Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
Cells ; 12(24)2023 12 06.
Article En | MEDLINE | ID: mdl-38132095

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.


Megakaryocytes , Thromboxanes , Humans , Megakaryocytes/metabolism , Delayed-Action Preparations , Blood Platelets/metabolism , GTP-Binding Proteins/metabolism , Thromboxane A2/metabolism
2.
Future Med Chem ; 15(17): 1549-1552, 2023 09.
Article En | MEDLINE | ID: mdl-37702004

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


Anti-Inflammatory Agents, Non-Steroidal , Dinoprostone , Prostaglandin-E Synthases , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 2
3.
Future Med Chem ; 15(9): 757-767, 2023 05.
Article En | MEDLINE | ID: mdl-37248701

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.


Anti-Inflammatory Agents , Dinoprostone , Prostaglandin-E Synthases , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology
4.
FASEB Bioadv ; 4(12): 758-774, 2022 Dec.
Article En | MEDLINE | ID: mdl-36479208

The ß2AR is a prototypical G protein-coupled receptor (GPCR) known to orchestrate different cellular responses by the stimulation of specific signaling pathways. The best-established signaling pathways for the ß2AR are the canonical Gs pathway and the alternative ß arrestin 2 (ßarr2) pathway. Exploring each pathway separately remains a challenging task due to the dynamic nature of the receptor. Here, we fused the ß2AR with its cognate transducers, Gαs and ßarr2, using short linkers as a novel approach for restricting the conformation of the receptor and preferentially activating one of its two signaling pathways. We characterized the behavior of our fusion proteins ß2AR-Gαs and ß2AR-ßarr2 in HEK293 cells by measuring their constitutive activity, transducer recruitment, and pharmacological modulation. Our fusion proteins show (a) steric hindrance from the reciprocal endogenous transducers, (b) constitutive activity of the ß2AR for the signaling pathway activated by the tethered transducer, and (c) pharmacologic modulation by ß2AR ligands. Based on these characteristics, we further explored the possibility of a gain-of-function mechanism in the human lung non-tumorigenic epithelial cell line, BEAS-2B cells. This immortalized human bronchial epithelial cell line has immunomodulatory properties through cytokine release mediated by ß2AR stimulation. Our findings suggest that each signaling pathway of the ß2AR is biased toward either the Th1 or Th2 inflammatory response suggesting a role in regulating the immune phenotype of respiratory diseases. Our data imply that our fusion proteins can be used as tools to isolate the function elicited by a single signaling pathway in physiologically relevant cell types.

7.
Future Med Chem ; 13(13): 1091-1103, 2021 07.
Article En | MEDLINE | ID: mdl-34080888

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.


Benzene Derivatives/pharmacology , Cyclooxygenase 1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Intramolecular Oxidoreductases/metabolism , Protein Engineering , Benzene Derivatives/chemistry , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Microsomes/drug effects , Microsomes/enzymology
9.
J Neuroimmune Pharmacol ; 15(2): 292-308, 2020 06.
Article En | MEDLINE | ID: mdl-31897976

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.


Epoprostenol/biosynthesis , Hippocampus/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Up-Regulation/physiology , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Epoprostenol/genetics , Hippocampus/drug effects , Hippocampus/pathology , Iloprost/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Up-Regulation/drug effects
10.
Inflamm Res ; 69(1): 131-137, 2020 Jan.
Article En | MEDLINE | ID: mdl-31797003

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.


Celecoxib/pharmacology , Colon/drug effects , Colorectal Neoplasms/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Animals , Biomarkers/metabolism , Celecoxib/therapeutic use , Colon/metabolism , Colorectal Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/therapeutic use , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Rats, Inbred F344 , Treatment Outcome
11.
J Cell Mol Med ; 23(12): 8343-8354, 2019 12.
Article En | MEDLINE | ID: mdl-31628732

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.


Amino Acids/metabolism , Cyclooxygenase 1/metabolism , Recombinant Fusion Proteins/metabolism , Thromboxane-A Synthase/metabolism , Amino Acids/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase 1/genetics , Dinoprostone/metabolism , Epoprostenol/metabolism , HEK293 Cells , Hemorrhage/prevention & control , Hemostatics/metabolism , Hemostatics/pharmacology , Humans , Mice, Transgenic , Platelet Aggregation/drug effects , Prostaglandin H2/metabolism , Recombinant Fusion Proteins/genetics , Thromboxane A2/metabolism , Thromboxane-A Synthase/genetics
13.
Cancer Res ; 78(19): 5656-5667, 2018 10 01.
Article En | MEDLINE | ID: mdl-30217928

Overexpression and activation of the murine double minute 2 (MDM2) or nuclear factor of activated T cells 1 (NFAT1) oncoproteins frequently occur in pancreatic cancer. Most MDM2 inhibitors under development target MDM2-p53 binding and have little or no effect on cancers without functional p53, including pancreatic cancer. Some available compounds indirectly inhibit NFAT1 activity by interfering with calcineurin activity, but there are currently no specific inhibitors against NFAT1. Here we performed a high-throughput virtual and cell-based screening to yield a lead compound (MA242) that can directly bind both MDM2 and NFAT1 with high affinity, induce their protein degradation, and inhibit NFAT1-mediated transcription of MDM2 As a result of this binding, MA242 decreased cell proliferation and induced apoptosis in pancreatic cancer cell lines regardless of p53 status. MA242 alone or in combination with gemcitabine inhibited pancreatic tumor growth and metastasis without any host toxicity. Our data indicate that targeting both MDM2 and NFAT1 represents a novel and effective strategy to treat pancreatic cancer.Significance: These findings suggest that pharmacological inhibition of both MDM2 and NFAT1 is a promising strategy for the treatment of pancreatic cancer, even in tumors lacking functional p53. Cancer Res; 78(19); 5656-67. ©2018 AACR.


Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Heterocyclic Compounds, 3-Ring/pharmacology , NFATC Transcription Factors/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Animals , Apoptosis , Calcineurin/metabolism , Cell Line, Tumor , Cell Proliferation , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Discovery , Drug Screening Assays, Antitumor , Female , Mice , Mice, Nude , Neoplasm Transplantation , Protein Binding , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism , Gemcitabine
15.
Sci Rep ; 8(1): 1653, 2018 01 26.
Article En | MEDLINE | ID: mdl-29374184

Vascular prostanoids, isomerized from an intermediate prostaglandin (PG), H2, produced by cyclooxygenase (COX), exert various effects on the vascular system, both protective and destructive. During endothelial dysfunction, vascular protector prostacyclin/prostaglandin I2 (PGI2) is decreased, while inflammatory PGE2 and thrombotic TXA2 are increased. Therefore, our research aim was to reverse the event by controlling PGH2 metabolism by generating an in vivo model via enzymatic engineering of COX-1 and prostacyclin synthase (PGIS). The COX-1 and PGIS genes were linked to a 10-residue amino acid linker to form a Single-chain Enzyme Complex (SCHEC), COX-1-10aa-PGIS. Transgenic (CP-Tg) mice in a FVB/N background were generated using the pronuclear microinjection method. We first confirmed mRNA and protein expression of COX-1-10aa-PGIS in various CP-Tg mouse tissues, as well as upregulation of circulating PGI2. We then examined the cardiovascular function of these mice. Our CP-Tg mice exhibited marked resistance to vascular assault through induced carotid arterial blockage, acute thrombotic stroke and arterial arrest, angiotensin-induced peripheral vasoconstriction, and hepatic lipid accumulation after receiving a high-fat diet. They also had a longer lifespan compared with wild-type mice. This study raises the possibility of fighting cardiovascular diseases by regulating cellular arachidonic acid-derived PGH2 metabolites using enzymatic engineering.


Disease Models, Animal , Disease Resistance , Myocardial Infarction/pathology , Stroke/pathology , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Mice , Mice, Transgenic , Myocardial Infarction/prevention & control
16.
Arch Biochem Biophys ; 616: 20-29, 2017 02 15.
Article En | MEDLINE | ID: mdl-28065721

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.


Alprostadil/chemistry , Dinoprostone/chemistry , Receptors, Prostaglandin E, EP3 Subtype/chemistry , Receptors, Prostaglandin E, EP3 Subtype/genetics , Binding Sites , Calcium Signaling , Crystallography, X-Ray , DNA, Complementary/chemistry , HEK293 Cells , Humans , Ligands , Magnetic Resonance Spectroscopy , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Domains , Receptors, Adrenergic, beta-2/chemistry , Recombinant Proteins/chemistry , Signal Transduction
19.
Arch Biochem Biophys ; 603: 29-37, 2016 08 01.
Article En | MEDLINE | ID: mdl-27177970

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.


Arachidonic Acid/chemistry , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Prostaglandin H2/metabolism , Prostaglandin-E Synthases/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Catalytic Domain , Crystallography, X-Ray , Cyclooxygenase 2/genetics , Dose-Response Relationship, Drug , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Inflammation , Mutagenesis, Site-Directed , Prostaglandin-E Synthases/genetics , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Nat Commun ; 7: 11276, 2016 Apr 15.
Article En | MEDLINE | ID: mdl-27080438

Promoting the paracrine effects of human mesenchymal stem cell (hMSC) therapy may contribute to improvements in patient outcomes. Here we develop an innovative strategy to enhance the paracrine effects of hMSCs. In a mouse hindlimb ischaemia model, we examine the effects of hMSCs in which a novel triple-catalytic enzyme is introduced to stably produce prostacyclin (PGI2-hMSCs). We show that PGI2-hMSCs facilitate perfusion recovery and enhance running capability as compared with control hMSCs or iloprost (a stable PGI2 analogue). Transplanted PGI2-hMSCs do not incorporate long term into host tissue, but rather they mediate host regeneration and muscle mass gain in a paracrine manner. Mechanistically, this involves long noncoding RNA H19 in promoting PGI2-hMSC-associated survival and proliferation of host progenitor cells under hypoxic conditions. Together, our data reveal the novel ability of PGI2-hMSCs to stimulate host regenerative processes and improve physical function by regulating long noncoding RNA in resident progenitor cells.


Epoprostenol/metabolism , Hindlimb/blood supply , Ischemia/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , RNA, Long Noncoding/genetics , Animals , Cell Line , Cells, Cultured , Combined Modality Therapy , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , HEK293 Cells , Humans , Iloprost/pharmacology , Mice, Inbred NOD , Mice, SCID , Motor Activity , Running , Transplantation, Heterologous , Treatment Outcome , Vasodilator Agents/pharmacology
...