Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(25): e2316615121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38861602

RESUMEN

Many cancer-driving protein targets remain undruggable due to a lack of binding molecular scaffolds. In this regard, octahedral metal complexes with unique and versatile three-dimensional structures have rarely been explored as inhibitors of undruggable protein targets. Here, we describe antitumor iridium(III) pyridinium-N-heterocyclic carbene complex 1a, which profoundly reduces the viability of lung and breast cancer cells as well as cancer patient-derived organoids at low micromolar concentrations. Compound 1a effectively inhibits the growth of non-small-cell lung cancer and triple-negative breast cancer xenograft tumors, impedes the metastatic spread of breast cancer cells, and can be modified into an antibody-drug conjugate payload to achieve precise tumor delivery in mice. Identified by thermal proteome profiling, an important molecular target of 1a in cellulo is Girdin, a multifunctional adaptor protein that is overexpressed in cancer cells and unequivocally serves as a signaling hub for multiple pivotal oncogenic pathways. However, specific small-molecule inhibitors of Girdin have not yet been developed. Notably, 1a exhibits high binding affinity to Girdin with a Kd of 1.3 µM and targets the Girdin-linked EGFR/AKT/mTOR/STAT3 cancer-driving pathway, inhibiting cancer cell proliferation and metastatic activity. Our study reveals a potent Girdin-targeting anticancer compound and demonstrates that octahedral metal complexes constitute an untapped library of small-molecule inhibitors that can fit into the ligand-binding pockets of key oncoproteins.


Asunto(s)
Antineoplásicos , Iridio , Metano , Animales , Humanos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Iridio/química , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metano/análogos & derivados , Metano/química , Metano/farmacología , Proteínas de Microfilamentos/metabolismo , Metástasis de la Neoplasia , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Masculino
2.
Chem Sci ; 15(14): 5349-5359, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38577372

RESUMEN

Silver compounds have favorable properties as promising anticancer drug candidates, such as low side effects, anti-inflammatory properties, and high potential to overcome drug resistance. However, the exact mechanism by which Ag(i) confers anticancer activity remains unclear, which hinders further development of anticancer applications of silver compounds. Here, we combine thermal proteome profiling, cysteine profiling, and ubiquitome profiling to study the molecular mechanisms of silver(i) complexes supported by non-toxic thiourea (TU) ligands. Through the formation of AgTU complexes, TU ligands deliver Ag+ ions to cancer cells and tumour xenografts to elicit inhibitory potency. Our chemical proteomics studies show that AgTU acts on the ubiquitin-proteasome system (UPS) and disrupts protein homeostasis, which has been identified as a main anticancer mechanism. Specifically, Ag+ ions are released from AgTU in the cellular environment, directly target the 19S proteasome regulatory complex, and may oxidize its cysteine residues, thereby inhibiting proteasomal activity and accumulating ubiquitinated proteins. After AgTU treatment, proteasome subunits are massively ubiquitinated and aberrantly aggregated, leading to impaired protein homeostasis and paraptotic death of cancer cells. This work reveals the unique anticancer mechanism of Ag(i) targeting the 19S proteasome regulatory complex and opens up new avenues for optimizing silver-based anticancer efficacy.

3.
Am J Stem Cells ; 12(3): 51-59, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37736270

RESUMEN

OBJECTIVE: This study investigated if silver nanoparticles (AgNps) could promote the proliferation and osteogenic differentiation of mouse embryonic fibroblasts. METHODS: Mouse embryonic fibroblasts were divided into two groups: Group 1 cells were cultured in DMEM/F12 medium and Group 2 cells were cultured in osteogenic medium. Both groups were then treated with 16, 32, or 100 µM AgNps. Fibroblast proliferation and viability were measured using BrdU and MTT methods at varying time points. Alizarin red staining and alkaline phosphatase (ALP) activity were measured to observe fibroblast differentiation into osteoblasts. Proteomics (cytokine array) was used to detect 111 different cytokines during differentiation. RESULTS: AgNps stimulated proliferation of mouse embryonic fibroblasts at a concentration of 16 µM. Marked enhancement of calcium mineralization was observed in cells cultured with AgNps compared with cells cultured without AgNps. Group 2 cells displayed nodules around the center where the cell density was high. ALP activity of mouse embryonic fibroblasts cultured in osteogenic medium increased during the whole culture period. Addition of AgNps at concentrations of 32 µM and 100 µM induced higher ALP activity at days 7 and 14. Proteomic array results show that low density lipoprotein receptor (LDL-R) and proprotein convertase subtilisin/kexin type 9 (PCSK-9) were significantly increased, while osteoprotegerin (OPG) was significantly reduced in medium containing 16 µM AgNPs. CONCLUSION: AgNps could promote differentiation of mouse embryonic fibroblasts into osteoblastic cells. LDL-R and PCSK-9, as well as OPG, may play a critical role in this process.

4.
Chem Commun (Camb) ; 59(19): 2747-2750, 2023 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-36757177

RESUMEN

Dihydroartemisinin non-covalently binds liver fatty acid binding protein (FABP1) with micromolar affinity, acts as a FABP1-dependent peroxisome proliferator-activated receptor alpha agonist and inhibits metastatic hepatocellular carcinoma growth.


Asunto(s)
Artemisininas , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Proteínas de Unión a Ácidos Grasos/metabolismo , Hígado/metabolismo
5.
Cell Rep ; 41(7): 111647, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36384131

RESUMEN

Identifying signals that govern the differentiation of tumor-infiltrating CD8+ T cells (CD8+ TILs) toward exhaustion can improve current therapeutic approaches for cancer. Here, we show that type I interferons (IFN-Is) act as environmental cues, enhancing terminal CD8+ T cell exhaustion in tumors. We find enrichment of IFN-I-stimulated genes (ISGs) within exhausted CD8+ T cells (Tex cells) in patients across various cancer types, with heightened ISG levels correlating with poor response to immune checkpoint blockade (ICB) therapy. In preclinical models, CD8+ TILs devoid of IFN-I signaling develop less exhaustion features, provide better tumor control, and show greater response to ICB-mediated rejuvenation. Mechanistically, chronic IFN-I stimulation perturbs lipid metabolism and redox balance in Tex cells, leading to aberrant lipid accumulation and elevated oxidative stress. Collectively, these defects promote lipid peroxidation, which potentiates metabolic and functional exhaustion of Tex cells. Thus, cell-intrinsic IFN-I signaling regulates the extent of CD8+ TIL exhaustion and has important implications for immunotherapy.


Asunto(s)
Enfermedad Injerto contra Huésped , Interferón Tipo I , Neoplasias , Humanos , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1/metabolismo , Peroxidación de Lípido , Neoplasias/metabolismo , Interferón Tipo I/metabolismo , Lípidos
6.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33883283

RESUMEN

Vimentin is a cytoskeletal intermediate filament protein that plays pivotal roles in tumor initiation, progression, and metastasis, and its overexpression in aggressive cancers predicted poor prognosis. Herein described is a highly effective antitumor and antimetastatic metal complex [PtII(C^N^N)(NHC2Bu)]PF6 (Pt1a; HC^N^N = 6-phenyl-2,2'-bipyridine; NHC= N-heterocyclic carbene) that engages vimentin via noncovalent binding interactions with a distinct orthogonal structural scaffold. Pt1a displays vimentin-binding affinity with a dissociation constant of 1.06 µM from surface plasmon resonance measurements and fits into a pocket between the coiled coils of the rod domain of vimentin with multiple hydrophobic interactions. It engages vimentin in cellulo, disrupts vimentin cytoskeleton, reduces vimentin expression in tumors, suppresses xenograft growth and metastasis in different mouse models, and is well tolerated, attributable to biotransformation to less toxic and renal-clearable platinum(II) species. Our studies uncovered the practical therapeutic potential of platinum(II)‒NHC complexes as effective targeted chemotherapy for combating metastatic and cisplatin-resistant cancers.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Compuestos Organoplatinos/uso terapéutico , Vimentina/efectos de los fármacos , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Femenino , Células HCT116 , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Simulación de Dinámica Molecular , Compuestos Organoplatinos/metabolismo , Compuestos Organoplatinos/farmacología , Ratas , Vimentina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Chem Sci ; 12(46): 15229-15238, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34976343

RESUMEN

Self-assembly of platinum(ii) complexes to form supramolecular structures/nanostructures due to intermolecular ligand π-π stacking and metal-ligand dispersive interactions is widely used to develop functional molecular materials, but the application of such non-covalent molecular interactions has scarcely been explored in medical science. Herein is described the unprecedented biological properties of platinum(ii) complexes relevant to induction of cancer cell death via manifesting such intermolecular interactions. With conjugation of a glucose moiety to the planar platinum(ii) terpyridyl scaffold, the water-soluble complex [Pt(tpy)(C[triple bond, length as m-dash]CArOGlu)](CF3SO3) (1a, tpy = 2,2':6',2''-terpyridine, Glu = glucose) is able to self-assemble into about 100 nm nanoparticles in physiological medium, be taken up by lung cancer cells via energy-dependent endocytosis, and eventually transform into other superstructures distributed in endosomal/lysosomal and mitochondrial compartments apparently following cleavage of the glycosidic linkage. Accompanying the formation of platinum-containing superstructures are increased autophagic vacuole formation, lysosomal membrane permeabilization, and mitochondrial membrane depolarization, as well as anti-tumor activity of 1a in a mouse xenograft model. These findings highlight the dynamic, multi-stage extracellular and intracellular supramolecular self-assembly of planar platinum(ii) complexes driven by modular intermolecular interactions with potential anti-cancer application.

8.
Front Chem ; 8: 587207, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240849

RESUMEN

The use of gold in medicine has a long history. Recent clinical applications include anti-inflammatory agents for the treatment of rheumatoid arthritis (chrysotherapy), and is currently being developed as potential anticancer chemotherapeutics. Gold(III), being isoelectronic to platinum(II) as in cisplatin, is of great interest but it is inherently unstable and redox-reactive under physiological conditions. Coordination ligands containing C and/or N donor atom(s) such as porphyrin, pincer-type cyclometalated and/or N-heterocyclic carbene (NHC) can be employed to stabilize gold(III) ion for the preparation of anticancer active compounds. In this review, we described our recent work on the anticancer properties of gold(III) compounds and the identification of molecular targets involved in the mechanisms of action. We also summarized the chemical formulation strategies that have been adopted for the delivery of cytotoxic gold compounds, and for ameliorating the in vivo toxicity.

9.
Pharm Res ; 37(11): 220, 2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-33051728

RESUMEN

PURPOSE: Gold porphyrin (AuP) is a complex that has been shown to be potent against various tumors. A biocompatible interpenetrating network (IPN) system comprised of polyethyleneglycol diacrylate (PEGdA) and chemically-modified gelatin has been shown to be an effective implantable drug depot to deliver AuP locally. Here we designed IPN microparticles complexed with AuP to facilitate intravenous administration and to diminish systemic toxicity. METHODS: We have synthesized and optimized an IPN microparticle formulation complexed with AuP. Tumor cell cytotoxicity, antitumor activity, and survival rate in lung cancer bearing nude mice were analyzed. RESULTS: IPN microparticles maintained AuP bioactivity against lung cancer cells (NCI-H460). In vivo study showed no observable systemic toxicity in nude mice bearing NCI-H460 xenografts after intravenous injection of 6 mg/kg AuP formulated with IPN microparticles. An anti-tumor activity level comparable to free AuP was maintained. Mice treated with 6 mg/kg AuP in IPN microparticles showed 100% survival rate while the survival rate of mice treated with free AuP was much less. Furthermore, microparticle-formulated AuP significantly reduced the intratumoral microvasculature when compared with the control. CONCLUSION: AuP in IPN microparticles can reduce the systemic toxicity of AuP without compromising its antitumor activity. This work highlighted the potential application of AuP in IPN microparticles for anticancer chemotherapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Oro/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Metaloporfirinas/farmacología , Administración Intravenosa , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/química , Animales , Línea Celular Tumoral , Composición de Medicamentos , Oro/administración & dosificación , Oro/química , Humanos , Neoplasias Pulmonares/patología , Metaloporfirinas/administración & dosificación , Metaloporfirinas/química , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Analyst ; 145(19): 6237-6242, 2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32839801

RESUMEN

HPV-induced cervical cancer is one of the most lethal cancers. Therefore, the development of a reliable and accurate method for the early diagnosis of HPV infections is highly important. Here, gold nanoparticles (AuNPs) were utilized as mass tags in an immuno-capture LI-MS assay for the detection of HPV marker proteins. Through the optimization of the amount of antibodies and surface charges on AuNPs, high antigen detection efficiency with minimal non-specific binding was achieved. With optimized antibody-conjugated AuNPs, low attomole amount of HPV proteins in HeLa cell lysate was quantified.


Asunto(s)
Oro , Nanopartículas del Metal , Biomarcadores , Células HeLa , Humanos , Proteínas
11.
Sci Rep ; 10(1): 6338, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286492

RESUMEN

Burn injury is common, and antimicrobial agents are often applied immediately to prevent wound infection and excessive inflammatory response. Although inflammation is essential for clearing bacteria and creating an environment conducive to the healing process, it is unclear what time-frame inflammation should be present for optimal wound healing. This study critically investigated the role of early inflammation in burn wound healing, and also revealed the molecular mechanisms underlying the pro-healing effects of silver nanoparticles (AgNPs). We created a burn injury mouse model using wild-type and Smad3-/- mice, which were topically treated with AgNPs at different post-burn days, and examined the healing processes of the various groups. We also delineated the molecular pathways underlying the anti-inflammation and pro-healing effects of AgNPs by morphological and histological analysis, immuno-histochemistry, and western blotting. Our results showed that (1) AgNPs regulated pro-inflammatory cytokine IL-6 production of keratinocytes and neutrophils infiltration through KGF-2/p38 signaling pathway, (2) Topical AgNPs treatment immediately after burn injury significantly supressed early inflammation but resulted in delayed healing, (3) A short delay in AgNPs application (post-burn day 3 in our model) allowed early inflammation in a controlled manner, and led to optimal burn wound healing. Thus, our current study showed that some degree of early inflammation was beneficial, but prolonged inflammation was detrimental for burn wound healing. Further evaluation and clinical translation of this finding is warranted.


Asunto(s)
Quemaduras/tratamiento farmacológico , Nanopartículas del Metal/administración & dosificación , Nanopartículas del Metal/uso terapéutico , Plata/uso terapéutico , Cicatrización de Heridas , Animales , Quemaduras/metabolismo , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Nanopartículas del Metal/química , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Plata/administración & dosificación , Plata/química , Proteína smad3/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Proc Natl Acad Sci U S A ; 117(3): 1321-1329, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31896586

RESUMEN

Cysteine thiols of many cancer-associated proteins are attractive targets of anticancer agents. Herein, we unequivocally demonstrate a distinct thiol-targeting property of gold(III) mesoporphyrin IX dimethyl ester (AuMesoIX) and its anticancer activities. While the binding of cysteine thiols with metal complexes usually occurs via M-S bond formation, AuMesoIX is unique in that the meso-carbon atom of the porphyrin ring is activated by the gold(III) ion to undergo nucleophilic aromatic substitution with thiols. AuMesoIX was shown to modify reactive cysteine residues and inhibit the activities of anticancer protein targets including thioredoxin, peroxiredoxin, and deubiquitinases. Treatment of cancer cells with AuMesoIX resulted in the formation of gold-bound sulfur-rich protein aggregates, oxidative stress-mediated cytotoxicity, and accumulation of ubiquitinated proteins. Importantly, AuMesoIX exhibited effective antitumor activity in mice. Our study has uncovered a gold(III)-induced ligand scaffold reactivity for thiol targeting that can be exploited for anticancer applications.


Asunto(s)
Antineoplásicos/química , Cisteína/química , Oro/química , Mesoporfirinas/química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Enzimas Desubicuitinizantes/química , Enzimas Desubicuitinizantes/metabolismo , Células HCT116 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Peroxirredoxinas/química , Peroxirredoxinas/metabolismo , Unión Proteica , Tiorredoxinas/química , Tiorredoxinas/metabolismo , Distribución Tisular
13.
Metallomics ; 11(11): 1925-1936, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31631207

RESUMEN

Auranofin (AuRF) has been reported to display anticancer activity and has entered several clinical trials; however, its mechanism of action remains largely unknown. In this work, the anticancer mechanism of auranofin was investigated using a proteomics strategy entailing subcellular fractionation prior to mass spectrometric analysis. Bioinformatics analysis of the nuclear sub-proteomes revealed that tumor suppressor p14ARF is a key regulator of transcription. Through independent analysis, we validated that up-regulation of p14ARF is associated with E2F-dependent transcription and increased p53 expression. Our analyses further reveal that 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), which is the rate-determining enzyme of the mevalonate pathway, is a novel target of auranofin with half maximal inhibitory concentration at micromolar levels. The auranofin-induced cancer cell death could be partially reverted by the addition of downstream products of the mevalonate pathway (mevalonolactone or geranyleranyl pyrophosphate (GGPP)), implying that auranofin may target the mevalonate pathway to exert its anticancer effect.


Asunto(s)
Antineoplásicos/farmacología , Auranofina/farmacología , Hidroximetilglutaril-CoA Reductasas/metabolismo , Terapia Molecular Dirigida , Línea Celular Tumoral , Factores de Transcripción E2F/metabolismo , Oro/farmacología , Humanos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
Angew Chem Int Ed Engl ; 58(32): 10914-10918, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31165553

RESUMEN

New anticancer platinum(II) compounds with distinctive modes of action are appealing alternatives to combat the drug resistance and improve the efficacy of clinically used platinum chemotherapy. Herein, we describe a rare example of an antitumor PtII complex targeting a tumor-associated protein, rather than DNA, under cellular conditions. Complex [(bis-NHC)Pt(bt)]PF6 (1 a; Hbt=1-(3-hydroxybenzo[b]thiophen-2-yl)ethanone) overcomes cisplatin resistance in cancer cells and displays significant tumor growth inhibition in mice with higher tolerable doses compared to cisplatin. The cellular Pt species shows little association with DNA, and localizes in the cytoplasm as revealed by nanoscale secondary ion mass spectrometry. An unbiased thermal proteome profiling experiment identified asparagine synthetase (ASNS) as a molecular target of 1 a. Accordingly, 1 a treatment reduced the cellular asparagine levels and inhibited cancer cell proliferation, which could be reversed by asparagine supplementation. A bis-NHC-ligated Pt species generated from the hydrolysis of 1 a forms adducts with thiols and appears to target an active-site cysteine of ASNS.


Asunto(s)
Antineoplásicos/farmacología , Aspartatoamoníaco Ligasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Compuestos Organoplatinos/farmacología , Antineoplásicos/química , Aspartatoamoníaco Ligasa/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Cisplatino/química , Cisplatino/farmacología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Humanos , Ligandos , Estructura Molecular , Compuestos Organoplatinos/química , Relación Estructura-Actividad
15.
Pharm Res ; 36(4): 61, 2019 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850894

RESUMEN

PURPOSE: Interpenetrating network system (IPN), consisting of polyethylene glycol (PEG) -diacrylate (PEGdA) and modified gelatin, is a biocompatible and biodegradable hydrogel and has been studied for the local delivery of bioactive molecules and drugs. Gold(III) porphyrin(AuP) is a stable metal compound in the development for anticancer application when administered systemically. The aim of this work is to develop a novel formulation for AuP based on IPN for local delivery. METHODS: IPN loaded with AuP hydrogel was optimized and synthesized. Drug release kinetics, cytotoxicity against tumor cells, and antitumor activity in lung cancer bearing nude mice were studied. RESULTS: AuP released from the IPN followed a first order kinetics in vitro. The AuP loaded IPN showed higher cytotoxicity against human lung cancer cell lines compared to IPN only. In mice bearing human lung cancer xenograft, AuP loaded IPN inhibited tumor growth and reduced angiogenesis. No sign of systemic toxicity was observed for all treatment groups. CONCLUSION: AuP loaded IPN provides an improved formulation over systemic delivery for tumor inhibition to complement surgical intervention. Graphical Abstract Injectable multifunctional matrix of polyethylene glycol and gelatin derivatives for the delivery of gold porphyrinto inhibit tumor growth.


Asunto(s)
Compuestos de Oro/farmacología , Xenoinjertos/efectos de los fármacos , Hidrogeles/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Materiales Biocompatibles/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Gelatina/química , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Cinética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Polietilenglicoles/química , Trasplante Heterólogo/métodos
16.
Chem Sci ; 10(1): 293-309, 2019 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-30746082

RESUMEN

A panel of iridium(iii) porphyrin complexes containing axial N-heterocyclic carbene (NHC) ligand(s) were synthesized and characterized. X-ray crystal structures of the bis-NHC complexes [IrIII(ttp)(IMe)2]+ (2a), [IrIII(oep)(BIMe)2]+ (2d), [IrIII(oep)(I i Pr)2]+ (2e) and [IrIII(F20tpp)(IMe)2]+ (2f) display ruffled porphyrin rings with mesocarbon displacements of 0.483-0.594 Å and long Ir-CNHC bonds of 2.100-2.152 Å. Variable-temperature 1H NMR analysis of 2a reveals that the macrocycle porphyrin ring inversion takes place in solution with an activation barrier of 40 ± 1 kJ mol-1. The UV-vis absorption spectra of IrIII(por)-NHC complexes display split Soret bands. TD-DFT calculations and resonance Raman experiments show that the higher-energy Soret band is derived from the 1MLCT dπ(Ir) → π*(por) transition. The near-infrared phosphorescence of IrIII(por)-NHC complexes from the porphyrin-based 3(π, π*) state features broad emission bands at 701-754 nm with low emission quantum yields and short lifetimes (Φ em < 0.01; τ < 4 µs). [IrIII(por)(IMe)2]+ complexes (por = ttp and oep) are efficient photosensitizers for 1O2 generation (Φ so = 0.64 and 0.88) and are catalytically active in the light-induced aerobic oxidation of secondary amines and arylboronic acid. The bis-NHC complexes exhibit potent dark cytotoxicity towards a panel of cancer cells with IC50 values at submicromolar levels. The cytotoxicity of these complexes could be further enhanced upon light irradiation with IC50 values as low as nanomolar levels in association with the light-induced generation of reactive oxygen species (ROS). Bioimaging of [IrIII(oep)(IMe)2]+ (2c) treated cells indicates that this Ir complex mainly targets the endoplasmic reticulum. [IrIII(oep)(IMe)2]+ catalyzes the photoinduced generation of singlet oxygen and triggers protein oxidation, cell cycle arrest, apoptosis and the inhibition of angiogenesis. It also causes pronounced photoinduced inhibition of tumor growth in a mouse model of human cancer.

18.
Anal Chim Acta ; 1055: 1-6, 2019 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-30782360

RESUMEN

The development of sensitive and specific analytical methods is critical for the discovery of molecular biomarkers, which assists disease diagnosis and understanding biological processes. Herein, a highly sensitive method is developed using antibody-conjugated plasmonic metal nanoparticles for the detection of targeted biomarkers down to low attomole level via coupling of immunoassay techniques with laser ionization mass spectrometry (LI-MS). The conjugated antibodies target specific antigens, while the metal nanoparticles act as mass tags and ion reservoirs for the signal amplification. With the characteristic localized surface plasmon resonance (LSPR) properties, gold (AuNPs) and silver nanoparticles (AgNPs) undergo explosive ionization upon laser irradiation to generate abundant characteristic mass reporter ions for strong MS signal amplification. With the antibody-conjugated NPs, detection of trace proteins in various biological samples with complex matrix environment, including urine, cell lysates, and animal tissues was demonstrated.


Asunto(s)
Oro/química , Límite de Detección , Nanopartículas del Metal/química , Proteínas/metabolismo , Resonancia por Plasmón de Superficie/métodos , Animales , Biomarcadores/metabolismo , Línea Celular Tumoral , Gonadotropina Coriónica/metabolismo , Riñón/metabolismo , Espectrometría de Masas , Ratas
19.
J Proteome Res ; 17(7): 2470-2479, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29812950

RESUMEN

Dehydroeffusol (DHE) is a phenanthrene isolated from the Chinese medicinal plant Juncus effusus. Biological evaluation of DHE reveals in vitro and in vivo anticancer effects. We performed a shotgun proteomic analysis using liquid chromatography-tandem mass spectrometry to investigate the changes in the protein profiles in cancer cells upon DHE treatment. DHE affected cancer-associated signaling pathways, including NF-κB, ß-catenin, and endoplasmic reticulum stress. Through quantitative pathway and key node analysis of the proteomics data, activating transcription factor 2 (ATF-2) and c-Jun kinase (JNK) were found to be the key components in DHE's modulated biological pathways. Based on the pathway analysis as well as chemical similarity to estradiol, DHE is proposed to be a phytoestrogen. The proteomic, bioinformatic, and chemoinformatic analyses were further verified with individual cell-based experiments. Our study demonstrates a workflow for identifying the mechanisms of action of DHE through shotgun proteomic analysis.


Asunto(s)
Antineoplásicos/farmacología , Fenantrenos/farmacología , Fitoquímicos/farmacología , Factor de Transcripción Activador 2/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias/patología , Fitoestrógenos , Poaceae/química , Proteómica/métodos , Transducción de Señal/efectos de los fármacos
20.
Curr Opin Chem Biol ; 43: 30-36, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29136524

RESUMEN

Transition metal compounds are a rich source for anticancer drug development. Judicious application of coordination ligands is a critical success factor in the design of effective anti-tumor compounds. N-heterocyclic carbenes (NHC) are stable ligands that have strong donor strengths in stabilizing metal ions and versatility in structural modifications to provide diverse scaffolds for biological molecular targeting. Remarkable advances have been achieved in the development of metal NHC complexes as anticancer as well as theranostic agents. NHC complexes of gold, platinum and palladium have been designed to elicit potent cancer cell cytotoxicity, effective anti-tumor activities in animal models as well as selective binding to molecular targets (e.g. protein thiols, DNA G-quadraplexes, mismatched DNA). The mechanisms of action of some of these complexes have been elucidated.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Oro/química , Compuestos Heterocíclicos/química , Metano/análogos & derivados , Paladio/química , Platino (Metal)/química , Animales , Modelos Animales de Enfermedad , Humanos , Ligandos , Metano/química , Sondas Moleculares/química , Compuestos de Sulfhidrilo/química , Nanomedicina Teranóstica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA