Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Braz. J. Pharm. Sci. (Online) ; 58: e20041, 2022. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1403703

RESUMO

Abstract This study aimed to develop promising and innovative mucoadhesive gel systems containing dexamethasone-loaded nanoparticle to increase the effectiveness of treatment for oral precancerous lesions and to reduce side effects. In this respect, a dexamethasone-loaded nanoparticle formulation was prepared by using emulsification/solvent evaporation method. The nanoparticle has high zeta potential (-10.3±0.5 mV), low particle size (218.42±2.1), low polydispersity index (0.070±0.014) and high encapsulation efficiency (95.018±2.982%). To improve the mucosal retention time, the dexamethasone-loaded nanoparticle was dispersed in mucoadhesive gel using gellan gum. The developed gels offered appropriate pH value, high drug content, suitable mechanical and mucoadhesive performance and appropriate viscosity for mucosal administration. All formulations exhibited plastic flow and typical gel-type mechanical spectra after the determined frequency value. The developed formulations exhibited extended drug release as intended for these systems. Cytotoxicity was tested by MTT assay in human epithelioid carcinoma cell (HeLa) in vitro. The MTT assay showed that the blank formulations were non-toxic to cells. It was observed that the bioactivity of the free dexamethasone was potentiated by mucoadhesive gels containing dexamethasone-loaded nanoparticle in HeLa cells. Results from this study indicate that mucoadhesive gels are effective for the local treatment of precancerous lesions. Our findings showed that the developed formulations were worthy of further studies.


Assuntos
Dexametasona/agonistas , Neoplasias Bucais/prevenção & controle , Administração Bucal , Géis/efeitos adversos , Antissépticos Bucais/análise , Técnicas In Vitro/métodos , Preparações Farmacêuticas/administração & dosagem , Carcinoma/classificação , Nanopartículas/classificação , Administração através da Mucosa , Liberação Controlada de Fármacos , Concentração de Íons de Hidrogênio
2.
Mol Med Rep ; 18(1): 421-428, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29749488

RESUMO

The long­term survival rate in paediatric acute lymphoblastic leukaemia (ALL) exceeds 80%; however, the outcome of adult ALL remains to be poor. Glucocorticoids (GCs) are the preferred drugs in the traditional treatment of ALL patients. In the anti­leukaemia molecular mechanisms of GCs, c­Myc inhibition serves a critical role. In the present study, a c­Myc inhibitor that increased the sensitivity to GCs in NALM6 cells of the B­cell­ALL cell line and CEM cells of the T­cell­ALL cell line was investigated. The data demonstrated that 10058­F4, a c­Myc inhibitor, increased the growth inhibition, G0/G1 phase arrest and apoptosis of the NALM6 and CEM cells as induced by dexamethasone (DXM), a type of GC. Additionally, 10058­F4 reinforced the decreased expressions of c­Myc, cyclin­dependent kinase (CDK)­4 and CDK6 in the NALM6 and CEM cells treated with DXM. These findings indicated that DXM in combination with the c­Myc inhibitor 10058­F4 may be a novel, potent therapeutic strategy for the treatment of ALL.


Assuntos
Dexametasona/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Tiazóis/farmacologia , Linhagem Celular Tumoral , Quinase 4 Dependente de Ciclina/biossíntese , Quinase 4 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/biossíntese , Quinase 6 Dependente de Ciclina/genética , Dexametasona/agonistas , Sinergismo Farmacológico , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Tiazóis/agonistas
3.
J Biol Chem ; 289(14): 10235-51, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24558046

RESUMO

Cross-talk between the glucocorticoid receptor (GR) and other receptors is emerging as a mechanism for fine-tuning cellular responses. We have previously shown that gonadotropin-releasing hormone (GnRH) ligand-independently activates the GR and synergistically modulates glucocorticoid-induced transcription of an endogenous gene in LßT2 pituitary gonadotrope precursor cells. Here, we investigated GR and GnRH receptor (GnRHR) cross-talk that involves co-localization with lipid rafts in LßT2 cells. We report that the GnRHR and a small population of the GR co-localize with the lipid raft protein flotillin-1 (Flot-1) at the plasma membrane and that the GR is present in a complex with Flot-1, independent of the presence of ligands. We found that the SGK-1 gene is up-regulated by Dex and GnRH alone, whereas a combination of both ligands resulted in a synergistic increase in SGK-1 mRNA levels. Using siRNA-mediated knockdown and antagonist strategies, we show that the gene-specific synergistic transcriptional response requires the GR, GnRHR, and Flot-1 as well as the protein kinase C pathway. Interestingly, although several GR cofactors are differentially recruited to the SGK-1 promoter in the presence of Dex and GnRH, GR levels remain unchanged compared with Dex treatment alone, suggesting that lipid raft association of the GR has a role in enhancing its transcriptional output in the nucleus. Finally, we show that Dex plus GnRH synergistically inhibit cell proliferation in a manner dependent on SGK-1 and Flot-1. Collectively the results support a mechanism whereby GR and GnRHR cross-talk within Flot-1-containing lipid rafts modulates cell proliferation via PKC activation and SGK-1 up-regulation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Microdomínios da Membrana/enzimologia , Proteína Quinase C/biossíntese , Animais , Células COS , Chlorocebus aethiops , Dexametasona/agonistas , Sinergismo Farmacológico , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Glucocorticoides/agonistas , Hormônio Liberador de Gonadotropina/agonistas , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Microdomínios da Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
4.
J Biol Chem ; 286(42): 36228-37, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21868377

RESUMO

Calcitriol, the active form of vitamin D, in combination with the glucocorticoid dexamethasone (Dex) has been shown to increase the antitumor effects of calcitriol in squamous cell carcinoma. In this study we found that pretreatment with Dex potentiates calcitriol effects by inhibiting cell growth and increasing vitamin D receptor (VDR) and VDR-mediated transcription. Treatment with actinomycin D inhibits Vdr mRNA synthesis, indicating that Dex regulates VDR expression at transcriptional level. Real time PCR shows that treatment with Dex increases Vdr transcripts in a time- and a dose-dependent manner, indicating that Dex directly regulates expression of Vdr. RU486, an inhibitor of glucocorticoids, inhibits Dex-induced Vdr expression. In addition, the silencing of glucocorticoid receptor (GR) abolishes the induction of Vdr by Dex, indicating that Dex increases Vdr transcripts in a GR-dependent manner. A fragment located 5.2 kb upstream of Vdr transcription start site containing two putative glucocorticoid response elements (GREs) was evaluated using a luciferase-based reporter assay. Treatment with 100 nm Dex induces transcription of luciferase driven by the fragment. Deletion of the GRE distal to transcription start site was sufficient to abolish Dex induction of luciferase. Also, chromatin immunoprecipitation reveals recruitment of GR to distal GRE with Dex treatment. We conclude that Dex increases VDR and vitamin D effects by increasing Vdr de novo transcription in a GR-dependent manner.


Assuntos
Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Di-Hidroxicolecalciferóis/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores de Calcitriol/biossíntese , Transcrição Gênica/efeitos dos fármacos , Animais , Anti-Inflamatórios/agonistas , Anti-Inflamatórios/antagonistas & inibidores , Sequência de Bases , Linhagem Celular , Dactinomicina/farmacologia , Dexametasona/agonistas , Dexametasona/antagonistas & inibidores , Di-Hidroxicolecalciferóis/agonistas , Antagonismo de Drogas , Sinergismo Farmacológico , Regulação da Expressão Gênica/fisiologia , Antagonistas de Hormônios/farmacologia , Camundongos , Mifepristona/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Receptores de Calcitriol/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta/fisiologia , Deleção de Sequência , Transcrição Gênica/fisiologia
5.
Patol Fiziol Eksp Ter ; (1): 10-3, 2010.
Artigo em Russo | MEDLINE | ID: mdl-20731121

RESUMO

It was established that in a mononuclear fraction polyoxidonium inhibited the TNF-alpha production and stimulated the IL-1 beta and IL-6 production. In LPS-induced mononuclear cultures polyoxidonium inhibited the IL-6 production and had no statistically significant effect on the synthesis of TNF-alpha and IL-1 beta. Polyoxidonium had no effect on TNF-alpha, IL-6 and IL-1 beta production by purified monocytes. The addition of polyoxidonium with dexamethasone to the cultures only in monocyte fraction enhanced the IL-1 beta production as compared with the effect of dexamethasone alone. Data obtained allow suggesting that under certain conditions polyoxidonium could alleviate pronounced suppressive influence of glucocorticoids on a secretory activity of effectors of innate immunity.


Assuntos
Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Monócitos/metabolismo , Piperazinas/farmacologia , Polímeros/farmacologia , Fator de Necrose Tumoral alfa/biossíntese , Adulto , Anti-Inflamatórios/agonistas , Dexametasona/agonistas , Sinergismo Farmacológico , Feminino , Humanos , Masculino , Piperazinas/agonistas
6.
J Appl Toxicol ; 30(8): 769-74, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20669259

RESUMO

Bisphenol-A (BPA) is a primary monomer in polycarbonate plastics and epoxy resins. BPA may be released into the environment following its formation via hydrolysis of ester bonds of the polymers. It has been detected in human plasma, placenta, amniotic fluid, amniotic chord, urine and saliva. BPA disrupts normal cell function by acting as an estrogen agonist as well as an androgen antagonist. The present study was carried out to investigate whether BPA can bind to human glucocorticoid receptor (GR) and elucidate its mode of interaction. BPA has been successfully docked in silico into the ligand binding site of GR using the program Discovery Studio 2.0. The structure has been compared with other agonist and antagonist bound structures of GR. It is found that the mode of interactions and binding energy of BPA were similar to that of DEXA and cortisol, two known agonists of GR. This reveals that BPA can bind to GR as an agonist. Hence, BPA may produce biological effects similar to that produced by glucocorticoids.


Assuntos
Disruptores Endócrinos/agonistas , Fenóis/agonistas , Receptores de Glucocorticoides/metabolismo , Compostos Benzidrílicos , Ligação Competitiva , Dexametasona/agonistas , Dexametasona/farmacologia , Disruptores Endócrinos/efeitos adversos , Glucocorticoides/metabolismo , Humanos , Hidrocortisona/agonistas , Hidrocortisona/farmacologia , Modelos Moleculares , Fenóis/efeitos adversos
7.
Oncol Rep ; 11(5): 1053-8, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15069546

RESUMO

To look for new candidates for agents to use in maintenance therapy for myeloma patients, the growth inhibitory effects of a 3-hydroxy-3-mehtylglutaryl coenzyme A (HMG-CoA) reductase inhibitor (statin), simvastatin, was analyzed using human myeloma cell lines. Several investigations have indicated growth reduction in certain lineages of cancer cells including one report on myeloma, and inhibitory effects of statins on GTPases and involving MAP-kinases. Most (12 out of 13) myeloma lines examined showed growth inhibition when cultured with various concentrations (1-30 microM) of simvastatin in a dose-dependent manner. Simvastatin in combination with other biological response modifiers such as ATRA or DEX had additional effects on growth. In addition, anti-oxides prevented the simvastatin-induced growth inhibition and apoptosis. Furthermore, myeloma cells treated with simvastatin clearly showed inactivation of various MAP-kinase pathways such as ERK1/2, MEK1/2, JNK, and p38. Based on these findings, statins may be suitable for clinical usage in maintenance therapy for myeloma patients.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mieloma Múltiplo/patologia , Sinvastatina/farmacologia , Antioxidantes/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dexametasona/agonistas , Dexametasona/farmacologia , Humanos , Interferon-alfa/agonistas , Interferon-alfa/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , MAP Quinase Quinase 4 , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Sinvastatina/uso terapêutico , Tretinoína/agonistas , Tretinoína/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
J Interferon Cytokine Res ; 21(3): 133-5, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11331035

RESUMO

We studied the effect of recombinant interferon-beta1b (IFN-beta1b) on the sensitivity to glucocorticoids (GC) and on the number of GC receptors (GCR) in the human monocytic cell line THP-1. We found that IFN-beta1b augments the suppressive effect that dexamethasone has on the stimulated production of tumor necrosis factor-alpha (TNF-alpha), most likely related to the increased number of GCR observed after exposure to IFN-beta1b. This provides a possible clue to the mechanism of action of IFN-beta in multiple sclerosis.


Assuntos
Glucocorticoides/agonistas , Glucocorticoides/farmacologia , Interferon beta/farmacologia , Receptores de Glucocorticoides/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Sítios de Ligação , Linhagem Celular , Dexametasona/agonistas , Dexametasona/metabolismo , Dexametasona/farmacologia , Glucocorticoides/metabolismo , Humanos , Interferon beta-1a , Interferon beta-1b , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Esclerose Múltipla/metabolismo , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA