Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Anal Chem ; 96(17): 6666-6673, 2024 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-38623755

RESUMEN

Nitric oxide (NO) is a crucial signal molecule closely linked to the biological immune response, especially in macrophage polarization. When activated, macrophages enter a pro-inflammatory state and produce NO, a marker for the M1 phenotype. In contrast, the anti-inflammatory M2 phenotype does not produce NO. We developed a mitochondria-targeted two-photon iridium-based complex (Ir-ImNO) probe that can detect endogenous NO and monitor macrophages' different immune response states using various imaging techniques, such as one- and two-photon phosphorescence imaging and phosphorescence lifetime imaging. Ir-ImNO was used to monitor the immune activation of macrophages in mice. This technology aims to provide a clear and comprehensive visualization of macrophage immune responses.


Asunto(s)
Macrófagos , Mitocondrias , Óxido Nítrico , Óxido Nítrico/análisis , Óxido Nítrico/metabolismo , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Mitocondrias/metabolismo , Mitocondrias/química , Ratones , Células RAW 264.7 , Iridio/química , Imagen Multimodal , Colorantes Fluorescentes/química , Ratones Endogámicos C57BL , Imagen Óptica
2.
Angew Chem Int Ed Engl ; 63(31): e202405679, 2024 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-38771671

RESUMEN

An optimal cancer chemotherapy regimen should effectively address the drug resistance of tumors while eliciting antitumor-immune responses. Research has shown that non-apoptotic cell death, such as pyroptosis and ferroptosis, can enhance the immune response. Despite this, there has been limited investigation and reporting on the mechanisms of oncosis and its correlation with immune response. Herein, we designed and synthesized a Ru(II) complex that targeted the nucleus and mitochondria to induce cell oncosis. Briefly, the Ru(II) complex disrupts the nucleus and mitochondria DNA, which active polyADP-ribose polymerase 1, accompanied by ATP consumption and porimin activation. Concurrently, mitochondrial damage and endoplasmic reticulum stress result in the release of Ca2+ ions and increased expression of Calpain 1. Subsequently, specific pore proteins porimin and Calpain 1 promote cristae destruction or vacuolation, ultimately leading to cell membrane rupture. The analysis of RNA sequencing demonstrates that the Ru(II) complex can initiate the oncosis-associated pathway and activate both innate and adaptive immunity. In vivo experiments have confirmed that oncosis promotes dendritic cell maturation and awakens adaptive cytotoxic T lymphocytes but also activates the innate immune by inducing the polarization of macrophages towards an M1 phenotype.


Asunto(s)
Inmunidad Adaptativa , Complejos de Coordinación , Inmunidad Innata , Rutenio , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Rutenio/química , Rutenio/farmacología , Humanos , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Animales , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral
3.
Anal Chem ; 95(43): 15956-15964, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37856322

RESUMEN

Nitric oxide (NO) serves as a ubiquitous and fundamental signaling molecule involved in intricate effects on both physiological and pathological processes. NO, biosynthesized by nitric oxide synthase (NOS) or generated from nitrite, can form nitrosation reagent N2O3 (4NO + O2 = 2N2O3) through its oxidation or quickly produce peroxynitrite anion ONOO- (NO + •O2- = ONOO-) by reacting with superoxide anion (•O2-). However, most of the existing luminescent probes for NO just focus on specificity and utilize only a single signal to distinguish products N2O3 or ONOO-. In most of the present work, they differentiate one product from another simply by fluorescence signal or fluorescence intensity, which is not enough to distinguish accurately the behavior of NO in living cells. Herein, a new mitochondria-targeted and two-photon near-infrared (NIR) phosphorescent iridium(III) complex, known as Ir-NBD, has been designed for accurate detection and simultaneous imaging of two downstream products of endogenous NO, i.e., N2O3 and ONOO-. Ir-NBD exhibits a rapid response to N2O3 and ONOO- in enhanced phosphorescence intensity, increased phosphorescence lifetime, and an exceptionally high two-photon cross-section, reaching values of 78 and 85 GM, respectively, after the reaction. Furthermore, we employed multiple imaging methods, phosphorescence intensity imaging, and phosphorescence lifetime imaging together to image even distinguish N2O3 and ONOO- by probe Ir-NBD. Thus, coupled with its excellent photometrics, Ir-NBD enabled the detection of the basal level of intracellular NO accurately by responding to N2O3 and ONOO- in the lipopolysaccharide-stimulated macrophage model in virtue of fluorescence signal and phosphorescence lifetime imaging, revealing precisely the endogenous mitochondrial NO distribution during inflammation in a cell environment.


Asunto(s)
Iridio , Óxido Nítrico , Óxido Nítrico/metabolismo , Oxidación-Reducción , Mitocondrias/metabolismo , Fotones , Ácido Peroxinitroso/metabolismo , Colorantes Fluorescentes/metabolismo
4.
J Am Chem Soc ; 144(9): 4091-4101, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35171598

RESUMEN

Despite the clinical success of photodynamic therapy (PDT), the application of this medical technique is intrinsically limited by the low oxygen concentrations found in cancer tumors, hampering the production of therapeutically necessary singlet oxygen (1O2). To overcome this limitation, we report on a novel mitochondria-localized iridium(III) endoperoxide prodrug (2-O-IrAn), which, upon two-photon irradiation in NIR, synergistically releases a highly cytotoxic iridium(III) complex (2-IrAn), singlet oxygen, and an alkoxy radical. 2-O-IrAn was found to be highly (photo-)toxic in hypoxic tumor cells and multicellular tumor spheroids (MCTS) in the nanomolar range. To provide cancer selectivity and improve the pharmacological properties of 2-O-IrAn, it was encapsulated into a biotin-functionalized polymer. The generated nanoparticles were found to nearly fully eradicate the tumor inside a mouse model within a single treatment. This study presents, to the best of our knowledge, the first example of an iridium(III)-based endoperoxide prodrug for synergistic photodynamic therapy/photoactivated chemotherapy, opening up new avenues for the treatment of hypoxic tumors.


Asunto(s)
Neoplasias , Fotoquimioterapia , Profármacos , Animales , Línea Celular Tumoral , Hipoxia/tratamiento farmacológico , Iridio/farmacología , Ratones , Mitocondrias , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Profármacos/farmacología , Profármacos/uso terapéutico , Oxígeno Singlete/uso terapéutico
5.
Angew Chem Int Ed Engl ; 61(28): e202205429, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35532958

RESUMEN

The clinical application of photodynamic therapy is hindered by the high glutathione concentration, poor cancer-targeting properties, poor drug loading into delivery systems, and an inefficient activation of the cell death machinery in cancer cells. To overcome these limitations, herein, the formulation of a promising IrIII complex into a biodegradable coordination polymer (IrS NPs) is presented. The nanoparticles were found to remain stable under physiological conditions but deplete glutathione and disintegrate into the monomeric metal complexes in the tumor microenvironment, causing an enhanced therapeutic effect. The nanoparticles were found to selectively accumulate in the mitochondria where these trigger cell death by hybrid apoptosis and ferroptosis pathways through the photoinduced production of singlet oxygen and superoxide anion radicals. This study presents the first example of a coordination polymer that can efficiently cause cancer cell death by apoptosis and ferroptosis upon irradiation, providing an innovative approach for cancer therapy.


Asunto(s)
Complejos de Coordinación , Ferroptosis , Fotoquimioterapia , Apoptosis , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , Glutatión , Iridio/farmacología , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Polímeros/farmacología
6.
Angew Chem Int Ed Engl ; 61(33): e202204866, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35736788

RESUMEN

The application of G-quadruplex stabilizers presents a promising anticancer strategy. However, the molecular crowding conditions within cells diminish the potency of current G-quadruplex stabilizers. Herein, chiral RuII -PtII dinuclear complexes were developed as highly potent G-quadruplex stabilizers even under challenging molecular crowding conditions. The compounds were encapsulated with biotin-functionalized DNA cages to enhance sub-cellular localization and provide cancer selectivity. The nanoparticles were able to efficiently inhibit the endogenous activities of telomerase in cisplatin-resistant cancer cells and cause cell death by apoptosis. The nanomaterials demonstrated high antitumor activity towards cisplatin-resistant tumor cells as well as tumor-bearing mice. To the best of our knowledge, this study presents the first example of a RuII -PtII dinuclear complex as a G-quadruplex stabilizer with an anti-cancer effect towards drug-resistant tumors inside an animal model.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , G-Cuádruplex , Neoplasias , Rutenio , Telomerasa , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Cisplatino/metabolismo , Cisplatino/farmacología , Complejos de Coordinación/metabolismo , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , ADN , Ratones , Rutenio/metabolismo , Rutenio/farmacología , Telomerasa/genética , Telómero
7.
Anal Chem ; 93(22): 8062-8070, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34037386

RESUMEN

Peroxynitrite (ONOO-) and glutathione (GSH), two unique reactive species, play an essential regulating role in the oxidation and antioxidation in the living body and are closely associated with various physiological and pathological processes, like cancer, cardiovascular disorders, diabetes, inflammation, Alzheimer's disease, and hepatotoxicity. Thus, it is crucial to study mitochondria ONOO-/GSH redox cycles by an effective molecular tool. In this work, a mitochondria-targeting and redox-reversible near-infrared (NIR) phosphorescent iridium complex, Ir-diol, has been synthesized and used for the detection and imaging of a cellular redox state by visualizing endogenous ONOO-/GSH content. Ir-diol shows excellent photophysical properties, including NIR emission (the maximum emissive wavelength for 704 nm, approximately) and high phosphorescent quantum yield (Φ = 0.136) and exhibits high sensitivity and selectivity toward ONOO-/GSH redox cycles in aqueous solution and living cells. Therefore, these features, combined with low cytotoxicity and excellent cell permeability, enable probe Ir-diol to monitor the changes of the intracellular ONOO-/GSH level induced by drug both in vitro and in vivo.


Asunto(s)
Colorantes Fluorescentes , Iridio , Glutatión/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Ácido Peroxinitroso
8.
Small ; 17(1): e2005086, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33284508

RESUMEN

The epigenetic dysregulation and hypoxia are two important factors that drive tumor malignancy, and N6 -methyladenosine (m6 A) in mRNA is involved in the regulation of gene expression. Herein, a nanocatalyst OsSx -PEG (PEG = poly(ethylene glycol)) nanoparticles (NPs) as O2 modulator is developed to improve tumor hypoxia. OsSx -PEG NPs can significantly downregulate genes involved in hypoxia pathway. Interestingly, OsSx -PEG NPs elevate RNA m6 A methylation levels to cause the m6 A-dependent mRNA degradation of the hypoxia-related genes. Moreover, OsSx -PEG NPs can regulate the expression of RNA m6 A methyltransferases and demethylases. Finally, DOX@OsSx -PEG (DOX = doxorubicin; utilized as a model drug) NPs modulate tumor hypoxia and regulate mRNA m6 A methylation of hypoxia-related genes in vivo. As the first report about relationship between catalytic nanomaterials and RNA modifications, the research opens a new avenue for unveiling the underlying action mechanisms of hypoxia-modulating nanomaterials and shows potential of regulating RNA modification to overcome chemoresistance.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Doxorrubicina , Humanos , Hipoxia , Metilación , Polietilenglicoles
9.
Angew Chem Int Ed Engl ; 60(8): 4150-4157, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33174359

RESUMEN

The organoplatinum(II) complex [Pt(C^N^N)(Cl)] (C^N^N=5,6-diphenyl-2,2'-bipyridine, Pt1) can assemble into nanoaggregates via π-π stacking and complementary hydrogen bonds, rather than Pt-Pt interactions. Pt1 exhibits ratiometric dual emission, including rare blue emission (λem =445 nm) and assembly-induced yellow emission (λem =573 nm), under one- and two-photon excitation. Pt1 displays blue emission in cells with an intact membrane due to its low cellular uptake. In cells where the membrane is disrupted, uptake of the complex is increased and at higher concentrations yellow emission is observed. The ratio of yellow to blue emission shows a linear relationship to the loss of cell membrane integrity. Pt1 is, to our knowledge, the first example of an assembly-induced two-photon ratiometric dual emission organoplatinum complex. The excellent and unique characteristics of the complex enabled its use for the tracking of cell apoptosis, necrosis, and the inflammation process in zebrafish.


Asunto(s)
Complejos de Coordinación/química , Microscopía de Fluorescencia por Excitación Multifotónica , Platino (Metal)/química , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/metabolismo , Complejos de Coordinación/farmacología , Humanos , Inflamación/inducido químicamente , Inflamación/diagnóstico por imagen , Larva/química , Larva/metabolismo , Piridinas/química , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
10.
Angew Chem Int Ed Engl ; 60(9): 4657-4665, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33217194

RESUMEN

Immunogenic cell death (ICD) is a vital component of therapeutically induced anti-tumor immunity. An iridium(III) complex (Ir1), containing an N,N-bis(2-chloroethyl)-azane derivate, as an endoplasmic reticulum-localized ICD inducer for non-small cell lung cancer (NSCLC) is reported. The characteristic discharge of damage-associated molecular patterns (DAMPs), that is, cell surface exposure of calreticulin (CRT), extracellular exclusion of high mobility group box 1 (HMGB1), and ATP, were generated by Ir1 in A549 lung cancer cells, accompanied by an increase in endoplasmic reticulum stress and reactive oxygen species (ROS). The vaccination of immunocompetent mice with Ir1-treated dying cells elicited an antitumor CD8+ T cell response and Foxp3+ T cell depletion, which eventually resulted in long-acting anti-tumor immunity by the activation of ICD in lung cancer cells. Ir1 is the first Ir-based complex that is capable of developing an immunomodulatory response by immunogenic cell death.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/química , Estrés del Retículo Endoplásmico/efectos de los fármacos , Muerte Celular Inmunogénica/efectos de los fármacos , Iridio/química , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Calreticulina/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , Retículo Endoplásmico/metabolismo , Femenino , Proteína HMGB1/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Tasa de Supervivencia
11.
Angew Chem Int Ed Engl ; 60(28): 15340-15343, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33899272

RESUMEN

G-quadruplexes (G4s) are prevalent in oncogenes and are potential antitumor drug targets. However, binding selectivity of compounds to G4s still faces challenges. Herein, we report a platinum(II) complex (Pt1), whose affinity to G4-DNA is activated by adaptive binding and selectivity controlled by binding kinetics. The resolved structure of Pt1/VEGF-G4 (a promoter G4) shows that Pt1 matches 3'-G-tetrad of VEGF-G4 through Cl- -dissociation and loop rearrangement of VEGF-G4. Binding rate constants are determined by coordination bond breakage/formation, correlating fully with affinities. The selective rate-determining binding step, Cl- -dissociation upon G4-binding, is 2-3 orders of magnitude higher than dsDNA. Pt1 potently targets G4 in living cells, effectively represses VEGF expression, and inhibits vascular growth in zebrafish. We show adaptive G4-binding activation and controlled by kinetics, providing a complementary design principle for compounds targeting G4 or similar biomolecules.


Asunto(s)
Antineoplásicos/farmacología , G-Cuádruplex/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Antineoplásicos/química , Sitios de Unión/efectos de los fármacos , Células HeLa , Humanos , Cinética , Estructura Molecular , Compuestos Organoplatinos/química
12.
Anal Chem ; 92(8): 6003-6009, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32212607

RESUMEN

Nitric oxide (NO) and superoxide anions (O2•-) are two noteworthy reactive species implicated in various physiological and pathological processes, such as ROS-induced lysosomal cell death. The interaction ("crosstalk") between them may form a new mediator peroxynitrite (ONOO-) which has implications for cancer, diabetes, Alzheimer's disease, and liver-damage. It is therefore essential to investigate lysosomal NO/O2•- crosstalk in vivo through ONOO--responsive molecular tools in order to fully comprehend the physiological and pathological mechanisms involved. In this study, a lysosome-targeting iridium(III) complex, Ir-NIR, has been investigated as a near-infrared (NIR) phosphorescent probe for visualizing NO/O2•- crosstalk by the phosphorescent detection of endogenous ONOO- levels in vivo. Ir-NIR exhibits a rapid (within 200 s), highly sensitive, and approximately 100-fold enhanced response to ONOO- in phosphorescence intensity. Thus, these characteristics, coupled with good cell permeability and low cytotoxicity, enable the probe to be used to detect intracellular ONOO- living organisms both in vitro and in vivo.


Asunto(s)
Complejos de Coordinación/química , Colorantes Fluorescentes/química , Iridio/química , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Superóxidos/metabolismo , Animales , Células Cultivadas , Complejos de Coordinación/síntesis química , Femenino , Colorantes Fluorescentes/síntesis química , Humanos , Rayos Infrarrojos , Mediciones Luminiscentes , Lisosomas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Confocal , Estructura Molecular , Óxido Nítrico/química , Ácido Peroxinitroso/análisis , Superóxidos/química
13.
J Biol Inorg Chem ; 25(4): 597-607, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32232583

RESUMEN

Cyclometalated iridium(III) complexes represent a promising approach to developing new anticancer metallodrugs. In this work, three phosphorescent cyclometalated iridium(III) complexes Ir1-Ir3 have been explored as mitochondria-targeted anticancer agents. All three complexes display higher antiproliferative activity than cisplatin against the cancer cells screened, and with the IC50 values ranging from 0.23 to 5.6 µM. Colocalization studies showed that these complexes are mainly localized in the mitochondria. Mechanism studies show that these complexes exert their anticancer efficacy through initiating a series of events related to mitochondrial dysfunction, including depolarization of mitochondrial membrane potential (MMP), elevation of intracellular reactive oxygen species (ROS) levels, and induction of apoptosis. Mitochondria-targted cyclometalated iridium complexes induce apoptosis through depolarized mitochondria, elevation of intracellular ROS and activated caspase.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Iridio/farmacología , Mitocondrias/efectos de los fármacos , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Iridio/química , Mitocondrias/metabolismo , Estructura Molecular , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad
14.
Inorg Chem ; 59(17): 12632-12642, 2020 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-32838518

RESUMEN

Multifunctional platinumIV anticancer prodrugs have the potential to enrich the anticancer properties and overcome the clinical problems of drug resistance and side effects of platinumII anticancer agents. Herein, we develop dual and triple action platinumIV complexes with targeted and biological active functionalities. One complex (PFL) that consists of cisplatin, tegafur, and lonidamine exhibits strong cytotoxicity against triple negative breast cancer (TNBC) cells. Cellular uptake and distribution studies reveal that PFL mainly accumulates in mitochondria. As a result, PFL disrupts the mitochondrial ultrastructure and induces significant alterations in the mitochondrial membrane potential, which further leads to an increase in production of reactive oxygen species (ROS) and a decrease in ATP synthesis in MDA-MB-231 TNBCs. Western blot analysis reveals the formation of ternary complex of thymidylate synthase, which shows the intracellular conversion of tegafur into 5-FU after its release from PFL. Furthermore, treatment with PFL impairs the mitochondrial function, leading to the inhibition of glycolysis and mitochondrial respiration and induction of apoptosis through the mitochondrial pathway. The RNA-sequencing experiment shows that PFL can perturb the pathways involved in DNA synthesis, DNA damage, metabolism, and transcriptional activity. These findings demonstrate that PFL intervenes in several cellular processes including DNA damage, thymidylate synthase inhibition, and perturbation of the mitochondrial bioenergetics to kill the cancer cells. The results highlight the significance of a triple-action prodrug for efficient anticancer therapy for TNBCs.


Asunto(s)
Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Inhibidores Enzimáticos/química , Platino (Metal)/química , Profármacos/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/patología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Liberación de Fármacos , Fluorouracilo/metabolismo , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Profármacos/química , Especies Reactivas de Oxígeno/metabolismo , Transcripción Genética/efectos de los fármacos
15.
Angew Chem Int Ed Engl ; 59(24): 9719-9726, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32173994

RESUMEN

G-quadruplex DNA show structural polymorphism, leading to challenges in the use of selective recognition probes for the accurate detection of G-quadruplexes in vivo. Herein, we present a tripodal cationic fluorescent probe, NBTE, which showed distinguishable fluorescence lifetime responses between G-quadruplexes and other DNA topologies, and fluorescence quantum yield (Φf ) enhancement upon G-quadruplex binding. We determined two NBTE-G-quadruplex complex structures with high Φf values by NMR spectroscopy. The structures indicated NBTE interacted with G-quadruplexes using three arms through π-π stacking, differing from that with duplex DNA using two arms, which rationalized the higher Φf values and lifetime response of NBTE upon G-quadruplex binding. Based on photon counts of FLIM, we detected the percentage of G-quadruplex DNA in live cells with NBTE and found G-quadruplex DNA content in cancer cells is 4-fold that in normal cells, suggesting the potential applications of this probe in cancer cell detection.


Asunto(s)
ADN/química , G-Cuádruplex , Línea Celular Tumoral , ADN/análisis , Humanos , Fotones
16.
Angew Chem Int Ed Engl ; 59(8): 3315-3321, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31828932

RESUMEN

Reported is the FeIII -activated lysosome-targeting prodrug FerriIridium for gastric cancer theranostics. It contains a meta-imino catechol group that can selectively bond to, and be oxidized by, free FeIII inside the cell. Subsequent oxidative rearrangement releases FeII and hydrolyses the amine bond under acidic conditions, forming an aminobipyridyl Ir complex and 2-hydroxybenzoquinone. Thus, FeII catalyzes the Fenton reaction, transforming hydrogen peroxide into hydroxyl radicals, the benzoquinone compounds interfere with the respiratory chain, and conversion of the prodrug into the Ir complex leads to an increase in phosphorescence and toxicity. These properties, combined with the high FeIII content and acidity of cancer cells, make FerriIridium a selective and efficient theranostic agent (IC50 =9.22 µm for AGS cells vs. >200 µm for LO2 cells). FerriIridium is the first metal-based compound that has been developed for chemotherapy using FeIII to enhance both selectivity and potency.


Asunto(s)
Iridio/química , Hierro/química , Profármacos/química , Neoplasias Gástricas/tratamiento farmacológico , Animales , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares
17.
Angew Chem Int Ed Engl ; 59(42): 18556-18562, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-32557982

RESUMEN

Hypoxia and the acidic microenvironment play a vital role in tumor metastasis and angiogenesis, generally compromising the chemotherapeutic efficacy. This provides a tantalizing angle for the design of platinum(IV) prodrugs for the effective and selective killing of solid tumors. Herein, two carbonic anhydrase IX (CAIX)-targeting platinum(IV) prodrugs have been developed, named as CAIXplatins. Based on their strong affinity for and inhibition of CAIX, CAIXplatins can not only overcome hypoxia and the acidic microenvironment, but also inhibit metabolic pathways of hypoxic cancer cells, resulting in a significantly enhanced therapeutic effect on hypoxic MDA-MB-231 tumors both in vitro and in vivo compared with cisplatin/oxaliplatin, accompanied with excellent anti-metastasis and anti-angiogenesis activities. Furthermore, the cancer selectivity indexes of CAIXplatins are 70-90 times higher than those of cisplatin/oxaliplatin with effectively alleviated side-effects.


Asunto(s)
Anhidrasa Carbónica IX/antagonistas & inhibidores , Hipoxia de la Célula , Complejos de Coordinación/química , Platino (Metal)/química , Profármacos/química , Animales , Anhidrasa Carbónica IX/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Análisis por Conglomerados , Complejos de Coordinación/metabolismo , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Larva/efectos de los fármacos , Larva/metabolismo , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Profármacos/metabolismo , Profármacos/farmacología , Profármacos/uso terapéutico , Proteoma/análisis , Proteoma/efectos de los fármacos , Proteómica , Pez Cebra/crecimiento & desarrollo
18.
Angew Chem Int Ed Engl ; 59(42): 18755-18762, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-32634290

RESUMEN

The development and malignancy of cancer cells are closely related to the changes of the epigenome. In this work, a mitochondria-targeted rhenium(I) complex (DFX-Re3), integrating the clinical iron chelating agent deferasirox (DFX), has been designed. By relocating iron to the mitochondria and changing the key metabolic species related to epigenetic modifications, DFX-Re3 can elevate the methylation levels of histone, DNA, and RNA. As a consequence, DFX-Re3 affects the events related to apoptosis, RNA polymerases, and T-cell receptor signaling pathways. Finally, it is shown that DFX-Re3 induces immunogenic apoptotic cell death and exhibits potent antitumor activity in vivo. This study provides a new approach for the design of novel epigenetic drugs that can recode the cancer epigenome by intervening in mitochondrial metabolism and iron homeostasis.


Asunto(s)
Complejos de Coordinación/química , Hierro/metabolismo , Mitocondrias/metabolismo , Renio/química , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , Deferasirox/química , Evaluación Preclínica de Medicamentos , Epigenómica , Histonas/metabolismo , Humanos , Quelantes del Hierro/química , Metilación/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , ARN Polimerasa II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Angew Chem Int Ed Engl ; 59(46): 20697-20703, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32735748

RESUMEN

The efficacy of photodynamic therapy is typically reliant on the local concentration and diffusion of oxygen. Due to the hypoxic microenvironment found in solid tumors, oxygen-independent photosensitizers are in great demand for cancer therapy. We herein report an iridium(III) anthraquinone complex as a mitochondrion-localized carbon-radical initiator. Its emission is turned on under hypoxic conditions after reduction by reductase. Furthermore, its two-photon excitation properties (λex =730 nm) are highly desirable for imaging. Upon irradiation, the reduced form of the complex generates carbon radicals, leading to a loss of mitochondrial membrane potential and cell death (IC50light =2.1 µm, IC50dark =58.2 µm, PI=27.7). The efficacy of the complex as a PDT agent was also demonstrated under hypoxic conditions in vivo. To the best of our knowledge, it is the first metal-complex-based theranostic agent which can generate carbon radicals for oxygen-independent two-photon photodynamic therapy.


Asunto(s)
Carbono/química , Hipoxia de la Célula , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/farmacología , Humanos , Mitocondrias/metabolismo , NADP/metabolismo , Neoplasias/patología , Fotoquimioterapia/métodos , Fotones , Análisis Espectral/métodos , Microambiente Tumoral
20.
Angew Chem Int Ed Engl ; 59(38): 16631-16637, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32533618

RESUMEN

Inducing necroptosis in cancer cells is an effective approach to circumvent drug-resistance. Metal-based triggers have, however, rarely been reported. Ruthenium(II) complexes containing 1,1-(pyrazin-2-yl)pyreno[4,5-e][1,2,4]triazine were developed with a series of different ancillary ligands (Ru1-7). The combination of the main ligand with bipyridyl and phenylpyridyl ligands endows Ru7 with superior nucleus-targeting properties. As a rare dual catalytic inhibitor, Ru7 effectively inhibits the endogenous activities of topoisomerase (topo) I and II and kills cancer cells by necroptosis. The cell signaling pathway from topo inhibition to necroptosis was elucidated. Furthermore, Ru7 displays significant antitumor activity against drug-resistant cancer cells in vivo. To the best of our knowledge, Ru7 is the first Ru-based necroptosis-inducing chemotherapeutic agent.


Asunto(s)
Complejos de Coordinación/farmacología , ADN-Topoisomerasas de Tipo I/metabolismo , Necroptosis/efectos de los fármacos , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Rutenio/farmacología , Inhibidores de Topoisomerasa/farmacología , Animales , Biocatálisis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , ADN-Topoisomerasas de Tipo II/metabolismo , Humanos , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Rutenio/química , Inhibidores de Topoisomerasa/síntesis química , Inhibidores de Topoisomerasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA