Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 338
Filtrar
1.
Biochem Pharmacol ; 224: 116221, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38641308

RESUMEN

Gain-of-function mutation in the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) catalytic subunit alpha gene (PIK3CA) is a significant factor in head and neck cancer (HNC). Patients with HNC harboring PIK3CA mutations receive therapeutic benefits from the use of non-steroidal anti-inflammatory drugs (NSAIDs). However, the molecular mechanisms underlying these effects remain unknown. Here, we examined the Detroit562 and FaDu cell lines as HNC models with and without a hyperactive PIK3CA mutation (H1047R), respectively, regarding their possible distinct responses to the NSAIDs celecoxib and sulindac sulfide (SUS). Detroit562 cells exhibited relatively high PI3K/Akt pathway-dependent cyclooxygenase-2 (COX-2) expression, associated with cell proliferation. Celecoxib treatment restricted cell proliferation and upregulated endoplasmic reticulum (ER) stress-related markers, including GRP78, C/EBP-homologous protein, activating transcription factor 4, death receptor 5, and reactive oxygen species (ROS). These effects were much stronger in Detroit562 cells than in FaDu cells and were largely COX-2-independent. SUS treatment yielded similar results. Salubrinal (an ER stress inhibitor) and N-acetyl-L-cysteine (a ROS scavenger) prevented NSAID-induced ROS generation and ER stress, respectively, indicating crosstalk between ER and oxidative stress. In addition, celecoxib and/or SUS elevated cleaved caspase-3 levels, Bcl-2-associated X protein/Bcl-2-interacting mediator of cell death expression, and mitochondrial damage, which was more pronounced in Detroit562 than in FaDu cells. Salubrinal and N-acetyl-L-cysteine attenuated celecoxib-induced mitochondrial dysfunction. Collectively, our results suggest that celecoxib and SUS efficiently suppress activating PIK3CA mutation-harboring HNC progression by inducing ER and oxidative stress and mitochondrial dysfunction, leading to apoptotic cell death, further supporting NSAID treatment as a useful strategy for oncogenic PIK3CA-mutated HNC therapy.


Asunto(s)
Celecoxib , Fosfatidilinositol 3-Quinasa Clase I , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Neoplasias de Cabeza y Cuello , Mitocondrias , Especies Reactivas de Oxígeno , Sulindac , Humanos , Celecoxib/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Sulindac/farmacología , Sulindac/análogos & derivados , Especies Reactivas de Oxígeno/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Línea Celular Tumoral , Antineoplásicos/farmacología , Mutación , Proliferación Celular/efectos de los fármacos
2.
J Ocul Pharmacol Ther ; 38(1): 102-113, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34964663

RESUMEN

Purpose: Dry eye disease (DED) is classified as aqueous deficient, evaporative, or mixed. We investigated the therapeutic effect of the novel anti-inflammatory drug phosphosulindac (PS) in rabbit models of DED encompassing its pathogenesis, and its transition to chronicity. Methods: We treated three rabbit models of DED with PS (hydrogel formulation) or vehicle topically applied 1 × /day. We induced aqueous-deficient DED (acute and chronic) by injecting Concanavalin A into lacrimal glands; evaporative DED by injecting into the upper eyelid inactivated Mycobacterium tuberculosis in complete Freund's adjuvant; and mixed DED through desiccative stress, induced by holding open the eye for 3 h. We determined corneal sensitivity, tear break-up time (TBUT), Schirmer's tear test (STT), tear osmolality, and fluorescein staining of the ocular surface. Results: PS reversed all abnormal DED parameters. In acute DED, PS dose dependently normalized corneal sensitivity and tear osmolality; and improved TBUT, STT, and fluorescein staining. PS normalized corneal sensitivity and improved all other parameters in chronic aqueous-deficient DED. In evaporative DED, PS normalized corneal sensitivity and improved TBUT and fluorescein staining (osmolality and STT were not significantly changed in this model). In the desiccative stress model, PS improved TBUT and fluorescein staining but had no effect on STT or tear osmolality. Conclusions: PS rapidly reversed almost all DED parameters in its three subtypes. The normalization of the suppressed corneal sensitivity suggests the possibility of marked symptomatic relief by PS. The hydrogel formulation allows once-daily dosing. PS merits further development as a potential treatment for DED.


Asunto(s)
Antiinflamatorios/farmacología , Síndromes de Ojo Seco/patología , Compuestos Organofosforados/farmacología , Sulindac/análogos & derivados , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/efectos adversos , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Hidrogeles , Aparato Lagrimal/efectos de los fármacos , Compuestos Organofosforados/administración & dosificación , Compuestos Organofosforados/efectos adversos , Concentración Osmolar , Conejos , Sulindac/administración & dosificación , Sulindac/efectos adversos , Sulindac/farmacología , Lágrimas/efectos de los fármacos
3.
Exp Eye Res ; 213: 108827, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34742691

RESUMEN

Drug development, resource- and time-intensive, extensively employs cell-based assays to assess the efficacy and safety of candidate drugs. The widely used immortalized cell lines, experimentally convenient, have limited predictive value. In contrast, ex-vivo models more faithfully reproduce diseases but are technically challenging to establish. To address this need, we developed a simplified process for ex-vivo cell culture, demonstrating its feasibility in ocular surface cells. Conjunctival cells were harvested by impression cytology and grown on mixed cellulose ester membrane filters (MCFs). Human and rabbit conjunctival cells cultured on MCFs are 100% viable at 24 h, and 43% viable at 72 h. A gene expression study evaluating 84 genes involved in ocular inflammation demonstrated that ex-vivo culturing maintains intact the expression of two thirds of these genes in human cells. That these cells are suitable for the assessment of ocular drugs was demonstrated by studying the effect of phosphosulindac (PS), a small molecule under development for the treatment of dry eye disease, in both human and rabbit conjunctival cells. PS, for example, suppressed the expression of CXCL10, a cytokine participating in the pathogenesis of dry eye disease, in human and in rabbit conjunctival cells cultured ex-vivo by 32% and 70%, respectively. Conjunctival cells cultured ex-vivo can be transfected to evaluate mechanistic questions. We successfully transfected such cells with a plasmid expressing luciferase under the control of an IFN-γ-responsive promoter or its control plasmid. IFN-γ stimulated luciferase expression by 85% in cells with the responsive plasmid but not in controls; PS significantly suppressed this induction by 37% without affecting the control plasmid. These findings demonstrate that human and rabbit conjunctival cells cultured ex-vivo with our method are viable and maintain their biological integrity; respond to biological and pharmacological agents; and are transfectable with informative plasmids. The unique advantage of this method is to potentially accelerate the development of novel drugs for the treatment of ocular surface diseases, and to advance our understanding of ocular surface pathophysiology.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Conjuntiva/efectos de los fármacos , Evaluación de Medicamentos/métodos , Síndromes de Ojo Seco/tratamiento farmacológico , Compuestos Organofosforados/uso terapéutico , Sulindac/análogos & derivados , Adulto , Anciano , Animales , Técnicas de Cultivo de Célula , Supervivencia Celular , Celulosa/análogos & derivados , Celulosa/química , Quimiocina CXCL10/metabolismo , Conjuntiva/metabolismo , Desarrollo de Medicamentos , Femenino , Perfilación de la Expresión Génica , Humanos , Luciferasas/metabolismo , Masculino , Persona de Mediana Edad , Plásmidos , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Sulindac/uso terapéutico , Obtención de Tejidos y Órganos , Transfección
4.
Sci Rep ; 11(1): 16956, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34417523

RESUMEN

Osteoarthritis (OA), a most common and highly prevalent joint disease, is closely associated with dysregulated expression and modification of RXRα. However, the role of RXRα in the pathophysiology of OA remains unknown. The present study aimed to investigate whether RXRα modulator, such as K-80003 can treat OA. Experimental OA was induced by intra-articular injection of monosodium iodoacetate (MIA) in the knee joint of rats. Articular cartilage degeneration was assessed using Safranin-O and fast green staining. Synovial inflammation was measured using hematoxylin and eosin (H&E) staining and enzyme-linked immunosorbent assay (ELISA). Expressions of MMP-13, ADAMTS-4 and ERα in joints were analyzed by immunofluorescence staining. Western blot, RT-PCR and co-Immunoprecipitation (co-IP) were used to assess the effects of K-80003 on RXRα-ERα interaction. Retinoid X receptor α (RXRα) modulator K-80003 prevented the degeneration of articular cartilage, reduced synovial inflammation, and alleviated osteoarthritic pain in rats. Furthermore, K-80003 markedly inhibited IL-1ß-induced p65 nuclear translocation and IκBα degradation, and down-regulate the expression of HIF-2α, proteinases (MMP9, MMP13, ADAMTS-4) and pro-inflammatory factors (IL-6 and TNFα) in primary chondrocytes. Additionally, knockdown of ERα with siRNA blocked these effects of K-80003 in chondrocytes. In conclusion, RXRα modulators K-80003 suppresses inflammatory and catabolic responses in OA, suggesting that targeting RXRα-ERα interaction by RXRα modulators might be a novel therapeutic approach for OA treatment.


Asunto(s)
Inflamación/complicaciones , Inflamación/metabolismo , Osteoartritis/complicaciones , Osteoartritis/metabolismo , Receptor alfa X Retinoide/metabolismo , Sulindac/análogos & derivados , Animales , Cartílago/diagnóstico por imagen , Cartílago/patología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/patología , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/metabolismo , Células HEK293 , Humanos , Inflamación/diagnóstico por imagen , Articulaciones/efectos de los fármacos , Articulaciones/patología , Masculino , FN-kappa B/metabolismo , Osteoartritis/diagnóstico por imagen , Dolor/complicaciones , Sustancias Protectoras/farmacología , Unión Proteica/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sulindac/farmacología , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología , Sinovitis/complicaciones , Sinovitis/patología , Regulación hacia Arriba
5.
J Ocul Pharmacol Ther ; 37(6): 321-330, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34152861

RESUMEN

Purpose: Inflammation of the ocular surface is central to dry eye disease (DED). The anti-inflammatory agent phospho-sulindac (PS) at a high dose was efficacious against DED in a rabbit model. We assessed the dose, formulation and structure dependence of PS's effect. Methods: In rabbits with concanavalin A-induced DED we evaluated a range of PS concentrations (0.05%-1.6%) and dosing frequencies, assessed the duration of its effect with PS in 2 solution formulations and one emulsion formulation, and compared the efficacy of PS to that of sulindac, and of the structurally similar phospho-ibuprofen amide. We determined tear breakup time (TBUT) (tear stability), Schirmer's tear test (tear production), and by esthesiometry corneal sensitivity (symptoms). We also determined the biodistribution in the eye of topically applied PS. Results: PS in a solution formulation, given as eye drops q.i.d. was efficacious starting at a dose of 0.1%. The effect was apparent after 2 days of treatment and lasted at least 8 days after the last dose. Both signs (evidenced by TBUT and Schirmer's test) and symptoms (measured by corneal sensitivity) improved significantly. The best formulation was the solution formulation; a cyclodextrin-based formulation was also successful but the emulsion formulation was not. PS and its metabolites were essentially restricted to the anterior chamber of the eye. Sulindac and phospho-ibuprofen amide had no efficacy on DED. Conclusions: PS is efficacious against DED. Its effect, encompassing signs, and symptoms, are dose, formulation, and structure dependent. PS has therapeutic promise and merits further development.


Asunto(s)
Composición de Medicamentos , Síndromes de Ojo Seco/tratamiento farmacológico , Compuestos Organofosforados/administración & dosificación , Compuestos Organofosforados/química , Sulindac/análogos & derivados , Administración Tópica , Animales , Síndromes de Ojo Seco/metabolismo , Síndromes de Ojo Seco/patología , Masculino , Compuestos Organofosforados/farmacocinética , Conejos , Sulindac/administración & dosificación , Sulindac/química , Sulindac/farmacocinética , Distribución Tisular
6.
Molecules ; 25(17)2020 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-32842685

RESUMEN

Distant metastatic colorectal cancer (CRC) is present in approximately 25% of patients at initial diagnosis, and eventually half of CRC patients will develop metastatic disease. The 5-year survival rate for patients with metastatic CRC is a mere 12.5%; thus, there is an urgent need to investigate the molecular mechanisms of cancer progression in CRC. High expression of human high-mobility group A2 (HMGA2) is related to tumor progression, a poor prognosis, and a poor response to therapy for CRC. Therefore, HMGA2 is an attractive target for cancer therapy. In this study, we identified aspirin and sulindac sulfide as novel potential inhibitors of HMGA2 using a genome-wide mRNA signature-based approach. In addition, aspirin and sulindac sulfide induced cytotoxicity of CRC cells stably expressing HMGA2 by inhibiting cell proliferation and migration. Moreover, a gene set enrichment analysis (GSEA) revealed that gene sets related to inflammation were positively correlated with HMGA2 and that the main molecular function of these genes was categorized as a G-protein-coupled receptor (GPCR) activity event. Collectively, this is the first study to report that aspirin and sulindac sulfide are novel potential inhibitors of HMGA2, which can induce cytotoxicity of CRC cells stably expressing HMGA2 by inhibiting cell proliferation and migration through influencing inflammatory-response genes, the majority of which are involved in GPCR signaling.


Asunto(s)
Aspirina/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Citotoxinas/farmacología , Proteína HMGA2/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Sulindac/análogos & derivados , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteína HMGA2/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Sulindac/farmacología
7.
Pharm Res ; 37(3): 44, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31993760

RESUMEN

PURPOSE: This prospective study aimed to evaluate the effects of genetic polymorphisms in sulindac-related metabolizing enzyme genes including FMO3 and AOX1 on the population pharmacokinetics of sulindac in 58 pregnant women with preterm labor. METHODS: Plasma samples were collected at 1.5, 4, and 10 h after first oral administration of sulindac. Plasma concentrations of sulindac and its active metabolite (sulindac sulfide) were determined, and pharmacokinetic analysis was performed with NONMEM 7.3. RESULTS: The mean maternal and gestational ages at the time of dosing were 32.5 ± 4.4 (range, 20-41) years and 27.4 ± 4.4 (range, 16.4-33.4) weeks, respectively. In the population pharmacokinetic analysis, one depot compartment model of sulindac with absorption lag time best described the data. The metabolism of sulindac and sulindac sulfide was described using Michaelis-Menten kinetics. In stepwise modeling, gestational age impacted volume of distribution (Vc), and FMO3 rs2266782 was shown by the Michaelis constant to affect conversion of sulindac sulfide to sulindac (KM32); these were retained in the final model. CONCLUSIONS: Genetic polymorphisms of FMO3 and AOX1 could affect the pharmacokinetics of sulindac in women who undergo preterm labor. The results of this study could help clinicians develop individualized treatment plans for administering sulindac.


Asunto(s)
Aldehído Oxidasa/genética , Antiinflamatorios/farmacocinética , Trabajo de Parto Prematuro/metabolismo , Oxigenasas/genética , Polimorfismo Genético/fisiología , Sulindac/farmacocinética , Adulto , Aldehído Oxidasa/metabolismo , Femenino , Genotipo , Edad Gestacional , Humanos , Modelos Biológicos , Oxigenasas/metabolismo , Embarazo , Estudios Prospectivos , Transducción de Señal , Sulindac/análogos & derivados , Sulindac/metabolismo
8.
Drug Des Devel Ther ; 13: 4127-4134, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31827319

RESUMEN

Introduction: The ionizing radiation exposure of the normal cell causes damage to DNA, which leads to cell dysfunction or even cell death. However, it is necessary to identify new radio protectives in order to protect normal cells. Sulindac sulfide (SS) is a metabolite of sulindac (a non-steroidal anti-inflammatory drug) known as a cyclooxygenase inhibitor. Free radicals and reactive oxygen species are generated in the IR-exposed cells. Also, the induced inflammation process causes damage in DNA. PURPOSE: In this research, the radioprotective effect of SS was investigated against genotoxicity and lipid peroxidation induced by ionizing radiation in the human blood lymphocytes. METHODS: In this study, the human blood samples were pretreated with SS at different concentrations (10, 25, 50, 100 and 250 µM) and then were exposed to IR at a dose of 1.5 Gy. The micronucleus (MN) assay was used to indicate the radioprotective effects of SS on exposed cells. Total antioxidant activity of the SS was measured by using FRAP and DPPH assay. Also, the malondialdehyde (MDA) levels and the activity of superoxide dismutase (SOD) on the exposed cells were evaluated. RESULTS: It was found that SS decreased the percentage of MN induced by IR in exposed cells. Maximum reduction in the frequency of MN was observed at 250 µM of SS (87%) that provides the highest degree of protection against IR. On the other hand, pretreatment at 250 µM of SS inhibited IR-induced oxidative stress, which led to a decrease in the MN frequencies and MDA levels, while SOD activity showed an increase in the exposed cells. CONCLUSION: It could be concluded that SS as a good radioprotective agent protects the human normal cells against the oxidative stress and genetic damage induced by IR.


Asunto(s)
Antioxidantes/farmacología , Daño del ADN , Sulindac/análogos & derivados , Relación Dosis-Respuesta a Droga , Humanos , Peroxidación de Lípido/efectos de los fármacos , Linfocitos/efectos de los fármacos , Malondialdehído/análisis , Estructura Molecular , Protección Radiológica , Relación Estructura-Actividad , Sulindac/farmacología , Superóxido Dismutasa/metabolismo
9.
Br J Pharmacol ; 176(14): 2559-2572, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30943581

RESUMEN

BACKGROUND AND PURPOSE: Atherosclerosis is a chronic inflammatory disease, and retinoid X receptor-α (RXRα) is an intriguing anti-atherosclerosis target. This study investigated whether and how an RXRα modulator, K-80003, derived from a non-steroidal anti-inflammatory drug attenuates atherosclerotic plaque progression and destabilization. EXPERIMENTAL APPROACH: Our previously established ApoE-/- mouse model of carotid vulnerable plaque progression was treated with K-80003 or vehicle for 4 or 8 weeks. Samples of carotid arteries and serum were collected to determine atherosclerotic lesion size, histological features, expression of related proteins, and lipid profiles. In vitro studies were carried out in 7-ketocholesterol (7-KC)-stimulated macrophages treated with or without K-80003. KEY RESULTS: K-80003 significantly reduced lesion size, plaque rupture, macrophage infiltration, and inflammatory cytokine levels. Plaque macrophages positive for nuclear p65 (RelA) NF-κB subunit were markedly reduced after K-80003 treatment. Also, K-80003 treatment inhibited 7-KC-induced p65 nuclear translocation, IκBα degradation, and transcription of NF-κB target genes. In addition, K-80003 inhibited NF-κB pathway mainly through the reduction of p62/sequestosome 1 (SQSTM1), probably due to promotion of autophagic flux by K-80003. Mechanistically, cytoplasmic localization of RXRα was associated with decreased autophagic flux. Increasing cytoplasmic RXRα expression by overexpression of RXRα/385 mutant decreased autophagic flux in RAW264.7 cells. Finally, K-80003 strongly inhibited 7-KC-induced RXRα cytoplasmic translocation. CONCLUSIONS AND IMPLICATIONS: K-80003 suppressed atherosclerotic plaque progression and destabilization by promoting macrophage autophagic flux and consequently inhibited the p62/SQSTM1-mediated NF-κB proinflammatory pathway. Thus, targeting RXRα-mediated autophagy-inflammation axis by its noncanonical modulator may represent a promising strategy to treat atherosclerosis.


Asunto(s)
Apolipoproteínas E/metabolismo , Placa Aterosclerótica/tratamiento farmacológico , Sulindac/análogos & derivados , Animales , Apolipoproteínas E/deficiencia , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Células RAW 264.7 , Sulindac/administración & dosificación , Sulindac/efectos adversos , Sulindac/farmacología
10.
Nat Commun ; 10(1): 1463, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30931933

RESUMEN

Retinoid X receptor-alpha (RXRα) is a potent regulator of inflammatory responses; however, its therapeutic potential for inflammatory cancer remains to be explored. We previously discovered that RXRα is abnormally cleaved in tumor cells and tissues, producing a truncated RXRα (tRXRα). Here, we show that transgenic expression of tRXRα in mice accelerates the development of colitis-associated colon cancer (CAC). The tumorigenic effect of tRXRα is primarily dependent on its expression in myeloid cells, which results in interleukin-6 (IL-6) induction and STAT3 activation. Mechanistic studies reveal an extensive interaction between tRXRα and TRAF6 in the cytoplasm of macrophages, leading to TRAF6 ubiquitination and subsequent activation of the NF-κB inflammatory pathway. K-80003, a tRXRα modulator derived from nonsteroidal anti-inflammatory drug (NSAID) sulindac, suppresses the growth of tRXRα-mediated colorectal tumor by inhibiting the NF-κB-IL-6-STAT3 signaling cascade. These results provide new insight into tRXRα action and identify a promising tRXRα ligand for treating CAC.


Asunto(s)
Carcinogénesis/genética , Colitis/genética , Neoplasias Colorrectales/genética , Interleucina-6/inmunología , Receptor alfa X Retinoide/genética , Factor de Transcripción STAT3/inmunología , Animales , Carcinogénesis/inmunología , Colitis/inmunología , Colitis Ulcerosa/inmunología , Colon/efectos de los fármacos , Colon/inmunología , Colon/metabolismo , Neoplasias Colorrectales/inmunología , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Células HCT116 , Humanos , Inflamación , Macrófagos/inmunología , Ratones , FN-kappa B/inmunología , Receptor alfa X Retinoide/inmunología , Transducción de Señal , Sulindac/análogos & derivados , Sulindac/farmacología , Factor 6 Asociado a Receptor de TNF/inmunología
11.
Br J Cancer ; 120(5): 537-546, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30739913

RESUMEN

BACKGROUND: Nonsteroidal anti-inflammatory drugs (NSAIDs) have been proposed as chemopreventive agents for many tumours; however, the mechanism responsible for their anti-neoplastic activity remains elusive and the side effects due to cyclooxygenase (COX) inhibition prevent this clinical application. METHODS: Molecular biology, in silico, cellular and in vivo tools, including innovative in vivo imaging and classical biochemical assays, were applied to identify and characterise the COX-independent anti-cancer mechanism of NSAIDs. RESULTS: Here, we show that tumour-protective functions of NSAIDs and exisulind (a sulindac metabolite lacking anti-inflammatory activity) occur through a COX-independent mechanism. We demonstrate these NSAIDs counteract carcinogen-induced proliferation by inhibiting the sirtuin 1 (SIRT1) deacetylase activity, augmenting acetylation and activity of the tumour suppressor p53 and increasing the expression of the antiproliferative gene p21. These properties are shared by all NSAIDs except for ketoprofen lacking anti-cancer properties. The clinical interest of the mechanism identified is underlined by our finding that p53 is activated in mastectomy patients undergoing intraoperative ketorolac, a treatment associated with decreased relapse risk and increased survival. CONCLUSION: Our study, for the first-time, links NSAID chemopreventive activity with direct SIRT1 inhibition and activation of the p53/p21 anti-oncogenic pathway, suggesting a novel strategy for the design of tumour-protective drugs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Anticarcinógenos/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Sirtuina 1/efectos de los fármacos , Sulindac/análogos & derivados , Proteína p53 Supresora de Tumor/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Anticarcinógenos/efectos adversos , Línea Celular Tumoral , Simulación por Computador , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidores de la Ciclooxigenasa/efectos adversos , Humanos , Ketorolaco/efectos adversos , Ketorolaco/uso terapéutico , Ratones , Modelos Moleculares , Sirtuina 1/metabolismo , Sulindac/farmacología , Proteína p53 Supresora de Tumor/metabolismo
12.
Bioorg Chem ; 85: 413-419, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30665035

RESUMEN

Retinoid X receptor alpha (RXRα), a central member of the nuclear receptor superfamily and a key regulator of many signal transduction pathways, has been an attractive drug target. We previously discovered that an N-terminally truncated form of RXRα can be induced by specific ligands to form homotetramers, which, as a result of conformational selection, forms the basis for inhibiting the nongenomic activation of RXRα. Here, we report the identification and characterization of atorvastatin as a new RXRα tetramer stabilizer by using structure-based virtual screening and demonstrate that virtual library screening can be used to aid in identifying RXRα ligands that can induce its tetramerization. In this study, docking was applied to screen the FDA-approved small molecule drugs in the DrugBank 4.0 collection. Two compounds were selected and purchased for testing. We showed that the selected atorvastatin could bind to RXRα to promote RXRα-LBD tetramerization. We also showed that atorvastatin possessed RXRα-dependent apoptotic effects. In addition, we used a chemical approach to aid in the studies of the binding mode of atorvastatin.


Asunto(s)
Atorvastatina/farmacología , Multimerización de Proteína/efectos de los fármacos , Receptor alfa X Retinoide/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Atorvastatina/química , Atorvastatina/metabolismo , Sitios de Unión , Evaluación Preclínica de Medicamentos , Humanos , Ligandos , Células MCF-7 , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Estabilidad Proteica/efectos de los fármacos , Sulindac/análogos & derivados , Sulindac/metabolismo
13.
Int J Pharm ; 557: 273-279, 2019 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-30597269

RESUMEN

We studied the pharmacokinetics, biodistribution and metabolism of phospho-sulindac (PS), a novel agent efficacious in the treatment of dry eye, formulated in nanoparticles (PS-NPs) following its topical administration to the eye of New Zealand White rabbits. The nanoparticles were spherical with effective diameter = 108.9 ±â€¯41.7 nm, zeta potential = -21.70 ±â€¯3.78 mV, drug loading = 7%, and entrapment efficiency = 46.4%. Of the total PS delivered topically to the eye, >95% was retained in the anterior segment, predominantly in the cornea (Cmax = 101.3 µM; Tmax = 1 h; T1/2 = 2.6 h; area AUC0-16h = 164.4 µM·h) and conjunctiva (Cmax = 89.4 µM; Tmax = 0.25 h; T1/2 = 3.1 h; AUC0-16h = 63.5 µM·h), the tissues most affected by dry eye disease. No PS or its metabolites were detected in the systemic circulation. PS was metabolized to PS sulfide and PS sulfone; all three molecules were hydrolyzed to sulindac, which was converted to sulindac sulfide and sulindac sulfone. A solution formulation of PS provided lower PS levels in ocular tissues but higher levels of PS metabolites, compared to PS-NPs. Therefore, NPs represent an effective formulation for the topical ocular administration of PS for anterior segment diseases, such as dry eye disease.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ojo/metabolismo , Nanopartículas/administración & dosificación , Compuestos Organofosforados/administración & dosificación , Sulindac/análogos & derivados , Administración Intravenosa , Administración Tópica , Animales , Masculino , Nanopartículas/química , Compuestos Organofosforados/sangre , Compuestos Organofosforados/química , Compuestos Organofosforados/farmacocinética , Conejos , Sulindac/administración & dosificación , Sulindac/sangre , Sulindac/química , Sulindac/farmacocinética , Distribución Tisular
14.
Biochem Biophys Res Commun ; 505(4): 1203-1210, 2018 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-30327144

RESUMEN

Sulindac sulfone is a metabolite of sulindac, a non-steroidal anti-inflammatory drug (NSAID), without anti-inflammatory ability. However, sulindac sulfone has been reported to significantly reduce polyps in patients with colorectal adenomatous polyposis in clinical trials. Thus, sulindac sulfone is expected to be useful for the chemoprevention of neoplasia with few side effects related to anti-inflammatory ability. To date, the molecular targets of sulindac sulfone have not yet fully investigated. Therefore, in order to newly identify sulindac sulfone-binding proteins, we generated sulindac sulfone-fixed FG beads and purified sulindac sulfone-binding proteins from human colon cancer HT-29 cells. we identified mitochondrial outer membrane proteins voltage-dependent anion channel (VDAC) 1 and VDAC2 as novel molecular targets of sulindac sulfone, and sulindac sulfone directly bound to both VDAC1 and VDAC2. Double knockdown of VDAC1 and VDAC2 by siRNA inhibited growth and arrested the cell cycle at G1 phase in HT-29 cells. Depletion of VDAC1 and VDAC2 also inhibited the mTORC1 pathway with a reduction in cyclin D1. Interestingly, these effects were consistent with those of sulindac sulfone against human colon cancer cells, suggesting that sulindac sulfone negatively regulates the function of VDAC1 and VDAC2. In the present study, our data suggested that VDAC1 and VDAC2 are direct targets of sulindac sulfone which suppresses the mTORC1 pathway and induces G1 arrest.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Sulindac/análogos & derivados , Canal Aniónico 1 Dependiente del Voltaje/antagonistas & inhibidores , Canal Aniónico 2 Dependiente del Voltaje/antagonistas & inhibidores , Adenosina Trifosfato/biosíntesis , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Puntos de Control del Ciclo Celular , Neoplasias del Colon/patología , Células HT29 , Humanos , Sulindac/química , Sulindac/metabolismo , Sulindac/farmacología , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Canal Aniónico 2 Dependiente del Voltaje/metabolismo
15.
Future Med Chem ; 10(7): 743-753, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29671617

RESUMEN

AIM: Experimental and epidemiological studies and clinical trials suggest that nonsteroidal anti-inflammatory drugs possess antitumor potential. Sulindac, a widely used nonsteroidal anti-inflammatory drug, can prevent adenomatous colorectal polyps and colon cancer, especially in patients with familial adenomatous polyposis. Sulindac sulfide amide (SSA) is an amide-linked sulindac sulfide analog that showed in vivo antitumor activity in a human colon tumor xenograft model. Results/methodology: A new analog series with heterocyclic rings such as oxazole or thiazole at the C-2 position of sulindac was prepared and screened against prostate, colon and breast cancer cell lines to probe the effect of these novel substitutions on the activity of sulindac analogs. CONCLUSION: In general, replacement of the amide function of SSA analogs had a negative impact on the cell lines tested. A small number of hits incorporating rigid oxazole or thiazole groups in the sulindac scaffold in place of the amide linkage show comparable activity to our lead agent SSA.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/prevención & control , Oxazoles/química , Sulindac/análogos & derivados , Sulindac/uso terapéutico , Tiazoles/química , Antiinflamatorios no Esteroideos/química , Antineoplásicos/química , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Xenoinjertos , Humanos , Masculino , Espectroscopía de Protones por Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray , Sulindac/química
16.
Transl Res ; 198: 58-72, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29702077

RESUMEN

Dry eye disease (DED) currently has no satisfactory treatment partly because of the lack of informative animal models. We evaluated the anti-inflammatory phosphosulindac (PS) for the treatment of DED using a new rabbit model of DED based on the concanavalin A (Con A) acute DED model: we injected all lacrimal glands with Con A weekly under ultrasound guidance, which prolonged DED to >3 weeks, and thoroughly assessed efficacy with tear break-up time (TBUT), tear osmolarity, Schirmer test, and tear lactoferrin levels. Rabbits with DED (n = 8-10 eyes per group) were treated topically with PS or vehicle 3×/day for 21days. PS restored TBUT, tear osmolarity, and lactoferrin levels (P < 0.0001-0.04) to normal but did not significantly improve the results of the Schirmer test. PS showed no side effects and was much more efficacious than cyclosporine or lifitegrast. In the cornea, PS suppressed the activation of nuclear factor kappa-B, the levels of transforming growth factor beta, interleukin-1 beta, interleukin-6, and interleukin-8, and the levels of matrix metalloproteinase (MMP)-1 and MMP-9, and MMP activity. Levels of prostaglandin E2 (PGE2) in tears and cornea were preserved in PS-treated rabbits. Ketorolac and diclofenac, two ophthalmic NSAIDs causing corneal melt, nearly completely suppressed PGE2 levels but had no effect on MMPs. The effects of PS on PGE2 and MMPs likely account for its apparent ocular safety. Our results establish an animal model for acute and chronic DED suitable for drug efficacy studies and indicate that PS merits evaluation for DED.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Concanavalina A/toxicidad , Modelos Animales de Enfermedad , Síndromes de Ojo Seco/tratamiento farmacológico , Compuestos Organofosforados/uso terapéutico , Conejos , Sulindac/análogos & derivados , Administración Oftálmica , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Células Cultivadas , Enfermedad Crónica , Citocinas/metabolismo , Dinoprostona/metabolismo , Humanos , Lactoferrina/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Compuestos Organofosforados/administración & dosificación , Concentración Osmolar , Sulindac/administración & dosificación , Sulindac/uso terapéutico , Lágrimas/metabolismo
17.
Bioorg Med Chem Lett ; 27(20): 4614-4621, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28935266

RESUMEN

Sulindac is a non-steroidal anti-inflammatory drug (NSAID) that has shown significant anticancer activity. Sulindac sulfide amide (1) possessing greatly reduced COX-related inhibition relative to sulindac displayed in vivo antitumor activity that was comparable to sulindac in a human colon tumor xenograft model. Inspired by these observations, a panel of diverse sulindac amide derivatives have been synthesized and their activity probed against three cancer cell lines (prostate, colon and breast). A neutral analog, compound 79 was identified with comparable potency relative to lead 1 and activity against a panel of lymphoblastic leukemia cell lines. Several new series also show good activity relative to the parent (1), including five analogs that also possess nanomolar inhibitory potencies against acute lymphoblastic leukemia cells. Several new analogs identified may serve as anticancer lead candidates for further development.


Asunto(s)
Amidas/química , Antineoplásicos/química , Neoplasias/tratamiento farmacológico , Sulindac/análogos & derivados , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Relación Estructura-Actividad , Sulindac/química , Sulindac/farmacología , Sulindac/uso terapéutico
18.
Nat Commun ; 8: 16066, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28714476

RESUMEN

Retinoid X receptor-alpha (RXRα) binds to DNA either as homodimers or heterodimers, but it also forms homotetramers whose function is poorly defined. We previously discovered that an N-terminally-cleaved form of RXRα (tRXRα), produced in tumour cells, activates phosphoinositide 3-kinase (PI3K) signalling by binding to the p85α subunit of PI3K and that K-80003, an anti-cancer agent, inhibits this process. Here, we report through crystallographic and biochemical studies that K-80003 binds to and stabilizes tRXRα tetramers via a 'three-pronged' combination of canonical and non-canonical mechanisms. K-80003 binding has no effect on tetramerization of RXRα, owing to the head-tail interaction that is absent in tRXRα. We also identify an LxxLL motif in p85α, which binds to the coactivator-binding groove on tRXRα and dissociates from tRXRα upon tRXRα tetramerization. These results identify conformational selection as the mechanism for inhibiting the nongenomic action of tRXRα and provide molecular insights into the development of RXRα cancer therapeutics.


Asunto(s)
Antineoplásicos/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Receptor alfa X Retinoide/antagonistas & inhibidores , Sulindac/análogos & derivados , Células A549 , Animales , Fosfatidilinositol 3-Quinasa Clase Ia , Cristalografía por Rayos X , Células HEK293 , Células Hep G2 , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor alfa X Retinoide/metabolismo , Transducción de Señal , Sulindac/farmacología
19.
J Clin Invest ; 127(8): 3167-3176, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28737505

RESUMEN

Proinflammatory leukotrienes (LTs) are produced by 5-lipoxygenase (5-LO) aided by 5-LO-activating protein (FLAP). LT biosynthesis inhibitors are currently under clinical investigation as treatments for respiratory and cardiovascular diseases. Here, we have revealed a sex bias in the efficiency of clinically relevant LT biosynthesis inhibitors, showing that their effects are superior in females. We found that androgens cause these sex differences by impeding the LT-biosynthetic 5-LO/FLAP complex assembly. Lower doses of the FLAP inhibitor MK886 were required to reduce LTB4 levels in exudates of female versus male mice and rats. Following platelet-activating factor-induced shock, MK886 increased survival exclusively in female mice, and this effect was abolished by testosterone administration. FLAP inhibitors and the novel-type 5-LO inhibitors licofelone and sulindac sulfide exhibited higher potencies in human blood from females, and bioactive 5-LO/FLAP complexes were formed in female, but not male, human and murine leukocytes. Supplementation of female blood or leukocytes with 5α-dihydrotestosterone abolished the observed sex differences. Our data suggest that females may benefit from anti-LT therapy to a greater extent than males, prompting consideration of sex issues in LT modifier development.


Asunto(s)
Andrógenos/metabolismo , Leucotrienos/biosíntesis , Factores Sexuales , Testosterona/administración & dosificación , Proteínas Activadoras de la 5-Lipooxigenasa/metabolismo , Animales , Araquidonato 5-Lipooxigenasa/metabolismo , Dihidrotestosterona/metabolismo , Femenino , Humanos , Hidroxiurea/análogos & derivados , Hidroxiurea/farmacología , Leucocitos/metabolismo , Inhibidores de la Lipooxigenasa/farmacología , Masculino , Ratones , Pirroles/administración & dosificación , Ratas , Ratas Wistar , Sulindac/administración & dosificación , Sulindac/análogos & derivados , Testosterona/metabolismo
20.
Oncogene ; 36(32): 4597-4609, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28368394

RESUMEN

Chronic inflammation plays an important role in cancer development and progression. Cyclooxygenases-2 (COX-2) is a key enzyme in generating prostaglandins causing inflammation, is often found to be overexpressed in prostate cancer (PCa) and is correlated with PCa cell invasion and metastasis. We aim to investigate the molecular mechanism of how COX-2 promotes PCa cell invasion and metastasis and to evaluate the effect of COX-2 inhibitors in a selected model of PCa progression. Our results showed that the expression of COX-2 and Interleukin 1ß (IL-1ß) was upregulated in highly invasive PCa cells and was correlated with the activated levels of membrane-anchored serine protease matriptase. The expression levels of COX-2 were increased and were correlated with matriptase levels in PCa specimens. Moreover, results showed that COX-2 overexpression or a COX-2 product Prostaglandin E2 (PGE2) caused an increase in matriptase activation and PCa cell invasion, whereas COX-2 silencing antagonized matriptase activation and cell invasion. In addition, the inhibition of COX-2-mediated matriptase activation by Celebrex and sulindac sulfide suppressed the androgen-independent and COX2-overexpressing PCa PC-3 cell invasion, tumor growth and lung metastasis in an orthotopic xenograft model. Our results indicate that COX-2/matriptase signaling contributes to the invasion, tumor growth and metastasis of COX-2-overexpressing and androgen-independent PCa cells.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Ciclooxigenasa 2/metabolismo , Proteínas de la Membrana/biosíntesis , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Serina Endopeptidasas/biosíntesis , Animales , Celecoxib/farmacología , Celecoxib/uso terapéutico , Movimiento Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/metabolismo , Células HEK293 , Humanos , Inflamación/enzimología , Interleucina-2/metabolismo , Masculino , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Próstata/enzimología , Sulindac/análogos & derivados , Sulindac/farmacología , Sulindac/uso terapéutico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA