Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Biochem ; 12: 65, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22177013

ABSTRACT

BACKGROUND: Nucleotide pyrophosphatase/phosphodiesterase 7 (NPP7) is the only member of the mammalian NPP enzyme family that has been confirmed to act as a sphingomyelinase, hydrolyzing sphingomyelin (SM) to form phosphocholine and ceramide. NPP7 additionally hydrolyzes lysophosphatidylcholine (LPC), a substrate preference shared with the NPP2/autotaxin(ATX) and NPP6 mammalian family members. This study utilizes a synergistic combination of molecular modeling validated by experimental site-directed mutagenesis to explore the molecular basis for the unique ability of NPP7 to hydrolyze SM. RESULTS: The catalytic function of NPP7 against SM, LPC, platelet activating factor (PAF) and para-nitrophenylphosphorylcholine (pNPPC) is impaired in the F275A mutant relative to wild type NPP7, but different impacts are noted for mutations at other sites. These results are consistent with a previously described role of F275 to interact with the choline headgroup, where all substrates share a common functionality. The L107F mutation showed enhanced hydrolysis of LPC, PAF and pNPPC but reduced hydrolysis of SM. Modeling suggests this difference can be explained by the gain of cation-pi interactions with the choline headgroups of all four substrates, opposed by increased steric crowding against the sphingoid tail of SM. Modeling also revealed that the long and flexible hydrophobic tails of substrates exhibit considerable dynamic flexibility in the binding pocket, reducing the entropic penalty that might otherwise be incurred upon substrate binding. CONCLUSIONS: Substrate recognition by NPP7 includes several important contributions, ranging from cation-pi interactions between F275 and the choline headgroup of all substrates, to tail-group binding pockets that accommodate the inherent flexibility of the lipid hydrophobic tails. Two contributions to the unique ability of NPP7 to hydrolyze SM were identified. First, the second hydrophobic tail of SM occupies a second hydrophobic binding pocket. Second, the leucine residue present at position 107 contrasts with a conserved phenylalanine in NPP enzymes that do not utilize SM as a substrate, consistent with the observed reduction in SM hydrolysis by the NPP7-L107F mutant.


Subject(s)
Lysophosphatidylcholines/metabolism , Platelet Activating Factor/metabolism , Sphingomyelin Phosphodiesterase/chemistry , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Amino Acid Sequence , Animals , Biocatalysis , Humans , Kinetics , Lysophosphatidylcholines/chemistry , Models, Molecular , Molecular Sequence Data , Platelet Activating Factor/chemistry , Sequence Alignment , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelins/chemistry , Substrate Specificity
2.
J Med Chem ; 53(8): 3095-105, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20349977

ABSTRACT

Autotaxin (ATX) is a secreted glycoprotein with lysophospholipase D (LPLD) activity that generates the bioactive lipid lysophosphatidic acid (LPA) from lysophosphatidylcholine (LPC). Both ATX and LPA have been linked to the promotion and progression of cancer as well as cardiovascular disease and obesity. Despite the fact that ATX inhibitors have the potential to be useful chemotherapeutics for multiple indications, few examples of potent ATX inhibitors are described in the current literature. Here we describe the development of pharmacophore models for the inhibition of ATX by nonlipids and apply these tools to the discovery of additional ATX inhibitors using the NCI open chemical repository database. From this database of > 250,000 compounds, 168 candidate inhibitors were identified. Of these candidates, 106 were available for testing and 33 were identified as active (those that inhibited ATX activity by > or =50% at a single 10 microM concentration), a 31% hit rate. Five of these compounds had IC(50) < 1.5 microM and the most potent compound possessed a K(i) of 271 nM.


Subject(s)
Enzyme Inhibitors/chemistry , Models, Molecular , Multienzyme Complexes/antagonists & inhibitors , Phosphodiesterase I/antagonists & inhibitors , Pyrophosphatases/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Databases, Factual , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Multienzyme Complexes/chemistry , Phosphodiesterase I/chemistry , Phosphoric Diester Hydrolases , Pyrophosphatases/chemistry , Stereoisomerism , Sulfonic Acids/chemistry
3.
J Med Chem ; 53(3): 1056-66, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20041668

ABSTRACT

Autotaxin (ATX, NPP2) has recently been shown to be the lysophospholipase D responsible for synthesis of the bioactive lipid lysophosphatidic acid (LPA). LPA has a well-established role in cancer, and the production of LPA is consistent with the cancer-promoting actions of ATX. Increased ATX and LPA receptor expression have been found in numerous cancer cell types. The current study has combined ligand-based computational approaches (binary quantitative structure-activity relationship), medicinal chemistry, and experimental enzymatic assays to optimize a previously identified small molecule ATX inhibitor, H2L 7905958 (1). Seventy prospective analogs were analyzed via computational screening, from which 30 promising compounds were synthesized and screened to assess efficacy, potency, and mechanism of inhibition. This approach has identified four analogs as potent as or more potent than the lead. The most potent analog displayed an IC(50) of 900 nM with respect to ATX-mediated FS-3 hydrolysis with a K(i) of 700 nM, making this compound approximately 3-fold more potent than the previously described lead.


Subject(s)
Enzyme Inhibitors/pharmacology , Lysophospholipids/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Phosphodiesterase I/chemistry , Phosphodiesterase I/metabolism , Pipemidic Acid/chemistry , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Computer-Aided Design , Drug Evaluation, Preclinical , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Female , Humans , Lysophosphatidylcholines/metabolism , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Quantitative Structure-Activity Relationship , Tumor Cells, Cultured
4.
Bioorg Med Chem ; 18(2): 769-76, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20005724

ABSTRACT

Autotaxin (ATX) is a member of the ecto-nucleotide pyrophosphatase/phosphodiesterase (NPP) family and is a lysophospholipase D that cleaves the choline headgroup from lysophosphatidylcholine to generate the bioactive lipid lysophosphatidic acid (LPA). Enhanced expression of ATX and specific receptors for LPA in numerous cancer cell types has created an interest in studying ATX as a potential chemotherapeutic target. Likewise, ATX has been linked to several additional human diseases including multiple sclerosis, diabetes, obesity, neuropathic pain, and Alzheimer's disease. ATX inhibitors reported to date consist of metal ion chelators, lipid-like product analogs, and non-lipid small molecules. In the current research, we examined the pharmacology of the best of our previously reported non-lipid small molecule inhibitors. Here, these six inhibitors were studied utilizing the synthetic fluorescent lysophospholipid substrate FS-3, the nucleotide substrate pNP-TMP and the endogenous substrate LPC (16:0). All six compounds inhibited FS-3 hydrolysis >or=50%, whereas only three inhibited the hydrolysis of pNP-TMP to this degree. None of the six compounds blocked LPC 16:0 hydrolysis within the desired 50% inhibition range. The most potent analog (5, H2L 7905958) displayed an IC(50) of 1.6microM (K(i)=1.9microM, competitive inhibition) with respect to ATX-mediated FS-3 hydrolysis and an IC(50) of 1.2microM (K(i)=K(i)(')=6.5microM, non-competitive inhibition) against ATX-mediated pNP-TMP hydrolysis. All six inhibitors were specific for ATX as they were without affect on two additional lipid preferring NPP isoforms.


Subject(s)
Enzyme Inhibitors/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Phosphodiesterase I/antagonists & inhibitors , Pyrophosphatases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Kinetics , Molecular Weight , Multienzyme Complexes/metabolism , Phosphodiesterase I/metabolism , Phosphoric Diester Hydrolases , Pyrophosphatases/metabolism , Quantitative Structure-Activity Relationship , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...