Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters










Publication year range
1.
Biochemistry ; 41(9): 2970-81, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11863435

ABSTRACT

Huprine X is a novel acetylcholinesterase (AChE) inhibitor, with one of the highest affinities reported for a reversible inhibitor. It is a synthetic hybrid that contains the 4-aminoquinoline substructure of one anti-Alzheimer drug, tacrine, and a carbobicyclic moiety resembling that of another AChE inhibitor, (-)-huperzine A. Cocrystallization of huprine X with Torpedo californica AChE yielded crystals whose 3D structure was determined to 2.1 A resolution. The inhibitor binds to the anionic site and also hinders access to the esteratic site. Its aromatic portion occupies the same binding site as tacrine, stacking between the aromatic rings of Trp84 and Phe330, whereas the carbobicyclic unit occupies the same binding pocket as (-)-huperzine A. Its chlorine substituent was found to lie in a hydrophobic pocket interacting with rings of the aromatic residues Trp432 and Phe330 and with the methyl groups of Met436 and Ile439. Steady-state inhibition data show that huprine X binds to human AChE and Torpedo AChE 28- and 54-fold, respectively, more tightly than tacrine. This difference stems from the fact that the aminoquinoline moiety of huprine X makes interactions similar to those made by tacrine, but additional bonds to the enzyme are made by the huperzine-like substructure and the chlorine atom. Furthermore, both tacrine and huprine X bind more tightly to Torpedo than to human AChE, suggesting that their quinoline substructures interact better with Phe330 than with Tyr337, the corresponding residue in the human AChE structure. Both (-)-huperzine A and huprine X display slow binding properties, but only binding of the former causes a peptide flip of Gly117.


Subject(s)
Acetylcholinesterase/chemistry , Aminoquinolines/chemistry , Cholinesterase Inhibitors/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Torpedo/metabolism , Alkaloids , Aminoquinolines/pharmacology , Animals , Binding Sites , Chlorine/chemistry , Cholinesterase Inhibitors/pharmacology , Computer Simulation , Crystallization , Crystallography, X-Ray , Heterocyclic Compounds, 4 or More Rings/pharmacology , Kinetics , Ligands , Models, Molecular , Protein Conformation , Sesquiterpenes/chemistry , Species Specificity , Tacrine/chemistry
2.
Biochem J ; 359(Pt 2): 305-13, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11583576

ABSTRACT

The somatic genetic defect in paroxysmal nocturnal haemoglobinuria (PNH) involves a block in the transfer of GlcNAc from UDP-GlcNAc to phosphatidylinositol (PI), the first step in the biosynthetic pathway for glycosylphosphatidylinositols (GPIs). We asked whether an exogenous lipid corresponding to an early intermediate in this pathway can be taken up by cells in culture and proceed through the GPI pathway. This approach could offer a strategy to bypass the block in PNH. To address this question we incubated HeLa D cells with sn-1-alkyl-sn-2-lyso-GlcN-[(3)H]PI (lyso-alkyl-GlcN-[(3)H]PI) for 24 h and analysed the cellular lipids. We found three lipid products: unaltered lyso-alkyl-GlcN-[(3)H]PI, GlcN-[(3)H]PI and GlcN(acyl)[(3)H]PI (GlcN-PI with a fatty acid acyl group on inositol). Since the latter two lipids are intermediates in the GPI biosynthetic pathway, this observation demonstrates that an exogenous lipid can enter and proceed partially through this pathway. However, the conversion of GlcN(acyl)PI to downstream mannosylated GPI intermediates in the GPI pathway was inefficient, both for GlcN(acyl)PI produced from the exogenous lipid as well as from that obtained by metabolic labelling with [(3)H]inositol. We investigated this poor conversion by examining whether GlcN(acyl)PI, radioactively labelled sequentially by [(14)C]inositol and [(3)H]inositol, resided in one compartment and could be readily metabolized to downstream intermediates. Isotope ratios indicated that the turnover of GlcN(acyl)PI was slower than those of either downstream mannosylated GPIs or even GPI anchors on proteins, the final products of GPI pathway. This result is incompatible with the one-compartment model and indicates that GlcN(acyl)PI in HeLa D cells accumulates largely in a compartment that is inert to subsequent mannosylation.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Biological Transport, Active , Cell Compartmentation , Glycosylphosphatidylinositols/chemistry , Glycosylphosphatidylinositols/pharmacology , HeLa Cells , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/metabolism , Humans , Models, Biological
3.
J Cell Biochem ; 82(2): 234-45, 2001.
Article in English | MEDLINE | ID: mdl-11527149

ABSTRACT

Isolated glycosylphosphatidylinositol (GPI)-anchored proteins, when added to cells in vitro, incorporate into their surface membranes and, once incorporated, exert their native functions. Virtually any protein of interest, if expressed as a GPI-reanchored derivative, can be modified to acquire this capacity. Such transfer of proteins directly to cells, termed "protein engineering" or "painting" constitutes an alternative to conventional gene transfer for manipulating cell surface composition that has many potential applications. Previous studies with incorporated GPI-anchored proteins have focused almost entirely on their extracellular functions. In this study, biotinylated human erythrocyte (E(hu)) decay accelerating factor, E(hu) acetylcholinesterase, and GPI-reanchored murine B7-1 and B7-2 were used as GPI-anchored reporters to characterize their plasma membrane organization and cell signalling properties following addition to Hela or Chinese hamster ovary cells. For each reporter, three types of cell-association were documented; (1) nonphysiological attachment and/or incomplete insertion, (2) uncomplexed membrane integration, and (3) organization into TX-100-resistant microdomains. Transit from the first two compartments into the third, i.e., microdomains, progressed slowly, continuing even after 24 to 36 h and was associated with the acquisition of cell signalling capacity. All four reporters, incorporated in two different detergents, behaved similarly. When organized in microdomains, caveolin and other GPI proteins co-isolated with the incorporated reporter. These results have implications for protein engineering of cells in general, and in particular, for cells such as modified tumor cell immunogens administered to patients for therapeutic purposes.


Subject(s)
Cell Membrane/metabolism , Glycosylphosphatidylinositols/pharmacology , Acetylcholinesterase/metabolism , Animals , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen , Biotinylation , CD55 Antigens/metabolism , CHO Cells , Cancer Vaccines , Cell Compartmentation , Centrifugation, Density Gradient , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel , Glycosylphosphatidylinositols/administration & dosage , Glycosylphosphatidylinositols/metabolism , HeLa Cells , Humans , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Mice , Phosphorylation , Protein Engineering , Protein Processing, Post-Translational , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection
4.
Biochemistry ; 40(19): 5682-90, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11341833

ABSTRACT

Cholinesterases use a Glu-His-Ser catalytic triad to enhance the nucleophilicity of the catalytic serine. We have previously shown by proton NMR that horse serum butyryl cholinesterase, like serine proteases, forms a short, strong hydrogen bond (SSHB) between the Glu-His pair upon binding mechanism-based inhibitors, which form tetrahedral adducts, analogous to the tetrahedral intermediates in catalysis [Viragh, C., et al. (2000) Biochemistry 39, 16200-16205]. We now extend these studies to human acetylcholinesterase, a 136 kDa homodimer. The free enzyme at pH 7.5 shows a proton resonance at 14.4 ppm assigned to an imidazole NH of the active-site histidine, but no deshielded proton resonances between 15 and 21 ppm. Addition of a 3-fold excess of the mechanism-based inhibitor m-(N,N,N-trimethylammonio)trifluoroacetophenone (TMTFA) induced the complete loss of the 14.4 ppm signal and the appearance of a broad, deshielded resonance of equal intensity with a chemical shift delta of 17.8 ppm and a D/H fractionation factor phi of 0.76 +/- 0.10, consistent with a SSHB between Glu and His of the catalytic triad. From an empirical correlation of delta with hydrogen bond lengths in small crystalline compounds, the length of this SSHB is 2.62 +/- 0.02 A, in agreement with the length of 2.63 +/- 0.03 A, independently obtained from phi. Upon addition of a 3-fold excess of the mechanism-based inhibitor 4-nitrophenyl diethyl phosphate (paraoxon) to the free enzyme at pH 7.5, and subsequent deethylation, two deshielded resonances of unequal intensity appeared at 16.6 and 15.5 ppm, consistent with SSHBs with lengths of 2.63 +/- 0.02 and 2.65 +/- 0.02 A, respectively, suggesting conformational heterogeneity of the active-site histidine as a hydrogen bond donor to either Glu-327 of the catalytic triad or to Glu-199, also in the active site. Conformational heterogeneity was confirmed with the methylphosphonate ester anion adduct of the active-site serine, which showed two deshielded resonances of equal intensity at 16.5 and 15.8 ppm with phi values of 0.47 +/- 0.10 and 0.49 +/- 0.10 corresponding to average hydrogen bond lengths of 2.59 +/- 0.04 and 2.61 +/- 0.04 A, respectively. Similarly, lowering the pH of the free enzyme to 5.1 to protonate the active-site histidine (pK(a) = 6.0 +/- 0.4) resulted in the appearance of two deshielded resonances, at 17.7 and 16.4 ppm, consistent with SSHBs with lengths of 2.62 +/- 0.02 and 2.63 +/- 0.02 A, respectively. The NMR-derived distances agree with those found in the X-ray structures of the homologous acetylcholinesterase from Torpedo californica complexed with TMTFA (2.66 +/- 0.28 A) and sarin (2.53 +/- 0.26 A) and at low pH (2.52 +/- 0.25 A). However, the order of magnitude greater precision of the NMR-derived distances establishes the presence of SSHBs at the active site of acetylcholinesterase, and detect conformational heterogeneity of the active-site histidine. We suggest that the high catalytic power of cholinesterases results in part from the formation of a SSHB between Glu and His of the catalytic triad.


Subject(s)
Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protons , Acetophenones/chemistry , Animals , Binding Sites , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Dimerization , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Nitrophenols , Nuclear Magnetic Resonance, Biomolecular/methods , Organophosphonates/chemistry , Paraoxon/chemistry , Recombinant Proteins/chemistry , Torpedo
5.
J Biol Chem ; 276(26): 23282-7, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11313335

ABSTRACT

Three-dimensional structures of acetylcholinesterase (AChE) reveal a narrow and deep active site gorge with two sites of ligand binding, an acylation site at the base of the gorge, and a peripheral site near the gorge entrance. Recent studies have shown that the peripheral site contributes to catalytic efficiency by transiently binding substrates on their way to the acylation site, but the question of whether the peripheral site makes other contributions to the catalytic process remains open. A possible role for ligand binding to the peripheral site that has long been considered is the initiation of a conformational change that is transmitted allosterically to the acylation site to alter catalysis. However, evidence for conformational interactions between these sites has been difficult to obtain. Here we report that thioflavin T, a fluorophore widely used to detect amyloid structure in proteins, binds selectively to the AChE peripheral site with an equilibrium dissociation constant of 1.0 microm. The fluorescence of the bound thioflavin T is increased more than 1000-fold over that of unbound thioflavin T, the greatest enhancement of fluorescence for the binding of a fluorophore to AChE yet observed. Furthermore, when the acylation site ligands edrophonium or m-(N, N,N-trimethylammonio)trifluoroacetophenone form ternary complexes with AChE and thioflavin T, the fluorescence is quenched by factors of 2.7-4.2. The observation of this partial quenching of thioflavin T fluorescence is a major advance in the study of AChE for two reasons. First, it allows thioflavin T to be used as a reporter for ligand reactions at the acylation site. Second, it indicates that ligand binding to the acylation site initiates a change in the local AChE conformation at the peripheral site that quenches the fluorescence of bound thioflavin T. The data provide strong evidence in support of a conformational interaction between the two AChE sites.


Subject(s)
Acetylcholinesterase/metabolism , Fluorescent Dyes/chemistry , Thiazoles/chemistry , Acetophenones/metabolism , Acylation , Benzothiazoles , Binding Sites , Cholinesterase Inhibitors/metabolism , Coloring Agents/chemistry , Dose-Response Relationship, Drug , Edrophonium/metabolism , Fluorescent Dyes/metabolism , Fluorescent Dyes/pharmacology , Humans , Propidium/chemistry , Protein Conformation , Thiazoles/metabolism , Thiazoles/pharmacology
6.
Mol Pharmacol ; 59(3): 619-26, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179458

ABSTRACT

The beta-site amyloid precursor protein-cleaving enzyme (BACE) cleaves the amyloid precursor protein to produce the N terminus of the amyloid beta peptide, a major component of the plaques found in the brains of Alzheimer's disease patients. Sequence analysis of BACE indicates that the protein contains the consensus sequences found in most known aspartyl proteases, but otherwise has only modest homology with aspartyl proteases of known three-dimensional structure (i.e., pepsin, renin, or cathepsin D). Because BACE has been shown to be one of the two proteolytic activities responsible for the production of the Abeta peptide, this enzyme is a prime target for the design of therapeutic agents aimed at reducing Abeta for the treatment of Alzheimer's disease. Toward this ultimate goal, we have expressed a recombinant, truncated human BACE in a Drosophila melanogaster S2 cell expression system to generate high levels of secreted BACE protein. The protein was convenient to purify and was enzymatically active and specific for cleaving the beta-secretase site of human APP, as demonstrated with soluble APP as the substrate in novel sandwich enzyme-linked immunosorbent assay and Western blot assays. Further kinetic analysis revealed no catalytic differences between this recombinant, secreted BACE, and brain BACE. Both showed a strong preference for substrates that contained the Swedish mutation, where NL is substituted for KM immediately upstream of the cleavage site, relative to the wild-type sequence, and both showed the same extent of inhibition by a peptide-based inhibitor. The capability to produce large quantities of BACE enzyme will facilitate protein structure determination and inhibitor development efforts that may lead to the evolution of useful Alzheimer's disease treatments.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Peptide Hydrolases/metabolism , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Cells, Cultured , Chromatography, High Pressure Liquid , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Endopeptidases , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Solubility , Transfection
7.
J Biol Chem ; 275(44): 34260-5, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10954708

ABSTRACT

In mammalian brain, acetylcholinesterase (AChE) exists mostly as a tetramer of 70-kDa catalytic subunits that are linked through disulfide bonds to a hydrophobic subunit P of approximately 20 kDa. To characterize P, we reduced the disulfide bonds in purified bovine brain AChE and sequenced tryptic fragments from bands in the 20-kDa region. We obtained sequences belonging to at least two distinct proteins: the P protein and another protein that was not disulfide-linked to catalytic subunits. Both proteins were recognized in Western blots by antisera raised against specific peptides. We cloned cDNA encoding the second protein in a cDNA library from bovine substantia nigra and obtained rat and human homologs. We call this protein mCutA because of its homology to a bacterial protein (CutA). We could not demonstrate a direct interaction between mCutA and AChE in vitro in transfected cells. However, in a mouse neuroblastoma cell line that produced membrane-bound AChE as an amphiphilic tetramer, the expression of mCutA antisense mRNA eliminated cell surface AChE and decreased the level of amphiphilic tetramer in cell extracts. mCutA therefore appears necessary for the localization of AChE at the cell surface; it may be part of a multicomponent complex that anchors AChE in membranes, together with the hydrophobic P protein.


Subject(s)
Acetylcholinesterase/metabolism , Proteins/metabolism , Acetylcholinesterase/chemistry , Amino Acid Sequence , Animals , Biopolymers , Blotting, Western , Brain/enzymology , Cattle , Cloning, Molecular , Humans , Membrane Proteins , Mice , Molecular Sequence Data , Proteins/chemistry , Proteins/genetics , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured
8.
Protein Sci ; 9(6): 1063-72, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10892800

ABSTRACT

We have crystallized Drosophila melanogaster acetylcholinesterase and solved the structure of the native enzyme and of its complexes with two potent reversible inhibitors, 1,2,3,4-tetrahydro-N-(phenylmethyl)-9-acridinamine and 1,2,3,4-tetrahydro-N-(3-iodophenyl-methyl)-9-acridinamine--all three at 2.7 A resolution. The refined structure of D. melanogaster acetylcholinesterase is similar to that of vertebrate acetylcholinesterases, for example, human, mouse, and fish, in its overall fold, charge distribution, and deep active-site gorge, but some of the surface loops deviate by up to 8 A from their position in the vertebrate structures, and the C-terminal helix is shifted substantially. The active-site gorge of the insect enzyme is significantly narrower than that of Torpedo californica AChE, and its trajectory is shifted several angstroms. The volume of the lower part of the gorge of the insect enzyme is approximately 50% of that of the vertebrate enzyme. Upon binding of either of the two inhibitors, nine aromatic side chains within the active-site gorge change their conformation so as to interact with the inhibitors. Some differences in activity and specificity between the insect and vertebrate enzymes can be explained by comparison of their three-dimensional structures.


Subject(s)
Acetylcholinesterase/chemistry , Aminoacridines/chemistry , Cholinesterase Inhibitors/chemistry , Drosophila melanogaster/enzymology , Acetylcholinesterase/metabolism , Amino Acid Sequence , Aminoacridines/metabolism , Animals , Cholinesterase Inhibitors/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
9.
Biochemistry ; 39(26): 7753-63, 2000 Jul 04.
Article in English | MEDLINE | ID: mdl-10869180

ABSTRACT

Studies of ligand binding to acetylcholinesterase (AChE) have demonstrated two sites of interaction. An acyl-enzyme intermediate is formed at the acylation site, and catalytic activity can be inhibited by ligand binding to a peripheral site. The three-dimensional structures of AChE-ligand complexes reveal a narrow and deep active site gorge and indicate that ligands specific for the acylation site at the base of the gorge must first traverse the peripheral site near the gorge entrance. In recent studies attempting to clarify the role of the peripheral site in the catalytic pathway for AChE, we showed that ligands which bind specifically to the peripheral site can slow the rates at which other ligands enter and exit the acylation site, a feature we called steric blockade [Szegletes, T., Mallender, W. D., and Rosenberry, T. L. (1998) Biochemistry 37, 4206-4216]. We also demonstrated that cationic substrates can form a low-affinity complex at the peripheral site that accelerates catalytic hydrolysis at low substrate concentrations but results in substrate inhibition at high concentrations because of steric blockade of product release [Szegletes, T., Mallender, W. D., Thomas, P. J., and Rosenberry, T. L. (1999) Biochemistry 38, 122-133]. In this report, we demonstrate that a key residue in the human AChE peripheral site with which the substrate acetylthiocholine interacts is D74. We extend our kinetic model to evaluate the substrate affinity for the peripheral site, indicated by the equilibrium dissociation constant K(S), from the dependence of the substrate hydrolysis rate on substrate concentration. For human AChE, a K(S) of 1.9+/-0.7 mM obtained by fitting this substrate inhibition curve agreed with a K(S) of 1.3+/-1.0 mM measured directly from acetylthiocholine inhibition of the binding of the neurotoxin fasciculin to the peripheral site. For Torpedo AChE, a K(S) of 0.5+/- 0.2 mM obtained from substrate inhibition agreed with a K(S) of 0.4+/- 0.2 mM measured with fasciculin. Introduction of the D72G mutation (corresponding to D74G in human AChE) increased the K(S) to 4-10 mM in the Torpedo enzyme and to about 33 mM in the human enzyme. While the turnover number k(cat) was unchanged in the human D74G mutant, the roughly 20-fold decrease in acetylthiocholine affinity for the peripheral site in D74G resulted in a corresponding decrease in k(cat)/K(app), the second-order hydrolysis rate constant, in the mutant. In addition, we show that D74 is important in conveying to the acylation site an inhibitory conformational effect induced by the binding of fasciculin to the peripheral site. This inhibitory effect, measured by the relative decrease in the first-order phosphorylation rate constant k(OP) for the neutral organophosphate 7-[(methylethoxyphosphonyl)oxy]-4-methylcoumarin (EMPC) that resulted from fasciculin binding, decreased from 0.002 in wild-type human AChE to 0.24 in the D74G mutant.


Subject(s)
Acetylcholinesterase/metabolism , Acetylthiocholine/metabolism , Acetylcholinesterase/genetics , Amino Acid Substitution , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Catalysis , Humans , Ligands , Models, Biological , Mutation , Phosphorylation , Recombinant Proteins/metabolism , Substrate Specificity , Torpedo
10.
Mol Pharmacol ; 57(2): 409-17, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10648652

ABSTRACT

Inhibitors of the enzyme acetylcholinesterase (AChE) slow and sometimes reverse the cognitive decline experienced by individuals with Alzheimer's disease. Huperzine A, a natural product used in traditional Chinese herbal medicine, and tacrine (Cognex) are among the potent AChE inhibitors used in this treatment, but the search for more selective inhibitors continues. We report herein the synthesis and characterization of (-)-12-amino-3-chloro-9-ethyl-6,7, 10,11-tetrahydro-7,11-methanocycloocta[b]quinoline hydrochloride (huprine X), a hybrid that combines the carbobicyclic substructure of huperzine A with the 4-aminoquinoline substructure of tacrine. Huprine X inhibited human AChE with an inhibition constant K(I) of 26 pM, indicating that it binds to this enzyme with one of the highest affinities yet reported. Under equivalent assay conditions, this affinity was 180 times that of huperzine A, 1200 times that of tacrine, and 40 times that of E2020 (donepezil, Aricept), the most selective AChE inhibitor currently approved for therapeutic use. The association and dissociation rate constants for huprine X with AChE were determined, and the location of its binding site on the enzyme was probed in competition studies with the peripheral site inhibitor propidium and the acylation site inhibitor edrophonium. Huprine X showed no detectable affinity for the edrophonium-AChE complex. In contrast, huprine X did form a ternary complex with propidium and AChE, although its affinity for the free enzyme was found to be 17 times its affinity for the propidium-AChE complex. These data indicated that huprine X binds to the enzyme acylation site in the active site gorge but interferes slightly with the binding of peripheral site ligands.


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Aminoquinolines/pharmacology , Cholinesterase Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Acetylcholinesterase/drug effects , Acylation , Aminoquinolines/chemical synthesis , Aminoquinolines/therapeutic use , Binding, Competitive , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/therapeutic use , Erythrocytes/enzymology , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Intercalating Agents/pharmacology , Kinetics , Propidium/pharmacology
11.
Biochem J ; 343 Pt 3: 627-35, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10527942

ABSTRACT

Glycosylphosphatidylinositol (GPI)-anchored proteins are resistant to solubilization with Triton X-100 at 4 degrees C, and they can be recovered in Triton-insoluble membranes (TIMs) that float to a characteristic buoyant density. Because the GPI structure itself has been shown to target GPI-anchored proteins to TIMs, we investigated the association of GPI-anchor intermediates with TIMs. GPI-anchor biosynthesis involves a pathway of some 10 steps that take place in the endoplasmic reticulum (ER). These intermediates include glucosaminyl-acylphosphatidylinositol [GlcN-(acyl)PI] and later mannosylated GPIs, denoted H6, H7 and H8, that are present not only in the ER but also in other cell compartments, including the plasma membrane. At least two-thirds of the GlcN-(acyl)PI in HeLa D cells and mannosylated GPIs in K562 cells were found in TIMs. Although previous reports have considered TIMs to be derived primarily from the plasma membrane, we recovered TIMs from subcellular fractions enriched in ER membranes. The ER marker calnexin and GPI-anchored proteins as well as N-acetylglucosaminyl-phosphatidylinositol and mannosylated GPIs were present in ER-TIMs. Interestingly, GlcN-PI and H7 were less enriched in ER-TIM than the other GPIs, suggesting that ER-TIMs might reflect a compartmentalization of the GPI-anchor biosynthetic pathway in the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Glycosylphosphatidylinositols/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Animals , CHO Cells , Carbohydrate Sequence , Cell Fractionation , Choline/metabolism , Cricetinae , Cysteine/metabolism , Endoplasmic Reticulum/ultrastructure , Glucosamine/metabolism , HeLa Cells , Humans , Inositol/metabolism , Intracellular Membranes/ultrastructure , K562 Cells , Mannose/metabolism , Membrane Proteins/isolation & purification , Models, Chemical , Molecular Sequence Data , Polyethylene Glycols , Radioisotope Dilution Technique , Sulfur Radioisotopes , Tritium , Uridine Diphosphate N-Acetylglucosamine/metabolism
12.
J Biol Chem ; 274(38): 26810-4, 1999 Sep 17.
Article in English | MEDLINE | ID: mdl-10480887

ABSTRACT

The Alzheimer's amyloid protein (Abeta) is released from the larger amyloid beta-protein precursor (APP) by unidentified enzymes referred to as beta- and gamma-secretase. beta-Secretase cleaves APP on the amino side of Abeta producing a large secreted derivative (sAPPbeta) and an Abeta-bearing C-terminal derivative that is subsequently cleaved by gamma-secretase to release Abeta. Alternative cleavage of the APP by alpha-secretase at Abeta16/17 releases the secreted derivative sAPPalpha. In yeast, alpha-secretase activity has been attributed to glycosylphosphatidylinositol (GPI)-anchored aspartyl proteases. To examine the role of GPI-anchored proteins, we specifically removed these proteins from the surface of mammalian cells using phosphatidylinositol-specific phospholipase C (PI-PLC). PI-PLC treatment of fetal guinea pig brain cultures substantially reduced the amount of Abeta40 and Abeta42 in the medium but had no effect on sAPPalpha. A mutant CHO cell line (gpi85), which lacks GPI-anchored proteins, secreted lower levels of Abeta40, Abeta42, and sAPPbeta than its parental line (GPI+). When this parental line was treated with PI-PLC, Abeta40, Abeta42, and sAPPbeta decreased to levels similar to those observed in the mutant line, and the mutant line was resistant to these effects of PI-PLC. These findings provide strong evidence that one or more GPI-anchored proteins play an important role in beta-secretase activity and Abeta secretion in mammalian cells. The cell-surface GPI-anchored protein(s) involved in Abeta biogenesis may be excellent therapeutic target(s) in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Glycosylphosphatidylinositols/metabolism , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Brain/enzymology , CHO Cells , Cells, Cultured , Cricetinae , Endopeptidases/metabolism , Enzyme Activation , Guinea Pigs , Humans , Hydrolysis , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Protein Kinase C/metabolism , Type C Phospholipases/metabolism
13.
Chem Biol Interact ; 119-120: 85-97, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10421442

ABSTRACT

The active site gorge of acetylcholinesterase (AChE) contains two sites of ligand binding, an acylation site near the base of the gorge and a peripheral site at its mouth. We recently introduced a steric blockade model which demonstrated that small peripheral site ligands like propidium can inhibit substrate hydrolysis simply by decreasing the substrate association and dissociation rate constants without altering the equilibrium constant for substrate binding to the acylation site. We now employ our nonequilibrium kinetic analysis to extend this model to include blockade of the dissociation of substrate hydrolysis products by bound peripheral site ligand. We also report here that acetylthiocholine can bind to the AChE peripheral site with an equilibrium dissociation constant K(S) of about 1 mM. This value was determined from the effect of the acetylthiocholine concentration on the rate at which fasciculin associates with the peripheral site. When substrate binding to the peripheral site is incorporated into our steric blockade model, hydrolysis rates at low substrate concentration appear to be accelerated while substrate inhibition of hydrolysis occurs at high substrate concentration. The model predicts that hydrolysis rates for substrates which equilibrate with the acylation site prior to the acylation step should not be inhibited by bound peripheral site ligand. Organophosphates equilibrate with AChE prior to phosphorylating the active site serine residue, and as predicted propidium had little effect on the phosphorylation rate constants for the fluorogenic organophosphate ethylmethyl-phosphonylcoumarin (EMPC). The 2nd-order phosphorylation rate constant kOP/K(OP) was decreased 3-fold by a high concentration of propidium and the 1st-order rate constant kOP increased somewhat. In contrast to propidium, when the neurotoxin fasciculin bound to the AChE peripheral site both a steric blockade and a conformational change in the acylation site appeared to occur. With saturating fasciculin, kOP/K(OP) decreased by a factor of more than 750 and kOP decreased 300-fold. These data suggest that new peripheral site ligands may be designed to have selective effects on AChE phosphorylation.


Subject(s)
Acetylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Models, Chemical , Acetylcholine/metabolism , Acetylcholinesterase/blood , Acetylcholinesterase/metabolism , Binding Sites , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/pharmacokinetics , Erythrocytes/enzymology , Humans , Hydrolysis , Indicators and Reagents/metabolism , Indicators and Reagents/pharmacology , Kinetics , Ligands , Phosphorylation , Propidium/metabolism , Propidium/pharmacology , Protein Conformation , Stereoisomerism , Substrate Specificity
14.
J Biol Chem ; 274(13): 8491-9, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085081

ABSTRACT

Structural analysis of acetylcholinesterase (AChE) has revealed two sites of ligand interaction in the active site gorge: an acylation site at the base of the gorge and a peripheral site at its mouth. A goal of our studies is to understand how ligand binding to the peripheral site alters the reactivity of substrates and organophosphates at the acylation site. Kinetic rate constants were determined for the phosphorylation of AChE by two fluorogenic organophosphates, 7-[(diethoxyphosphoryl)oxy]-1-methylquinolinium iodide (DEPQ) and 7-[(methylethoxyphosphonyl)oxy]-4-methylcoumarin (EMPC), by monitoring release of the fluorescent leaving group. Rate constants obtained with human erythrocyte AChE were in good agreement with those obtained for recombinant human AChE produced from a high level Drosophila S2 cell expression system. First-order rate constants kOP were 1,600 +/- 300 min-1 for DEPQ and 150 +/- 11 min-1 for EMPC, and second-order rate constants kOP/KOP were 193 +/- 13 microM-1 min-1 for DEPQ and 0.7-1.0 +/- 0.1 microM-1 min-1 for EMPC. Binding of the small ligand propidium to the AChE peripheral site decreased kOP/KOP by factors of 2-20 for these organophosphates. Such modest inhibitory effects are consistent with our recently proposed steric blockade model (Szegletes, T., Mallender, W. D., and Rosenberry, T. L. (1998) Biochemistry 37, 4206-4216). Moreover, the binding of propidium resulted in a clear increase in kOP for EMPC, suggesting that molecular or electronic strain caused by the proximity of propidium to EMPC in the ternary complex may promote phosphorylation. In contrast, the binding of the polypeptide neurotoxin fasciculin to the peripheral site of AChE dramatically decreased phosphorylation rate constants. Values of kOP/KOP were decreased by factors of 10(3) to 10(5), and kOP was decreased by factors of 300-4,000. Such pronounced inhibition suggested a conformational change in the acylation site induced by fasciculin binding. As a note of caution to other investigators, measurements of phosphorylation of the fasciculin-AChE complex by AChE inactivation gave misleading rate constants because a small fraction of the AChE was resistant to inhibition by fasciculin.


Subject(s)
Acetylcholinesterase/metabolism , Ligands , Organophosphorus Compounds/metabolism , Acylation , Coumarins/pharmacology , Elapid Venoms/pharmacology , Erythrocytes/enzymology , Humans , Kinetics , Molecular Structure , Phosphorylation , Propidium/pharmacology , Protein Binding , Quinolines/pharmacology , Recombinant Proteins/metabolism
15.
Biochemistry ; 38(1): 122-33, 1999 Jan 05.
Article in English | MEDLINE | ID: mdl-9890890

ABSTRACT

Two sites of ligand interaction in acetylcholinesterase (AChE) were first demonstrated in ligand binding studies and later confirmed by crystallography, site-specific mutagenesis, and molecular modeling: an acylation site at the base of the active site gorge and a peripheral site at its mouth. We recently introduced a steric blockade model which demonstrated how small peripheral site ligands such as propidium may inhibit substrate hydrolysis [Szegletes, T., Mallender, W. D., and Rosenberry, T. L. (1998) Biochemistry 37, 4206-4216]. In this model, the only effect of a bound peripheral site ligand is to decrease the association and dissociation rate constants for an acylation site ligand without altering the equilibrium constant for ligand binding to the acylation site. Here, we first provide evidence that not only rate constants for substrates but also dissociation rate constants for their hydrolysis products are decreased by bound peripheral site ligand. Previous reaction schemes for substrate hydrolysis by AChE were extended to include product dissociation steps, and acetylthiocholine hydrolysis rates in the presence of propidium under nonequilibrium conditions were simulated with assigned rate constants in the program SCoP. We next showed that cationic substrates such as acetylthiocholine and 7-acetoxy-N-methylquinolinium (M7A) bind to the peripheral site as well as to the acylation site. The neurotoxin fasciculin was used to report specifically on interactions at the peripheral site. Analysis of inhibition of fasciculin association rates by these substrates revealed KS values of about 1 mM for the peripheral site binding of acetylthiocholine and 0.2 mM for the binding of M7A. The AChE reaction scheme was further extended to include substrate binding to the peripheral site as the initial step in the catalytic pathway. Simulations of the steric blockade model with this scheme were in reasonable agreement with observed substrate inhibition for acetylthiocholine and M7A and with mutual competitive inhibition in mixtures of acetylthiocholine and M7A. Substrate inhibition was explained by blockade of product dissociation when substrate is bound to the peripheral site. However, our analyses indicate that the primary physiologic role of the AChE peripheral site is to accelerate the hydrolysis of acetylcholine at low substrate concentrations.


Subject(s)
Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/metabolism , Acetylthiocholine/metabolism , Binding, Competitive , Catalysis , Cholinesterase Inhibitors/pharmacology , Elapid Venoms/chemistry , Elapid Venoms/metabolism , Humans , Hydrolysis , Ligands , Models, Chemical , Protein Binding/drug effects , Stereoisomerism , Substrate Specificity/drug effects
16.
Biochemistry ; 37(12): 4206-16, 1998 Mar 24.
Article in English | MEDLINE | ID: mdl-9521743

ABSTRACT

The active site gorge of acetylcholinesterase (AChE) contains two sites of ligand binding, an acylation site near the base of the gorge with a catalytic triad characteristic of serine hydrolases, and a peripheral site at the mouth of the gorge some 10-20 A from the acylation site. Many ligands that bind exclusively to the peripheral site inhibit substrate hydrolysis at the acylation site, but the mechanistic interpretation of this inhibition has been unclear. Previous interpretations have been based on analyses of inhibition patterns obtained from steady-state kinetic models that assume equilibrium ligand binding. These analyses indicate that inhibitors bound to the peripheral site decrease acylation and deacylation rate constants and/or decrease substrate affinity at the acylation site by factors of up to 100. Conformational interactions have been proposed to account for such large inhibitory effects transmitted over the distance between the two sites, but site-specific mutagenesis has failed to reveal residues that mediate the proposed conformational linkage. Since examination of individual rate constants in the AChE catalytic pathway reveals that assumptions of equilibrium ligand binding cannot be justified, we introduce here an alternative nonequilibrium analysis of the steady-state inhibition patterns. This analysis incorporates a steric blockade hypothesis which assumes that the only effect of a bound peripheral site ligand is to decrease the association and dissociation rate constants for an acylation site ligand without altering the equilibrium constant for ligand binding to the acylation site. Simulations based on this nonequilibrium steric blockade model were in good agreement with experimental data for inhibition by the peripheral site ligands propidium and gallamine at low concentrations of either acetylthiocholine or phenyl acetate if binding of these ligands slows substrate association and dissociation rate constants by factors of 5-70. Direct measurements with the acylation site ligands huperzine A and m-(N,N, N-trimethylammonio)trifluoroacetophenone showed that bound propidium decreased the association rate constants 49- and 380-fold and the dissociation rate constants 10- and 60-fold, respectively, relative to the rate constants for these acylation site ligands with free AChE, in reasonable agreement with the nonequilibrium steric blockade model. We conclude that this model can account for the inhibition of AChE by small peripheral site ligands such as propidium without invoking any conformational interaction between the peripheral and acylation sites.


Subject(s)
Acetylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Acetophenones/metabolism , Acetophenones/pharmacology , Acylation/drug effects , Alkaloids , Binding Sites/drug effects , Binding, Competitive/drug effects , Enzyme Activation/drug effects , Humans , Hydrolysis/drug effects , Ligands , Models, Chemical , Propidium/metabolism , Propidium/pharmacology , Sesquiterpenes/metabolism , Sesquiterpenes/pharmacology
17.
Biochem J ; 321 ( Pt 3): 837-44, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9032473

ABSTRACT

Glucosaminyl(acyl)phosphatidylinositol [GlcN(acyl)PI], the third intermediate in the mammalian glycosylphosphatidylinositol (GPI) anchor pathway, is undetectable in most cells. This intermediate was previously shown to accumulate, however, in murine lymphoma mutant E and in yeast mutant dpm1, both of which lack dolicholphosphomannose synthase activity. Here we report that a mammalian HeLa S3 subline, denoted D, produces large amounts of GlcN(acyl)PI. The level of GlcN(acyl)PI in this subline is twice that in the murine lymphoma mutant E and 4 times that in the parental S3 line. This HeLa D subline differs from the previously reported mutants that accumulate GlcN(acyl)PI because no defects in the synthesis or utilization of dolicholphosphomannose were found. Kinetic analysis indicated that in this HeLa subline there is an increased rate of synthesis of GlcN(acyl)PI, whereas the rate of metabolism for this GPI is comparable to that in wild-type cells. Furthermore, HeLa D cells accumulate GlcN(acyl)PI without a block in the synthesis of the downstream mannosylated GPI anchor precursors and GPI-anchored proteins. These findings might be relevant for understanding the regulation of the GPI pathway.


Subject(s)
Glucosamine/analogs & derivatives , Glycosylphosphatidylinositols/metabolism , Mannosyltransferases/metabolism , Phosphatidylinositols/metabolism , Chromatography, Gel , Chromatography, Thin Layer , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Inositol/metabolism , Kinetics , Mannosides/biosynthesis , Microscopy, Phase-Contrast , Oligosaccharides/biosynthesis , Radioimmunoassay
18.
Mol Biol Cell ; 7(4): 613-30, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8730103

ABSTRACT

Drosophila has a single glycoinositol phospholipid (GPI)-anchored form of acetylcholinesterase (AChE) encoded by the Ace locus. To assess the role that GPI plays in the physiology, of AChE, we have replaced the wild-type GPI-AChE with a chimeric transmembrane form (TM-AChE) in the nervous system of the fly. Ace null alleles provided a genetic background completely lacking in endogenous GPI-AChE, and Ace minigene P transposon constructs were used to express both GPI- and TM-AChE forms in the tissues where AChE is normally expressed. Control experiments with the GPI-AChE minigene demonstrated a threshold between 9 and 12% of normal AChE activity for adult viability. Ace mutant flies were rescued by GPI-AChE minigene lines that expressed 12-40% of normal activity and were essentially unchanged from wild-type flies in behavior. TM-AChE minigene lines were able to rescue Ace null alleles, although with a slightly higher threshold than that for GPI-AChE. Although rescued flies expressing GPI-AChE at a level of 12% of normal activity were viable, flies expressing 13-16% of normal activity from the TM-AChE transgene died shortly after eclosion. Flies expressing TM-AChE at about 30% of normal levels were essentially unchanged from wild-type flies in gross behavior but had a reduced lifespan secondary to subtle coordination defects. These flies also showed reduced locomotor activity and performed poorly in a grooming assay. However, light level and electron microscopic immunocytochemistry showed no differences in the localization of GPI- and TM-AChE. Furthermore, endogenous and ectopic-induced expression of both AChEs in epithelial tissues of the adult and embryo, respectively, showed that they were sorted identically. Most epithelial cells sorted GPI- and TM-AChE to the apical surface, but cuticle-secreting epithelia sorted both proteins basolaterally. Our data suggest that rather than having a primary role in protein sorting, the GPI anchor or AChE plays some other more subtle cellular role in neuronal physiology.


Subject(s)
Acetylcholinesterase/genetics , Drosophila/enzymology , Glycosylphosphatidylinositols/physiology , Neurons/cytology , Acetylcholinesterase/metabolism , Alleles , Animals , Drosophila/embryology , Drosophila/genetics , Epithelial Cells , Glycosylphosphatidylinositols/genetics , Microscopy, Electron , Motor Activity/genetics , Neurons/enzymology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Tissue Distribution
19.
Mol Biol Cell ; 7(4): 595-611, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8730102

ABSTRACT

Despite advances in understanding the cell biology of glycoinositol phospholipid (GPI)-anchored proteins in cultured cells, the in vivo functions of GPI anchors have remained elusive. We have focused on Drosophila acetylcholinesterase (AChE) as a model GPI-anchored protein that can be manipulated in vivo with sophisticated genetic techniques. In Drosophila, AChE is found only as a GPI-anchored G2 form encoded by the Ace locus on the third chromosome. To pursue our goal of replacing wild-type GPI-anchored AChE with forms that have alternative anchor structures in transgenic files, we report the construction of two secreted forms of Drosophila AChE (SEC1 and SEC2) and a chimeric form (TM-AChE) anchored by the transmembrane and cytoplasmic domains of herpes simplex virus type 1 glycoprotein C. To confirm that the biochemical properties of these AChEs were unchanged from GPI-AChE except as predicted, we made stably transfected Drosophila Schneider Line 2(S2) cells expressing each of the four forms. TM-AChE, SEC1, and SEC2 had the same catalytic activity and quaternary structure as wild type. TM-AChE was expressed as an amphiphilic membrane-bound protein resistant to an enzyme that cleaves GPI-AChE (phosphatidylinositol-specific phospholipase C), and the same percentage of TM-AChE and GPI-AChE was on the cell surface according to immunofluorescence and pharmacological data. SEC1 and SEC2 were constructed by truncating the C-terminal signal peptide initially present in GPI-AChE: in SEC1 the last 25 residues of this 34-residue peptide were deleted while in SEC2 the last 29 were deleted. Both SEC1 and SEC2 were efficiently secreted and are very stable in culture medium; with one cloned SEC1-expressing line, AChE accumulated to as high as 100 mg/liter. Surprisingly, 5-10% of SEC1 was attached to a GPI anchor, but SEC2 showed no GPI anchoring. Since no differences in catalytic activity were observed among the four AChEs, and since the same percentage of GPI-AChE and TM-AChE were on the cell surface, we contend that in vivo experiments in which GPI-AChE is replaced can be interpreted solely on the basis of the altered anchoring domain.


Subject(s)
Acetylcholinesterase/metabolism , Drosophila/enzymology , Glycosylphosphatidylinositols/metabolism , Acetylcholinesterase/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Cell Line , Centrifugation, Density Gradient , DNA, Complementary/chemistry , Echothiophate Iodide/pharmacology , Electrophoresis, Polyacrylamide Gel , Glycosylphosphatidylinositols/genetics , Molecular Sequence Data , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Phosphoric Diester Hydrolases/pharmacology , Protein Processing, Post-Translational , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Signal Transduction , Simplexvirus/genetics , Viral Envelope Proteins/genetics
20.
Biochem J ; 314 ( Pt 3): 817-25, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8615775

ABSTRACT

Purified bovine erythrocyte acetylcholinesterase (AChE) was radiomethylated on its amine groups and incubated with bacterial phosphatidylinositol-specific phospholipase C to remove the lipid portion of the AChE glycoinositol phospholipid (GPI) anchor, and a C-terminal tryptic fragment that contained the residual GPI glycan was isolated by HPLC. Analysis by electrospray-ionization mass spectrometry revealed a parent ion of m/z 3798. The fragmentation patterns produced by collision-induced dissociation mass spectrometry of the +4 and +5 states of the parent ion indicated a 23-amino acid peptide in amide linkage to ethanolamine-P04-Hex-Hex-Hex(PO4-ethanolamine)(HexNAc)-Hex N(Me)2-inositol phosphate. The glycan structure is completely consistent with that obtained previously for the GPI anchor of human erythrocyte AChE except for the addition of the HexNAc substituent. A nearly complete peptide sequence was deduced from the fragmentation patterns, although four assignments were based only on single fragments of very low abundance. To resolve this uncertainty, a segment of bovine genomic DNA corresponding to the C-terminal AChE sequence was amplified by PCR. DNA sequencing established the 23-amino acid peptide sequence to be FLPKLLSATASEAPCTCSGPAHG, in agreement with the MS data and consistent with results from Edman protein sequencing. Dimerization of AChE polypeptides is mediated by intersubunit disulphide bonding in this C-terminal segment, but the bovine AChE contained two cysteine residues in a ...CTC... motif, in contrast with human AChE which contains only a single cysteine in this segment. Although bovine AChE contained no free thiol groups reactive with iodo[14C]acetamide, partial reduction and alkylation with iodo[14C]acetamide revealed that conversion into monomers occurred with an overall incorporation of only one alkyl group per monomer. An identical level of alkylation was observed when dimeric human AChE was converted into monomers by partial reduction. The question of whether the bovine AChE contains one or two intersubunit disulphide linkages is considered.


Subject(s)
Acetylcholinesterase/blood , Acetylcholinesterase/chemistry , DNA, Complementary , Erythrocytes/enzymology , Glycosylphosphatidylinositols/blood , Acetylcholinesterase/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Carbohydrate Sequence , Cattle , Chromatography, High Pressure Liquid , DNA Primers , Disulfides , Exons , Glycosylphosphatidylinositols/chemistry , Glycosylphosphatidylinositols/isolation & purification , Humans , Macromolecular Substances , Mass Spectrometry , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Polymerase Chain Reaction , Rats , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Trypsin
SELECTION OF CITATIONS
SEARCH DETAIL
...