Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cell Sci ; 136(5)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36861884

RESUMEN

The pathological accumulation of cholesterol is a signature feature of Niemann-Pick type C (NPC) disease, in which excessive lipid levels induce Purkinje cell death in the cerebellum. NPC1 encodes a lysosomal cholesterol-binding protein, and mutations in NPC1 drive cholesterol accumulation in late endosomes and lysosomes (LE/Ls). However, the fundamental role of NPC proteins in LE/L cholesterol transport remains unclear. Here, we demonstrate that NPC1 mutations impair the projection of cholesterol-containing membrane tubules from the surface of LE/Ls. A proteomic survey of purified LE/Ls identified StARD9 as a novel lysosomal kinesin responsible for LE/L tubulation. StARD9 contains an N-terminal kinesin domain, a C-terminal StART domain, and a dileucine signal shared with other lysosome-associated membrane proteins. Depletion of StARD9 disrupts LE/L tubulation, paralyzes bidirectional LE/L motility and induces accumulation of cholesterol in LE/Ls. Finally, a novel StARD9 knock-out mouse recapitulates the progressive loss of Purkinje cells in the cerebellum. Together, these studies identify StARD9 as a microtubule motor protein responsible for LE/L tubulation and provide support for a novel model of LE/L cholesterol transport that becomes impaired in NPC disease.


Asunto(s)
Cinesinas , Células de Purkinje , Animales , Ratones , Cinesinas/genética , Proteómica , Transporte Biológico , Lisosomas , Ratones Noqueados
2.
J Bacteriol ; 202(11)2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32205460

RESUMEN

Streptococcus pyogenes, or group A Streptococcus (GAS), is both a pathogen and an asymptomatic colonizer of human hosts and produces a large number of surface-expressed and secreted factors that contribute to a variety of infection outcomes. The GAS-secreted cysteine protease SpeB has been well studied for its effects on the human host; however, despite its broad proteolytic activity, studies on how this factor is utilized in polymicrobial environments are lacking. Here, we utilized various forms of SpeB protease to evaluate its antimicrobial and antibiofilm properties against the clinically important human colonizer Staphylococcus aureus, which occupies niches similar to those of GAS. For our investigation, we used a skin-tropic GAS strain, AP53CovS+, and its isogenic ΔspeB mutant to compare the production and activity of native SpeB protease. We also generated active and inactive forms of recombinant purified SpeB for functional studies. We demonstrate that SpeB exhibits potent biofilm disruption activity at multiple stages of S. aureus biofilm formation. We hypothesized that the surface-expressed adhesin SdrC in S. aureus was cleaved by SpeB, which contributed to the observed biofilm disruption. Indeed, we found that SpeB cleaved recombinant SdrC in vitro and in the context of the full S. aureus biofilm. Our results suggest an understudied role for the broadly proteolytic SpeB as an important factor for GAS colonization and competition with other microorganisms in its niche.IMPORTANCEStreptococcus pyogenes (GAS) causes a range of diseases in humans, ranging from mild to severe, and produces many virulence factors in order to be a successful pathogen. One factor produced by many GAS strains is the protease SpeB, which has been studied for its ability to cleave and degrade human proteins, an important factor in GAS pathogenesis. An understudied aspect of SpeB is the manner in which its broad proteolytic activity affects other microorganisms that co-occupy niches similar to that of GAS. The significance of the research reported herein is the demonstration that SpeB can degrade the biofilms of the human pathogen Staphylococcus aureus, which has important implications for how SpeB may be utilized by GAS to successfully compete in a polymicrobial environment.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas , Exotoxinas/metabolismo , Staphylococcus aureus/enzimología , Staphylococcus aureus/fisiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/enzimología , Proteínas Bacterianas/genética , Exotoxinas/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Staphylococcus aureus/genética , Streptococcus pyogenes/genética
3.
Nat Commun ; 10(1): 5567, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804467

RESUMEN

SPOR domains are widely present in bacterial proteins that recognize cell-wall peptidoglycan strands stripped of the peptide stems. This type of peptidoglycan is enriched in the septal ring as a product of catalysis by cell-wall amidases that participate in the separation of daughter cells during cell division. Here, we document binding of synthetic denuded glycan ligands to the SPOR domain of the lytic transglycosylase RlpA from Pseudomonas aeruginosa (SPOR-RlpA) by mass spectrometry and structural analyses, and demonstrate that indeed the presence of peptide stems in the peptidoglycan abrogates binding. The crystal structures of the SPOR domain, in the apo state and in complex with different synthetic glycan ligands, provide insights into the molecular basis for recognition and delineate a conserved pattern in other SPOR domains. The biological and structural observations presented here are followed up by molecular-dynamics simulations and by exploration of the effect on binding of distinct peptidoglycan modifications.


Asunto(s)
Pared Celular/química , Proteínas de Escherichia coli/química , Escherichia coli/química , Peptidoglicano/química , Dominios Proteicos , Bacillus subtilis/química , Bacillus subtilis/metabolismo , Secuencia de Carbohidratos , Pared Celular/metabolismo , Cristalografía por Rayos X , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Lipoproteínas/química , Lipoproteínas/metabolismo , Simulación de Dinámica Molecular , Peptidoglicano/metabolismo , Unión Proteica , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/metabolismo
4.
ACS Chem Biol ; 12(10): 2552-2557, 2017 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-28937735

RESUMEN

We have produced draft whole-genome sequences for two bacterial strains reported to produce the bulgecins as well as NRPS-derived monobactam ß-lactam antibiotics. We propose classification of ATCC 31363 as Paraburkholderia acidophila. We further reaffirm that ATCC 31433 (Burkholderia ubonensis subsp. mesacidophila) is a taxonomically distinct producer of bulgecins with notable gene regions shared with Paraburkholderia acidophila. We use RAST multiple-gene comparison and MASH distancing with published genomes to order the draft contigs and identify unique gene regions for characterization. Forty-eight natural-product gene clusters are presented from PATRIC (RASTtk) and antiSMASH annotations. We present evidence that the 10 genes that follow the sulfazecin and isosulfazecin pathways in both species are likely involved in bulgecin A biosynthesis.


Asunto(s)
Burkholderiaceae/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano , Glicopéptidos/metabolismo , Burkholderiaceae/genética , Glicopéptidos/química , Glicopéptidos/genética , Familia de Multigenes
5.
Chembiochem ; 18(17): 1696-1702, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28591487

RESUMEN

The major constituent of bacterial cell walls is peptidoglycan, which, in its crosslinked form, is a polymer of considerable complexity that encases the entire bacterium. A functional cell wall is indispensable for survival of the organism. There are several dozen enzymes that assemble and disassemble the peptidoglycan dynamically within each bacterial generation. Understanding of the nature of these transformations is critical knowledge for these events. Octasaccharide peptidoglycans were prepared and studied with seven recombinant cell-wall-active enzymes (SltB1, MltB, RlpA, mutanolysin, AmpDh2, AmpDh3, and PBP5). With the use of highly sensitive mass spectrometry methods, we described the breadth of reactions that these enzymes catalyzed with peptidoglycan and shed light on the nature of the cell wall alteration performed by these enzymes. The enzymes exhibit broadly distinct preferences for their substrate peptidoglycans in the reactions that they catalyze.


Asunto(s)
Bacterias/metabolismo , Pared Celular/metabolismo , Enzimas/metabolismo , Biocatálisis , Cromatografía Líquida de Alta Presión , Endopeptidasas/genética , Endopeptidasas/metabolismo , Enzimas/genética , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Espectrometría de Masas , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Peptidoglicano/análisis , Peptidoglicano/química , Peptidoglicano/metabolismo , Pseudomonas aeruginosa/enzimología , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Streptomyces griseus/enzimología , Especificidad por Sustrato , Transferasas/genética , Transferasas/metabolismo
6.
Angew Chem Int Ed Engl ; 56(10): 2735-2739, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28128504

RESUMEN

An enzyme superfamily, the lytic transglycosylases (LTs), occupies the space between the two membranes of Gram-negative bacteria. LTs catalyze the non-hydrolytic cleavage of the bacterial peptidoglycan cell-wall polymer. This reaction is central to the growth of the cell wall, for excavating the cell wall for protein insertion, and for monitoring the cell wall so as to initiate resistance responses to cell-wall-acting antibiotics. The nefarious Gram-negative pathogen Pseudomonas aeruginosa encodes eleven LTs. With few exceptions, their substrates and functions are unknown. Each P. aeruginosa LT was expressed as a soluble protein and evaluated with a panel of substrates (both simple and complex mimetics of their natural substrates). Thirty-one distinct products distinguish these LTs with respect to substrate recognition, catalytic activity, and relative exolytic or endolytic ability. These properties are foundational to an understanding of the LTs as catalysts and as antibiotic targets.


Asunto(s)
Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Pseudomonas aeruginosa/enzimología , Biocatálisis , Pared Celular/química , Pared Celular/metabolismo , Conformación Molecular , Pseudomonas aeruginosa/citología
7.
J Am Chem Soc ; 139(4): 1448-1451, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28079369

RESUMEN

A complex link exists between cell-wall recycling/repair and the manifestation of resistance to ß-lactam antibiotics in many Enterobacteriaceae and Pseudomonas aeruginosa. This process is mediated by specific cell-wall-derived muropeptide products. These muropeptides are internalized into the cytoplasm and bind to the transcriptional regulator AmpR, which controls the cytoplasmic events that lead to expression of ß-lactamase, an antibiotic-resistance determinant. The effector-binding domain (EBD) of AmpR was purified to homogeneity. We document that the EBD exists exclusively as a dimer, even at a concentration as low as 1 µM. The EBD binds to the suppressor ligand UDP-N-acetyl-ß-d-muramyl-l-Ala-γ-d-Glu-meso-DAP-d-Ala-d-Ala and binds to two activator muropeptides, N-acetyl-ß-d-glucosamine-(1→4)-1,6-anhydro-N-acetyl-ß-d-muramyl-l-Ala-γ-d-Glu-meso-DAP-d-Ala-d-Ala and 1,6-anhydro-N-acetyl-ß-d-muramyl-l-Ala-γ-d-Glu-meso-DAP-d-Ala-d-Ala, as assessed by non-denaturing mass spectrometry. The EBD does not bind to 1,6-anhydro-N-acetyl-ß-d-muramyl-l-Ala-γ-d-Glu-meso-DAP. This binding selectivity revises the dogma in the field. The crystal structure of the EBD dimer was solved to 2.2 Å resolution. The EBD crystallizes in a "closed" conformation, in contrast to the "open" structure required to bind the muropeptides. Structural issues of this ligand recognition are addressed by molecular dynamics simulations, which reveal significant differences among the complexes with the effector molecules.


Asunto(s)
Proteínas Bacterianas/química , Péptidos/química , Pseudomonas aeruginosa/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Péptidos/metabolismo , Dominios Proteicos , Pseudomonas aeruginosa/metabolismo
8.
Structure ; 25(2): 295-304, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-28089449

RESUMEN

Active in the aqueous cellular environment where a massive excess of water is perpetually present, enzymes that catalyze the transfer of an electrophile to a non-water nucleophile (transferases) require specific strategies to inhibit mechanistically related hydrolysis reactions. To identify principles that confer transferase versus hydrolase reaction specificity, we exploited two enzymes that use highly similar catalytic apparatuses to catalyze the transglycosylation (a transferase reaction) or hydrolysis of α-1,3-glucan linkages in the cyclic tetrasaccharide cycloalternan (CA). We show that substrate binding to non-catalytic domains and a conformationally stable active site promote CA transglycosylation, whereas a distinct pattern of active site conformational change is associated with CA hydrolysis. These findings defy the classic view of induced-fit conformational change and illustrate a mechanism by which a stable hydrophobic binding site can favor transferase activity and disfavor hydrolysis. Application of these principles could facilitate the rational reengineering of transferases with desired catalytic properties.


Asunto(s)
Actinomycetales/enzimología , Glucosidasas/química , Glicósido Hidrolasas/química , Listeria monocytogenes/enzimología , Oligosacáridos/química , Agua/química , Actinomycetales/genética , Secuencias de Aminoácidos , Sitios de Unión , Biocatálisis , Conformación de Carbohidratos , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Glucosidasas/genética , Glucosidasas/metabolismo , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Glicosilación , Hidrólisis , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Listeria monocytogenes/genética , Modelos Moleculares , Oligosacáridos/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Agua/metabolismo
10.
Angew Chem Int Ed Engl ; 55(24): 6882-6, 2016 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-27111486

RESUMEN

Muropeptides are a group of bacterial natural products generated from the cell wall in the course of its turnover. These compounds are cell-wall recycling intermediates and are also involved in signaling within the bacterium. However, the identity of these signaling molecules remains elusive. The identification and characterization of 20 muropeptides from Pseudomonas aeruginosa is described. The least abundant of these metabolites is present at 100 and the most abundant at 55,000 molecules per bacterium. Analysis of these muropeptides under conditions of induction of resistance to a ß-lactam antibiotic identified two signaling muropeptides (N-acetylglucosamine-1,6-anhydro-N-acetylmuramyl pentapeptide and 1,6-anhydro-N-acetylmuramyl pentapeptide). Authentic synthetic samples of these metabolites were shown to activate expression of ß-lactamase in the absence of any ß-lactam antibiotic, thus indicating that they serve as chemical signals in this complex biochemical pathway.


Asunto(s)
Antibacterianos/farmacología , Péptidos/metabolismo , Pseudomonas aeruginosa/química , Resistencia betalactámica/efectos de los fármacos , beta-Lactamas/farmacología , Antibacterianos/química , Conformación Molecular , Péptidos/química , Pseudomonas aeruginosa/metabolismo , beta-Lactamasas/metabolismo , beta-Lactamas/química
11.
ACS Chem Neurosci ; 6(10): 1658-64, 2015 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-26241578

RESUMEN

SB-3CT is a potent and selective inhibitor of matrix metalloproteinase (MMP)-2 and -9, which has shown efficacy in an animal model of severe traumatic brain injury (TBI). However, SB-3CT is poorly water-soluble and is metabolized primarily to p-hydroxy SB-3CT (2), a more potent inhibitor than SB-3CT. We synthesized the O-phosphate prodrug (3) of compound 2 to enhance its water solubility by more than 2000-fold. The prodrug 3 was a poor MMP inhibitor, but readily hydrolyzed to the active 2 in human blood. Pharmacokinetics and brain distribution studies in mice showed that 2 crossed the blood-brain barrier (BBB) and achieved therapeutic concentrations in the brain. The prodrug 3/compound 2 was evaluated in a mouse model of severe TBI and found to significantly decrease the brain lesion volume and improve neurological outcomes. MMP-9 inhibition by a water-soluble thiirane inhibitor is a promising therapy for treatment of TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Compuestos Heterocíclicos con 1 Anillo/uso terapéutico , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Sulfonas/uso terapéutico , Animales , Área Bajo la Curva , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/fisiopatología , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Compuestos Heterocíclicos con 1 Anillo/farmacología , Concentración 50 Inhibidora , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Endogámicos C57BL , Examen Neurológico , Desempeño Psicomotor/efectos de los fármacos , Solubilidad , Sulfonas/farmacología , Agua/metabolismo
12.
J Am Chem Soc ; 137(1): 190-200, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25495032

RESUMEN

Pseudomonas aeruginosa is an opportunistic Gram-negative bacterial pathogen. A primary contributor to its ability to resist ß-lactam antibiotics is the expression, following detection of the ß-lactam, of the AmpC ß-lactamase. As AmpC expression is directly linked to the recycling of the peptidoglycan of the bacterial cell wall, an important question is the identity of the signaling molecule(s) in this relationship. One mechanism used by clinical strains to elevate AmpC expression is loss of function of penicillin-binding protein 4 (PBP4). As the mechanism of the ß-lactams is PBP inactivation, this result implies that the loss of the catalytic function of PBP4 ultimately leads to induction of antibiotic resistance. PBP4 is a bifunctional enzyme having both dd-carboxypeptidase and endopeptidase activities. Substrates for both the dd-carboxypeptidase and the 4,3-endopeptidase activities were prepared by multistep synthesis, and their turnover competence with respect to PBP4 was evaluated. The endopeptidase activity is specific to hydrolysis of 4,3-cross-linked peptidoglycan. PBP4 catalyzes both reactions equally well. When P. aeruginosa is grown in the presence of a strong inducer of AmpC, the quantities of both the stem pentapeptide (the substrate for the dd-carboxypeptidase activity) and the 4,3-cross-linked peptidoglycan (the substrate for the 4,3-endopeptidase activity) increase. In the presence of ß-lactam antibiotics these altered cell-wall segments enter into the muropeptide recycling pathway, the conduit connecting the sensing event in the periplasm and the unleashing of resistance mechanisms in the cytoplasm.


Asunto(s)
Proteínas de Unión a las Penicilinas/metabolismo , Pseudomonas aeruginosa/química , Antibacterianos/farmacología , Biocatálisis , Conformación Molecular , Proteínas de Unión a las Penicilinas/química , Proteínas de Unión a las Penicilinas/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/metabolismo , Resistencia betalactámica/efectos de los fármacos
13.
ACS Chem Biol ; 9(9): 2058-66, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-24988330

RESUMEN

The lytic transglycosylases are essential bacterial enzymes that catalyze the nonhydrolytic cleavage of the glycan strands of the bacterial cell wall. We describe here the structural and catalytic properties of MltC, one of the seven lytic transglycosylases found in the genome of the Gram-negative bacterium Escherichia coli. The 2.3 Å resolution X-ray structure of a soluble construct of MltC shows a unique, compared to known lytic transglycosylase structures, two-domain structure characterized by an expansive active site of 53 Å length extending through an interface between the domains. The structures of three complexes of MltC with cell wall analogues suggest the positioning of the peptidoglycan in the active site both as a substrate and as a product. One complex is suggested to correspond to an intermediate in the course of sequential and exolytic cleavage of the peptidoglycan. Moreover, MltC partitioned its reactive oxocarbenium-like intermediate between trapping by the C6-hydroxyl of the muramyl moiety (lytic transglycosylase activity, the major path) and by water (muramidase activity). Genomic analysis identifies the presence of an MltC homologue in no less than 791 bacterial genomes. While the role of MltC in cell wall assembly and maturation remains uncertain, we propose a functional role for this enzyme as befits the uniqueness of its two-domain structure.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Glicosiltransferasas/química , Glicosiltransferasas/metabolismo , Dominio Catalítico , Pared Celular/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/citología , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Modelos Moleculares , Mutación , Peptidoglicano/química , Conformación Proteica , Estructura Terciaria de Proteína
14.
Biochemistry ; 53(10): 1548-50, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24564530

RESUMEN

ß-Lactam antibiotics have faced obsolescence with the emergence of methicillin-resistant Staphylococcus aureus (MRSA). A complex set of events ensues upon exposure of MRSA to these antibiotics, which culminates in proteolysis of BlaI or MecI, two gene repressors, and results in the induction of resistance. We report studies on the mechanism of binding of these gene repressors to the operator regions by fluorescence anisotropy. Within the range of in vivo concentrations for BlaI and MecI, these proteins interact with their regulatory elements in a reversible manner, as both a monomer and a dimer.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Staphylococcus aureus Resistente a Meticilina/metabolismo , Proteínas Represoras/metabolismo , beta-Lactamas/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Regiones Operadoras Genéticas , Operón , Regiones Promotoras Genéticas , Proteínas Represoras/química , Proteínas Represoras/genética
15.
ACS Chem Biol ; 9(1): 105-10, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24053680

RESUMEN

A complication of diabetes is the inability of wounds to heal in diabetic patients. Diabetic wounds are refractory to healing due to the involvement of activated matrix metalloproteinases (MMPs), which remodel the tissue resulting in apoptosis. There are no readily available methods that identify active unregulated MMPs. With the use of a novel inhibitor-tethered resin that binds exclusively to the active forms of MMPs, coupled with proteomics, we quantified MMP-8 and MMP-9 in a mouse model of diabetic wounds. Topical treatment with a selective MMP-9 inhibitor led to acceleration of wound healing, re-epithelialization, and significantly attenuated apoptosis. In contrast, selective pharmacological inhibition of MMP-8 delayed wound healing, decreased re-epithelialization, and exhibited high apoptosis. The MMP-9 activity makes the wounds refractory to healing, whereas that of MMP-8 is beneficial. The treatment of diabetic wounds with a selective MMP-9 inhibitor holds great promise in providing heretofore-unavailable opportunities for intervention of this disease.


Asunto(s)
Complicaciones de la Diabetes/tratamiento farmacológico , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Animales , Complicaciones de la Diabetes/enzimología , Complicaciones de la Diabetes/patología , Metaloproteinasa 8 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/química , Ratones , Modelos Moleculares
16.
J Org Chem ; 78(23): 12224-8, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24251875

RESUMEN

An efficient synthesis of olefins by the coupling of stabilized, semistabilized, and nonstabilized phosphorus ylides with various carbonyl compounds in the presence of silver carbonate is reported. Wittig olefination of aromatic, heteroaromatic, and aliphatic aldehydes (yields >63%) and a ketone (yield 42%) are demonstrated. These reactions proceed overnight at room temperature, under weakly basic conditions, and as such extend the applicability of the Wittig reaction to base-sensitive reactants.


Asunto(s)
Alquenos/síntesis química , Carbonatos/química , Compuestos de Plata/química , Alquenos/química , Cristalografía por Rayos X , Modelos Moleculares , Estructura Molecular
17.
J Am Chem Soc ; 135(34): 12604-7, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23931161

RESUMEN

Bacterial cell wall is a polymer of considerable complexity that is in constant equilibrium between synthesis and recycling. AmpDh3 is a periplasmic zinc protease of Pseudomonas aeruginosa , which is intimately involved in cell-wall remodeling. We document the hydrolytic reactions that this enzyme performs on the cell wall. The process removes the peptide stems from the peptidoglycan, the major constituent of the cell wall. We document that the majority of the reactions of this enzyme takes place on the polymeric insoluble portion of the cell wall, as opposed to the fraction that is released from it. We show that AmpDh3 is tetrameric both in crystals and in solution. Based on the X-ray structures of the enzyme in complex with two synthetic cell-wall-based ligands, we present for the first time a model for a multivalent anchoring of AmpDh3 onto the cell wall, which lends itself to its processive remodeling.


Asunto(s)
Pared Celular/metabolismo , Metaloproteasas/metabolismo , Pseudomonas aeruginosa/enzimología , Zinc/metabolismo , Pared Celular/enzimología , Cristalografía por Rayos X , Metaloproteasas/química , Modelos Moleculares , Conformación Molecular , Pseudomonas aeruginosa/citología , Pseudomonas aeruginosa/metabolismo , Zinc/química
18.
J Am Chem Soc ; 135(28): 10318-10321, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23819763

RESUMEN

The zinc protease AmpDh2 is a virulence determinant of Pseudomonas aeruginosa, a problematic human pathogen. The mechanism of how the protease manifests virulence is not known, but it is known that it turns over the bacterial cell wall. The reaction of AmpDh2 with the cell wall was investigated, and nine distinct turnover products were characterized by LC/MS/MS. The enzyme turns over both the cross-linked and noncross-linked cell wall. Three high-resolution X-ray structures, the apo enzyme and two complexes with turnover products, were solved. The X-ray structures show how the dimeric protein interacts with the inner leaflet of the bacterial outer membrane and that the two monomers provide a more expansive surface for recognition of the cell wall. This binding surface can accommodate the 3D solution structure of the cross-linked cell wall.


Asunto(s)
Proteínas Bacterianas/química , Metaloproteasas/química , Pseudomonas aeruginosa/enzimología , Factores de Virulencia/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Metaloproteasas/metabolismo , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Factores de Virulencia/metabolismo
19.
J Am Chem Soc ; 135(13): 4950-3, 2013 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-23510438

RESUMEN

A group of Gram-negative bacteria, including the problematic pathogen Pseudomonas aeruginosa, has linked the steps in cell-wall recycling with the ability to manifest resistance to ß-lactam antibiotics. A key step at the crossroads of the two events is performed by the protease AmpD, which hydrolyzes the peptide in the metabolite that influences these events. In contrast to other organisms that harbor this elaborate system, the genomic sequences of P. aeruginosa reveal it to have three paralogous genes for this protease, designated as ampD, ampDh2, and ampDh3. The recombinant gene products were purified to homogeneity, and their functions were assessed by the use of synthetic samples of three bacterial metabolites in cell-wall recycling and of three surrogates of cell-wall peptidoglycan. The results unequivocally identify AmpD as the bona fide recycling enzyme and AmpDh2 and AmpDh3 as enzymes involved in turnover of the bacterial cell wall itself. These findings define for the first time the events mediated by these three enzymes that lead to turnover of a key cell-wall recycling metabolite as well as the cell wall itself in its maturation.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Metaloproteasas/química , Metaloproteasas/metabolismo , Pseudomonas aeruginosa/enzimología , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Cromatografía Líquida de Alta Presión
20.
J Am Chem Soc ; 135(9): 3311-4, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23421439

RESUMEN

The reactions of all seven Escherichia coli lytic transglycosylases with purified bacterial sacculus are characterized in a quantitative manner. These reactions, which initiate recycling of the bacterial cell wall, exhibit significant redundancy in the activities of these enzymes along with some complementarity. These discoveries underscore the importance of the functions of these enzymes for recycling of the cell wall.


Asunto(s)
Pared Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Escherichia coli/metabolismo , Glicósido Hidrolasas/metabolismo , Pared Celular/química , Pared Celular/enzimología , Escherichia coli/citología , Conformación Molecular , Sáculo y Utrículo/química , Sáculo y Utrículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...