Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Carcinog ; 53(8): 659-66, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23475608

RESUMEN

Matrix metalloproteinases (MMPs) and monocyte chemoattractant protein-1 (MCP-1, CCL2) are known to be upregulated in many tumors. Their roles in tumor invasion and metastasis are being uncovered. How they are related to each other and involved in tumor progression remains to be determined. Earlier it was reported that I-BOP-initiated activation of thromboxane A2 receptor (TP) induced the release of MMP-1, MMP-3, and MMP-9 from lung cancer A549 cells overexpressing TPα (A549-TPα). Herein it was found that MMP-1, but not MMP-3 or MMP-9, induced the expression of MCP-1 in A549 cells. Conditioned medium (CM) from I-BOP activated, MMP-1 siRNA pretreated A549-TPα cells induced greatly attenuated expression of MCP-1 in A549 cells indicating that MMP-1 in the CM contributed significantly to the expression of MCP-1. MMP-1 was shown to activate protease-activated receptor 2 (PAR2) instead of commonly assumed PAR1 to increase the expression of MCP-1 in A549 cells. This conclusion was reached from the following findings: (1) expression of MCP-1 induced by trypsin, a PAR2 agonist, and also PAR2 agonist peptide, was inhibited by a PAR2 antagonist; (2) expression of MCP-1 induced by MMP-1 and by CM from I-BOP activated A549-TPα cells was blocked by a PAR2 antagonist but not by other PAR antagonists; (3) expression of MCP-1 induced by MMP-1 and by CM from I-BOP activated A549-TPα cells was attenuated significantly by pretreatment of cells with PAR2-siRNA. These results suggest that PAR2 is a novel MMP-1 target mediating MMP-1-induced signals in A549 lung cancer cells.


Asunto(s)
Adenocarcinoma/metabolismo , Quimiocina CCL2/metabolismo , Neoplasias Pulmonares/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Receptor PAR-2/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Western Blotting , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Quimiocina CCL2/genética , Medios de Cultivo Condicionados/farmacología , Ácidos Grasos Insaturados/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Metaloproteinasa 1 de la Matriz/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor PAR-2/antagonistas & inhibidores , Receptor PAR-2/genética , Receptores de Tromboxano A2 y Prostaglandina H2/antagonistas & inhibidores , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
2.
Biochem J ; 453(3): 447-54, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23849058

RESUMEN

Cocaine is a widely abused drug without an FDA (Food and Drug Administration)-approved medication. It has been recognized that an ideal anti-cocaine medication would accelerate cocaine metabolism producing biologically inactive metabolites via a route similar to the primary cocaine-metabolizing pathway, i.e. human BChE (butyrylcholinesterase)-catalysed hydrolysis. However, the native human BChE has a low catalytic activity against cocaine. We recently designed and discovered a BChE mutant (A199S/F227A/S287G/A328W/Y332G) with a high catalytic activity (kcat=5700 min-1, Km=3.1 µM) specifically for cocaine, and the mutant was proven effective in protecting mice from acute cocaine toxicity of a lethal dose of cocaine (180 mg/kg of body weight, LD100). Further characterization in animal models requires establishment of a high-efficiency stable cell line for the BChE mutant production at a relatively larger scale. It has been extremely challenging to develop a high-efficiency stable cell line expressing BChE or its mutant. In the present study, we successfully developed a stable cell line efficiently expressing the BChE mutant by using a lentivirus-based repeated-transduction method. The scaled-up protein production enabled us to determine for the first time the in vivo catalytic activity and the biological half-life of this high-activity mutant of human BChE in accelerating cocaine clearance. In particular, it has been demonstrated that the BChE mutant (administered to mice 1 min prior to cocaine) can quickly metabolize cocaine and completely eliminate cocaine-induced hyperactivity in rodents, implying that the BChE mutant may be developed as a promising therapeutic agent for cocaine abuse treatment.


Asunto(s)
Butirilcolinesterasa/metabolismo , Cocaína/metabolismo , Ingeniería de Proteínas/métodos , Animales , Butirilcolinesterasa/genética , Células CHO , Línea Celular , Trastornos Relacionados con Cocaína , Cricetinae , Femenino , Humanos , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
3.
Cancer Metastasis Rev ; 30(3-4): 409-17, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22020925

RESUMEN

15-Hydroxyprostaglandin dehydrogenase (15-PGDH) is a key prostaglandin catabolic enzyme catalyzing the oxidation and inactivation of prostaglandin E(2) (PGE(2)) synthesized from the cyclooxygenase (COX) pathway. Accumulating evidence indicates that 15-PGDH may function as a tumor suppressor antagonizing the action of COX-2 oncogene. 15-PGDH has been found to be down-regulated contributing to elevated levels of PGE(2) in most tumors. The expression of 15-PGDH and COX-2 appears to be regulated reciprocally in cancer cells. Down-regulation of 15-PGDH in tumors is due, in part, to transcriptional repression and epigenetic silencing. Numerous agents have been found to up-regulate 15-PGDH by down-regulation of transcriptional repressors and by attenuation of the turnover of the enzyme. Up-regulation of 15-PGDH may provide a viable approach to cancer chemoprevention. Further catabolism of 15-keto-prostaglandin E(2) is catalyzed by 15-keto-prostaglandin-∆(13)-reductase (13-PGR), which also exhibits LTB(4)-12-hydroxydehydrogenase (LTB(4)-12-DH) activity. 13-PGR/LTB(4)-12-DH behaves as a tumor suppressor as well. This review summarizes current knowledge of the expression and function of 15-PGDH and 13-PGR/LTB(4)-12-DH in lung and other tissues during tumor progression. Future directions of research on these prostaglandin catabolic enzymes as tumor suppressors are also discussed.


Asunto(s)
15-Oxoprostaglandina 13-Reductasa/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Neoplasias/enzimología , Proteínas Supresoras de Tumor/metabolismo , 15-Oxoprostaglandina 13-Reductasa/genética , Animales , Ciclooxigenasa 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Neoplasias/patología , Neoplasias/terapia , Prostaglandinas/metabolismo , Proteínas Supresoras de Tumor/genética
4.
Proc Natl Acad Sci U S A ; 106(23): 9409-13, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19470469

RESUMEN

Pharmacologic inhibitors of the prostaglandin-synthesizing COX-2 oncogene prevent the development of premalignant human colon adenomas. However, resistance to treatment is common. In this study, we show that the adenoma prevention activity of the COX-2 inhibitor celecoxib requires the concomitant presence of the 15-hydroxyprostaglandin dehydrogenase (15-PGDH) tumor suppressor gene, and that loss of 15-PGDH expression imparts resistance to celecoxib's anti-tumor effects. We first demonstrate that the adenoma-preventive activity of celecoxib is abrogated in mice genetically lacking 15-PGDH. In FVB mice, celecoxib prevents 85% of azoxymethane-induced tumors >1 mm in size, but is essentially inactive in preventing tumor induction in 15-PGDH-null animals. Indeed, celecoxib treated 15-PGDH null animals develop more tumors than do celecoxib naive WT mice. In parallel with the loss of tumor prevention activity, celecoxib-mediated suppression of colonic PGE(2) levels is also markedly attenuated in 15-PGDH-null versus WT mice. Finally, as predicted by the murine models, humans with low colonic 15-PGDH levels also exhibit celecoxib resistance. Specifically, in a colon adenoma prevention trial, in all cases tested, individuals who developed new adenomas while receiving celecoxib treatment were also found as having low colonic 15-PGDH levels.


Asunto(s)
Adenoma/prevención & control , Neoplasias del Colon/prevención & control , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Pirazoles/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Celecoxib , Colon/metabolismo , Colon/patología , Colonoscopía , Control de Medicamentos y Narcóticos , Humanos , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Pirazoles/metabolismo , Sulfonamidas/metabolismo
5.
Mol Pharmacol ; 79(2): 290-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20971807

RESUMEN

Cocaine is a widely abused drug without a U.S. Food and Drug Administration-approved medication. There is a recognized, promising anticocaine medication to accelerate cocaine metabolism, producing biologically inactive metabolites via a route similar to the primary cocaine-metabolizing pathway [i.e., cocaine hydrolysis catalyzed by butyrylcholinesterase (BChE) in plasma]. An ideal, therapeutically valuable mutant of human BChE should have not only a significantly improved catalytic activity against (-)-cocaine but also certain selectivity for (-)-cocaine over neurotransmitter acetylcholine (ACh), such that one would not expect systemic administration of the BChE mutant to interrupt cholinergic transmission. The present study accounting for the mutation-caused changes of the catalytic activities of BChE against both (-)-cocaine and ACh by means of molecular modeling and site-directed mutagenesis has led to identification of three BChE mutants that have not only a considerably improved catalytic efficiency against (-)-cocaine but also the desirable selectivity for (-)-cocaine over ACh. Two representative BChE mutants have been confirmed to be potent in actual protection of mice from acute toxicity (convulsion and lethality) of a lethal dose of cocaine (180 mg/kg). Pretreatment with the BChE mutant (i.e., 1 min before cocaine administration) dose-dependently protected mice against cocaine-induced convulsions and lethality. In particular, all mice pretreated with the mutant (e.g., 0.02 mg or more of A199S/F227A/S287G/A328W/E441D BChE) survived. The in vivo data reveal the primary factor (i.e., the relative catalytic efficiency), determining the efficacy in practical protection of mice from the acute cocaine toxicity and future direction for further improving the efficacy of the enzyme in the cocaine overdose treatment.


Asunto(s)
Butirilcolinesterasa/metabolismo , Cocaína/farmacocinética , Inactivación Metabólica , Acetilcolina/metabolismo , Animales , Butirilcolinesterasa/genética , Cocaína/toxicidad , Humanos , Enlace de Hidrógeno , Masculino , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
6.
Prostaglandins Other Lipid Mediat ; 96(1-4): 37-40, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21763448

RESUMEN

NSAIDs are known to be inhibitors of cyclooxygenase-2 (COX-2) accounting for their anti-inflammatory and anti-tumor activities. However, the anti-tumor activity cannot be totally attributed to their COX-2 inhibitory activity as these drugs can also inhibit the growth and tumor formation of COX-2-null cell lines. Several potential targets aside from COX-2 for NSAIDs have been proposed. 15-Hydroxyprostaglandin dehydrogenase (15-PGDH), a key prostaglandin catabolic enzyme, was recently shown to be a tumor suppressor. Effects of NSAIDs on 15-PGDH expression were therefore studied. Flurbiprofen, indomethacin and other NSAIDs stimulated 15-PGDH activity in colon cancer HT29 cells as well as in lung cancer A549 cells and glioblastoma T98G cells. (R)-flurbiprofen and sulindac sulfone, COX-2 inactive analogs, also stimulated 15-PGDH activity indicating induction of 15-PGDH is independent of COX-2 inhibition. Stimulation of 15-PGDH expression and activity by NSAIDs was examined in detail in colon cancer HT29 cells using flurbiprofen as a stimulant. Flurbiprofen stimulated 15-PGDH expression and activity by increasing transcription and translation and by decreasing the turnover of 15-PGDH. Mechanism of stimulation of 15-PGDH expression is not clear. Protease(s) involved in the turnover of 15-PGDH remains to be identified. However, flurbiprofen down-regulated matrix metalloproteinase-9 (MMP-9) which was shown to degrade 15-PGDH, but up-regulated tissue inhibitor of metalloproteinase-1 (TIMP-1), an inhibitor of MMP-9 contributing further to a slower turnover of 15-PGDH. Taken together, NSAIDs may up-regulate 15-PGDH by increasing the protein expression as well as decreasing the turnover of 15-PGDH in cancer cells.


Asunto(s)
Adenocarcinoma/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica/inmunología , Glioblastoma/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Neoplasias Pulmonares/metabolismo , Transducción de Señal/inmunología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Ciclooxigenasa 2/inmunología , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Activación Enzimática , Flurbiprofeno/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/inmunología , Glioblastoma/patología , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/inmunología , Indometacina/farmacología , Cinética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Transducción de Señal/genética , Sulindac/análogos & derivados , Sulindac/farmacología , Inhibidor Tisular de Metaloproteinasa-1/agonistas , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Regulación hacia Arriba
7.
Bioorg Med Chem ; 19(20): 6077-86, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21920764

RESUMEN

Microsomal prostaglandin E synthase-1 (mPGES-1) is an inducible prostaglandin E synthase after exposure to pro-inflammatory stimuli and, therefore, represents a novel target for therapeutic treatment of acute and chronic inflammatory disorders. It is essential to identify mPGES-1 inhibitors with novel scaffolds as new leads or hits for the purpose of drug design and discovery that aim to develop the next-generation anti-inflammatory drugs. Herein we report novel mPGES-1 inhibitors identified through a combination of large-scale structure-based virtual screening, flexible docking, molecular dynamics simulations, binding free energy calculations, and in vitro assays on the actual inhibitory activity of the computationally selected compounds. The computational studies are based on our recently developed three-dimensional (3D) structural model of mPGES-1 in its open state. The combined computational and experimental studies have led to identification of new mPGES-1 inhibitors with new scaffolds. In particular, (Z)-5-benzylidene-2-iminothiazolidin-4-one is a promising novel scaffold for the further rational design and discovery of new mPGES-1 inhibitors. To our best knowledge, this is the first time a 3D structural model of the open state mPGES-1 is used in structure-based virtual screening of a large library of available compounds for the mPGES-1 inhibitor identification. The positive experimental results suggest that our recently modeled trimeric structure of mPGES-1 in its open state is ready for the structure-based drug design and discovery.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/química , Diseño de Fármacos , Humanos , Modelos Moleculares , Prostaglandina-E Sintasas , Unión Proteica , Relación Estructura-Actividad , Termodinámica
8.
Exp Cell Res ; 316(14): 2251-9, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20632471

RESUMEN

IL-4, an anti-inflammatory cytokine, was found to stimulate 15-PGDH activity in A549 and other lung cancer cells. Increase in 15-PGDH activity was due to increased transcription and decreased protein turnover of 15-PGDH. MMP-9 was shown to result in decreased levels of 15-PGDH in A549 cells. IL-4 induced down-regulation of MMP-9 mRNA and up-regulation of TIMP-1, an inhibitor of MMP-9. These data suggest that IL-4-induced down-regulation of MMP-9 activity may contribute to up-regulation of 15-PGDH in A549 cells. The role of JAK-STAT6 in IL-4-induced 15-PGDH expression was examined by using inhibitors. Inhibitors of JAKs, kaempferol and JAK inhibitor I, attenuated IL-4-stimulated STAT6 phosphorylation and 15-PGDH activity in a comparable concentration-dependent manner. Furthermore, JAK inhibitor I blocked IL-4-induced down-regulation of MMP-9 mRNA and up-regulation of TIMP-1 but not IL-4-stimulated up-regulation of 15-PGDH mRNA. These results indicate that JAK-STAT6 participated in IL-4-induced up-regulation of 15-PGDH through inhibition of MMP-9-mediated degradation. The roles of other signaling kinases in IL-4-induced 15-PGDH expression were also examined by using various inhibitors. Inhibitors of various MAPKs, PI-3K and PKC attenuated significantly IL-4-stimulated 15-PGDH activity indicating that MAPKs, PI-3K/Akt and PKC pathways participated in IL-4-induced up-regulation of 15-PGDH. Our results indicate that IL-4 up-regulates 15-PGDH by increased gene transcription and decreased protein turnover and the up-regulation can be mediated by JAK-STAT6 as well as MAPKs, PI-3K/Akt and PKC pathways.


Asunto(s)
Activación Enzimática/efectos de los fármacos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Interleucina-4/farmacología , Neoplasias Pulmonares/enzimología , Regulación hacia Arriba/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos
9.
Bioorg Med Chem ; 18(4): 1428-33, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20122835

RESUMEN

Prostaglandins have a short life in vivo because they are metabolized rapidly by oxidation to 15-ketoprostaglandins catalyzed by a cytosolic enzyme known as NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH). Previously, CT-8, a thiazolidinedione analogue, was found to be a potent inhibitor of 15-PGDH. Structure-activity analysis indicated that the N-methylation of thiazolidine-2,4-dione, CT-8, abolished the inhibitory activity, whereas the introduction of an ethyl hydroxyl group at amine in CT-8 still had a good inhibitory effect. Based on the structures of the thiazolidinediones analogues and inhibitory activity, a range of benzylidene thiazolidinedione derivatives were synthesized with different substituents on the phenyl ring and their inhibitory activity was evaluated. Replacement of the cyclohexylethyl group of CT-8 with the hetero five-member ring increased the inhibitory potency. However, replacement of the cyclohexylethyl group with a hetero six-member ring decreased the inhibitory potency significantly. It was found that compound 2 (5-(4-(2-(thiophen-2-yl)ethoxy)benzylidene)thiazolidine-2,4-dione) was the most potent inhibitor that was effective in the nanomolar range.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Tiazolidinas/síntesis química , Espectroscopía de Resonancia Magnética , Relación Estructura-Actividad , Tiazolidinas/química , Tiazolidinas/farmacología
10.
Biophys J ; 96(5): 1931-8, 2009 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-19254552

RESUMEN

It is recognized that an ideal anti-cocaine treatment is to accelerate cocaine metabolism by producing biologically inactive metabolites via a route similar to the primary cocaine-metabolizing pathway, i.e., butyrylcholinesterase (BChE)-catalyzed hydrolysis of cocaine. BChE mutants with a higher catalytic activity against (-)-cocaine are highly desired for use as an exogenous enzyme in humans. To develop a rational design for high-activity mutants, we carried out free-energy perturbation (FEP) simulations on various mutations of the transition-state structures in addition to the corresponding free-enzyme structures by using an extended FEP procedure. The FEP simulations on the mutations of both the free-enzyme and transition-state structures allowed us to calculate the mutation-caused shift of the free-energy change from the free enzyme (BChE) to the transition state, and thus to theoretically predict the mutation-caused shift of the catalytic efficiency (k(cat)/K(M)). The computational predictions are supported by the kinetic data obtained from the wet experiments, demonstrating that the FEP-based computational design approach is promising for rational design of high-activity mutants of an enzyme. One of the BChE mutants designed and discovered in this study has an approximately 1800-fold improved catalytic efficiency against (-)-cocaine compared to wild-type BChE. The high-activity mutant may be therapeutically valuable.


Asunto(s)
Butirilcolinesterasa/química , Simulación por Computador , Modelos Químicos , Ingeniería de Proteínas , Algoritmos , Butirilcolinesterasa/genética , Catálisis , Cocaína/química , Diseño Asistido por Computadora , Humanos , Cinética , Modelos Moleculares , Proteínas Mutantes/química , Mutación
11.
Carcinogenesis ; 30(9): 1606-13, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570744

RESUMEN

Previous studies implicate that activation of thromboxane A(2) receptor (TP) induced cell proliferation and transformation in several cell lines. We report here that the activation of TP by its agonist, [1S-[1alpha, 2alpha (Z), 3beta (1E, 3S*), 4alpha]]-7-[3-[3-hydroxy-4-(4-iodophenoxy)-1-butenyl]-7-oxabicyclo [2.2.1] hept-2-yl]-5-heptenoic acid (I-BOP), induced Nurr1 expression and stimulated proliferation of human lung cancer cells. Nurr1, an orphan nuclear receptor in the nuclear receptor subfamily 4A subfamily, has been implicated in cell proliferation, differentiation and apoptosis. I-BOP markedly induced Nurr1 messenger RNA and protein levels as compared with other subfamily members, Nur77 and Nor-1. The signaling pathways of I-BOP-induced Nurr1 expression were examined by using various inhibitors of signaling molecules. The induction of Nurr1 expression by I-BOP appeared to be mediated through protein kinase A (PKA)/cAMP response element binding (CREB), protein kinase C and mitogen-activated protein kinase/extracellular signal-regulated kinase pathways and not related to epidermal growth factor receptor and prostaglandin E(2) pathways. Transcriptional activation of Nurr1 gene by I-BOP was further investigated at the promoter level in H157 cells. 5'-Deletion analysis, site-directed mutagenesis and luciferase reporter assay demonstrated that Nurr1 expression was induced by I-BOP in a PKA/CREB-dependent manner. Further studies have revealed that Nurr1 may mediate cyclin D1 expression and I-BOP-induced cell proliferation in H157 cells since small interfering RNA of Nurr1 blocked I-BOP-induced cyclin D1 expression and cell proliferation and also decreased cell growth rate. These results provide strong evidence that Nurr1 plays a significant role in cell proliferation and may mediate TP agonist-induced proliferation in lung cancer cells.


Asunto(s)
Proteínas de Unión al ADN/genética , Neoplasias Pulmonares/patología , Receptores de Tromboxano A2 y Prostaglandina H2/fisiología , Factores de Transcripción/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Proliferación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Ciclina D1/genética , Dinoprostona/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Grasos Insaturados/farmacología , Regulación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Fosforilación , Proteína Quinasa C/fisiología , Transducción de Señal
12.
Am J Physiol Gastrointest Liver Physiol ; 297(3): G559-66, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19608733

RESUMEN

Multiple lines of evidence have suggested a role for both bile acids and prostaglandins (PG) in gastrointestinal carcinogenesis. Levels of PGE(2) are determined by both synthesis and catabolism. Previously, bile acid-mediated induction of cyclooxygenase-2 (COX-2) was found to stimulate PGE(2) synthesis. NAD(+)-dependent 15-hydroxyprostaglandin dehydrogenase (15-PGDH), the key enzyme responsible for the catabolism of PGE(2), has been linked to colorectal carcinogenesis. In this study, we determined whether bile acids altered the expression of 15-PGDH in human colon cancer cell lines. Treatment with unconjugated bile acids (chenodeoxycholate and deoxycholate) suppressed the transcription of 15-PGDH, resulting in reduced amounts of 15-PGDH mRNA, protein, and enzyme activity. Conjugated bile acids were less potent suppressors of 15-PGDH expression than unconjugated bile acids. Treatment with chenodeoxycholate activated protein kinase C (PKC), leading in turn to increased extracellular signal-regulated kinase (ERK) 1/2 activity. Small molecules that inhibited bile acid-mediated activation of PKC and ERK1/2 also blocked the downregulation of 15-PGDH. Bile acids induced early growth response factor-1 (Egr-1) and Snail, a repressive transcription factor that bound to the 15-PGDH promoter. Silencing Egr-1 or Snail blocked chenodeoxycholate-mediated downregulation of 15-PGDH. Together, these data indicate that bile acids activate the signal transduction pathway PKC --> ERK1/2 --> Egr-1 --> Snail and thereby suppress 15-PGDH transcription. Bile acids appear to increase the release of PGs from cells by downregulating catabolism in addition to stimulating synthesis. These results provide new mechanistic insights into the link between bile acids and gastrointestinal carcinogenesis.


Asunto(s)
Ácido Quenodesoxicólico/metabolismo , Colon/enzimología , Ácido Desoxicólico/metabolismo , Regulación Enzimológica de la Expresión Génica , Hidroxiprostaglandina Deshidrogenasas/genética , ARN Mensajero/metabolismo , Transcripción Genética , Sitios de Unión , Dinoprostona/metabolismo , Regulación hacia Abajo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Células HT29 , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Regiones Promotoras Genéticas , Proteína Quinasa C/metabolismo , Interferencia de ARN , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Tiempo , Factores de Transcripción/metabolismo
13.
Arch Biochem Biophys ; 487(2): 139-45, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19501039

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) are known to inhibit prostaglandin synthetic enzyme, cyclooxygenases (COXs), as well as to exhibit anti-tumor activity although at much higher concentrations. 15-Hydroxyprostaglandin dehyrogenase (15-PGDH), a key prostaglandin catabolic enzyme, was recently shown to be a tumor suppressor. Effects of NSAIDs on 15-PGDH expression were therefore examined. Flurbiprofen and several other NSAIDs were found to induce 15-PGDH expression in human colon cancer HT29 cells. Flurbiprofen, the most active one, was also shown to induce 15-PGDH expression in other types of cancer cells. Induction of 15-PGDH expression appeared to occur at the stage of mRNA as levels of 15-PGDH mRNA were increased by flurbiprofen in HT29 cells. Levels of 15-PGDH were also found to be regulated at the stage of protein turnover. MEK inhibitors, PD98059 and U-0126, which inhibited ERK phosphorylation were shown to elevate 15-PGDH levels very significantly. These inhibitors did not appear to alter 15-PGDH mRNA levels but down-regulate matrix metalloproteinase-9 (MMP-9). This protease was shown to degrade and inactivate 15-PGDH suggesting that elevation of 15-PGDH levels could be due to inhibition of MMP-9 expression by these inhibitors. Similarly, flurbiprofen was also demonstrated to inhibit ERK activation and to down-regulate MMP-9 expression. Furthermore, flurbiprofen was shown to induce the expression of tissue inhibitor of metalloproteinase-1 (TIMP-1), an inhibitor of MMP-9. The turnover of 15-PGDH was found to prolong in the presence of flurbiprofen as compared to that in the absence of this drug. Taken together, these results indicate that flurbiprofen up-regulates 15-PGDH by increasing the expression and decreasing the degradation of 15-PGDH in HT29 cells.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Neoplasias del Colon/patología , Flurbiprofeno/farmacología , Hidroxiprostaglandina Deshidrogenasas/genética , Regulación hacia Arriba/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Células HT29 , Humanos , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Metaloproteinasa 9 de la Matriz/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/genética
14.
Mol Endocrinol ; 22(1): 105-12, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17872381

RESUMEN

Parturition is a complex mammalian physiological process whose fundamental determinants have remained elusive. The increasing incidence of human preterm birth, a leading cause of infant mortality, highlights the importance of further understanding mechanisms regulating the timing of birth. Parturition is initiated in most nonprimate mammals, including mice, through a decrease in circulating progesterone caused by elevated prostaglandins. In humans, other higher primates, and guinea pigs, no consistent decrease in circulating progesterone occurs before the onset of labor. The divergence in endocrine control of labor initiation between most mammals compared with the great apes and guinea pigs gives rise to the question: how could a mechanism for the initiation of labor not requiring the withdrawal of progesterone evolve? Here, we genetically modulate prostaglandin signaling to determine the role of prostaglandin catabolism in the timing of birth. We find spontaneous preterm labor in the absence of progesterone withdrawal in 15-hydroxyprostaglandin dehydrogenase hypomorphic mice. The onset of labor in these hypomorphic mice is preceded by prematurely increased concentrations of prostaglandin E(2) and F(2alpha). Moreover, genetic crosses demonstrate a role for fetal genotype in birth timing. Together, these findings demonstrate a 15-hydroxyprostaglandin dehydrogenase-dependent shift in the physiology of murine parturition to one resembling the physiology of higher primates. Thus, endocrine control of labor has the capacity to plastically adapt to changes in genetically determined prostaglandin signals.


Asunto(s)
Animales Recién Nacidos/sangre , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Progesterona/sangre , Animales , Animales Recién Nacidos/metabolismo , Northern Blotting , Dinoprost/sangre , Dinoprost/metabolismo , Dinoprostona/sangre , Dinoprostona/metabolismo , Femenino , Feto/metabolismo , Genotipo , Edad Gestacional , Hidroxiprostaglandina Deshidrogenasas/genética , Hibridación in Situ , Hígado/metabolismo , Ratones , Modelos Genéticos , Ovario/metabolismo , Placenta/metabolismo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Útero/metabolismo
15.
Cancer Res ; 67(12): 5587-93, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575121

RESUMEN

Evidence indicates that the induction of cyclooxygenase-2 (COX-2) and high prostaglandin E2 (PGE2) levels contribute to the pathogenesis of non-small-cell lung cancer (NSCLC). In addition to overproduction by COX-2, PGE2 concentrations also depend upon the levels of the PGE2 catabolic enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH). We find a dramatic down-regulation of PGDH protein in NSCLC cell lines and in resected human tumors when compared with matched normal lung. Affymetrix array analysis of 10 normal lung tissue samples and 49 resected lung tumors revealed a much lower expression of PGDH transcripts in all NSCLC histologic groups. In addition, treatment with the epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI) erlotinib increased the expression of 15-PGDH in a subset of NSCLC cell lines. This effect may be due in part to an inhibition of the extracellular signal-regulated kinase (ERK) pathway as treatment with mitogen-activated protein kinase kinase (MEK) inhibitor U0126 mimics the erlotinib results. We show by quantitative reverse transcription-PCR that the transcript levels of ZEB1 and Slug transcriptional repressors are dramatically reduced in a responsive cell line upon EGFR and MEK/ERK inhibition. In addition, the Slug protein, but not ZEB1, binds to the PGDH promoter and represses transcription. As these repressors function by recruiting histone deacetylases to promoters, it is likely that PGDH is repressed by an epigenetic mechanism involving histone deacetylation, resulting in increased PGE2 activity in tumors. This effect is reversible in a subset of NSCLC upon treatment with an EGFR TKI.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Receptores ErbB/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Neoplasias Pulmonares/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Epigénesis Genética , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Proteínas de Homeodominio/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidores de Proteínas Quinasas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Transcripción Genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
16.
Sci Rep ; 9(1): 17612, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31772225

RESUMEN

Prostaglandins (PGs) have critical signaling functions in a variety of processes including the establishment and maintenance of pregnancy, and the initiation of labor. Most PGs are non-enzymatically degraded, however, the two PGs most prominently implicated in the termination of pregnancy, including the initiation of labor, prostaglandin E2 (PGE2) and prostaglandin F2α (PGF2α), are enzymatically degraded by 15-hydroxyprostaglandin dehydrogenase (15-HPGD). The role of PG metabolism by 15-HPGD in the maintenance of pregnancy remains largely unknown, as direct functional studies are lacking. To test the hypothesis that 15-PGDH-mediated PG metabolism is essential for pregnancy maintenance and normal labor timing, we generated and analyzed pregnancy in 15-HPGD knockout mice (Hpgd-/-). We report here that pregnancies resulting from matings between 15-HPGD KO mice (Hpgd-/- X Hpgd-/-KO mating) are terminated at mid gestation due to a requirement for embryo derived 15-HPGD. Aside from altered implantation site spacing, pregnancies from KO matings look grossly and histologically normal at days post coitum (dpc) 6.5 and 7.5 of pregnancy. However, virtually all of these pregnancies are resorbed by dpc 8.5. This resorption is preceded by elevation of PGF2∝ but is not preceded by a decrease in circulating progesterone, suggesting that pregnancy loss is a local inflammatory phenomenon rather than a centrally mediated phenomena. This pregnancy loss can be temporarily deferred by indomethacin treatment, but treated pregnancies are not maintained to term and indomethacin treatment increases maternal mortality. We conclude that PG metabolism to inactive products by embryo derived 15-HPGD is essential for pregnancy maintenance in mice, and may serve a similar function during human pregnancy.


Asunto(s)
Aborto Espontáneo/genética , Hidroxiprostaglandina Deshidrogenasas/fisiología , Mantenimiento del Embarazo/fisiología , Aborto Espontáneo/enzimología , Aborto Espontáneo/prevención & control , Animales , Ciclooxigenasa 1/biosíntesis , Ciclooxigenasa 1/genética , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Dinoprost/metabolismo , Dinoprostona/metabolismo , Implantación del Embrión , Femenino , Feto/enzimología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Genotipo , Edad Gestacional , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Hidroxiprostaglandina Deshidrogenasas/deficiencia , Hidroxiprostaglandina Deshidrogenasas/genética , Indometacina/farmacología , Indometacina/uso terapéutico , Indometacina/toxicidad , Muerte Materna/etiología , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Mantenimiento del Embarazo/efectos de los fármacos , Progesterona/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética
17.
J Am Chem Soc ; 130(36): 12148-55, 2008 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-18710224

RESUMEN

Cocaine is recognized as the most reinforcing of all drugs of abuse. There is no anticocaine medication available. The disastrous medical and social consequences of cocaine addiction have made the development of an anticocaine medication a high priority. It has been recognized that an ideal anticocaine medication is one that accelerates cocaine metabolism producing biologically inactive metabolites via a route similar to the primary cocaine-metabolizing pathway, i.e., cocaine hydrolysis catalyzed by plasma enzyme butyrylcholinesterase (BChE). However, wild-type BChE has a low catalytic efficiency against the abused cocaine. Design of a high-activity enzyme mutant is extremely challenging, particularly when the chemical reaction process is rate-determining for the enzymatic reaction. Here we report the design and discovery of a high-activity mutant of human BChE by using a novel, systematic computational design approach based on transition-state simulations and activation energy calculations. The novel computational design approach has led to discovery of the most efficient cocaine hydrolase, i.e., a human BChE mutant with an approximately 2000-fold improved catalytic efficiency, promising for therapeutic treatment of cocaine overdose and addiction as an exogenous enzyme in human. The encouraging discovery resulted from the computational design not only provides a promising anticocaine medication but also demonstrates that the novel, generally applicable computational design approach is promising for rational enzyme redesign and drug discovery.


Asunto(s)
Butirilcolinesterasa/química , Cocaína/química , Diseño de Fármacos , Hidrolasas/química , Animales , Butirilcolinesterasa/metabolismo , Cocaína/metabolismo , Simulación por Computador , Hidrolasas/metabolismo , Hidrolasas/farmacología , Cinética , Masculino , Ratones , Modelos Moleculares , Teoría Cuántica , Estereoisomerismo , Termodinámica
18.
Mol Carcinog ; 47(6): 466-77, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18058808

RESUMEN

Oncogenic Ras mutations are early genetic events in colorectal cancer that induce cyclooxygenase (COX)-2 expression and prostaglandin E(2) (PGE(2)) biosynthesis. PGE(2), a downstream product of COX-2, promotes cancer progression by modulating proliferation, apoptosis and angiogenesis. 15-hydroxyprostaglandin dehydrogenase (PGDH) degrades PGE(2) and is down-regulated in colorectal cancer, suggesting that PGDH plays a role in regulating PGE(2) levels and that PGDH over-expression could attenuate Ras-mediated tumorigenesis. Lentiviral transduction was used to express GFP (18.GFP), K-Ras(V12) (18.K-Ras(V12)), PGDH (18.PGDH) or both K-Ras(V12) and PGDH (18.K-Ras(V12).PGDH) in nontumorigenic rat intestinal epithelial (IEC-18) cells. 18.K-Ras(V12) cells exhibited increased phosphorylation of MAP kinases and CREB, proliferation rates, COX-2 and microsomal prostaglandin E synthase (mPGES)-1 expression and PGE(2) and PGI(2) levels. 18.PGDH and 18.K-Ras(V12).PGDH cells had 10(4)-fold increases in PGDH activity with decreased PGE(2) and PGI(2) levels, COX-2 and mPGES-1 expression and proliferation rates. 18.GFP, 18.PGDH, and 18.K-Ras(V12).PGDH cells were unable to grow in soft agar media whereas 18.K-Ras(V12) cells exhibited anchorage-independent cell growth. Xenografts of implanted 18.K-Ras(V12) cells in nu/nu mice produced rapid (2 wk) tumors with uniform antibody staining for COX-2 and mPGES-1 throughout the tumor and elevated PGE(2) levels. Xenografts of 18.K-Ras(V12).PGDH cells exhibited delayed (8 wk) tumor formation with negligible COX-2 and mPGES-1 expression and significantly decreased PGE(2) levels. 18.K-Ras(V12).PGDH tumors had decreased staining of the proliferative marker, Ki-67, and a significant increase in apoptosis in the central region of the tumor. Based on these data, we conclude that PGDH expression suppresses K-Ras(V12)-mediated tumorigenesis in intestinal epithelial cells.


Asunto(s)
Genes ras , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Neoplasias Experimentales/prevención & control , Animales , Secuencia de Bases , División Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Cartilla de ADN , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Fosforilación , Prostaglandinas/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Cancer Res ; 66(15): 7818-23, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16885386

RESUMEN

Prostaglandin E(2) plays a growth-stimulatory role in breast cancer, and the rate-limiting enzyme in its synthesis, cyclooxygenase-2, is often overexpressed in these cancers. Little is known about the role of the key prostaglandin catabolic enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in breast cancer pathogenesis. Using a pharmacologically based screen for epigenetically silenced genes, we found low levels of 15-PGDH in MDA-MB-231 cells [estrogen receptor (ER) negative] but high levels in MCF-7 cells (ER positive) and observed its up-regulation following demethylation treatment. Further analysis revealed methylation of the 15-PGDH promoter in one breast cancer cell line and 30% of primary tumors. Analysis of 15-PGDH expression revealed low levels in 40% of primary breast tumors and identified a correlation between 15-PGDH and ER expression. Transfection assays showed that transient up-regulation of 15-PGDH levels in MDA-MB-231 cells resulted in a decreased clonal growth, and stable up-regulation significantly decreased the ability of these cells to form tumors in athymic mice. In contrast, transient silencing of 15-PGDH in MCF-7 cells resulted in their enhanced proliferation, and a stable silencing in these cells enhanced cell cycle entry in vitro and tumorigenicity in vivo. Forced expression of 15-PGDH inhibited the ER pathway and silencing of 15-PGDH up-regulated expression of aromatase. In addition, 15-PGDH levels were down-regulated by estrogen but up-regulated by the tumor suppressor gene CAAT/enhancer binding protein alpha. Our results indicate for the first time that 15-PGDH may be a novel tumor suppressor gene in breast cancer, and suggest that this enzyme can modulate the ER pathway.


Asunto(s)
Neoplasias de la Mama/enzimología , Genes Supresores de Tumor , Hidroxiprostaglandina Deshidrogenasas/biosíntesis , Animales , Secuencia de Bases , Neoplasias de la Mama/genética , Procesos de Crecimiento Celular , Línea Celular Tumoral , Metilación de ADN , Regulación hacia Abajo , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Receptores de Estrógenos/biosíntesis , Transfección , Regulación hacia Arriba
20.
Arch Biochem Biophys ; 467(1): 20-30, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17880908

RESUMEN

Both 12-hydroxyheptadecatrienoic acid (12-HHT) and thromboxane A2 (TXA2) are products derived from prostaglandin H2 (PGH2) catalyzed by thromboxane synthase. Whether or not they exhibit similar actions remains to be determined. While TXA2-induced activation of extracellular signal-regulated kinases (ERKs) has been extensively studied, 12-HHT-induced activation of ERKs has not been explored. We reported for the first time that 12-HHT induced activation of ERKs in human prostate cancer cell line, PC3. We also compared the mechanisms of 12-HHT- and I-BOP-, a TXA2 mimetic, mediated ERK activation in PC3 cells. The activation of ERKs induced by either agent was shown to involve protein kinase C (PKC)-, protein kinase A (PKA)-, Src kinase and phosphoinositide-3 kinase (PI-3K)-dependent mechanisms in addition to the transactivation of the EGF receptor (EGFR) and the involvement of matrix metalloproteinases (MMPs) based on the sensitivity of the activation to their respective inhibitors. JNK/SAPK and p38 MAPK pathways were responsive to I-BOP but not to 12-HHT stimulation. Both 12-HHT- and I-BOP-induced activations of ERKs were also examined in other human prostate cancer cells, human lung cancer cells, and human lung fibroblast. I-BOP appeared to induce activation of ERKs in most cell lines, whereas 12-HHT induced activation of ERKs only in lung fibroblast in addition to PC3 cells. It appears that TPs are more generally expressed and the potential 12-HHT receptor (s) is expressed in limited specific cell types. Our results suggest that increased expression of thromboxane synthase as seen in prostate tumor may stimulate tumorigenesis as a consequence of concurrent increased synthesis of two fatty acids capable of activating ERKs.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ácidos Grasos Insaturados/farmacología , Regulación Enzimológica de la Expresión Génica , Bioquímica/métodos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Activación Enzimática , Receptores ErbB/metabolismo , Humanos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Modelos Biológicos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores de Tromboxanos/metabolismo , Tromboxano-A Sintasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA