Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cancer ; 23(1): 109, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769556

RESUMEN

Breast cancer (BC) is the most frequent malignant cancer diagnosis and is a primary factor for cancer deaths in women. The clinical subtypes of BC include estrogen receptor (ER) positive, progesterone receptor (PR) positive, human epidermal growth factor receptor 2 (HER2) positive, and triple-negative BC (TNBC). Based on the stages and subtypes of BC, various treatment methods are available with variations in the rates of progression-free disease and overall survival of patients. However, the treatment of BC still faces challenges, particularly in terms of drug resistance and recurrence. The study of epigenetics has provided new ideas for treating BC. Targeting aberrant epigenetic factors with inhibitors represents a promising anticancer strategy. The KDM5 family includes four members, KDM5A, KDM5B, KDM5C, and KDMD, all of which are Jumonji C domain-containing histone H3K4me2/3 demethylases. KDM5 proteins have been extensively studied in BC, where they are involved in suppressing or promoting BC depending on their specific upstream and downstream pathways. Several KDM5 inhibitors have shown potent BC inhibitory activity in vitro and in vivo, but challenges still exist in developing KDM5 inhibitors. In this review, we introduce the subtypes of BC and their current therapeutic options, summarize KDM5 family context-specific functions in the pathobiology of BC, and discuss the outlook and pitfalls of KDM5 inhibitors in this disease.


Asunto(s)
Neoplasias de la Mama , Histona Demetilasas , Terapia Molecular Dirigida , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/genética , Biomarcadores de Tumor
2.
Bioorg Chem ; 144: 107176, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38330721

RESUMEN

Repurposing drugs can significantly reduce the time and costs associated with drug discovery and development. However, many drug compounds possess intrinsic fluorescence, resulting in aberrations such as auto-fluorescence, scattering and quenching, in fluorescent high-throughput screening assays. To overcome these drawbacks, time-resolved technologies have received increasing attention. In this study, we have developed a rapid and efficient screening platform based on time-resolved emission spectroscopy in order to screen for inhibitors of the DNA repair enzyme, uracil-DNA glycosylase (UDG). From a database of 1456 FDA/EMA-approved drugs, sodium stibogluconate was discovered as a potent UDG inhibitor. This compound showed synergistic cytotoxicity against 5-fluorouracil-resistant cancer cells. This work provides a promising future for time-resolved technologies for high-throughput screening (HTS), allowing for the swift identification of bioactive compounds from previously overlooked scaffolds due to their inherent fluorescence properties.


Asunto(s)
Neoplasias de la Próstata , Uracil-ADN Glicosidasa , Humanos , Masculino , Uracil-ADN Glicosidasa/química , Oligonucleótidos , Gluconato de Sodio Antimonio , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Detección Precoz del Cáncer
3.
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
4.
Med Res Rev ; 43(5): 1438-1469, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37012609

RESUMEN

Histone methylation plays a key function in modulating gene expression, and preserving genome integrity and epigenetic inheritance. However, aberrations of histone methylation are commonly observed in human diseases, especially cancer. Lysine methylation mediated by histone methyltransferases can be reversed by lysine demethylases (KDMs), which remove methyl marks from histone lysine residues. Currently, drug resistance is a main impediment for cancer therapy. KDMs have been found to mediate drug tolerance of many cancers via altering the metabolic profile of cancer cells, upregulating the ratio of cancer stem cells and drug-tolerant genes, and promoting the epithelial-mesenchymal transition and metastatic ability. Moreover, different cancers show distinct oncogenic addictions for KDMs. The abnormal activation or overexpression of KDMs can alter gene expression signatures to enhance cell survival and drug resistance in cancer cells. In this review, we describe the structural features and functions of KDMs, the KDMs preferences of different cancers, and the mechanisms of drug resistance resulting from KDMs. We then survey KDM inhibitors that have been used for combating drug resistance in cancer, and discuss the opportunities and challenges of KDMs as therapeutic targets for cancer drug resistance.


Asunto(s)
Histonas , Neoplasias , Humanos , Histonas/química , Lisina/química , Lisina/metabolismo , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética
5.
Bioconjug Chem ; 34(10): 1727-1737, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37750807

RESUMEN

Glutathione S-transferase is heterogeneously expressed in breast cancer cells and is therefore emerging as a potential diagnostic biomarker for studying the heterogeneity of breast cancers. However, available fluorescent probes for GSTs depend heavily on GSTs-catalyzed glutathione (GSH) nucleophilic substitution reactions, making them susceptible to interference by the high concentration of nucleophilic species in the cellular environment. Moreover, the functions of subcellular GSTs are generally overlooked due to the lack of suitable luminescence probes. Herein, we report a highly selective affinity-based luminescence probe 1 for GST in breast cancer cells through tethering a GST inhibitor, ethacrynic acid, to an iridium(III) complex. Compared to activity-based probes which require the use of GSH, this probe could image GST-pi in the mitochondria by directly adducting to GST-pi (or potentially GST-pi/GS) in living cells. Probe 1 possesses desirable photophysical properties including a lifetime of 911 ns, a Stokes shift of 343 nm, and high photostability. The "turn on" luminescence mode of the probe enables highly selective detection of the GST with a limit of detection of 1.01 µM, while its long emission lifetime allows sensitive detection in organic dye-spiked autofluorescence samples by a time-resolved mode. The probe was further applied to specifically and quantitatively visualize MDA-MB-231 cells via specific binding to mitochondrial GST, and could differentiate breast cell lines based on their expression levels of GST. To the best of our knowledge, this probe is the first affinity-based iridium(III) imaging probe for the subcellular GST. Our work provides a valuable tool for unmasking the diverse roles of a subcellular GST in living systems, as well as for studying the heterogeneity of breast cancers.


Asunto(s)
Neoplasias de la Mama , Glutatión Transferasa , Humanos , Femenino , Glutatión Transferasa/metabolismo , Neoplasias de la Mama/diagnóstico por imagen , Iridio , Ácido Etacrínico , Mitocondrias/metabolismo , Glutatión/metabolismo
6.
Bioorg Chem ; 140: 106765, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37582330

RESUMEN

Janus kinases (JAKs) play a critical role in modulating the function and expression of inflammatory cytokines related to rheumatoid arthritis (RA). Herein, we report the design, synthesis, and structure-activity relationships (SARs) of a series of novel quinazoline derivatives as JAK inhibitors. Among these inhibitors, compound 11n showed high potency against JAKs (JAK1/JAK2/JAK3/TYK2, IC50 = 0.40, 0.83, 2.10, 1.95 nM), desirable metabolic characters, and excellent pharmacokinetic properties. In collagen-induced arthritis (CIA) models, compound 11n exhibited significant reduction in joint swelling with good safety, which could be served as a potential therapeutic candidate for the treatment of inflammatory diseases.


Asunto(s)
Artritis Reumatoide , Inhibidores de las Cinasas Janus , Humanos , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/uso terapéutico , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Quinasas Janus , Relación Estructura-Actividad , Artritis Reumatoide/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
7.
Sens Actuators B Chem ; 377: 133006, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36439053

RESUMEN

Common reference methods for COVID-19 variant diagnosis include viral sequencing and PCR-based methods. However, sequencing is tedious, expensive, and time-consuming, while PCR-based methods have high risk of insensitive detection in variant-prone regions and are susceptible to potential background signal interference in biological samples. Here, we report a loop-mediated interference reduction isothermal nucleic acid amplification (LM-IR-INA) strategy for highly sensitive single-base mutation detection in viral variants. This strategy exploits the advantages of nicking endonuclease-mediated isothermal amplification, luminescent iridium(III) probes, and time-resolved emission spectroscopy (TRES). Using the LM-IR-INA strategy, we established a luminescence platform for diagnosing COVID-19 D796Y single-base substitution detection with a detection limit of 2.01 × 105 copies/µL in a linear range of 6.01 × 105 to 3.76 × 108 copies/µL and an excellent specificity with a variant/wild-type ratio of significantly less than 0.0625%. The developed TRES-based method was also successfully applied to detect D796Y single-base substitution sequence in complicated biological samples, including throat and blood, and was a superior to steady-state technique. LM-IR-INA was also demonstrated for detecting the single-base substitution D614G as well as the multiple-base mutation H69/V70del without mutual interference, indicating that this approach has the potential to be used as a universal viral variant detection strategy.

8.
Mikrochim Acta ; 190(5): 171, 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-37016086

RESUMEN

A new method is presented for the one-step synthesis and real-time monitoring of iridium(III) complex-functionalized AuNPs from the precursor gold(III) chloride (AuCl3). The functionalized AuNPs with an average size of 8 - 20 nm were obtained by the reduction of Au3+ ions by the alkyne group of iridium(III) complexes, which was accompanied by the anchoring iridium(III) complexes on the surface of the nanoparticles. Meanwhile, the luminescence of the iridium(III) complexes was effectively quenched due to distance-dependent fluorescence quenching by AuNPs, thereby enabling luminescence monitoring of the formation process of the functionalized AuNPs and obtaining scattering information and spectral information in real time. Moreover, this method was applied to the determination of Au3+ ions in buffer with a limit of detection of 0.38 µM at 700 nm in luminescence mode, while the detection limit for absorbance was 10.04 µM. Importantly, the multimodal detection strategy alleviates interference from other metal ions. Furthermore, the iridium(III) alkyne complexes were capable of imaging mitochondrial Au3+ ions in living cells. Taken together, this work opens a new avenue for convenient synthesis and monitoring formation of functionalized AuNPs, and also provides a tool for selective determination of Au3+ ions in solution and in cellulo.

9.
Nano Lett ; 21(23): 9862-9868, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34780188

RESUMEN

Hybrid upconversion nanosystems have been reported to improve the low absorption efficiency of lanthanide-doped upconversion nanoparticles (UCNPs). However, the low quantum yield and poor photostability of NIR dyes pose challenges for practical uses. Here, we introduce a bulky moiety, 4-(1,2,2-triphenylvinyl)-1,1'-biphenyl (TPEO), to enhance its quantum yield by suppressing the bond rotation and improve the stability by deactivating the photoinduced oxidization. Compared with the conventional IR806, the formed NIR dye, TPEO-Cy, has been characterized to deliver three times higher quantum yield and seven times better photostability. Moreover, we take advantage of the strong affinity of sulfonate chains on the TPEO-Cy to bind to the surface of UCNPs. Taking together the synergistic effect, we have achieved a 242-fold upconversion emission enhancement over the benchmark of IR806-sensitized system and an ∼800 000-fold increase than the bare UCNPs. Our design of the NIR dyes suggests a new scope to search for more efficient upconversion nanohybrids.


Asunto(s)
Elementos de la Serie de los Lantanoides , Nanopartículas , Colorantes , Nanopartículas/química
10.
Bioorg Chem ; 114: 105158, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34378541

RESUMEN

Acetylation of NF-κB's RelA subunit at lysine-310 (AcLys310) helps to maintain constitutive NF-κB activity in cancers such as triple-negative breast cancer (TNBC). Bromodomain-containing factor BRD4 binds to acetylated RelA to promote the activity of NF-κB. Hence, interfering with the acetylated RelA-BRD4 interaction is a potential strategy for treating NF-κB-driven TNBC. Here, a new compound 13a was obtained by structural optimization and modification of our previously reported compound. In comparison with the well-known BRD4 inhibitor (+)-JQ1, 13a showed more potent anticancer activity in NF-κB-active MDA-MB-231 cells. Mechanistically, 13a antagonized the protein-protein interaction (PPI) between BRD4 and acetylated RelA, decreased levels of IL-6, IL-8, Snail, Vimentin, and ZEB1, induced cell senescence and DNA damage, and weakened the adhesion, metastasis, and invasion ability of TNBC cells. Our results provide insights into avenues for the further development of potent BRD4-acetylated RelA PPI inhibitors. Moreover, our findings highlight the effectiveness and feasibility of blocking the interaction between BRD4 and acetylated RelA against NF-κB-active cancers, and of screening antagonists of this PPI.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Indoles/farmacología , FN-kappa B/antagonistas & inhibidores , Ácidos Pentanoicos/farmacología , Factores de Transcripción/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antineoplásicos/síntesis química , Antineoplásicos/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indoles/química , Modelos Moleculares , Estructura Molecular , FN-kappa B/metabolismo , Ácidos Pentanoicos/química , Relación Estructura-Actividad , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
11.
Angew Chem Int Ed Engl ; 59(41): 17897-17902, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32649787

RESUMEN

Gastrin-releasing peptide receptor (GRPr) plays proliferative and inflammatory roles in living systems. Here, we report a highly selective GRPr antagonist (JMV594)-tethered iridium(III) complex for probing GRPr in living cancer cells and immune cells. This probe exhibited desirable photophysical properties and also displayed negligible cytotoxicity, overcoming the inherent toxicity of the iridium(III) complex. Its long emission lifetime enabled its luminescence signal to be readily distinguished from the interfering fluorescence of organic dyes by using a time-resolved technique. This probe selectively visualized living cancer cells via specific binding to GRPr, while it also modulated the function of GRPr on TNF-α secretion in immune cells. To our knowledge, this is the first peptide-conjugated iridium(III) complex developed as a GRPr bioimaging probe and modulator of GRPr activity. This theranostic agent shows great potential at unmasking the diverse roles of GRPr in living systems.


Asunto(s)
Péptidos/metabolismo , Medicina de Precisión , Receptores de Bombesina/metabolismo , Células A549 , Animales , Humanos , Ratones , Células RAW 264.7 , Análisis Espectral/métodos
12.
Angew Chem Int Ed Engl ; 57(40): 13091-13095, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29968419

RESUMEN

Lysine-specific demethylase 5A (KDM5A) has recently become a promising target for epigenetic therapy. In this study, we designed and synthesized metal complexes bearing ligands with reported demethylase and p27 modulating activities. The Rh(III) complex 1 was identified as a direct, selective and potent inhibitor of KDM5A that directly abrogate KDM5A demethylase activity via antagonizing the KDM5A-tri-/di-methylated histone 3 protein-protein interaction (PPI) in vitro and in cellulo. Complex 1 induced accumulation of H3K4me3 and H3K4me2 levels in cells, causing growth arrest at G1 phase in the triple-negative breast cancer (TNBC) cell lines, MDA-MB-231 and 4T1. Finally, 1 exhibited potent anti-tumor activity against TNBC xenografts in an in vivo mouse model, presumably via targeting of KDM5A and hence upregulating p27. Moreover, complex 1 was less toxic compared with two clinical drugs, cisplatin and doxorubicin. To our knowledge, complex 1 is the first metal-based KDM5A inhibitor reported in the literature. We anticipate that complex 1 may be used as a novel scaffold for the further development of more potent epigenetic agents against cancers, including TNBC.


Asunto(s)
Complejos de Coordinación/química , Proteína 2 de Unión a Retinoblastoma/antagonistas & inhibidores , Rodio/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular , Complejos de Coordinación/uso terapéutico , Complejos de Coordinación/toxicidad , Femenino , Histonas/antagonistas & inhibidores , Histonas/metabolismo , Humanos , Iridio/química , Ratones , Ratones Endogámicos BALB C , Proteína 2 de Unión a Retinoblastoma/metabolismo , Trasplante Heterólogo , Neoplasias de la Mama Triple Negativas/patología
13.
Anal Chem ; 89(21): 11679-11684, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-28969424

RESUMEN

ß-Galactosidase (ß-gal) is an important biomarker for ovarian cancers. In this work, we designed and synthesized a novel iridium(III)-based probe 1 for discriminating ovarian carcinoma cell lines from normal cell lines. The probe could detect ß-gal even in the presence of a highly autofluorescent background. The probe also showed a good linear response to ß-gal between 0 and 30 U/mL, with a detection limit of 0.51 U/mL. Importantly, complex 1 could selectively "light up" ovarian carcinoma cells, while exhibiting negligible luminescence in normal cells. Overall, complex 1 could be potentially used as a useful probe for detecting ß-gal expression in the context of ovarian cancer diagnostics.


Asunto(s)
Sustancias Luminiscentes/metabolismo , Imagen Óptica/métodos , Neoplasias Ováricas/patología , beta-Galactosidasa/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Femenino , Humanos , Iridio/química , Sustancias Luminiscentes/química
14.
Chemistry ; 23(20): 4929-4935, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28211965

RESUMEN

NF-κB is a critical transcription factor that plays an important role in mediating inflammation, the immune response, and cell proliferation. The activation of NF-κB leads to an enhancement of proinflammatory mediator expression, which is implicated in the pathogenesis of a variety of diseases. Therefore, methods that allow the intracellular tracking of NF-κB are particularly attractive because they can provide information regarding the pathways or stimulation responses that are involved in the activation of NF-κB. In this work, we report a novel platform to track intracellular NF-κB by employing the conjugated iridium(III) complex 1, which was synthesized through the unique combination of a luminescent iridium(III) moiety with the natural product oridonin. Experiments conducted with p50 knockdown cells revealed that complex 1 could detect the p50 subunit of NF-κB in cellulo. Furthermore, complex 1 tracked NF-κB translocation induced by tumor necrosis factor-α (TNF-α) as a model stimulus, without affecting the translocation process itself. To the best of our knowledge, complex 1 is the first metal-based compound that has been reported to be capable of monitoring intracellular NF-κB in living cells.


Asunto(s)
Complejos de Coordinación/síntesis química , Diterpenos de Tipo Kaurano/química , Iridio/química , FN-kappa B/metabolismo , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Células HeLa , Humanos , Microscopía Fluorescente , Subunidad p50 de NF-kappa B/deficiencia , Subunidad p50 de NF-kappa B/genética , Activación Transcripcional/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
15.
Biochim Biophys Acta Gen Subj ; 1861(2): 256-263, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27888146

RESUMEN

BACKGROUND: Cancer of the skin is by far the most common of all cancers. Melanoma accounts for only about 1% of skin cancers but causes a large majority of skin cancer deaths. Autotaxin (ATX), also known as ectonucleotide pyrophosphatase/phosphodiesterase 2 (ENPP2), regulates physiological and pathological functions of lysophosphatidic acid (LPA), and is thus an important therapeutic target. METHODS: We synthesized ten metal-based complexes and a novel cyclometalated rhodium(III) complex 1 was identified as an ATX enzymatic inhibitor using multiple methods, including ATX enzymatic assay, thermal shift assay, western immunoblotting and so on. RESULTS: Protein thermal shift assays showed that 1 increased the melting temperature (Tm) of ATX by 3.5°C. 1 also reduced ATX-LPA mediated downstream survival signal pathway proteins such as ERK and AKT, and inhibited the activation of the transcription factor nuclear factor κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3). 1 also exhibited strong anti-proliferative activity against A2058 melanoma cells (IC50=0.58µM). Structure-activity relationship indicated that both the rhodium(III) center and the auxiliary ligands of complex 1 are important for bioactivity. CONCLUSIONS: 1 represents a promising scaffold for the development of small-molecule ATX inhibitors for anti-tumor applications. To our knowledge, complex 1 is the first metal-based ATX inhibitor reported to date. GENERAL SIGNIFICANCE: Rhodium complexes will have the increased attention in therapeutic and bioanalytical applications.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Melanoma/tratamiento farmacológico , Hidrolasas Diéster Fosfóricas/metabolismo , Rodio/farmacología , Línea Celular Tumoral , Humanos , Lisofosfolípidos/farmacología , Melanoma/metabolismo , Complejos Multienzimáticos/metabolismo , FN-kappa B/metabolismo , Fosfodiesterasa I/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
16.
Anal Chem ; 88(1): 981-7, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26607385

RESUMEN

In this study, an unreported Ir(III) complex 1 was identified by screening as a versatile G-quadruplex probe. It exhibited highly selective response for different G-quadruplex DNA over double strand, single strand and triplex DNA. Compared with the organic G-quadruplex probe thioflavin T, complex 1 displays a longer lifetime, a larger Stokes shift, comparable G-quadruplex/ssDNA enhancement ratios, and higher G-quadruplex/triplex DNA enhancement ratios. In consideration of the encouraging G-quadruplex probe performance of complex 1, we employed 1 to develop a G-quadruplex-based detection system for the detection of insulin as a "proof-of-principle" concept. We also demonstrate an optimization process that enhanced the sensitivity of this sensing assay. Compared to previously reported methods, our "mix-and-detect" detection methodology is easy operated, quick, and cost-effective. A detection limit as low as 80 pM for insulin can be achieved by this sensing approach, with a linear relationship between luminescence intensity and insulin concentration established from 80 pM to 20 nM. Moreover, this assay could work effectively in diluted human serum.


Asunto(s)
Colorantes Fluorescentes/química , G-Cuádruplex , Insulina/sangre , Iridio/química , Compuestos Organometálicos/química , Humanos , Luminiscencia , Mediciones Luminiscentes , Estructura Molecular
17.
Sci Technol Adv Mater ; 17(1): 109-114, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27877862

RESUMEN

In this study, we report the use of a cyclometalated luminescent iridium(III) complex for the visualization of thiols. The detection of glutathione (GSH) by complex 1 is achieved through the reduction of its phendione N^N donor, which influences the metal-to-ligand charge-transfer (MLCT) of the complex. Complex 1 produced a maximum threefold luminescence enhancement at 587 nm in response to GSH. The linear detection range of 1 for GSH is between 0.2 and 2 M equivalents of GSH, with a detection limit of 1.67 µM. Complex 1 also displays good selectivity for thiols over other amino acids.

18.
Sci Technol Adv Mater ; 16(6): 065004, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27877846

RESUMEN

We report herein the use of an exonuclease III and G-quadruplex probe to construct a G-quadruplex-based luminescence detection platform for Hg2+. Unlike common DNA-based Hg2+ detection methods, when using the dsDNA probe to monitor the hairpin formation, the intercalation of the dsDNA probe may be influenced by the distortion of dsDNA. This 'mix-and-detect' methodology utilized the G-quadruplex probe as the signal transducer and is simple, rapid, convenient to use and can detect down to 20 nM of Hg2+.

19.
Eur J Med Chem ; 264: 115995, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38043488

RESUMEN

Hepatocellular carcinoma (HCC) is a major contributor to global mortality rates, but current treatment options have limitations. Advanced theranostics are needed to effectively integrate diagnosis and therapeutic of HCC. Glycyrrhetinic acid (GA) has abundant binding sites with glycyrrhetinic acid receptors (GA-Rs) on the surface of HCC cells and has also been reported to possess ligands with mitochondrial-targeting capability but with limited efficacy. Herein, we report a near-infrared (NIR) luminescent theranostic complex 1 through conjugating an iridium(III) complex to GA, which exhibits the desired photophysical properties and promotes mitochondrial-targeting capability. Complex 1 was selectively taken up by HepG2 liver cancer cells and was imaged within mitochondria with NIR emission. Complex 1 targeted mitochondria and opened mitochondrial permeability transition pores (MPTPs), resulting in ROS accumulation, mitochondrial damage, disruption of Bax/Bcl-2 equilibrium, and tumor cell apoptosis, resulting in significantly improved anticancer activity compared to GA. This work offers a methodology for developing multifunctional theranostic probes with amplified specificity and efficacy.


Asunto(s)
Carcinoma Hepatocelular , Ácido Glicirretínico , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Medicina de Precisión , Iridio/farmacología , Iridio/química , Ácido Glicirretínico/farmacología , Ácido Glicirretínico/química , Mitocondrias/metabolismo , Línea Celular Tumoral
20.
Int J Biol Macromol ; 259(Pt 1): 129211, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38184034

RESUMEN

The overexpression and overactivation of epidermal growth factor receptor (EGFR) are frequently observed in human cancers, including squamous cell carcinoma and adenocarcinoma. In this study, a covalent EGFR probe was developed by conjugating afatinib to an iridium(III) scaffold. Complex 1 showed enhanced luminescence in living epidermoid squamous carcinoma A431 cells compared to other cell lines, via engaging EGFR as confirmed via CETSA and knockdown experiments. Moreover, complex 1 inhibited downstream targets of EGFR in cellulo with repression persisting after removal of the complex, indicating an irreversible mode of inhibition. Finally, complex 1 showed potent antiproliferative activity against A431 cells with comparable potency to afatinib alone. To our knowledge, complex 1 is the first EGFR covalent inhibitor based on an iridium scaffold reported in the literature, with the potential to be further explored as a theranostic agent in the future.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Pulmonares , Humanos , Afatinib , Iridio/farmacología , Quinazolinas/farmacología , Receptores ErbB/metabolismo , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA