Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38562742

RESUMO

Antibiotics have dose-dependent effects on exposed bacteria. The medicinal use of antibiotics relies on their growth-inhibitory activities at sufficient concentrations. At subinhibitory concentrations, exposure effects vary widely among different antibiotics and bacteria. Bacillus subtilis responds to bacteriostatic translation inhibitors by mobilizing a population of cells (MOB-Mobilized Bacillus) to spread across a surface. How B. subtilis regulates the antibiotic-induced mobilization is not known. In this study, we used chloramphenicol to identify regulatory functions that B. subtilis requires to coordinate cell mobilization following subinhibitory exposure. We measured changes in gene expression and metabolism and mapped the results to a network of regulatory proteins that direct the mobile response. Our data reveal that several transcriptional regulators coordinately control the reprogramming of metabolism to support mobilization. The network regulates changes in glycolysis, nucleotide metabolism, and amino acid metabolism that are signature features of the mobilized population. Among the hundreds of genes with changing expression, we identified two, pdhA and pucA, where the magnitudes of their changes in expression, and in the abundance of associated metabolites, reveal hallmark metabolic features of the mobilized population. Using reporters of pdhA and pucA expression, we visualized the separation of major branches of metabolism in different regions of the mobilized population. Our results reveal a regulated response to chloramphenicol exposure that enables a population of bacteria in different metabolic states to mount a coordinated mobile response.

2.
Curr Opin Microbiol ; 51: 64-71, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31344518

RESUMO

Microorganisms produce biologically active natural products, some of which are useful as antibiotics and other medicines. A great demand for new antibiotics exists due to the diversity of pathogens and their mechanisms of drug resistance. Antibiotics were discovered as natural metabolites that enable a microorganism to suppress the growth of a competitor. Although the pace of discovery has slowed dramatically, new approaches to identifying antibiotics show promise for the future. Among many modern approaches to discovery, co-culturing different species and understanding the molecular bases of their interactions is opening new windows to antibiotic discovery. Here we review several examples to illustrate how co-culturing as an approach is producing new insights into the biology of specialized metabolism. Understanding the varied functions of specialized metabolites, combined with use of innovative and advanced analytical tools, indicates that studies of microbial interactions will enhance the discovery of new antibiotics and other natural products.


Assuntos
Antibacterianos/farmacologia , Fenômenos Fisiológicos Bacterianos , Descoberta de Drogas , Interações Microbianas , Antibacterianos/metabolismo , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Produtos Biológicos/metabolismo , Produtos Biológicos/farmacologia
3.
mSphere ; 3(1)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29507890

RESUMO

Competitive interactions between bacteria reveal physiological adaptations that benefit fitness. Bacillus subtilis is a Gram-positive species with several adaptive mechanisms for competition and environmental stress. Biofilm formation, sporulation, and motility are the outcomes of widespread changes in a population of B. subtilis. These changes emerge from complex, regulated pathways for adapting to external stresses, including competition from other species. To identify competition-specific functions, we cultured B. subtilis with multiple species of Streptomyces and observed altered patterns of growth for each organism. In particular, when plated on agar medium near Streptomyces venezuelae, B. subtilis initiates a robust and reproducible mobile response. To investigate the mechanistic basis for the interaction, we determined the type of motility used by B. subtilis and isolated inducing metabolites produced by S. venezuelae. Bacillus subtilis has three defined forms of motility: swimming, swarming, and sliding. Streptomyces venezuelae induced sliding motility specifically in our experiments. The inducing agents produced by S. venezuelae were identified as chloramphenicol and a brominated derivative at subinhibitory concentrations. Upon further characterization of the mobile response, our results demonstrated that subinhibitory concentrations of chloramphenicol, erythromycin, tetracycline, and spectinomycin all activate a sliding motility response by B. subtilis. Our data are consistent with sliding motility initiating under conditions of protein translation stress. This report underscores the importance of hormesis as an early warning system for potential bacterial competitors and antibiotic exposure. IMPORTANCE Antibiotic resistance is a major challenge for the effective treatment of infectious diseases. Identifying adaptive mechanisms that bacteria use to survive low levels of antibiotic stress is important for understanding pathways to antibiotic resistance. Furthermore, little is known about the effects of individual bacterial interactions on multispecies communities. This work demonstrates that subinhibitory amounts of some antibiotics produced by streptomycetes induce active motility in B. subtilis, which may alter species interaction dynamics among species-diverse bacterial communities in natural environments. The use of antibiotics at subinhibitory concentrations results in many changes in bacteria, including changes in biofilm formation, small-colony variants, formation of persisters, and motility. Identifying the mechanistic bases of these adaptations is crucial for understanding how bacterial communities are impacted by antibiotics.

4.
J Antibiot (Tokyo) ; 71(3): 372-381, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29348524

RESUMO

The linearmycin family of polyketides was originally classified as antifungal metabolites. However, in addition to antifungal activity, we previously found that linearmycins cause cellular lysis and colony degradation of the Gram-positive bacterium Bacillus subtilis. We recently showed that Streptomyces sp. strain Mg1 incorporates linearmycins into extracellular vesicles, which are capable of lysing B. subtilis. However, the mechanism of linearmycin-induced lysis was hitherto unexplored. Therefore, we sought to determine how linearmycin-laden vesicles cause lysis. In this study, we found that linearmycins inhibited the growth of all Gram-positive bacteria that we tested, but lysis was limited to some Bacillus species. Next, we found that linearmycin-induced lysis occurred even when cellular metabolism and growth were inhibited, which suggested that linearmycins possess the intrinsic capacity to lyse cells, unlike cell-wall targeting antibiotics. We showed that linearmycin exposure caused changes consistent with rapid depolarization of the B. subtilis cytoplasmic membrane, which was correlated with a loss of viability. Finally, using liposomes as in vitro membrane models, we demonstrated that linearmycins are capable of disrupting lipid bilayers without any other cellular components. Taken together, our results strongly indicate that the cytoplasmic membrane is the direct antibacterial target of linearmycins.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Policetídeos/farmacologia , Antibacterianos/isolamento & purificação , Antifúngicos , Bacillus/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Bicamadas Lipídicas , Lipossomos , Potenciais da Membrana/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Policetídeos/isolamento & purificação
5.
Cell Chem Biol ; 24(10): 1238-1249.e7, 2017 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-28919037

RESUMO

Specialized metabolites support bacterial competitive fitness as antibiotics, signals, pigments, and metal scavengers. Little is known about how specialized metabolites are processed and trafficked for their diverse competitive functions. Linearmycins A and B are linear polyketides with antifungal and antibacterial activity but are colony-localized in imaging mass spectrometry of Streptomyces sp. Mg1 (S. sp. Mg1). To decipher a connection between colony localization and antibiotic activity, we identified the linearmycin gene cluster and investigated linearmycin production and distribution by S. sp. Mg1. Our results uncover a large family of variant linearmycins with limited solubility in aqueous solution. We hypothesized that extracellular vesicles may traffic the lipid-like linearmycins. We found that vesicles isolated from culture supernatants contained linearmycins. Surprisingly, abolishing production of linearmycins in S. sp. Mg1 also diminished extracellular vesicle production. Our results reveal integration of linearmycin biosynthesis with production of extracellular vesicles, suggesting a deep connection between specialized metabolism and bacterial membrane physiology.


Assuntos
Membrana Celular/metabolismo , Vesículas Extracelulares/metabolismo , Policetídeos/metabolismo , Streptomyces/citologia , Streptomyces/metabolismo , Bacillus subtilis/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Deleção de Genes , Família Multigênica/genética , Mutação , Policetídeos/farmacologia , Streptomyces/genética
6.
J Bacteriol ; 199(18)2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28461449

RESUMO

Bacteria use two-component signaling systems to adapt and respond to their competitors and changing environments. For instance, competitor bacteria may produce antibiotics and other bioactive metabolites and sequester nutrients. To survive, some species of bacteria escape competition through antibiotic production, biofilm formation, or motility. Specialized metabolite production and biofilm formation are relatively well understood for bacterial species in isolation. How bacteria control these functions when competitors are present is not well studied. To address fundamental questions relating to the competitive mechanisms of different species, we have developed a model system using two species of soil bacteria, Bacillus subtilis and Streptomyces sp. strain Mg1. Using this model, we previously found that linearmycins produced by Streptomyces sp. strain Mg1 cause lysis of B. subtilis cells and degradation of colony matrix. We identified strains of B. subtilis with mutations in the two-component signaling system yfiJK operon that confer dual phenotypes of specific linearmycin resistance and biofilm morphology. We determined that expression of the ATP-binding cassette (ABC) transporter yfiLMN operon, particularly yfiM and yfiN, is necessary for biofilm morphology. Using transposon mutagenesis, we identified genes that are required for YfiLMN-mediated biofilm morphology, including several chaperones. Using transcriptional fusions, we found that YfiJ signaling is activated by linearmycins and other polyene metabolites. Finally, using a truncated YfiJ, we show that YfiJ requires its transmembrane domain to activate downstream signaling. Taken together, these results suggest coordinated dual antibiotic resistance and biofilm morphology by a single multifunctional ABC transporter promotes competitive fitness of B. subtilisIMPORTANCE DNA sequencing approaches have revealed hitherto unexplored diversity of bacterial species in a wide variety of environments that includes the gastrointestinal tract of animals and the rhizosphere of plants. Interactions between different species in bacterial communities have impacts on our health and industry. However, many approaches currently used to study whole bacterial communities do not resolve mechanistic details of interspecies interactions, including how bacteria sense and respond to their competitors. Using a competition model, we have uncovered dual functions for a previously uncharacterized two-component signaling system involved in specific antibiotic resistance and biofilm morphology. Insights gleaned from signaling within interspecies interaction models build a more complete understanding of gene functions important for bacterial communities and will enhance community-level analytical approaches.


Assuntos
Antibacterianos/metabolismo , Antibiose , Bacillus subtilis/fisiologia , Biofilmes/crescimento & desenvolvimento , Viabilidade Microbiana , Streptomyces/fisiologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Fusão Gênica Artificial , Bacillus subtilis/efeitos dos fármacos , Elementos de DNA Transponíveis , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Mutagênese Insercional , Mutação , Transdução de Sinais , Streptomyces/metabolismo
7.
Front Microbiol ; 7: 1234, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27551280

RESUMO

In the environment, bacteria live in complex multispecies communities. These communities span in scale from small, multicellular aggregates to billions or trillions of cells within the gastrointestinal tract of animals. The dynamics of bacterial communities are determined by pairwise interactions that occur between different species in the community. Though interactions occur between a few cells at a time, the outcomes of these interchanges have ramifications that ripple through many orders of magnitude, and ultimately affect the macroscopic world including the health of host organisms. In this review we cover how bacterial competition influences the structures of bacterial communities. We also emphasize methods and insights garnered from culture-dependent pairwise interaction studies, metagenomic analyses, and modeling experiments. Finally, we argue that the integration of multiple approaches will be instrumental to future understanding of the underlying dynamics of bacterial communities.

8.
J Bacteriol ; 198(16): 2145-55, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27246570

RESUMO

Microbial communities span many orders of magnitude, ranging in scale from hundreds of cells on a single particle of soil to billions of cells within the lumen of the gastrointestinal tract. Bacterial cells in all habitats are members of densely populated local environments that facilitate competition between neighboring cells. Accordingly, bacteria require dynamic systems to respond to the competitive challenges and the fluctuations in environmental circumstances that tax their fitness. The assemblage of bacteria into communities provides an environment where competitive mechanisms are developed into new strategies for survival. In this minireview, we highlight a number of mechanisms used by bacteria to compete between species. We focus on recent discoveries that illustrate the dynamic and multifaceted functions used in bacterial competition and discuss how specific mechanisms provide a foundation for understanding bacterial community development and function.


Assuntos
Bactérias/classificação , Bactérias/metabolismo , Fenômenos Fisiológicos Bacterianos , Biodiversidade , Meio Ambiente , Sistemas de Secreção Tipo VI
9.
PLoS Genet ; 12(1): e1005807, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26752286

RESUMO

[This corrects the article DOI: 10.1371/journal.pgen.1005722.].

10.
PLoS Genet ; 11(12): e1005722, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26647299

RESUMO

Bacteria have diverse mechanisms for competition that include biosynthesis of extracellular enzymes and antibiotic metabolites, as well as changes in community physiology, such as biofilm formation or motility. Considered collectively, networks of competitive functions for any organism determine success or failure in competition. How bacteria integrate different mechanisms to optimize competitive fitness is not well studied. Here we study a model competitive interaction between two soil bacteria: Bacillus subtilis and Streptomyces sp. Mg1 (S. Mg1). On an agar surface, colonies of B. subtilis suffer cellular lysis and progressive degradation caused by S. Mg1 cultured at a distance. We identify the lytic and degradative activity (LDA) as linearmycins, which are produced by S. Mg1 and are sufficient to cause lysis of B. subtilis. We obtained B. subtilis mutants spontaneously resistant to LDA (LDAR) that have visibly distinctive morphology and spread across the agar surface. Every LDAR mutant identified had a missense mutation in yfiJK, which encodes a previously uncharacterized two-component signaling system. We confirmed that gain-of-function alleles in yfiJK cause a combination of LDAR, changes in colony morphology, and motility. Downstream of yfiJK are the yfiLMN genes, which encode an ATP-binding cassette transporter. We show that yfiLMN genes are necessary for LDA resistance. The developmental phenotypes of LDAR mutants are genetically separable from LDA resistance, suggesting that the two competitive functions are distinct, but regulated by a single two-component system. Our findings suggest that a subpopulation of B. subtilis activate an array of defensive responses to counter lytic stress imposed by competition. Coordinated regulation of development and antibiotic resistance is a streamlined mechanism to promote competitive fitness of bacteria.


Assuntos
Bacillus subtilis/genética , Resistência Microbiana a Medicamentos/genética , Aptidão Genética , Streptomyces/genética , Antibacterianos/farmacologia , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/crescimento & desenvolvimento , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Mutação , Streptomyces/efeitos dos fármacos , Streptomyces/crescimento & desenvolvimento
11.
Appl Environ Microbiol ; 80(18): 5603-10, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25002419

RESUMO

Myxococcus xanthus and Bacillus subtilis are common soil-dwelling bacteria that produce a wide range of secondary metabolites and sporulate under nutrient-limiting conditions. Both organisms affect the composition and dynamics of microbial communities in the soil. However, M. xanthus is known to be a predator, while B. subtilis is not. A screen of various prey led to the finding that M. xanthus is capable of consuming laboratory strains of B. subtilis, while the ancestral strain, NCIB3610, was resistant to predation. Based in part on recent characterization of several strains of B. subtilis, we were able to determine that the pks gene cluster, which is required for production of bacillaene, is the major factor allowing B. subtilis NCIB3610 cells to resist predation by M. xanthus. Furthermore, purified bacillaene was added exogenously to domesticated strains, resulting in resistance to predation. Lastly, we found that M. xanthus is incapable of consuming B. subtilis spores even from laboratory strains, indicating the evolutionary fitness of sporulation as a survival strategy. Together, the results suggest that bacillaene inhibits M. xanthus predation, allowing sufficient time for development of B. subtilis spores.


Assuntos
Anti-Infecciosos/metabolismo , Bacillus subtilis/crescimento & desenvolvimento , Bacillus subtilis/metabolismo , Myxococcus xanthus/metabolismo , Polienos/metabolismo , Esporos Bacterianos/crescimento & desenvolvimento , Viabilidade Microbiana
12.
Genome Announc ; 1(4)2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23908282

RESUMO

We report a draft genome assembly of Streptomyces sp. strain Mg1, a competitive soil isolate with multiple secondary metabolite gene clusters.

13.
Science ; 339(6123): 1071-4, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23371552

RESUMO

The conserved kinases Mps1 and Ipl1/Aurora B are critical for enabling chromosomes to attach to microtubules so that partner chromosomes will be segregated correctly from each other, but the precise roles of these kinases have been unclear. We imaged live yeast cells to elucidate the stages of chromosome-microtubule interactions and their regulation by Ipl1 and Mps1 through meiosis I. Ipl1 was found to release kinetochore-microtubule (kMT) associations after meiotic entry, liberating chromosomes to begin homologous pairing. Surprisingly, most chromosome pairs began their spindle interactions with incorrect kMT attachments. Ipl1 released these improper connections, whereas Mps1 triggered the formation of new force-generating microtubule attachments. This microtubule release and reattachment cycle could prevent catastrophic chromosome segregation errors in meiosis.


Assuntos
Segregação de Cromossomos/fisiologia , Cromossomos Fúngicos/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Meiose/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , Aurora Quinases , Segregação de Cromossomos/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Cinetocoros/enzimologia , Meiose/genética , Microtúbulos/enzimologia , Mutação , Proteínas Serina-Treonina Quinases/genética , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
14.
Proc Natl Acad Sci U S A ; 109(32): 13082-7, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22826229

RESUMO

Many species of bacteria secrete natural products that inhibit the growth or development of competing species. In turn, competitors may develop or acquire resistance to antagonistic molecules. Few studies have investigated the interplay of these countervailing forces in direct competition between two species. We have used an imaging mass spectrometry (IMS) approach to track metabolites exchanged between Bacillus subtilis and Streptomyces sp. Mg1 cultured together. Surfactin is a cyclic lipopeptide produced by B. subtilis that inhibits the formation of aerial hyphae by streptomycetes. IMS analysis exposed an addition of 18 mass units to surfactin in the agar proximal to Streptomyces sp. Mg1 but not other streptomycetes tested. The spatially resolved change in the mass of surfactin indicated hydrolysis of the molecule. We observed that the aerial growth of Streptomyces sp. Mg1 was resistant to inhibition by surfactin, which suggests that hydrolysis was a mechanism of resistance. To identify possible enzymes from Streptomyces sp. Mg1 with surfactin hydrolase activity, we isolated secreted proteins and identified candidates by mass spectrometry. We purified one candidate enzyme that hydrolyzed surfactin in vitro. We tested the role of this enzyme in surfactin resistance by deleting the corresponding gene from the S. Mg1 genome. We observed that aerial growth by the ΔsfhA mutant strain was now sensitive to surfactin. Our results identify an enzyme that hydrolyzes surfactin and confers resistance to aerial growth inhibition, which demonstrates the effective use of an IMS approach to track natural product modifications during interspecies competition.


Assuntos
Bacillus subtilis/metabolismo , Farmacorresistência Bacteriana/fisiologia , Lipopeptídeos/metabolismo , Interações Microbianas/fisiologia , Peptídeos Cíclicos/metabolismo , Streptomyces/metabolismo , Bacillus subtilis/fisiologia , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Hidrolases/genética , Hidrolases/metabolismo , Espectroscopia de Ressonância Magnética , Streptomyces/enzimologia , Streptomyces/fisiologia , Espectrometria de Massas em Tandem
15.
Antonie Van Leeuwenhoek ; 102(3): 435-45, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22777252

RESUMO

Soil streptomycetes are saprotrophic bacteria that secrete numerous secondary metabolites and enzymes for extracellular functions. Many streptomycetes produce antibiotics thought to protect vegetative mycelia from competing organisms. Here we report that an organism isolated from soil, Streptomyces sp. Mg1, actively degrades colonies and causes cellular lysis of Bacillus subtilis when the organisms are cultured together. We predicted that the inhibition and degradation of B. subtilis colonies in this competition depends upon a combination of secreted factors, including small molecule metabolites and enzymes. To begin to unravel this complex competitive phenomenon, we use a MALDI imaging mass spectrometry strategy to map the positions of metabolites secreted by both organisms. In this report, we show that Streptomyces sp. Mg1 produces the macrolide antibiotic chalcomycin A, which contributes to inhibition of B. subtilis growth in combination with other, as yet unidentified factors. We suggest that efforts to understand competitive and cooperative interactions between bacterial species benefit from assays that pair living organisms and probe the complexity of metabolic exchanges between them.


Assuntos
Antibacterianos/metabolismo , Antibiose , Bacillus subtilis/efeitos dos fármacos , Bacteriólise , Macrolídeos/metabolismo , Streptomyces/fisiologia , Microbiologia do Solo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Streptomyces/isolamento & purificação , Streptomyces/metabolismo
16.
Proc Natl Acad Sci U S A ; 107(37): 16286-90, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20805502

RESUMO

During bacterial cannibalism, a differentiated subpopulation harvests nutrients from their genetically identical siblings to allow continued growth in nutrient-limited conditions. Hypothesis-driven imaging mass spectrometry (IMS) was used to identify metabolites active in a Bacillus subtilis cannibalism system in which sporulating cells lyse nonsporulating siblings. Two candidate molecules with sequences matching the products of skfA and sdpC, genes for the proposed cannibalistic factors sporulation killing factor (SKF) and sporulation delaying protein (SDP), respectively, were identified and the structures of the final products elucidated. SKF is a cyclic 26-amino acid (aa) peptide that is posttranslationally modified with one disulfide and one cysteine thioether bridged to the α-position of a methionine, a posttranslational modification not previously described in biology. SDP is a 42-residue peptide with one disulfide bridge. In spot test assays on solid medium, overproduced SKF and SDP enact a cannibalistic killing effect with SDP having higher potency. However, only purified SDP affected B. subtilis cells in liquid media in fluorescence microscopy and growth assays. Specifically, SDP treatment delayed growth in a concentration-dependent manner, caused increases in cell permeability, and ultimately caused cell lysis accompanied by the production of membrane tubules and spheres. Similarly, SDP but not SKF was able to inhibit the growth of the pathogens Staphylococcus aureus and Staphylococcus epidermidis with comparable IC(50) to vancomycin. This investigation, with the identification of SKF and SDP structures, highlights the strength of IMS in investigations of metabolic exchange of microbial colonies and also demonstrates IMS as a promising approach to discover novel biologically active molecules.


Assuntos
Bacillus subtilis/química , Proteínas de Bactérias/análise , Espectrometria de Massas/métodos , Bacillus subtilis/metabolismo , Estrutura Molecular , Esporos Bacterianos
17.
Annu Rev Microbiol ; 63: 99-118, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19566421

RESUMO

Our view of bacteria, from the earliest observations through the heyday of antibiotic discovery, has shifted dramatically. We recognize communities of bacteria as integral and functionally important components of diverse habitats, ranging from soil collectives to the human microbiome. To function as productive communities, bacteria coordinate metabolic functions, often requiring shifts in growth and development. The hallmark of cellular development, which we characterize as physiological change in response to environmental stimuli, is a defining feature of many bacterial interspecies interactions. Bacterial communities rely on chemical exchanges to provide the cues for developmental change. Traditional methods in microbiology focus on isolation and characterization of bacteria in monoculture, separating the organisms from the surroundings in which interspecies chemical communication has relevance. Developing multispecies experimental systems that incorporate knowledge of bacterial physiology and metabolism with insights from biodiversity and metagenomics shows great promise for understanding interspecies chemical communication in the microbial world.


Assuntos
Bactérias/crescimento & desenvolvimento , Bactérias/metabolismo , Fenômenos Fisiológicos Bacterianos , Percepção de Quorum , Transdução de Sinais , Modelos Biológicos
18.
Chem Biol ; 14(3): 303-12, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17379145

RESUMO

We have developed a phage-display method for high-throughput mining of bacterial gene clusters encoding the natural-product biosynthetic enzymes, polyketide synthases (PKSs) and nonribosomal peptide synthetases (NRPSs). This method uses the phosphopantetheinyl transferase activity of Sfp to specifically biotinylate NRPS and PKS carrier-protein domains expressed from a library of random genome fragments fused to a gene encoding a phage coat protein. Subsequently, the biotinylated phages are enriched through selection on streptavidin-coated plates. Using this method, we isolated phage clones from the multiple NRPS and PKS gene clusters encoded in the genomes of Bacillus subtilis and Myxococcus xanthus. Due to the rapid and unambiguous identification of carrier domains, this method will provide an efficient tool for high-throughput cloning of NRPS and PKS gene clusters from many individual bacterial genomes and multigenome environmental DNA.


Assuntos
Bacillus subtilis/enzimologia , Genes Sintéticos , Biblioteca de Peptídeos , Bacillus subtilis/genética , Proteínas de Bactérias/genética , Família Multigênica , Myxococcus xanthus/enzimologia , Myxococcus xanthus/genética , Peptídeo Sintases/genética , Policetídeo Sintases/genética
19.
Proc Natl Acad Sci U S A ; 104(5): 1506-9, 2007 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17234808

RESUMO

The approximately 80-kb pksX gene cluster in Bacillus subtilis encodes an unusual hybrid polyketide/nonribosomal peptide synthase that has been linked to the production of the uncharacterized antibiotic bacillaene. Multiple copies of this synthase, each similar in size to the ribosome, assemble into a single organelle-like complex with a mass of tens to hundreds of megadaltons. The resource requirements of the assembled megacomplex suggest that bacillaene has an important biological role. By coupling a differential NMR spectroscopic technique with genetically manipulated strains of B. subtilis, we were able to characterize the structure of this unusual secondary metabolite, which could not be predicted by using bioinformatic analysis. We report that bacillaene is a linear molecule with two amide bonds: the first links an alpha-hydroxy carboxylic acid to a omega-amino carboxylic acid containing a conjugated hexaene, and the second links the hexaene-containing carboxylic acid to an (omega-1) amino carboxylic acid containing a conjugated triene. Knowledge of bacillaene's structure has enabled us to annotate the pksX gene cluster and should facilitate the study of bacillaene's biosynthesis as well as its biological role in B. subtilis.


Assuntos
Bacillus subtilis/metabolismo , Biologia Computacional/métodos , Família Multigênica , Peptídeo Sintases/química , Polienos/química , Polienos/metabolismo , Bioensaio , Ácidos Carboxílicos/química , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Modelos Químicos , Peptídeos/química , Espectrofotometria , Estereoisomerismo
20.
Proc Natl Acad Sci U S A ; 104(1): 305-10, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17190806

RESUMO

Nonribosomal peptide synthetases (NRPS), polyketide synthases (PKS), and hybrid NRPS/PKS are of particular interest, because they produce numerous therapeutic agents, have great potential for engineering novel compounds, and are the largest enzymes known. The predicted masses of known enzymatic assembly lines can reach almost 5 megadaltons, dwarfing even the ribosome (approximately 2.6 megadaltons). Despite their uniqueness and importance, little is known about the organization of these enzymes within the native producer cells. Here we report that an 80-kb gene cluster, which occupies approximately 2% of the Bacillus subtilis genome, encodes the subunits of approximately 2.5 megadalton active hybrid NRPS/PKS. Many copies of the NRPS/PKS assemble into a single organelle-like membrane-associated complex of tens to hundreds of megadaltons. Such an enzymatic megacomplex is unprecedented in bacterial subcellular organization and has important implications for engineering novel NRPS/PKSs.


Assuntos
Bacillus subtilis/enzimologia , Família Multigênica , Peptídeo Sintases/genética , Policetídeo Sintases/genética , Sequência de Aminoácidos , Bacillus subtilis/genética , Microscopia Crioeletrônica , Dados de Sequência Molecular , Engenharia de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...