Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
mSystems ; 6(3): e0034121, 2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34156292

ABSTRACT

Planobispora rosea is the natural producer of the potent thiopeptide antibiotic GE2270A. Here, we present the results of a metabolomics and transcriptomics analysis of P. rosea during production of GE2270A. The data generated provides useful insights into the biology of this genetically intractable bacterium. We characterize the details of the shutdown of protein biosynthesis and the respiratory chain associated with the end of the exponential growth phase. We also provide the first description of the phosphate regulon in P. rosea. Based on the transcriptomics data, we show that both phosphate and iron are limiting P. rosea growth in our experimental conditions. Additionally, we identified and validated a new biosynthetic gene cluster associated with the production of the siderophores benarthin and dibenarthin in P. rosea. Together, the metabolomics and transcriptomics data are used to inform and refine the very first genome-scale metabolic model for P. rosea, which will be a valuable framework for the interpretation of future studies of the biology of this interesting but poorly characterized species. IMPORTANCE Planobispora rosea is a genetically intractable bacterium used for the production of GE2270A on an industrial scale. GE2270A is a potent thiopeptide antibiotic currently used as a precursor for the synthesis of two compounds under clinical studies for the treatment of Clostridium difficile infection and acne. Here, we present the very first systematic multi-omics investigation of this important bacterium, which provides a much-needed detailed picture of the dynamics of metabolism of P. rosea while producing GE2270A.

3.
Sci Rep ; 5: 16498, 2015 11 13.
Article in English | MEDLINE | ID: mdl-26563586

ABSTRACT

To date, characterization of ancient oral (dental calculus) and gut (coprolite) microbiota has been primarily accomplished through a metataxonomic approach involving targeted amplification of one or more variable regions in the 16S rRNA gene. Specifically, the V3 region (E. coli 341-534) of this gene has been suggested as an excellent candidate for ancient DNA amplification and microbial community reconstruction. However, in practice this metataxonomic approach often produces highly skewed taxonomic frequency data. In this study, we use non-targeted (shotgun metagenomics) sequencing methods to better understand skewed microbial profiles observed in four ancient dental calculus specimens previously analyzed by amplicon sequencing. Through comparisons of microbial taxonomic counts from paired amplicon (V3 U341F/534R) and shotgun sequencing datasets, we demonstrate that extensive length polymorphisms in the V3 region are a consistent and major cause of differential amplification leading to taxonomic bias in ancient microbiome reconstructions based on amplicon sequencing. We conclude that systematic amplification bias confounds attempts to accurately reconstruct microbiome taxonomic profiles from 16S rRNA V3 amplicon data generated using universal primers. Because in silico analysis indicates that alternative 16S rRNA hypervariable regions will present similar challenges, we advocate for the use of a shotgun metagenomics approach in ancient microbiome reconstructions.


Subject(s)
Gene Amplification , Metagenome/genetics , Metagenomics/methods , Microbiota/genetics , RNA, Ribosomal, 16S/genetics , Archaeology , Bacteria/classification , Bacteria/genetics , Dental Calculus/microbiology , Female , Gastrointestinal Microbiome/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Methanobrevibacter/classification , Methanobrevibacter/genetics , Nucleic Acid Conformation , Phylogeny , RNA, Ribosomal, 16S/chemistry
4.
Am J Pathol ; 185(12): 3290-303, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26458765

ABSTRACT

Type 17 helper T-cell cytokines have been implicated in the pathogenesis of inflammatory bowel disease, a chronic condition affecting the gastrointestinal tract, but information regarding their contribution to pathology in different regions of the gut is lacking. By using a murine model of bacteria-induced typhlocolitis, we investigated the role of IL-17A, IL-17F, and IL-22 in cecal versus colonic inflammation. Cecal, but not colonic, pathology in C57BL/6 mice inoculated with Helicobacter hepaticus plus anti-IL-10 receptor (IL-10R) monoclonal antibody was exacerbated by co-administration of anti-IL-17A monoclonal antibody, suggesting a disease-protective role for IL-17A in the cecum. In contrast, anti-IL-17F had no effect on H. hepaticus-induced intestinal pathology. Neutralization of IL-22 prevented the development of colonic, but not cecal, inflammation in H. hepaticus-infected anti-IL-10R-treated mice, demonstrating a pathogenic role for IL-22 in the colon. Analysis of transcript levels revealed differential expression of IL-22R, IL-22 binding protein, and IL-23R between cecum and colon, a finding that may help explain why these tissues respond differently after anti-IL-22 treatment. Analysis of microarray data from healthy human intestine further revealed significant differences in cytokine receptor transcript levels (including IL-22RA1 and IL-23R) in distinct parts of the human gut. Together, our findings demonstrate that individual type 17 helper T-cell cytokines can have proinflammatory or anti-inflammatory effects in different regions of the intestine, an observation that may have implications for interventions against human inflammatory bowel disease.


Subject(s)
Colitis/microbiology , Helicobacter Infections/immunology , Helicobacter hepaticus , Interleukin-17/immunology , Interleukins/immunology , Typhlitis/microbiology , Animals , Antibodies, Monoclonal/immunology , Colitis/immunology , Colitis/prevention & control , Female , Gene Expression/immunology , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , Interleukins/biosynthesis , Interleukins/genetics , Intestines/immunology , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptors, Cytokine/biosynthesis , Typhlitis/immunology , Interleukin-22
5.
FEBS Lett ; 588(10): 1869-72, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24735724

ABSTRACT

Staphylococcus aureus and Staphylococcus epidermidis cause dangerous and difficult to treat medical device-related infections through their ability to form biofilms. Extracellular poly-N-acetylglucosamine (PNAG) facilitates biofilm formation and is a vaccination target, yet details of its biosynthesis by the icaADBC gene products is limited. IcaC is the proposed transporter for PNAG export, however a comparison of the Ica proteins to homologous exo-polysaccharide synthases suggests that the common IcaAD protein components both synthesise and transport the PNAG. The limited distribution of icaC to the Staphylococcaceae and its membership of a family of membrane-bound acyltransferases, leads us to suggest that IcaC is responsible for the known O-succinylation of PNAG that occurs in staphylococci, identifying a potentially new therapeutic target specific for these bacteria.


Subject(s)
Bacterial Proteins/physiology , Biofilms/growth & development , Staphylococcus aureus/physiology , Staphylococcus epidermidis/physiology , Acetylglucosamine/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biosynthetic Pathways/genetics , Gene Order , Humans , Operon/genetics , Phylogeny , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism
6.
Proc Biol Sci ; 279(1740): 2965-73, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22513857

ABSTRACT

Symbiotic nitrogen recycling enables animals to thrive on nitrogen-poor diets and environments. It traditionally refers to the utilization of animal waste nitrogen by symbiotic micro-organisms to synthesize essential amino acids (EAAs), which are translocated back to the animal host. We applied metabolic modelling and complementary metabolite profiling to investigate nitrogen recycling in the symbiosis between the pea aphid and the intracellular bacterium Buchnera, which synthesizes EAAs. The results differ from traditional notions of nitrogen recycling in two important respects. First, aphid waste ammonia is recycled predominantly by the host cell (bacteriocyte) and not Buchnera. Host cell recycling is mediated by shared biosynthetic pathways for four EAAs, in which aphid transaminases incorporate ammonia-derived nitrogen into carbon skeletons synthesized by Buchnera to generate EAAs. Second, the ammonia substrate for nitrogen recycling is derived from bacteriocyte metabolism, such that the symbiosis is not a sink for nitrogenous waste from other aphid organs. Host cell-mediated nitrogen recycling may be general among insect symbioses with shared EAA biosynthetic pathways generated by the loss of symbiont genes mediating terminal reactions in EAA synthesis.


Subject(s)
Aphids/metabolism , Aphids/microbiology , Buchnera/metabolism , Nitrogen/metabolism , Symbiosis , Amino Acids, Essential/metabolism , Animals , Aphids/cytology , Buchnera/genetics , Buchnera/growth & development , Models, Biological , Pisum sativum/parasitology
7.
BMC Syst Biol ; 3: 24, 2009 Feb 21.
Article in English | MEDLINE | ID: mdl-19232131

ABSTRACT

BACKGROUND: In silico analyses provide valuable insight into the biology of obligately intracellular pathogens and symbionts with small genomes. There is a particular opportunity to apply systems-level tools developed for the model bacterium Escherichia coli to study the evolution and function of symbiotic bacteria which are metabolically specialised to overproduce specific nutrients for their host and, remarkably, have a gene complement that is a subset of the E. coli genome. RESULTS: We have reconstructed and analysed the metabolic network of the gamma-proteobacterium Buchnera aphidicola (symbiont of the pea aphid) as a model for using systems-level approaches to discover key traits of symbionts with small genomes. The metabolic network is extremely fragile with > 90% of the reactions essential for viability in silico; and it is structured so that the bacterium cannot grow without producing the essential amino acid, histidine, which is released to the insect host. Further, the amount of essential amino acid produced by the bacterium in silico can be controlled by host supply of carbon and nitrogen substrates. CONCLUSION: This systems-level analysis predicts that the fragility of the bacterial metabolic network renders the symbiotic bacterium intolerant of drastic environmental fluctuations, whilst the coupling of histidine production to growth prevents the bacterium from exploiting host nutrients without reciprocating. These metabolic traits underpin the sustained nutritional contribution of B. aphidicola to the host and, together with the impact of host-derived substrates on the profile of nutrients released from the bacteria, point to a dominant role of the host in controlling the symbiosis.


Subject(s)
Buchnera/metabolism , Metabolic Networks and Pathways , Models, Biological , Amino Acids, Essential/biosynthesis , Buchnera/genetics , Buchnera/growth & development , Computer Simulation , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Genes, Bacterial , Histidine/biosynthesis , Metabolic Networks and Pathways/genetics , Purines/biosynthesis , Species Specificity , Symbiosis/genetics , Symbiosis/physiology , Systems Biology
SELECTION OF CITATIONS
SEARCH DETAIL
...