and their combination with fluconazole (FLC). Of which, the combination of the lead iridium(III) complex Ir2 and FLC showed strong antifungal activity against drug-resistant C. albicans. Mechanism studies have shown that they can inhibit the formation of hyphae and biofilm, damage mitochondrial function and accumulate intracellular ROS. Therefore, iridium(III) complexes combined with FLC can be used as a promising treatment to exert anti-drug-resistant C. albicans activity, in order to improve the treatment efficiency of fungal infection.


Asunto(s)
Antifúngicos , Fluconazol , Fluconazol/farmacología , Antifúngicos/farmacología , Candida albicans , Iridio/farmacología , Piridinas/farmacología
16.
Materials (Basel) ; 16(13)2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37445016

RESUMEN

Two novel ionic red/near-infrared Ir(III) complexes (Ir1 and Ir2) were reasonably designed and prepared using 2-(1-isoquinolinyl)-9,10-anthraquinone as the main ligand and 4,4'-dimethyl-2,2'-bipyridyl and 4,4'-dimethoxy-2,2'-bipyridyl as the auxiliary ligands, respectively. Both complexes showed bright phosphorescence in solution (peak at 618 nm with a shoulder at 670 nm). Interestingly, the phosphorescence peak of two Ir(III) complexes showed a blue-shift of about 36 nm after being ground. Simultaneously, both complexes exhibited mechanical force-induced enhanced emission, and the intensity of the luminescence for Ir1 and Ir2 increased by around two times compared to the one before being ground, respectively. Powder X-ray diffraction (PXRD) and time-dependent density functional theory (TD-DFT) calculation were utilized to understand well the mechanism of this phenomenon and suggested that the destruction of the well-ordered crystalline nature and the decline in triplet-triplet annihilation maybe responsible for the pressure-induced blue-shift and the enhancement of the phosphorescence.

17.
Molecules ; 28(12)2023 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-37375420

RESUMEN

Two novel neutral phosphorescent iridium(III) complexes (Ir1 and Ir2) were rationally designed and synthesized with high yields using 10,11,12,13-tetrahydrodibenzo[a,c]phenazine as the main ligand. The two complexes showed bright-red phosphorescence (625 nm for Ir1, and 620 nm for Ir2, in CH2Cl2), high-luminescence quantum efficiency (0.32 for Ir1, and 0.35 for Ir2), obvious solvatochromism and good thermostability. Then, they were used to fabricate high-efficiency red OLEDs via vacuum evaporation; the maximum current efficiency, power efficiency, and external quantum efficiency of the red devices based on Ir1 and Ir2 are 13.47/15.22 cd/A, 10.35/12.26 lm/W, and 10.08/7.48%, respectively.

18.
Molecules ; 28(3)2023 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-36770897

RESUMEN

Improvements in the design of receptors for the detection and quantification of anions are desirable and ongoing in the field of anion chemistry, and remarkable progress has been made in this direction. In this regard, the development of luminescent chemosensors for sensing anions is an imperative and demanding sub-area in supramolecular chemistry. This decade, in particular, witnessed advancements in chemosensors based on ruthenium and iridium complexes for anion sensing by virtue of their modular synthesis and rich chemical and photophysical properties, such as visible excitation wavelength, high quantum efficiency, high luminescence intensity, long lifetimes of phosphorescence, and large Stokes shifts, etc. Thus, this review aims to summarize the recent advances in the development of ruthenium(II) and iridium(III)-based complexes for their application as luminescent chemosensors for anion sensing. In addition, the focus was devoted to designing aspects of polypyridyl complexes of these two transition metals with different recognition motifs, which upon interacting with different inorganic anions, produces desirable quantifiable outputs.

19.
J Inorg Biochem ; 241: 112134, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36706490

RESUMEN

In this study, two new iridium(III) polypyridyl complexes [Ir(bzq)2(DIPH)](PF6) (bzq = deprotonated benzo[h]quinoline, DIPH = 4-(2,5-dibromo-4-(1H-imidazo[4,5-f][1,10]phenanthrolim-2-yl)-4-hydroxybutan-2-one) (Ir1) and [Ir(piq)2(DIPH)](PF6) (piq = deprotonated 1-phenylisoquinoline) (Ir2) were synthesized and characterized by elemental analysis, HRMS, 1H and 13C NMR. The cytotoxic activity of Ir1, Ir2, Ir1lipo and Ir2lipo against cancer cells SGC-7901, HepG2, A549, HeLa, B16 and normal NIH3T3 cells in vitro was evaluated using 3-(4,5-dimethylthiazole-2-yl)-2,5-biphenyl tetrazolium bromide (MTT) method. Ir1 and Ir2 showed no cytotoxic activity, but their liposome-entrapped Ir1 (Ir1lipo) and Ir2 (Ir2lipo) showed significant cellular activity, especially sensitive to SGC-7901 with IC50 values of 4.7 ± 0.2 and 12.4 ± 0.5 µM, respectively. The cellular uptake, endoplasmic reticulum (ER) localization, autophagy, tubulin polymerization, glutathione (GSH), malondialdehyde (MDA) and release of cytochrome c were investigated to explore the mechanisms of apoptosis. The calreticulin (CRT), heat shock protein 70 (HSP70), high mobility group box 1 (HMGB1) were also explored. Western blotting showed that Ir1lipo and Ir2lipo inhibited PI3K (phosphoinositide-3 kinase), AKT (protein kinase B), p-AKT and activated Bcl-2 (B-cell lymphoma-2) protein and apoptosis-regulated factor caspase 3 (cysteinyl aspartate specific proteinase-3) and cleaving PARP (poly ADP-ribose polymerase). The results demonstrated that Ir1lipo and Ir2lipo induce cell apoptosis through targeting the endoplasmic reticulum (ER), cause oxidative stress damage, inhibiting PI3K/AKT signaling pathway, immunogenic cell death (ICD) and inhibit the cell growth at G2/M phase.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Ratones , Animales , Humanos , Línea Celular Tumoral , Liposomas/farmacología , Iridio/química , Proteínas Proto-Oncogénicas c-akt , Complejos de Coordinación/química , Células 3T3 NIH , Fosfatidilinositol 3-Quinasas , Proliferación Celular , Antineoplásicos/farmacología , Apoptosis
20.
J Inorg Biochem ; 238: 112054, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36335745

RESUMEN

Ligand HMSPIP (2-(4-(methylsulfonyl)phenyl)-1H-imidazo[4,5-f][1,10]phenanthroline) and its iridium(III) complexes [Ir(ppy)2(HMSPIP)]PF6 (ppy = 2-phenylpyridine, Ir1) and [Ir(bzq)2(HMSPIP)]PF6 (bzq = benzo[h]quinoline, Ir2) were synthesized. The complexes were characterized by 1H NMR, 13C NMR, and UV/Vis spectra. The cytotoxicity of the complexes toward cancer cells were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method, the scratch wound healing and colony-forming were also investigated. MTT assay certificated that the complexes show high toxic effect on the HeLa cells. The cell cycle assay illustrated that the complexes blocked cell growth at G0/G1 phase in HeLa cells. A series of subsequent experiments showed that the complexes first enter the endoplasmic reticulum (ER) and then enter the mitochondria, leading to an increase in intracellular Ca2+ and reactive oxygen species (ROS) content, depolarizing mitochondrial membrane potential (MMP), and ultimately resulting in apoptosis. In addition, the experimental results revealed that the complexes not only increase the level of ROS but also inhibit the production of GSH and eventually produce large amounts of MDA and further leading to cell death. Taken together, we consider that the complexes can be used as potential candidate drugs for HeLa cancer treatment.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Humanos , Iridio/química , Células HeLa , Especies Reactivas de Oxígeno/metabolismo , Complejos de Coordinación/química , Línea Celular Tumoral , Antineoplásicos/química , Mitocondrias , Retículo Endoplásmico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA