Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Cell Mol Med ; 23(12): 8343-8354, 2019 12.
Article in English | MEDLINE | ID: mdl-31628732

ABSTRACT

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.


Subject(s)
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
2.
Sci Rep ; 8(1): 1653, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374184

ABSTRACT

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.


Subject(s)
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
3.
Biochemistry ; 54(23): 3707-15, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-25988363

ABSTRACT

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.


Subject(s)
Arachidonic Acid/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Endoplasmic Reticulum, Smooth/enzymology , Intramolecular Oxidoreductases/metabolism , Models, Biological , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/genetics , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dinoprostone/metabolism , Endoplasmic Reticulum, Smooth/metabolism , Epoprostenol/metabolism , HEK293 Cells , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/genetics , Molecular Weight , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Prostaglandin H2/metabolism , Prostaglandin-E Synthases , Protein Engineering , Protein Structure, Secondary , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
4.
Life Sci ; 116(1): 43-50, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25139833

ABSTRACT

AIM: Many cancers originate and flourish in a prolonged inflammatory environment. Our aim is to understand the mechanisms of how the pathway of prostaglandin E2 (PGE2) biosynthesis and signaling can promote cancer growth in inflammatory environment at cellular and animal model levels. MAIN METHODS: In this study, a chronic inflammation pathway was mimicked with a stable cell line that over-expressed a novel human enzyme consisting of cyclooxygenase isoform-2 (COX-2) linked to microsomal (PGE2 synthase-1 (mPGES-1)) for the overproduction of pathogenic PGE2. This PGE2-producing cell line was co-cultured and co-implanted with three human cancer cell lines including prostate, lung, and colon cancers in vitro and in vivo, respectively. KEY FINDINGS: Increases in cell doubling rates for the three cancer cell types in the presence of the PGE2-producing cell line were clearly observed. In addition, one of the four human PGE2 subtype receptors, EP1, was used as a model to identify PGE2-signaling involved in promoting the cancer cell growth. This finding was further proven in vivo by co-implanting the PGE2-producing cells line and the EP1-positive cancer cells into the immune deficient mice, after that, it was observed that the PGE2-producing cells promoted all three types of cancer formation in the mice. SIGNIFICANCE: This study clearly demonstrated that the human COX-2 linked to mPGES-1 is a pathway that, when mediated by the EP, is linked to promoting cancer growth in a chronic inflammatory environment. The identified pathway could be used as a novel target for developing and advancing anti-inflammation and anti-cancer interventions.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Inflammation/pathology , Intramolecular Oxidoreductases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Colonic Neoplasms/pathology , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microsomes/metabolism , Prostaglandin-E Synthases , Prostatic Neoplasms/pathology , Receptors, Prostaglandin E/metabolism
5.
Circulation ; 128(9): 982-94, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23841984

ABSTRACT

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.


Subject(s)
Endothelial Cells/transplantation , Epoprostenol/genetics , Genetic Therapy , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/therapy , Hypertrophy, Right Ventricular/therapy , Monocrotaline/adverse effects , Stem Cell Transplantation , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Epoprostenol/metabolism , Familial Primary Pulmonary Hypertension , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/mortality , Hypertrophy, Right Ventricular/pathology , Infusions, Intravenous , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Male , Rats , Rats, Inbred F344 , Survival Rate , Tissue Engineering , Transfection , Treatment Outcome
7.
Int J Cardiol ; 163(2): 163-9, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-21722977

ABSTRACT

BACKGROUND: For decades, there have been many ongoing attempts to use prostaglandin I(2) (PGI(2)) to treat heart diseases, such as pulmonary arterial hypertension. However, the short half life of PGI(2) has limited the therapeutic impact potential. METHODS: Here, we have engineered a novel adipose tissue-derived cell that constantly produces PGI(2,) through transfecting of an engineered cDNA of a hybrid enzyme (human COX-1-10-aa-PGIS) which has superior triple catalytic functions in directly converting arachidonic acid into PGI(2). RESULTS: The gene-transfected cells were further converted into a stable cell line, in which cells constantly express the hybrid enzyme and are capable of producing large-amounts of PGI(2). In a comparison between un-transfected- and gene-transfected cells, it was determined that the majority of the endogenous AA metabolism shifted from that of unwanted PGE(2) (in un-transfected cells) to that of the preferred PGI(2) (in gene-transfected cells) with a PGI(2)/PGE(2) ratio change from 0.03 to 25. The PGI(2)-producing cell line not only exhibited an approximate 50-fold increase in PGI(2) biosynthesis, but also demonstrated superior anti-platelet aggregation in vitro, and increased reperfusion in the mouse ischemic hindlimb model in vivo. CONCLUSIONS: The cells, which have an ability to increase the biosynthesis of the vascular protector, PGI(2), while reducing that of the vascular inflammatory mediator, PGE(2), provide a dual effect on vascular protection, which is not available through any existing drug treatments. Thus, the current finding has potential to be an experimental intervention for PGI(2)-deficient heart diseases, such as pulmonary arterial hypertension.


Subject(s)
Cells, Cultured/metabolism , Epoprostenol/biosynthesis , Heart Diseases/drug therapy , Animals , Cyclooxygenase 1/biosynthesis , Humans , Mice , Transfection
8.
BMC Complement Altern Med ; 12: 143, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22938033

ABSTRACT

BACKGROUND: The molecular mechanisms of dietary oils (such as fish oil) and unsaturated fatty acids, which are widely used by the public for anti-inflammation and vascular protection, have not been settled yet. In this study, prostaglandin E(2) (PGE(2))-mediated calcium signaling was used to screen dietary oils and eight unsaturated fatty acids for identification of their anti-inflammatory mechanisms. Isolated fat/stromal cells expressing endogenous PGE(2) receptors and an HEK293 cell line specifically expressing the recombinant human PGE(2) receptor subtype-1 (EP(1)) were cultured and used in live cell calcium signaling assays. The different dietary oils and unsaturated fatty acids were used to affect cell signaling under the specific stimulation of a pathological amount of inflammatory PGE(2). RESULTS: It was identified that fish oil best inhibited the PGE(2) signaling in the primary cultured stromal cells. Second, docosahexaenoic acid (DHA), found in abundance in fish oil, was identified as a key factor of inhibition of PGE(2) signaling. Eicosapentaenoic acid (EPA), another major fatty acid found in fish oil and tested in this study was found to have small effect on EP(1) signaling. The study suggested one of the four PGE(2) subtype receptors, EP(1) as the key target for the fish oil and DHA target. These findings were further confirmed by using the recombinant EP(1) expressed in HEK293 cells as a target. CONCLUSION: This study demonstrated the new mechanism behind the positive effects of dietary fish oils in inhibiting inflammation originates from the rich concentration of DHA, which can directly inhibit the inflammatory EP(1)-mediated PGE(2) receptor signaling, and that the inflammatory response stimulated by PGE(2) in the fat stromal cells, which directly related to metabolic diseases, could be down regulated by fish oil and DHA. These findings also provided direct evidence to support the use of dietary oils and unsaturated fatty acids for protection against heart disease, pain, and cancer resulted from inflammatory PGE(2).


Subject(s)
Anti-Inflammatory Agents/chemistry , Dietary Fats, Unsaturated/analysis , Dinoprostone/immunology , Fat Body/cytology , Fatty Acids, Unsaturated/chemistry , Inflammation/metabolism , Stromal Cells/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Calcium/metabolism , Cells, Cultured , Dietary Fats, Unsaturated/metabolism , Digestion , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacology , Down-Regulation/drug effects , Fat Body/drug effects , Fat Body/metabolism , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacology , Fish Oils/chemistry , Fish Oils/pharmacology , Gastrointestinal Tract/metabolism , HEK293 Cells , Humans , Inflammation/drug therapy , Inflammation/immunology , Mice , Models, Biological , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/immunology
9.
J Cell Physiol ; 227(7): 2907-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21938725

ABSTRACT

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.


Subject(s)
Cell Engineering/methods , Endothelium, Vascular/metabolism , Epoprostenol/biosynthesis , Epoprostenol/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Stem Cells/metabolism , 4-Aminopyridine/metabolism , Animals , Apoptosis/genetics , Cell Growth Processes/genetics , Culture Media, Conditioned/metabolism , Cyclooxygenase 1/genetics , Cytochrome P-450 Enzyme System/genetics , DNA, Complementary/genetics , Endothelium, Vascular/cytology , Epoprostenol/metabolism , Half-Life , Hypoxia/genetics , Hypoxia/metabolism , Intramolecular Oxidoreductases/genetics , Membrane Proteins/genetics , Muscle, Smooth, Vascular/cytology , Neovascularization, Physiologic , Phenotype , Potassium Channels/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection/methods
10.
Biochemistry ; 50(10): 1691-9, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21250659

ABSTRACT

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).


Subject(s)
Adipocytes/metabolism , Down-Regulation , Epoprostenol/metabolism , MicroRNAs/genetics , Up-Regulation , Animals , Cells, Cultured , Humans , Mice , Proto-Oncogene Proteins c-akt/genetics
11.
Life Sci ; 88(1-2): 24-30, 2011 Jan 03.
Article in English | MEDLINE | ID: mdl-21035466

ABSTRACT

AIM: Our aim is to understand the molecular mechanisms of the selective nonsteroidal anti-inflammatory drugs (NSAID), cyclooxygenase-2 (COX-2) inhibitors', higher "priority" to reduce synthesis of the vascular protector, prostacyclin (PGI2), compared to that of nonselective NSAIDs. MAIN METHODS: COX-1 or COX-2 was co-expressed with PGI2 synthase (PGIS) in COS-7 cells. The Km and initial velocity (½t Vmax) of the coupling reaction between COX-1 and COX-2 to PGIS were established. The experiment was further confirmed by a kinetics study using hybrid enzymes linking COX-1 or COX-2 to PGIS. Finally, COX-1 or COX-2 and PGIS were respectively fused to red (RFP) and cyanic (CFP) fluorescence proteins, and co-expressed in cells. The distances between COXs and PGIS were compared by FRET. KEY FINDINGS: The Km for converting arachidonic acid (AA) to PGI2 by COX-2 coupled to PGIS is ~2.0µM; however, it was 3-fold more (~6.0µM) for COX-1 coupled to PGIS. The Km and ½t Vmax for COX-2 linked to PGIS were ~2.0µM and 20s, respectively, which were 2-5 folds faster than that of COX-1 linked to PGIS. The FRET study found that the distance between COX-2-RFP and PGIS-CFP is shorter than that between COX-1-RFP and PGIS-CFP. SIGNIFICANCE: The study provided strong evidence suggesting that the low Km, faster ½t Vmax, and closer distance are the basis for COX-2 dominance over COX-1 (coupled to PGIS) in PGI2 synthesis, and further demonstrated the mechanisms of selective COX-2 inhibitors with higher potential to reduce synthesis of the vascular protector, PGI2.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Epoprostenol/biosynthesis , Animals , Arachidonic Acid/metabolism , Blotting, Western , COS Cells , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Chlorocebus aethiops , Chromatography, High Pressure Liquid , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Electrophoresis, Polyacrylamide Gel , Microscopy, Fluorescence
12.
Int J Cardiol ; 150(3): 253-9, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-20451268

ABSTRACT

INTRODUCTION: An emerging technology using human embryonic stem cells (hESCs) to regenerate infarcted heart tissue has been underdeveloped. However, because non-steroidal anti-inflammatory drugs (NSAIDs), such as aspirin, are taken during the infarction, it becomes critical to know whether the NSAIDs have negative impacts on heart tissue regeneration when using hESCs. METHODS: Mass spectrometry (LC/MS/MS) and high performance liquid chromatography (HPLC) analyses were used to analyze the functional presence of the elaborate prostanoids' biosynthesis and signaling systems in hESCs. The detected endogenous arachidonic acid (AA) released in the hESC membranes reflects the activity of phospholipase which directly controls the biosyntheses of the prostanoids. RESULTS: The complete inhibition of the endogenous prostaglandin E(2) (PGE(2)) biosynthesis by the cyclooxygenase-2 (COX-2) inhibitor, NS398, confirmed that the major prostanoids synthesized in the hESCs are mediated by the COX-2 enzyme. We also found that PGE(2) and the prostacyclin (PGI(2)) metabolite, 6-keto-PGF(1α), are present in the undifferentiated hESCs. CONCLUSION: This indicated different cyclooxygenase (COX)-downstream synthases and metabolizing enzymes are involved in the AA products' signaling through the COX-1 and COX-2 pathways. The presence of many enzymes' and receptors' [(COX-1, COX-2, microsomal prostaglandin E synthase (mPGES), cytosolic prostaglandin E synthase (cPGES), prostaglandin I synthase (PGIS), the PGE(2) subtype receptors (EP(1), EP(2), and EP(4)) and the prostacyclin receptor (IP)] involvement in the prostanoid biosynthesis and activity was confirmed by western blot. The studies implied the negative effects of NSAIDs, such as aspirin and COX-2 inhibitors, which suppress prostanoid production during tissue regeneration for infarcted heart when using hESCs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Arachidonic Acid/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Heart/drug effects , Regeneration/drug effects , Arachidonic Acid/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Drug Delivery Systems/methods , Feeder Cells/drug effects , Feeder Cells/metabolism , Growth Inhibitors/adverse effects , Heart/physiology , Humans , Prostaglandins/biosynthesis , Regeneration/physiology
13.
Protein Eng Des Sel ; 22(12): 733-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19850676

ABSTRACT

Cyclooxygenase isoform-2 (COX-2) and microsomal prostaglandin E(2) synthase-1 (mPGES-1) are inducible enzymes that become up-regulated in inflammation and some cancers. It has been demonstrated that their coupling reaction of converting arachidonic acid (AA) into prostaglandin (PG) E(2) (PGE(2)) is responsible for inflammation and cancers. Understanding their coupling reactions at the molecular and cellular levels is a key step toward uncovering the pathological processes in inflammation. In this paper, we describe a structure-based enzyme engineering which produced a novel hybrid enzyme that mimics the coupling reactions of the inducible COX-2 and mPGES-1 in the native ER membrane. Based on the hypothesized membrane topologies and structures, the C-terminus of COX-2 was linked to the N-terminus of mPGES-1 through a transmembrane linker to form a hybrid enzyme, COX-2-10aa-mPGES-1. The engineered hybrid enzyme expressed in HEK293 cells exhibited strong triple-catalytic functions in the continuous conversion of AA into PGG(2) (catalytic-step 1), PGH(2) (catalytic-step 2) and PGE(2) (catalytic-step 3), a pro-inflammatory mediator. In addition, the hybrid enzyme was also able to directly convert dihomo-gamma-linolenic acid (DGLA) into PGG(1), PGH(1) and then PGE(1) (an anti-inflammatory mediator). The hybrid enzyme retained similar K(d) and V(max) values to that of the parent enzymes, suggesting that the configuration between COX-2 and mPGES-1 (through the transmembrane domain) could mimic the native conformation and membrane topologies of COX-2 and mPGES-1 in the cells. The results indicated that the quick coupling reaction between the native COX-2 and mPGES-1 (in converting AA into PGE(2)) occurred in a way so that both enzymes are localized near each other in a face-to-face orientation, where the COX-2 C-terminus faces the mPGES-1 N-terminus in the ER membrane. The COX-2-10aa-mPGES-1 hybrid enzyme engineering may be a novel approach in creating inflammation cell and animal models, which are particularly valuable targets for the next generation of NSAID screening.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arachidonic Acid/metabolism , Dinoprostone/metabolism , Protein Engineering , Biocatalysis , Cell Line , Chromatography, High Pressure Liquid , Cyclooxygenase 2/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Immunoenzyme Techniques , Inflammation/enzymology , Inflammation/metabolism , Intramolecular Oxidoreductases/metabolism , Kinetics , Prostaglandin-E Synthases
14.
FEBS J ; 275(23): 5820-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19021758

ABSTRACT

It remains a challenge to achieve the stable and long-term expression (in human cell lines) of a previously engineered hybrid enzyme [triple-catalytic (Trip-cat) enzyme-2; Ruan KH, Deng H & So SP (2006) Biochemistry45, 14003-14011], which links cyclo-oxygenase isoform-2 (COX-2) to prostacyclin (PGI(2)) synthase (PGIS) for the direct conversion of arachidonic acid into PGI(2) through the enzyme's Trip-cat functions. The stable upregulation of the biosynthesis of the vascular protector, PGI(2), in cells is an ideal model for the prevention and treatment of thromboxane A(2) (TXA(2))-mediated thrombosis and vasoconstriction, both of which cause stroke, myocardial infarction, and hypertension. Here, we report another case of engineering of the Trip-cat enzyme, in which human cyclo-oxygenase isoform-1, which has a different C-terminal sequence from COX-2, was linked to PGI(2) synthase and called Trip-cat enzyme-1. Transient expression of recombinant Trip-cat enzyme-1 in HEK293 cells led to 3-5-fold higher expression capacity and better PGI(2)-synthesizing activity as compared to that of the previously engineered Trip-cat enzyme-2. Furthermore, an HEK293 cell line that can stably express the active new Trip-cat enzyme-1 and constantly synthesize the bioactive PGI(2) was established by a screening approach. In addition, the stable HEK293 cell line, with constant production of PGI(2), revealed strong antiplatelet aggregation properties through its unique dual functions (increasing PGI(2) production while decreasing TXA(2) production) in TXA(2) synthase-rich plasma. This study has optimized engineering of the active Trip-cat enzyme, allowing it to become the first to stably upregulate PGI(2) biosynthesis in a human cell line, which provides a basis for developing a PGI(2)-producing therapeutic cell line for use against vascular diseases.


Subject(s)
Cyclooxygenase 1/metabolism , Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/biosynthesis , Intramolecular Oxidoreductases/metabolism , Recombinant Fusion Proteins/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Catalysis , Cell Line , Cloning, Molecular , Cyclooxygenase 1/genetics , Cytochrome P-450 Enzyme System/genetics , Endoplasmic Reticulum/metabolism , Epoprostenol/pharmacology , Gene Expression , Humans , Intramolecular Oxidoreductases/genetics , Kinetics , Models, Molecular , Platelet Aggregation/drug effects , Protein Engineering/methods , Recombinant Fusion Proteins/genetics , Thromboxane B2/metabolism
15.
Arch Biochem Biophys ; 480(1): 41-50, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18835243

ABSTRACT

Recently, we reported that a novel hybrid enzyme (TriCat enzyme), engineered by linking human cyclooxygenase-2 (COX-2) with prostacyclin (PGI(2)) synthase (PGIS) together through a transmembrane domain, was able to directly integrate the triple catalytic (TripCat) functions of COX-2 and PGIS and effectively convert arachidonic acid (AA) into the vascular protector, PGI(2) [K.H. Ruan, H. Deng, S.P. So, Biochemistry 45 (2006) 14003-14011]. In order to confirm the important biological activity and evaluate its therapeutic potential, it is critical to characterize the properties of the enzyme using the purified protein. The TriCat enzyme cDNA was subcloned into a baculovirus vector and its protein was expressed in Sf-9 cells in large-scale with a high-yield ( approximately 4% of the total membrane protein), as confirmed by Western blot and protein staining. The Sf-9 cells' membrane fraction, rich in TriCat enzyme, exhibited strong TriCat functions (K(m)=3 microM and K(cat)=100 molecules/min) for the TriCat enzyme and was 3-folds faster in converting AA to PGI(2) than the combination of the individual COX-2 and PGIS. Another superiority of the TriCat enzyme is its dual effect on platelet aggregation: it completely inhibited platelet aggregation at the low concentration of 2 microg/ml and then displayed the ability to reverse the initially aggregated platelets to their non-aggregated state. Furthermore, multiple substrate-binding sites were confirmed in the single protein by high-resolution NMR spectroscopy, using partially purified TriCat enzyme. These studies have clearly demonstrated that the isolated TriCat enzyme protein functions in the selective biosynthesis of the vascular protector, PGI(2), and revealed its potential for anti-thrombosis therapeutics.


Subject(s)
Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Animals , Arachidonic Acid/metabolism , Baculoviridae/genetics , COS Cells , Cell Line , Chlorocebus aethiops , Cyclooxygenase 2/isolation & purification , Cyclooxygenase 2/pharmacology , Cytochrome P-450 Enzyme System/isolation & purification , Cytochrome P-450 Enzyme System/pharmacology , DNA, Complementary/genetics , Drug Design , Epoprostenol/biosynthesis , Genetic Vectors , Humans , In Vitro Techniques , Intramolecular Oxidoreductases/isolation & purification , Intramolecular Oxidoreductases/pharmacology , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Platelet Aggregation/drug effects , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Spodoptera
16.
FEBS Lett ; 582(19): 2863-8, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18652829

ABSTRACT

A peptide constrained to a conformation of second extracellular loop of human prostaglandin-E(2) (PGE(2)) receptor subtype3 (hEP3) was synthesized. The contacts between the peptide residues at S211 and R214, and PGE(2) were first identified by NMR spectroscopy. The results were used as a guide for site-directed mutagenesis of the hEP3 protein. The S211L and R214L mutants expressed in HEK293 cells lost binding to [(3)H]PGE(2). This study found that the non-conserved S211 and R214 of the hEP3 are involved in PGE(2) recognition, and implied that the corresponding residues in other subtype receptors could be important to distinguish the different configurations of PGE(2) ligand recognition sites.


Subject(s)
Dinoprostone/metabolism , Receptors, Prostaglandin E/metabolism , Amino Acid Sequence , Cell Line , Conserved Sequence , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Peptides/genetics , Peptides/metabolism , Point Mutation , Protein Conformation , Radioligand Assay , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Spectrometry, Fluorescence
17.
FEBS J ; 275(1): 128-37, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18042246

ABSTRACT

The residues in the second extracellular loop (eLP2) of the prostanoid receptors, which are important for specific ligand recognition, were previously predicted in our earlier studies of the thromboxane A2 receptor (TP) using a combination of NMR spectroscopy and recombinant protein approaches. To further test this hypothesis, another prostanoid receptor, the prostacyclin receptor (IP), which has opposite biological characteristics to that of TP, was used as a model for these studies. A set of recombinant human IPs with site-directed mutations at the nonconserved eLP2 residues were constructed using an Ala-scanning approach, and then expressed in HEK293 and COS-7 cells. The expression levels of the recombinant receptors were six-fold higher in HEK293 cells than in COS-7 cells. The residues important for ligand recognition and binding within the N-terminal segment (G159, Q162, and C165) and the C-terminal segment (L172, R173, M174, and P179) of IP eLP2 were identified by mutagenesis analyses. The molecular mechanisms for the specific ligand recognition of IP were further demonstrated by specific site-directed mutagenesis using different amino acid residues with unique chemical properties for the key residues Q162, L172, R173, and M174. A comparison with the corresponding functional residues identified in TP eLP2 revealed that three (Q162, R173, and M174) of the four residues are nonconserved, and these are proposed to be involved in specific ligand recognition. We discuss the importance of G159 and P179 in ligand recognition through configuration of the loop conformation is discussed. These studies have further indicated that characterization of the residues in the eLP2 regions for all eight prostanoid receptors could be an effective approach for uncovering the molecular mechanisms of the ligand selectivities of the G-protein-coupled receptors.


Subject(s)
Receptors, Prostaglandin/chemistry , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Receptors, Epoprostenol , Receptors, Prostaglandin/metabolism
18.
Biochemistry ; 45(47): 14003-11, 2006 Nov 28.
Article in English | MEDLINE | ID: mdl-17115695

ABSTRACT

Prostacyclin (PGI2), a vascular protector with vasodilation and antithrombotic properties, is synthesized by coupling reactions of cyclooxygenase (COX, the first enzyme) with PGI2 synthase (PGIS, the second enzyme) using arachidonic acid (AA) as an initial substrate. The first COX product, prostaglandin H2 (PGH2) is also a command substrate for other prostanoid enzymes that produce distinct eicosanoids, such as thromboxane A2 (TXA2). The actions of TXA2 to cause vasoconstriction and platelet aggregation oppose the vasodilatory and anti-aggregatory effects of PGI2. Specifically upregulating PGI2 biosynthesis is an ideal model for the prevention and treatment of the TXA2-mediated thrombosis involved in strokes and myocardial infarctions. Here, we report that a single protein was constructed by linking COX-2 and PGIS together to form a new fusion enzyme through a transmembrane domain with 10 or 22 residues. The engineered protein expressed in HEK293 and COS-7 cells was able to continually convert AA to prostaglandin (PG) G2 (catalytic step 1), PGH2 (catalytic step 2), and PGI2 (catalytic step 3). The studies first demonstrate that a single protein with three catalytic functions could directly synthesize PGI2 from AA with a Km of approximately 3.2 microM. Specific upregulation of PGI2 biosynthesis through expression of the engineered single protein in the cells has shown strong activity in inhibiting platelet aggregation induced by AA in vitro, which creates a great potential for the fusion enzyme to be used as one of the new therapeutic interventions for strokes and heart attacks. The studies have also provided a model linking COX with its downstream enzymes to specifically regulate biosynthesis of eicosanoids which have potent biological functions.


Subject(s)
Arachidonic Acid/metabolism , Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/metabolism , Intramolecular Oxidoreductases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Engineering , Amino Acid Sequence , Animals , Base Sequence , Catalysis , Cell Line , Cytochrome P-450 Enzyme System/chemistry , DNA Primers , Humans , Intramolecular Oxidoreductases/chemistry , Prostaglandin-Endoperoxide Synthases/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
19.
Arch Biochem Biophys ; 435(2): 372-81, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15708381

ABSTRACT

To mimic the native conditions, the cyclooxygenase (COX)/prostaglandin I(2) synthase (PGIS) coupling reaction system was used to determine the coordination of PGIS with COX for the biosynthesis of prostacyclin (PGI(2)) using arachidonic acid (AA) as a substrate in a membrane-bound environment. The membrane-bound PGIS exhibited a faster isomerization of PGH(2) produced by COX to PGI(2) than the detergent-solubilized PGIS. To determine whether the N-terminal domain of PGIS responds to the facilitation of PGH(2) movement (presentation) from COX to the active site of PGIS, the first 20 residues of PGIS (Delta20-PGIS) were deleted and expressed in COS-7 cells. Delta20-PGIS retained membrane-bound properties and exhibited a slower substrate presentation property. Furthermore, a chimeric molecule (PGIS/TXAS(8-27)) with the replacement of the first 20 residues of PGIS by the corresponding membrane anchor region (residues 8-27) of thromboxane A(2) synthase was created to evaluate the mechanism influencing the biosynthesis of PGI(2) in coordination with COX. The chimera revealed a multiple fold delay in the PGH(2) presentation in low range concentrations of AA (0.3-3muM) at 30s reactions. However, the delay could be recovered by a longer incubation time in high range concentrations of AA (>10muM), but not in low range concentrations of AA. These results demonstrated that the N-terminal domain of PGIS plays a role in the facilitation of the substrate presentation to the PGIS active site in low concentrations of AA, which may be a physiological condition. The TXAS N-terminal domain could not replace the function of the corresponding domain of PGIS, indicating that the facilitation of the substrate presentation is specific.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/metabolism , Intramolecular Oxidoreductases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Recombinant Fusion Proteins/metabolism , Thromboxane-A Synthase/metabolism , Animals , Arachidonic Acid/metabolism , Binding Sites , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cytochrome P-450 Enzyme System/genetics , Epoprostenol/genetics , Intramolecular Oxidoreductases/genetics , Mutation/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/genetics , Thromboxane-A Synthase/genetics
20.
Eur J Biochem ; 271(14): 3006-16, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15233797

ABSTRACT

To overcome the difficulty of characterizing the structures of the extracellular loops (eLPs) of G protein-coupled receptors (GPCRs) other than rhodopsin, we have explored a strategy to generate a three-dimensional structural model for a GPCR, the thromboxane A(2) receptor. This three-dimensional structure was completed by the assembly of the NMR structures of the computation-guided constrained peptides that mimicked the extracellular loops and connected to the conserved seven transmembrane domains. The NMR structure-based model reveals the structural features of the eLPs, in which the second extracellular loop (eLP(2)) and the disulfide bond between the first extracellular loop (eLP(1)) and eLP(2) play a major role in forming the ligand recognition pocket. The eLP(2) conformation is dynamic and regulated by the oxidation and reduction of the disulfide bond, which affects ligand docking in the initial recognition. The reduced form of the thromboxane A(2) receptor experienced a decrease in ligand binding activity due to the rearrangement of the eLP(2) conformation. The ligand-bound receptor was, however, resistant to the reduction inactivation because the ligand covered the disulfide bond and stabilized the eLP(2) conformation. This molecular mechanism of ligand recognition is the first that may be applied to other prostanoid receptors and other GPCRs.


Subject(s)
Protein Structure, Tertiary , Receptors, Thromboxane A2, Prostaglandin H2/chemistry , Amino Acid Sequence , Animals , Binding Sites , Disulfides/chemistry , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Structure, Secondary , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...