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.
Biochemistry ; 60(12): 908-917, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33721990

ABSTRACT

We report the atomic-resolution (1.3 Å) X-ray crystal structure of an open conformation of the dapE-encoded N-succinyl-l,l-diaminopimelic acid desuccinylase (DapE, EC 3.5.1.18) from Neisseria meningitidis. This structure [Protein Data Bank (PDB) entry 5UEJ] contains two bound sulfate ions in the active site that mimic the binding of the terminal carboxylates of the N-succinyl-l,l-diaminopimelic acid (l,l-SDAP) substrate. We demonstrated inhibition of DapE by sulfate (IC50 = 13.8 ± 2.8 mM). Comparison with other DapE structures in the PDB demonstrates the flexibility of the interdomain connections of this protein. This high-resolution structure was then utilized as the starting point for targeted molecular dynamics experiments revealing the conformational change from the open form to the closed form that occurs when DapE binds l,l-SDAP and cleaves the amide bond. These simulations demonstrated closure from the open to the closed conformation, the change in RMS throughout the closure, and the independence in the movement of the two DapE subunits. This conformational change occurred in two phases with the catalytic domains moving toward the dimerization domains first, followed by a rotation of catalytic domains relative to the dimerization domains. Although there were no targeting forces, the substrate moved closer to the active site and bound more tightly during the closure event.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Amidohydrolases/chemistry , Enzyme Inhibitors/pharmacology , Molecular Dynamics Simulation , Sulfates/pharmacology , Amidohydrolases/metabolism , Crystallography, X-Ray , Neisseria meningitidis/enzymology
2.
Antibiotics (Basel) ; 9(9)2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32933028

ABSTRACT

Inhibitors of the bacterial enzyme dapE-encoded N-succinyl-L,L-diaminopimelic acid desuccinylase (DapE; EC 3.5.1.18) hold promise as antibiotics with a new mechanism of action. Herein we describe the discovery of a new series of indoline sulfonamide DapE inhibitors from a high-throughput screen and the synthesis of a series of analogs. Inhibitory potency was measured by a ninhydrin-based DapE assay recently developed by our group. Molecular docking experiments suggest active site binding with the sulfonamide acting as a zinc-binding group (ZBG).

3.
PLoS One ; 13(4): e0196010, 2018.
Article in English | MEDLINE | ID: mdl-29698518

ABSTRACT

A new enzymatic assay for the bacterial enzyme succinyl-diaminopimelate desuccinylase (DapE, E.C. 3.5.1.18) is described. This assay employs N6-methyl-N2-succinyl-L,L-diaminopimelic acid (N6-methyl-L,L-SDAP) as the substrate with ninhydrin used to detect cleavage of the amide bond of the modified substrate, wherein N6-methylation enables selective detection of the primary amine enzymatic product. Molecular modeling supported preparation of the mono-N6-methylated-L,L-SDAP as an alternate substrate for the assay, given binding in the active site of DapE predicted to be comparable to the endogenous substrate. The alternate substrate for the assay, N6-methyl-L,L-SDAP, was synthesized from the tert-butyl ester of Boc-L-glutamic acid employing a Horner-Wadsworth-Emmons olefination followed by an enantioselective reduction employing Rh(I)(COD)(S,S)-Et-DuPHOS as the chiral catalyst. Validation of the new ninhydrin assay was demonstrated with known inhibitors of DapE from Haemophilus influenza (HiDapE) including captopril (IC50 = 3.4 [± 0.2] µM, 3-mercaptobenzoic acid (IC50 = 21.8 [±2.2] µM, phenylboronic acid (IC50 = 316 [± 23.6] µM, and 2-thiopheneboronic acid (IC50 = 111 [± 16] µM. Based on these data, this assay is simple and robust, and should be amenable to high-throughput screening, which is an important step forward as it opens the door to medicinal chemistry efforts toward the discovery of DapE inhibitors that can function as a new class of antibiotics.


Subject(s)
Amidohydrolases/metabolism , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Enzyme Assays , Spectrophotometry , Amidohydrolases/genetics , Anti-Bacterial Agents/chemistry , Bacterial Proteins/genetics , Binding Sites , Catalysis , Catalytic Domain , Coordination Complexes/chemistry , Diaminopimelic Acid/chemical synthesis , Diaminopimelic Acid/chemistry , Diaminopimelic Acid/metabolism , Haemophilus influenzae/enzymology , Kinetics , Molecular Docking Simulation , Ninhydrin/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Rhodium/chemistry , Stereoisomerism , Substrate Specificity
4.
Biochemistry ; 57(5): 574-584, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29272107

ABSTRACT

The X-ray crystal structure of the dapE-encoded N-succinyl-l,l-diaminopimelic acid desuccinylase from Haemophilus influenzae (HiDapE) bound by the products of hydrolysis, succinic acid and l,l-DAP, was determined at 1.95 Å. Surprisingly, the structure bound to the products revealed that HiDapE undergoes a significant conformational change in which the catalytic domain rotates ∼50° and shifts ∼10.1 Å (as measured at the position of the Zn atoms) relative to the dimerization domain. This heretofore unobserved closed conformation revealed significant movements within the catalytic domain compared to that of wild-type HiDapE, which results in effectively closing off access to the dinuclear Zn(II) active site with the succinate carboxylate moiety bridging the dinculear Zn(II) cluster in a µ-1,3 fashion forming a bis(µ-carboxylato)dizinc(II) core with a Zn-Zn distance of 3.8 Å. Surprisingly, His194.B, which is located on the dimerization domain of the opposing chain ∼10.1 Å from the dinuclear Zn(II) active site, forms a hydrogen bond (2.9 Å) with the oxygen atom of succinic acid bound to Zn2, forming an oxyanion hole. As the closed structure forms upon substrate binding, the movement of His194.B by more than ∼10 Å is critical, based on site-directed mutagenesis data, for activation of the scissile carbonyl carbon of the substrate for nucleophilic attack by a hydroxide nucleophile. Employing the HiDapE product-bound structure as the starting point, a reverse engineering approach called product-based transition-state modeling provided structural models for each major catalytic step. These data provide insight into the catalytic reaction mechanism and also the future design of new, potent inhibitors of DapE enzymes.


Subject(s)
Amidohydrolases/chemistry , Bacterial Proteins/chemistry , Haemophilus influenzae/enzymology , Amidohydrolases/genetics , Amidohydrolases/metabolism , Amino Acid Substitution , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalysis , Catalytic Domain , Crystallography, X-Ray , Diaminopimelic Acid/metabolism , Dimerization , Haemophilus influenzae/genetics , Hydrogen Bonding , Models, Molecular , Mutagenesis, Site-Directed , Neisseria meningitidis/enzymology , Neisseria meningitidis/genetics , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Rotation , Substrate Specificity , Succinic Acid/metabolism , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...