Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Biol Chem ; 295(12): 3759-3770, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32029475

RESUMO

Menaquinone (vitamin K2) plays a vital role in energy generation and environmental adaptation in many bacteria, including the human pathogen Mycobacterium tuberculosis (Mtb). Although menaquinone levels are known to be tightly linked to the cellular redox/energy status of the cell, the regulatory mechanisms underpinning this phenomenon are unclear. The first committed step in menaquinone biosynthesis is catalyzed by MenD, a thiamine diphosphate-dependent enzyme comprising three domains. Domains I and III form the MenD active site, but no function has yet been ascribed to domain II. Here, we show that the last cytosolic metabolite in the menaquinone biosynthesis pathway, 1,4-dihydroxy-2-naphthoic acid (DHNA), binds to domain II of Mtb-MenD and inhibits its activity. Using X-ray crystallography of four apo- and cofactor-bound Mtb-MenD structures, along with several spectroscopy assays, we identified three arginine residues (Arg-97, Arg-277, and Arg-303) that are important for both enzyme activity and the feedback inhibition by DHNA. Among these residues, Arg-277 appeared to be particularly important for signal propagation from the allosteric site to the active site. This is the first evidence of feedback regulation of the menaquinone biosynthesis pathway in bacteria, identifying a protein-level regulatory mechanism that controls menaquinone levels within the cell and may therefore represent a good target for disrupting menaquinone biosynthesis in M. tuberculosis.


Assuntos
Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/metabolismo , Vitamina K 2/metabolismo , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico , Sequência de Aminoácidos , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Sítios de Ligação , Biocatálise , Domínio Catalítico , Cristalografia por Raios X , Humanos , Mutagênese Sítio-Dirigida , Mycobacterium tuberculosis/enzimologia , Naftóis/química , Naftóis/metabolismo , Naftóis/farmacologia , Conformação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Alinhamento de Sequência
2.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 4): 862-72, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25849397

RESUMO

Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis. Access to iron in host macrophages depends on iron-chelating siderophores called mycobactins and is strongly correlated with Mtb virulence. Here, the crystal structure of an Mtb enzyme involved in mycobactin biosynthesis, MbtN, in complex with its FAD cofactor is presented at 2.30 Šresolution. The polypeptide fold of MbtN conforms to that of the acyl-CoA dehydrogenase (ACAD) family, consistent with its predicted role of introducing a double bond into the acyl chain of mycobactin. Structural comparisons and the presence of an acyl carrier protein, MbtL, in the same gene locus suggest that MbtN acts on an acyl-(acyl carrier protein) rather than an acyl-CoA. A notable feature of the crystal structure is the tubular density projecting from N(5) of FAD. This was interpreted as a covalently bound polyethylene glycol (PEG) fragment and resides in a hydrophobic pocket where the substrate acyl group is likely to bind. The pocket could accommodate an acyl chain of 14-21 C atoms, consistent with the expected length of the mycobactin acyl chain. Supporting this, steady-state kinetics show that MbtN has ACAD activity, preferring acyl chains of at least 16 C atoms. The acyl-binding pocket adopts a different orientation (relative to the FAD) to other structurally characterized ACADs. This difference may be correlated with the apparent ability of MbtN to catalyse the formation of an unusual cis double bond in the mycobactin acyl chain.


Assuntos
Enoil-(Proteína de Transporte de Acila) Redutase (NADPH, B-Específica)/química , Mycobacterium tuberculosis/enzimologia , Tuberculose/microbiologia , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Flavina-Adenina Dinucleotídeo/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Oxazóis/metabolismo , Conformação Proteica , Alinhamento de Sequência
3.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2297-308, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26527146

RESUMO

The tryptophan-biosynthesis pathway is essential for Mycobacterium tuberculosis (Mtb) to cause disease, but not all of the enzymes that catalyse this pathway in this organism have been identified. The structure and function of the enzyme complex that catalyses the first committed step in the pathway, the anthranilate synthase (AS) complex, have been analysed. It is shown that the open reading frames Rv1609 (trpE) and Rv0013 (trpG) encode the chorismate-utilizing (AS-I) and glutamine amidotransferase (AS-II) subunits of the AS complex, respectively. Biochemical assays show that when these subunits are co-expressed a bifunctional AS complex is obtained. Crystallization trials on Mtb-AS unexpectedly gave crystals containing only AS-I, presumably owing to its selective crystallization from solutions containing a mixture of the AS complex and free AS-I. The three-dimensional structure reveals that Mtb-AS-I dimerizes via an interface that has not previously been seen in AS complexes. As is the case in other bacteria, it is demonstrated that Mtb-AS shows cooperative allosteric inhibition by tryptophan, which can be rationalized based on interactions at this interface. Comparative inhibition studies on Mtb-AS-I and related enzymes highlight the potential for single inhibitory compounds to target multiple chorismate-utilizing enzymes for TB drug discovery.


Assuntos
Antranilato Sintase/antagonistas & inibidores , Antranilato Sintase/química , Mycobacterium tuberculosis/enzimologia , Triptofano/metabolismo , Tuberculose/microbiologia , Antranilato Sintase/metabolismo , Vias Biossintéticas , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Modelos Moleculares , Mycobacterium tuberculosis/metabolismo , Conformação Proteica , Multimerização Proteica , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
4.
Biochem J ; 461(1): 87-98, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24712732

RESUMO

AnPRT (anthranilate phosphoribosyltransferase), required for the biosynthesis of tryptophan, is essential for the virulence of Mycobacterium tuberculosis (Mtb). AnPRT catalyses the Mg2+-dependent transfer of a phosphoribosyl group from PRPP (5'-phosphoribosyl-1'-pyrophosphate) to anthranilate to form PRA (5'-phosphoribosyl anthranilate). Mtb-AnPRT was shown to catalyse a sequential reaction and significant substrate inhibition by anthranilate was observed. Antimycobacterial fluoroanthranilates and methyl-substituted analogues were shown to act as alternative substrates for Mtb-AnPRT, producing the corresponding substituted PRA products. Structures of the enzyme complexed with anthranilate analogues reveal two distinct binding sites for anthranilate. One site is located over 8 Å (1 Å=0.1 nm) from PRPP at the entrance to a tunnel leading to the active site, whereas in the second, inner, site anthranilate is adjacent to PRPP, in a catalytically relevant position. Soaking the analogues for variable periods of time provides evidence for anthranilate located at transient positions during transfer from the outer site to the inner catalytic site. PRPP and Mg2+ binding have been shown to be associated with the rearrangement of two flexible loops, which is required to complete the inner anthranilate-binding site. It is proposed that anthranilate first binds to the outer site, providing an unusual mechanism for substrate capture and efficient transfer to the catalytic site following the binding of PRPP.


Assuntos
Antranilato Fosforribosiltransferase/química , Antranilato Fosforribosiltransferase/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/enzimologia , Antranilato Fosforribosiltransferase/farmacologia , Proteínas de Bactérias/farmacologia , Catálise , Cristalização , Modelos Moleculares , Mycobacterium tuberculosis/patogenicidade , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/fisiologia , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Fatores de Virulência/farmacologia
5.
J Biol Chem ; 288(3): 1643-52, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23179721

RESUMO

D-Xylulokinase (XK; EC 2.7.1.17) catalyzes the ATP-dependent phosphorylation of d-xylulose (Xu) to produce xylulose 5-phosphate (Xu5P). In mammals, XK is the last enzyme in the glucuronate-xylulose pathway, active in the liver and kidneys, and is linked through its product Xu5P to the pentose-phosphate pathway. XK may play an important role in metabolic disease, given that Xu5P is a key regulator of glucose metabolism and lipogenesis. We have expressed the product of a putative human XK gene and identified it as the authentic human d-xylulokinase (hXK). NMR studies with a variety of sugars showed that hXK acts only on d-xylulose, and a coupled photometric assay established its key kinetic parameters as K(m)(Xu) = 24 ± 3 µm and k(cat) = 35 ± 5 s(-1). Crystal structures were determined for hXK, on its own and in complexes with Xu, ADP, and a fluorinated inhibitor. These reveal that hXK has a two-domain fold characteristic of the sugar kinase/hsp70/actin superfamily, with glycerol kinase as its closest relative. Xu binds to domain-I and ADP to domain-II, but in this open form of hXK they are 10 Å apart, implying that a large scale conformational change is required for catalysis. Xu binds in its linear keto-form, sandwiched between a Trp side chain and polar side chains that provide exquisite hydrogen bonding recognition. The hXK structure provides a basis for the design of specific inhibitors with which to probe its roles in sugar metabolism and metabolic disease.


Assuntos
Difosfato de Adenosina/química , Fosfotransferases (Aceptor do Grupo Álcool)/química , Xilulose/análogos & derivados , Difosfato de Adenosina/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Escherichia coli/genética , Expressão Gênica , Humanos , Ligação de Hidrogênio , Cinética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Pentosefosfatos/química , Pentosefosfatos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Xilulose/metabolismo
6.
Chembiochem ; 15(6): 852-64, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24623674

RESUMO

The emergence of extensively drug-resistant strains of Mycobacterium tuberculosis (Mtb) highlights the need for new therapeutics to treat tuberculosis. We are attempting to fast-track a targeted approach to drug design by generating analogues of a validated hit from molecular library screening that shares its chemical scaffold with a current therapeutic, the anti-arthritic drug Lobenzarit (LBZ). Our target, anthranilate phosphoribosyltransferase (AnPRT), is an enzyme from the tryptophan biosynthetic pathway in Mtb. A bifurcated hydrogen bond was found to be a key feature of the LBZ-like chemical scaffold and critical for enzyme inhibition. We have determined crystal structures of compounds in complex with the enzyme that indicate that the bifurcated hydrogen bond assists in orientating compounds in the correct conformation to interact with key residues in the substrate-binding tunnel of Mtb-AnPRT. Characterising the inhibitory potency of the hit and its analogues in different ways proved useful, due to the multiple substrates and substrate binding sites of this enzyme. Binding in a site other than the catalytic site was found to be associated with partial inhibition. An analogue, 2-(2-5-methylcarboxyphenylamino)-3-methylbenzoic acid, that bound at the catalytic site and caused complete, rather than partial, inhibition of enzyme activity was found. Therefore, we designed and synthesised an extended version of the scaffold on the basis of this observation. The resultant compound, 2,6-bis-(2-carboxyphenylamino)benzoate, is a 40-fold more potent inhibitor of the enzyme than the original hit and provides direction for further structure-based drug design.


Assuntos
Antituberculosos/química , Mycobacterium tuberculosis/enzimologia , Triptofano/biossíntese , ortoaminobenzoatos/química , Antranilato Fosforribosiltransferase/antagonistas & inibidores , Antranilato Fosforribosiltransferase/metabolismo , Antituberculosos/metabolismo , Antituberculosos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Ligação de Hidrogênio , Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Relação Estrutura-Atividade , ortoaminobenzoatos/metabolismo , ortoaminobenzoatos/farmacologia
7.
Anal Biochem ; 465: 53-62, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25016192

RESUMO

The production and analysis of individual structural domains is a common strategy for studying large or complex proteins, which may be experimentally intractable in their full-length form. However, identifying domain boundaries is challenging if there is little structural information concerning the protein target. One experimental procedure for mapping domains is to screen a library of random protein fragments for solubility, since truncation of a domain will typically expose hydrophobic groups, leading to poor fragment solubility. We have coupled fragment solubility screening with global data analysis to develop an effective method for identifying structural domains within a protein. A gene fragment library is generated using mechanical shearing, or by uracil doping of the gene and a uracil-specific enzymatic digest. A split green fluorescent protein (GFP) assay is used to screen the corresponding protein fragments for solubility when expressed in Escherichia coli. The soluble fragment data are then analyzed using two complementary approaches. Fragmentation "hotspots" indicate possible interdomain regions. Clustering algorithms are used to group related fragments, and concomitantly predict domain location. The effectiveness of this Domain Seeking procedure is demonstrated by application to the well-characterized human protein p85α.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/química , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Uracila/química , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Solubilidade
8.
Nat Commun ; 15(1): 1310, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38346985

RESUMO

Poly-γ-glutamate tails are a distinctive feature of archaeal, bacterial, and eukaryotic cofactors, including the folates and F420. Despite decades of research, key mechanistic questions remain as to how enzymes successively add glutamates to poly-γ-glutamate chains while maintaining cofactor specificity. Here, we show how poly-γ-glutamylation of folate and F420 by folylpolyglutamate synthases and γ-glutamyl ligases, non-homologous enzymes, occurs via processive addition of L-glutamate onto growing γ-glutamyl chain termini. We further reveal structural snapshots of the archaeal γ-glutamyl ligase (CofE) in action, crucially including a bulged-chain product that shows how the cofactor is retained while successive glutamates are added to the chain terminus. This bulging substrate model of processive poly-γ-glutamylation by terminal extension is arguably ubiquitous in such biopolymerisation reactions, including addition to folates, and demonstrates convergent evolution in diverse species from archaea to humans.


Assuntos
Ácido Fólico , Ácido Glutâmico , Humanos , Peptídeo Sintases/metabolismo , Bactérias/metabolismo , Processamento de Proteína Pós-Traducional
9.
Biochemistry ; 52(10): 1776-87, 2013 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-23363292

RESUMO

Anthranilate phosphoribosyltransferase (AnPRT, EC 2.4.2.18) is a homodimeric enzyme that catalyzes the reaction between 5'-phosphoribosyl 1'-pyrophosphate (PRPP) and anthranilate, as part of the tryptophan biosynthesis pathway. Here we present the results of the first chemical screen for inhibitors against Mycobacterium tuberculosis AnPRT (Mtb-AnPRT), along with crystal structures of Mtb-AnPRT in complex with PRPP and several inhibitors. Previous work revealed that PRPP is bound at the base of a deep cleft in Mtb-AnPRT and predicted two anthranilate binding sites along the tunnel leading to the PRPP binding site. Unexpectedly, the inhibitors presented here almost exclusively bound at the entrance of the tunnel, in the presumed noncatalytic anthranilate binding site, previously hypothesized to have a role in substrate capture. The potencies of the inhibitors were measured, yielding Ki values of 1.5-119 µM, with the strongest inhibition displayed by a bianthranilate compound that makes hydrogen bond and salt bridge contacts with Mtb-AnPRT via its carboxyl groups. Our results reveal how the substrate capture mechanism of AnPRT can be exploited to inhibit the enzyme's activity and provide a scaffold for the design of improved Mtb-AnPRT inhibitors that may ultimately form the basis of new antituberculosis drugs with a novel mode of action.


Assuntos
Antranilato Fosforribosiltransferase/antagonistas & inibidores , Antranilato Fosforribosiltransferase/química , Mycobacterium tuberculosis/enzimologia , Antranilato Fosforribosiltransferase/genética , Antituberculosos/farmacologia , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Cinética , Modelos Moleculares , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Fosforribosil Pirofosfato/metabolismo , Especificidade por Substrato , ortoaminobenzoatos/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-24316828

RESUMO

In Mycobacterium tuberculosis, the protein MbtN (Rv1346) catalyzes the formation of a double bond in the fatty-acyl moiety of the siderophore mycobactin, which is used by this organism to acquire essential iron. MbtN is homologous to acyl-CoA dehydrogenases, whose general role is to catalyze the α,ß-dehydrogenation of fatty-acyl-CoA conjugates. Mycobactins, however, contain a long unsaturated fatty-acid chain with an unusual cis double bond conjugated to the carbonyl group of the mycobactin core. To characterize the role of MbtN in the dehydrogenation of this fatty-acyl moiety, the enzyme has been expressed, purified and crystallized. The crystals diffracted to 2.3 Å resolution at a synchrotron source and were found to belong to the hexagonal space group H32, with unit-cell parameters a = b = 139.10, c = 253.09 Å, α = ß = 90, γ = 120°.


Assuntos
Acil-CoA Desidrogenases/química , Proteínas de Bactérias/química , Mycobacterium tuberculosis/química , Acil-CoA Desidrogenases/genética , Acil-CoA Desidrogenases/isolamento & purificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Cristalização , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Mycobacterium tuberculosis/genética , Oxazóis/química , Oxazóis/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Sideróforos/química , Sideróforos/metabolismo
11.
ACS Infect Dis ; 9(8): 1658-1673, 2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37488090

RESUMO

Millions of people are infected by the dengue and Zika viruses each year, resulting in significant morbidity and mortality. Galidesivir is an adenosine nucleoside analog that can attenuate flavivirus replication in cell-based assays and animal models of infection. Galidesivir is converted to the triphosphorylated form by host kinases and subsequently incorporated into viral RNA by viral RNA polymerases. This has been proposed to lead to the delayed termination of RNA synthesis. Here, we report direct in vitro testing of the effects of Galidesivir triphosphate on dengue-2 and Zika virus polymerase activity. Galidesivir triphosphate was chemically synthesized, and inhibition of RNA synthesis followed using a dinucleotide-primed assay with a homopolymeric poly(U) template. Galidesivir triphosphate was equipotent against dengue-2 and Zika polymerases, with IC50 values of 42 ± 12 µM and 47 ± 5 µM, respectively, at an ATP concentration of 20 µM. RNA primer extension assays show that the dengue-2 polymerase stalls while attempting to add a Galidesivir nucleotide to the nascent RNA chain, evidenced by the accumulation of RNA products truncated immediately upstream of Galidesivir incorporation sites. Nevertheless, Galidesivir is incorporated at isolated sites with low efficiency, leading to the subsequent synthesis of full-length RNA with no evidence of delayed chain termination. The incorporation of Galidesivir at consecutive sites is strongly disfavored, highlighting the potential for modulation of inhibitory effects of nucleoside analogs by the template sequence. Our results suggest that attenuation of dengue replication by Galidesivir may not derive from the early termination of RNA synthesis following Galidesivir incorporation.


Assuntos
Dengue , Infecção por Zika virus , Zika virus , Animais , Antivirais/farmacologia , Adenosina/farmacologia , RNA Viral/genética , Nucleotidiltransferases , Zika virus/genética
12.
Philos Trans R Soc Lond B Biol Sci ; 378(1871): 20220035, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36633276

RESUMO

Menaquinones (MKs) are electron carriers in bacterial respiratory chains. In Staphylococcus aureus (Sau), MKs are essential for aerobic and anaerobic respiration. As MKs are redox-active, their biosynthesis likely requires tight regulation to prevent disruption of cellular redox balance. We recently found that the Mycobacterium tuberculosis MenD, the first committed enzyme of the MK biosynthesis pathway, is allosterically inhibited by the downstream metabolite 1,4-dihydroxy-2-naphthoic acid (DHNA). To understand if this is a conserved mechanism in phylogenetically distant genera that also use MK, we investigated whether the Sau-MenD is allosterically inhibited by DHNA. Our results show that DHNA binds to and inhibits the SEPHCHC synthase activity of Sau-MenD enzymes. We identified residues in the DHNA binding pocket that are important for catalysis (Arg98, Lys283, Lys309) and inhibition (Arg98, Lys283). Furthermore, we showed that exogenous DHNA inhibits the growth of Sau, an effect that can be rescued by supplementing the growth medium with MK-4. Our results demonstrate that, despite a lack of strict conservation of the DHNA binding pocket between Mtb-MenD and Sau-MenD, feedback inhibition by DHNA is a conserved mechanism in Sau-MenD and hence the Sau MK biosynthesis pathway. These findings may have implications for the development of anti-staphylococcal agents targeting MK biosynthesis. This article is part of the theme issue 'Reactivity and mechanism in chemical and synthetic biology'.


Assuntos
Naftalenos , Staphylococcus aureus , Vitamina K 2/farmacologia , Vitamina K 2/metabolismo , Staphylococcus aureus/metabolismo , Retroalimentação , Naftalenos/farmacologia
13.
Biochemistry ; 51(11): 2289-97, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22352945

RESUMO

α-Isopropylmalate synthase (α-IPMS) catalyzes the metal-dependent aldol reaction between α-ketoisovalerate (α-KIV) and acetyl-coenzyme A (AcCoA) to give α-isopropylmalate (α-IPM). This reaction is the first committed step in the biosynthesis of leucine in bacteria. α-IPMS is homodimeric, with monomers consisting of (ß/α)(8) barrel catalytic domains fused to a C-terminal regulatory domain, responsible for binding leucine and providing feedback regulation for leucine biosynthesis. In these studies, we demonstrate that removal of the regulatory domain from the α-IPMS enzymes of both Neisseria meningitidis (NmeIPMS) and Mycobacterium tuberculosis (MtuIPMS) results in enzymes that are unable to catalyze the formation of α-IPM, although truncated NmeIPMS was still able to slowly hydrolyze AcCoA. The lack of catalytic activity of these truncation variants was confirmed by complementation studies with Escherichia coli cells lacking the α-IPMS gene, where transformation with the plasmids encoding the truncated α-IPMS enzymes was not able to rescue α-IPMS activity. X-ray crystal structures of both truncation variants reveal that both proteins are dimeric and that the catalytic sites of the proteins are intact, although the divalent metal ion that is thought to be responsible for activating substrate α-KIV is displaced slightly relative to its position in the substrate-bound, wild-type structure. Isothermal titration calorimetry and WaterLOGSY nuclear magnetic resonance experiments demonstrate that although these truncation variants are not able to catalyze the reaction between α-KIV and AcCoA, they are still able to bind the substrate α-KIV. It is proposed that the regulatory domain is crucial for ensuring protein dynamics necessary for competent catalysis.


Assuntos
2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/metabolismo , Acetilcoenzima A/química , Acetilcoenzima A/metabolismo , Sítios de Ligação , Catálise , Domínio Catalítico , Cristalografia por Raios X , Hemiterpenos , Cetoácidos/química , Cetoácidos/metabolismo , Cinética , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/metabolismo , Neisseria meningitidis/enzimologia , Neisseria meningitidis/metabolismo , Especificidade por Substrato
14.
Biochemistry ; 51(24): 4868-79, 2012 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-22607697

RESUMO

MbtI is the salicylate synthase that catalyzes the first committed step in the synthesis of the iron chelating compound mycobactin in Mycobacterium tuberculosis. We previously developed a series of aromatic inhibitors against MbtI based on the reaction intermediate for this enzyme, isochorismate. The most potent of these inhibitors had hydrophobic substituents, ranging in size from a methyl to a phenyl group, appended to the terminal alkene of the enolpyruvyl group. These compounds exhibited low micromolar inhibition constants against MbtI and were at least an order of magnitude more potent than the parental compound for the series, which carries a native enolpyruvyl group. In this study, we sought to understand how the substituted enolpyruvyl group confers greater potency, by determining cocrystal structures of MbtI with six inhibitors from the series. A switch in binding mode at the MbtI active site is observed for inhibitors carrying a substituted enolpyruvyl group, relative to the parental compound. Computational studies suggest that the change in binding mode, and higher potency, is due to the effect of the substituents on the conformational landscape of the core inhibitor structure. The crystal structures and fluorescence-based thermal shift assays indicate that substituents larger than a methyl group are accommodated in the MbtI active site through significant but localized flexibility in the peptide backbone. These findings have implications for the design of improved inhibitors of MbtI, as well as other chorismate-utilizing enzymes from this family.


Assuntos
Domínio Catalítico , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Liases/antagonistas & inibidores , Liases/metabolismo , Mycobacterium tuberculosis/enzimologia , Ácido Corísmico/metabolismo , Cristalografia por Raios X , Inibidores Enzimáticos/química , Liases/química , Modelos Moleculares , Ligação Proteica , Piruvatos/química , Piruvatos/metabolismo , Piruvatos/farmacologia , Soluções , Temperatura
15.
J Biol Chem ; 286(47): 40706-16, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21956117

RESUMO

Enzymes of the de novo purine biosynthetic pathway have been identified as essential for the growth and survival of Mycobacterium tuberculosis and thus have potential for the development of anti-tuberculosis drugs. The final two steps of this pathway are carried out by the bifunctional enzyme 5-aminoimidazole-4-carboxamide ribonucleotide transformylase/inosine monophosphate cyclohydrolase (ATIC), also known as PurH. This enzyme has already been the target of anti-cancer drug development. We have determined the crystal structures of the M. tuberculosis ATIC (Rv0957) both with and without the substrate 5-aminoimidazole-4-carboxamide ribonucleotide, at resolutions of 2.5 and 2.2 Å, respectively. As for other ATIC enzymes, the protein is folded into two domains, the N-terminal domain (residues 1-212) containing the cyclohydrolase active site and the C-terminal domain (residues 222-523) containing the formyltransferase active site. An adventitiously bound nucleotide was found in the cyclohydrolase active site in both structures and was identified by NMR and mass spectral analysis as a novel 5-formyl derivative of an earlier intermediate in the biosynthetic pathway 4-carboxy-5-aminoimidazole ribonucleotide. This result and other studies suggest that this novel nucleotide is a cyclohydrolase inhibitor. The dimer formed by M. tuberculosis ATIC is different from those seen for human and avian ATICs, but it has a similar ∼50-Å separation of the two active sites of the bifunctional enzyme. Evidence in M. tuberculosis ATIC for reactivity of half-the-sites in the cyclohydrolase domains can be attributed to ligand-induced movements that propagate across the dimer interface and may be a common feature of ATIC enzymes.


Assuntos
Hidroximetil e Formil Transferases/química , Hidroximetil e Formil Transferases/metabolismo , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Mycobacterium tuberculosis/enzimologia , Nucleotídeo Desaminases/química , Nucleotídeo Desaminases/metabolismo , Nucleotídeos de Purina/biossíntese , Nucleotídeos de Purina/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Animais , Domínio Catalítico , Cristalografia por Raios X , Humanos , Ligantes , Modelos Moleculares , Mycobacterium tuberculosis/metabolismo , Multimerização Proteica , Ribonucleotídeos/metabolismo
16.
Org Biomol Chem ; 10(46): 9223-36, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23108268

RESUMO

Mycobacterium tuberculosis salicylate synthase (MbtI) catalyses the first committed step in the biosynthesis of mycobactin T, an iron-chelating siderophore essential for the virulence and survival of M. tuberculosis. Co-crystal structures of MbtI with members of a first generation inhibitor library revealed large inhibitor-induced rearrangements within the active site of the enzyme. This plasticity of the MbtI active site was probed via the preparation of a library of inhibitors based on a 2,3-dihydroxybenzoate scaffold with a range of substituted phenylacrylate side chains appended to the C3 position. Most compounds exhibited moderate inhibitory activity against the enzyme, with inhibition constants in the micromolar range, while several dimethyl ester variants possessed promising anti-tubercular activity in vitro.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Hidroxibenzoatos/química , Liases/antagonistas & inibidores , Mycobacterium tuberculosis/enzimologia , Bibliotecas de Moléculas Pequenas/síntese química , Acrilatos/química , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Ésteres , Cinética , Liases/metabolismo , Espectroscopia de Ressonância Magnética , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Modelos Moleculares , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/efeitos dos fármacos , Ligação Proteica , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade
17.
Viruses ; 13(9)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34578318

RESUMO

The paramyxoviral phosphoprotein (P protein) is the non-catalytic subunit of the viral RNA polymerase, and coordinates many of the molecular interactions required for RNA synthesis. All paramyxoviral P proteins oligomerize via a centrally located coiled-coil that is connected to a downstream binding domain by a dynamic linker. The C-terminal region of the P protein coordinates interactions between the catalytic subunit of the polymerase, and the viral nucleocapsid housing the genomic RNA. The inherent flexibility of the linker is believed to facilitate polymerase translocation. Here we report biophysical and structural characterization of the C-terminal region of the P protein from Menangle virus (MenV), a bat-borne paramyxovirus with zoonotic potential. The MenV P protein is tetrameric but can dissociate into dimers at sub-micromolar protein concentrations. The linker is globally disordered and can be modeled effectively as a worm-like chain. However, NMR analysis suggests very weak local preferences for alpha-helical and extended beta conformation exist within the linker. At the interface between the disordered linker and the structured C-terminal binding domain, a gradual disorder-to-order transition occurs, with X-ray crystallographic analysis revealing a dynamic interfacial structure that wraps the surface of the binding domain.


Assuntos
Paramyxoviridae/metabolismo , Fosfoproteínas/química , Proteínas Virais/química , Domínio Catalítico , Cristalografia por Raios X , RNA Polimerases Dirigidas por DNA , Modelos Moleculares , Paramyxoviridae/genética , Fosfoproteínas/genética , Ligação Proteica , Domínios Proteicos , RNA Viral , Proteínas Virais/genética
19.
Anal Biochem ; 396(1): 13-22, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19699711

RESUMO

Methylation and acetylation of lysines are crucial posttranslational modifications that regulate gene transcription and have been shown to be misregulated in many forms of cancers. Western blot, immunoprecipitation, and immunofluorescence are commonly used to characterize histone acetylation and methylation. However, these approaches are limited by the availability, site specificity, and cross-reactivity of antibodies. Mass spectrometry is emerging as an additional powerful tool for histone characterization. The isobaric nature of trimethylation and acetylation (42.0470 and 42.0106 Da, respectively) confounds histone characterization by means other than high-resolution/high-mass accuracy mass spectrometry. In this study, we adapted methodology that exploits difference in the relative retention time of acetylated and methylated peptides to unequivocally distinguish between these two modifications even with low-mass accuracy mass spectrometers. The approach was tested on tryptic digest of Saccharomyces cerevisiae histones. We found that acetylation resulted in increased retention in reversed-phase chromatography, whereas methylation, including trimethylation, showed little change in retention. For example, the acetylated forms of peptide (27)KSAPSTGGVKKPHR(40) eluted at 15.63 min, whereas the methylated forms eluted at 13.89 min. In addition, the effect of acetylation was cumulative as observed in the case of peptide (9)KSTGGKAPR(17), whose unmodified, monoacetylated, and diacetylated isoforms eluted at 7.43, 10.47, and 16.49 min, respectively. The modification patterns of the peptides in question were subsequently verified by high-mass accuracy tandem mass spectrometry.


Assuntos
Cromatografia de Fase Reversa/métodos , Histonas/metabolismo , Espectrometria de Massas/métodos , Processamento de Proteína Pós-Traducional , Acetilação , Sequência de Aminoácidos , Cromatografia Líquida , Histonas/química , Metilação , Dados de Sequência Molecular , Peso Molecular , Peptídeos/análise , Peptídeos/química , Saccharomyces cerevisiae/metabolismo , Fatores de Tempo
20.
Org Biomol Chem ; 8(15): 3534-42, 2010 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-20532401

RESUMO

Several 2-amino-4-carboxypyridine, 4- and 5-carboxypyridone-based compounds were prepared and tested against three members of the chorismate-utilising enzyme family, anthranilate synthase, isochorismate synthase and salicylate synthase. Most compounds exhibited low micromolar inhibition of these three enzymes. The most potent inhibitor was a 4-carboxypyridone analogue bearing a lactate side chain on the pyridyl nitrogen which exhibited inhibition constants of 5, 91 and 54 muM against anthranilate synthase, isochorismate synthase and salicylate synthase respectively.


Assuntos
Aminopiridinas/química , Aminopiridinas/farmacologia , Ácido Corísmico/metabolismo , Ácidos Isonicotínicos/química , Ácidos Isonicotínicos/farmacologia , Piridinas/química , Piridinas/farmacologia , Piridonas/química , Piridonas/farmacologia , Aminopiridinas/síntese química , Aminopiridinas/metabolismo , Antranilato Sintase/antagonistas & inibidores , Antranilato Sintase/química , Antranilato Sintase/metabolismo , Bactérias/enzimologia , Domínio Catalítico , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Transferases Intramoleculares/antagonistas & inibidores , Transferases Intramoleculares/química , Transferases Intramoleculares/metabolismo , Ácidos Isonicotínicos/síntese química , Ácidos Isonicotínicos/metabolismo , Liases/antagonistas & inibidores , Liases/química , Liases/metabolismo , Modelos Moleculares , Piridinas/síntese química , Piridinas/metabolismo , Piridonas/síntese química , Piridonas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA