Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Colloids Surf B Biointerfaces ; 234: 113738, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199189

RESUMEN

Tumor recurrence and wound healing represent significant burdens for tumor patients after the surgical removal of melanomas. Wound dressings with wound healing and anticancer therapeutic abilities could help to solve these issues. Thus, a hybrid hydrogel made of polyvinyl alcohol (PVA) and polyethylene imine (PEI) was prepared by cross-linking imine bond and boronic acid bond. This hydrogel was loaded with ruthenium nanorods (Ru NRs) and glucose oxidase (GOx) and named as nanocomposite hydrogel (Ru/GOx@Hydrogel), exhibiting remarkable photothermal/photodynamic/starvation antitumor therapy and wound repair abilities. Ru NRs are bifunctional phototherapeutic agents that simultaneously exhibit intrinsic photothermal and photodynamic functions. Three-dimensional composite hydrogel loaded with GOx can also consume glucose in the presence of O2 during tumor starvation therapy. Near-infrared (NIR) light-triggered hyperthermia can not only promote the consumption of glucose, but also facilitate the ablation of residual cancer cells. The antitumor effect of the Ru/GOx@Hydrogel resulted in significant improvements, compared to those observed with either phototherapy or starvation therapy alone. Additionally, the postoperative wound was substantially healed after treatment with Ru/GOx@Hydrogel and NIR irradiation. Therefore, the Ru/GOx@Hydrogel can be used as a multi-stimulus-responsive nanoplatform that could facilitate on-demand controlled drug release, and be used as a promising postoperative adjuvant in combination therapy.


Asunto(s)
Hipertermia Inducida , Nanotubos , Neoplasias , Rutenio , Humanos , Glucosa Oxidasa , Rutenio/farmacología , Polietileneimina , Alcohol Polivinílico , Hidrogeles/química , Neoplasias/terapia , Glucosa
2.
Inorg Chem ; 62(51): 21181-21200, 2023 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-38079387

RESUMEN

Ru(II) polypyridyl complexes have gained widespread attention as photosensitizers for photodynamic therapy (PDT). Herein, we systematically investigate a series of the type [Ru(phen)2(IP-nT)]2+, featuring 1,10-phenanthroline (phen) coligands and imidazo[4,5-f][1,10]phenanthroline ligands tethered to n = 0-4 thiophene rings (IP-nT). The complexes were characterized and investigated for their electrochemical, spectroscopic, and (photo)biological properties. The electrochemical oxidation of the nT unit shifted by -350 mV as n = 1 → 4 (+920 mV for Ru-1T, +570 mV for Ru-4T); nT reductions were observed in complexes Ru-3T (-2530 mV) and Ru-4T (-2300 mV). Singlet oxygen quantum yields ranged from 0.53 to 0.88, with Ru-3T and Ru-4T being equally efficient (∼0.88). Time-resolved absorption spectra of Ru-0T-1T were dominated by metal-to-ligand charge-transfer (3MLCT) states (τTA = 0.40-0.85 µs), but long-lived intraligand charge-transfer (3ILCT) states were observed in Ru-2T-4T (τTA = 25-148 µs). The 3ILCT energies of Ru-3T and Ru-4T were computed to be 1.6 and 1.4 eV, respectively. The phototherapeutic efficacy against melanoma cells (SK-MEL-28) under broad-band visible light (400-700 nm) increases as n = 0 → 4: Ru-0T was inactive up to 300 µM, Ru-1T-2T were moderately active (EC50 ∼ 600 nM, PI = 200), and Ru-3T (EC50 = 57 nM, PI > 1100) and Ru-4T (EC50 = 740 pM, PI = 114,000) were the most phototoxic. The activity diminishes with longer wavelengths of light and is completely suppressed for all complexes except Ru-3T and Ru-4T in hypoxia. Ru-4T is the more potent and robust PS in 1% O2 over seven biological replicates (avg EC50 = 1.3 µM, avg PI = 985). Ru-3T exhibited hypoxic activity in five of seven replicates, underscoring the need for biological replicates in compound evaluation. Singlet oxygen sensitization is likely responsible for phototoxic effects of the compounds in normoxia, but the presence of redox-active excited states may facilitate additional photoactive pathways for complexes with three or more thienyl groups. The 3ILCT state with its extended lifetime (30-40× longer than the 3MLCT state for Ru-3T and Ru-4T) implicates its predominant role in photocytotoxicity.


Asunto(s)
Fotoquimioterapia , Rutenio , Fenantrolinas/farmacología , Fenantrolinas/química , Oxígeno Singlete/química , Rutenio/farmacología , Rutenio/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Ligandos
3.
Biomacromolecules ; 24(12): 5940-5950, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-38033171

RESUMEN

Polymer micelles/vesicles made of a red-light-responsive Ru(II)-containing block copolymer (PolyRu) are elaborated as a model system for anticancer phototherapy. PolyRu is composed of PEG and a hydrophobic polypeptoid bearing thioether side chains, 40% of which are coordinated with [Ru(2,2':6',2″-terpyridine)(2,2'-biquinoline)](PF6)2 via the Ru-S bond, resulting in a 67 wt % Ru complex loading capacity. Red-light illumination induces the photocleavage of the Ru-S bond and produces [Ru(2,2':6',2″-terpyridine)(2,2'-biquinoline)(H2O)](PF6)2. Meanwhile, ROS are generated under the photosensitization of the Ru complex and oxidize hydrophobic thioether to hydrophilic sulfoxide, causing the disruption of micelles/vesicles. During the disruption, ROS generation and Ru complex release are synergistically enhanced. PolyRu micelles/vesicles are taken up by cancer cells while they exhibit very low cytotoxicity in the dark. In contrast, they show much higher cytotoxicity under red-light irradiation. PolyRu micelles/vesicles are promising nanoassembly prototypes that protect metallodrugs in the dark but exhibit light-activated anticancer effects with spatiotemporal control for photoactivated chemotherapy and photodynamic therapy.


Asunto(s)
Complejos de Coordinación , Rutenio , Especies Reactivas de Oxígeno , Rutenio/farmacología , Rutenio/química , Liberación de Fármacos , Micelas , Fototerapia/métodos , Polímeros/química , Sulfuros , Complejos de Coordinación/farmacología , Complejos de Coordinación/química
4.
Molecules ; 28(22)2023 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-38005258

RESUMEN

Photodynamic therapy (PDT) is an anticancer/antibacterial strategy in which photosensitizers (PSs), light, and molecular oxygen generate reactive oxygen species and induce cell death. PDT presents greater selectivity towards tumor cells than conventional chemotherapy; however, PSs have limitations that have prompted the search for new molecules featuring more favorable chemical-physical characteristics. Curcumin and its derivatives have been used in PDT. However, low water solubility, rapid metabolism, interference with other drugs, and low stability limit curcumin use. Chemical modifications have been proposed to improve curcumin activity, and metal-based PSs, especially ruthenium(II) complexes, have attracted considerable attention. This study aimed to characterize six Ru(II)-arene curcuminoids for anticancer and/or antibacterial PDT. The hydrophilicity, photodegradation rates, and singlet oxygen generation of the compounds were evaluated. The photodynamic effects on human colorectal cancer cell lines were also assessed, along with the ability of the compounds to induce ROS production, apoptotic, necrotic, and/or autophagic cell death. Overall, our encouraging results indicate that the Ru(II)-arene curcuminoid derivatives are worthy of further investigation and could represent an interesting option for cancer PDT. Additionally, the lack of significant in vivo toxicity on the larvae of Galleria mellonella is an important finding. Finally, the photoantimicrobial activity of HCurc I against Gram-positive bacteria is indeed promising.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Curcumina , Fotoquimioterapia , Rutenio , Humanos , Fármacos Fotosensibilizantes/química , Rutenio/farmacología , Rutenio/química , Curcumina/farmacología , Diarilheptanoides , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Antineoplásicos/química , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
5.
mSphere ; 8(5): e0019023, 2023 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-37646510

RESUMEN

The rapid dissemination of antibiotic resistance combined with the decline in the discovery of novel antibiotics represents a major challenge for infectious disease control that can only be mitigated by investments in novel treatment strategies. Alternative antimicrobials, including silver, have regained interest due to their diverse mechanisms of inhibiting microbial growth. One such example is AGXX, a broad-spectrum antimicrobial that produces highly cytotoxic reactive oxygen species (ROS) to inflict extensive macromolecular damage. Due to the connections identified between ROS production and antibiotic lethality, we hypothesized that AGXX could potentially increase the activity of conventional antibiotics. Using the gram-negative pathogen Pseudomonas aeruginosa, we screened possible synergistic effects of AGXX on several antibiotic classes. We found that the combination of AGXX and aminoglycosides tested at sublethal concentrations led to a rapid exponential decrease in bacterial survival and restored the sensitivity of a kanamycin-resistant strain. ROS production contributes significantly to the bactericidal effects of AGXX/aminoglycoside treatments, which is dependent on oxygen availability and can be reduced by the addition of ROS scavengers. Additionally, P. aeruginosa strains deficient in ROS detoxifying/repair genes were more susceptible to AGXX/aminoglycoside treatment. We further demonstrate that this synergistic interaction was associated with a significant increase in outer and inner membrane permeability, resulting in increased antibiotic influx. Our study also revealed that AGXX/aminoglycoside-mediated killing requires an active proton motive force across the bacterial membrane. Overall, our findings provide an understanding of cellular targets that could be inhibited to increase the activity of conventional antimicrobials. IMPORTANCE The emergence of drug-resistant bacteria coupled with the decline in antibiotic development highlights the need for novel alternatives. Thus, new strategies aimed at repurposing conventional antibiotics have gained significant interest. The necessity of these interventions is evident especially in gram-negative pathogens as they are particularly difficult to treat due to their outer membrane. This study highlights the effectiveness of the antimicrobial AGXX in potentiating aminoglycoside activities against P. aeruginosa. The combination of AGXX and aminoglycosides not only reduces bacterial survival rapidly but also significantly re-sensitizes aminoglycoside-resistant P. aeruginosa strains. In combination with gentamicin, AGXX induces increased endogenous oxidative stress, membrane damage, and iron-sulfur cluster disruption. These findings emphasize AGXX's potential as a route of antibiotic adjuvant development and shed light on potential targets to enhance aminoglycoside activity.


Asunto(s)
Antiinfecciosos , Rutenio , Aminoglicósidos/farmacología , Pseudomonas aeruginosa , Rutenio/farmacología , Plata/farmacología , Especies Reactivas de Oxígeno , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , Bacterias
6.
J Mater Chem B ; 11(24): 5607-5618, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37170629

RESUMEN

Radiotherapy has been extensively applied to cancer therapy in clinical trials. However, radiation resistance and dose limitation generally hamper the efficacy of radiotherapy. There is an urgent need for radiosensitizers with high efficiency and safety to enhance the anti-tumor effect of radiotherapy. In this paper, a selenium-containing (Se) ruthenium (Ru) complex (RuSe) was designed as a radiosensitizer to synergistically augment the killing effect of radiotherapy on nasopharyngeal carcinoma cells. In this system, the heavy atomic effect of Ru enhances the photoelectron production triggered by X-rays, thus inducing a burst of reactive oxygen species (ROS). In addition, Se atoms with a strong polarization property were introduced into the ligand of the metal complex to enhance the tumor chemo/radiotherapy effect. Consequently, RuC with a weak atomic polarization effect, as a comparison for RuSe, was also rationally explored to elucidate the role of Se atoms on chemo/radiotherapy sensitization. Indeed, compared with RuC, RuSe at a sub-toxic dose was able to potentiate the lethality of radiotherapy after preconditioning with cancer cells, by inducing ROS over-production, decreasing the mitochondrial membrane potential, and arresting the cell cycle at the sub-G1 phase. Furthermore, upon radiation, RuSe was superior to RuC, by inducing apoptotic cell death by activating caspase-3, -8, and -9. In summary, this study not only demonstrates an effective and safe strategy for the application of RuSe complexes to the cancer-targeted chemo/radiotherapy of human cancers, but also sheds light on the potential mechanisms of such Se-containing drugs as efficient radiotherapy sensitizers.


Asunto(s)
Neoplasias Nasofaríngeas , Fármacos Sensibilizantes a Radiaciones , Rutenio , Selenio , Humanos , Selenio/farmacología , Rayos X , Rutenio/farmacología , Especies Reactivas de Oxígeno/metabolismo , Carcinoma Nasofaríngeo/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias Nasofaríngeas/tratamiento farmacológico
7.
J Med Chem ; 66(10): 6922-6937, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37185020

RESUMEN

Synergistic drug combinations can extend the use of poly(ADP-ribose) polymerase inhibitors (PARPi) such as Olaparib to BRCA-proficient tumors and overcome acquired or de novo drug resistance. To identify new synergistic combinations for PARPi, we screened a "micro-library" comprising a mix of commercially available drugs and DNA-binding ruthenium(II) polypyridyl complexes (RPCs) for Olaparib synergy in BRCA-proficient triple-negative breast cancer cells. This identified three hits: the natural product Curcumin and two ruthenium(II)-rhenium(I) polypyridyl metallomacrocycles. All combinations identified were effective in BRCA-proficient breast cancer cells, including an Olaparib-resistant cell line, and spheroid models. Mechanistic studies indicated that synergy was achieved via DNA-damage enhancement and resultant apoptosis. Combinations showed low cytotoxicity toward non-malignant breast epithelial cells and low acute and developmental toxicity in zebrafish embryos. This work identifies RPC metallomacrocycles as a novel class of agents for cancer combination therapy and provides a proof of concept for the inclusion of metallocompounds within drug synergy screens.


Asunto(s)
Neoplasias Ováricas , Rutenio , Humanos , Animales , Femenino , Rutenio/farmacología , Rutenio/uso terapéutico , Pez Cebra , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , ADN , Línea Celular Tumoral
8.
J Inorg Biochem ; 242: 112175, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36898296

RESUMEN

Compounds modified with selenium atom as potential antibacterial agents have been exploited to combat the nondrug-resistant bacterial infection. In this study, we designed and synthesized four ruthenium complexes retouching of selenium-ether. Fortunately, those four ruthenium complexes shown excellent antibacterial bioactive (MIC: 1.56-6.25 µg/mL) against Staphylococcus aureus (S. aureus), and the most active complex Ru(II)-4 could kill S. aureus by targeting the membrane integrity and avoid the bacteria to evolve drug resistance. Moreover, Ru(II)-4 was found to significantly inhibit the formation of biofilms and biofilm eradicate capacity. In toxicity experiments, Ru(II)-4 exhibited poor hemolysis and low mammalian toxicity. To illustrate the antibacterial mechanism: we conducted scanning electron microscope (SEM), fluorescent staining, membrane rupture and DNA leakage assays. Those results demonstrated that Ru(II)-4 could destroy the integrity of bacterial cell membrane. Furthermore, both G. mellonella wax worms infection model and mouse skin infection model were established to evaluate the antibacterial activity of Ru(II)-4 in vivo, the results indicated that Ru(II)-4 was a potential candidate for combating S. aureus infections, and almost non-toxic to mouse tissue. Thus, all the results indicated that introducing selenium-atom into ruthenium compounds were a promising strategy for developing interesting antibacterial agents.


Asunto(s)
Complejos de Coordinación , Infecciones por Bacterias Grampositivas , Rutenio , Selenio , Animales , Ratones , Staphylococcus aureus , Rutenio/farmacología , Complejos de Coordinación/farmacología , Selenio/farmacología , Antibacterianos/farmacología , Bacterias , Resistencia a Medicamentos , Pruebas de Sensibilidad Microbiana , Mamíferos
9.
J Biomol Struct Dyn ; 41(9): 4143-4153, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35514135

RESUMEN

The interactions of ruthenium(II) complex with Glucose inhibited division protein A (GidA protein) was studied through various spectroscopic techniques with the ultimate goal of preparing adducts with good selectivity for cancer cells. In all the cases, formation of a tight metal-protein conjugate was observed. The influence of pH, reducing agents and chelators on the formation of adduct was analysed by UV- visible spectroscopy. While there was no effect on the addition of sodium ascorbate, some alterations on some selected bands were seen on the UV-visible spectra on the addition of EDTA. The adduct was stable in the pH range of 5-8. Addition of ruthenium(II) complex effectively quenched the intrinsic fluorescence of GidA and it occurred through static quenching. The effect of ruthenium(II) complex on the conformation of GidA has been examined by analyzing CD spectrum. Though, there was some conformational changes observed in the presence of ruthenium(II) complex, α- helix in the secondary structure of GidA retained its identity. Molecular docking of ruthenium(II) complex with GidA also indicated that GidA docks through hydrophobic interaction. The stable semisynthetic complex (ruthenium(II) complex with GidA) was checked for topoisomerase II inhibition. Relaxation and decatenation assay proved topoisomerase II inhibition of semisynthetic complex.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Neoplasias , Rutenio , Humanos , Inhibidores de Topoisomerasa II/farmacología , Simulación del Acoplamiento Molecular , Proteína Estafilocócica A , Rutenio/farmacología , Rutenio/química , Neoplasias/tratamiento farmacológico , ADN-Topoisomerasas de Tipo II/metabolismo
10.
Eur J Med Chem ; 243: 114772, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36191406

RESUMEN

In this work we present the synthesis and characterization of six new ruthenium compounds with general formulae [Ru(L)(dppb)(bipy)]PF6 and [Ru(L)(dppe)2]PF6 where L = salicylic acid (Sal), 4-aminosalicylic acid (AmSal) or 2,4-dihydroxybenzoic acid (DiSal), dppb = 1,4-bis(diphenylphosphino)butane, dppe = 1,2-bis(diphenylphosphino)ethane and bipy = 2,2'-bipyridine. The complexes were characterized by elemental analysis, molar conductivity, cyclic voltammetry, NMR, UV-vis and IR spectroscopies, and two by X-ray crystallography. The 31P{1H} NMR spectra of the complexes with the general formula [Ru(L)(dppe)2]PF6 showed that the phosphorus signals are solvent-dependent. Aprotic solvents, which form strong hydrogen bonds with the complexes, inhibit the free rotation of the salicylic acid-based, modifying the diphosphine cone angles, leading to distortion of the phosphorus signals in the NMR spectra. The cytotoxicity of the complexes was evaluated in MCF-7, MDA-MB-231, SKBR3 human breast tumor cells, and MCF-10 non-tumor cell lines. The complexes with the structural formula [Ru(L)(dppe)2]PF6 were the most cytotoxic, and the complex [Ru(AmSal)(dppe)2]PF6 with L = 4-aminosalicylic acid ligand was the most selective for the MDA-MB-231 cell line. This complex interacts with the transferrin and induces apoptosis through the intrinsic pathway, as demonstrated by increased levels of proteins involved in apoptotic cell death.


Asunto(s)
Ácido Aminosalicílico , Antineoplásicos , Complejos de Coordinación , Neoplasias , Rutenio , Humanos , Rutenio/farmacología , Rutenio/química , Complejos de Coordinación/química , Ácido Salicílico/farmacología , Ácido Aminosalicílico/farmacología , Amobarbital/farmacología , Apoptosis , Antineoplásicos/química , Fósforo/farmacología , Línea Celular Tumoral
11.
J Med Chem ; 65(19): 13041-13051, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36134739

RESUMEN

The curative effect of sorafenib in hepatocellular carcinoma (HCC) is limited and sorafenib resistance remains a major obstacle for HCC. To overcome this obstacle, a new photoactive sorafenib-Ru(II) complex Ru-Sora has been designed. Upon irradiation (λ = 465 nm), Ru-Sora rapidly releases sorafenib and generates reactive oxygen species, which can oxidize intracellular substances such as GSH. Cellular experiments show that irradiated Ru-Sora is highly cytotoxic toward Hep-G2 cells, including sorafenib-resistant Hep-G2-SR cells. Compared to sorafenib, Ru-Sora has a significant photoactivated chemotherapeutic effect against Hep-G2-SR cancer cells and 3D Hep-G2 multicellular tumor spheroids. Furthermore, Ru-Sora inducing apoptosis and ferroptosis is proved by GSH depletion, GPX4 downregulation, and lipid peroxide accumulation. Metabolomics results suggest that Ru-Sora exerts photocytotoxicity by disrupting the purine metabolism, which is expected to inhibit tumor development. This study provides a promising strategy for enhancing chemotherapy and combating drug-resistant HCC disease.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Ferroptosis , Neoplasias Hepáticas , Profármacos , Rutenio , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Células Hep G2 , Humanos , Peróxidos Lipídicos/farmacología , Neoplasias Hepáticas/patología , Profármacos/farmacología , Profármacos/uso terapéutico , Purinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Rutenio/farmacología , Rutenio/uso terapéutico , Sorafenib/farmacología
12.
J Mater Chem B ; 10(38): 7760-7771, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36069420

RESUMEN

Phototherapy, which mainly includes photothermal therapy (PTT) and photodynamic therapy (PDT), is one of the most promising strategies for cancer therapeutics. Ruthenium as a metal nanomaterial shows great potential as a phototherapy agent. Herein, we developed flower-like ruthenium nanoparticles (FRuNPs) to enhance cancer phototherapy. Compared with spherical ruthenium nanoparticles (SRuNPs) of a similar size, FRuNPs exhibited more enhanced near-infrared (NIR) absorption. FRuNPs exhibited a superior photothermal effect, which significantly improved the efficiency of PTT. Intracellular reactive oxide species (ROS) generation was recognized as the primary mechanism of PDT treatment. FRuNPs mediated the generation of ROS when exposed to 808 nm laser irradiation. Moreover, with the synergistic effect of PTT and PDT in FRuNPs, the phototherapeutic effects were obviously enhanced. In vitro phototherapy of MCF-7 cells in the presence of FRuNPs led to nearly 100% cell death under irradiation with an 808 nm laser. And in vivo FRuNPs further showed the capacity of in completely clearing the tumor tissues and improving the hypoxia environment with 14 days for interval laser irradiation. These results indicate that FRuNPs have versatile potential for tumor phototherapy.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Rutenio , Humanos , Rayos Láser , Nanopartículas/uso terapéutico , Óxidos , Fototerapia/métodos , Especies Reactivas de Oxígeno , Rutenio/farmacología
13.
Chem Commun (Camb) ; 58(65): 9068-9071, 2022 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-35894452

RESUMEN

Herein, we construct a series of Ru(II) metallacycles with multimodal chemo-phototherapeutic properties, which exhibited much higher anticancer activity and better cancer-cell selectivity than cisplatin. The antitumor mechanism could be ascribed to the activation of caspase 3/7 and the resulting apoptosis. These results open new possibilities for Ru(II) metallacycles in biomedicine.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Neoplasias , Rutenio , Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Humanos , Imidazoles/farmacología , Ligandos , Neoplasias/tratamiento farmacológico , Fototerapia , Rutenio/farmacología
14.
Acta Biomater ; 147: 198-208, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35643198

RESUMEN

Corneal collagen crosslinking (CXL) is a commonly used minimally invasive surgical technique to prevent the progression of corneal ectasias, such as keratoconus. Unfortunately, riboflavin/UV-A light-based CXL procedures have not been successfully applied to all patients, and result in frequent complications, such as corneal haze and endothelial damage. We propose a new method for corneal crosslinking by using a Ruthenium (Ru) based water-soluble photoinitiator and visible light (430 nm). Tris(bipyridine)ruthenium(II) ([Ru(bpy)3]2+) and sodium persulfate (SPS) mixture covalently crosslinks free tyrosine, histidine, and lysine groups under visible light (400-450 nm), which prevents UV-A light-induced cytotoxicity in an efficient and time saving collagen crosslinking procedure. In this study, we investigated the effects of the Ru/visible blue light procedure on the viability and toxicity of human corneal epithelium, limbal, and stromal cells. Then bovine corneas crosslinked with ruthenium mixture and visible light were characterized, and their biomechanical properties were compared with the customized riboflavin/UV-A crosslinking approach in the clinics. Crosslinked corneas with a ruthenium-based CXL approach showed significantly higher young's modulus compared to riboflavin/UV-A light-based method applied to corneas. In addition, crosslinked corneas with both methods were characterized to evaluate the hydrodynamic behavior, optical transparency, and enzymatic resistance. In all biomechanical, biochemical, and optical tests used here, corneas that were crosslinked with ruthenium-based approach demonstrated better results than that of corneas crosslinked with riboflavin/ UV-A. This study is promising to be translated into a non-surgical therapy for all ectatic corneal pathologies as a result of mild conditions introduced here with visible light exposure and a nontoxic ruthenium-based photoinitiator to the cornea. STATEMENT OF SIGNIFICANCE: Keratoconus, one of the most frequent corneal diseases, could be treated with riboflavin and ultraviolet light-based photo-crosslinking application to the cornea of the patients. Unfortunately, this method has irreversible side effects and cannot be applied to all keratoconus patients. In this study, we exploited the photoactivation behavior of an organoruthenium compound to achieve corneal crosslinking. Ruthenium-based organic complex under visible light demonstrated significantly better biocompatibility and superior biomechanical results than riboflavin and ultraviolet light application. This study promises to translate into a new fast, efficient non-surgical therapy option for all ectatic corneal pathologies.


Asunto(s)
Queratocono , Fotoquimioterapia , Rutenio , Animales , Bovinos , Colágeno/farmacología , Córnea/patología , Reactivos de Enlaces Cruzados/farmacología , Humanos , Queratocono/tratamiento farmacológico , Queratocono/patología , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Riboflavina/farmacología , Rutenio/farmacología , Rayos Ultravioleta
15.
Stem Cell Rev Rep ; 18(7): 2234-2261, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35103937

RESUMEN

Mesenchymal stem cells (MSCs) exhibit regenerative and reparative properties. However, most MSC-related studies remain to be translated for regular clinical usage, partly due to challenges in pre-transplantation cell labelling and post-transplantation cell tracking. Amidst this, there are growing concerns over the toxicity of commonly used gadolinium-based contrast agents that mediate in-vivo cell detection via MRI. This urges to search for equally effective but less toxic alternatives that would facilitate and enhance MSC detection post-administration and provide therapeutic benefits in-vivo. MSCs labelled with iron oxide nanoparticles (IONPs) have shown promising results in-vitro and in-vivo. Thus, it would be useful to revisit these studies before inventing new labelling approaches. Aiming to inform regenerative medicine and augment clinical applications of IONP-labelled MSCs, this review collates and critically evaluates the utility of IONPs in enhancing MSC detection and therapeutics. It explains the rationale, principle, and advantages of labelling MSCs with IONPs, and describes IONP-induced intracellular alterations and consequent cellular manifestations. By exemplifying clinical pathologies, it examines contextual in-vitro, animal, and clinical studies that used IONP-labelled bone marrow-, umbilical cord-, adipose tissue- and dental pulp-derived MSCs. It compiles and discusses studies involving MSC-labelling of IONPs in combinations with carbohydrates (Venofer, ferumoxytol, dextran, glucosamine), non-carbohydrate polymers [poly(L-lysine), poly(lactide-co-glycolide), poly(L-lactide), polydopamine], elements (ruthenium, selenium, gold, zinc), compounds/stains (silica, polyethylene glycol, fluorophore, rhodamine B, DAPI, Prussian blue), DNA, Fibroblast growth Factor-2 and the drug doxorubicin. Furthermore, IONP-labelling of MSC exosomes is reviewed. Also, limitations of IONP-labelling are addressed and methods of tackling those challenges are suggested.


Asunto(s)
Células Madre Mesenquimatosas , Rutenio , Selenio , Animales , Medios de Contraste , Dextranos/farmacología , Doxorrubicina/farmacología , Compuestos Férricos , Sacarato de Óxido Férrico/farmacología , Óxido Ferrosoférrico , Factor 2 de Crecimiento de Fibroblastos/farmacología , Gadolinio/farmacología , Glucosamina/farmacología , Oro/farmacología , Nanopartículas Magnéticas de Óxido de Hierro , Polietilenglicoles/farmacología , Poliglactina 910/farmacología , Polilisina/farmacología , Rutenio/farmacología , Selenio/farmacología , Dióxido de Silicio/farmacología , Zinc/farmacología
16.
Chem Commun (Camb) ; 56(20): 3019-3022, 2020 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-32048647

RESUMEN

Nucleus-targeting NPs based on RuO2 (RuO2NPs) were developed by controlling the size and the surface charge of nanoparticles (NPs). This study not only demonstrates a facile approach for the fabrication of ultrasmall CS-RuO2NPs with good biocompatibility and excellent photothermal properties but also their unique potential for the nucleus-targeted low-temperature PTT.


Asunto(s)
Nanopartículas/química , Imagen Óptica , Óxidos/química , Técnicas Fotoacústicas , Rutenio/química , Temperatura , Núcleo Celular/efectos de los fármacos , Humanos , Rayos Infrarrojos , Células MCF-7 , Óxidos/farmacología , Tamaño de la Partícula , Fototerapia , Rutenio/farmacología , Propiedades de Superficie
17.
Nanoscale ; 11(39): 18209-18223, 2019 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-31560010

RESUMEN

Rheumatoid arthritis (RA) is a degenerative joint disease caused by autoimmunity; for the effective treatment of RA while avoiding the side effects of conventional drugs, we have proposed a new therapeutic strategy to eliminate the inflammatory response in RA by regulating the immune system that promotes the transformation of M1-type macrophages to M2-type macrophages. Herein, we designed and synthesized a core-shell nanocomposite (QRu-PLGA-RES-DS NPs), which showed an effective therapeutic effect on RA by accurately inducing the polarization of M2 macrophages. In this system, the quadrilateral ruthenium nanoparticles (QRuNPs) with a photothermal effect were utilized as a core and the thermosensitive molecular poly (lactic-co-glycolic acid) (PLGA) modified with the targeted molecule dextran sulfate (DS) was employed as a shell. Then, the nanocarrier QRu-PLGA-DS NPs effectively improved the water solubility and targeting of resveratrol (RES) through self-assembly. Therefore, the QRu-PLGA-RES-DS NPs significantly enhanced the ability of RES to reverse the M1 type macrophages to the M2 type macrophages through an accurate release. In vivo experiments further demonstrated that the QRu-PLGA-RES-DS NPs could effectively accumulate in the lesion area with an exogenous stimulus, and this significantly enhanced the transformation of the M2 type macrophages and decreased the recruitment of the M1 type macrophages. Furthermore, the QRu-PLGA-RES-DS NPs effectively treated RA by eliminating the inflammatory response; in addition, photoacoustic imaging (PA) of the QRu NPs provided image guidance for the distribution and analysis of nanomedicine in inflammatory tissues. Hence, this therapeutic strategy promotes the biological applications of Ru-based nanoparticles in disease treatment.


Asunto(s)
Hipertermia Inducida , Macrófagos/metabolismo , Nanocompuestos , Fototerapia , Resveratrol , Fiebre Reumática/terapia , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Macrófagos/patología , Ratones , Nanocompuestos/química , Nanocompuestos/uso terapéutico , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacocinética , Ácido Poliglicólico/farmacología , Células RAW 264.7 , Resveratrol/farmacocinética , Resveratrol/farmacología , Fiebre Reumática/metabolismo , Fiebre Reumática/patología , Rutenio/química , Rutenio/farmacocinética , Rutenio/farmacología
18.
Biomaterials ; 219: 119377, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31374478

RESUMEN

Natural killer (NK) cells-based therapy has been used widely for cancer treatment in clinic trails. However, the immunotherapeutic efficacy of this method has been greatly hindered by tumor evasion and diminished activities of NK cells. In the present study, a selenium (Se)-bearing ruthenium (Ru) complex (RuSe) was designed that could synergistically potentiate NK cell-mediated killing against prostate cancer cells. As expected, pretreatment of cancer cells with subtoxic doses of RuSe effectively augmented the lysis potency of NK cells, with up to 2.46-fold enhancement than NK cells alone, against PC3 cells. More importantly, low concentrations of RuSe could augment the tumor destroying potency of NK cells derived from 10 clinical patients, with the enhancement range from 0.78- to 11.9-fold against PC3 cells and 0.67- to 3.8-fold against LNCAP cells. Mechanistic studies revealed that the sensitizing effect of RuSe primarily depended on TRAIL/TRAIL-R and Fas/FasL-mediated signaling. Furthermore, the increased expression level of these ligands highly relied on ROS overproduction-triggered DNA damage and the downstream ATM and ATR pathways. Furthermore, RuSe potently activated and synergized with NK cells to restrain tumor growth in vivo without causing toxic side effects on major organs. Taken together, the current study not only provides a strategy for application of metal complexes in chemo-immunotherapy but also sheds light on the potential roles and mechanisms of action on such Se-containing drugs as efficient immune-sensitizing agents for NK cell-based immunotherapy.


Asunto(s)
Proteína Ligando Fas/metabolismo , Inmunoterapia , Células Asesinas Naturales/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Rutenio/farmacología , Selenio/farmacología , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Degranulación de la Célula/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Daño del ADN , Humanos , Células Asesinas Naturales/efectos de los fármacos , Masculino , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Muerte Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Receptor fas/metabolismo
19.
ACS Appl Mater Interfaces ; 10(37): 31106-31113, 2018 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-30178992

RESUMEN

To improve the efficiency of cancer therapy, we developed multifoliate PEGylated PtRu bimetallic nanocomplexes (PtRu-PEG BNCs) as multifunctional theranostic nanoagents for computed tomography (CT) imaging and synergistic thermoradiotherapy. The synthesized PtRu-PEG BNCs with uniform size and morphology exhibit excellent stability, notable photothermal effect, and good biocompatibility. As compared with other platinum nanomaterials, the PtRu-PEG BNCs are able to absorb near-infrared laser energy and present excellent photothermal conversion efficiency (44.5%). Multifoliate PtRu-PEG BNCs can be applied to CT imaging and radiotherapy (RT) because of the presence of platinum. Unlike a single therapy method, the integration of photothermal therapy with RT can effectively induce cell apoptosis and generate an obvious synergistic effect. Hence, the as-prepared nanocomplexes can be used as multifunctional theranostic nanoagents.


Asunto(s)
Nanoconjugados/química , Fototerapia/métodos , Platino (Metal)/química , Rutenio/química , Tomografía Computarizada por Rayos X/métodos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Calor , Ratones , Platino (Metal)/farmacología , Rutenio/farmacología , Nanomedicina Teranóstica
20.
Adv Healthc Mater ; 7(5)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29327505

RESUMEN

Utilizing the size-dependent adsorption properties of ruthenium carbonyl clusters (Ru-carbon monoxide (CO)) onto graphene oxide (GO), a facile CO-release platform for in situ vasodilation as a treatment for stroke-related vascular diseases is developed. The rate and amount of formation of the CO-release-active RuII (CO)2 species can be modulated by a simple mixing procedure at room temperature. The subsequent thermally induced oxidation of RuII (CO)2 to RuO2 on the GO surface results in the release of CO. Further modulation of thermal and CO-release properties can be achieved via a hybridization of medium- and high-nuclearity of Ru-CO clusters that produces a RuO2 /RuII (CO)2 /6 Ru-CO-GO composite, where 6 Ru-CO-GO provides a photothermally activated reservoir of RuII (CO)2 species and the combined infrared absorption properties of GO and RuO2 provides photothermal response for in situ CO-release. The RuO2 /RuII (CO)2 /6 Ru-CO-GO composite does not produce any cytotoxicity and the efficacy of the composite is further demonstrated in a cortical photothrombotic ischemia rat model.


Asunto(s)
Complejos de Coordinación , Grafito , Hipertermia Inducida , Fototerapia , Rutenio , Accidente Cerebrovascular/terapia , Animales , Línea Celular , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/farmacología , Grafito/química , Grafito/farmacocinética , Grafito/farmacología , Humanos , Ratas , Rutenio/química , Rutenio/farmacocinética , Rutenio/farmacología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA