Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Open Biol ; 11(8): 210098, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34375548

RESUMEN

Glycolysis and gluconeogenesis are central pathways of metabolism across all domains of life. A prominent enzyme in these pathways is phosphoglucose isomerase (PGI), which mediates the interconversion of glucose-6-phosphate and fructose-6-phosphate. The predatory bacterium Bdellovibrio bacteriovorus leads a complex life cycle, switching between intraperiplasmic replicative and extracellular 'hunter' attack-phase stages. Passage through this complex life cycle involves different metabolic states. Here we present the unliganded and substrate-bound structures of the B. bacteriovorus PGI, solved to 1.74 Å and 1.67 Å, respectively. These structures reveal that an induced-fit conformational change within the active site is not a prerequisite for the binding of substrates in some PGIs. Crucially, we suggest a phenylalanine residue, conserved across most PGI enzymes but substituted for glycine in B. bacteriovorus and other select organisms, is central to the induced-fit mode of substrate recognition for PGIs. This enzyme also represents the smallest conventional PGI characterized to date and probably represents the minimal requirements for a functional PGI.


Asunto(s)
Bdellovibrio bacteriovorus/enzimología , Fructosafosfatos/metabolismo , Glucosa-6-Fosfato Isomerasa/química , Glucosa-6-Fosfato Isomerasa/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Modelos Moleculares , Unión Proteica , Conformación Proteica , Homología de Secuencia , Especificidad por Sustrato
2.
Nat Commun ; 12(1): 1228, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33623032

RESUMEN

Bacterial hybrid malic enzymes (MaeB grouping, multidomain) catalyse the transformation of malate to pyruvate, and are a major contributor to cellular reducing power and carbon flux. Distinct from other malic enzyme subtypes, the hybrid enzymes are regulated by acetyl-CoA, a molecular indicator of the metabolic state of the cell. Here we solve the structure of a MaeB protein, which reveals hybrid enzymes use the appended phosphotransacetylase (PTA) domain to form a hexameric sensor that communicates acetyl-CoA occupancy to the malic enzyme active site, 60 Å away. We demonstrate that allostery is governed by a large-scale rearrangement that rotates the catalytic subunits 70° between the two states, identifying MaeB as a new model enzyme for the study of ligand-induced conformational change. Our work provides the mechanistic basis for metabolic control of hybrid malic enzymes, and identifies inhibition-insensitive variants that may find utility in synthetic biology.


Asunto(s)
Bdellovibrio bacteriovorus/enzimología , Malato Deshidrogenasa/metabolismo , Acetilcoenzima A/metabolismo , Regulación Alostérica , Apoproteínas/química , Sitios de Unión , Biocatálisis , Cinética , Malato Deshidrogenasa/química , Modelos Moleculares , Movimiento (Física) , Dominios Proteicos
3.
Nat Commun ; 11(1): 4817, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32968056

RESUMEN

Lysozymes are among the best-characterized enzymes, acting upon the cell wall substrate peptidoglycan. Here, examining the invasive bacterial periplasmic predator Bdellovibrio bacteriovorus, we report a diversified lysozyme, DslA, which acts, unusually, upon (GlcNAc-) deacetylated peptidoglycan. B. bacteriovorus are known to deacetylate the peptidoglycan of the prey bacterium, generating an important chemical difference between prey and self walls and implying usage of a putative deacetyl-specific "exit enzyme". DslA performs this role, and ΔDslA strains exhibit a delay in leaving from prey. The structure of DslA reveals a modified lysozyme superfamily fold, with several adaptations. Biochemical assays confirm DslA specificity for deacetylated cell wall, and usage of two glutamate residues for catalysis. Exogenous DslA, added ex vivo, is able to prematurely liberate B. bacteriovorus from prey, part-way through the predatory lifecycle. We define a mechanism for specificity that invokes steric selection, and use the resultant motif to identify wider DslA homologues.


Asunto(s)
Bdellovibrio bacteriovorus/enzimología , Bdellovibrio bacteriovorus/metabolismo , Muramidasa/química , Muramidasa/metabolismo , Periplasma/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bdellovibrio bacteriovorus/genética , Pared Celular , Escherichia coli , Regulación Bacteriana de la Expresión Génica , Modelos Moleculares , Muramidasa/genética , Mutación , Peptidoglicano/metabolismo , Fenotipo , Conformación Proteica , Especificidad por Sustrato
4.
J Bacteriol ; 202(18)2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32601070

RESUMEN

Bdellovibrio bacteriovorus is an obligate predatory bacterium that invades and kills a broad range of Gram-negative prey cells, including human pathogens. Its potential therapeutic application has been the subject of increased research interest in recent years. However, an improved understanding of the fundamental molecular aspects of the predatory life cycle is crucial for developing this bacterium as a "living antibiotic." During intracellular growth, B. bacteriovorus secretes an arsenal of hydrolases, which digest the content of the host cell to provide growth nutrients for the predator, e.g., prey DNA is completely degraded by the nucleases. Here, we have, on a genetic and molecular level, characterized two secreted DNases from B. bacteriovorus, Bd0934 and Bd3507, and determined the temporal expression profile of other putative secreted nucleases. We conclude that Bd0934 and Bd3507 are likely a part of the predatosome but are not essential for the predation, host-independent growth, prey biofilm degradation, and self-biofilm formation. The detailed temporal expression analysis of genes encoding secreted nucleases revealed that these enzymes are produced in a sequential orchestrated manner. This work contributes to our understanding of the sequential breakdown of the prey nucleic acid by the nucleases secreted during the predatory life cycle of B. bacteriovorusIMPORTANCE Antibiotic resistance is a major global concern with few available new means to combat it. From a therapeutic perspective, predatory bacteria constitute an interesting tool. They not only eliminate the pathogen but also reduce the overall pool of antibiotic resistance genes through secretion of nucleases and complete degradation of exogenous DNA. Molecular knowledge of how these secreted DNases act will give us further insight into how antibiotic resistance, and the spread thereof, can be limited through the action of predatory bacteria.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bdellovibrio bacteriovorus/enzimología , Biopelículas , Endonucleasas/metabolismo , Bdellovibrio bacteriovorus/crecimiento & desarrollo , Escherichia coli , Regulación Bacteriana de la Expresión Génica
5.
ISME J ; 12(8): 2090-2095, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29849167

RESUMEN

Bdellovibrio bacteriovorus HD100 is a predatory bacterium which lives by invading the periplasm of Gram-negative bacteria and consuming them from within. Although B. bacteriovorus HD100 attacks only Gram-negative bacterial strains, our work here shows attack-phase predatory cells also benefit from interacting with Gram-positive biofilms. Using Staphylococcus aureus biofilms, we show this predator degrades the biofilm matrix, obtains nutrients and uses these to produce and secrete proteolytic enzymes to continue this process. When exposed to S. aureus biofilms, the transcriptome of B. bacteriovorus HD100 was analogous to that seen when present intraperiplasmically, suggesting it is responding similarly as when in a prey. Moreover, two of the induced proteases (Bd2269 and Bd2692) were purified and their activities against S. aureus biofilms verified. In addition, B. bacteriovorus HD100 gained several clear benefits from its interactions with S. aureus biofilms, including increased ATP pools and improved downstream predatory activities when provided prey.


Asunto(s)
Bdellovibrio bacteriovorus/fisiología , Biopelículas , Staphylococcus aureus/fisiología , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bdellovibrio bacteriovorus/enzimología , Bdellovibrio bacteriovorus/genética , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo
6.
Nat Microbiol ; 2(12): 1648-1657, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28974693

RESUMEN

Modification of essential bacterial peptidoglycan (PG)-containing cell walls can lead to antibiotic resistance; for example, ß-lactam resistance by L,D-transpeptidase activities. Predatory Bdellovibrio bacteriovorus are naturally antibacterial and combat infections by traversing, modifying and finally destroying walls of Gram-negative prey bacteria, modifying their own PG as they grow inside prey. Historically, these multi-enzymatic processes on two similar PG walls have proved challenging to elucidate. Here, with a PG-labelling approach utilizing timed pulses of multiple fluorescent D-amino acids, we illuminate dynamic changes that predator and prey walls go through during the different phases of bacteria:bacteria invasion. We show formation of a reinforced circular port-hole in the prey wall, L,D-transpeptidaseBd-mediated D-amino acid modifications strengthening prey PG during Bdellovibrio invasion, and a zonal mode of predator elongation. This process is followed by unconventional, multi-point and synchronous septation of the intracellular Bdellovibrio, accommodating odd- and even-numbered progeny formation by non-binary division.


Asunto(s)
Aminoácidos Diaminos/metabolismo , Aminoácidos/metabolismo , Bdellovibrio bacteriovorus/metabolismo , Pared Celular/química , Pared Celular/metabolismo , Peptidoglicano/química , Peptidoglicano/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Bdellovibrio/metabolismo , Bdellovibrio bacteriovorus/citología , Bdellovibrio bacteriovorus/enzimología , Bdellovibrio bacteriovorus/genética , Escherichia coli/metabolismo , Genes Bacterianos/genética , Bacterias Gramnegativas/metabolismo , Peptidil Transferasas/genética , Peptidil Transferasas/metabolismo , Eliminación de Secuencia , Factores de Tiempo
7.
FEBS J ; 284(18): 3029-3049, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28710792

RESUMEN

Many enzymes form homooligomers, yet the functional significance of self-association is seldom obvious. Herein, we examine the connection between oligomerization and catalytic function for proline utilization A (PutA) enzymes. PutAs are bifunctional enzymes that catalyze both reactions of proline catabolism. Type A PutAs are the smallest members of the family, possessing a minimal domain architecture consisting of N-terminal proline dehydrogenase and C-terminal l-glutamate-γ-semialdehyde dehydrogenase modules. Type A PutAs form domain-swapped dimers, and in one case (Bradyrhizobium japonicum PutA), two of the dimers assemble into a ring-shaped tetramer. Whereas the dimer has a clear role in substrate channeling, the functional significance of the tetramer is unknown. To address this question, we performed structural studies of four-type A PutAs from two clades of the PutA tree. The crystal structure of Bdellovibrio bacteriovorus PutA covalently inactivated by N-propargylglycine revealed a fold and substrate-channeling tunnel similar to other PutAs. Small-angle X-ray scattering (SAXS) and analytical ultracentrifugation indicated that Bdellovibrio PutA is dimeric in solution, in contrast to the prediction from crystal packing of a stable tetrameric assembly. SAXS studies of two other type A PutAs from separate clades also suggested that the dimer predominates in solution. To assess whether the tetramer of B. japonicum PutA is necessary for catalytic function, a hot spot disruption mutant that cleanly produces dimeric protein was generated. The dimeric variant exhibited kinetic parameters similar to the wild-type enzyme. These results implicate the domain-swapped dimer as the core structural and functional unit of type A PutAs. ENZYMES: Proline dehydrogenase (EC 1.5.5.2); l-glutamate-γ-semialdehyde dehydrogenase (EC 1.2.1.88). DATABASES: The atomic coordinates and structure factor amplitudes have been deposited in the Protein Data Bank under accession number 5UR2. The SAXS data have been deposited in the SASBDB under the following accession codes: SASDCP3 (BbPutA), SASDCQ3 (DvPutA 1.5 mg·mL-1 ), SASDCX3 (DvPutA 3.0 mg·mL-1 ), SASDCY3 (DvPutA 4.5 mg·mL-1 ), SASDCR3 (LpPutA 3.0 mg·mL-1 ), SASDCV3 (LpPutA 5.0 mg·mL-1 ), SASDCW3 (LpPutA 8.0 mg·mL-1 ), SASDCS3 (BjPutA 2.3 mg·mL-1 ), SASDCT3 (BjPutA 4.7 mg·mL-1 ), SASDCU3 (BjPutA 7.0 mg·mL-1 ), SASDCZ3 (R51E 2.3 mg·mL-1 ), SASDC24 (R51E 4.7 mg·mL-1 ), SASDC34 (R51E 7.0 mg·mL-1 ).


Asunto(s)
Alquinos/química , Proteínas Bacterianas/química , Bdellovibrio bacteriovorus/química , Bradyrhizobium/química , Glicina/análogos & derivados , Proteínas de la Membrana/química , Prolina/química , Alquinos/metabolismo , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bdellovibrio bacteriovorus/enzimología , Sitios de Unión , Bradyrhizobium/enzimología , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Glicina/química , Glicina/metabolismo , Cinética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Filogenia , Prolina/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Dispersión del Ángulo Pequeño , Homología Estructural de Proteína , Especificidad por Sustrato , Termodinámica , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...