Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Lipid Res ; 52(2): 374-82, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21062953

ABSTRACT

Endothelial lipase (EL) is a phospholipase A1 (PLA1) enzyme that hydrolyzes phospholipids at the sn-1 position to produce lysophospholipids and free fatty acids. Measurement of the PLA1 activity of EL is usually accomplished by the use of substrates that are also hydrolyzed by lipases in other subfamilies such as PLA2 enzymes. In order to distinguish PLA1 activity of EL from PLA2 enzymatic activity in cell-based assays, cell supernatants, and other nonhomogeneous systems, a novel fluorogenic substrate with selectivity toward PLA1 hydrolysis was conceived and characterized. This substrate was preferred by PLA1 enzymes, such as EL and hepatic lipase, and was cleaved with much lower efficiency by lipases that exhibit primarily triglyceride lipase activity, such as LPL or a lipase with PLA2 activity. The phospholipase activity detected by the PLA1 substrate could be inhibited with the small molecule esterase inhibitor ebelactone B. Furthermore, the PLA1 substrate was able to detect EL activity in human umbilical vein endothelial cells in a cell-based assay. This substrate is a useful reagent for identifying modulators of PLA1 enzymes, such as EL, and aiding in characterizing their mechanisms of action.


Subject(s)
Boron Compounds/metabolism , Endothelium/enzymology , Lysophospholipids/metabolism , Phospholipases A1/analysis , Animals , Fluorescent Dyes/metabolism , Humans , Lactones/pharmacology , Lipase/antagonists & inhibitors , Lipase/metabolism , Mice , Phospholipases A1/antagonists & inhibitors
2.
J Lipid Res ; 51(12): 3559-67, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20805092

ABSTRACT

Acyl-CoA:diacylglycerol acyltransferase (DGAT) catalyzes the terminal step in triglyceride (TG) synthesis using diacylglycerol (DAG) and fatty acyl-CoA as substrates. In the liver, the production of VLDL permits the delivery of hydrophobic TG from the liver to peripheral tissues for energy metabolism. We describe here a novel high-content, high-throughput LC/MS/MS-based cellular assay for determining DGAT activity. We treated endogenous DGAT-expressing cells with stable isotope-labeled [¹³C18]oleic acid. The [¹³C18]oleoyl-incorporated TG and DAG lipid species were profiled. The TG synthesis pathway assay was optimized to a one-step extraction, followed by LC/MS/MS quantification. Further, we report a novel LC/MS/MS method for tracing hepatic TG synthesis and VLDL-TG secretion in vivo by administering [¹³C18]oleic acid to rats. The [¹³C18]oleic acid-incorporated VLDL-TG was detected after one-step extraction without conventional separation of TG and recovery by derivatizing [¹³C18]oleic acid for detection. Using potent and selective DGAT1 inhibitors as pharmacological tools, we measured changes in [¹³C18]oleoyl-incorporated TG and DAG and demonstrated that DGAT1 inhibition significantly reduced [¹³C18]oleoyl-incorporated VLDL-TG. This DGAT1-selective assay will enable researchers to discern differences between the roles of DGAT1 and DGAT2 in TG synthesis in vitro and in vivo.


Subject(s)
Diacylglycerol O-Acyltransferase/metabolism , Enzyme Assays/methods , Liver/enzymology , Animals , Carbon Radioisotopes/metabolism , Cells, Cultured , Chromatography, Liquid , Hepatocytes/cytology , Hepatocytes/enzymology , Humans , Insecta/cytology , Insecta/enzymology , Insecta/virology , Kidney/cytology , Kidney/embryology , Kidney/enzymology , Lipoproteins, VLDL/chemistry , Lipoproteins, VLDL/metabolism , Male , Oleic Acid/metabolism , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Triglycerides/chemistry , Triglycerides/metabolism
3.
Magn Reson Chem ; 47 Suppl 1: S20-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19565469

ABSTRACT

NMR-based metabolomics of mouse urine was used in conjunction with the traditional staining and imaging of aortas for the characterization of disease advancement, that is, plaque formation in untreated and drug-treated apolipoprotein-E (apoE) knockout mice. The metabolomics approach with multivariate analysis was able to differentiate the captopril-treated from the untreated mice in general agreement with the staining results. Principal component analysis showed a pattern shift in both the drug-treated and untreated samples as a function of time that could possibly be explained as the effect of aging. Allantoin, a marker attributed to captopril treatment was elevated in the drug-treated mice. From partial least squares-discriminant analysis, xanthine and ascorbate were elevated in the untreated mice and were possible markers of plaque formation in the apoE knockout mice. Several additional peaks in the spectra characterizing the study endpoint were found but their respective metabolite identities were unknown.


Subject(s)
Atherosclerosis/physiopathology , Biomarkers/urine , Metabolomics , Animals , Antihypertensive Agents/pharmacology , Apolipoproteins E/deficiency , Captopril/pharmacology , Disease Models, Animal , Magnetic Resonance Spectroscopy , Mice , Mice, Knockout , Reference Standards
4.
Methods ; 37(3): 280-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16308157

ABSTRACT

Microarray technology enables high-throughput testing of gene expression to investigate various neuroscience related questions. This in turn creates a demand for scalable methods to confirm microarray results and the opportunity to use this information to discover and test novel pathways and therapeutic applications. Discovery of new central nervous system (CNS) treatments requires a comprehensive understanding of multiple aspects including the biology of a target, the pathophysiology of a disease/disorder, and the selection of successful lead compounds as well as efficient biomarker and drug disposition strategies such as absorption (how a drug is absorbed), distribution (how a drug spreads through an organism), metabolism (chemical conversion of a drug, if any, and into which substances), and elimination (how is a drug eliminated) (ADME). Understanding of the toxicity is also of paramount importance. These approaches, in turn, require novel high-content integrative assay technologies that provide thorough information about changes in cell biology. To increase efficiency of profiling, characterization, and validation, we established a new screening strategy that combines high-content image-based testing on Array Scan (Cellomics) with a confocal system and the multiplexed TaqMan RT-PCR method for quantitative mRNA expression analysis. This approach could serve as an interface between high-throughput microarray testing and specific application of markers discovered in the course of a microarray experiment. Markers could pinpoint activation or inhibition of a molecular pathway related, for instance, to neuronal viability. We demonstrate the successful testing of the same cell population in an image-based translocational assay followed by poly(A) mRNA capture and multiplexed single tube RT-PCR. In addition, Ciphergen ProteinChip analysis can be performed on the supernatant, thus allowing significant complementarity in the data output and interpretation by also including the capture and initial analysis of proteins in the integrative approach presented. We have determined various conditions including the number of cells, RT and PCR optimization, which are necessary for successful detection and consequent assay integration. We also show the successful convergence of various different approaches and multiplexing of different targets within a single real-time PCR tube. This novel integrative technological approach has utility for CNS drug discovery, target and biomarker identification, selection and characterization as well as for the study of toxicity- and adverse event-associated molecular mechanisms.


Subject(s)
Drug Evaluation, Preclinical/methods , Gene Expression Profiling/methods , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Drug Industry/methods , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Confocal , Protein Array Analysis/methods , Protein Kinase C/analysis , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Transport , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reproducibility of Results
5.
Anal Biochem ; 331(1): 122-9, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15246004

ABSTRACT

Sphingosine kinase catalyses the phosphorylation of sphingosine to generate sphingosine 1-phosphate, a lipid signaling molecule implicated in roles in a diverse range of mammalian cell processes through its action as both a ligand for G-protein-coupled cell-surface receptors and an apparent intracellular second messenger. This paper describes a rapid, sensitive, and reproducible assay for sphingosine kinase activity using biotinylated sphingosine (biotinyl-Sph) as a substrate and capturing the phosphorylated product with streptavidin-coated membranes. We have shown that both human sphingosine kinase 1 and 2 (hSK1 and hSK2) can efficiently phosphorylate biotinyl-Sph, with K(m) values similar to those of sphingosine. The assay utilizing this substrate has high sensitivity for hSK1 and hSK2, with detection limits in the low-femtomole range for both purified recombinant enzymes. Importantly, we have also demonstrated the capacity of this assay to measure endogenous sphingosine kinase activity in crude cell extracts and to follow changes in this activity following sphingosine kinase activation. Together, these results demonstrate the potential utility of this assay in both cell-based analysis of sphingosine kinase signaling pathways and high-throughput screens for agents affecting sphingosine kinase activity in vitro.


Subject(s)
Bacterial Proteins/chemistry , Biotin/chemistry , Membranes, Artificial , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Sphingosine/chemistry , Streptavidin/chemistry , Humans , Kinetics , Substrate Specificity
6.
Arterioscler Thromb Vasc Biol ; 24(6): 1118-23, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15130911

ABSTRACT

OBJECTIVE: Recently, mice made deficient in growth arrest-specific gene 6 product (Gas6) or in which Gas6 gene expression was inhibited were shown to have platelet dysfunction and to be less susceptible to thrombosis. The aim of this study was to define and characterize the relevant Gas6 receptor or receptors involved in platelet function. METHODS AND RESULTS: Using RT-PCR and Western blot analysis we found that mer was the predominantly expressed subtype in mouse and human platelets, whereas axl and rse were not detected. We generated mer-deficient mice by targeted disruption of the mer receptor gene. Platelets derived from mer-deficient mice had decreased platelet aggregation in responses to low concentrations of collagen, U46619, and PAR4 thrombin receptor agonist peptide in vitro. However, the response to ADP was not different from wild-type platelets. Knockout of the mer gene protected mice from collagen/epinephrine-induced pulmonary thromoembolism and inhibited ferric chloride-induced thrombosis in vivo. Tail bleeding times, coagulation parameters, and peripheral blood cell counts in mer-deficient mice were similar to wild-type mice. CONCLUSIONS: Our data provide the first evidence that mer, presumably through activation by its ligand Gas6, participates in regulation of platelet function in vitro and platelet-dependent thrombosis in vivo.


Subject(s)
Blood Platelets/enzymology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Blood Coagulation Tests , Blood Platelets/physiology , Blotting, Western , Chlorides , Collagen/pharmacology , Collagen/toxicity , Epinephrine/toxicity , Female , Ferric Compounds/toxicity , Humans , Intercellular Signaling Peptides and Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligopeptides/pharmacology , Oncogene Proteins/analysis , Platelet Aggregation/drug effects , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Thrombin/agonists , Reverse Transcriptase Polymerase Chain Reaction , Thromboembolism/chemically induced , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
7.
J Biomol Screen ; 8(1): 65-71, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12854999

ABSTRACT

Target validation is one of rate-limiting steps in the modern drug discovery. The authors developed a strategy of combining adenovirus-mediated gene transfer for efficient target functionality validation, both in vivo and in vitro, with baculovirus expression to produce sufficient quantities of protein for high-throughput screening (HTS). The incorporation of green fluorescent protein (GFP) in the adenovirus vectors accelerates recombinant adenovirus plaque purification, whereas the use of epitope and affinity tags facilitates the identification and purification of recombinant protein. In this generalized scheme, the flexible modular design of viral vectors facilitates the transition between target validation and HTS. In the example presented, functional target validation in vivo was achieved by overexpressing the target gene in cell-based models and in the mouse cortex following adenovirus-mediated gene delivery. In this context, target overexpression resulted in the accumulation of a disease-related biomarker both in vitro and in vivo. A baculovirus-based expressional system was then generated to produce enough target protein for HTS. Thus, the use of these viral expression systems represents a generalized method for rapid target functionality validation and HTS assay development, which could be applied to numerous target candidates being elucidated in gene discovery programs.


Subject(s)
Adenoviridae , Genetic Vectors , Recombinant Proteins/analysis , Computational Biology
8.
Biochem J ; 374(Pt 1): 97-107, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12795636

ABSTRACT

We have identified a novel serine protease designated EOS by sequence identity searches. The deduced protein contains 284 amino acids with an active form containing 248 amino acids starting from an Ile-Val-Gly-Gly motif. The active form comprises a catalytic triad of conserved amino acids: His77, Asp126 and Ser231. It shares 44% identity with beta-tryptase and belongs to the S1 trypsin-like serine-protease family. Interestingly, this gene also maps to human chromosome 16p13.3. The purified protease showed amidolytic activity, cleaving its substrates before arginine residues. Tissue distribution by immunohistochemistry analysis demonstrated that EOS is highly expressed in spleen and moderately expressed in intestine, colon, lung and brain. We confirmed this expression pattern at the mRNA level by performing in situ hybridization. The results from both immunohistochemistry and in situ hybridization indicate that EOS is associated with macrophages. We corroborated this observation by double immunofluorescence using the anti-EOS antibody and an anti-CD68 antibody, a macrophage specific marker. Furthermore, we have detected a dramatic increase in immune staining of EOS in cultured U937 cells treated with PMA, which represent activated macrophages. This up-regulation is also reflected by elevated EOS mRNA in the PMA-treated U937 cells detected by Northern blotting. Since macrophages have important roles in various pathological conditions, such as wound healing, atherosclerosis and numerous inflammatory diseases, the localization of this novel serine protease to active macrophages may help to further the elucidation of the roles of this gene product in modulating these disorders.


Subject(s)
Chromosomes, Human, Pair 16 , Macrophages/enzymology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Colon/enzymology , DNA Primers , Humans , Lung/enzymology , Molecular Sequence Data , Organ Specificity , Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Spleen/enzymology , U937 Cells
9.
J Pharmacol Exp Ther ; 304(2): 855-61, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538843

ABSTRACT

Although it is well recognized that human platelet responses to alpha-thrombin are mediated by the protease-activated receptors PAR-1 and PAR-4, their role and relative importance in platelet-dependent human disease has not yet been elucidated. Because the expression profile of PARs in platelets from nonprimates differs from humans, we used cynomolgus monkeys to evaluate the role of PAR-1 in thrombosis. Based on reverse transcription-polymerase chain reaction, PAR expression in platelets from cynomolgus monkeys consisted primarily of PAR-1 and PAR-4, thereby mirroring the profile of human platelets. We probed the role of PAR-1 in a primate model of vascular injury-induced thrombosis with the selective PAR-1 antagonist (alpha S)-N-[(1S)-3-amino-1-[[(phenylmethyl)amino]carbonyl]propyl]-alpha-[[[[[1-(2,6-dichlorophenyl)methyl]-3-(1-pyrrolidinylmethyl)-1H-indazol-6-yl]amino]carbonyl]amino]-3,4-difluorobenzenepropanamide (RWJ-58259). After pretreatment with RWJ-58259 or vehicle, both carotid arteries of anesthetized monkeys were electrolytically injured and blood flow was monitored for 60 min. Time to occlusion was significantly extended after RWJ-58259 administration (27 +/- 3 to 53 +/- 8 min; p < 0.048). Vessels from three of the five treated animals remained patent. Ex vivo platelet aggregation measurements indicated complete PAR-1 inhibition, as well as an operational PAR-4 response. Immunohistochemical staining of mural thrombi with antibodies to the platelet marker CD61 and fibrinogen indicated that RWJ-58259 significantly reduced thrombus platelet deposition. Drug treatment had no effect on key hematological or coagulation parameters. Our results provide direct evidence that PAR-1 is the primary receptor that mediates alpha-thrombin's prothrombotic actions in primates and suggest that PAR-1 antagonists may have potential for the treatment of thrombotic disorders in humans.


Subject(s)
Carotid Artery, Common/drug effects , Receptors, Thrombin/antagonists & inhibitors , Thrombosis/prevention & control , Urea/analogs & derivatives , Animals , Carotid Artery, Common/pathology , Female , Indazoles/chemistry , Indazoles/pharmacology , Indazoles/therapeutic use , Macaca fascicularis , Male , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Receptor, PAR-1 , Thrombosis/drug therapy , Thrombosis/pathology , Urea/chemistry , Urea/pharmacology , Urea/therapeutic use
10.
J Biol Chem ; 278(13): 11714-20, 2003 Mar 28.
Article in English | MEDLINE | ID: mdl-12522105

ABSTRACT

Protease-activated receptor (PAR)-4 is a low affinity thrombin receptor with slow activation and desensitization kinetics relative to PAR-1. This study provides novel evidence that cardiomyocytes express functional PAR-4 whose signaling phenotype is distinct from PAR-1 in cardiomyocytes. AYPGKF, a modified PAR-4 agonist with increased potency at PAR-4, activates p38-mitogen-activated protein kinase but is a weak activator of phospholipase C, extracellular signal-regulated kinase, and cardiomyocyte hypertrophy; AYPGKF and thrombin, but not the PAR-1 agonist SFLLRN, activate Src. The observation that AYPGKF and thrombin activate Src in cardiomyocytes cultured from PAR-1(-/-) mice establishes that Src activation is via PAR-4 (and not PAR-1) in cardiomyocytes. Further studies implicate Src and epidermal growth factor receptor (EGFR) kinase activity in the PAR-4-dependent p38-mitogen-activated protein kinase signaling pathway. Thrombin phosphorylates EGFRs and ErbB2 via a PP1-sensitive pathway in PAR-1(-/-) cells that stably overexpress PAR-4; the Src-mediated pathway for EGFR/ErbB2 transactivation underlies the protracted phases of thrombin-dependent extracellular signal-regulated kinase activation in PAR-1(-/-) cells that overexpress PAR-4 and in cardiomyocytes. These studies identify a unique signaling phenotype for PAR-4 (relative to other cardiomyocyte G protein-coupled receptors) that is predicted to contribute to cardiac remodeling and influence the functional outcome at sites of cardiac inflammation.


Subject(s)
Myocardium/metabolism , Receptors, Thrombin/metabolism , src-Family Kinases/metabolism , Animals , Base Sequence , Blotting, Western , DNA Primers , Mice , Mice, Inbred C57BL , Myocardium/cytology , Myocardium/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
11.
J Biol Chem ; 277(51): 49545-53, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12393916

ABSTRACT

Sphingosine kinase catalyzes the formation of sphingosine 1-phosphate, a lipid second messenger that has been implicated in a number of agonist-driven cellular responses including mitogenesis, anti-apoptosis, and expression of inflammatory molecules. Despite the importance of sphingosine kinase, very little is known regarding its structure or mechanism of catalysis. Moreover, sphingosine kinase does not contain recognizable catalytic or substrate-binding sites, based on sequence motifs found in other kinases. Here we have elucidated the nucleotide-binding site of human sphingosine kinase 1 (hSK1) through a combination of site-directed mutagenesis and affinity labeling with the ATP analogue, FSBA. We have shown that Gly(82) of hSK1 is involved in ATP binding since mutation of this residue to alanine resulted in an enzyme with an approximately 45-fold higher K(m)((ATP)). We have also shown that Lys(103) is important in catalysis since an alanine substitution of this residue ablates catalytic activity. Furthermore, we have shown that this residue is covalently modified by FSBA. Our data, combined with amino acid sequence comparison, suggest a motif of SGDGX(17-21)K is involved in nucleotide binding in the sphingosine kinases. This motif differs in primary sequence from all previously identified nucleotide-binding sites. It does, however, share some sequence and likely structural similarity with the highly conserved glycine-rich loop, which is known to be involved in anchoring and positioning the nucleotide in the catalytic site of many protein kinases.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine/analogs & derivatives , Nucleotides/chemistry , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Adenosine/pharmacology , Adenosine Triphosphate/pharmacology , Affinity Labels/pharmacology , Alanine/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Azides/pharmacology , Baculoviridae/metabolism , Binding Sites , Blotting, Western , Calmodulin/metabolism , Catalysis , Catalytic Domain , Cell Line , DNA, Complementary/metabolism , Gene Deletion , Humans , Insecta , Kinetics , Lysine/chemistry , Mass Spectrometry , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Photoaffinity Labels/pharmacology , Protein Binding , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Substrate Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...