Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 30(10): 2401-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20231358

ABSTRACT

We previously reported that vascular endothelial growth factor (VEGF)-dependent activation of phospholipase Cgamma1 (PLCgamma) regulated tube stability by competing with phosphoinositide 3-kinase (PI3K) for their common substrate. Here we describe an additional mechanism by which PLCgamma promoted regression of tubes and blood vessels. Namely, it increased the level of autotaxin (ATX), which is a secreted form of lysophospholipase D that produces lysophosphatidic acid (LPA). LPA promoted motility of endothelial cells, leading to disorganization/regression of tubes in vitro. Furthermore, mice that under- or overexpressed members of this intrinsic destabilization pathway showed either delayed or accelerated, respectively, regression of blood vessels. We conclude that endothelial cells can be instructed to engage a PLCgamma-dependent intrinsic destabilization pathway that results in the production of soluble regression factors such as ATX and LPA. These findings are likely to potentiate ongoing efforts to prevent, manage, and eradicate numerous angiogenesis-based diseases such as proliferative diabetic retinopathy and solid tumors.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lysophospholipids , Multienzyme Complexes/metabolism , Neovascularization, Physiologic/physiology , Phosphodiesterase I/metabolism , Phospholipase C gamma/metabolism , Pyrophosphatases/metabolism , Animals , Calcineurin/metabolism , Cattle , Cells, Cultured , Endothelial Cells/cytology , Enzyme Activation , Eye/blood supply , Humans , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Transgenic , Multienzyme Complexes/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphodiesterase I/genetics , Phospholipase C gamma/genetics , Phosphoric Diester Hydrolases , Pyrophosphatases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
Science ; 323(5919): 1288, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19265000
3.
Lipids Health Dis ; 8: 4, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19222837

ABSTRACT

BACKGROUND: The secreted enzyme autotaxin (ATX) stimulates tumor cell migration, tumorigenesis, angiogenesis, and metastasis. ATX hydrolyzes nucleotides, but its hydrolysis of lysophospholipids to produce lysophosphatidic acid (LPA) accounts for its biological activities. ATX has been identified only as a constitutively active enzyme, and regulation of its activity is largely unexplored. In spite of its presence in plasma along with abundant putative substrate LPC, the product LPA is found in plasma at unexpectedly low concentrations. It is plausible that the LPA-producing activity of ATX is regulated by its expression and by access to substrate(s). For this reason studying the interaction of enzyme with substrate is paramount to understanding the regulation of LPA production. RESULTS: In this study we determine ATX hydrolytic activities toward several artificial and natural substrates. Two novel point mutations near the enzyme active site (H226Q and H434Q) confer attenuated activity toward all substrates tested. The Vmax for LPC compounds depends upon chain length and saturation; but this order does not differ among wild type and mutants. However the mutant forms show disproportionately low activity toward two artificial substrates, pNpTMP and FS-3. The mutant forms did not significantly stimulate migration responses at concentrations that produced a maximum response for WT-ATX, but this defect could be rescued by inclusion of exogenous LPC. CONCLUSION: H226Q-ATX and H434Q-ATX are the first point mutations of ATX/NPP2 demonstrated to differentially impair substrate hydrolysis, with hydrolysis of artificial substrates being disproportionately lower than that of LPC. This implies that H226 and H434 are important for substrate interaction. Assays that rely on hydrolyses of artificial substrates (FS-3 and pNpTMP), or that rely on hydrolysis of cell-derived substrate, might fail to detect certain mutated forms of ATX that are nonetheless capable of producing LPA in the presence of sufficient exogenous substrate. H420Q-ATX could not be differentiated from WT-ATX, indicating that histidine at position 420 is not required for any of the activities of ATX tested in this study.


Subject(s)
Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutant Proteins/metabolism , Phosphodiesterase I/genetics , Phosphodiesterase I/metabolism , Point Mutation/genetics , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Amino Acid Substitution/drug effects , Cell Movement/drug effects , Fatty Acids/metabolism , Humans , Hydrolysis/drug effects , Immunoblotting , Kinetics , Lysophospholipids/pharmacology , Mutant Proteins/genetics , Phosphoric Diester Hydrolases , Substrate Specificity/drug effects
4.
Mol Pharmacol ; 73(2): 587-600, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18006645

ABSTRACT

Lysophosphatidic acid (LPA), via interaction with its G-protein coupled receptors, is involved in various pathological conditions. Extracellular LPA is mainly produced by the enzyme autotaxin (ATX). Using fibroblast-like synoviocytes (FLS) isolated from synovial tissues of patients with rheumatoid arthritis (RA), we studied the expression profile of LPA receptors, LPA-induced cell migration, and interleukin (IL)-8 and IL-6 production. We report that FLS express LPA receptors LPA(1-3). Moreover, exogenously applied LPA induces FLS migration and secretion of IL-8/IL-6, whereas the LPA(3) agonist l-sn-1-O-oleoyl-2-methyl-glyceryl-3-phosphothionate (2S-OMPT) stimulates cytokine synthesis but not cell motility. The LPA-induced FLS motility and cytokine production are suppressed by LPA(1/3) receptor antagonists diacylglycerol pyrophosphate and (S)-phosphoric acid mono-(2-octadec-9-enoylamino-3-[4-(pyridine-2-ylmethoxy)-phenyl]-propyl) ester (VPC32183). Signal transduction through p42/44 mitogen-activated protein kinase (MAPK), p38 MAPK, and Rho kinase is involved in LPA-mediated cytokine secretion, whereas LPA-induced cell motility requires p38 MAPK and Rho kinase but not p42/44 MAPK. Treatment of FLS with tumor necrosis factor-alpha (TNF-alpha) increases LPA(3) mRNA expression and correlates with enhanced LPA- or OMPT-induced cytokine production. LPA-mediated superproduction of cytokines by TNF-alpha-primed FLS is abolished by LPA(1/3) receptor antagonists. We also report the presence of ATX in synovial fluid of patients with RA. LPA(1/3) receptor antagonists and ATX inhibitors reduce the synovial fluid-induced cell motility. Together the data suggest that LPA(1) and LPA(3) may contribute to the pathogenesis of RA through the modulation of FLS migration and cytokine production. The above results provide novel insights into the relevance of LPA receptors in FLS biology and as potential therapeutic targets for the treatment of RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Gene Expression Regulation/physiology , Receptors, Lysophosphatidic Acid/biosynthesis , Receptors, Lysophosphatidic Acid/physiology , Synovial Fluid/cytology , Synovial Fluid/metabolism , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/genetics , Cell Movement/physiology , Cells, Cultured , Cytokines/biosynthesis , Humans , Receptors, Lysophosphatidic Acid/genetics , Synovial Fluid/physiology
5.
Cell Signal ; 19(6): 1328-38, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17307336

ABSTRACT

Lysophosphatidic acid (LPA) stimulates sphingosine-1-phosphate (S1P)-sensitive motility in NIH3T3 clone7 cells. S1P inhibits motility only when added to the bottom well of the Boyden chamber, suggesting that pseudopodia can respond to their microenvironment. In order to study and localize this effect, we utilized a Transwell insert system to isolate pseudopodia. LPA stimulates protrusion of pseudopodia that are enriched in RhoA compared to cell bodies. Removal of LPA results in slow retraction with loss of vinculin-rich adhesion complexes and prolonged activation of RhoA. However, RhoA, ROCK and mDia are not required for this process. In contrast, rapid retraction, induced by adding S1P to the bottom well, is associated with a quick spike of activated RhoA and coalescence of adhesion complexes that colocalize with the ends of stress fibers. S1P-induced retraction requires RhoA and ROCK but is only delayed by inhibition of mDia. These data indicate that pseudopodia sense and integrate signals initiated by localized bioactive lipids, affecting both cellular polarity and their own function in motility.


Subject(s)
Lysophospholipids/pharmacology , Pseudopodia/drug effects , Pseudopodia/enzymology , Sphingosine/analogs & derivatives , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Cell Polarity/drug effects , Enzyme Activation/drug effects , Formins , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microtubules/drug effects , Microtubules/metabolism , NIH 3T3 Cells , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Sphingosine/pharmacology , Vinculin/metabolism , rac GTP-Binding Proteins/metabolism , rho-Associated Kinases , rhoA GTP-Binding Protein/antagonists & inhibitors
6.
Cancer Lett ; 250(1): 53-62, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17189669

ABSTRACT

The histone deacetylase inhibitor, trichostatin A (TSA), and the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (Aza-dC), induced epigenetic regulation of sphingosine-1-phosphate (S1P) receptors in human melanoma cells, switching S1P from motility inhibitor to stimulator. Quantitative PCR revealed increased expression of S1P(1) and S1P(3), associated with S1P-induced chemotaxis, and decreased expression of S1P(2), associated with motility inhibition. Expression of lysophosphatidic acid (LPA) receptors was less affected. The TSA effect was reversible suggesting no mutational change, and Aza-dC treatment resulted in demethylation of a putative S1P(1) promoter. S1P receptors, therefore, appear to be susceptible to epigenetic regulation, accompanied by altered cellular functionality.


Subject(s)
Azacitidine/analogs & derivatives , Cell Movement/drug effects , Epigenesis, Genetic , Hydroxamic Acids/pharmacology , Lysophospholipids/pharmacology , Melanoma/genetics , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , DNA Methylation , Decitabine , Gene Expression Regulation , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Lysophospholipid , Sphingosine/pharmacology
7.
Mol Cell Biol ; 26(4): 1297-306, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449643

ABSTRACT

Programmed cell death 4 (Pdcd4) suppresses neoplastic transformation by inhibiting the activation of c-Jun and consequently AP-1-dependent transcription. We report that Pdcd4 blocks c-Jun activation by inhibiting the expression of mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1)/hematopoietic progenitor kinase 1, a kinase upstream of Jun N-terminal kinase (JNK). cDNA microarray analysis of Pdcd4-overexpressing RKO human colon carcinoma cells revealed MAP4K1 as the sole target of Pdcd4 on the JNK activation pathway. Cotransfection of a MAP4K1 promoter-reporter with Pdcd4 demonstrated inhibition of transcription from the MAP4K1 promoter. Ectopic expression of Pdcd4 in metastatic RKO cells suppressed invasion. MAP4K1 activity is functionally significant in invasion, as overexpression of a dominant negative MAP4K1 (dnMAP4K1) mutant in RKO cells inhibited not only c-Jun activation but also invasion. Overexpression of a MAP4K1 cDNA in Pdcd4-transfected cells rescued the kinase activity of JNK. Thus, Pdcd4 suppresses tumor progression in human colon carcinoma cells by the novel mechanism of down-regulating MAP4K1 transcription, with consequent inhibition of c-Jun activation and AP-1-dependent transcription.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Colonic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Base Sequence , Cell Line, Tumor , Cell Movement , Cloning, Molecular , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA, Neoplasm/genetics , Down-Regulation , Enzyme Activation , Extracellular Matrix/enzymology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transfection
8.
Biochem Biophys Res Commun ; 340(1): 175-82, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16356477

ABSTRACT

We recently coined CST6 as a novel candidate tumor suppressor gene for breast cancer. CST6 indeed is expressed in the normal human breast epithelium, but little or not at all in breast carcinomas and breast cancer cell lines. Moreover, ectopic expression of CST6 in human breast cancer cells suppressed cell proliferation, migration, invasion, and orthotopic tumor growth. To obtain insights into the molecular mechanism by which CST6 exhibits its pleiotropic effects on tumor cells, we compared global gene expression profiles in mock- and CST6-transfected human MDA-MB-435S cells. Out of 12,625 transcript species, 61 showed altered expression. These included genes for extracellular matrix components, cytokines, kinases, and phosphatases, as well as several key transcription factors. TaqMan PCR assays were used to confirm the microarray data for 7 out of 11 genes. One down-regulated gene product, secreted autotaxin/lyso-phospholipase D, was of particular interest because its down-regulation by CST6 could explain most of CST6's effect on the breast cancer cells. This study thus provides the first evidence that CST6 plays a role in the modulation of genes, particularly, genes that are highly relevant to breast cancer progression.


Subject(s)
Breast Neoplasms/metabolism , Cystatins/metabolism , Gene Expression Regulation, Neoplastic , Glucose-6-Phosphate Isomerase/metabolism , Glycoproteins/metabolism , Multienzyme Complexes/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/blood supply , Cell Line, Tumor , Cystatin M , Down-Regulation , Humans , Mitosis , Phosphodiesterase I , Phosphoric Diester Hydrolases , Pyrophosphatases
9.
Biochem Biophys Res Commun ; 337(3): 967-75, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16219296

ABSTRACT

Autotaxin (ATX) was originally identified as a potent tumor cell motility-stimulating factor that displays multiple enzymatic activities including ATPase, Type I nucleotide pyrophosphatase/phosphodiesterase, and lysophospholipase D, depending on its substrates. We demonstrate herein that ATX is a key regulator of extracellular lysophosphatidic acid (LPA) that can act as survival factor, in addition to its mitogenic activity in mouse fibroblasts. Introduction of atx gene into NIH3T3 cells resulted in resistance to conditional apoptosis induced by serum-deprivation, and exogenous ATX protein prevented cells from death by starvation. Flow cytometric analysis showed that co-treatment of ATX with lysophosphatidylcholine as substrate rescued NIH3T3 cells from cellular apoptosis, and this survival activity of ATX was also demonstrated by caspase-3 degradation and PARP cleavage resulting from the enzymatic activity of extracellular ATX. Furthermore, the effect of ATX in preventing apoptosis appears to be mediated through the G-protein-coupled receptor pathway followed by the activation of phosphoinositide 3-kinase and Akt pathway leading to enhanced cell survival. These findings provide novel insights into understanding the functions of ATX as a key regulator of bioactive phospholipids and suggest interventions to correct dysfunction in conditions of tumor cell growth and metastasis.


Subject(s)
Apoptosis/drug effects , Fibroblasts/metabolism , Lysophospholipids/metabolism , Multienzyme Complexes/administration & dosage , Phosphodiesterase I/administration & dosage , Phosphoric Diester Hydrolases/drug effects , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/administration & dosage , Serum Albumin/metabolism , Animals , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Mice , NIH 3T3 Cells , Substrate Specificity
10.
Physiology (Bethesda) ; 20: 194-200, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15888576

ABSTRACT

Recent advances in understanding the complex biology of the microenvironment that underlies tumor invasion and migration have revealed novel and promising therapeutic targets. Pharmacological blockade of intra- and extracellular signaling events that regulate migration and survival of multiple cell types may disrupt the host-tumor conspiracy that allows escape from normal developmental regulation.


Subject(s)
Cell Movement/physiology , Neoplasm Invasiveness/physiopathology , Neoplasms/pathology , Neoplasms/physiopathology , Animals , Humans
11.
Lipids Health Dis ; 4: 5, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15737239

ABSTRACT

BACKGROUND: Autotaxin (ATX, NPP-2), originally purified as a potent tumor cell motility factor, is now known to be the long-sought plasma lysophospholipase D (LPLD). The integrity of the enzymatic active site, including three crucial histidine moieties, is required for motility stimulation, as well as LPLD and 5'nucleotide phosphodiesterase (PDE) activities. Except for relatively non-specific chelation agents, there are no known inhibitors of the ATX LPLD activity. RESULTS: We show that millimolar concentrations of L-histidine inhibit ATX-stimulated but not LPA-stimulated motility in two tumor cell lines, as well as inhibiting enzymatic activities. Inhibition is reversed by 20-fold lower concentrations of zinc salt. L-histidine has no significant effect on the Km of LPLD, but reduces the Vmax by greater than 50%, acting as a non-competitive inhibitor. Several histidine analogs also inhibit the LPLD activity of ATX; however, none has greater potency than L-histidine and all decrease cell viability or adhesion. CONCLUSION: L-histidine inhibition of LPLD is not a simple stoichiometric chelation of metal ions but is more likely a complex interaction with a variety of moieties, including the metal cation, at or near the active site. The inhibitory effect of L-histidine requires all three major functional groups of histidine: the alpha amino group, the alpha carboxyl group, and the metal-binding imidazole side chain. Because of LPA's involvement in pathological processes, regulation of its formation by ATX may give insight into possible novel therapeutic approaches.


Subject(s)
Cytokines/pharmacology , Histidine/pharmacology , Lysophospholipids/biosynthesis , Multienzyme Complexes/pharmacology , Neoplasms/metabolism , Phosphodiesterase I/pharmacology , Pyrophosphatases/pharmacology , Cations, Divalent/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chelating Agents/pharmacology , Enzyme Activation/drug effects , Histidine/analogs & derivatives , Humans , Molecular Structure , Neoplasms/pathology , Phosphoric Diester Hydrolases/metabolism , Substrate Specificity , Zinc/chemistry , Zinc/pharmacology
12.
Indian J Med Res ; 119 Suppl: 88-94, 2004 May.
Article in English | MEDLINE | ID: mdl-15232170

ABSTRACT

BACKGROUND & OBJECTIVES: To develop a broad strain coverage GAS vaccine, several strategies have been investigated which included multi-epitope approaches as well as targeting the M protein conserved Cregion. These approaches, however, have relied on the use of adjuvants that are toxic for human application. The development of safe and effective adjuvants for human use is a key issue in the development of effective vaccines. In this study, we investigated the lipid polylysine core peptide (LCP) system as a self-adjuvanting GAS vaccine delivery approach. METHODS: An LCP-GAS construct was synthesised incorporating multiple copies of a protective peptide epitope (J8) from the conserved carboxy terminal C-repeat region of the M protein. B10.BR mice were immunized parenterally with the LCP-J8 construct, with or without conventional adjuvant, prior to the assessment of immunogenicity and the induction of serum opsonic antibodies. RESULTS: Our data demonstrated immunogenicity of LCP-J8 when coadministered in complete Freund's adjuvant (CFA), or administered in the absence of conventional adjuvant. In both cases, immunization led to the induction of high-titre J8 peptide-specific serum IgG antibody responses, and the induction of heterologous opsonic antibodies that did not cross-react with human heart tissue proteins. INTERPRETATION & CONCLUSION: These data indicated the potential of a novel self-adjuvanting LCP vaccine delivery system incorporating a synthetic GAS M protein C-region peptide immunogen in the induction of broadly protective immune responses, and pointed to the potential application of this system in human vaccine development against infectious diseases.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Vaccines/administration & dosage , Lipids/chemistry , Peptides/chemistry , Streptococcus/immunology , Amino Acid Sequence , Antibodies, Bacterial/biosynthesis , Bacterial Vaccines/chemistry , Bacterial Vaccines/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Molecular Sequence Data
13.
Oncogene ; 23(13): 2357-66, 2004 Mar 25.
Article in English | MEDLINE | ID: mdl-14691447

ABSTRACT

We have used chicken cDNA microarrays to investigate gene-expression changes induced during transformation of chick embryo fibroblasts (CEF) by the viral Jun oncoprotein encoded by ASV17. This analysis reveals that v-Jun induces increases and decreases of varying magnitude in the expression of genes involved in diverse cellular functions, most of which have not been detected in previous screens for putative v-Jun targets. In all, 27 individual genes were identified, whose expression is increased threefold or more in v-Jun-transformed cells, including genes involved in energy generation, protein synthesis, and gene transcription. Interestingly, this group includes the hypoxia-inducible factor-1 alpha (Hif-1alpha) transcription factor and the glycolytic enzyme enolase, suggesting that adaptation to hypoxia could play a role in tumorigenesis by v-Jun. We also identified 32 genes whose expression is decreased threefold or more, including chaperones, components of the cytoskeleton, and, unexpectedly, DNA replication factors. The gene whose expression is upregulated most dramatically (approximately 100-fold) encodes Autotaxin (ATX), a secreted tumor motility-promoting factor with lysophospholipase D activity. Strikingly, v-Jun-transformed CEF secrete catalytically active ATX and chemotactic activity, which can be detected in conditioned medium. ATX is not detectably expressed in normal CEF or CEF transformed by the v-Src or v-Myc oncoproteins, indicating that induction of this putative autocrine/paracrine factor is a specific consequence of cell transformation by v-Jun. ATX has been implicated in both angiogenesis and invasion, and could therefore play an important role in tumorigenesis by v-Jun in vivo.


Subject(s)
Glucose-6-Phosphate Isomerase/metabolism , Glycoproteins/metabolism , Multienzyme Complexes , Oncogene Protein p65(gag-jun)/metabolism , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Chick Embryo , Down-Regulation , Oligonucleotide Array Sequence Analysis , Phosphodiesterase I , Pyrophosphatases , Up-Regulation
14.
Cancer Res ; 63(17): 5446-53, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500380

ABSTRACT

Autotaxin (ATX) is an exoenzyme that potently induces tumor cell motility, and enhances experimental metastasis and angiogenesis. ATX was shown recently to be identical to serum lysophospholipase D activity, producing lysophosphatidic acid (LPA) from lyso-glycerophospholipids. LPA, itself a strong chemoattractant for tumor cells, may mediate the actions of ATX. We now extend the substrate specificity to sphingosylphosphorylcholine (SPC), which ATX hydrolyzes to sphingosine-1-phosphate (S1P). Under migration assay conditions, this novel reaction for the production of S1P has a substrate (SPC) K(m) = 0.23 +/- 0.07 mM. In our responder cell lines (NIH3T3 clone7 and A2058), S1P exerts maximal biological effects at concentrations of 10-100 nM and is mimicked in its biological effects by ATX plus SPC. These effects include inhibition of ATX- and LPA-stimulated motility, and elevation of activated Rho. In NIH3T3 clone7 cells stimulated with platelet-derived growth factor and treated with 10-25 nM S1P, motility is not inhibited and activation of Rho is unaffected, indicating that S1P possesses specificity in its effects. The exoenzyme ATX can potentially regulate diverse processes such as motility and angiogenesis via the S1P family of receptors. Because ATX hydrolyzes nucleotides, lyso-glycerophospholipids, and phosphosphingolipids into bioactive products, it possesses the ability, depending on the availability of substrates, to act as positive or negative regulator of receptor-mediated activity in the cellular microenvironment.


Subject(s)
Glucose-6-Phosphate Isomerase/pharmacology , Glycoproteins/pharmacology , Lysophospholipids , Multienzyme Complexes , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/metabolism , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine/metabolism , 3T3 Cells , Animals , COS Cells , Catalysis , Cell Movement/physiology , Chlorocebus aethiops , Hydrolysis/drug effects , Mice , Phosphodiesterase I , Phosphoric Diester Hydrolases , Pyrophosphatases , Receptors, Cell Surface/biosynthesis , Receptors, Lysophospholipid , rho GTP-Binding Proteins/metabolism
15.
Eur J Biochem ; 270(14): 2971-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12846830

ABSTRACT

Diadenosine polyphosphates (ApnAs) act as extracellular signaling molecules in a broad variety of tissues. They were shown to be hydrolyzed by surface-located enzymes in an asymmetric manner, generating AMP and Apn-1 from ApnA. The molecular identity of the enzymes responsible remains unclear. We analyzed the potential of NPP1, NPP2, and NPP3, the three members of the ecto-nucleotide pyrophosphatase/phosphodiesterase family, to hydrolyze the diadenosine polyphosphates diadenosine 5',5"'-P1,P3-triphosphate (Ap3A), diadenosine 5',5"'-P1,P4-tetraphosphate (Ap4A), and diadenosine 5',5"'-P1,P5-pentaphosphate, (Ap5A), and the diguanosine polyphosphate, diguanosine 5',5"'-P1,P4-tetraphosphate (Gp4G). Each of the three enzymes hydrolyzed Ap3A, Ap4A, and Ap5A at comparable rates. Gp4G was hydrolyzed by NPP1 and NPP2 at rates similar to Ap4A, but only at half this rate by NPP3. Hydrolysis was asymmetric, involving the alpha,beta-pyrophosphate bond. ApnA hydrolysis had a very alkaline pH optimum and was inhibited by EDTA. Michaelis constant (Km) values for Ap3A were 5.1 micro m, 8.0 micro m, and 49.5 micro m for NPP1, NPP2, and NPP3, respectively. Our results suggest that NPP1, NPP2, and NPP3 are major enzyme candidates for the hydrolysis of extracellular diadenosine polyphosphates in vertebrate tissues.


Subject(s)
Adenine Nucleotides/metabolism , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism , Adenine Nucleotides/chemistry , Animals , CHO Cells , Calcium Chloride/pharmacology , Catalysis , Cell Membrane/metabolism , Cricetinae , Edetic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Magnesium Chloride/pharmacology , Pyrophosphatases/antagonists & inhibitors , Subcellular Fractions/metabolism , Substrate Specificity , Transfection
16.
Cancer Res ; 63(9): 2042-5, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12727817

ABSTRACT

The exo-enzyme autotaxin/NPP2 (ATX/NPP2) is a potent stimulator of cell migration, invasion, metastasis, and angiogenesis. Recently, ATX/NPP2 was found to possess lysophospholipase D (lyso-LPD) activity, generating the bioactive mediator lysophosphatidic acid from precursors. In the present study, we used site-directed mutagenesis to delineate the active domain of lysophospholipid catalytic activity and to examine potential overlap with the nucleotide phosphodiesterase domain. We found four amino acid residues obligatory for the phosphodiesterase, lyso-PLD, and migration-stimulating activities of ATX/NPP2, suggesting that 5'-nucleotide phosphodiesterase (PDE) and lyso-PLD share a common reaction mechanism and inviting design of enzymatic inhibitors as therapeutic agents for neoplastic disease.


Subject(s)
Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Multienzyme Complexes , Phosphoric Diester Hydrolases/metabolism , Point Mutation , Animals , COS Cells , Cell Movement/genetics , Chlorocebus aethiops , Humans , Mutagenesis, Site-Directed , Phosphodiesterase I , Phosphoric Diester Hydrolases/genetics , Protein Structure, Tertiary , Pyrophosphatases , Receptors, Purinergic P1/physiology , Structure-Activity Relationship
17.
Infect Immun ; 71(5): 2373-83, 2003 May.
Article in English | MEDLINE | ID: mdl-12704107

ABSTRACT

This study demonstrates the effectiveness of a novel self-adjuvanting vaccine delivery system for multiple different synthetic peptide immunogens by use of lipid core peptide (LCP) technology. An LCP formulation incorporating two different protective epitopes of the surface antiphagocytic M protein of group A streptococci (GAS)--the causative agents of rheumatic fever and subsequent rheumatic heart disease--was tested in a murine parenteral immunization and GAS challenge model. Mice were immunized with the LCP-GAS formulation, which contains an M protein amino-terminal type-specific peptide sequence (8830) in combination with a conserved non-host-cross-reactive carboxy-terminal C-region peptide sequence (J8) of the M protein. Our data demonstrated immunogenicity of the LCP-8830-J8 formulation in B10.BR mice when coadministered in complete Freund's adjuvant and in the absence of a conventional adjuvant. In both cases, immunization led to induction of high-titer GAS peptide-specific serum immunoglobulin G antibody responses and induction of highly opsonic antibodies that did not cross-react with human heart tissue proteins. Moreover, mice were completely protected from GAS infection when immunized with LCP-8830-J8 in the presence or absence of a conventional adjuvant. Mice were not protected, however, following immunization with an LCP formulation containing a control peptide from a Schistosoma sp. These data support the potential of LCP technology in the development of novel self-adjuvanting multi-antigen component vaccines and point to the potential application of this system in the development of human vaccines against infectious diseases.


Subject(s)
Antigens, Bacterial , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Peptide Fragments/immunology , Streptococcal Vaccines/administration & dosage , Streptococcus pyogenes/immunology , Vaccines, Synthetic/administration & dosage , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Cross Reactions , Drug Delivery Systems , Female , Immunoglobulin G/blood , Immunoglobulin G/classification , Lipids , Mice , Molecular Sequence Data , Phagocytosis , Streptococcal Vaccines/immunology
18.
Infect Immun ; 70(7): 3953-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065542

ABSTRACT

Cytokine mRNA levels were assessed in Burkholderia pseudomallei-susceptible BALB/c mice and B. pseudomallei-resistant C57BL/6 mice following administration of a sublethal dose of less virulent (LV) B. pseudomallei, a candidate immunogen tested for protection against a highly virulent (HV) challenge. Compared on the basis of the bacterial loads, the cytokine patterns induced by HV and LV B. pseudomallei were similar, involving gamma interferon, interleukin-10, and other cytokines. Partial cross-protection between B. pseudomallei strains is shown to be associated with cytokine profiles involving both type 1 and type 2 cytokines.


Subject(s)
Cytokines/immunology , Melioidosis/prevention & control , Animals , Burkholderia pseudomallei/growth & development , Burkholderia pseudomallei/immunology , Burkholderia pseudomallei/pathogenicity , Cross Reactions , Cytokines/genetics , Disease Models, Animal , Liver/immunology , Liver/microbiology , Melioidosis/immunology , Melioidosis/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/immunology , Spleen/microbiology , Vaccination , Virulence
19.
Vaccine ; 20(21-22): 2816-25, 2002 Jun 21.
Article in English | MEDLINE | ID: mdl-12034109

ABSTRACT

Infection with group A streptococci (GAS) can lead to rheumatic fever (RF) and rheumatic heart disease (RHD) which are a major health concern particularly in indigenous populations worldwide, and especially in Australian Aboriginals. A primary route of GAS infection is via the upper respiratory tract, and therefore, a major goal of research is the development of a mucosal-based GAS vaccine. The majority of the research to date has focused on the GAS M protein since immunity to GAS is mediated by M protein type-specific opsonic antibodies. There are two major impediments to the development of a vaccine-the variability in M proteins and the potential for the induction of an autoimmune response. To develop a safe and broad-based vaccine, we have therefore focused on the GAS M protein conserved C-region, and have identified peptides, J8 and the closely related J8 peptide (J14), which may be important in protective immunity to GAS infection. Using a mucosal animal model system, our data have shown a high degree of throat GAS colonisation in B10.BR mice 24h following intranasal immunisation with the mucosal adjuvant, cholera toxin B subunit (CTB), and/or diptheria toxoid (dT) carrier, or PBS alone, and challenge with the M1 GAS strain. However, GAS colonisation of the throat was significantly reduced following intranasal immunisation of mice with the vaccine candidate J8 conjugated to dT or J14-dT when administered with CTB. Moreover, J8-dT/CTB and J14-dT/CTB-immunised mice had a significantly higher survival when compared to CTB and PBS-immunised control mice. These data indicate that immunity to GAS infection can be evoked by intranasal immunisation with a GAS M protein C-region peptide vaccine that contains a protective B cell epitope and lacks a T cell autoepitope.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Streptococcus pyogenes/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , Conserved Sequence/immunology , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunization , Immunoglobulin A , Immunoglobulin G , Mice , Molecular Sequence Data , Pharynx/microbiology , Streptococcus pyogenes/growth & development , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
20.
Infect Immun ; 70(5): 2734-8, 2002 May.
Article in English | MEDLINE | ID: mdl-11953422

ABSTRACT

The study reported here investigated the immunogenicity and protective potential of a lipid core peptide (LCP) construct containing a conserved region determinant of M protein, defined as peptide J8. Parenteral immunization of mice with LCP-J8 led to the induction of high-titer serum immunoglobulin G J8-specific antibodies when the construct was coadministered with complete Freund's adjuvant (CFA) or administered alone. LCP-J8 in CFA had significantly enhanced immunogenicity compared with the monomeric peptide J8 given in CFA. Moreover, LCP-J8/CFA and LCP-J8 antisera opsonized four different group A streptococcal (GAS) strains, and the antisera did not cross-react with human heart tissue proteins. These data indicate the potential of an LCP-based M protein conserved region GAS vaccine in the induction of broadly protective immune responses in the absence of a conventional adjuvant.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Peptide Fragments/immunology , Streptococcal Vaccines/immunology , Streptococcus pyogenes/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Female , Immunization , Immunoglobulin G/classification , Mice , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL