Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Mol Microbiol ; 111(6): 1592-1603, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30875449

RESUMO

Nitrate is available to microbes in many environments due to sustained use of inorganic fertilizers on agricultural soils and many bacterial and archaeal lineages have the capacity to express respiratory (Nar) and assimilatory (Nas) nitrate reductases to utilize this abundant respiratory substrate and nutrient for growth. Here, we show that in the denitrifying bacterium Paracoccus denitrificans, NarJ serves as a chaperone for both the anaerobic respiratory nitrate reductase (NarG) and the assimilatory nitrate reductase (NasC), the latter of which is active during both aerobic and anaerobic nitrate assimilation. Bioinformatic analysis suggests that the potential for this previously unrecognized role for NarJ in functional maturation of other cytoplasmic molybdenum-dependent nitrate reductases may be phylogenetically widespread as many bacteria contain both Nar and Nas systems.


Assuntos
Proteínas de Bactérias/metabolismo , Nitrato Redutase/metabolismo , Nitratos/metabolismo , Paracoccus denitrificans/enzimologia , Aerobiose , Anaerobiose , Proteínas de Bactérias/genética , Chaperonas Moleculares/metabolismo , Molibdênio/metabolismo , Nitrato Redutase/genética , Oxirredução , Paracoccus denitrificans/genética
2.
J Immunol ; 198(1): 335-343, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27895168

RESUMO

During Salmonella Typhimurium infection, intestinal CX3CR1+ cells can either extend transepithelial cellular processes to sample luminal bacteria or, very early after infection, migrate into the intestinal lumen to capture bacteria. However, until now, the biological relevance of the intraluminal migration of CX3CR1+ cells remained to be determined. We addressed this by using a combination of mouse strains differing in their ability to carry out CX3CR1-mediated sampling and intraluminal migration. We observed that the number of S. Typhimurium traversing the epithelium did not differ between sampling-competent/migration-competent C57BL/6 and sampling-deficient/migration-competent BALB/c mice. In contrast, in sampling-deficient/migration-deficient CX3CR1-/- mice the numbers of S. Typhimurium penetrating the epithelium were significantly higher. However, in these mice the number of invading S. Typhimurium was significantly reduced after the adoptive transfer of CX3CR1+ cells directly into the intestinal lumen, consistent with intraluminal CX3CR1+ cells preventing S. Typhimurium from infecting the host. This interpretation was also supported by a higher bacterial fecal load in CX3CR1+/gfp compared with CX3CR1gfp/gfp mice following oral infection. Furthermore, by using real-time in vivo imaging we observed that CX3CR1+ cells migrated into the lumen moving through paracellular channels within the epithelium. Also, we reported that the absence of CX3CR1-mediated sampling did not affect Ab responses to a noninvasive S. Typhimurium strain that specifically targeted the CX3CR1-mediated entry route. These data showed that the rapidly deployed CX3CR1+ cell-based mechanism of immune exclusion is a defense mechanism against pathogens that complements the mucous and secretory IgA Ab-mediated system in the protection of intestinal mucosal surface.


Assuntos
Mucosa Intestinal/imunologia , Salmonelose Animal/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Receptor 1 de Quimiocina CX3C , Movimento Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imunofluorescência , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Receptores de Quimiocinas/imunologia , Receptores de Quimiocinas/metabolismo , Salmonella typhimurium/imunologia
3.
Biochem J ; 474(11): 1769-1787, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28385879

RESUMO

Transcriptional adaptation to nitrate-dependent anabolism by Paracoccus denitrificans PD1222 was studied. A total of 74 genes were induced in cells grown with nitrate as N-source compared with ammonium, including nasTSABGHC and ntrBC genes. The nasT and nasS genes were cotranscribed, although nasT was more strongly induced by nitrate than nasS The nasABGHC genes constituted a transcriptional unit, which is preceded by a non-coding region containing hairpin structures involved in transcription termination. The nasTS and nasABGHC transcripts were detected at similar levels with nitrate or glutamate as N-source, but nasABGHC transcript was undetectable in ammonium-grown cells. The nitrite reductase NasG subunit was detected by two-dimensional polyacrylamide gel electrophoresis in cytoplasmic fractions from nitrate-grown cells, but it was not observed when either ammonium or glutamate was used as the N-source. The nasT mutant lacked both nasABGHC transcript and nicotinamide adenine dinucleotide (NADH)-dependent nitrate reductase activity. On the contrary, the nasS mutant showed similar levels of the nasABGHC transcript to the wild-type strain and displayed NasG protein and NADH-nitrate reductase activity with all N-sources tested, except with ammonium. Ammonium repression of nasABGHC was dependent on the Ntr system. The ntrBC and ntrYX genes were expressed at low levels regardless of the nitrogen source supporting growth. Mutational analysis of the ntrBCYX genes indicated that while ntrBC genes are required for nitrate assimilation, ntrYX genes can only partially restore growth on nitrate in the absence of ntrBC genes. The existence of a regulation mechanism for nitrate assimilation in P. denitrificans, by which nitrate induction operates at both transcriptional and translational levels, is proposed.


Assuntos
Adaptação Fisiológica , Regulação Bacteriana da Expressão Gênica , Modelos Biológicos , Nitratos/metabolismo , Ciclo do Nitrogênio , Paracoccus denitrificans/fisiologia , Compostos de Amônio/metabolismo , Proteínas de Bactérias/agonistas , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Metabolismo Energético , Perfilação da Expressão Gênica , Ácido Glutâmico/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Nitrato Redutase (NADH)/antagonistas & inibidores , Nitrato Redutase (NADH)/química , Nitrato Redutase (NADH)/genética , Nitrato Redutase (NADH)/metabolismo , Paracoccus denitrificans/enzimologia , Paracoccus denitrificans/crescimento & desenvolvimento , Proteômica/métodos , RNA Bacteriano/metabolismo , RNA Mensageiro/metabolismo , Elementos Reguladores de Transcrição , Proteínas Repressoras/agonistas , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transativadores/agonistas , Transativadores/antagonistas & inibidores , Transativadores/genética , Transativadores/metabolismo
4.
Environ Microbiol ; 19(12): 4953-4964, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29076595

RESUMO

Bacterial denitrification is a respiratory process that is a major source and sink of the potent greenhouse gas nitrous oxide. Many denitrifying bacteria can adjust to life in both oxic and anoxic environments through differential expression of their respiromes in response to environmental signals such as oxygen, nitrate and nitric oxide. We used steady-state oxic and anoxic chemostat cultures to demonstrate that the switch from aerobic to anaerobic metabolism is brought about by changes in the levels of expression of relatively few genes, but this is sufficient to adjust the configuration of the respirome to allow the organism to efficiently respire nitrate without the significant release of intermediates, such as nitrous oxide. The regulation of the denitrification respirome in strains deficient in the transcription factors FnrP, Nnr and NarR was explored and revealed that these have both inducer and repressor activities, possibly due to competitive binding at similar DNA binding sites. This may contribute to the fine tuning of expression of the denitrification respirome and so adds to the understanding of the regulation of nitrous oxide emission by denitrifying bacteria in response to different environmental signals.


Assuntos
Anaerobiose/fisiologia , Respiração Celular/fisiologia , Desnitrificação/fisiologia , Óxido Nítrico/metabolismo , Óxido Nitroso/metabolismo , Oxigênio/metabolismo , Paracoccus denitrificans/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Respiração Celular/genética , Desnitrificação/genética , Nitratos/metabolismo , Oxirredutases/genética , Paracoccus denitrificans/genética , Fatores de Transcrição/genética
5.
Proc Natl Acad Sci U S A ; 110(49): 19926-31, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24248380

RESUMO

Global agricultural emissions of the greenhouse gas nitrous oxide (N2O) have increased by around 20% over the last 100 y, but regulation of these emissions and their impact on bacterial cellular metabolism are poorly understood. Denitrifying bacteria convert nitrate in soils to inert di-nitrogen gas (N2) via N2O and the biochemistry of this process has been studied extensively in Paracoccus denitrificans. Here we demonstrate that expression of the gene encoding the nitrous oxide reductase (NosZ), which converts N2O to N2, is regulated in response to the extracellular copper concentration. We show that elevated levels of N2O released as a consequence of decreased cellular NosZ activity lead to the bacterium switching from vitamin B12-dependent to vitamin B12-independent biosynthetic pathways, through the transcriptional modulation of genes controlled by vitamin B12 riboswitches. This inhibitory effect of N2O can be rescued by addition of exogenous vitamin B12.


Assuntos
Cobre/farmacologia , Fertilizantes/análise , Regulação Bacteriana da Expressão Gênica/fisiologia , Óxido Nitroso/metabolismo , Oxirredutases/metabolismo , Paracoccus denitrificans/metabolismo , Vitamina B 12/metabolismo , Agricultura/métodos , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Aquecimento Global , Análise em Microsséries , Óxido Nitroso/toxicidade , Oxirredutases/genética , Paracoccus denitrificans/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Riboswitch/fisiologia , Vitamina B 12/genética
6.
Mol Microbiol ; 87(3): 526-38, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23190111

RESUMO

OmpR is a multifunctional DNA binding regulator with orthologues in many enteric bacteria that exhibits classical regulator activity as well as nucleoid-associated protein-like characteristics. In the enteric pathogen Salmonella enterica, using chromatin immunoprecipitation of OmpR:FLAG and nucleotide sequencing, 43 putative OmpR binding sites were identified in S. enterica serovar Typhi, 22 of which were associated with OmpR-regulated genes. Mutation of a sequence motif (TGTWACAW) that was associated with the putative OmpR binding sites abrogated binding of OmpR:6×His to the tviA upstream region. A core set of 31 orthologous genes were found to exhibit OmpR-dependent expression in both S. Typhi and S. Typhimurium. S. Typhimurium-encoded orthologues of two divergently transcribed OmpR-regulated operons (SL1068-71 and SL1066-67) had a putative OmpR binding site in the inter-operon region in S. Typhi, and were characterized using in vitro and in vivo assays. These operons are widely distributed within S. enterica but absent from the closely related Escherichia coli. SL1066 and SL1067 were required for growth on N-acetylmuramic acid as a sole carbon source. SL1068-71 exhibited sequence similarity to sialic acid uptake systems and contributed to colonization of the ileum and caecum in the streptomycin-pretreated mouse model of colitis.


Assuntos
Perfilação da Expressão Gênica , Regulon , Salmonella typhi/genética , Salmonella typhi/patogenicidade , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidade , Transativadores/metabolismo , Animais , Sítios de Ligação , Ceco/microbiologia , Imunoprecipitação da Cromatina , Colite/microbiologia , DNA Bacteriano/química , DNA Bacteriano/genética , Modelos Animais de Doenças , Íleo/microbiologia , Camundongos , Salmonelose Animal/microbiologia , Análise de Sequência de DNA , Transativadores/genética , Virulência
7.
Adv Appl Microbiol ; 83: 87-144, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23651595

RESUMO

The ability to sense and respond to the environment is essential for the survival of all living organisms. Bacterial pathogens such as Salmonella enterica are of particular interest due to their ability to sense and adapt to the diverse range of conditions they encounter, both in vivo and in environmental reservoirs. During this cycling from host to non-host environments, Salmonella encounter a variety of environmental insults ranging from temperature fluctuations, nutrient availability and changes in osmolarity, to the presence of antimicrobial peptides and reactive oxygen/nitrogen species. Such fluctuating conditions impact on various areas of bacterial physiology including virulence, growth and antimicrobial resistance. A key component of the success of any bacterial pathogen is the ability to recognize and mount a suitable response to the discrete chemical and physical stresses elicited by the host. Such responses occur through a coordinated and complex programme of gene expression and protein activity, involving a range of transcriptional regulators, sigma factors and two component regulatory systems. This review briefly outlines the various stresses encountered throughout the Salmonella life cycle and the repertoire of regulatory responses with which Salmonella counters. In particular, how these Gram-negative bacteria are able to alleviate disruption in periplasmic envelope homeostasis through a group of stress responses, known collectively as the Envelope Stress Responses, alongside the mechanisms used to overcome nitrosative stress, will be examined in more detail.


Assuntos
Salmonella enterica , Salmonella typhimurium , Fenômenos Fisiológicos Bacterianos , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Fator sigma , Virulência
8.
Biochem J ; 442(1): 85-93, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22084975

RESUMO

The bacterial envelope is the interface with the surrounding environment and is consequently subjected to a barrage of noxious agents including a range of compounds with antimicrobial activity. The ESR (envelope stress response) pathways of enteric bacteria are critical for maintenance of the envelope against these antimicrobial agents. In the present study, we demonstrate that the periplasmic protein ZraP contributes to envelope homoeostasis and assign both chaperone and regulatory function to ZraP from Salmonella Typhimurium. The ZraP chaperone mechanism is catalytic and independent of ATP; the chaperone activity is dependent on the presence of zinc, which is shown to be responsible for the stabilization of an oligomeric ZraP complex. Furthermore, ZraP can act to repress the two-component regulatory system ZraSR, which itself is responsive to zinc concentrations. Through structural homology, ZraP is a member of the bacterial CpxP family of periplasmic proteins, which also consists of CpxP and Spy. We demonstrate environmental co-expression of the CpxP family and identify an important role for these proteins in Salmonella's defence against the cationic antimicrobial peptide polymyxin B.


Assuntos
Proteínas de Escherichia coli/genética , Chaperonas Moleculares/metabolismo , Proteínas Periplásmicas/metabolismo , Proteínas Repressoras/metabolismo , Salmonella typhimurium/genética , Cristalografia por Raios X , Farmacorresistência Bacteriana/fisiologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/biossíntese , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Indóis/farmacologia , Proteínas de Membrana/biossíntese , Periplasma/efeitos dos fármacos , Periplasma/metabolismo , Proteínas Periplásmicas/biossíntese , Polimixina B/farmacologia , Salmonella typhimurium/metabolismo , Zinco/metabolismo
9.
Biochem J ; 441(2): 755-62, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22039967

RESUMO

The production of cytotoxic nitric oxide (NO) and conversion into the neuropharmacological agent and potent greenhouse gas nitrous oxide (N2O) is linked with anoxic nitrate catabolism by Salmonella enterica serovar Typhimurium. Salmonella can synthesize two types of nitrate reductase: a membrane-bound form (Nar) and a periplasmic form (Nap). Nitrate catabolism was studied under nitrate-rich and nitrate-limited conditions in chemostat cultures following transition from oxic to anoxic conditions. Intracellular NO production was reported qualitatively by assessing transcription of the NO-regulated genes encoding flavohaemoglobin (Hmp), flavorubredoxin (NorV) and hybrid cluster protein (Hcp). A more quantitative analysis of the extent of NO formation was gained by measuring production of N2O, the end-product of anoxic NO-detoxification. Under nitrate-rich conditions, the nar, nap, hmp, norV and hcp genes were all induced following transition from the oxic to anoxic state, and 20% of nitrate consumed in steady-state was released as N2O when nitrite had accumulated to millimolar levels. The kinetics of nitrate consumption, nitrite accumulation and N2O production were similar to those of wild-type in nitrate-sufficient cultures of a nap mutant. In contrast, in a narG mutant, the steady-state rate of N2O production was ~30-fold lower than that of the wild-type. Under nitrate-limited conditions, nap, but not nar, was up-regulated following transition from oxic to anoxic metabolism and very little N2O production was observed. Thus a combination of nitrate-sufficiency, nitrite accumulation and an active Nar-type nitrate reductase leads to NO and thence N2O production, and this can account for up to 20% of the nitrate catabolized.


Assuntos
Membrana Celular/enzimologia , Nitrato Redutases/metabolismo , Nitrito Redutases/metabolismo , Óxido Nitroso/metabolismo , Periplasma/enzimologia , Salmonella typhimurium/enzimologia , Aerobiose , Anaerobiose , Hipóxia Celular , Regulação Bacteriana da Expressão Gênica , Nitratos/metabolismo , Nitritos/metabolismo , Salmonella typhimurium/metabolismo
10.
Environ Microbiol ; 14(7): 1788-800, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22642644

RESUMO

Denitrifying bacteria convert nitrate (NO(3) (-) ) to dinitrogen (N(2) ) gas through an anaerobic respiratory process in which the potent greenhouse gas nitrous oxide (N(2) O) is a free intermediate. These bacteria can be grouped into classes that synthesize a nitrite (NO(2) (-) ) reductase (Nir) that is solely dependent on haem-iron as a cofactor (e.g. Paracoccus denitrificans) or a Nir that is solely dependent on copper (Cu) as a cofactor (e.g. Achromobacter xylosoxidans). Regardless of which form of Nir these groups synthesize, they are both dependent on a Cu-containing nitrous oxide reductase (NosZ) for the conversion of N(2) O to N(2) . Agriculture makes a major contribution to N(2) O release and it is recognized that a number of agricultural lands are becoming Cu-limited but are N-rich because of fertilizer addition. Here we utilize continuous cultures to explore the denitrification phenotypes of P. denitrificans and A. xylosoxidans at a range of extracellular NO(3) (-) , organic carbon and Cu concentrations. Quite distinct phenotypes are observed between the two species. Notably, P. denitrificans emits approximately 40% of NO(3) (-) consumed as N(2) O under NO(3) (-) -rich Cu-deficient conditions, while under the same conditions A. xylosoxidans releases approximately 40% of the NO(3) (-) consumed as NO(2) (-) . However, the denitrification phenotypes are very similar under NO(3) (-) -limited conditions where denitrification intermediates do not accumulate significantly. The results have potential implications for understanding denitrification flux in a range of agricultural environments.


Assuntos
Achromobacter denitrificans/metabolismo , Cobre/metabolismo , Desnitrificação , Óxido Nitroso/metabolismo , Paracoccus denitrificans/metabolismo , Achromobacter denitrificans/genética , Carbono/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Oxirredutases/metabolismo , Paracoccus denitrificans/genética , Fenótipo
11.
Microbiology (Reading) ; 157(Pt 3): 848-858, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21148205

RESUMO

The alternative sigma factor σ(E) (rpoE) is essential for survival in vivo of Salmonella Typhimurium but is dispensable during growth in the laboratory. We have been identifying σ(E)-regulated genes and studying their regulation and function to elucidate their potential role in the severe attenuation of S. Typhimurium rpoE mutants. In this study we identify five promoters that control the rseP, yaeT (bamA), skp region. A confirmed σ(E)-dependent promoter, yaeTp1, and a second downstream promoter, yaeTp2, are located within the upstream gene rseP and direct expression of the downstream genes. The only known function of RseP is σ(E) activation, and it is therefore not expected to be essential for S. Typhimurium in vitro. However, it proved impossible to delete the entire rseP gene due to the presence of internal promoters that regulate the essential gene yaeT. We could inactivate rseP by deleting the first third of the gene, leaving the yaeT promoters intact. Like the rpoE mutant, the rseP mutant exhibited severe attenuation in vivo. We were able to delete the entire coding sequence of skp, which encodes a periplasmic chaperone involved in targeting misfolded outer-membrane proteins to the ß-barrel assembly machinery. The skp mutant was attenuated in mice after oral and parenteral infection. Virulence could be complemented by providing skp in trans but only by linking it to a heterologous σ(E)-regulated promoter. The reason the skp mutant is attenuated is currently enigmatic, but we know it is not through increased sensitivity to a variety of RpoE-activating host stresses, such as H(2)O(2), polymyxin B and high temperature, or through altered secretion of effector proteins by either the Salmonella pathogenicity island (SPI)-1 or the SPI-2 type III secretion system.


Assuntos
Regulação Bacteriana da Expressão Gênica , Chaperonas Moleculares/metabolismo , Periplasma/metabolismo , Salmonella typhimurium/patogenicidade , Febre Tifoide/patologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Feminino , Proteínas Fúngicas/metabolismo , Resposta ao Choque Térmico , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Chaperonas Moleculares/genética , Mutação , Regiões Promotoras Genéticas , Regulon , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Fator sigma/genética , Fator sigma/metabolismo , Endonucleases Específicas para DNA e RNA de Cadeia Simples/metabolismo , Transcrição Gênica , Febre Tifoide/microbiologia , Virulência
12.
Biochem Soc Trans ; 39(6): 1876-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22103543

RESUMO

The nitrogen cycle is based on several redox reactions that are mainly accomplished by prokaryotic organisms, some archaea and a few eukaryotes, which use these reactions for assimilatory, dissimilatory or respiratory purposes. One group is the Enterobacteriaceae family of Gammaproteobacteria, which have their natural habitats in soil, marine environments or the intestines of humans and other warm-blooded animals. Some of the genera are pathogenic and usually associated with intestinal infections. Our body possesses several physical and chemical defence mechanisms to prevent pathogenic enteric bacteria from invading the gastrointestinal tract. One response of the innate immune system is to activate macrophages, which produce the potent cytotoxin nitric oxide (NO). However, some pathogens have evolved the ability to detoxify NO to less toxic compounds, such as the neuropharmacological agent and greenhouse gas nitrous oxide (N2O), which enables them to overcome the host's attack. The same mechanisms may be used by bacteria producing NO endogenously as a by-product of anaerobic nitrate respiration. In the present review, we provide a brief introduction into the NO detoxification mechanisms of two members of the Enterobacteriaceae family: Escherichia coli and Salmonella enterica serovar Typhimurium. These are discussed as comparative non-pathogenic and pathogenic model systems in order to investigate the importance of detoxifying NO and producing N2O for the pathogenicity of enteric bacteria.


Assuntos
Citotoxinas/biossíntese , Citotoxinas/metabolismo , Enterobacteriaceae/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Saúde , Humanos , Inativação Metabólica , Óxido Nitroso/metabolismo
13.
Front Microbiol ; 12: 698286, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34408730

RESUMO

Metagenomics and metatranscriptomics are powerful methods to uncover key micro-organisms and processes driving biogeochemical cycling in natural ecosystems. Databases dedicated to depicting biogeochemical pathways (for example, metabolism of dimethylsulfoniopropionate (DMSP), which is an abundant organosulfur compound) from metagenomic/metatranscriptomic data are rarely seen. Additionally, a recognized normalization model to estimate the relative abundance and environmental importance of pathways from metagenomic and metatranscriptomic data has not been organized to date. These limitations impact the ability to accurately relate key microbial-driven biogeochemical processes to differences in environmental conditions. Thus, an easy-to-use, specialized tool that infers and visually compares the potential for biogeochemical processes, including DMSP cycling, is urgently required. To solve these issues, we developed DiTing, a tool wrapper to infer and compare biogeochemical pathways among a set of given metagenomic or metatranscriptomic reads in one step, based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) and a manually created DMSP cycling gene database. Accurate and specific formulae for over 100 pathways were developed to calculate their relative abundance. Output reports detail the relative abundance of biogeochemical pathways in both text and graphical format. DiTing was applied to simulated metagenomic data and resulted in consistent genetic features of simulated benchmark genomic data. Subsequently, when applied to natural metagenomic and metatranscriptomic data from hydrothermal vents and the Tara Ocean project, the functional profiles predicted by DiTing were correlated with environmental condition changes. DiTing can now be confidently applied to wider metagenomic and metatranscriptomic datasets, and it is available at https://github.com/xuechunxu/DiTing.

14.
Microorganisms ; 8(9)2020 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-32867361

RESUMO

The Earth's oceans are a huge body of water with physicochemical properties and microbial community profiles that change with depth, which in turn influences their biogeochemical cycling potential. The differences between microbial communities and their functional potential in surface to hadopelagic water samples are only beginning to be explored. Here, we used metagenomics to investigate the microbial communities and their potential to drive biogeochemical cycling in seven different water layers down the vertical profile of the Challenger Deep (0-10,500 m) in the Mariana Trench, the deepest natural point in the Earth's oceans. We recovered 726 metagenome-assembled genomes (MAGs) affiliated to 27 phyla. Overall, biodiversity increased in line with increased depth. In addition, the genome size of MAGs at ≥4000 m layers was slightly larger compared to those at 0-2000 m. As expected, surface waters were the main source of primary production, predominantly from Cyanobacteria. Intriguingly, microbes conducting an unusual form of nitrogen metabolism were identified in the deepest waters (>10,000 m), as demonstrated by an enrichment of genes encoding proteins involved in dissimilatory nitrate to ammonia conversion (DNRA), nitrogen fixation and urea transport. These likely facilitate the survival of ammonia-oxidizing archaea α lineage, which are typically present in environments with a high ammonia concentration. In addition, the microbial potential for oxidative phosphorylation and the glyoxylate shunt was enhanced in >10,000 m waters. This study provides novel insights into how microbial communities and their genetic potential for biogeochemical cycling differs through the Challenger deep water column, and into the unique adaptive lifestyle of microbes in the Earth's deepest seawater.

15.
Infect Immun ; 77(7): 3117-26, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19380470

RESUMO

Salmonella is a widespread zoonotic enteropathogen that causes gastroenteritis and fatal typhoidal disease in mammals. During systemic infection of mice, Salmonella enterica serovar Typhimurium resides and replicates in macrophages within the "Salmonella-containing vacuole" (SCV). It is surprising that the substrates and metabolic pathways necessary for growth of S. Typhimurium within the SCV of macrophages have not been identified yet. To determine whether S. Typhimurium utilized sugars within the SCV, we constructed a series of S. Typhimurium mutants that lacked genes involved in sugar transport and catabolism and tested them for replication in mice and macrophages. These mutants included a mutant with a mutation in the pfkAB-encoded phosphofructokinase, which catalyzes a key committing step in glycolysis. We discovered that a pfkAB mutant is severely attenuated for replication and survival within RAW 264.7 macrophages. We also show that disruption of the phosphoenolpyruvate:carbohydrate phosphotransferase system by deletion of the ptsHI and crr genes reduces S. Typhimurium replication within RAW 264.7 macrophages. We discovered that mutants unable to catabolize glucose due to deletion of ptsHI, crr, and glk or deletion of ptsG, manXYZ, and glk showed reduced replication within RAW 264.7 macrophages. This study proves that S. Typhimurium requires glycolysis for infection of mice and macrophages and that transport of glucose is required for replication within macrophages.


Assuntos
Glucose/metabolismo , Glicólise , Macrófagos/microbiologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Animais , Linhagem Celular , Contagem de Colônia Microbiana , Feminino , Deleção de Genes , Genes Bacterianos , Fígado/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Salmonelose Animal , Salmonella typhimurium/genética , Baço/microbiologia , Virulência
16.
Nat Rev Microbiol ; 4(5): 383-94, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16715050

RESUMO

Despite being nutrient rich, the tissues and fluids of vertebrates are hostile to microorganisms, and most bacteria that attempt to take advantage of this environment are rapidly eliminated by host defences. Pathogens have evolved various means to promote their survival in host tissues, including stress responses that enable bacteria to sense and adapt to adverse conditions. Many different stress responses have been described, some of which are responsive to one or a small number of cues, whereas others are activated by a broad range of insults. The surface layers of pathogenic bacteria directly interface with the host and can bear the brunt of the attack by the host armoury. Several stress systems that respond to perturbations in the microbial cell outside of the cytoplasm have been described and are known collectively as extracytoplasmic or envelope stress responses (ESRs). Here, we review the role of the ESRs in the pathogenesis of Gram-negative bacterial pathogens.


Assuntos
Adaptação Fisiológica , Proteínas de Bactérias/fisiologia , Bactérias Gram-Negativas/fisiologia , Proteínas de Membrana/fisiologia , Transdução de Sinais/fisiologia , Animais , Regulação Bacteriana da Expressão Gênica , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/patogenicidade , Infecções por Bactérias Gram-Negativas/microbiologia , Humanos
17.
Artigo em Inglês | MEDLINE | ID: mdl-31552196

RESUMO

The Gram-negative bacterial envelope is an essential interface between the intracellular and harsh extracellular environment. Envelope stress responses (ESRs) are crucial to the maintenance of this barrier and function to detect and respond to perturbations in the envelope, caused by environmental stresses. Pathogenic bacteria are exposed to an array of challenging and stressful conditions during their lifecycle and, in particular, during infection of a host. As such, maintenance of envelope homeostasis is essential to their ability to successfully cause infection. This review will discuss our current understanding of the σE- and Cpx-regulated ESRs, with a specific focus on their role in the virulence of a number of model pathogens.


Assuntos
Membrana Celular/fisiologia , Parede Celular/fisiologia , Bactérias Gram-Negativas/fisiologia , Estresse Fisiológico , Adaptação Fisiológica , Regulação Bacteriana da Expressão Gênica , Redes Reguladoras de Genes , Bactérias Gram-Negativas/genética
18.
Artigo em Inglês | MEDLINE | ID: mdl-31396489

RESUMO

The host presents an array of environments which induce bacterial stress including changes in pH, antimicrobial compounds and reactive oxygen species. The bacterial envelope sits at the interface between the intracellular and extracellular environment and its maintenance is essential for Salmonella cell viability under a range of conditions, including during infection. In this study, we aimed to understand the contribution of the σH- and σE-regulated small heat shock proteins IbpA, IbpB, and AgsA and the putative σE-regulated stress response protein STM1250 to the Salmonella envelope stress response. Due to shared sequence identity, regulatory overlap, and the specificity of STM1250 and AgsA to Salmonella sp., we hypothesized that functional overlap exists between these four stress response proteins, which might afford a selective advantage during Salmonella exposure to stress. We present here new roles for three small heat shock proteins and a putative stress response protein in Salmonella that are not limited to heat shock. We have shown that, compared to WT, a quadruple mutant is significantly more sensitive to hydrogen peroxide, has a lower minimum bactericidal concentration to the cationic antimicrobial peptide polymyxin B, and is attenuated in macrophages.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Macrófagos/imunologia , Viabilidade Microbiana , Estresse Oxidativo , Salmonella typhimurium/imunologia , Estresse Fisiológico , Animais , Proteínas de Bactérias/genética , Deleção de Genes , Redes Reguladoras de Genes , Proteínas de Choque Térmico/deficiência , Proteínas de Choque Térmico/metabolismo , Macrófagos/microbiologia , Camundongos , Modelos Biológicos , Células RAW 264.7 , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/fisiologia , Fator sigma/deficiência , Fator sigma/metabolismo , Virulência
19.
mBio ; 10(4)2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31387903

RESUMO

Global atmospheric loading of the climate-active gas nitrous oxide (N2O) continues to increase. A significant proportion of anthropogenic N2O emissions arises from microbial transformation of nitrogen-based fertilizers during denitrification, making microbial N2O emissions a key target for greenhouse gas reduction strategies. The genetic, physiological, and environmental regulation of microbially mediated N2O flux is poorly understood and therefore represents a critical knowledge gap in the development of successful mitigation approaches. We have previously mapped the transcriptional landscape of the model soil-denitrifying bacterium Paracoccus denitrificans Here, we show that a single bacterial small RNA (sRNA) can control the denitrification rate of P. denitrificans by stalling denitrification at nitrite reduction to limit production of downstream pathway intermediates and N2O emissions. Overexpression of sRNA-29 downregulates nitrite reductase and limits NO and N2O production by cells. RNA sequencing (RNA-seq) analysis revealed 53 genes that are controlled by sRNA-29, one of which is a previously uncharacterized GntR-type transcriptional regulator. Overexpression of this regulator phenocopies sRNA-29 overexpression and allows us to propose a model whereby sRNA-29 enhances levels of the regulator to repress denitrification under appropriate conditions. Our identification of a new regulatory pathway controlling the core denitrification pathway in bacteria highlights the current chasm in knowledge regarding genetic regulation of this pivotal biogeochemical process, which needs to be closed to support future biological and chemical N2O mitigation strategies.IMPORTANCE N2O is an important greenhouse gas and a major cause of ozone depletion. Denitrifying bacteria play vital roles in the production and consumption of N2O in many environments. Complete denitrification consists of the conversion of a soluble N-oxyanion, nitrate (NO3-), to an inert gaseous N-oxide, dinitrogen (N2). Incomplete denitrification can occur if conditions are prohibitive, for example, under conditions of low soil copper concentrations, leading to emission of N2O rather than N2 Although enzymatically well characterized, the genetic drivers that regulate denitrification in response to environmental and physiological cues are not fully understood. This study identified a new regulatory sRNA-based control mechanism for denitrification in the model denitrifying bacterium P. denitrificans Overexpression of this sRNA slows the rate of denitrification. This report highlights that there are gaps in understanding the regulation of this important pathway which need to be filled if strategies for N2O mitigation can be rationally and carefully developed.


Assuntos
Regulação Bacteriana da Expressão Gênica , Óxido Nitroso/metabolismo , Paracoccus denitrificans/genética , Paracoccus denitrificans/metabolismo , RNA Bacteriano/genética , Desnitrificação , Óxido Nítrico/metabolismo , RNA Bacteriano/metabolismo , Microbiologia do Solo
20.
PLoS Pathog ; 2(8): e81, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16933988

RESUMO

Histone-like nucleoid structuring protein (H-NS) is a modular protein that is associated with the bacterial nucleoid. We used chromatin immunoprecipitation to determine the binding sites of H-NS and RNA polymerase on the Salmonella enterica serovar Typhimurium chromosome. We found that H-NS does not bind to actively transcribed genes and does not co-localize with RNA polymerase. This shows that H-NS principally silences gene expression by restricting the access of RNA polymerase to the DNA. H-NS had previously been shown to preferentially bind to curved DNA in vitro. In fact, at the genomic level we discovered that the level of H-NS binding correlates better with the AT-content of DNA. This is likely to have evolutionary consequences because we show that H-NS binds to many Salmonella genes acquired by lateral gene transfer, and functions as a gene silencer. The removal of H-NS from the cell causes un-controlled expression of several Salmonella pathogenicity islands, and we demonstrate that this has deleterious consequences for bacterial fitness. Our discovery of this novel role for H-NS may have implications for the acquisition of foreign genes by enteric bacteria.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Regulação Bacteriana da Expressão Gênica , Inativação Gênica , Transferência Genética Horizontal , Salmonella enterica/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Evolução Biológica , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Salmonella enterica/metabolismo , Salmonella enterica/patogenicidade
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa