Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Bacteriol ; 206(3): e0033323, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38411059

RESUMEN

Although bacterial peptidoglycan (PG) is highly conserved, some natural variations in PG biosynthesis and structure have evolved. Understanding the mechanisms and limits of such variation will inform our understanding of antibiotic resistance, innate immunity, and the evolution of bacteria. We have explored the constraints on PG evolution by blocking essential steps in PG biosynthesis in Vibrio fischeri and then selecting mutants with restored prototrophy. Here, we attempted to select prototrophic suppressors of a D-glutamate auxotrophic murI racD mutant. No suppressors were isolated on unsupplemented lysogeny broth salts (LBS), despite plating >1011 cells, nor were any suppressors generated through mutagenesis with ethyl methanesulfonate. A single suppressor was isolated on LBS supplemented with iso-D-gln, although the iso-D-gln subsequently appeared irrelevant. This suppressor has a genomic amplification formed by the creation of a novel junction that fuses proB to a gene encoding a putative broad-spectrum racemase of V. fischeri, bsrF. An engineered bsrF allele lacking the putative secretion signal (ΔSS-bsrF) also suppressed D-glu auxotrophy, resulting in PG that was indistinguishable from the wild type. The ΔSS-bsrF allele similarly suppressed the D-alanine auxotrophy of an alr mutant and restored prototrophy to a murI alr double mutant auxotrophic for both D-ala and D-glu. The ΔSS-bsrF allele increased resistance to D-cycloserine but had no effect on sensitivity to PG-targeting antibiotics penicillin, ampicillin, or vancomycin. Our work helps define constraints on PG evolution and reveals a periplasmic broad-spectrum racemase in V. fischeri that can be co-opted for PG biosynthesis, with concomitant D-cycloserine resistance. IMPORTANCE: D-Amino acids are used and produced by organisms across all domains of life, but often, their origins and roles are not well understood. In bacteria, D-ala and D-glu are structural components of the canonical peptidoglycan cell wall and are generated by dedicated racemases Alr and MurI, respectively. The more recent discovery of additional bacterial racemases is broadening our view and deepening our understanding of D-amino acid metabolism. Here, while exploring alternative PG biosynthetic pathways in Vibrio fischeri, we unexpectedly shed light on an unusual racemase, BsrF. Our results illustrate a novel mechanism for the evolution of antibiotic resistance and provide a new avenue for exploring the roles of non-canonical racemases and D-amino acids in bacteria.


Asunto(s)
Alanina Racemasa , Ácido Glutámico , Ácido Glutámico/metabolismo , Aliivibrio fischeri/genética , Aliivibrio fischeri/metabolismo , Racemasas y Epimerasas/metabolismo , Cicloserina , Peptidoglicano/metabolismo , Aminoácidos/metabolismo , Alanina Racemasa/metabolismo
2.
Protein Sci ; 32(10): e4781, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37703013

RESUMEN

The 11 lytic transglycosylases of Pseudomonas aeruginosa have overlapping activities in the turnover of the cell-wall peptidoglycan. Rare lipoprotein A (RlpA) is distinct among the 11 by its use of only peptidoglycan lacking peptide stems. The spatial localization of RlpA and its interactome within P. aeruginosa are unknown. We employed suppression of introduced amber codons at sites in the rlpA gene for the introduction of the unnatural-amino-acids Νζ -[(2-azidoethoxy)carbonyl]-l-lysine (compound 1) and Nζ -[[[3-(3-methyl-3H-diazirin-3-yl)propyl]amino]carbonyl]-l-lysine (compound 2). In live P. aeruginosa, full-length RlpA incorporating compound 1 into its sequence was fluorescently tagged using strained-promoted alkyne-azide cycloaddition and examined by fluorescence microscopy. RlpA is present at low levels along the sidewall length of the bacterium, and at higher levels at the nascent septa of replicating bacteria. In intact P. aeruginosa, UV photolysis of full-length RlpA having compound 2 within its sequence generated a transient reactive carbene, which engaged in photoaffinity capture of neighboring proteins. Thirteen proteins were identified. Three of these proteins-PBP1a, PBP5, and MreB-are members of the bacterial divisome. The use of the complementary methodologies of non-canonical amino-acid incorporation, photoaffinity proximity analysis, and fluorescent microscopy confirm a dominant septal location for the RlpA enzyme of P. aeruginosa, as a divisome-associated activity. This accomplishment adds to the emerging recognition of the value of these methodologies for identification of the intracellular localization of bacterial proteins.


Asunto(s)
Lipoproteína(a) , Pseudomonas aeruginosa , Lipoproteína(a)/metabolismo , Codón de Terminación/metabolismo , Peptidoglicano/metabolismo , Lisina/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(24): e2302580120, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37276423

RESUMEN

AmiA and AmiB are peptidoglycan-hydrolyzing enzymes from Escherichia coli that are required to break the peptidoglycan layer during bacterial cell division and maintain integrity of the cell envelope. In vivo, the activity of AmiA and AmiB is tightly controlled through their interactions with the membrane-bound FtsEX-EnvC complex. Activation of AmiA and AmiB requires access to a groove in the amidase-activating LytM domain of EnvC which is gated by ATP-driven conformational changes in FtsEX-EnvC complex. Here, we present a high-resolution structure of the isolated AmiA protein, confirming that it is autoinhibited in the same manner as AmiB and AmiC, and a complex of the AmiB enzymatic domain bound to the activating EnvC LytM domain. In isolation, the active site of AmiA is blocked by an autoinhibitory helix that binds directly to the catalytic zinc and fills the volume expected to accommodate peptidoglycan binding. In the complex, binding of the EnvC LytM domain induces a conformational change that displaces the amidase autoinhibitory helix and reorganizes the active site for activity. Our structures, together with complementary mutagenesis work, defines the conformational changes required to activate AmiA and/or AmiB through their interaction with their cognate activator EnvC.


Asunto(s)
Proteínas de Escherichia coli , Proteínas de Escherichia coli/metabolismo , Peptidoglicano/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Escherichia coli/metabolismo , Amidohidrolasas/metabolismo , Proteínas Bacterianas/metabolismo
4.
Environ Microbiol ; 23(10): 5823-5836, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33830599

RESUMEN

The bacterial cell wall is made of peptidoglycan (PG), a polymer that is essential for maintenance of cell shape and survival. Many bacteria alter their PG chemistry as a strategy to adapt their cell wall to external challenges. Therefore, identifying these environmental cues is important to better understand the interplay between microbes and their habitat. Here, we used the soil bacterium Pseudomonas putida to uncover cell wall modulators from plant extracts and found canavanine (CAN), a non-proteinogenic amino acid. We demonstrated that cell wall chemical editing by CAN is licensed by P. putida BSAR, a broad-spectrum racemase which catalyses production of dl-CAN from l-CAN, which is produced by many legumes. Importantly, d-CAN diffuses to the extracellular milieu thereby having a potential impact on other organisms inhabiting the same niche. Our results show that d-CAN alters dramatically the PG structure of Rhizobiales (e.g., Agrobacterium tumefaciens, Sinorhizobium meliloti), impairing PG crosslinkage and cell division. Using A. tumefaciens, we demonstrated that the detrimental effect of d-CAN is suppressed by a single amino acid substitution in the cell division PG transpeptidase penicillin binding protein 3a. Collectively, this work highlights the role of amino acid racemization in cell wall chemical editing and fitness.


Asunto(s)
Alphaproteobacteria , Peptidoglicano , Alphaproteobacteria/metabolismo , Proteínas Bacterianas/metabolismo , Canavanina/análisis , Canavanina/metabolismo , Pared Celular/metabolismo , Morfogénesis , Peptidoglicano/metabolismo
5.
Sci Rep ; 10(1): 5874, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32246076

RESUMEN

Complete Freund's adjuvant (CFA) has historically been one of the most useful tools of immunologists. Essentially comprised of dead mycobacteria and mineral oil, we asked ourselves what is special about the mycobacterial part of this adjuvant, and could it be recapitulated synthetically? Here, we demonstrate the essentiality of N-glycolylated peptidoglycan plus trehalose dimycolate (both unique in mycobacteria) for the complete adjuvant effect using knockouts and chemical complementation. A combination of synthetic N-glycolyl muramyl dipeptide and minimal trehalose dimycolate motif GlcC14C18 was able to upregulate dendritic cell effectors, plus induce experimental autoimmunity qualitatively similar but quantitatively milder compared to CFA. This research outlines how to substitute CFA with a consistent, molecularly-defined adjuvant which may inform the design of immunotherapeutic agents and vaccines benefitting from cell-mediated immunity. We also anticipate using synthetic microbe-associated molecular patterns (MAMPs) to study mycobacterial immunity and immunopathogenesis.


Asunto(s)
Adyuvante de Freund/metabolismo , Mycobacterium/metabolismo , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animales , Células Dendríticas/metabolismo , Femenino , Adyuvante de Freund/farmacología , Inmunidad Celular/efectos de los fármacos , Lectinas Tipo C/metabolismo , Ganglios Linfáticos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Adaptadora de Señalización NOD2/metabolismo , Peptidoglicano/metabolismo
6.
Microbiologyopen ; 9(5): e1015, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32134563

RESUMEN

Listeria monocytogenes is the causative agent of human listeriosis which has high hospitalization and mortality rates for individuals with weakened immune systems. The survival and dissemination of L. monocytogenes in adverse environments can be reinforced by the formation of biofilms. Therefore, this study aimed to understand the mechanisms underlying listerial biofilm development. Given that both nutrient availability and quorum sensing (QS) have been known as the factors influencing biofilm development, we hypothesized that the signal from a sentinel metabolite S-adenosylmethionine (SAM) and Agr-based QS could be synchronous in L. monocytogenes to modulate nutrient availability, the synthesis of extracellular polymeric substances (EPSs), and biofilm formation. We performed biofilm assays and quantitative real-time PCR to investigate how biofilm volumes and the expression of genes for the synthesis of EPS were affected by SAM supplementation, agr deletion, or both. We found that exogenously applied SAM induced biofilm formation and that the expression of genes encoding the EPS synthesis machineries was regulated by SAM and/or Agr QS. Moreover, the gene transcription of components acting in the methyl cycle for SAM synthesis and Agr QS was affected by the signals from the other system. In summary, we reveal an interconnection at the transcriptional level between metabolism and QS in L. monocytogenes and highlight the critical role of metabolite-oriented QS in biofilm development.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Matriz Extracelular de Sustancias Poliméricas/genética , Matriz Extracelular de Sustancias Poliméricas/metabolismo , Listeria monocytogenes/fisiología , Percepción de Quorum/genética , S-Adenosilmetionina/metabolismo , ADN Bacteriano/genética , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/genética , Mutación , Peptidoglicano/genética , Peptidoglicano/metabolismo
7.
FEBS Open Bio ; 9(12): 2025-2040, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31050202

RESUMEN

Multidrug-resistant Staphylococcus aureus infections place a huge burden on the healthcare sector and the wider community. An increasing rate of infections caused by methicillin-resistant Staphylococcus aureus (MRSA) has necessitated the development of alternative agents. We previously reported that usnic acid (UA) has activity against MRSA; here, we report the effect of UA in combination with norfloxacin on the drug resistance of MRSA clinical isolates. We observed that the combination of UA-norfloxacin significantly reduces the bacterial burden in mouse models infected with S. aureus, without causing any detectable associated toxicity. Proteomic analysis indicated that UA-norfloxacin induces oxidative stress within cells, which leads to membrane damage and inhibits metabolic activity and biosynthesis of peptidoglycan and fatty acids. Collectively, this study provides evidence that UA in combination with norfloxacin may be a potential candidate for development into a resistance-modifying agent for the treatment of invasive MRSA infections.


Asunto(s)
Benzofuranos/uso terapéutico , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Animales , Antibacterianos/farmacología , Resistencia a Medicamentos , Sinergismo Farmacológico , Ácidos Grasos/biosíntesis , Ácidos Grasos/metabolismo , Masculino , Staphylococcus aureus Resistente a Meticilina/metabolismo , Ratones , Pruebas de Sensibilidad Microbiana , Norfloxacino/farmacología , Peptidoglicano/metabolismo , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/metabolismo
8.
Dev Comp Immunol ; 98: 13-19, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30980872

RESUMEN

Peptidoglycan recognition proteins (PGRPs) are a family of pattern recognition receptors (PRRs) involved in host antibacterial responses, and their functions have been characterized in most invertebrate and vertebrate animals. However, little information is available regarding the function of frog PGRPs. In this study, a short-type PGRP (termed Xl-PGRP-S) gene was identified in the African clawed frog, Xenopus laevis. The predicted protein of Xl-PGRP-S contains several structural features known in PGRPs, including a typical PGRP domain and two closely spaced conserved cysteines. Xl-PGRP-S gene was constitutively expressed in all tissues examined, with the highest expression level observed in muscle. As a typical PRR, Xl-PGRP-S is inducible after peptidoglycan (PGN) stimulation, and has an ability to bind PGN. In addition, Xl-PGRP-S has been proven to have Zn2+-dependent amidase activity and antibacterial activity against Edwardsiella tarda. The present study represents the first discovery on the function of frog PGRPs, thus contributing to a better understanding of the functional evolution of PGRPs in early tetrapods.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Secuencia de Aminoácidos , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , Edwardsiella tarda/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Peptidoglicano/metabolismo , Filogenia , Unión Proteica , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/farmacología , Xenopus laevis/metabolismo , Zinc/metabolismo
9.
Pol J Microbiol ; 67(3): 315-320, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30451448

RESUMEN

Listeria monocytogenes is Gram-positive bacterial pathogen, a causative agent of food poisoning and systemic disease - listeriosis. This species is still susceptible to several conventionally used antibiotics but an increase in its resistance has been reported. For this reason the search for new, alternative therapies is an urgent task. Silver nanoparticles seem to be the promising antibacterial agent. Minimal inhibitory concentration of silver nanoparticles was determined. Sublethal concentrations were used in study of nanosilver effect on cells lysis by estimation of the number of cells surviving the treatment with 0.25 or 0.5 of minimal inhibitory concentrations of silver nanoparticles. Autolysis of isolated peptidoglycan was studied by measuring the absorbance of preparation subjected to nanosilver treatment. Silver nanoparticles effect on L. monocytogenes envelopes permeability was determined by measuring the efflux of cF, DNA and proteins. It was demonstrated that nanosilver enhanced the lysis of L. monocytogenes cells and, to the lesser extent, autolysis of isolated peptidoglycan. The increase in the efflux of carboxyfluoresceine, DNA and proteins was also noted. The obtained results allow to postulate that L. monocytogenes peptidoglycan, constituting the main component of cell wall, is the target of silver nanoparticles activity against this pathogen.


Asunto(s)
Antibacterianos/farmacología , Bacteriólisis , Permeabilidad de la Membrana Celular/efectos de los fármacos , Listeria monocytogenes/efectos de los fármacos , Peptidoglicano/metabolismo , Plata/farmacología , Nanopartículas del Metal/química , Pruebas de Sensibilidad Microbiana
10.
Clin Dermatol ; 36(3): 376-389, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29908580

RESUMEN

The gut is the largest lymphoid organ in the body. The human microbiome is composed of trillions of bacteria. The DNA of these bacteria dwarfs the human genome. Diet and ethanol can cause rapid shifts in the number and types of bacteria in the gut. The psoriatic microbiome is similar to that seen in alcoholics; there is a decrease in bacterial diversity and overgrowth of bacteria in the small bowel. Psoriatics often have liver disease and deficiencies in bile acids. Psoriasis is a disease characterized by a leaky gut. All of the comorbidities of this disease are due to systemic endotoxemia. Bacterial peptidoglycans absorbed from the gut have direct toxic effects on the liver and skin. Their absorption, as well as endotoxin absorption, must be eliminated to treat psoriasis successfully. Endotoxin absorption is markedly increased by ethanol and peppers. Bioflavonoids, such as quercetin and citrus bioflavonoids, prevent this absorption. Bile acids, given orally, break up endotoxin in the intestinal lumen. Pathogens, including Helicobacter pylori and Streptococcus pyogenes, must be eliminated with antimicrobial therapy for any treatment to work. A complete protocol for curing psoriasis is provided.


Asunto(s)
Ácidos y Sales Biliares/uso terapéutico , Endotoxinas/metabolismo , Flavonoides/uso terapéutico , Microbioma Gastrointestinal , Peptidoglicano/metabolismo , Psoriasis/tratamiento farmacológico , Psoriasis/microbiología , Animales , Antibacterianos/uso terapéutico , Traslocación Bacteriana , Variación Biológica Individual , Dieta , Endotoxemia/complicaciones , Endotoxemia/tratamiento farmacológico , Tracto Gastrointestinal/inmunología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori , Humanos
11.
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
12.
Bioconjug Chem ; 28(9): 2310-2315, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28737895

RESUMEN

Peptidoglycan (PG) scaffolds are critical components of bacterial cell walls. They counter internal turgor pressure to prevent lysis and protect against external insults. It was recently discovered that various types of bacteria release large quantities of PG building blocks (d-amino acids) into their surrounding medium. Contrarily, cultured bacteria were also found to incorporate d-amino acids (both natural and synthetic) from the medium directly into their PG scaffold. These two processes may potentially function, in concert, to metabolically remodel PG in live host organisms. However, demonstration that bacteria can decorate their cell surfaces with exogenous d-amino acids was limited to in vitro culture conditions. We present the first evidence that bacteria remodel their PG with exogenous d-amino acids in a live host animal. A tetrazine click partner was conjugated onto the side chain of a d-amino acid to capture incorporation into the bacterial PG scaffold using a complementary click-reactive fluorophore. Staphylococcus aureus infected Caenorhabditis elegans treated with exogenous d-amino acids readily revealed in vivo PG labeling. These results suggest that extracellular d-amino acids may provide pathogens with a mode of late-stage in vivo cell-surface remodeling.


Asunto(s)
Aminoácidos/metabolismo , Caenorhabditis elegans/microbiología , Pared Celular/metabolismo , Interacciones Huésped-Patógeno , Peptidoglicano/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Animales , Caenorhabditis elegans/fisiología , Modelos Animales de Enfermedad , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/veterinaria
13.
J Am Chem Soc ; 139(31): 10597-10600, 2017 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-28727445

RESUMEN

Antibiotic-resistant strains of Staphylococcus aureus pose a major threat to human health and there is an ongoing need for new antibiotics to treat resistant infections. In a high throughput screen (HTS) of 230 000 small molecules designed to identify bioactive wall teichoic acid (WTA) inhibitors, we identified one hit, which was expanded through chemical synthesis into a small panel of potent compounds. We showed that these compounds target TarG, the transmembrane component of the two-component ATP-binding cassette (ABC) transporter TarGH, which exports WTA precursors to the cell surface for attachment to peptidoglycan. We purified, for the first time, a WTA transporter and have reconstituted ATPase activity in proteoliposomes. We showed that this new compound series inhibits TarH-catalyzed ATP hydrolysis even though the binding site maps to TarG near the opposite side of the membrane. These are the first ABC transporter inhibitors shown to block ATPase activity by binding to the transmembrane domain. The compounds have potential as therapeutic agents to treat S. aureus infections, and purification of the transmembrane transporter will enable further development.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Staphylococcus aureus/efectos de los fármacos , Ácidos Teicoicos/farmacología , Adenosina Trifosfatasas/antagonistas & inhibidores , Sitios de Unión , Pared Celular/química , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Modelos Biológicos , Estructura Molecular , Peptidoglicano/química , Peptidoglicano/metabolismo , Unión Proteica/efectos de los fármacos
14.
Proc Natl Acad Sci U S A ; 114(27): 7019-7024, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28630348

RESUMEN

Phosphoglycosyl transferases (PGTs) are integral membrane proteins with diverse architectures that catalyze the formation of polyprenol diphosphate-linked glycans via phosphosugar transfer from a nucleotide diphosphate-sugar to a polyprenol phosphate. There are two PGT superfamilies that differ significantly in overall structure and topology. The polytopic PGT superfamily, represented by MraY and WecA, has been the subject of many studies because of its roles in peptidoglycan and O-antigen biosynthesis. In contrast, less is known about a second, extensive superfamily of PGTs that reveals a core structure with dual domain architecture featuring a C-terminal soluble globular domain and a predicted N-terminal membrane-associated domain. Representative members of this superfamily are the Campylobacter PglCs, which initiate N-linked glycoprotein biosynthesis and are implicated in virulence and pathogenicity. Despite the prevalence of dual domain PGTs, their mechanism of action is unknown. Here, we present the mechanistic analysis of PglC, a prototypic dual domain PGT from Campylobacter concisus Using a luminescence-based assay, together with substrate labeling and kinetics-based approaches, complementary experiments were carried out that support a ping-pong mechanism involving a covalent phosphosugar intermediate for PglC. Significantly, mass spectrometry-based approaches identified Asp93, which is part of a highly conserved AspGlu dyad found in all dual domain PGTs, as the active-site nucleophile of the enzyme involved in the formation of the covalent adduct. The existence of a covalent phosphosugar intermediate provides strong support for a ping-pong mechanism of PglC, differing fundamentally from the ternary complex mechanisms of representative polytopic PGTs.


Asunto(s)
Antibacterianos/química , Proteínas Bacterianas/química , Campylobacter/enzimología , Transferasas/química , Ácido Aspártico/química , Dominio Catalítico , Ácido Glutámico/química , Cinética , Luminiscencia , Modelos Químicos , Peptidoglicano/metabolismo , Especificidad por Sustrato , Azúcares/química
15.
Appl Environ Microbiol ; 83(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28003196

RESUMEN

Libraries of defined mutants are valuable research tools but necessarily lack gene knockouts that are lethal under the conditions used in library construction. In this study, we augmented a Vibrio fischeri mutant library generated on a rich medium (LBS, which contains [per liter] 10 g of tryptone, 5 g of yeast extract, 20 g of NaCl, and 50 mM Tris [pH 7.5]) by selecting transposon insertion mutants on supplemented LBS and screening for those unable to grow on LBS. We isolated strains with insertions in alr, glr (murI), glmS, several heme biosynthesis genes, and ftsA, as well as a mutant disrupted 14 bp upstream of ftsQ Mutants with insertions in ftsA or upstream of ftsQ were recovered by addition of Mg2+ to LBS, but their cell morphology and motility were affected. The ftsA mutant was more strongly affected and formed cells or chains of cells that appeared to wind back on themselves helically. Growth of mutants with insertions in glmS, alr, or glr was recovered with N-acetylglucosamine (NAG), d-alanine, or d-glutamate, respectively. We hypothesized that NAG, d-alanine, or d-glutamate might be available to V. fischeri in the Euprymna scolopes light organ; however, none of these mutants colonized the host effectively. In contrast, hemA and hemL mutants, which are auxotrophic for δ-aminolevulinate (ALA), colonized at wild-type levels, although mutants later in the heme biosynthetic pathway were severely impaired or unable to colonize. Our findings parallel observations that legume hosts provide Bradyrhizobium symbionts with ALA, but they contrast with virulence phenotypes of hemA mutants in some pathogens. The results further inform our understanding of the symbiotic light organ environment.IMPORTANCE By supplementing a rich yeast-based medium, we were able to recover V. fischeri mutants with insertions in conditionally essential genes, and further characterization of these mutants provided new insights into this bacterium's symbiotic environment. Most notably, we show evidence that the squid host can provide V. fischeri with enough ALA to support its growth in the light organ, paralleling the finding that legumes provide Bradyrhizobium ALA in symbiotic nodules. Taken together, our results show how a simple method of augmenting already rich media can expand the reach and utility of defined mutant libraries.


Asunto(s)
Aliivibrio fischeri/genética , Aliivibrio fischeri/metabolismo , Elementos Transponibles de ADN/genética , Decapodiformes/microbiología , Simbiosis/genética , Simbiosis/fisiología , Alanina/metabolismo , Aliivibrio/genética , Aliivibrio/crecimiento & desarrollo , Aliivibrio fischeri/crecimiento & desarrollo , Aliivibrio fischeri/fisiología , Ácido Aminolevulínico/metabolismo , Animales , Proteínas Bacterianas/genética , Decapodiformes/fisiología , Biblioteca de Genes , Genes Bacterianos/genética , Ácido Glutámico/metabolismo , Hemina/metabolismo , Interacciones Huésped-Patógeno/fisiología , Luz , Proteínas de la Membrana/genética , Mutación , Peptidoglicano/metabolismo , Fenotipo , Photobacterium/genética , Photobacterium/metabolismo , Virulencia
16.
Antonie Van Leeuwenhoek ; 109(6): 827-32, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27027520

RESUMEN

During an investigation of microbial diversity in medicinal herbs, a novel actinobacterium, strain NEAU-KD1(T) was isolated from the rhizosphere of Peucedanum praeruptorum Dunn collected from Xianglu Mountain in Heilongjiang Province, northeast China and characterized using a polyphasic approach. The organism was found to have the typical chemotaxonomic and morphological characteristics of the genus Mumia. Cells were observed to be non-spore-forming and irregular cocci. The cell wall was found to contain LL-diaminopimelic acid as the cell wall diamino acid. The whole-cell sugars were detected as galactose and rhamnose and the predominant menaquinone was identified as MK-9(H4). The polar lipids were found to consist of diphosphatidylglycerol, phosphatidylglycerol, phosphoglycolipid and five unidentified phospholipids. The major cellular fatty acids were determined to be composed of C16:0, 10-methyl C18:0 and C18:1ω7c. The phylogenetic analysis based on 16S rRNA gene sequence also indicated that strain NEAU-KD1(T) belongs to the genus Mumia and with high sequence similarity to Mumia flava NBRC 109973(T) (97.6 % sequence similarity). The results of DNA-DNA hybridization and the phenotypic characteristics indicated that strain NEAU-KD1(T) could be distinguished from its close phylogenetic relative. Thus, strain NEAU-KD1(T) can be concluded to represent a novel species of the genus Mumia, for which the name Mumia xiangluensis sp. nov. is proposed. The type strain is NEAU-KD1(T) (=CGMCC 4.7305(T) = DSM 101040(T)).


Asunto(s)
Propionibacteriaceae/clasificación , Propionibacteriaceae/aislamiento & purificación , Rizosfera , Tracheophyta/microbiología , Técnicas de Tipificación Bacteriana , Metabolismo de los Hidratos de Carbono , Pared Celular/metabolismo , China , Cloruros/metabolismo , ADN Bacteriano/genética , ADN Ribosómico/genética , Ácido Diaminopimélico/metabolismo , Ácidos Grasos/metabolismo , Peptidoglicano/metabolismo , Fosfolípidos/metabolismo , Filogenia , Propionibacteriaceae/genética , Propionibacteriaceae/metabolismo , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Microbiología del Suelo
17.
PLoS One ; 10(4): e0120620, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25874956

RESUMEN

Curcumin is a natural dietary compound with antimicrobial activity against various gram positive and negative bacteria. This study aims to investigate the proteome level alterations in Bacillus subtilis due to curcumin treatment and identification of its molecular/cellular targets to understand the mechanism of action. We have performed a comprehensive proteomic analysis of B. subtilis AH75 strain at different time intervals of curcumin treatment (20, 60 and 120 min after the drug exposure, three replicates) to compare the protein expression profiles using two complementary quantitative proteomic techniques, 2D-DIGE and iTRAQ. To the best of our knowledge, this is the first comprehensive longitudinal investigation describing the effect of curcumin treatment on B. subtilis proteome. The proteomics analysis revealed several interesting targets such UDP-N-acetylglucosamine 1-carboxyvinyltransferase 1, putative septation protein SpoVG and ATP-dependent Clp protease proteolytic subunit. Further, in silico pathway analysis using DAVID and KOBAS has revealed modulation of pathways related to the fatty acid metabolism and cell wall synthesis, which are crucial for cell viability. Our findings revealed that curcumin treatment lead to inhibition of the cell wall and fatty acid synthesis in addition to differential expression of many crucial proteins involved in modulation of bacterial metabolism. Findings obtained from proteomics analysis were further validated using 5-cyano-2,3-ditolyl tetrazolium chloride (CTC) assay for respiratory activity, resazurin assay for metabolic activity and membrane integrity assay by potassium and inorganic phosphate leakage measurement. The gene expression analysis of selected cell wall biosynthesis enzymes has strengthened the proteomics findings and indicated the major effect of curcumin on cell division.


Asunto(s)
Antibacterianos/farmacología , Bacillus subtilis/efectos de los fármacos , Curcumina/farmacología , Proteoma/efectos de los fármacos , Bacillus subtilis/genética , Bacillus subtilis/crecimiento & desarrollo , Bacillus subtilis/metabolismo , Bacillus subtilis/ultraestructura , Proteínas Bacterianas/genética , Pared Celular/metabolismo , Simulación por Computador , Evaluación Preclínica de Medicamentos , Electroforesis en Gel Bidimensional/métodos , Ácidos Grasos/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Modelos Biológicos , Peptidoglicano/metabolismo , Fosfatos/metabolismo , Potasio/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Espectrometría de Masas en Tándem/métodos , Factores de Tiempo
18.
Antimicrob Agents Chemother ; 59(4): 2337-42, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25645840

RESUMEN

A pyridodiazepine amine inhibitor of Helicobacter pylori glutamate racemase (MurI) was characterized. The compound was selectively active against H. pylori, and growth suppression was shown to be mediated through the inhibition of MurI by several methods. In killing kinetics experiments, the compound showed concentration-independent activity, with about a 2-log loss of viability in 24 h. A demonstration of efficacy in a mouse infection model was attempted but not achieved, and this was attributed to the failure to attain extended exposure levels above the MIC for >95% of the time. This index and magnitude were derived from pharmacokinetic-pharmacodynamic (PK-PD) studies with amoxicillin, another inhibitor of peptidoglycan biosynthesis that showed slow killing kinetics similar to those of the pyridodiazepine amines. These studies indicate that MurI and other enzymes involved in peptidoglycan biosynthesis may be less desirable targets for monotherapy directed against H. pylori if once-a-day dosing is required.


Asunto(s)
Isomerasas de Aminoácido/antagonistas & inhibidores , Antibacterianos/uso terapéutico , Azepinas/uso terapéutico , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/enzimología , Piridinas/uso terapéutico , Amoxicilina/farmacocinética , Amoxicilina/farmacología , Animales , Antibacterianos/farmacocinética , Azepinas/farmacocinética , Femenino , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/sangre , Infecciones por Helicobacter/microbiología , Cinética , Ratones , Pruebas de Sensibilidad Microbiana , Peptidoglicano/metabolismo , Piridinas/farmacocinética , Ratas , Ratas Sprague-Dawley
19.
J Bacteriol ; 196(24): 4239-44, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25246478

RESUMEN

The mycobacterial cell wall frequently has been used as a target for drug development, and d-glutamate, synthesized by glutamate racemase (MurI), is an important component of peptidoglycan. While the essentiality of the murI gene has been shown in several bacterial species, including Escherichia coli, Bacillus anthracis, and Streptococcus pneumoniae, studies in mycobacteria have not yet provided definitive results. This study aimed to determine whether murI is indeed essential and can serve as a possible target for structure-aided drug design. We have achieved this goal by creating a ΔmurI strain of Mycobacterium smegmatis, a close relative of Mycobacterium tuberculosis. The deletion of the murI gene in M. smegmatis could be achieved only in minimal medium supplemented with D-glutamate, demonstrating that MurI is essential for growth and that glutamate racemase is the only source of D-glutamate for peptidoglycan synthesis in M. smegmatis.


Asunto(s)
Isomerasas de Aminoácido/genética , Isomerasas de Aminoácido/metabolismo , Genes Esenciales , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/crecimiento & desarrollo , Pared Celular/metabolismo , Medios de Cultivo/química , Eliminación de Gen , Ácido Glutámico/metabolismo , Mycobacterium smegmatis/genética , Peptidoglicano/metabolismo
20.
PLoS One ; 8(2): e57579, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23469030

RESUMEN

The peptidoglycan (PG) is a macromolecular component of the bacterial cell wall that maintains the shape and integrity of the cell. The PG of Caulobacter crescentus, unlike that of many other Gram-negative bacteria, has repeatedly been shown to contain significant amounts of glycine. This compositional peculiarity has been deemed an intrinsic characteristic of this species. By performing a comprehensive qualitative and quantitative analysis of the C. crescentus PG by high-performance liquid chromatography (HPLC) and mass spectrometry (MS), we show here that glycine incorporation into the C. crescentus PG depends on the presence of exogenous glycine in the growth medium. High levels of glycine were detected at the fifth position of the peptide side chains of PG isolated from C. crescentus cells grown in the complex laboratory medium PYE or in defined medium (M2G) supplemented with casamino acids or glycine alone. In contrast, glycine incorporation was undetectable when cells were grown in M2G medium lacking glycine. Remarkably, glycine incorporation into C. crescentus peptidoglycan occurred even in the presence of low millimolar to sub-millimolar concentrations of free glycine. High glycine content in the PG had no obvious effects on growth rates, mode of PG incorporation or cell morphology. Hence, the C. crescentus PG is able to retain its physiological functions in cell growth and morphogenesis despite significant alterations in its composition, in what we deem to be unprecedented plasticity.


Asunto(s)
Caulobacter crescentus/metabolismo , Medios de Cultivo , Glicina/metabolismo , Peptidoglicano/metabolismo , Caulobacter crescentus/crecimiento & desarrollo , Cromatografía Líquida de Alta Presión , Microscopía Electrónica de Transmisión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA