Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Inflamm Res ; 69(1): 131-137, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31797003

RESUMO

OBJECTIVE: This study sought to evaluate short-term treatment with COX-2 inhibitors and acute changes in colonic PGE2 levels as predictors of long-term efficacy in a genetic model of colorectal cancer. METHODS: Celecoxib oral suspension (40 mg/kg BID) was dosed to Apc-mutant Pirc (F344/NTac-Apcam1137) rats for 4 days (short-term group), or the equivalent dose of 1500 ppm celecoxib was administered in the diet for 4 months (long-term group). Percent inhibition of colonic PGE2 was calculated, and the reduction in colonic PGE2 was assessed in relation to suppression of adenomatous colon polyps. RESULTS: Colonic mucosa PGE2 was fourfold higher in Pirc than in F344 wild-type rats (21 vs. 5.6 pg/mg epithelial tissue), due at least in part to higher COX-2 expression, and this was confirmed by elevated PGE2-d11 levels in Pirc colonic S9 incubations. In the 4-day study, dose-dependent reductions in PGE2 were observed in colonic epithelium (-33% (P>0.05) and -57% (P=0.0012)), after low- and high-dose celecoxib treatments of 4 mg/kg and 40 mg/kg (bid), respectively. In the 4-month study, 1500 ppm celecoxib suppressed colonic epithelium PGE2 by 43.5%, and tumor multiplicity by 80% (P<0.0015). Suppression of plasma 6-keto PGF1α also was corroborated following long-term treatment with 1500 ppm celecoxib (P<0.05). CONCLUSIONS: Acute changes in colonic mucosa PGE2 provided a rapid means of predicting long-term chemopreventive effects from celecoxib, and might be useful for screening of new COX-2 inhibitor compounds.


Assuntos
Celecoxib/farmacologia , Colo/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Animais , Biomarcadores/metabolismo , Celecoxib/uso terapêutico , Colo/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Ratos Endogâmicos F344 , Resultado do Tratamento
2.
J Cell Mol Med ; 23(12): 8343-8354, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31628732

RESUMO

Uncontrollable bleeding is still a worldwide killer. In this study, we aimed to investigate a novel approach to exhibit effective haemostatic properties, which could possibly save lives in various bleeding emergencies. According to the structure-based enzymatic design, we have engineered a novel single-chain hybrid enzyme complex (SCHEC), COX-1-10aa-TXAS. We linked the C-terminus of cyclooxygenase-1 (COX-1) to the N-terminus of the thromboxane A2 (TXA2 ) synthase (TXAS), through a 10-amino acid residue linker. This recombinant COX-1-10aa-TXAS can effectively pass COX-1-derived intermediate prostaglandin (PG) H2 (PGH2 ) to the active site of TXAS, resulting in an effective chain reaction property to produce the haemostatic prostanoid, TXA2 , rapidly. Advantageously, COX-1-10aa-TXAS constrains the production of other pro-bleeding prostanoids, such as prostacyclin (PGI2 ) and prostaglandin E2 (PGE2 ), through reducing the common substrate, PGH2 being passed to synthases which produce aforementioned prostanoids. Therefore, based on these multiple properties, this novel COX-1-10aa-TXAS indicated a powerful anti-bleeding ability, which could be used to treat a variety of bleeding situations and could even be useful for bleeding prone situations, including nonsteroidal anti-inflammatory drugs (NSAIDs)-resulted TXA2 -deficient and PGI2 -mediated bleeding disorders. This novel SCHEC has a great potential to be developed into a biological haemostatic agent to treat severe haemorrhage emergencies, which will prevent the complications of blood loss and save lives.


Assuntos
Aminoácidos/metabolismo , Ciclo-Oxigenase 1/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Tromboxano-A Sintase/metabolismo , Aminoácidos/genética , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Ciclo-Oxigenase 1/genética , Dinoprostona/metabolismo , Epoprostenol/metabolismo , Células HEK293 , Hemorragia/prevenção & controle , Hemostáticos/metabolismo , Hemostáticos/farmacologia , Humanos , Camundongos Transgênicos , Agregação Plaquetária/efeitos dos fármacos , Prostaglandina H2/metabolismo , Proteínas Recombinantes de Fusão/genética , Tromboxano A2/metabolismo , Tromboxano-A Sintase/genética
3.
Arch Biochem Biophys ; 616: 20-29, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28065721

RESUMO

Key residues and binding mechanisms of PGE1 and PGE2 on prostanoid receptors are poorly understood due to the lack of X-ray structures for the receptors. We constructed a human EP3 (hEP3) model through integrative homology modeling using the X-ray structure of the ß2-adrenergic receptor transmembrane domain and NMR structures of the thromboxane A2 receptor extracellular loops. PGE1 and PGE2 docking into the hEP3 model showed differing configurations within the extracellular ligand recognition site. While PGE2 could form possible binding contact with S211, PGE1 is unable to form similar contacts. Therefore, S211 could be the critical residue for PGE2 recognition, but is not a significant for PGE1. This prediction was confirmed using HEK293 cells transfected with hEP3 S211L cDNA. The S211L cells lost PGE2 binding and signaling. Interestingly, the S211L cells retained PGE1-mediated signaling. It indicates that S211 within the second extracellular loop is a key residue involved in turning down PGE2 signaling. Our study provided information that S211L within EP3 is the key residue to distinguish PGE1 and PGE2 binding to mediate diverse biological functions at the initial recognition step. The S211L mutant could be used as a model for studying the binding mechanism and signaling pathway specifically mediated by PGE1.


Assuntos
Alprostadil/química , Dinoprostona/química , Receptores de Prostaglandina E Subtipo EP3/química , Receptores de Prostaglandina E Subtipo EP3/genética , Sítios de Ligação , Sinalização do Cálcio , Cristalografia por Raios X , DNA Complementar/química , Células HEK293 , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Domínios Proteicos , Receptores Adrenérgicos beta 2/química , Proteínas Recombinantes/química , Transdução de Sinais
4.
Arch Biochem Biophys ; 603: 29-37, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27177970

RESUMO

Through linking inducible cyclooxygenase (COX)-2 with microsomal prostaglandin E2 (PGE2) synthase-1 (mPGES-1), a Single-Chain Enzyme Complex (SCEC, COX-2-10aa-mPGES-1) was engineered to mimic a specific inflammatory PGE2 biosynthesis from omega-6 fatty acid, arachidonic acid (AA), by eliminating involvements of non-inducible COX-1 and other PGE2 synthases. Using the SCEC, we characterized coupling reactions between COX-2 and mPGES-1 at 1:1 ratio of inflammatory PGE2 production. AA demonstrated two phase activities to regulate inflammatory PGE2 production. In the first phase (<2 µM), AA was a COX-2 substrate and converted to increasing production of PGE2. In the second phase with a further increased AA level (2-10 µM), AA bound to mPGES-1 and inhibited the PGE2 production. The SCEC study was identical to the co-expression of COX-2 and mPGES-1. This was further confirmed by using mPGES-1 and PGH2 as a direct enzyme target and substrate, respectively. Furthermore, the carboxylic acid group of AA binding to R67 and R70 of mPGES-1 was identified by X-ray structure-based docking and mutagenesis. mPGES-1 mutants, R70A, R70K, R67A and R67K, lost 40-100% binding to [(14)C]-AA. To conclude, a cellular model, in which AA is involved in self-controlling initial initiating and later resolving inflammation by its two phase activities, was discussed.


Assuntos
Ácido Araquidônico/química , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Prostaglandina H2/metabolismo , Prostaglandina-E Sintases/metabolismo , Anti-Inflamatórios não Esteroides/uso terapêutico , Domínio Catalítico , Cristalografia por Raios X , Ciclo-Oxigenase 2/genética , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Inflamação , Mutagênese Sítio-Dirigida , Prostaglandina-E Sintases/genética , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
Biochemistry ; 54(23): 3707-15, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-25988363

RESUMO

In vascular inflammation, prostaglandin E2 (PGE2) is largely biosynthesized by microsomal PGE2 synthase-1 (mPGES-1), competing with other downstream eicosanoid-synthesizing enzymes, such as PGIS, a synthase of a vascular protector prostacyclin (PGI2), to isomerize the cyclooxygenase (COX)-2-derived prostaglandin H2 (PGH2). In this study, we found that a majority of the product from the cells co-expressing human COX-2, mPGES-1, and PGIS was PGE2. We hypothesize that the molecular and cellular mechanisms are related to the post-translational endoplasmic reticulum (ER) arrangement of those enzymes. A set of fusion enzymes, COX-2-linker [10 amino acids (aa)]-PGIS and COX-2-linker (22 amino acids)-PGIS, were created as "The Bioruler", in which the 10 and 22 amino acids are defined linkers with known helical structures and distances (14.4 and 30.8 Å, respectively). Our experiments have shown that the efficiency of PGI2 biosynthesis was reduced when the separation distance increased from 10 to 22 amino acids. When COX-2-10aa-PGIS (with a 14.4 Å separation) was co-expressed with mPGES-1 on the ER membrane, a major product was PGE2, but not PGI2. However, expression of COX-2-10aa-PGIS and mPGES-1 on a separated ER with a distance of ≫30.8 Å reduced the level of PGE2 production. These data indicated that the mPGES-1 is "complex-likely" colocalized with COX-2 within a distance of 14.4 Å. In addition, the cells co-expressing COX-1-10aa-PGIS and mPGES-1 produced PGI2 mainly, but not PGE2. This indicates that mPGES-1 is expressed much farther from COX-1. These findings have led to proposed models showing the different post-translational ER organization between COX-2 and COX-1 with respect to the topological arrangement of the mPGES-1 during vascular inflammation.


Assuntos
Ácido Araquidônico/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Retículo Endoplasmático Liso/enzimologia , Oxirredutases Intramoleculares/metabolismo , Modelos Biológicos , Ciclo-Oxigenase 1/química , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/genética , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Dinoprostona/metabolismo , Retículo Endoplasmático Liso/metabolismo , Epoprostenol/metabolismo , Células HEK293 , Humanos , Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/genética , Peso Molecular , Oligopeptídeos/química , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Prostaglandina H2/metabolismo , Prostaglandina-E Sintases , Engenharia de Proteínas , Estrutura Secundária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
6.
Circulation ; 128(9): 982-94, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23841984

RESUMO

BACKGROUND: Intravenous prostacyclin is approved for treating pulmonary arterial hypertension (PAH), but it has a short half-life and must be delivered systemically via an indwelling intravenous catheter. We hypothesize that localized jugular vein delivery of prostacyclin-producing cells may provide sustained therapeutic effects without the limitations of systemic delivery. METHODS AND RESULTS: We generated a vector expressing a human cyclooxygenase isoform 1 and prostacyclin synthase fusion protein that produces prostacyclin from arachidonic acid. Endothelial-like progenitor cells (ELPCs) were transfected with the cyclooxygenase isoform 1-prostacyclin synthase plasmid and labeled with lentivirus expressing nuclear-localized red fluorescent protein (nuRFP). The engineered ELPCs (expressing cyclooxygenase isoform 1-prostacyclin synthase and nuRFP) were tested in rats with monocrotaline (MCT)-induced PAH. In PAH prevention studies, treatment with engineered ELPCs or control ELPCs (expressing nuRFP alone) attenuated MCT-induced right ventricular systolic pressure increase, right ventricular hypertrophy, and pulmonary vessel wall thickening. Engineered ELPCs were more effective than control ELPCs in all variables evaluated. In PAH reversal studies, engineered ELPCs or control ELPCs increased the survival rate of rats with established PAH and decreased right ventricular hypertrophy. Engineered ELPCs provided a survival benefit 2 weeks earlier than did control ELPCs. Microarray-based gene ontology analysis of the right ventricle revealed that a number of MCT-altered genes and neurotransmitter pathways (dopamine, serotonin, and γ-aminobutyric acid) were restored after ELPC-based prostacyclin gene therapy. CONCLUSIONS: Cyclooxygenase isoform 1-prostacyclin synthase-expressing ELPCs reversed MCT-induced PAH. A single jugular vein injection offered survival benefits for at least 4 weeks and may provide a promising option for PAH patients.


Assuntos
Células Endoteliais/transplante , Epoprostenol/genética , Terapia Genética , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/terapia , Hipertrofia Ventricular Direita/terapia , Monocrotalina/efeitos adversos , Transplante de Células-Tronco , Animais , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Modelos Animais de Doenças , Epoprostenol/metabolismo , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita/mortalidade , Hipertrofia Ventricular Direita/patologia , Infusões Intravenosas , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Masculino , Ratos , Ratos Endogâmicos F344 , Taxa de Sobrevida , Engenharia Tecidual , Transfecção , Resultado do Tratamento
7.
J Sex Med ; 10(6): 1476-87, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23551902

RESUMO

INTRODUCTION: Erectile dysfunction (ED) is a very common complication after radical prostatectomy. COX-2-10aa-PGIS is a newly engineered protein with COX-2 and prostacyclin synthase activities that converts arachidonic acid directly to prostacyclin (prostaglandin I2 [PGI2]). PGI2 is a potent smooth muscle relaxant. AIM: The purpose of this study was to explore the effect and mechanism of COX-2-10aa-PGIS gene therapy in penile rehabilitation. METHODS: Bilateral cavernous nerve crush (BCNC) in adult Sprague-Dawley rats was used to mimic radical prostatectomy-induced ED. Sprague-Dawley rats were randomly assigned into four groups: 1. sham surgery; 2. BCNC; 3. BCNC + null control recombinant adenovirus intracavernous injection; and 4. BCNC + Ad-COX2-10aa-PGIS intracavernous injection. Twenty-eight days later, intracavernosal pressure (ICP) was recorded under cavernous nerve stimulation; in the meantime, the mean arterial pressure (MAP) was monitored. At the end of the measurement, the penis was harvested and processed for (i) immunohistochemistry analysis of endothelial nitric oxide synthase (eNOS), alpha-smooth muscle actin (α-SMA), and transforming growth factor beta-1 (TGF-ß1); (ii) Masson's trichrome stain for smooth muscle/collagen ratios; (iii) Western blot of eNOS, α-SMA, TGF-ß1, and COX2-10aa-PGIS; and (iv) terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay for apoptosis. MAIN OUTCOME MEASURES: Erectile function was evaluated by ICP/MAP. Smooth muscle and endothelium functions in corpora cavernosum were assessed by Masson's trichrome stain, immunohistochemistry, and Western blot. Apoptosis was identified by TUNEL assay. RESULTS: The results were the following: 1. COX2-10aa-PGIS gene therapy improved erectile function (82%, compared with control) in the BCNC rat model; 2. COX2-10aa-PGIS gene therapy increased eNOS (121%) and α-SMA (118%) expression and decreased TGF-ß1 (45%) expression; 3. COX2-10aa-PGIS gene therapy reduced cell apoptosis after cavernous nerve injury (64%); and 4. COX2-10aa-PGIS gene therapy improved smooth muscle/collagen ratios (81%). CONCLUSION: Our data demonstrated that COX2-10aa-PGIS improved erectile function after cavernous nerve injury through antifibrotic and anti-apoptotic mechanisms.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Sistema Enzimático do Citocromo P-450/biossíntese , Disfunção Erétil/terapia , Terapia Genética/métodos , Oxirredutases Intramoleculares/biossíntese , Ereção Peniana , Pênis/irrigação sanguínea , Pênis/inervação , Actinas/metabolismo , Adenoviridae/genética , Animais , Apoptose , Ácido Araquidônico/metabolismo , Pressão Sanguínea , Ciclo-Oxigenase 2/genética , Sistema Enzimático do Citocromo P-450/genética , Modelos Animais de Doenças , Estimulação Elétrica , Epoprostenol/metabolismo , Vetores Genéticos , Oxirredutases Intramoleculares/genética , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Compressão Nervosa , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Fator de Crescimento Transformador beta1/metabolismo
8.
Future Med Chem ; 15(17): 1549-1552, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37702004

RESUMO

Tweetable abstract This work describes novel evidence of the relationship between NSAIDs and three prostaglandin E2 synthases.


Assuntos
Anti-Inflamatórios não Esteroides , Dinoprostona , Prostaglandina-E Sintases , Anti-Inflamatórios não Esteroides/farmacologia , Ciclo-Oxigenase 2
9.
Future Med Chem ; 15(9): 757-767, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37248701

RESUMO

Aim: The objective of this study was to synthesize and validate a set of compounds that selectively inhibit mPGES-1, with the potential to be developed into a novel anti-inflammatory drug. Methods: The synthesized compounds were characterized using 1H NMR spectroscopy and LC-MS to confirm their structure. Cellular and enzymatic assays were used to demonstrate their inhibitory activity on prostaglandin E2 production. Results: Docking studies revealed that compounds containing fluoro-, chloro- and methyl- groups displayed strong inhibitory activity against prostaglandin E2. The inhibitory activity of synthesized trimethyl and trifluoro was further validated using enzymatic and cell migration assays. Conclusion: The findings demonstrated that the synthesized compounds possess significant potential as a new generation of nonsteroidal anti-inflammatory drugs that selectively target mPGES-1 with fewer side effects.


Assuntos
Anti-Inflamatórios , Dinoprostona , Prostaglandina-E Sintases , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios não Esteroides/farmacologia
10.
Cells ; 12(24)2023 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-38132095

RESUMO

In this study, we reported that novel single-chain fusion proteins linking thromboxane A2 (TXA2) receptor (TP) to a selected G-protein α-subunit q (SC-TP-Gαq) or to α-subunit s (SC-TP-Gαs) could be stably expressed in megakaryocytes (MKs). We tested the MK-released platelet-linked particles (PLPs) to be used as a vehicle to deliver the overexpressed SC-TP-Gαq or the SC-TP-Gαs to regulate human platelet function. To understand how the single-chain TP-Gα fusion proteins could regulate opposite platelet activities by an identical ligand TXA2, we tested their dual functions-binding to ligands and directly linking to different signaling pathways within a single polypeptide chain-using a 3D structural model. The immature MKs were cultured and transfected with cDNAs constructed from structural models of the individual SC-TP-Gαq and SC-TP-Gαs, respectively. After transient expression was identified, the immature MKs stably expressing SC-TP-Gαq or SC-TP-Gαs (stable cell lines) were selected. The stable cell lines were induced into mature MKs which released PLPs. Western blot analysis confirmed that the released PLPs were carrying the recombinant SC-TP-Gαq or SC-TP-Gαs. Flow cytometry analysis showed that the PLPs carrying SC-TP-Gαq were able to perform the activity by promoting platelet aggregation. In contrast, PLPs carrying SC-TP-Gαs reversed Gq to Gs signaling to inhibit platelet aggregation. This is the first time demonstrating that SC-TP-Gαq and SC-TP-Gαs were successfully overexpressed in MK cells and released as PLPs with proper folding and programmed biological activities. This bio-engineering led to the formation of two sets of biologically active PLP forms mediating calcium and cAMP signaling, respectively. As a result, these PLPs are able to bind to identical endogenous TXA2 with opposite activities, inhibiting and promoting platelet aggregation as reprogrammed for therapeutic process. Results also demonstrated that the nucleus-free PLPs could be used to deliver recombinant membrane-bound GPCRs to regulate cellular activity in general.


Assuntos
Megacariócitos , Tromboxanos , Humanos , Megacariócitos/metabolismo , Preparações de Ação Retardada , Plaquetas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Tromboxano A2/metabolismo
11.
J Biol Chem ; 286(37): 32575-85, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21795694

RESUMO

Tumor suppressor protein p53, our most critical defense against tumorigenesis, can be made powerless by mechanisms such as mutations and inhibitors. Fortilin, a 172-amino acid polypeptide with potent anti-apoptotic activity, is up-regulated in many human malignancies. However, the exact mechanism by which fortilin exerts its anti-apoptotic activity remains unknown. Here we present significant insight. Fortilin binds specifically to the sequence-specific DNA binding domain of p53. The interaction of fortilin with p53 blocks p53-induced transcriptional activation of Bax. In addition, fortilin, but not a double point mutant of fortilin lacking p53 binding, inhibits p53-dependent apoptosis. Furthermore, cells with wild-type p53 and fortilin, but not cells with wild-type p53 and the double point mutant of fortilin lacking p53 binding, fail to induce Bax gene and apoptosis, leading to the formation of large tumor in athymic mice. Our results suggest that fortilin is a novel p53-interacting molecule and p53 inhibitor and that it is a logical molecular target in cancer therapy.


Assuntos
Apoptose , Biomarcadores Tumorais/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/genética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Mutação Puntual , Ligação Proteica , Transplante Heterólogo , Proteína Tumoral 1 Controlada por Tradução , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2/biossíntese , Proteína X Associada a bcl-2/genética
12.
J Cell Physiol ; 227(7): 2907-16, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21938725

RESUMO

Prostacyclin (PGI2) is a potent vasodilator and important mediator of vascular homeostasis; however, its clinical use is limited because of its short (<2-min) half-life. Thus, we hypothesize that the use of engineered endothelial progenitor cells (EPCs) that constitutively secrete high levels of PGI2 may overcome this limitation of PGI2 therapy. A cDNA encoding COX-1-10aa-PGIS, which links human cyclooxygenase-1 (COX-1) to prostacyclin synthase (PGIS), was delivered via nucleofection into outgrowth EPCs derived from rat bone marrow mononuclear cells. PGI2-secreting strains (PGI2-EPCs) were established by continuous subculturing of transfected cells under G418 selection. Genomic PCR, RT-PCR, and Western blot analyses confirmed the overexpression of COX-1-10aa-PGIS in PGI2-EPCs. PGI2-EPCs secreted significantly higher levels of PGI2 in vitro than native EPCs (P < 0.05) and showed higher intrinsic angiogenic capability; conditioned medium (CM) from PGI2-EPCs promoted better tube formation than CM from native EPCs (P < 0.05). Cell- and paracrine-mediated in vitro angiogenesis was attenuated when COX-1-10aa-PGIS protein expression was knocked down. Whole-cell patch-clamp studies showed that 4-aminopyridine-sensitive K(+) current density was increased significantly in rat smooth muscle cells (rSMCs) cocultured under hypoxia with PGI2-EPCs (7.50 ± 1.59 pA/pF; P < 0.05) compared with rSMCs cocultured with native EPCs (3.99 ± 1.26 pA/pF). In conclusion, we successfully created EPC strains that overexpress an active novel enzyme resulting in consistent secretion of PGI2. PGI2-EPCs showed enhanced intrinsic proangiogenic properties and provided favorable paracrine-mediated cellular protections, including promoting in vitro angiogenesis of native EPCs and hyperpolarization of SMCs under hypoxia.


Assuntos
Engenharia Celular/métodos , Endotélio Vascular/metabolismo , Epoprostenol/biossíntese , Epoprostenol/genética , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Células-Tronco/metabolismo , 4-Aminopiridina/metabolismo , Animais , Apoptose/genética , Processos de Crescimento Celular/genética , Meios de Cultivo Condicionados/metabolismo , Ciclo-Oxigenase 1/genética , Sistema Enzimático do Citocromo P-450/genética , DNA Complementar/genética , Endotélio Vascular/citologia , Epoprostenol/metabolismo , Meia-Vida , Hipóxia/genética , Hipóxia/metabolismo , Oxirredutases Intramoleculares/genética , Proteínas de Membrana/genética , Músculo Liso Vascular/citologia , Neovascularização Fisiológica , Fenótipo , Canais de Potássio/metabolismo , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção/métodos
13.
Biochemistry ; 50(10): 1691-9, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21250659

RESUMO

Prostacyclin (PGI(2)) is a key vascular protector, metabolized from endogenous arachidonic acid (AA). Its actions are mediated through the PGI(2) receptor (IP) and nuclear receptor, peroxisome proliferator-activated receptor γ (PPARγ). Here, we found that PGI(2) is involved in regulating cellular microRNA (miRNA) expression through its receptors in a mouse adipose tissue-derived primary culture cell line expressing a novel hybrid enzyme gene (COX-1-10aa-PGIS), cyclooxygenase-1 (COX-1) and PGI(2) synthase (PGIS) linked with a 10-amino acid linker. The triple catalytic functions of the hybrid enzyme in these cells successfully redirected the endogenous AA metabolism toward a stable and dominant production of PGI(2). The miRNA microarray analysis of the cell line with upregulated PGI(2) revealed a significant upregulation (711, 148b, and 744) and downregulation of miRNAs of interest, which were reversed by antagonists of the IP and PPARγ receptors. Furthermore, we also found that the insulin-mediated lipid deposition was inhibited in the PGI(2)-upregulated adipocytes. The study also initiated a discussion that suggested that the endogenous PGI(2) inhibition of lipid deposition in adipocytes could involve miRNA-mediated inhibition of expression of the targeted genes. This indicated that PGI(2)-miRNA regulation could exist in broad pathophysiological processes involving PGI(2) (i.e., apoptosis, vascular inflammation, cancer, embryo implantation, and obesity).


Assuntos
Adipócitos/metabolismo , Regulação para Baixo , Epoprostenol/metabolismo , MicroRNAs/genética , Regulação para Cima , Animais , Células Cultivadas , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-akt/genética
14.
BMC Complement Altern Med ; 11: 11, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21299883

RESUMO

BACKGROUND: Conventionally the active ingredients in herbal extracts are separated into individual components, by fractionation, desalting, and followed by high-performance liquid chromatography (HPLC). In this study we have tried to directly screen water-soluble fractions of herbs with potential active ingredients before purification or extraction. We propose that the herbal extracts mimicking prostaglandin E(1) (PGE(1)) and E(2) (PGE(2)) can be identified in the water-soluble non-purified fraction. PGE(1) is a potent anti-inflammatory molecule used for treating peripheral vascular diseases while PGE(2) is an inflammatory molecule. METHODS: We used cell-based assays (CytoFluor multi-well plate reader and fluorescence microscopy) in which a calcium signal was generated by the recombinant EP(1) receptor stably expressed in HEK293 cells (human embryonic kidney). PGE(1) and PGE(2) were tested for their ability to generate a calcium signal. Ninety-six water soluble fractions of Treasures of the east (single Chinese herb dietary supplements) were screened. RESULTS: After screening, the top ten stimulators were identified. The identified herbs were then desalted and the calcium fluorescent signal reconfirmed using fluorescence microscopy. Among these top ten agonists identified, seven stimulated the calcium signaling (1-40 µM concentration) using fluorescence microscopy. CONCLUSIONS: Fluorescence microscopy and multi-well plate readers can be used as a target specific method for screening water soluble fractions with active ingredients at a very early stage, before purification. Our future work consists of purifying and separating the active ingredients and repeating fluorescence microscopy. Under ordinary circumstances we would have to purify the compounds first and then test all the extracts from 96 herbs. Conventionally, for screening natural product libraries, the procedure followed is the automated separation of all constituents into individual components using fractionation and high performance liquid chromatography. We, however, demonstrated that the active ingredients of the herbal extracts can be tested before purification using an agonist sensitive, quick and simple cell-based signaling assay for ligands mimicking the agonists, PGE(1) and PGE(2).


Assuntos
Alprostadil/agonistas , Dinoprostona/agonistas , Descoberta de Drogas/métodos , Medicamentos de Ervas Chinesas/farmacologia , Receptores de Prostaglandina E/agonistas , Transdução de Sinais , Cálcio/metabolismo , Medicamentos de Ervas Chinesas/química , Células HEK293 , Humanos , Ligantes , Microscopia de Fluorescência , Proteínas Recombinantes
15.
Future Med Chem ; 13(13): 1091-1103, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34080888

RESUMO

Aim: This study investigated our Enzymelinks, COX-2-10aa-mPGES-1 and COX-2-10aa-PGIS, as cellular cross-screening targets for quick identification of lead compounds to inhibit inflammatory PGE2 biosynthesis while maintaining prostacyclin synthesis. Methods: We integrated virtual and wet cross-screening using Enzymelinks to rapidly identify lead compounds from a large compound library. Results: From 380,000 compounds virtually cross-screened with the Enzymelinks, 1576 compounds were identified and used for wet cross-screening using HEK293 cells that overexpressed individual Enzymelinks as targets. The top 15 lead compounds that inhibited mPGES-1 activity were identified. The top compound that specifically inhibited inflammatory PGE2 biosynthesis alone without affecting COX-2 coupled to PGI2 synthase (PGIS) for PGI2 biosynthesis was obtained. Conclusion: Enzymelink technology could advance cyclooxygenase pathway-targeted drug discovery to a significant degree.


Assuntos
Derivados de Benzeno/farmacologia , Ciclo-Oxigenase 1/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Oxirredutases Intramoleculares/metabolismo , Engenharia de Proteínas , Derivados de Benzeno/química , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Microssomos/efeitos dos fármacos , Microssomos/enzimologia
16.
Biochemistry ; 49(30): 6365-74, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20590159

RESUMO

The human thromboxane A(2) (TXA(2)) receptor (TP) is known to mediate platelet aggregation and vasoconstriction. The receptor predominantly interacts with the Gq protein, thereby activating phospholipase C and increasing the intracellular calcium level. In this study, we synthesized a 15-residue peptide corresponding to the C-terminal domain of the Gq protein alpha subunit (Galphaq-Ct peptide) and characterized its interaction with recombinant TP purified from a baculovirus expression system in the presence and absence of an agonist using fluorescence and NMR spectroscopic studies. With fluorescence binding assays, we demonstrated that the Galphaq-Ct peptide was bound to TP, in the absence of the agonist, with a K(d) value of approximately 17 muM. Interestingly, upon addition of the agonist, U46619, the Galphaq-Ct peptide's binding affinity for this activated TP was reduced, thereby increasing the K(d) value to approximately 240 muM. NMR experiments demonstrated that the TP-bound Galphaq-Ct peptide shows a different affinity and conformation, in the absence and presence of the agonist, U46619. This suggested there is the possibility of ligand-free constitutive TP signaling through Galpha binding. Thus, an HEK293 cell line that stably expresses human TP and lacks the ability to produce TXA(2) was created by gene transfer and G418 selection. In comparison with the control cells, the stable cell line showed significant Galpha-mediated ligand-free calcium signaling. The study indicates a promising new outlook for the examination of prostanoid receptor-G-protein interactions in greater detail using integrated NMR spectroscopy, the purified receptor, and the stable cell line.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Sinalização do Cálcio , Linhagem Celular , Humanos , Espectroscopia de Ressonância Magnética , Ligação Proteica , Conformação Proteica , Receptores de Prostaglandina , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Espectrometria de Fluorescência
17.
J Neuroimmune Pharmacol ; 15(2): 292-308, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31897976

RESUMO

Cellular arachidonic acid (AA), an unsaturated fatty acid found ubiquitously in plasma membranes, is metabolized to different prostanoids, such as prostacyclin (PGI2) and prostaglandin E2 (PGE2), by the three-step reactions coupling the upstream cyclooxygenase (COX) isoforms (COX-1 and COX-2) with the corresponding individual downstream synthases. While the vascular actions of these prostanoids are well-characterized, their specific roles in the hippocampus, a major brain area for memory, are poorly understood. The major obstacle for its understanding in the brain was to mimic the biosynthesis of each prostanoid. To solve the problem, we utilized Single-Chain Hybrid Enzyme Complexes (SCHECs), which could successfully control cellular AA metabolites to the desired PGI2 or PGE2. Our in vitro studies suggested that neurons with higher PGI2 content and lower PGE2 content exhibited survival protection and resistance to Amyloid-ß-induced neurotoxicity. Further extending to an in vivo model, the hybrid of PGI2-producing transgenic mice and Alzheimer's disease (AD) mice showed restored long-term memory. These findings suggested that the vascular prostanoids, PGI2 and PGE2, exerted significant regulatory influences on neuronal protection (by PGI2), or damage (by PGE2) in the hippocampus, and raised a concern that the wide uses of aspirin in cardiovascular diseases may exert negative impacts on neurodegenerative protection. Graphic Abstract Our study intended to understand the crosstalk of prostanoids in the hippocampus, a major brain area impacted in AD, by using hybrid enzymes to redirect the synthesis of prostanoids to PGE2 and PGI2, respectively. Our data indicated that during inflammation, the vascular mediators, PGI2 and PGE2, exerted significant regulatory influences on neuronal protection (by PGI2), or damage (by PGE2) in the hippocampus. These findings also raised a concern that the widely uses of non-steroidal anti-inflammatory drugs in cardiovascular diseases may exert negative impacts on neurodegenerative protection.


Assuntos
Epoprostenol/biossíntese , Hipocampo/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Regulação para Cima/fisiologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Epoprostenol/genética , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Iloprosta/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Regulação para Cima/efeitos dos fármacos
18.
Biochemistry ; 48(14): 3157-65, 2009 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-19170518

RESUMO

The binding of an agonist to a G protein-coupled receptor (GPCR) causes its coupling to different G proteins, which mediate signaling. However, the binding of an antagonist to the same site of the GPCR could not induce coupling. To understand the molecular mechanism involved, the structural flexibility of the purified human thromboxane A2 receptor (TP) was characterized by spectroscopic approaches, while bound to an agonist or antagonist. Circular dichroism not only revealed that the purified TP adopted more than 50% helical conformation in solution but also showed that the antagonist, SQ29,548, could induce more of a beta-sheet structure in the TP than that of the agonist, U46619. Also, fluorescence studies showed that the antagonist induced the intrinsic Trp fluorescence signal change more than the agonist. Furthermore, three of the nine tryptophan residues involved in the different ligand-based structural changes were demonstrated by NMR spectroscopy. Low pH-induced changes in the receptor conformation and molecular interaction field dramatically increased the agonist binding but did not significantly affect the antagonist binding. Different conformational changes were also observed in the TP reconstituted into phosphatidylcholine/phosphatidylserine/phosphatydylethanolamine-formed liposomes. These studies are the first to show a possible mechanism of the ligand-specific conformation-dependent agonist activation and antagonist blockage in the GPCR.


Assuntos
Receptores de Tromboxano A2 e Prostaglandina H2/química , Dicroísmo Circular , Humanos , Concentração de Íons de Hidrogênio , Ligantes , Lipossomos , Espectroscopia de Ressonância Magnética , Fosfolipídeos , Conformação Proteica , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/química , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Receptores de Tromboxano A2 e Prostaglandina H2/antagonistas & inibidores , Triptofano
19.
J Sex Med ; 6 Suppl 3: 328-33, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19267856

RESUMO

INTRODUCTION: Erectile dysfunction (ED) after radical prostatectomy (RP) is a very common condition. Prostacyclin (PGI(2)) is a prostaglandin with properties of vasodilation and anti-platelet aggregation. SuperEnzyme is a newly engineered protein with PGI(2) synthase activity that converts arachidonic acid directly to PGI(2). Transfection of SuperEnzyme into the penis to generate high levels of PGI(2) may increase penile blood inflow, alleviate hypoxia, and prevent apoptosis and fibrosis with potential use for ED after RP. AIM: The pathophysiology of ED after RP and the prostaglandin regulation was reviewed, and the possibly relevant mechanism of SuperEnzyme as a therapy for ED after RP was proposed. MAIN OUTCOME MEASURE: The rationale for SuperEnzyme as a possible therapy for ED after RP is analyzed. METHODS: We reviewed the publications on the proposed pathophysiology of ED after RP, the molecular regulation of prostaglandin and methods of SuperEnzyme engineering and transfection. RESULTS: ED after RP is involved in hypoxia, apoptosis and fibrosis, mainly due to the cavernosal nerve injury. Transfection of SuperEnzyme into the penis of an animal model to produce PGI(2) is feasible. Animal studies with the use of SuperEnzyme gene therapy are needed to provide new insight into metabolic and signaling pathways of PGI(2) in the penis and the role of PGI(2) signaling in the recovery of erectile function after RP. CONCLUSION: SuperEnzyme may be a potential candidate as a gene therapy for ED after RP.


Assuntos
Epoprostenol/biossíntese , Disfunção Erétil/fisiopatologia , Disfunção Erétil/terapia , Terapia Genética/métodos , Pênis/fisiopatologia , Disfunção Erétil/etiologia , Estudos de Viabilidade , Humanos , Masculino , Pênis/metabolismo , Complicações Pós-Operatórias , Prostatectomia
20.
Biochemistry ; 47(26): 6819-26, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18529068

RESUMO

Human thromboxane A2 receptor (TP), a G protein-coupled receptor (GPCR), is one of the most promising targets for developing the next generation of anti-thrombosis and hypertension drugs. However, obtaining a sufficient amount of the full-sized and active membrane protein has been the major obstacle for structural elucidation that reveals the molecular mechanisms of the receptor activation and drug designs. Here we report an approach for the simple, quick, and high-yield preparation of the purified and active full-sized TP in an amount suitable for structural studies. Glycosylated human TP was highly expressed in Sf-9 cells using an optimized baculovirus (BV) expression system. The active receptor was extracted and solubilized by different detergents for comparison and was finally purified to a nearly single band with a ratio of 1:0.9 +/- 0.05 (ligand:receptor molecule) in ligand binding using a Ni column with a relatively low yield. However, a high-yield purification (milligram quantity) of the TP protein, from a modulate scale of transfected Sf-9 cell culture, has been achieved by quick and simple purification steps, which include DNA digestion, dodecyl-maltoside detergent extraction, centrifugation, and FPLC purification. The purity and quantity of the purified TP, using the high-yield approach, were suitable for protein structural studies as evidenced by SDS-PAGE, Western blot analyses, ligand binding assays, and a feasibility test using high-resolution one-dimensional and two-dimensional (1)H NMR spectroscopic analyses. These studies provide a basis for the high-yield expression and purification of the GPCR for the structural and functional characterization using biophysics approaches.


Assuntos
Receptores de Tromboxano A2 e Prostaglandina H2/química , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Animais , Baculoviridae/genética , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , DNA Complementar/genética , Expressão Gênica , Vetores Genéticos/genética , Humanos , Ligantes , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Processamento de Proteína Pós-Traducional , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Receptores de Tromboxano A2 e Prostaglandina H2/isolamento & purificação , Spodoptera , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA