Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Chembiochem ; 25(2): e202300603, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-37934785

RESUMO

Mitochondrion has appeared as one of the important targets for anti-cancer therapy. Subsequently, small molecule anti-cancer drugs are directed to the mitochondria for improved therapeutic efficacy. However, simultaneous imaging and impairing mitochondria by a single probe remained a major challenge. To address this, herein Chimeric Small Molecules (CSMs) encompassing drugs, fluorophore and mitochondria homing moiety were designed and synthesized through a concise strategy. Screening of the CSMs in a panel of cancer cell lines (HeLa, MCF7, A549, and HCT-116) revealed that one of the CSMs comprising Indomethacin V exhibited remarkable cervical cancer cell (HeLa) killing (IC50 =0.97 µM). This lead CSM homed into the mitochondria of HeLa cells within 1 h followed by mitochondrial damage and reactive oxygen species (ROS) generation. This novel Indomethacin V-based CSM-mediated mitochondrial damage induced programmed cell death (apoptosis). We anticipate these CSMs can be used as tools to understand the drug effects in organelle chemical biology in diseased states.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Células HeLa , Antineoplásicos/química , Mitocôndrias/metabolismo , Indometacina/metabolismo , Indometacina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Linhagem Celular Tumoral , Neoplasias/metabolismo
2.
Eur J Pharm Biopharm ; 191: 114-123, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37652137

RESUMO

The potentiation of pharmacological effects can be achieved through several strategies, such as the association of substances and delivery in nanostructured systems. In practice, potentiation can be measured by the law of mass action and joint evaluation of the combination index (CI) and dose-response curves. In this context, this study aimed to evaluate the anti-inflammatory effect of the association of ß-caryophyllene and indomethacin in the free form and delivered in nanoemulsions using the in vitro model of LPS-stimulated murine macrophage. The results indicated potentiation of the anti-inflammatory effect of nanoemulsified substances compared to free substances, as well as synergistic action between the sesquiterpene and the selected NSAID. In comparison, the association of ß-caryophyllene and indomethacin in the free form inhibited the production of nitric oxide by 50% at 48.60 µg/mL (CI = 0.21), while the nanoemulsified association of these substances resulted in an IC50 of 1.45 µg/mL (CI = 0.14). In parallel, cytotoxicity assays on HaCaT and MRC-5 cell lines demonstrated the safety of IC50-equivalent concentrations of the anti-inflammatory action, and no irritating effects on the chorioallantoic membrane of embryonated eggs were observed (HET-CAM assay). The results suggest that ß-caryophyllene may be an alternative to replace an inert oily core in nanoemulsion systems when anti-inflammatory effects are desirable.


Assuntos
Indometacina , Lipopolissacarídeos , Camundongos , Animais , Indometacina/farmacologia , Indometacina/metabolismo , Lipopolissacarídeos/farmacologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/metabolismo , Macrófagos
3.
Redox Biol ; 58: 102529, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36375380

RESUMO

Antitumor immune polarization is a key predictor of clinical outcomes to cancer therapy. An emerging concept influencing clinical outcome involves the spatial location of CD8+ T cells, within the tumor. Our earlier work demonstrated immunosuppressive effects of NOS2 and COX2 tumor expression. Here, we show that NOS2/COX2 levels influence both the polarization and spatial location of lymphoid cells including CD8+ T cells. Importantly, elevated tumor NOS2/COX2 correlated with exclusion of CD8+ T cells from the tumor epithelium. In contrast, tumors expressing low NOS2/COX2 had increased CD8+ T cell penetration into the tumor epithelium. Consistent with a causative relationship between these observations, pharmacological inhibition of COX2 with indomethacin dramatically reduced tumor growth of the 4T1 model of TNBC in both WT and Nos2- mice. This regimen led to complete tumor regression in ∼20-25% of tumor-bearing Nos2- mice, and these animals were resistant to tumor rechallenge. Th1 cytokines were elevated in the blood of treated mice and intratumoral CD4+ and CD8+ T cells were higher in mice that received indomethacin when compared to control untreated mice. Multiplex immunofluorescence imaging confirmed our phenotyping results and demonstrated that targeted Nos2/Cox2 blockade improved CD8+ T cell penetration into the 4T1 tumor core. These findings are consistent with our observations in low NOS2/COX2 expressing breast tumors proving that COX2 activity is responsible for limiting the spatial distribution of effector T cells in TNBC. Together these results suggest that clinically available NSAID's may provide a cost-effective, novel immunotherapeutic approach for treatment of aggressive tumors including triple negative breast cancer.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Camundongos , Animais , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Orientação Espacial , Imunoterapia , Progressão da Doença , Linfócitos/metabolismo , Indometacina/farmacologia , Indometacina/metabolismo , Indometacina/uso terapêutico
4.
Platelets ; 33(8): 1293-1300, 2022 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-35535424

RESUMO

Src tyrosine kinases and spleen tyrosine kinase (Syk) have recently been shown to contribute to sustained platelet aggregation on collagen under arterial shear. In the present study, we have investigated whether Src and Syk are required for aggregation under minimal shear following activation of glycoprotein VI (GPVI) and have extended this to C-type lectin-like receptor-2 (CLEC-2) which signals through the same pathway. Aggregation was induced by the GPVI ligand collagen-related peptide (CRP) and the CLEC-2 ligand rhodocytin and monitored by light transmission aggregometry (LTA). Aggregation and tyrosine phosphorylation by both receptors were sustained for up to 50 min. The addition of inhibitors of Src, Syk or Bruton's tyrosine kinase (Btk) at 150 sec, by which time aggregation was maximal, induced rapid loss of tyrosine phosphorylation of their downstream proteins, but only Src kinase inhibition caused a weak (~10%) reversal in light transmission. A similar effect was observed when the inhibitors were combined with apyrase and indomethacin or glycoprotein IIb-IIIa (GPIIb-IIIa) antagonist, eptifibatide. On the other hand, activation of GPIIb-IIIa by GPVI in a diluted platelet suspension, as measured by binding of fluorescein isothiocyanate-labeled antibody specific for the activated GPIIb-IIIa (FITC-PAC1), was reversed on the addition of Src and Syk inhibitors showing that integrin activation is rapidly reversible in the absence of outside-in signals. The results demonstrate that Src but not Syk and Btk contribute to sustained aggregation as monitored by LTA, possibly as a result of inhibition of outside-in signaling from GPIIb-IIIa to the cytoskeleton through a Syk-independent pathway. This is in contrast to the role of Syk in supporting sustained aggregation on collagen under arterial shear.


Assuntos
Agregação Plaquetária , Glicoproteínas da Membrana de Plaquetas , Tirosina Quinase da Agamaglobulinemia/metabolismo , Apirase/farmacologia , Plaquetas/metabolismo , Colágeno/farmacologia , Eptifibatida/farmacologia , Fluoresceína-5-Isotiocianato/metabolismo , Fluoresceína-5-Isotiocianato/farmacologia , Humanos , Indometacina/metabolismo , Indometacina/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Lectinas Tipo C/metabolismo , Ligantes , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Proteínas Tirosina Quinases , Quinase Syk/metabolismo , Tirosina/metabolismo , Tirosina/farmacologia , Quinases da Família src/metabolismo
5.
Bioorg Med Chem ; 64: 116759, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35468536

RESUMO

Mitochondrion emerged as an important therapeutic target for anti-cancer strategy due to its involvement in cancer progression and development. However, progress of novel small molecules for selective targeting of mitochondria in cancer cells remained a major challenge. To address this, herein, through a concise synthetic strategy, we have synthesized a small molecule library of indomethacin and ibuprofen (non-steroidal anti-inflammatory drugs, NSAIDs) derivatives having triarylphosphonium moiety for mitochondria localization. Two of the library members were identified to induce mitochondrial damage through outer membrane permeabilization (MOMP) followed by generation of reactive oxygen species (ROS) leading to the remarkable MCF7 breast cancer cell death through apoptosis. These novel mitochondria targeted NSAID derivatives could open a new direction in understanding mitochondrial biology towards anti-cancer therapeutics in future.


Assuntos
Anti-Inflamatórios não Esteroides , Neoplasias , Anti-Inflamatórios não Esteroides/farmacologia , Apoptose , Ibuprofeno/metabolismo , Ibuprofeno/farmacologia , Indometacina/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
6.
Int J Mol Sci ; 21(11)2020 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-32471121

RESUMO

(1) Background: Compounds with multitarget activity are of interest in basic research to explore molecular foundations of promiscuous binding and in drug discovery as agents eliciting polypharmacological effects. Our study has aimed to systematically identify compounds that form complexes with proteins from distinct classes and compare their bioactive conformations and molecular properties. (2) Methods: A large-scale computational investigation was carried out that combined the analysis of complex X-ray structures, ligand binding modes, compound activity data, and various molecular properties. (3) Results: A total of 515 ligands with multitarget activity were identified that included 70 organic compounds binding to proteins from different classes. These multiclass ligands (MCLs) were often flexible and surprisingly hydrophilic. Moreover, they displayed a wide spectrum of binding modes. In different target structure environments, binding shapes of MCLs were often similar, but also distinct. (4) Conclusions: Combined structural and activity data analysis identified compounds with activity against proteins with distinct structures and functions. MCLs were found to have greatly varying shape similarity when binding to different protein classes. Hence, there were no apparent canonical binding shapes indicating multitarget activity. Rather, conformational versatility characterized MCL binding.


Assuntos
Quimioinformática , Proteínas/química , Proteínas/metabolismo , Cristalografia por Raios X , Ligação de Hidrogênio , Indometacina/química , Indometacina/metabolismo , Canamicina/química , Canamicina/metabolismo , Ligantes , Lipídeos/química , Ligação Proteica
7.
Prep Biochem Biotechnol ; 50(6): 592-597, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32003284

RESUMO

The laccase (Lac), manganese peroxidases (MnP), and lignin peroxidase enzymes produced by basidiomycete have been studied due to their potential in bioremediation, therefore, in this study, degradation of diclofenac (DCF), sulfamethoxazole (SMX), indomethacin (IND), gemfibrozil (GFB), and bezafibrate (BZF) by enzymes produced by Trametes maxima, Pleurotus sp., and Pycnosporus sanguineus grown in culture was evaluated. The degradation of drugs can mainly be attributed to MnP because a correlation between the activity of this enzyme and the degree of removal was found. The specific activity of Lac did not show correlation with drug removal, while lignin peroxidase was not expressed. Trametes maxima showed the highest specific activity of MnP (387.6 ± 67.4 U/mg) and efficiency removal 90.2% of DCF, 72.62% of SMX, 60.76% of IND, 43.39% of GFB, and 32.59% of BZF) followed by Pleurotus sp. with specific activity of MnP of 55.9 ± 8.5 U/mg and 89.47% of DCF, 47.61% of GFB and 73% of IND were removed, P. sanguineus had the lowest specific activity of 18 ± 1.3 U/mg and was able to remove only 42% of SMX and 10.59% of IND. In order to prove that MnP remove drugs instead of Lac, the pure Lac was tested and only degraded DCF.


Assuntos
Bezafibrato/metabolismo , Diclofenaco/metabolismo , Genfibrozila/metabolismo , Indometacina/metabolismo , Lacase/metabolismo , Peroxidases/metabolismo , Pleurotus/enzimologia , Polyporaceae/enzimologia , Sulfametoxazol/metabolismo , Biodegradação Ambiental , Fermentação , Lignina/metabolismo
8.
Cancer Lett ; 457: 86-97, 2019 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-31100409

RESUMO

Receptor tyrosine kinase (RTK)-mediated hyperactivation of the MAPK/Erk pathway is responsible for a large number of pathogenic outcomes including many cancers. Considerable effort has been directed at targeting this pathway with varying degrees of long term therapeutic success. Under non-stimulated conditions Erk is bound to the adaptor protein Shc preventing aberrant signalling by sequestering Erk from activation by Mek. Activated RTK recruits Shc, via its phosphotyrosine binding (PTB) domain (ShcPTB), precipitating the release of Erk to engage in a signalling response. Here we describe a novel approach to inhibition of MAP kinase signal transduction through attempting to preserve the Shc-Erk complex under conditions of activated receptor. A library of existing drug molecules was computationally screened for hits that would bind to the ShcPTB and block its interaction with the RTKs EGFR and ErbB2. The primary hit from the screen was indomethacin, a non-steroidal anti-inflammatory drug. Validation of this molecule in vitro and in cellular efficacy studies in cancer cells provides proof of principle of the approach to pathway down-regulation and a potential optimizable lead compound.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/farmacologia , Reposicionamento de Medicamentos , Indometacina/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Proteínas Adaptadoras da Sinalização Shc/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Movimento Celular/efeitos dos fármacos , Receptores ErbB/química , Receptores ErbB/metabolismo , Células HeLa , Humanos , Indometacina/química , Indometacina/metabolismo , Células MCF-7 , Simulação de Acoplamento Molecular , Invasividade Neoplásica , Neoplasias/enzimologia , Neoplasias/patologia , Fosforilação , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Adaptadoras da Sinalização Shc/química , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Relação Estrutura-Atividade
9.
J Pharm Sci ; 108(7): 2334-2340, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30776382

RESUMO

The in vitro dissolution absorption system 2 (IDAS2), a recent invention comprised a conventional dissolution vessel containing 2 permeation chambers with Caco-2 cell monolayers mounted with their apical side facing the dissolution media, permits simultaneous measurement of dissolution and permeation of drugs from intact clinical dosage forms. The objectives of this study were (1) to assess the utility of IDAS2 in the determination of the effect of particle size on in vitro performance of indomethacin and (2) to find out whether the behavior in IDAS2 of 2 indomethacin products differing in particle size is correlated with their in vivo behavior. Indomethacin dissolution and permeation across Caco-2 cell monolayers were simultaneously measured in IDAS2; the dissolution and permeation profiles were simultaneously modeled using a simple two-compartment model. Compared to microsized indomethacin, the nanosized formulation increased the dissolution rate constant by fivefold, whereas moderately increasing the permeation rate constant and the kinetic solubility. As a result, the drug amount permeated across the Caco-2 cell monolayers doubled in the nanosized versus microsized formulation. The in vitro results showed a good correlation with in vivo human oral pharmacokinetic parameters, thus emphasizing the physiological relevance of IDAS2 data in predicting in vivo absorption.


Assuntos
Absorção Fisiológica/fisiologia , Indometacina/química , Indometacina/metabolismo , Administração Oral , Células CACO-2 , Linhagem Celular Tumoral , Humanos , Absorção Intestinal/fisiologia , Tamanho da Partícula , Permeabilidade , Solubilidade
10.
J Mol Model ; 24(7): 150, 2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29869728

RESUMO

Inhibitors selective towards the second isoform of prostaglandin synthase (cyclooxygenase, COX-2) are promising nonsteroidal anti-inflammatory drugs and antitumor medications. Methylation of the carboxylate group in the relatively nonselective COX inhibitor indomethacin confers significant COX-2 selectivity. Several other modifications converting indomethacin into a COX-2 selective inhibitor have been reported. Earlier experimental and computational studies on neutral indomethacin derivatives suggest that the methyl ester derivative likely binds to COX-2 with a similar binding mode as that observed for the parent indomethacin. However, docking studies followed by molecular dynamics simulations revealed two possible binding modes in COX-2 for indomethacin methyl ester, which differs from the experimental binding mode found for indomethacin. Both alternative binding modes might explain the observed COX-2 selectivity of indomethacin methyl ester. Graphical abstract Binding of indomethacin methyl ester to cyclooxygenase-2.


Assuntos
Ciclo-Oxigenase 2/química , Ésteres , Indometacina/química , Sítios de Ligação , Ciclo-Oxigenase 2/metabolismo , Ésteres/química , Ligação de Hidrogênio , Indometacina/metabolismo , Ligantes , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica , Relação Estrutura-Atividade
11.
Chemistry ; 23(39): 9397-9406, 2017 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-28489292

RESUMO

Dual stimuli-responsive nanoparticles capable of fine-tuning drug release to augment therapeutic efficacy have become a promising tool for anticancer drug delivery. However, the rational design of these "smart" nanoparticles for a selective delivery and controlled release of multidrug combinations in cancer cells to achieve synergistic effects remain challenging. Here we report the pH/redox dual responsive nanoparticle FA-DOX-Ind-NP (FA=folic acid, DOX=doxorubicin, Ind=indomethacin, NP=nanoparticle) based on the novel tumor targeting and biodegradable poly(ß-amino ester) polymer, and demonstrate its high ability to enter into cancer cells and release a combination of the anticancer drug doxorubicin and the non-steroidal anti-inflammatory drug indomethacin to achieve synergistic chemo-anti-inflammatory effects and overcome multidrug resistance. This study highlights the great potential of tumor targeting and dual stimuli-responsive nanoparticles for an efficient delivery of multidrug combination to improve the cancer therapeutic efficacy.


Assuntos
Anti-Inflamatórios/química , Antineoplásicos/química , Portadores de Fármacos/química , Nanopartículas/química , Animais , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Antineoplásicos/metabolismo , Antineoplásicos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Doxorrubicina/química , Doxorrubicina/metabolismo , Liberação Controlada de Fármacos , Ácido Fólico , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Indometacina/química , Indometacina/metabolismo , Interleucina-6/metabolismo , Células MCF-7 , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Microscopia Confocal , Polímeros/química , Células RAW 264.7 , Fator de Necrose Tumoral alfa/metabolismo
12.
Cell Microbiol ; 17(9): 1391-404, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25856684

RESUMO

Indomethacin, a cyclooxygenase-1 and -2 inhibitor widely used in the clinic for its potent anti-inflammatory/analgesic properties, possesses antiviral activity against several viral pathogens; however, the mechanism of antiviral action remains elusive. We have recently shown that indomethacin activates the double-stranded RNA (dsRNA)-dependent protein kinase R (PKR) in human colon cancer cells. Because of the important role of PKR in the cellular defence response against viral infection, herein we investigated the effect of indomethacin on PKR activity during infection with the prototype rhabdovirus vesicular stomatitis virus. Indomethacin was found to activate PKR in an interferon- and dsRNA-independent manner, causing rapid (< 5 min) phosphorylation of eukaryotic initiation factor-2 α-subunit (eIF2α). These events resulted in shutting off viral protein translation and blocking viral replication (IC50 = 2 µM) while protecting host cells from virus-induced damage. Indomethacin did not affect eIF2α kinases PKR-like endoplasmic reticulum-resident protein kinase (PERK) and general control non-derepressible-2 (GCN2) kinase, and was unable to trigger eIF2α phosphorylation in the presence of PKR inhibitor 2-aminopurine. In addition, small-interfering RNA-mediated PKR gene silencing dampened the antiviral effect in indomethacin-treated cells. The results identify PKR as a critical target for the antiviral activity of indomethacin and indicate that eIF2α phosphorylation could be a key element in the broad spectrum antiviral activity of the drug.


Assuntos
Antivirais/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Indometacina/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Vesiculovirus/efeitos dos fármacos , Proteínas Virais/biossíntese , eIF-2 Quinase/metabolismo , Linhagem Celular , Ativadores de Enzimas/metabolismo , Humanos , Concentração Inibidora 50 , Fosforilação , Processamento de Proteína Pós-Traducional
13.
J Am Chem Soc ; 135(31): 11663-9, 2013 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-23862760

RESUMO

Identifying cancer cells and quantifying cancer-related events in particular organelles in a rapid and sensitive fashion are important for early diagnosis and for studies on pathology and therapeutics of cancers. Herein a smart "off-on" cyclooxygenase-2-specific fluorescence probe (ANQ-IMC-6), able to report the presence of cancer cells and to image Golgi-related events, has been designed and evaluated. Cyclooxygenase-2 (COX-2) has been used as imaging target in the probe design, since this enzyme is a biomarker of virtually all cancer cell lines. In the free state in aqueous solution, ANQ-IMC-6 mainly exists in a folded conformation where probe fluorescence is quenched through photoinduced electron transfer between the fluorophore acenaphtho[1,2-b]quinoxaline (ANQ) and the recognition group, indomethacin (IMC). Fluorescence is turned on, by restraining the photoinduced electron transfer, when ANQ-IMC-6 is forced to adopt the unfolded state following binding to COX-2 in the Golgi apparatus of cancer cells. ANQ-IMC-6 provides high signal-to-background staining and has been successfully used to rapidly differentiate cancer cells from normal cells when using flow cytometry and one- and two-photon fluorescence microscopic imaging. Furthermore, ANQ-IMC-6 may be able to visualize dynamic changes of the Golgi apparatus during cancer cell apoptosis, with possible application to early diagnosis.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Corantes Fluorescentes/metabolismo , Neoplasias/diagnóstico , Neoplasias/metabolismo , Acenaftenos/química , Acenaftenos/metabolismo , Linhagem Celular Tumoral , Corantes Fluorescentes/química , Complexo de Golgi/metabolismo , Humanos , Indometacina/química , Indometacina/metabolismo , Modelos Moleculares , Imagem Óptica/métodos , Quinoxalinas/química , Quinoxalinas/metabolismo
14.
Biochem Pharmacol ; 85(8): 1195-202, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23399640

RESUMO

Phospho-tyrosol-indomethacin (PTI; MPI 621), a novel anti-cancer agent, is more potent and safer than conventional indomethacin. Here, we show that PTI was extensively metabolized in vitro and in vivo. PTI was rapidly hydrolyzed by carboxylesterases to generate indomethacin as its major metabolite in the liver microsomes and rats. PTI additionally undergoes cytochromes P450 (CYP)-mediated hydroxylation at its tyrosol moiety and O-demethylation at its indomethacin moiety. Of the five major human CYPs, CYP3A4 and CYP2D6 catalyze the hydroxylation and O-demethylation reactions of PTI, respectively; whereas CYP1A2, 2C9 and 2C19 are inactive towards PTI. In contrast to PTI, indomethacin is primarily O-demethylated by CYP2C9, which prefers acidic substrates. The hydrolyzed and O-demethylated metabolites of PTI are further glucuronidated and sulfated, facilitating drug elimination and detoxification. We observed substantial inter-species differences in the metabolic rates of PTI. Among the liver microsomes from various species, PTI was the most rapidly hydrolyzed, hydroxylated and O-demethylated in mouse, human and rat liver microsomes, respectively. These results reflect the differential expression patterns of carboxylesterase and CYP isoforms among these species. Of the human microsomes from various tissues, PTI underwent more rapid carboxylesterase- and CYP-catalyzed reactions in liver and intestine microsomes than in kidney and lung microsomes. Together, our results establish the metabolic pathways of PTI, reveal significant inter-species differences in its metabolism, and provide insights into the underlying biochemical mechanisms.


Assuntos
Antineoplásicos/metabolismo , Indometacina/análogos & derivados , Organofosfatos/metabolismo , Animais , Glucuronídeos/metabolismo , Humanos , Técnicas In Vitro , Indometacina/metabolismo , Mucosa Intestinal/metabolismo , Rim/metabolismo , Pulmão/metabolismo , Camundongos , Microssomos Hepáticos/metabolismo , Ratos
15.
Curr Pain Headache Rep ; 17(4): 327, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23423598

RESUMO

Indomethacin, as a member of the non-steroidal anti-inflammatory drug class, plays a special role in the treatment of headaches. By definition, it is completely efficacious in the treatment of the primary headache disorders paroxysmal hemicrania and hemicrania continua. Therefore, indomethacin is also used as a tool for differential diagnosis in headache clinics. Indomethacin has a clear action as a cyclooxygenase inhibitor. Additional mechanisms and interactions with cell signaling pathways and inflammatory pathways are considered in this article. However, it is not known what mechanism or interaction with pathophysiological mechanisms is the key to indomethacin's specific pharmacology in headache therapy. Focusing on headache therapy, we summarize the current knowledge of pharmacology, treatment options, and recommendations for the use of indomethacin in primary headaches. New findings from the field of headache research, as well as from Alzheimer's disease and cancer research on the pharmacological actions of indomethacin and their potential implications on the pathophysiology of indomethacin sensitive headaches, are discussed.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Inibidores de Ciclo-Oxigenase/farmacologia , Inibidores de Ciclo-Oxigenase/uso terapêutico , Cefaleia/tratamento farmacológico , Indometacina/farmacologia , Indometacina/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Inibidores de Ciclo-Oxigenase/metabolismo , Cefaleia/diagnóstico , Cefaleia/metabolismo , Humanos , Indometacina/metabolismo , Resultado do Tratamento
16.
Endocrinology ; 153(11): 5415-27, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23054057

RESUMO

Prostaglandins (PGs) are ubiquitous membrane-derived, lipid-signaling molecules with wide ranging effects throughout the body. In the brain, PGE(2) is the key regulator of fever after inflammation but is also implicated in neural development and synaptic plasticity. The steroid hormone estradiol is also a key regulator of neural development and synaptic plasticity. Recently, we showed that administering cyclooxygenase (COX) inhibitors to block PGE(2) production increased the total length of Purkinje cell dendrites, the number of dendritic spines, and the level of spinophilin protein, which is enriched in dendritic spines. Correspondingly, PGE(2) administration into the cerebellum decreased spinophilin protein content. We now report that PGE(2) stimulates estradiol synthesis in the immature rat cerebellum via enhanced activity of the aromatase enzyme. Treatment with cyclooxygenase inhibitors reduced cerebellar aromatase activity and estradiol content whereas PGE(2) administration increased both. Treatment with either PGE(2) or estradiol stunted Purkinje neuron dendritic length and complexity and produced a corresponding reduction in spinophilin content. Treatment with formestane to inhibit aromatase activity led to excessive sprouting of the dendritic tree, whereas elevated estradiol had the opposite effect. Electrophysiological measurements from Purkinje neurons revealed novel sex differences in input resistance and membrane capacitance that were abolished by estradiol exposure, whereas a sex difference in the amplitude of the afterhyperpolarization after an action potential was not. Correlated changes in action potential threshold suggest that prolonged alterations in neuronal firing activity could be a consequence of increased estradiol content during the second week of life. These findings reveal a previously unappreciated role for PG-stimulated steroidogenesis in the developing brain and a new potential route for inflammation-mediated disruption of neuronal maturation.


Assuntos
Cerebelo/metabolismo , Dendritos/metabolismo , Dinoprostona/metabolismo , Estradiol/biossíntese , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Aromatase/metabolismo , Células Cultivadas , Cerebelo/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Dendritos/efeitos dos fármacos , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Dinoprostona/farmacologia , Estradiol/farmacologia , Feminino , Indometacina/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/metabolismo , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia
17.
PLoS One ; 7(8): e43965, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22937138

RESUMO

Aldo-keto reductase 1C3 (AKR1C3) catalyses the NADPH dependent reduction of carbonyl groups in a number of important steroid and prostanoid molecules. The enzyme is also over-expressed in prostate and breast cancer and its expression is correlated with the aggressiveness of the disease. The steroid products of AKR1C3 catalysis are important in proliferative signalling of hormone-responsive cells, while the prostanoid products promote prostaglandin-dependent proliferative pathways. In these ways, AKR1C3 contributes to tumour development and maintenance, and suggest that inhibition of AKR1C3 activity is an attractive target for the development of new anti-cancer therapies. Non-steroidal anti-inflammatory drugs (NSAIDs) are one well-known class of compounds that inhibits AKR1C3, yet crystal structures have only been determined for this enzyme with flufenamic acid, indomethacin, and closely related analogues bound. While the flufenamic acid and indomethacin structures have been used to design novel inhibitors, they provide only limited coverage of the NSAIDs that inhibit AKR1C3 and that may be used for the development of new AKR1C3 targeted drugs. To understand how other NSAIDs bind to AKR1C3, we have determined ten crystal structures of AKR1C3 complexes that cover three different classes of NSAID, N-phenylanthranilic acids (meclofenamic acid, mefenamic acid), arylpropionic acids (flurbiprofen, ibuprofen, naproxen), and indomethacin analogues (indomethacin, sulindac, zomepirac). The N-phenylanthranilic and arylpropionic acids bind to common sites including the enzyme catalytic centre and a constitutive active site pocket, with the arylpropionic acids probing the constitutive pocket more effectively. By contrast, indomethacin and the indomethacin analogues sulindac and zomepirac, display three distinctly different binding modes that explain their relative inhibition of the AKR1C family members. This new data from ten crystal structures greatly broadens the base of structures available for future structure-guided drug discovery efforts.


Assuntos
3-Hidroxiesteroide Desidrogenases/química , Anti-Inflamatórios não Esteroides/química , Hidroxiprostaglandina Desidrogenases/química , 3-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase , Anti-Inflamatórios não Esteroides/metabolismo , Ácido Flufenâmico/química , Ácido Flufenâmico/metabolismo , Flurbiprofeno/química , Flurbiprofeno/metabolismo , Hidroxiprostaglandina Desidrogenases/metabolismo , Ibuprofeno/química , Ibuprofeno/metabolismo , Indometacina/química , Indometacina/metabolismo , Ácido Meclofenâmico/química , Ácido Meclofenâmico/metabolismo , Ácido Mefenâmico/química , Ácido Mefenâmico/metabolismo , Naproxeno/química , Naproxeno/metabolismo , Sulindaco/química , Sulindaco/metabolismo , Tolmetino/análogos & derivados , Tolmetino/química , Tolmetino/metabolismo
18.
J Pharmacol Exp Ther ; 340(2): 422-32, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22085648

RESUMO

Phospho-nonsteroidal anti-inflammatory drugs (phospho-NSAIDs) are novel NSAID derivatives with improved anticancer activity and reduced side effects in preclinical models. Here, we studied the metabolism of phospho-NSAIDs by carboxylesterases and assessed the impact of carboxylesterases on the anticancer activity of phospho-NSAIDs in vitro and in vivo. The expression of human liver carboxylesterase (CES1) and intestinal carboxylesterase (CES2) in human embryonic kidney 293 cells resulted in the rapid intracellular hydrolysis of phospho-NSAIDs. Kinetic analysis revealed that CES1 is more active in the hydrolysis of phospho-sulindac, phospho-ibuprofen, phospho-naproxen, phospho-indomethacin, and phospho-tyrosol-indomethacin that possessed a bulky acyl moiety, whereas the phospho-aspirins are preferentially hydrolyzed by CES2. Carboxylesterase expression leads to a significant attenuation of the in vitro cytotoxicity of phospho-NSAIDs, suggesting that the integrity of the drug is critical for anticancer activity. Benzil and bis-p-nitrophenyl phosphate (BNPP), two carboxylesterase inhibitors, abrogated the effect of carboxylesterases and resensitized carboxylesterase-expressing cells to the potent cytotoxic effects of phospho-NSAIDs. In mice, coadministration of phospho-sulindac and BNPP partially protected the former from esterase-mediated hydrolysis, and this combination more effectively inhibited the growth of AGS human gastric xenografts in nude mice (57%) compared with phospho-sulindac alone (28%) (p = 0.037). Our results show that carboxylesterase mediates that metabolic inactivation of phospho-NSAIDs, and the inhibition of carboxylesterases improves the efficacy of phospho-NSAIDs in vitro and in vivo.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Carboxilesterase/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Organofosfatos/metabolismo , Organofosfatos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/sangue , Anti-Inflamatórios não Esteroides/uso terapêutico , Aspirina/análogos & derivados , Aspirina/metabolismo , Aspirina/farmacologia , Carboxilesterase/antagonistas & inibidores , Carboxilesterase/genética , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Hidrolases de Éster Carboxílico/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Interações Medicamentosas/fisiologia , Quimioterapia Combinada/métodos , Inibidores Enzimáticos/farmacologia , Feminino , Células HEK293 , Humanos , Hidrólise , Ibuprofeno/análogos & derivados , Ibuprofeno/metabolismo , Ibuprofeno/farmacologia , Indometacina/análogos & derivados , Indometacina/metabolismo , Indometacina/farmacologia , Concentração Inibidora 50 , Cinética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Naproxeno/análogos & derivados , Naproxeno/metabolismo , Naproxeno/farmacologia , Nitrofenóis/farmacologia , Nitrofenóis/uso terapêutico , Organofosfatos/sangue , Organofosfatos/uso terapêutico , Compostos Organofosforados/sangue , Compostos Organofosforados/metabolismo , Compostos Organofosforados/farmacologia , Compostos Organofosforados/uso terapêutico , Fenilglioxal/análogos & derivados , Fenilglioxal/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Sulindaco/análogos & derivados , Sulindaco/sangue , Sulindaco/metabolismo , Sulindaco/farmacologia , Sulindaco/uso terapêutico
19.
Int J Mol Sci ; 12(6): 3998-4008, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21747720

RESUMO

In the present work, we investigated the effect of non-steroidal anti-inflammatory drugs (NSAIDs) on the monophenolase and diphenolase activity of mushroom tyrosinase. The results showed that diflunisal and indomethacin inhibited both monophenolase and diphenolase activity. For monophenolase activity, the lag time was extended in the presence of diflunisal. In the presence of indomethacin, the lag time did not change. IC(50) values of monophenolase activity were estimated to be 0.112 mM (diflunisal) and 1.78 mM (indomethacin). Kinetic studies of monophenolase activity revealed that both diflunisal and indomethacin were non-competitive inhibitors. For diphenolase activity, IC(50) values were estimated to be 0.197 mM (diflunisal) and 0.509 mM (indomethacin). Diflunisal and indomethacin were also found to be non-competitive diphenolase inhibitors.


Assuntos
Agaricales/enzimologia , Anti-Inflamatórios não Esteroides/química , Catecol Oxidase/antagonistas & inibidores , Inibidores Enzimáticos/química , Monofenol Mono-Oxigenase/antagonistas & inibidores , Oxirredutases/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/metabolismo , Catecol Oxidase/metabolismo , Diflunisal/química , Diflunisal/metabolismo , Inibidores Enzimáticos/metabolismo , Indometacina/química , Indometacina/metabolismo , Concentração Inibidora 50 , Cinética , Monofenol Mono-Oxigenase/metabolismo , Oxirredutases/metabolismo , Ligação Proteica
20.
Biomacromolecules ; 12(9): 3124-30, 2011 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-21780829

RESUMO

A polymeric prodrug, PEGylated indomethacin (MPEG-indo), was prepared and then used to interact with α-cyclodextrin (α-CD) in their aqueous mixed system. This process could lead to the formation of supramolecular hydrogel under mild conditions and simultaneous encapsulation of MPEG-indo in the hydrogel matrix. For the formed supramolecular hydrogel, its gelation kinetics, mechanical strength, shear-thinning behavior and thixotropic response were investigated with respect to the effects of MPEG-indo and α-CD amounts by dynamic and steady rheological tests. Meanwhile, the possibility of using this hydrogel matrix as injectable drug delivery system was also explored. By in vitro release and cell viability tests, it was found that the encapsulated MPEG-indo could exhibit a controlled and sustained release behavior as well as maintain its biological activity.


Assuntos
Preparações de Ação Retardada/síntese química , Portadores de Fármacos/síntese química , Indometacina/metabolismo , Polietilenoglicóis/química , Polímeros/química , Pró-Fármacos/síntese química , alfa-Ciclodextrinas/química , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada/metabolismo , Preparações de Ação Retardada/farmacologia , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacologia , Células Hep G2 , Humanos , Indometacina/química , Cinética , Espectroscopia de Ressonância Magnética , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacologia , Resistência ao Cisalhamento , Viscosidade , Água
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA