Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
1.
J Ocul Pharmacol Ther ; 38(1): 102-113, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34964663

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Síndromes do Olho Seco/patologia , Compostos Organofosforados/farmacologia , Sulindaco/análogos & derivados , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/efeitos adversos , Preparações de Ação Retardada , Modelos Animais de Doenças , Hidrogéis , Aparelho Lacrimal/efeitos dos fármacos , Compostos Organofosforados/administração & dosagem , Compostos Organofosforados/efeitos adversos , Concentração Osmolar , Coelhos , Sulindaco/administração & dosagem , Sulindaco/efeitos adversos , Sulindaco/farmacologia , Lágrimas/efeitos dos fármacos
2.
Exp Eye Res ; 213: 108827, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34742691

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Túnica Conjuntiva/efeitos dos fármacos , Avaliação de Medicamentos/métodos , Síndromes do Olho Seco/tratamento farmacológico , Compostos Organofosforados/uso terapêutico , Sulindaco/análogos & derivados , Adulto , Idoso , Animais , Técnicas de Cultura de Células , Sobrevivência Celular , Celulose/análogos & derivados , Celulose/química , Quimiocina CXCL10/metabolismo , Túnica Conjuntiva/metabolismo , Desenvolvimento de Medicamentos , Feminino , Perfilação da Expressão Gênica , Humanos , Luciferases/metabolismo , Masculino , Pessoa de Meia-Idade , Plasmídeos , Coelhos , Reação em Cadeia da Polimerase em Tempo Real , Sulindaco/uso terapêutico , Obtenção de Tecidos e Órgãos , Transfecção
3.
Sci Rep ; 11(1): 16956, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34417523

RESUMO

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.


Assuntos
Inflamação/complicações , Inflamação/metabolismo , Osteoartrite/complicações , Osteoartrite/metabolismo , Receptor X Retinoide alfa/metabolismo , Sulindaco/análogos & derivados , Animais , Cartilagem/diagnóstico por imagem , Cartilagem/patologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Modelos Animais de Doenças , Receptor alfa de Estrogênio/metabolismo , Células HEK293 , Humanos , Inflamação/diagnóstico por imagem , Articulações/efeitos dos fármacos , Articulações/patologia , Masculino , NF-kappa B/metabolismo , Osteoartrite/diagnóstico por imagem , Dor/complicações , Substâncias Protetoras/farmacologia , Ligação Proteica/efeitos dos fármacos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Sulindaco/farmacologia , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/patologia , Sinovite/complicações , Sinovite/patologia , Regulação para Cima
4.
J Ocul Pharmacol Ther ; 37(6): 321-330, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34152861

RESUMO

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.


Assuntos
Composição de Medicamentos , Síndromes do Olho Seco/tratamento farmacológico , Compostos Organofosforados/administração & dosagem , Compostos Organofosforados/química , Sulindaco/análogos & derivados , Administração Tópica , Animais , Síndromes do Olho Seco/metabolismo , Síndromes do Olho Seco/patologia , Masculino , Compostos Organofosforados/farmacocinética , Coelhos , Sulindaco/administração & dosagem , Sulindaco/química , Sulindaco/farmacocinética , Distribuição Tecidual
5.
Molecules ; 25(17)2020 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-32842685

RESUMO

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.


Assuntos
Aspirina/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Citotoxinas/farmacologia , Proteína HMGA2/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Sulindaco/análogos & derivados , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proteína HMGA2/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo , Sulindaco/farmacologia
6.
Pharm Res ; 37(3): 44, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31993760

RESUMO

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.


Assuntos
Aldeído Oxidase/genética , Anti-Inflamatórios/farmacocinética , Trabalho de Parto Prematuro/metabolismo , Oxigenases/genética , Polimorfismo Genético/fisiologia , Sulindaco/farmacocinética , Adulto , Aldeído Oxidase/metabolismo , Feminino , Genótipo , Idade Gestacional , Humanos , Modelos Biológicos , Oxigenases/metabolismo , Gravidez , Estudos Prospectivos , Transdução de Sinais , Sulindaco/análogos & derivados , Sulindaco/metabolismo
7.
Drug Des Devel Ther ; 13: 4127-4134, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31827319

RESUMO

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.


Assuntos
Antioxidantes/farmacologia , Dano ao DNA , Sulindaco/análogos & derivados , Relação Dose-Resposta a Droga , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Malondialdeído/análise , Estrutura Molecular , Proteção Radiológica , Relação Estrutura-Atividade , Sulindaco/farmacologia , Superóxido Dismutase/metabolismo
8.
Nat Commun ; 10(1): 1463, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30931933

RESUMO

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.


Assuntos
Carcinogênese/genética , Colite/genética , Neoplasias Colorretais/genética , Interleucina-6/imunologia , Receptor X Retinoide alfa/genética , Fator de Transcrição STAT3/imunologia , Animais , Carcinogênese/imunologia , Colite/imunologia , Colite Ulcerativa/imunologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/metabolismo , Neoplasias Colorretais/imunologia , Meios de Cultivo Condicionados , Modelos Animais de Doenças , Células HCT116 , Humanos , Inflamação , Macrófagos/imunologia , Camundongos , NF-kappa B/imunologia , Receptor X Retinoide alfa/imunologia , Transdução de Sinais , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Fator 6 Associado a Receptor de TNF/imunologia
9.
Br J Pharmacol ; 176(14): 2559-2572, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30943581

RESUMO

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.


Assuntos
Apolipoproteínas E/metabolismo , Placa Aterosclerótica/tratamento farmacológico , Sulindaco/análogos & derivados , Animais , Apolipoproteínas E/deficiência , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Células RAW 264.7 , Sulindaco/administração & dosagem , Sulindaco/efeitos adversos , Sulindaco/farmacologia
10.
Br J Cancer ; 120(5): 537-546, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30739913

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Anticarcinógenos/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Sirtuína 1/efeitos dos fármacos , Sulindaco/análogos & derivados , Proteína Supressora de Tumor p53/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anticarcinógenos/efeitos adversos , Linhagem Celular Tumoral , Simulação por Computador , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidores de Ciclo-Oxigenase/efeitos adversos , Humanos , Cetorolaco/efeitos adversos , Cetorolaco/uso terapêutico , Camundongos , Modelos Moleculares , Sirtuína 1/metabolismo , Sulindaco/farmacologia , Proteína Supressora de Tumor p53/metabolismo
11.
Bioorg Chem ; 85: 413-419, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30665035

RESUMO

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.


Assuntos
Atorvastatina/farmacologia , Multimerização Proteica/efeitos dos fármacos , Receptor X Retinoide alfa/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Atorvastatina/química , Atorvastatina/metabolismo , Sítios de Ligação , Avaliação Pré-Clínica de Medicamentos , Humanos , Ligantes , Células MCF-7 , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Estabilidade Proteica/efeitos dos fármacos , Sulindaco/análogos & derivados , Sulindaco/metabolismo
12.
Int J Pharm ; 557: 273-279, 2019 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-30597269

RESUMO

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.


Assuntos
Sistemas de Liberação de Medicamentos , Olho/metabolismo , Nanopartículas/administração & dosagem , Compostos Organofosforados/administração & dosagem , Sulindaco/análogos & derivados , Administração Intravenosa , Administração Tópica , Animais , Masculino , Nanopartículas/química , Compostos Organofosforados/sangue , Compostos Organofosforados/química , Compostos Organofosforados/farmacocinética , Coelhos , Sulindaco/administração & dosagem , Sulindaco/sangue , Sulindaco/química , Sulindaco/farmacocinética , Distribuição Tecidual
13.
Biochem Biophys Res Commun ; 505(4): 1203-1210, 2018 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-30327144

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/farmacologia , Neoplasias do Colo/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Sulindaco/análogos & derivados , Canal de Ânion 1 Dependente de Voltagem/antagonistas & inibidores , Canal de Ânion 2 Dependente de Voltagem/antagonistas & inibidores , Trifosfato de Adenosina/biossíntese , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Pontos de Checagem do Ciclo Celular , Neoplasias do Colo/patologia , Células HT29 , Humanos , Sulindaco/química , Sulindaco/metabolismo , Sulindaco/farmacologia , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Canal de Ânion 2 Dependente de Voltagem/metabolismo
14.
Future Med Chem ; 10(7): 743-753, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29671617

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/prevenção & controle , Oxazóis/química , Sulindaco/análogos & derivados , Sulindaco/uso terapêutico , Tiazóis/química , Anti-Inflamatórios não Esteroides/química , Antineoplásicos/química , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Delgada , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Xenoenxertos , Humanos , Masculino , Espectroscopia de Prótons por Ressonância Magnética , Espectrometria de Massas por Ionização por Electrospray , Sulindaco/química
15.
Transl Res ; 198: 58-72, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29702077

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Concanavalina A/toxicidade , Modelos Animais de Doenças , Síndromes do Olho Seco/tratamento farmacológico , Compostos Organofosforados/uso terapêutico , Coelhos , Sulindaco/análogos & derivados , Administração Oftálmica , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Células Cultivadas , Doença Crônica , Citocinas/metabolismo , Dinoprostona/metabolismo , Humanos , Lactoferrina/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Compostos Organofosforados/administração & dosagem , Concentração Osmolar , Sulindaco/administração & dosagem , Sulindaco/uso terapêutico , Lágrimas/metabolismo
16.
Bioorg Med Chem Lett ; 27(20): 4614-4621, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28935266

RESUMO

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.


Assuntos
Amidas/química , Antineoplásicos/química , Neoplasias/tratamento farmacológico , Sulindaco/análogos & derivados , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Relação Estrutura-Atividade , Sulindaco/química , Sulindaco/farmacologia , Sulindaco/uso terapêutico
17.
J Clin Invest ; 127(8): 3167-3176, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28737505

RESUMO

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.


Assuntos
Androgênios/metabolismo , Leucotrienos/biossíntese , Fatores Sexuais , Testosterona/administração & dosagem , Proteínas Ativadoras de 5-Lipoxigenase/metabolismo , Animais , Araquidonato 5-Lipoxigenase/metabolismo , Di-Hidrotestosterona/metabolismo , Feminino , Humanos , Hidroxiureia/análogos & derivados , Hidroxiureia/farmacologia , Leucócitos/metabolismo , Inibidores de Lipoxigenase/farmacologia , Masculino , Camundongos , Pirróis/administração & dosagem , Ratos , Ratos Wistar , Sulindaco/administração & dosagem , Sulindaco/análogos & derivados , Testosterona/metabolismo
18.
Nat Commun ; 8: 16066, 2017 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-28714476

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Receptor X Retinoide alfa/antagonistas & inibidores , Sulindaco/análogos & derivados , Células A549 , Animais , Classe Ia de Fosfatidilinositol 3-Quinase , Cristalografia por Raios X , Células HEK293 , Células Hep G2 , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/metabolismo , Receptor X Retinoide alfa/metabolismo , Transdução de Sinais , Sulindaco/farmacologia
19.
Oncogene ; 36(32): 4597-4609, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28368394

RESUMO

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.


Assuntos
Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Ciclo-Oxigenase 2/metabolismo , Proteínas de Membrana/biossíntese , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Serina Endopeptidases/biossíntese , Animais , Celecoxib/farmacologia , Celecoxib/uso terapêutico , Movimento Celular/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Células HEK293 , Humanos , Inflamação/enzimologia , Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias da Próstata/enzimologia , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Sulindaco/uso terapêutico , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Biomed Res Int ; 2017: 4189678, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28331852

RESUMO

Sulindac is a nonsteroidal anti-inflammatory drug, which is clinically used for the ailments of various inflammations. This study investigated the allele frequencies of FMO3 E158K and E308G and evaluated the influences of these two genetic polymorphisms on the pharmacokinetics of sulindac and its metabolites in Chinese healthy male volunteers. Eight FMO3 wild-type (FMO3 HHDD) subjects and seven FMO3 homozygotes E158K and E308G mutant (FMO3 hhdd) subjects were recruited from 247 healthy male volunteers genotyped by PCR-RFLP method. The plasma concentrations of sulindac, sulindac sulfide, and sulindac sulfone were determined by UPLC, while the pharmacokinetic parameters of the two different FMO3 genotypes were compared with each other. The frequencies of FMO3 E158K and E308G were 20.3% and 20.1%, respectively, which were in line with Hardy-Weinberg equilibrium (D' = 0.977, r2 = 0.944). The mean values of Cmax, AUC0-24, and AUC0-∞ of sulindac were significantly higher in FMO3 hhdd group than those of FMO3 HHDD group (P < 0.05), while the pharmacokinetic parameters except Tmax of sulindac sulfide and sulindac sulfone showed no statistical difference between the two groups. The two FMO3 mutants were in close linkage disequilibrium and might play an important role in the pharmacokinetics of sulindac in Chinese healthy male volunteers.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Inflamação/tratamento farmacológico , Oxigenases/genética , Sulindaco/administração & dosagem , Adulto , Anti-Inflamatórios não Esteroides/farmacocinética , Frequência do Gene , Genótipo , Voluntários Saudáveis , Humanos , Inflamação/genética , Desequilíbrio de Ligação , Masculino , Polimorfismo de Fragmento de Restrição , Sulindaco/análogos & derivados , Sulindaco/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...