Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Proc Natl Acad Sci U S A ; 116(32): 15907-15913, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31320588

ABSTRACT

Mycobacterium tuberculosis (Mtb) is the etiological agent of tuberculosis. One-fourth of the global population is estimated to be infected with Mtb, accounting for ∼1.3 million deaths in 2017. As part of the immune response to Mtb infection, macrophages produce metabolites with the purpose of inhibiting or killing the bacterial cell. Itaconate is an abundant host metabolite thought to be both an antimicrobial agent and a modulator of the host inflammatory response. However, the exact mode of action of itaconate remains unclear. Here, we show that Mtb has an itaconate dissimilation pathway and that the last enzyme in this pathway, Rv2498c, also participates in l-leucine catabolism. Our results from phylogenetic analysis, in vitro enzymatic assays, X-ray crystallography, and in vivo Mtb experiments, identified Mtb Rv2498c as a bifunctional ß-hydroxyacyl-CoA lyase and that deletion of the rv2498c gene from the Mtb genome resulted in attenuation in a mouse infection model. Altogether, this report describes an itaconate resistance mechanism in Mtb and an l-leucine catabolic pathway that proceeds via an unprecedented (R)-3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) stereospecific route in nature.


Subject(s)
Leucine/metabolism , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Succinates/metabolism , Aerosols , Animals , Biocatalysis , Ligands , Lyases/metabolism , Malates/metabolism , Mice, Inbred C57BL , Phylogeny , Recombinant Proteins/metabolism , Stereoisomerism , Tuberculosis/microbiology , Tuberculosis/pathology
2.
Proc Natl Acad Sci U S A ; 115(15): 3912-3917, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581255

ABSTRACT

Ipilimumab, a monoclonal antibody that recognizes cytotoxic T lymphocyte antigen (CTLA)-4, was the first approved "checkpoint"-blocking anticancer therapy. In mouse tumor models, the response to antibodies against CTLA-4 depends entirely on expression of the Fcγ receptor (FcγR), which may facilitate antibody-dependent cellular phagocytosis, but the contribution of simple CTLA-4 blockade remains unknown. To understand the role of CTLA-4 blockade in the complete absence of Fc-dependent functions, we developed H11, a high-affinity alpaca heavy chain-only antibody fragment (VHH) against CTLA-4. The VHH H11 lacks an Fc portion, binds monovalently to CTLA-4, and inhibits interactions between CTLA-4 and its ligand by occluding the ligand-binding motif on CTLA-4 as shown crystallographically. We used H11 to visualize CTLA-4 expression in vivo using whole-animal immuno-PET, finding that surface-accessible CTLA-4 is largely confined to the tumor microenvironment. Despite this, H11-mediated CTLA-4 blockade has minimal effects on antitumor responses. Installation of the murine IgG2a constant region on H11 dramatically enhances its antitumor response. Coadministration of the monovalent H11 VHH blocks the efficacy of a full-sized therapeutic antibody. We were thus able to demonstrate that CTLA-4-binding antibodies require an Fc domain for antitumor effect.


Subject(s)
CTLA-4 Antigen/immunology , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fragments/administration & dosage , Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , CTLA-4 Antigen/chemistry , Cell Line, Tumor , Disease Models, Animal , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/immunology , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Immunotherapy , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Protein Domains
3.
Proc Natl Acad Sci U S A ; 111(23): 8535-40, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24872444

ABSTRACT

The rate of protein evolution is determined by a combination of selective pressure on protein function and biophysical constraints on protein folding and structure. Determining the relative contributions of these properties is an unsolved problem in molecular evolution with broad implications for protein engineering and function prediction. As a case study, we examined the structural divergence of the rapidly evolving o-succinylbenzoate synthase (OSBS) family, which catalyzes a step in menaquinone synthesis in diverse microorganisms and plants. On average, the OSBS family is much more divergent than other protein families from the same set of species, with the most divergent family members sharing <15% sequence identity. Comparing 11 representative structures revealed that loss of quaternary structure and large deletions or insertions are associated with the family's rapid evolution. Neither of these properties has been investigated in previous studies to identify factors that affect the rate of protein evolution. Intriguingly, one subfamily retained a multimeric quaternary structure and has small insertions and deletions compared with related enzymes that catalyze diverse reactions. Many proteins in this subfamily catalyze both OSBS and N-succinylamino acid racemization (NSAR). Retention of ancestral structural characteristics in the NSAR/OSBS subfamily suggests that the rate of protein evolution is not proportional to the capacity to evolve new protein functions. Instead, structural features that are conserved among proteins with diverse functions might contribute to the evolution of new functions.


Subject(s)
Bacterial Proteins/chemistry , Carbon-Carbon Lyases/chemistry , Genetic Variation , Protein Structure, Quaternary , Bacteria/enzymology , Bacteria/genetics , Bacterial Proteins/classification , Bacterial Proteins/genetics , Carbon-Carbon Lyases/classification , Carbon-Carbon Lyases/genetics , Catalytic Domain , Crystallography, X-Ray , Deinococcus/enzymology , Deinococcus/genetics , Enterococcus faecalis/enzymology , Enterococcus faecalis/genetics , Evolution, Molecular , INDEL Mutation , Listeria/enzymology , Listeria/genetics , Models, Molecular , Phylogeny , Protein Structure, Secondary , Protein Structure, Tertiary , Thermus thermophilus/enzymology , Thermus thermophilus/genetics
4.
J Am Chem Soc ; 138(3): 826-36, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26714575

ABSTRACT

5-Carboxyvanillate decarboxylase (LigW) catalyzes the conversion of 5-carboxyvanillate to vanillate in the biochemical pathway for the degradation of lignin. This enzyme was shown to require Mn(2+) for catalytic activity and the kinetic constants for the decarboxylation of 5-carboxyvanillate by the enzymes from Sphingomonas paucimobilis SYK-6 (kcat = 2.2 s(-1) and kcat/Km = 4.0 × 10(4) M(-1) s(-1)) and Novosphingobium aromaticivorans (kcat = 27 s(-1) and kcat/Km = 1.1 × 10(5) M(-1) s(-1)) were determined. The three-dimensional structures of both enzymes were determined in the presence and absence of ligands bound in the active site. The structure of LigW from N. aromaticivorans, bound with the substrate analogue, 5-nitrovanillate (Kd = 5.0 nM), was determined to a resolution of 1.07 Å. The structure of this complex shows a remarkable enzyme-induced distortion of the nitro-substituent out of the plane of the phenyl ring by approximately 23°. A chemical reaction mechanism for the decarboxylation of 5-carboxyvanillate by LigW was proposed on the basis of the high resolution X-ray structures determined in the presence ligands bound in the active site, mutation of active site residues, and the magnitude of the product isotope effect determined in a mixture of H2O and D2O. In the proposed reaction mechanism the enzyme facilitates the transfer of a proton to C5 of the substrate prior to the decarboxylation step.


Subject(s)
Biocatalysis , Carboxy-Lyases/metabolism , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/chemistry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Models, Molecular , Molecular Structure , Sphingomonadaceae/enzymology , Sphingomonas/enzymology , Substrate Specificity
5.
Biochemistry ; 54(3): 890-7, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25559274

ABSTRACT

N-Formimino-l-glutamate iminohydrolase (HutF), from Pseudomonas aeruginosa with a locus tag of Pa5106 ( gi|15600299 ), is a member of the amidohydrolase superfamily. This enzyme catalyzes the deamination of N-formimino-l-glutamate to N-formyl-l-glutamate and ammonia in the histidine degradation pathway. The crystal structure of Pa5106 was determined in the presence of the inhibitors N-formimino-l-aspartate and N-guanidino-l-glutaric acid at resolutions of 1.9 and 1.4 Å, respectively. The structure of an individual subunit is composed of two domains with the larger domain folding as a distorted (ß/α)8-barrel. The (ß/α)8-barrel domain is composed of eight ß-strands flanked by 11 α-helices, whereas the smaller domain is made up of eight ß-strands. The active site of Pa5106 contains a single zinc atom that is coordinated by His-56, His-58, His-232, and Asp-320. The nucleophilic solvent water molecule coordinates with the zinc atom at a distance of 2.0 Å and is hydrogen bonded to Asp-320 and His-269. The α-carboxylate groups of both inhibitors are hydrogen bonded to the imidazole moiety of His-206, the hydroxyl group of Tyr-121, and the side chain amide group of Gln-61. The side chain carboxylate groups of the two inhibitors are ion-paired with the guanidino groups of Arg-209 and Arg-82. Computational docking of high-energy tetrahedral intermediate forms of the substrate, N-formimino-l-glutamate, to the three-dimensional structure of Pa5106 suggests that this compound likely undergoes a re-faced nucleophilic attack at the formimino group by the metal-bound hydroxide. A catalytic mechanism of the reaction catalyzed by Pa5106 is proposed.


Subject(s)
Amidohydrolases/chemistry , Glutamates/metabolism , Pseudomonas aeruginosa/enzymology , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glutamates/chemistry , Ligands , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Secondary , Zinc/metabolism
6.
Proc Natl Acad Sci U S A ; 109(11): 4122-7, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22392983

ABSTRACT

The rapid advance in genome sequencing presents substantial challenges for protein functional assignment, with half or more of new protein sequences inferred from these genomes having uncertain assignments. The assignment of enzyme function in functionally diverse superfamilies represents a particular challenge, which we address through a combination of computational predictions, enzymology, and structural biology. Here we describe the results of a focused investigation of a group of enzymes in the enolase superfamily that are involved in epimerizing dipeptides. The first members of this group to be functionally characterized were Ala-Glu epimerases in Eschericiha coli and Bacillus subtilis, based on the operon context and enzymological studies; these enzymes are presumed to be involved in peptidoglycan recycling. We have subsequently studied more than 65 related enzymes by computational methods, including homology modeling and metabolite docking, which suggested that many would have divergent specificities;, i.e., they are likely to have different (unknown) biological roles. In addition to the Ala-Phe epimerase specificity reported previously, we describe the prediction and experimental verification of: (i) a new group of presumed Ala-Glu epimerases; (ii) several enzymes with specificity for hydrophobic dipeptides, including one from Cytophaga hutchinsonii that epimerizes D-Ala-D-Ala; and (iii) a small group of enzymes that epimerize cationic dipeptides. Crystal structures for certain of these enzymes further elucidate the structural basis of the specificities. The results highlight the potential of computational methods to guide experimental characterization of enzymes in an automated, large-scale fashion.


Subject(s)
Dipeptides/metabolism , Multigene Family , Phosphopyruvate Hydratase/metabolism , Racemases and Epimerases/metabolism , Sequence Homology, Amino Acid , Catalytic Domain , Cations , Cluster Analysis , Computational Biology , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Racemases and Epimerases/chemistry , Substrate Specificity
7.
Biochemistry ; 53(47): 7426-35, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25384249

ABSTRACT

5-Methylcytosine is found in all domains of life, but the bacterial cytosine deaminase from Escherichia coli (CodA) will not accept 5-methylcytosine as a substrate. Since significant amounts of 5-methylcytosine are produced in both prokaryotes and eukaryotes, this compound must eventually be catabolized and the fragments recycled by enzymes that have yet to be identified. We therefore initiated a comprehensive phylogenetic screen for enzymes that may be capable of deaminating 5-methylcytosine to thymine. From a systematic analysis of sequence homologues of CodA from thousands of bacterial species, we identified putative cytosine deaminases where a "discriminating" residue in the active site, corresponding to Asp-314 in CodA from E. coli, was no longer conserved. Representative examples from Klebsiella pneumoniae (locus tag: Kpn00632), Rhodobacter sphaeroides (locus tag: Rsp0341), and Corynebacterium glutamicum (locus tag: NCgl0075) were demonstrated to efficiently deaminate 5-methylcytosine to thymine with values of kcat/Km of 1.4 × 10(5), 2.9 × 10(4), and 1.1 × 10(3) M(-1) s(-1), respectively. These three enzymes also catalyze the deamination of 5-fluorocytosine to 5-fluorouracil with values of kcat/Km of 1.2 × 10(5), 6.8 × 10(4), and 2.0 × 10(2) M(-1) s(-1), respectively. The three-dimensional structure of Kpn00632 was determined by X-ray diffraction methods with 5-methylcytosine (PDB id: 4R85 ), 5-fluorocytosine (PDB id: 4R88 ), and phosphonocytosine (PDB id: 4R7W ) bound in the active site. When thymine auxotrophs of E. coli express these enzymes, they are capable of growth in media lacking thymine when supplemented with 5-methylcytosine. Expression of these enzymes in E. coli is toxic in the presence of 5-fluorocytosine, due to the efficient transformation to 5-fluorouracil.


Subject(s)
5-Methylcytosine/metabolism , Bacteria/enzymology , Cytosine Deaminase/metabolism , Amino Acid Sequence , Biocatalysis , Catalytic Domain , Cell Line , Cytosine Deaminase/chemistry , Flucytosine/metabolism , Flucytosine/toxicity , Models, Molecular , Molecular Sequence Data , Phylogeny , Thymine/metabolism
8.
Biochemistry ; 53(16): 2722-31, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24697546

ABSTRACT

The continued increase in the size of the protein sequence databases as a result of advances in genome sequencing technology is overwhelming the ability to perform experimental characterization of function. Consequently, functions are assigned to the vast majority of proteins via automated, homology-based methods, with the result that as many as 50% are incorrectly annotated or unannotated ( Schnoes et al. PLoS Comput. Biol. 2009 , 5 ( 12 ), e1000605 ). This manuscript describes a study of the D-mannonate dehydratase (ManD) subgroup of the enolase superfamily (ENS) to investigate how function diverges as sequence diverges. Previously, one member of the subgroup had been experimentally characterized as ManD [dehydration of D-mannonate to 2-keto-3-deoxy-D-mannonate (equivalently, 2-keto-3-deoxy-D-gluconate)]. In this study, 42 additional members were characterized to sample sequence-function space in the ManD subgroup. These were found to differ in both catalytic efficiency and substrate specificity: (1) high efficiency (kcat/KM = 10(3) to 10(4) M(-1) s(-1)) for dehydration of D-mannonate, (2) low efficiency (kcat/KM = 10(1) to 10(2) M(-1) s(-1)) for dehydration of d-mannonate and/or D-gluconate, and 3) no-activity with either D-mannonate or D-gluconate (or any other acid sugar tested). Thus, the ManD subgroup is not isofunctional and includes D-gluconate dehydratases (GlcDs) that are divergent from the GlcDs that have been characterized in the mandelate racemase subgroup of the ENS (Lamble et al. FEBS Lett. 2004 , 576 , 133 - 136 ) (Ahmed et al. Biochem. J. 2005 , 390 , 529 - 540 ). These observations signal caution for functional assignment based on sequence homology and lay the foundation for the studies of the physiological functions of the GlcDs and the promiscuous ManDs/GlcDs.


Subject(s)
Hydro-Lyases/chemistry , Hydro-Lyases/metabolism , Phosphopyruvate Hydratase/metabolism , Catalytic Domain , Crystallography, X-Ray , Gluconates/metabolism , Hydro-Lyases/genetics , Kinetics , Molecular Sequence Data , Mutation , Phosphopyruvate Hydratase/chemistry , Protein Conformation , Structure-Activity Relationship , Substrate Specificity , Sugar Acids/metabolism
9.
Biochemistry ; 52(6): 1101-12, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23327428

ABSTRACT

L-Histidinol phosphate phosphatase (HPP) catalyzes the hydrolysis of L-histidinol phosphate to L-histidinol and inorganic phosphate, the penultimate step in the biosynthesis of L-histidine. HPP from the polymerase and histidinol phosphatase (PHP) family of proteins possesses a trinuclear active site and a distorted (ß/α)(7)-barrel protein fold. This group of enzymes is closely related to the amidohydrolase superfamily of enzymes. The mechanism of phosphomonoester bond hydrolysis by the PHP family of HPP enzymes was addressed. Recombinant HPP from Lactococcus lactis subsp. lactis that was expressed in Escherichia coli contained a mixture of iron and zinc in the active site and had a catalytic efficiency of ~10(3) M(-1) s(-1). Expression of the protein under iron-free conditions resulted in the production of an enzyme with a 2 order of magnitude improvement in catalytic efficiency and a mixture of zinc and manganese in the active site. Solvent isotope and viscosity effects demonstrated that proton transfer steps and product dissociation steps are not rate-limiting. X-ray structures of HPP were determined with sulfate, L-histidinol phosphate, and a complex of L-histidinol and arsenate bound in the active site. These crystal structures and the catalytic properties of variants were used to identify the structural elements required for catalysis and substrate recognition by the HPP family of enzymes within the amidohydrolase superfamily.


Subject(s)
Arsenates/metabolism , Escherichia coli/enzymology , Histidine/metabolism , Histidinol-Phosphatase/chemistry , Lactococcus lactis/enzymology , Amino Acid Sequence , Catalysis , Catalytic Domain , Computational Biology , Crystallography, X-Ray , Escherichia coli/genetics , Histidinol-Phosphatase/genetics , Histidinol-Phosphatase/metabolism , Hydrogen-Ion Concentration , Lactococcus lactis/genetics , Models, Molecular , Molecular Sequence Data , Multigene Family , Mutagenesis, Site-Directed , Mutation/genetics , Recombinant Proteins , Sequence Homology, Amino Acid
10.
Nature ; 448(7155): 775-9, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17603473

ABSTRACT

With many genomes sequenced, a pressing challenge in biology is predicting the function of the proteins that the genes encode. When proteins are unrelated to others of known activity, bioinformatics inference for function becomes problematic. It would thus be useful to interrogate protein structures for function directly. Here, we predict the function of an enzyme of unknown activity, Tm0936 from Thermotoga maritima, by docking high-energy intermediate forms of thousands of candidate metabolites. The docking hit list was dominated by adenine analogues, which appeared to undergo C6-deamination. Four of these, including 5-methylthioadenosine and S-adenosylhomocysteine (SAH), were tested as substrates, and three had substantial catalytic rate constants (10(5) M(-1 )s(-1)). The X-ray crystal structure of the complex between Tm0936 and the product resulting from the deamination of SAH, S-inosylhomocysteine, was determined, and it corresponded closely to the predicted structure. The deaminated products can be further metabolized by T. maritima in a previously uncharacterized SAH degradation pathway. Structure-based docking with high-energy forms of potential substrates may be a useful tool to annotate enzymes for function.


Subject(s)
Enzymes/chemistry , Enzymes/metabolism , Structure-Activity Relationship , Thermotoga maritima/enzymology , Adenine/analogs & derivatives , Adenine/metabolism , Catalysis , Crystallography, X-Ray , Deamination , Deoxyadenosines/metabolism , Homocysteine/analogs & derivatives , Homocysteine/metabolism , Inosine/analogs & derivatives , Inosine/metabolism , Models, Molecular , Nucleoside Deaminases/chemistry , Nucleoside Deaminases/metabolism , Protein Conformation , S-Adenosylhomocysteine/metabolism , Substrate Specificity , Thionucleosides/metabolism
11.
Biochemistry ; 51(8): 1762-73, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22313111

ABSTRACT

Two enzymes of unknown function from the cog1735 subset of the amidohydrolase superfamily (AHS), LMOf2365_2620 (Lmo2620) from Listeria monocytogenes str. 4b F2365 and Bh0225 from Bacillus halodurans C-125, were cloned, expressed, and purified to homogeneity. The catalytic functions of these two enzymes were interrogated by an integrated strategy encompassing bioinformatics, computational docking to three-dimensional crystal structures, and library screening. The three-dimensional structure of Lmo2620 was determined at a resolution of 1.6 Å with two phosphates and a binuclear zinc center in the active site. The proximal phosphate bridges the binuclear metal center and is 7.1 Å from the distal phosphate. The distal phosphate hydrogen bonds with Lys-242, Lys-244, Arg-275, and Tyr-278. Enzymes within cog1735 of the AHS have previously been shown to catalyze the hydrolysis of substituted lactones. Computational docking of the high-energy intermediate form of the KEGG database to the three-dimensional structure of Lmo2620 highly enriched anionic lactones versus other candidate substrates. The active site structure and the computational docking results suggested that probable substrates would likely include phosphorylated sugar lactones. A small library of diacid sugar lactones and phosphorylated sugar lactones was synthesized and tested for substrate activity with Lmo2620 and Bh0225. Two substrates were identified for these enzymes, D-lyxono-1,4-lactone-5-phosphate and l-ribono-1,4-lactone-5-phosphate. The k(cat)/K(m) values for the cobalt-substituted enzymes with these substrates are ~10(5) M(-1) s(-1).


Subject(s)
Amidohydrolases/chemistry , Bacillus/enzymology , Lactones/chemistry , Listeria monocytogenes/enzymology , Sugar Phosphates/chemistry , Amidohydrolases/metabolism , Catalytic Domain , Crystallography, X-Ray , Hydrogen Bonding , Hydrolysis , Kinetics , Lactones/metabolism , Models, Molecular , Structure-Activity Relationship , Substrate Specificity , Sugar Phosphates/metabolism
12.
Biochemistry ; 51(43): 8665-78, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23030629

ABSTRACT

The binding of a ligand to orotidine 5'-monophosphate decarboxylase (OMPDC) is accompanied by a conformational change from an open, inactive conformation (E(o)) to a closed, active conformation (E(c)). As the substrate traverses the reaction coordinate to form the stabilized vinyl carbanion/carbene intermediate, interactions that destabilize the carboxylate group of the substrate and stabilize the intermediate (in the E(c)·S(‡) complex) are enforced. Focusing on the OMPDC from Methanothermobacter thermautotrophicus, we find the "remote" 5'-phosphate group of the substrate activates the enzyme 2.4 × 10(8)-fold; the activation is equivalently described by an intrinsic binding energy (IBE) of 11.4 kcal/mol. We studied residues in the activation that (1) directly contact the 5'-phosphate group, (2) participate in a hydrophobic cluster near the base of the active site loop that sequesters the bound substrate from the solvent, and (3) form hydrogen bonding interactions across the interface between the "mobile" and "fixed" half-barrel domains of the (ß/α)(8)-barrel structure. Our data support a model in which the IBE provided by the 5'-phosphate group is used to allow interactions both near the N-terminus of the active site loop and across the domain interface that stabilize both the E(c)·S and E(c)·S(‡) complexes relative to the E(o)·S complex. The conclusion that the IBE of the 5'-phosphate group provides stabilization to both the E(c)·S and E(c)·S(‡) complexes, not just the E(c)·S(‡) complex, is central to understanding the structural origins of enzymatic catalysis as well as the requirements for the de novo design of enzymes that catalyze novel reactions.


Subject(s)
Methanobacteriaceae/enzymology , Orotidine-5'-Phosphate Decarboxylase/chemistry , Orotidine-5'-Phosphate Decarboxylase/metabolism , Phosphates/metabolism , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Methanobacteriaceae/chemistry , Methanobacteriaceae/genetics , Models, Molecular , Mutation , Orotidine-5'-Phosphate Decarboxylase/genetics , Phosphates/chemistry , Protein Binding , Protein Conformation , Thermodynamics
13.
Biochemistry ; 50(39): 8497-507, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21870810

ABSTRACT

The reaction catalyzed by orotidine 5'-monophosphate decarboxylase (OMPDC) is accompanied by exceptional values for rate enhancement (k(cat)/k(non) = 7.1 × 10(16)) and catalytic proficiency [(k(cat)/K(M))/k(non) = 4.8 × 10(22) M(-1)]. Although a stabilized vinyl carbanion/carbene intermediate is located on the reaction coordinate, the structural strategies by which the reduction in the activation energy barrier is realized remain incompletely understood. This laboratory recently reported that "substrate destabilization" by Asp 70 in the OMPDC from Methanothermobacter thermoautotrophicus (MtOMPDC) lowers the activation energy barrier by ∼5 kcal/mol (contributing ~2.7 × 10(3) to the rate enhancement) [Chan, K. K., Wood, B. M., Fedorov, A. A., Fedorov, E. V., Imker, H. J., Amyes, T. L., Richard, J. P., Almo, S. C., and Gerlt, J. A. (2009) Biochemistry 48, 5518-5531]. We now report that substitutions of hydrophobic residues in a pocket proximal to the carboxylate group of the substrate (Ile 96, Leu 123, and Val 155) with neutral hydrophilic residues decrease the value of k(cat) by as much as 400-fold but have a minimal effect on the value of k(ex) for exchange of H6 of the FUMP product analogue with solvent deuterium; we hypothesize that this pocket destabilizes the substrate by preventing hydration of the substrate carboxylate group. We also report that substitutions of Ser 127 that is proximal to O4 of the orotate ring decrease the value of k(cat)/K(M), with the S127P substitution that eliminates hydrogen bonding interactions with O4 producing a 2.5 × 10(6)-fold reduction; this effect is consistent with delocalization of the negative charge of the carbanionic intermediate on O4 that produces an anionic carbene intermediate and thereby provides a structural strategy for stabilization of the intermediate. These observations provide additional information about the identities of the active site residues that contribute to the rate enhancement and, therefore, insights into the structural strategies for catalysis.


Subject(s)
Orotidine-5'-Phosphate Decarboxylase/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Hydrogen Bonding , Kinetics , Methanobacteriaceae/enzymology , Orotidine-5'-Phosphate Decarboxylase/chemistry , Orotidine-5'-Phosphate Decarboxylase/genetics
14.
Biochemistry ; 50(22): 5077-85, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21545144

ABSTRACT

Cytosine deaminase (CDA) from E. coli is a member of the amidohydrolase superfamily. The structure of the zinc-activated enzyme was determined in the presence of phosphonocytosine, a mimic of the tetrahedral reaction intermediate. This compound inhibits the deamination of cytosine with a K(i) of 52 nM. The zinc- and iron-containing enzymes were characterized to determine the effect of the divalent cations on activation of the hydrolytic water. Fe-CDA loses activity at low pH with a kinetic pK(a) of 6.0, and Zn-CDA has a kinetic pK(a) of 7.3. Mutation of Gln-156 decreased the catalytic activity by more than 5 orders of magnitude, supporting its role in substrate binding. Mutation of Glu-217, Asp-313, and His-246 significantly decreased catalytic activity supporting the role of these three residues in activation of the hydrolytic water molecule and facilitation of proton transfer reactions. A library of potential substrates was used to probe the structural determinants responsible for catalytic activity. CDA was able to catalyze the deamination of isocytosine and the hydrolysis of 3-oxauracil. Large inverse solvent isotope effects were obtained on k(cat) and k(cat)/K(m), consistent with the formation of a low-barrier hydrogen bond during the conversion of cytosine to uracil. A chemical mechanism for substrate deamination by CDA was proposed.


Subject(s)
Cytosine Deaminase/chemistry , Escherichia coli Proteins/chemistry , Catalysis , Cytosine/chemistry , Cytosine/metabolism , Cytosine Deaminase/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Hydrogen Bonding , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Zinc/chemistry
15.
Biochemistry ; 50(25): 5555-7, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21604715

ABSTRACT

Cytosine deaminase (CDA) from Escherichia coli was shown to catalyze the deamination of isoguanine (2-oxoadenine) to xanthine. Isoguanine is an oxidation product of adenine in DNA that is mutagenic to the cell. The isoguanine deaminase activity in E. coli was partially purified by ammonium sulfate fractionation, gel filtration, and anion exchange chromatography. The active protein was identified by peptide mass fingerprint analysis as cytosine deaminase. The kinetic constants for the deamination of isoguanine at pH 7.7 are as follows: k(cat) = 49 s(-1), K(m) = 72 µM, and k(cat)/K(m) = 6.7 × 10(5) M(-1) s(-1). The kinetic constants for the deamination of cytosine are as follows: k(cat) = 45 s(-1), K(m) = 302 µM, and k(cat)/K(m) = 1.5 × 10(5) M(-1) s(-1). Under these reaction conditions, isoguanine is the better substrate for cytosine deaminase. The three-dimensional structure of CDA was determined with isoguanine in the active site.


Subject(s)
Escherichia coli Proteins/chemistry , Guanine Deaminase/chemistry , Catalysis , Cytosine/chemistry , Cytosine/metabolism , Deamination , Escherichia coli Proteins/metabolism , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/metabolism , Guanine Deaminase/metabolism , Kinetics , Substrate Specificity
16.
J Biol Chem ; 285(40): 30606-14, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20659898

ABSTRACT

Atrazine chlorohydrolase, TrzN (triazine hydrolase or atrazine chlorohydrolase 2), initiates bacterial metabolism of the herbicide atrazine by hydrolytic displacement of a chlorine substituent from the s-triazine ring. The present study describes crystal structures and reactivity of wild-type and active site mutant TrzN enzymes. The homodimer native enzyme structure, solved to 1.40 Å resolution, is a (ßα)(8) barrel, characteristic of members of the amidohydrolase superfamily. TrzN uniquely positions threonine 325 in place of a conserved aspartate that ligates the metal in most mononuclear amidohydrolases superfamily members. The threonine side chain oxygen atom is 3.3 Å from the zinc atom and 2.6 Å from the oxygen atom of zinc-coordinated water. Mutation of the threonine to a serine resulted in a 12-fold decrease in k(cat)/K(m), largely due to k(cat), whereas the T325D and T325E mutants had immeasurable activity. The structure and kinetics of TrzN are reminiscent of carbonic anhydrase, which uses a threonine to assist in positioning water for reaction with carbon dioxide. An isosteric substitution in the active site glutamate, E241Q, showed a large diminution in activity with ametryn, no detectable activity with atratone, and a 10-fold decrease with atrazine, when compared with wild-type TrzN. Activity with the E241Q mutant was nearly constant from pH 6.0 to 10.0, consistent with the loss of a proton-donating group. Structures for TrzN-E241Q were solved with bound ametryn and atratone to 1.93 and 1.64 Å resolution, respectively. Both structure and kinetic determinations suggest that the Glu(241) side chain provides a proton to N-1 of the s-triazine substrate to facilitate nucleophilic displacement at the adjacent C-2.


Subject(s)
Bacterial Proteins/chemistry , Hydrolases/chemistry , Protein Multimerization , Triazines/chemistry , Zinc/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Crystallography, X-Ray , Hydrogen-Ion Concentration , Hydrolases/genetics , Kinetics , Mutation, Missense , Protein Structure, Quaternary , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity
17.
J Exp Med ; 218(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34709351

ABSTRACT

HVEM is a TNF (tumor necrosis factor) receptor contributing to a broad range of immune functions involving diverse cell types. It interacts with a TNF ligand, LIGHT, and immunoglobulin (Ig) superfamily members BTLA and CD160. Assessing the functional impact of HVEM binding to specific ligands in different settings has been complicated by the multiple interactions of HVEM and HVEM binding partners. To dissect the molecular basis for multiple functions, we determined crystal structures that reveal the distinct HVEM surfaces that engage LIGHT or BTLA/CD160, including the human HVEM-LIGHT-CD160 ternary complex, with HVEM interacting simultaneously with both binding partners. Based on these structures, we generated mouse HVEM mutants that selectively recognized either the TNF or Ig ligands in vitro. Knockin mice expressing these muteins maintain expression of all the proteins in the HVEM network, yet they demonstrate selective functions for LIGHT in the clearance of bacteria in the intestine and for the Ig ligands in the amelioration of liver inflammation.


Subject(s)
Antigens, CD/metabolism , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Crystallography, X-Ray , Drosophila/cytology , Drosophila/genetics , Female , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutation , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Yersinia Infections/genetics , Yersinia Infections/pathology
18.
Biochemistry ; 49(17): 3514-6, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20369850

ABSTRACT

The structural factors responsible for the extraordinary rate enhancement ( approximately 10(17)) of the reaction catalyzed by orotidine 5'-monophosphate decarboxylase (OMPDC) have not been defined. Catalysis requires a conformational change that closes an active site loop and "clamps" the orotate base proximal to hydrogen-bonded networks that destabilize the substrate and stabilize the intermediate. In the OMPDC from Methanobacter thermoautotrophicus, a "remote" structurally conserved cluster of hydrophobic residues that includes Val 182 in the active site loop is assembled in the closed, catalytically active conformation. Substitution of these residues with Ala decreases k(cat)/K(m) with a minimal effect on k(cat), providing evidence that the cluster stabilizes the closed conformation. The intrinsic binding energies of the 5'-phosphate group of orotidine 5'-monophosphate for the mutant enzymes are similar to that for the wild type, supporting this conclusion.


Subject(s)
Methanobacteriaceae/enzymology , Orotidine-5'-Phosphate Decarboxylase/chemistry , Orotidine-5'-Phosphate Decarboxylase/metabolism , Amino Acid Substitution , Catalysis , Hydrogen Bonding , Models, Molecular , Mutagenesis, Site-Directed , Mutation/genetics , Orotidine-5'-Phosphate Decarboxylase/genetics , Protein Conformation , Structure-Activity Relationship
19.
Biochemistry ; 49(3): 611-22, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20000809

ABSTRACT

Human renal dipeptidase, an enzyme associated with glutathione metabolism and the hydrolysis of beta-lactams, is similar in sequence to a cluster of approximately 400 microbial proteins currently annotated as nonspecific dipeptidases within the amidohydrolase superfamily. The closest homologue to the human renal dipeptidase from a fully sequenced microbe is Sco3058 from Streptomyces coelicolor. Dipeptide substrates of Sco3058 were identified by screening a comprehensive series of l-Xaa-l-Xaa, l-Xaa-d-Xaa, and d-Xaa-l-Xaa dipeptide libraries. The substrate specificity profile shows that Sco3058 hydrolyzes a broad range of dipeptides with a marked preference for an l-amino acid at the N-terminus and a d-amino acid at the C-terminus. The best substrate identified was l-Arg-d-Asp (k(cat)/K(m) = 7.6 x 10(5) M(-1) s(-1)). The three-dimensional structure of Sco3058 was determined in the absence and presence of the inhibitors citrate and a phosphinate mimic of l-Ala-d-Asp. The enzyme folds as a (beta/alpha)(8) barrel, and two zinc ions are bound in the active site. Site-directed mutagenesis was used to probe the importance of specific residues that have direct interactions with the substrate analogues in the active site (Asp-22, His-150, Arg-223, and Asp-320). The solvent viscosity and kinetic effects of D(2)O indicate that substrate binding is relatively sticky and that proton transfers do not occurr during the rate-limiting step. A bell-shaped pH-rate profile for k(cat) and k(cat)/K(m) indicated that one group needs to be deprotonated and a second group must be protonated for optimal turnover. Computational docking of high-energy intermediate forms of l/d-Ala-l/d-Ala to the three-dimensional structure of Sco3058 identified the structural determinants for the stereochemical preferences for substrate binding and turnover.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Dipeptidases/chemistry , Dipeptidases/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dipeptides/chemistry , Dipeptides/metabolism , Humans , Streptomyces coelicolor/enzymology , Streptomyces coelicolor/metabolism , Structure-Activity Relationship , Substrate Specificity
20.
Biochemistry ; 49(31): 6791-803, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20604542

ABSTRACT

Two uncharacterized enzymes from the amidohydrolase superfamily belonging to cog1228 were cloned, expressed, and purified to homogeneity. The two proteins, Sgx9260c ( gi|44242006 ) and Sgx9260b ( gi|44479596 ), were derived from environmental DNA samples originating from the Sargasso Sea. The catalytic function and substrate profiles for Sgx9260c and Sgx9260b were determined using a comprehensive library of dipeptides and N-acyl derivative of l-amino acids. Sgx9260c catalyzes the hydrolysis of Gly-l-Pro, l-Ala-l-Pro, and N-acyl derivatives of l-Pro. The best substrate identified to date is N-acetyl-l-Pro with a value of k(cat)/K(m) of 3 x 10(5) M(-1) s(-1). Sgx9260b catalyzes the hydrolysis of l-hydrophobic l-Pro dipeptides and N-acyl derivatives of l-Pro. The best substrate identified to date is N-propionyl-l-Pro with a value of k(cat)/K(m) of 1 x 10(5) M(-1) s(-1). Three-dimensional structures of both proteins were determined by X-ray diffraction methods (PDB codes 3MKV and 3FEQ ). These proteins fold as distorted (beta/alpha)(8)-barrels with two divalent cations in the active site. The structure of Sgx9260c was also determined as a complex with the N-methylphosphonate derivative of l-Pro (PDB code 3N2C ). In this structure the phosphonate moiety bridges the binuclear metal center, and one oxygen atom interacts with His-140. The alpha-carboxylate of the inhibitor interacts with Tyr-231. The proline side chain occupies a small substrate binding cavity formed by residues contributed from the loop that follows beta-strand 7 within the (beta/alpha)(8)-barrel. A total of 38 other proteins from cog1228 are predicted to have the same substrate profile based on conservation of the substrate binding residues. The structure of an evolutionarily related protein, Cc2672 from Caulobacter crecentus, was determined as a complex with the N-methylphosphonate derivative of l-arginine (PDB code 3MTW ).


Subject(s)
Amidohydrolases/chemistry , Dipeptidases/chemistry , Catalytic Domain , Kinetics , Oceans and Seas , Protein Conformation , Substrate Specificity , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL