Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biochem J ; 453(3): 393-9, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23679855

ABSTRACT

The multifunctional nature of Alzheimer's disease calls for MTDLs (multitarget-directed ligands) to act on different components of the pathology, like the cholinergic dysfunction and amyloid aggregation. Such MTDLs are usually on the basis of cholinesterase inhibitors (e.g. tacrine or huprine) coupled with another active molecule aimed at a different target. To aid in the design of these MTDLs, we report the crystal structures of hAChE (human acetylcholinesterase) in complex with FAS-2 (fasciculin 2) and a hydroxylated derivative of huprine (huprine W), and of hBChE (human butyrylcholinesterase) in complex with tacrine. Huprine W in hAChE and tacrine in hBChE reside in strikingly similar positions highlighting the conservation of key interactions, namely, π-π/cation-π interactions with Trp86 (Trp82), and hydrogen bonding with the main chain carbonyl of the catalytic histidine residue. Huprine W forms additional interactions with hAChE, which explains its superior affinity: the isoquinoline moiety is associated with a group of aromatic residues (Tyr337, Phe338 and Phe295 not present in hBChE) in addition to Trp86; the hydroxyl group is hydrogen bonded to both the catalytic serine residue and residues in the oxyanion hole; and the chlorine substituent is nested in a hydrophobic pocket interacting strongly with Trp439. There is no pocket in hBChE that is able to accommodate the chlorine substituent.


Subject(s)
Alzheimer Disease/enzymology , Aminoquinolines/chemistry , Cholinesterase Inhibitors/chemistry , Cholinesterases/chemistry , Cholinesterases/metabolism , Crystallography, X-Ray/methods , Tacrine/chemistry , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Aminoquinolines/pharmacology , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterases/pharmacology , Humans
2.
Chem Res Toxicol ; 26(2): 280-9, 2013 Feb 18.
Article in English | MEDLINE | ID: mdl-23339663

ABSTRACT

Tri-o-cresyl-phosphate (TOCP) is a common additive in jet engine lubricants and hydraulic fluids suspected to have a role in aerotoxic syndrome in humans. TOCP is metabolized to cresyl saligenin phosphate (CBDP), a potent irreversible inhibitor of butyrylcholinesterase (BChE), a natural bioscavenger present in the bloodstream, and acetylcholinesterase (AChE), the off-switch at cholinergic synapses. Mechanistic details of cholinesterase (ChE) inhibition have, however, remained elusive. Also, the inhibition of AChE by CBDP is unexpected, from a structural standpoint, i.e., considering the narrowness of AChE active site and the bulkiness of CBDP. In the following, we report on kinetic X-ray crystallography experiments that provided 2.7-3.3 Å snapshots of the reaction of CBDP with mouse AChE and human BChE. The series of crystallographic snapshots reveals that AChE and BChE react with the opposite enantiomers and that an induced-fit rearrangement of Phe297 enlarges the active site of AChE upon CBDP binding. Mass spectrometry analysis of aging in either H(2)(16)O or H(2)(18)O furthermore allowed us to identify the inhibition steps, in which water molecules are involved, thus providing insights into the mechanistic details of inhibition. X-ray crystallography and mass spectrometry show the formation of an aged end product formed in both AChE and BChE that cannot be reactivated by current oxime-based therapeutics. Our study thus shows that only prophylactic and symptomatic treatments are viable to counter the inhibition of AChE and BChE by CBDP.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/metabolism , Organophosphorus Compounds/metabolism , Tritolyl Phosphates/metabolism , Acetylcholinesterase/chemistry , Animals , Butyrylcholinesterase/chemistry , CHO Cells , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Cricetinae , Crystallography, X-Ray , Humans , Mass Spectrometry , Mice , Molecular Dynamics Simulation , Organophosphorus Compounds/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tritolyl Phosphates/chemistry
4.
Proc Natl Acad Sci U S A ; 108(38): 15954-9, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21896761

ABSTRACT

Igs offer a versatile template for combinatorial and rational design approaches to the de novo creation of catalytically active proteins. We have used a covalent capture selection strategy to identify biocatalysts from within a human semisynthetic antibody variable fragment library that uses a nucleophilic mechanism. Specific phosphonylation at a single tyrosine within the variable light-chain framework was confirmed in a recombinant IgG construct. High-resolution crystallographic structures of unmodified and phosphonylated Fabs display a 15-Å-deep two-chamber cavity at the interface of variable light (V(L)) and variable heavy (V(H)) fragments having a nucleophilic tyrosine at the base of the site. The depth and structure of the pocket are atypical of antibodies in general but can be compared qualitatively with the catalytic site of cholinesterases. A structurally disordered heavy chain complementary determining region 3 loop, constituting a wall of the cleft, is stabilized after covalent modification by hydrogen bonding to the phosphonate tropinol moiety. These features and presteady state kinetics analysis indicate that an induced fit mechanism operates in this reaction. Mutations of residues located in this stabilized loop do not interfere with direct contacts to the organophosphate ligand but can interrogate second shell interactions, because the H3 loop has a conformation adjusted for binding. Kinetic and thermodynamic parameters along with computational docking support the active site model, including plasticity and simple catalytic components. Although relatively uncomplicated, this catalytic machinery displays both stereo- and chemical selectivity. The organophosphate pesticide paraoxon is hydrolyzed by covalent catalysis with rate-limiting dephosphorylation. This reactibody is, therefore, a kinetically selected protein template that has enzyme-like catalytic attributes.


Subject(s)
Antibodies/chemistry , Immunoglobulin Light Chains/chemistry , Immunoglobulin Variable Region/chemistry , Proteins/chemistry , Algorithms , Amino Acid Sequence , Animals , Antibodies/genetics , Antibodies/metabolism , Binding Sites/genetics , CHO Cells , Calorimetry , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Cricetinae , Cricetulus , Crystallography, X-Ray , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Conformation , Proteins/genetics , Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Thermodynamics
5.
J Biol Chem ; 286(19): 16783-9, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21454498

ABSTRACT

Nerve agents are chiral organophosphate compounds (OPs) that exert their acute toxicity by phosphorylating the catalytic serine of acetylcholinesterase (AChE). The inhibited cholinesterases can be reactivated using oximes, but a spontaneous time-dependent process called aging alters the adduct, leading to resistance toward oxime reactivation. Human butyrylcholinesterase (BChE) functions as a bioscavenger, protecting the cholinergic system against OPs. The stereoselectivity of BChE is an important parameter for its efficiency at scavenging the most toxic OPs enantiomer for AChE. Crystals of BChE inhibited in solution or in cristallo with racemic V-agents (VX, Russian VX, and Chinese VX) systematically show the formation of the P(S) adduct. In this configuration, no catalysis of aging seems possible as confirmed by the three-dimensional structures of the three conjugates incubated over a period exceeding a week. Crystals of BChE soaked in optically pure VX(R)-(+) and VX(S)-(-) solutions lead to the formation of the P(S) and P(R) adduct, respectively. These structural data support an in-line phosphonylation mechanism. Additionally, they show that BChE reacts with VX(R)-(+) in the presence of racemic mixture of V-agents, at odds with earlier kinetic results showing a moderate higher inhibition rate for VX(S)-(-). These combined results suggest that the simultaneous presence of both enantiomers alters the enzyme stereoselectivity. In summary, the three-dimensional data show that BChE reacts preferentially with P(R) enantiomer of V-agents and does not age, in complete contrast to AChE, which is selectively inhibited by the P(S) enantiomer and ages.


Subject(s)
Butyrylcholinesterase/chemistry , Neurotoxins/chemistry , Organothiophosphorus Compounds/chemistry , Acetylcholinesterase/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray/methods , Humans , Molecular Conformation , Organothiophosphorus Compounds/pharmacology , Oximes/chemistry , Protein Conformation , Recombinant Proteins/chemistry , Stereoisomerism
6.
Chem Res Toxicol ; 24(6): 797-808, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21438623

ABSTRACT

Aerotoxic syndrome is assumed to be caused by exposure to tricresyl phosphate (TCP), an antiwear additive in jet engine lubricants and hydraulic fluid. CBDP (2-(ortho-cresyl)-4H-1,2,3-benzodioxaphosphoran-2-one) is the toxic metabolite of triortho-cresylphosphate, a component of TCP. Human butyrylcholinesterase (BChE; EC 3.1.1.8) and human acetylcholinesterase (AChE; EC 3.1.1.7) are irreversibly inhibited by CBDP. The bimolecular rate constants of inhibition (k(i)), determined under pseudo-first-order conditions, displayed a biphasic time course of inhibition with k(i) of 1.6 × 10(8) M(-1) min(-1) and 2.7 × 10(7) M(-1) min(-1) for E and E' forms of BChE. The inhibition constants for AChE were 1 to 2 orders of magnitude slower than those for BChE. CBDP-phosphorylated cholinesterases are nonreactivatable due to ultra fast aging. Mass spectrometry analysis showed an initial BChE adduct with an added mass of 170 Da from cresylphosphate, followed by dealkylation to a structure with an added mass of 80 Da. Mass spectrometry in (18)O-water showed that (18)O was incorporated only during the final aging step to form phospho-serine as the final aged BChE adduct. The crystal structure of CBDP-inhibited BChE confirmed that the phosphate adduct is the ultimate aging product. CBDP is the first organophosphorus agent that leads to a fully dealkylated phospho-serine BChE adduct.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/toxicity , Neurotoxicity Syndromes/metabolism , Organophosphorus Compounds/toxicity , Acetylcholinesterase/chemistry , Air Pollution, Indoor , Aircraft , Butyrylcholinesterase/chemistry , Crystallography, X-Ray , Humans , Kinetics , Mass Spectrometry , Models, Molecular , Neurotoxicity Syndromes/enzymology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/enzymology
7.
Biochem J ; 434(1): 73-82, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21091433

ABSTRACT

OPs (organophosphylates) exert their acute toxicity through inhibition of acetylcholinesterase, by phosphylation of the catalytic serine residue. Engineering of human butyrylcholinesterase, by substitution of a histidine residue for the glycine residue at position 117, led to the creation of OP hydrolase activity. However, the lack of structural information and poor understanding of the hydrolytic mechanism of the G117H mutant has hampered further improvements in the catalytic activity. We have solved the crystallographic structure of the G117H mutant with a variety of ligands in its active site. A sulfate anion bound to the active site suggested the positioning for an OP prior to phosphylation. A fluoride anion was found in the active site when NaF was added to the crystallization buffer. In the fluoride complex, the imidazole ring from the His117 residue was substantially shifted, adopting a relaxed conformation probably close to that of the unliganded mutant enzyme. Additional X-ray structures were obtained from the transient covalent adducts formed upon reaction of the G117H mutant with the OPs echothiophate and VX [ethyl ({2-[bis(propan-2-yl)amino]ethyl}sulfanyl](methyl)phosphinate]. The position of the His117 residue shifted in response to the introduction of these adducts, overlaying the phosphylserine residue. These structural data suggest that the dephosphylation mechanism involves either a substantial conformational change of the His117 residue or an adjacent nucleophilic substitution by water.


Subject(s)
Butyrylcholinesterase/genetics , Butyrylcholinesterase/metabolism , Crystallography, X-Ray , Amino Acid Substitution , Butyrylcholinesterase/chemistry , Catalytic Domain , Echothiophate Iodide/pharmacology , Gene Expression Regulation, Enzymologic , Humans , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Organophosphorus Compounds , Organothiophosphorus Compounds/pharmacology , Protein Engineering , Receptors, Fc
8.
Chem Biol Interact ; 187(1-3): 44-8, 2010 Sep 06.
Article in English | MEDLINE | ID: mdl-20381476

ABSTRACT

Cholinesterases are the main target of organophosphorus nerve agents (OPs). Their inhibition results in cholinergic syndrome and death. The enzymes are inhibited by phosphylation of the catalytic serine enzyme, but can be reactivated by oximes to some extent. However, phosphylated cholinesterases undergo a side reaction that progressively prevents their reactivatability. This unimolecular reaction, termed "aging", has been investigated for decades. It was shown that most OP-ChE conjugates aged by O-dealkylation of an alkoxy substituent of the phosphorus atom, a mechanism involving the stabilization of a transient carbocation. In this paper we present structural data supporting a substitution-based mechanism for aging of the huBChE conjugate of an N-mono-methyl analogue of tabun. This mechanism involves an adjacent nucleophilic attack followed by Berry pseudorotation. A similar adjacent attack and subsequent rearrangement of the transition state have been recently proposed for tabun phosphylation of AChE. We suggest that a similar mechanism is also possible for oxime reactivation of phosphylated cholinesterases. This opens new perspectives in terms of reactivator design.


Subject(s)
Butyrylcholinesterase/metabolism , Cholinesterase Reactivators/pharmacology , Organophosphates/chemistry , Organophosphates/pharmacology , Butyrylcholinesterase/chemistry , Catalytic Domain , Enzyme Activation/drug effects , Humans , Models, Molecular , Phosphorylation/drug effects , Stereoisomerism
9.
J Med Chem ; 53(10): 4002-8, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20408548

ABSTRACT

Tabun is a warfare agent that inhibits human acetylcholinesterase (hAChE) by rapid phosphylation of the catalytic serine. A time-dependent reaction occurs on the tabun adduct, leading to an "aged" enzyme, resistant to oxime reactivators. The aging reaction may proceed via either dealkylation or deamidation, depending on the stereochemistry of the phosphoramidyl adduct. We solved the X-ray structure of aged tabun-hAChE complexed with fasciculin II, and we show that aging proceeds through O-dealkylation, in agreement with the aging mechanism that we determined for tabun-inhibited human butyrylcholinesterase and mouse acetylcholinesterase. Noteworthy, aging and binding of fasciculin II lead to an improved thermostability, resulting from additional stabilizing interactions between the two subdomains that face each other across the active site gorge. This first structure of hAChE inhibited by a nerve agent provides structural insight into the inhibition and aging mechanisms and a structural template for the design of molecules capable of reactivating aged hAChE.


Subject(s)
Acetylcholinesterase/chemistry , Chemical Warfare Agents/chemistry , Cholinesterase Inhibitors/chemistry , Organophosphates/chemistry , Catalytic Domain , Crystallography, X-Ray , Dealkylation , Elapid Venoms/chemistry , Enzyme Stability , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Denaturation , Solutions
10.
Protein Pept Lett ; 16(10): 1215-24, 2009.
Article in English | MEDLINE | ID: mdl-19508180

ABSTRACT

Human butyrylcholinesterase (BuChE) is a serine enzyme present in most organs and plasma. No clear physiological function has yet been assigned to BuChE, but it is a pharmacologically and toxicologically important enzyme that plays a role in degradation of numerous ester-containing drugs and poisonous esters. Thus, BuChE-based bioscavengers are an alternative for prophylaxis and treatments of intoxications by these compounds. Also, BuChE has been integrated in biosensors for detection of organophosphorus compounds and other cholinesterase inhibitors.


Subject(s)
Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Protein Structure, Tertiary , Apnea/enzymology , Apnea/prevention & control , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Butyrylcholinesterase/therapeutic use , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Humans , Kinetics , Structure-Activity Relationship , Substrate Specificity
11.
J Enzyme Inhib Med Chem ; 24(4): 1045-55, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19548794

ABSTRACT

Phosphotriesterase from Pseudomonas diminuta (PTE; EC 3.1.8.1) hydrolyzes organophosphate insecticides and chemical warfare agents. The two zinc cations in the active center can be substituted. Co(2+)-containing PTE is the most efficient but least stable isoform. Gel filtration showed that PTE is monomeric at the submicromolar concentrations used in kinetic assays. The analysis of the recombinant enzyme by X-ray fluorescence spectrometry and CCT-ICP-MS, confirms that recombinant Zn-PTE contains only Zn(2+) whereas Co-PTE has Zn(2+) and Co(2+) in equimolar amount, with Co(2+) most likely in the reported labile beta-site. We noted that recombinant PTE is unstable at low concentrations and must be stabilized by a protein environment. We tested the effect of excess of various metal cofactors on PTE-catalyzed hydrolysis of paraoxon. We notably observed that ZnCl(2) induces a non-competitive partial inhibition of Zn(2+)- and Co(2+)-PTE at pH 8.5 (apparent Ki=155 microM and 52 microM, respectively). Inhibition results from interactions with colloidal Zn(OH)(2) formed in alkaline buffer that alters the catalytic machinery. NiCl(2) caused a similar effect at higher concentrations (apparent Ki=3 mM). We observed that mutating His123, a surface residue close to an alleged allosteric site, dramatically altered the bacterial expression yield of Co(2+)-PTE, Ki for Zn(OH)(2) inhibition, k(cat) (up to 60 fold) for paraoxon hydrolysis, but not K(M). Issues addressed in this work are important for future biotechnological developments of PTE as a detoxifying enzyme.


Subject(s)
Phosphoric Triester Hydrolases/chemistry , Pseudomonas/enzymology , Recombinant Proteins , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Kinetics , Mass Spectrometry , Models, Molecular , Molecular Structure , Phosphoric Triester Hydrolases/genetics , Phosphoric Triester Hydrolases/metabolism , Pseudomonas/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, X-Ray Emission
12.
Biochem J ; 421(1): 97-106, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19368529

ABSTRACT

hBChE [human BChE (butyrylcholinesterase)] naturally scavenges OPs (organophosphates). This bioscavenger is currently in Clinical Phase I for pretreatment of OP intoxication. Phosphylated ChEs (cholinesterases) can undergo a spontaneous time-dependent process called 'aging' during which the conjugate is dealkylated, leading to creation of an enzyme that cannot be reactivated. hBChE inhibited by phosphoramidates such as tabun displays a peculiar resistance to oxime-mediated reactivation. We investigated the basis of oxime resistance of phosphoramidyl-BChE conjugates by determining the kinetics of inhibition, reactivation (obidoxime {1,1'-(oxybis-methylene) bis[4-(hydroxyimino) methyl] pyridinium dichloride}, TMB-4 [1,3-trimethylene-bis(4-hydroxyiminomethylpyridinium) dibromide], HLö 7 {1-[[[4-(aminocarbonyl) pyridinio]methoxy]methyl]-2,4-bis-[(hydroxyimino)methyl] pyridinium dimethanesulfonate)}, HI-6 {1-[[[4-(aminocarbonyl) pyridinio] methoxy] methyl]-2-[(hydroxyimino)methyl]pyridinium dichloride monohydrate} and aging, and the crystal structures of hBChE inhibited by different N-monoalkyl and N,N-dialkyl tabun analogues. The refined structures of aged hBChE conjugates show that aging proceeds through O-dealkylation of the P(R) enantiomer of N,N-diethyl and N-propyl analogues, with subsequent formation of a salt bridge preventing reactivation, similarly to a previous observation made on tabun-ChE conjugates. Interestingly, the N-methyl analogue projects its amino group towards the choline-binding pocket, so that aging proceeds through deamination. This orientation results from a preference of hBChE's acyl-binding pocket for larger than 2-atoms linear substituents. The correlation between the inhibitory potency and the N-monoalkyl chain length is related to increasingly optimized interactions with the acyl-binding pocket as shown by the X-ray structures. These kinetics and X-ray data lead to a structure-activity relationship that highlights steric and electronic effects of the amino substituent of phosphoramidate. This study provides the structural basis to design new oximes capable of reactivating phosphoramidyl-hBChE conjugates after intoxication, notably when hBChE is used as pretreatment, or to design BChE-based catalytic bioscavengers.


Subject(s)
Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Reactivators/pharmacology , Organophosphates/pharmacology , Oximes/pharmacology , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Cholinesterase Reactivators/chemistry , Humans , Kinetics , Models, Molecular , Molecular Structure , Oximes/chemistry , Protein Conformation , Structure-Activity Relationship , Time Factors
13.
J Am Chem Soc ; 130(47): 16011-20, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-18975951

ABSTRACT

Human butyrylcholinesterase (hBChE) hydrolyzes or scavenges a wide range of toxic esters, including heroin, cocaine, carbamate pesticides, organophosphorus pesticides, and nerve agents. Organophosphates (OPs) exert their acute toxicity through inhibition of acetylcholinesterase (AChE) by phosphorylation of the catalytic serine. Phosphylated cholinesterase (ChE) can undergo a spontaneous, time-dependent process called "aging", during which the OP-ChE conjugate is dealkylated. This leads to irreversible inhibition of the enzyme. The inhibition of ChEs by tabun and the subsequent aging reaction are of particular interest, because tabun-ChE conjugates display an extraordinary resistance toward most current oxime reactivators. We investigated the structural basis of oxime resistance for phosphoramidated ChE conjugates by determining the crystal structures of the non-aged and aged forms of hBChE inhibited by tabun, and by updating the refinement of non-aged and aged tabun-inhibited mouse AChE (mAChE). Structures for non-aged and aged tabun-hBChE were refined to 2.3 and 2.1 A, respectively. The refined structures of aged ChE conjugates clearly show that the aging reaction proceeds through O-dealkylation of the P(R) enantiomer of tabun. After dealkylation, the negatively charged oxygen forms a strong salt bridge with protonated His438N epsilon2 that prevents reactivation. Mass spectrometric analysis of the aged tabun-inhibited hBChE showed that both the dimethylamine and ethoxy side chains were missing from the phosphorus. Loss of the ethoxy is consistent with the crystallography results. Loss of the dimethylamine is consistent with acid-catalyzed deamidation during the preparation of the aged adduct for mass spectrometry. The reported 3D data will help in the design of new oximes capable of reactivating tabun-ChE conjugates.


Subject(s)
Cholinesterases/chemistry , Cholinesterases/metabolism , Organophosphates/chemistry , Alkylation , Catalytic Domain , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Crystallography, X-Ray , Humans , Mass Spectrometry , Models, Molecular , Molecular Structure , Phosphorylation
14.
Biochemistry ; 45(51): 15644-53, 2006 Dec 26.
Article in English | MEDLINE | ID: mdl-17176086

ABSTRACT

Kinesins are molecular motors that transport cargo along microtubules (MTs). To move forward the motor must attach to the MT in a defined orientation and detach from it in a process that is driven by ATP hydrolysis. The knowledge of the motor-MT interface is essential for a detailed understanding of how kinesins move along MTs and how they are related to other molecular motors such as myosins or dyneins. We have used the marine natural product adociasulfate-2 (AS-2), previously identified as a MT-competitive inhibitor of conventional kinesin, to infer the secondary structure elements forming the MT interface of two human mitotic kinesins, namely, CENP-E and Eg5. AS-2 inhibits both basal and MT-stimulated ATPase activities of CENP-E (IC50 of 8.6 and 1.3 microM, respectively) and Eg5 (IC50 of 3.5 and 5.3 microM, respectively) and is a MT-competitive inhibitor of CENP-E with a Ki of 0.35 microM. Binding of AS-2 to CENP-E also stimulates the ADP release from the nucleotide-binding pocket. AS-2 is a nonspecific kinesin inhibitor targeting several superfamily members including KHC, MPP1, MKLP1, RabK6, KIFC1, KIFC3, CENP-E, and Eg5. By measuring hydrogen/deuterium exchange with mass spectrometry we have shown that the formation of the CENP-E/AS-2 complex decreases the solvent accessibility of three neighboring peptides on the same face of CENP-E. We deduce that this is the site of MT attachment and conclude that loop L11, helix alpha4, loop L12, helix alpha5, loop L8, and strand beta5 constitute the main MT interface of the CENP-E motor domain. Similarly for Eg5/AS-2, a region of increased solvent accessibility locates the MT interface of Eg5.


Subject(s)
Kinesins/metabolism , Microtubules/metabolism , Sulfuric Acid Esters/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cattle , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster , Haliclona , Humans , Kinesins/chemistry , Microtubules/chemistry , Molecular Sequence Data , Sulfuric Acid Esters/chemistry , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...