Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Rev Genet ; 18(11): 690-699, 2017 11.
Article in English | MEDLINE | ID: mdl-28824167

ABSTRACT

Taxonomic and functional changes to the composition of the gut microbiome have been implicated in multiple human diseases. Recent microbiome genome-wide association studies reveal that variants in many human genes involved in immunity and gut architecture are associated with an altered composition of the gut microbiome. Although many factors can affect the microbial organisms residing in the gut, a number of recent findings support the hypothesis that certain host genetic variants predispose an individual towards microbiome dysbiosis. This condition, in which the normal microbiome population structure is disturbed, is a key feature in disorders of metabolism and immunity.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Genetic Variation , Dysbiosis/genetics , Dysbiosis/microbiology , Gastrointestinal Microbiome/genetics , Humans
2.
Nucleic Acids Res ; 49(19): e113, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34417598

ABSTRACT

DNA methylation is widespread amongst eukaryotes and prokaryotes to modulate gene expression and confer viral resistance. 5-Methylcytosine (m5C) methylation has been described in genomes of a large fraction of bacterial species as part of restriction-modification systems, each composed of a methyltransferase and cognate restriction enzyme. Methylases are site-specific and target sequences vary across organisms. High-throughput methods, such as bisulfite-sequencing can identify m5C at base resolution but require specialized library preparations and single molecule, real-time (SMRT) sequencing usually misses m5C. Here, we present a new method called RIMS-seq (rapid identification of methylase specificity) to simultaneously sequence bacterial genomes and determine m5C methylase specificities using a simple experimental protocol that closely resembles the DNA-seq protocol for Illumina. Importantly, the resulting sequencing quality is identical to DNA-seq, enabling RIMS-seq to substitute standard sequencing of bacterial genomes. Applied to bacteria and synthetic mixed communities, RIMS-seq reveals new methylase specificities, supporting routine study of m5C methylation while sequencing new genomes.


Subject(s)
5-Methylcytosine/metabolism , DNA Modification Methylases/metabolism , DNA Restriction Enzymes/metabolism , Escherichia coli K12/genetics , Genome, Bacterial , High-Throughput Nucleotide Sequencing/methods , Acinetobacter calcoaceticus/enzymology , Acinetobacter calcoaceticus/genetics , Aeromonas hydrophila/enzymology , Aeromonas hydrophila/genetics , Bacillus amyloliquefaciens/enzymology , Bacillus amyloliquefaciens/genetics , Base Sequence , Clostridium acetobutylicum/enzymology , Clostridium acetobutylicum/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Restriction Enzymes/genetics , Escherichia coli K12/enzymology , Gene Expression Regulation, Bacterial , Haemophilus/enzymology , Haemophilus/genetics , Haemophilus influenzae/enzymology , Haemophilus influenzae/genetics , Humans , Microbiota/genetics , Sequence Analysis, DNA , Skin/microbiology
3.
J Immunol ; 205(2): 414-424, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32522834

ABSTRACT

Genome-wide association studies have identified common genetic variants impacting human diseases; however, there are indications that the functional consequences of genetic polymorphisms can be distinct depending on cell type-specific contexts, which produce divergent phenotypic outcomes. Thus, the functional impact of genetic variation and the underlying mechanisms of disease risk are modified by cell type-specific effects of genotype on pathological phenotypes. In this study, we extend these concepts to interrogate the interdependence of cell type- and stimulation-specific programs influenced by the core autophagy gene Atg16L1 and its T300A coding polymorphism identified by genome-wide association studies as linked with increased risk of Crohn's disease. We applied a stimulation-based perturbational profiling approach to define Atg16L1 T300A phenotypes in dendritic cells and T lymphocytes. Accordingly, we identified stimulus-specific transcriptional signatures revealing T300A-dependent functional phenotypes that mechanistically link inflammatory cytokines, IFN response genes, steroid biosynthesis, and lipid metabolism in dendritic cells and iron homeostasis and lysosomal biogenesis in T lymphocytes. Collectively, these studies highlight the combined effects of Atg16L1 genetic variation and stimulatory context on immune function.


Subject(s)
Autophagy-Related Proteins/metabolism , Crohn Disease/metabolism , Dendritic Cells/physiology , Genotype , T-Lymphocytes/physiology , Animals , Autophagy-Related Proteins/genetics , Cells, Cultured , Crohn Disease/genetics , Genetic Predisposition to Disease , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Phenotype , Polymorphism, Genetic , Risk , Transcriptional Activation
4.
J Bacteriol ; 201(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31109990

ABSTRACT

The mechanisms by which bacteria uptake solutes across the cell membrane broadly impact their cellular energetics. Here, we use functional genomic, genetic, and biophysical approaches to reveal how Clostridium (Lachnoclostridium) phytofermentans, a model bacterium that ferments lignocellulosic biomass, uptakes plant hexoses using highly specific, nonredundant ATP-binding cassette (ABC) transporters. We analyze the transcription patterns of its 173 annotated sugar transporter genes to find those upregulated on specific carbon sources. Inactivation of these genes reveals that individual ABC transporters are required for uptake of hexoses and hexo-oligosaccharides and that distinct ABC transporters are used for oligosaccharides versus their constituent monomers. The thermodynamics of sugar binding shows that substrate specificity of these transporters is encoded by the extracellular solute-binding subunit. As sugars are not phosphorylated during ABC transport, we identify intracellular hexokinases based on in vitro activities. These mechanisms used by Clostridia to uptake plant hexoses are key to understanding soil and intestinal microbiomes and to engineer strains for industrial transformation of lignocellulose.IMPORTANCE Plant-fermenting Clostridia are anaerobic bacteria that recycle plant matter in soil and promote human health by fermenting dietary fiber in the intestine. Clostridia degrade plant biomass using extracellular enzymes and then uptake the liberated sugars for fermentation. The main sugars in plant biomass are hexoses, and here, we identify how hexoses are taken in to the cell by the model organism Clostridium phytofermentans We show that this bacterium uptakes hexoses using a set of highly specific, nonredundant ABC transporters. Once in the cell, the hexoses are phosphorylated by intracellular hexokinases. This study provides insight into the functioning of abundant members of soil and intestinal microbiomes and identifies gene targets to engineer strains for industrial lignocellulosic fermentation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Clostridium/metabolism , Hexoses/metabolism , ATP-Binding Cassette Transporters/genetics , Bacterial Proteins/genetics , Biological Transport , Clostridium/genetics
5.
Appl Environ Microbiol ; 83(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28363966

ABSTRACT

Increasing the resistance of plant-fermenting bacteria to lignocellulosic inhibitors is useful to understand microbial adaptation and to develop candidate strains for consolidated bioprocessing. Here, we study and improve inhibitor resistance in Clostridium phytofermentans (also called Lachnoclostridium phytofermentans), a model anaerobe that ferments lignocellulosic biomass. We survey the resistance of this bacterium to a panel of biomass inhibitors and then evolve strains that grow in increasing concentrations of the lignin phenolic, ferulic acid, by automated, long-term growth selection in an anaerobic GM3 automat. Ultimately, strains resist multiple inhibitors and grow robustly at the solubility limit of ferulate while retaining the ability to ferment cellulose. We analyze genome-wide transcription patterns during ferulate stress and genomic variants that arose along the ferulate growth selection, revealing how cells adapt to inhibitors through changes in gene dosage and regulation, membrane fatty acid structure, and the surface layer. Collectively, this study demonstrates an automated framework for in vivo directed evolution of anaerobes and gives insight into the genetic mechanisms by which bacteria survive exposure to chemical inhibitors.IMPORTANCE Fermentation of plant biomass is a key part of carbon cycling in diverse ecosystems. Further, industrial biomass fermentation may provide a renewable alternative to fossil fuels. Plants are primarily composed of lignocellulose, a matrix of polysaccharides and polyphenolic lignin. Thus, when microorganisms degrade lignocellulose to access sugars, they also release phenolic and acidic inhibitors. Here, we study how the plant-fermenting bacterium Clostridium phytofermentans resists plant inhibitors using the lignin phenolic, ferulic acid. We examine how the cell responds to abrupt ferulate stress by measuring changes in gene expression. We evolve increasingly resistant strains by automated, long-term cultivation at progressively higher ferulate concentrations and sequence their genomes to identify mutations associated with acquired ferulate resistance. Our study develops an inhibitor-resistant bacterium that ferments cellulose and provides insights into genomic evolution to resist chemical inhibitors.


Subject(s)
Clostridium/metabolism , Lignin/metabolism , Phenol/metabolism , Plants/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biodegradation, Environmental , Biological Evolution , Biomass , Cellulose/metabolism , Clostridium/genetics , Clostridium/growth & development , Fermentation
6.
PLoS Genet ; 10(11): e1004773, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25393313

ABSTRACT

Microbial metabolism of plant polysaccharides is an important part of environmental carbon cycling, human nutrition, and industrial processes based on cellulosic bioconversion. Here we demonstrate a broadly applicable method to analyze how microbes catabolize plant polysaccharides that integrates carbohydrate-active enzyme (CAZyme) assays, RNA sequencing (RNA-seq), and anaerobic growth screening. We apply this method to study how the bacterium Clostridium phytofermentans ferments plant biomass components including glucans, mannans, xylans, galactans, pectins, and arabinans. These polysaccharides are fermented with variable efficiencies, and diauxies prioritize metabolism of preferred substrates. Strand-specific RNA-seq reveals how this bacterium responds to polysaccharides by up-regulating specific groups of CAZymes, transporters, and enzymes to metabolize the constituent sugars. Fifty-six up-regulated CAZymes were purified, and their activities show most polysaccharides are degraded by multiple enzymes, often from the same family, but with divergent rates, specificities, and cellular localizations. CAZymes were then tested in combination to identify synergies between enzymes acting on the same substrate with different catalytic mechanisms. We discuss how these results advance our understanding of how microbes degrade and metabolize plant biomass.


Subject(s)
Clostridium/enzymology , Enzymes/genetics , Plants/metabolism , Polysaccharides/metabolism , Cell Wall/metabolism , Cellulose/genetics , Cellulose/metabolism , Cloning, Molecular , Enzymes/isolation & purification , Enzymes/metabolism , Fermentation , Glucose/metabolism , Humans , Sequence Analysis, RNA , Xylose/genetics , Xylose/metabolism
7.
Environ Microbiol ; 17(8): 2618-27, 2015 Aug.
Article in English | MEDLINE | ID: mdl-24798076

ABSTRACT

Recycling of plant biomass by a community of bacteria and fungi is fundamental to carbon flow in terrestrial ecosystems. Here we report how the plant fermenting, soil bacterium Clostridium phytofermentans enhances growth on cellulose by simultaneously lysing and consuming model fungi from soil. We investigate the mechanism of fungal lysis to show that among the dozens of different glycoside hydrolases C. phytofermentans secretes on cellulose, the most highly expressed enzymes degrade fungi rather than plant substrates. These enzymes, the GH18 Cphy1799 and Cphy1800, synergize to hydrolyse chitin, a main component of the fungal cell wall. Purified enzymes inhibit fungal growth and mutants lacking either GH18 grow normally on cellulose and other plant substrates, but have a reduced ability to hydrolyse chitinous substrates and fungal hyphae. Thus, C. phytofermentans boosts growth on cellulose by lysing fungi with its most highly expressed hydrolases, highlighting the importance of fungal interactions to the ecology of cellulolytic bacteria.


Subject(s)
Cellulose/metabolism , Chitin/metabolism , Clostridium/enzymology , Clostridium/growth & development , Fungi/metabolism , Glycoside Hydrolases/metabolism , Soil Microbiology , Cell Wall/metabolism , Ecosystem , Fermentation , Hydrolysis , Plants/metabolism , Soil
8.
Appl Environ Microbiol ; 81(16): 5440-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26048945

ABSTRACT

Novel processing strategies for hydrolysis and fermentation of lignocellulosic biomass in a single reactor offer large potential cost savings for production of biocommodities and biofuels. One critical challenge is retaining high enzyme production in the presence of elevated product titers. Toward this goal, the cellulolytic, ethanol-producing bacterium Clostridium phytofermentans was adapted to increased ethanol concentrations. The resulting ethanol-tolerant (ET) strain has nearly doubled ethanol tolerance relative to the wild-type level but also reduced ethanol yield and growth at low ethanol concentrations. The genome of the ET strain has coding changes in proteins involved in membrane biosynthesis, the Rnf complex, cation homeostasis, gene regulation, and ethanol production. In particular, purification of the mutant bifunctional acetaldehyde coenzyme A (CoA)/alcohol dehydrogenase showed that a G609D variant abolished its activities, including ethanol formation. Heterologous expression of Zymomonas mobilis pyruvate decarboxylase and alcohol dehydrogenase in the ET strain increased cellulose consumption and restored ethanol production, demonstrating how metabolic engineering can be used to overcome disadvantageous mutations incurred during adaptation to ethanol. We discuss how genetic changes in the ET strain reveal novel potential strategies for improving microbial solvent tolerance.


Subject(s)
Cellulose/metabolism , Clostridium/genetics , Clostridium/metabolism , Ethanol/metabolism , Metabolic Engineering , Metabolic Networks and Pathways/genetics , Adaptation, Biological , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Drug Tolerance , Ethanol/toxicity , Gene Expression , Pyruvate Decarboxylase/genetics , Pyruvate Decarboxylase/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Zymomonas/enzymology , Zymomonas/genetics
9.
Front Bioeng Biotechnol ; 11: 1324396, 2023.
Article in English | MEDLINE | ID: mdl-38239921

ABSTRACT

The Lachnospiraceae is a family of anaerobic bacteria in the class Clostridia with potential to advance the bio-economy and intestinal therapeutics. Some species of Lachnospiraceae metabolize abundant, low-cost feedstocks such as lignocellulose and carbon dioxide into value-added chemicals. Others are among the dominant species of the human colon and animal rumen, where they ferment dietary fiber to promote healthy gut and immune function. Here, we summarize recent studies of the physiology, cultivation, and genetics of Lachnospiraceae, highlighting their wide substrate utilization and metabolic products with industrial applications. We examine studies of these bacteria as Live Biotherapeutic Products (LBPs), focusing on in vivo disease models and clinical studies using them to treat infection, inflammation, metabolic syndrome, and cancer. We discuss key research areas including elucidation of intra-specific diversity and genetic modification of candidate strains that will facilitate the exploitation of Lachnospiraceae in industry and medicine.

10.
Mol Syst Biol ; 7: 461, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21245846

ABSTRACT

Fermentation of plant biomass by microbes like Clostridium phytofermentans recycles carbon globally and can make biofuels from inedible feedstocks. We analyzed C. phytofermentans fermenting cellulosic substrates by integrating quantitative mass spectrometry of more than 2500 proteins with measurements of growth, enzyme activities, fermentation products, and electron microscopy. Absolute protein concentrations were estimated using Absolute Protein EXpression (APEX); relative changes between treatments were quantified with chemical stable isotope labeling by reductive dimethylation (ReDi). We identified the different combinations of carbohydratases used to degrade cellulose and hemicellulose, many of which were secreted based on quantification of supernatant proteins, as well as the repertoires of glycolytic enzymes and alcohol dehydrogenases (ADHs) enabling ethanol production at near maximal yields. Growth on cellulose also resulted in diverse changes such as increased expression of tryptophan synthesis proteins and repression of proteins for fatty acid metabolism and cell motility. This study gives a systems-level understanding of how this microbe ferments biomass and provides a rational, empirical basis to identify engineering targets for industrial cellulosic fermentation.


Subject(s)
Bacterial Proteins/metabolism , Cellulose/metabolism , Clostridium/metabolism , Proteome/metabolism , Systems Biology/methods , Bacterial Proteins/analysis , Biofuels , Biomass , Carbon/metabolism , Cell Adhesion , Clostridium/cytology , Clostridium/enzymology , Clostridium/physiology , Glucose/metabolism , Linear Models , Mass Spectrometry , Metabolic Networks and Pathways , Microscopy, Electron, Scanning , Polysaccharides/metabolism , Proteome/analysis
11.
Microbiol Spectr ; 10(2): e0228821, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35412381

ABSTRACT

Transcription initiation is a tightly regulated process that is crucial for many aspects of prokaryotic physiology. High-throughput transcription start site (TSS) mapping can shed light on global and local regulation of transcription initiation, which in turn may help us understand and predict microbial behavior. In this study, we used Capp-Switch sequencing to determine the TSS positions in the genomes of three model solventogenic clostridia: Clostridium acetobutylicum ATCC 824, C. beijerinckii DSM 6423, and C. beijerinckii NCIMB 8052. We first refined the approach by implementing a normalization pipeline accounting for gene expression, yielding a total of 12,114 mapped TSSs across the species. We further compared the distributions of these sites in the three strains. Results indicated similar distribution patterns at the genome scale, but also some sharp differences, such as for the butyryl-CoA synthesis operon, particularly when comparing C. acetobutylicum to the C. beijerinckii strains. Lastly, we found that promoter structure is generally poorly conserved between C. acetobutylicum and C. beijerinckii. A few conserved promoters across species are discussed, showing interesting examples of how TSS determination and comparison can improve our understanding of gene expression regulation at the transcript level. IMPORTANCE Solventogenic clostridia have been employed in industry for more than a century, initially being used in the acetone-butanol-ethanol (ABE) fermentation process for acetone and butanol production. Interest in these bacteria has recently increased in the context of green chemistry and sustainable development. However, our current understanding of their genomes and physiology limits their optimal use as industrial solvent production platforms. The gene regulatory mechanisms of solventogenesis are still only partly understood, impeding efforts to increase rates and yields. Genome-wide mapping of transcription start sites (TSSs) for three model solventogenic Clostridium strains is an important step toward understanding mechanisms of gene regulation in these industrially important bacteria.


Subject(s)
Acetone , Clostridium acetobutylicum , Acetone/metabolism , Bacteria, Anaerobic , Butanols/metabolism , Clostridium/genetics , Clostridium/metabolism , Clostridium acetobutylicum/genetics , Clostridium acetobutylicum/metabolism , Fermentation
12.
ACS Synth Biol ; 11(12): 4077-4088, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36427328

ABSTRACT

Control of gene expression is fundamental to cell engineering. Here we demonstrate a set of approaches to tune gene expression in Clostridia using the model Clostridium phytofermentans. Initially, we develop a simple benchtop electroporation method that we use to identify a set of replicating plasmids and resistance markers that can be cotransformed into C. phytofermentans. We define a series of promoters spanning a >100-fold expression range by testing a promoter library driving the expression of a luminescent reporter. By insertion of tet operator sites upstream of the reporter, its expression can be quantitatively altered using the Tet repressor and anhydrotetracycline (aTc). We integrate these methods into an aTc-regulated dCas12a system with which we show in vivo CRISPRi-mediated repression of reporter and fermentation genes in C. phytofermentans. Together, these approaches advance genetic transformation and experimental control of gene expression in Clostridia.


Subject(s)
Clostridiales , Clostridium , Clostridiales/genetics , Promoter Regions, Genetic/genetics , Clostridium/genetics , Clostridium/metabolism , Gene Expression
13.
Nat Commun ; 13(1): 1244, 2022 03 10.
Article in English | MEDLINE | ID: mdl-35273143

ABSTRACT

Relative abundances of bacterial species in the gut microbiome have been linked to many diseases. Species of gut bacteria are ecologically differentiated by their abilities to metabolize different glycans, making glycan delivery a powerful way to alter the microbiome to promote health. Here, we study the properties and therapeutic potential of chemically diverse synthetic glycans (SGs). Fermentation of SGs by gut microbiome cultures results in compound-specific shifts in taxonomic and metabolite profiles not observed with reference glycans, including prebiotics. Model enteric pathogens grow poorly on most SGs, potentially increasing their safety for at-risk populations. SGs increase survival, reduce weight loss, and improve clinical scores in mouse models of colitis. Synthetic glycans are thus a promising modality to improve health through selective changes to the gut microbiome.


Subject(s)
Colitis , Gastrointestinal Microbiome , Animals , Bacteria/metabolism , Colitis/drug therapy , Health Promotion , Mice , Polysaccharides/metabolism
14.
Mol Microbiol ; 74(6): 1300-13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19775243

ABSTRACT

Summary Microbial cellulose degradation is a central part of the global carbon cycle and has great potential for the development of inexpensive, carbon-neutral biofuels from non-food crops. Clostridium phytofermentans has a repertoire of 108 putative glycoside hydrolases to break down cellulose and hemicellulose into sugars, which this organism then ferments primarily to ethanol. An understanding of cellulose degradation at the molecular level requires learning the different roles of these hydrolases. In this study, we show that interspecific conjugation with Escherichia coli can be used to transfer a plasmid into C. phytofermentans that has a resistance marker, an origin of replication that can be selectively lost, and a designed group II intron for efficient, targeted chromosomal insertions without selection. We applied these methods to disrupt the cphy3367 gene, which encodes the sole family 9 glycoside hydrolase (GH9) in the C. phytofermentans genome. The GH9-deficient strain grew normally on some carbon sources such as glucose, but had lost the ability to degrade cellulose. Although C. phytofermentans upregulates the expression of numerous enzymes to break down cellulose, this process thus relies upon a single, key hydrolase, Cphy3367.


Subject(s)
Cellulase/metabolism , Cellulose/metabolism , Clostridium/enzymology , Gene Silencing , Gene Targeting , Cellulase/genetics , Clostridium/genetics , Conjugation, Genetic , Escherichia coli/genetics , Introns , Models, Biological , Models, Chemical , Mutagenesis, Insertional/methods , Phylogeny , Sequence Homology, Amino Acid
15.
Nat Microbiol ; 5(3): 486-497, 2020 03.
Article in English | MEDLINE | ID: mdl-31959971

ABSTRACT

Inflammatory bowel diseases (IBD) are associated with alterations in gut microbial abundances and lumenal metabolite concentrations, but the effects of specific metabolites on the gut microbiota in health and disease remain largely unknown. Here, we analysed the influences of metabolites that are differentially abundant in IBD on the growth and physiology of gut bacteria that are also differentially abundant in IBD. We found that N-acylethanolamines (NAEs), a class of endogenously produced signalling lipids elevated in the stool of IBD patients and a T-cell transfer model of colitis, stimulated growth of species over-represented in IBD and inhibited that of species depleted in IBD in vitro. Using metagenomic sequencing, we recapitulated the effects of NAEs in complex microbial communities ex vivo, with Proteobacteria blooming and Bacteroidetes declining in the presence of NAEs. Metatranscriptomic analysis of the same communities identified components of the respiratory chain as important for the metabolism of NAEs, and this was verified using a mutant deficient for respiratory complex I. In this study, we identified NAEs as a class of metabolites that are elevated in IBD and have the potential to shift gut microbiota towards an IBD-like composition.


Subject(s)
Bacteria/drug effects , Bacteria/growth & development , Ethanolamines/pharmacology , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/drug therapy , Animals , Bacteria/genetics , Bacteroidetes/drug effects , Bacteroidetes/isolation & purification , Cohort Studies , Disease Models, Animal , Dysbiosis , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Gene Expression Profiling , Humans , Inflammatory Bowel Diseases/microbiology , Male , Metagenome , Mice , Mice, Inbred C57BL , Microbiota/drug effects , Proteobacteria/drug effects , Proteobacteria/isolation & purification , Tandem Mass Spectrometry , Whole Genome Sequencing
16.
mSphere ; 4(6)2019 12 11.
Article in English | MEDLINE | ID: mdl-31826971

ABSTRACT

Clostridia are a group of Gram-positive anaerobic bacteria of medical and industrial importance for which limited genetic methods are available. Here, we demonstrate an approach to make large genomic deletions and insertions in the model Clostridium phytofermentans by combining designed group II introns (targetrons) and Cre recombinase. We apply these methods to delete a 50-gene prophage island by programming targetrons to position markerless lox66 and lox71 sites, which mediate deletion of the intervening 39-kb DNA region using Cre recombinase. Gene expression and growth of the deletion strain showed that the prophage genes contribute to fitness on nonpreferred carbon sources. We also inserted an inducible fluorescent reporter gene into a neutral genomic site by recombination-mediated cassette exchange (RMCE) between genomic and plasmid-based tandem lox sites bearing heterospecific spacers to prevent intracassette recombination. These approaches generally enable facile markerless genome engineering in clostridia to study their genome structure and regulation.IMPORTANCE Clostridia are anaerobic bacteria with important roles in intestinal and soil microbiomes. The inability to experimentally modify the genomes of clostridia has limited their study and application in biotechnology. Here, we developed a targetron-recombinase system to efficiently make large targeted genomic deletions and insertions using the model Clostridium phytofermentans We applied this approach to reveal the importance of a prophage to host fitness and introduce an inducible reporter by recombination-mediated cassette exchange.


Subject(s)
Clostridiales/genetics , Gene Editing/methods , Genetics, Microbial/methods , Molecular Biology/methods , Carbon/metabolism , Clostridiales/growth & development , Clostridiales/metabolism , Clostridiales/virology , Gene Deletion , Genetic Fitness , Integrases , Introns , Prophages/genetics
17.
Science ; 363(6423): 181-187, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30630933

ABSTRACT

Phase variation, the reversible alternation between genetic states, enables infection by pathogens and colonization by commensals. However, the diversity of phase variation remains underexplored. We developed the PhaseFinder algorithm to quantify DNA inversion-mediated phase variation. A systematic search of 54,875 bacterial genomes identified 4686 intergenic invertible DNA regions (invertons), revealing an enrichment in host-associated bacteria. Invertons containing promoters often regulate extracellular products, underscoring the importance of surface diversity for gut colonization. We found invertons containing promoters regulating antibiotic resistance genes that shift to the ON orientation after antibiotic treatment in human metagenomic data and in vitro, thereby mitigating the cost of antibiotic resistance. We observed that the orientations of some invertons diverge after fecal microbiota transplant, potentially as a result of individual-specific selective forces.


Subject(s)
Bacteria/genetics , DNA, Intergenic/genetics , Drug Resistance, Bacterial/genetics , Gastrointestinal Microbiome , Promoter Regions, Genetic , Algorithms , DNA, Bacterial/genetics , Genome, Bacterial , Humans
18.
Mol Syst Biol ; 2: 53, 2006.
Article in English | MEDLINE | ID: mdl-17016519

ABSTRACT

Nitrogen (N) often limits biological productivity in the oceanic gyres where Prochlorococcus is the most abundant photosynthetic organism. The Prochlorococcus community is composed of strains, such as MED4 and MIT9313, that have different N utilization capabilities and that belong to ecotypes with different depth distributions. An interstrain comparison of how Prochlorococcus responds to changes in ambient nitrogen is thus central to understanding its ecology. We quantified changes in MED4 and MIT9313 global mRNA expression, chlorophyll fluorescence, and photosystem II photochemical efficiency (Fv/Fm) along a time series of increasing N starvation. In addition, the global expression of both strains growing in ammonium-replete medium was compared to expression during growth on alternative N sources. There were interstrain similarities in N regulation such as the activation of a putative NtcA regulon during N stress. There were also important differences between the strains such as in the expression patterns of carbon metabolism genes, suggesting that the two strains integrate N and C metabolism in fundamentally different ways.


Subject(s)
Gene Expression Regulation, Bacterial , Nitrogen/metabolism , Prochlorococcus/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Carbon/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Ecology , Energy Metabolism , Gene Expression Profiling , Oceans and Seas , Operon , PII Nitrogen Regulatory Proteins/physiology , Photosystem II Protein Complex/physiology , Prochlorococcus/growth & development , Prochlorococcus/metabolism , Prochlorococcus/radiation effects , Sigma Factor/physiology , Species Specificity , Transcription Factors/physiology , Water Microbiology
19.
Genome Med ; 9(1): 56, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615076

ABSTRACT

Recent advances in genome sequencing of single microbial cells enable the assignment of functional roles to members of the human microbiome that cannot currently be cultured. This approach can reveal the genomic basis of phenotypic variation between closely related strains and can be applied to the targeted study of immunogenic bacteria in disease.


Subject(s)
Bacteria/genetics , Genome, Bacterial , Genomics/methods , Microbiota , Sequence Analysis, DNA/methods , Humans
20.
Nucleic Acids Res ; 30(20): e107, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12384609

ABSTRACT

Oligonucleotide arrays are powerful tools to study changes in gene expression for whole genomes. These arrays can be synthesized by adapting photolithographic techniques used in microelectronics. Using this method, oligonucleotides are built base by base directly on the array surface by numerous cycles of photodeprotection and nucleotide addition. In this paper we examine strategies to reduce the number of synthesis cycles required to construct oligonucleotide arrays. By computer modeling oligonucleotide synthesis, we found that the number of required synthesis cycles could be significantly reduced by focusing upon how oligonucleotides are chosen from within genes and upon the order in which nucleotides are deposited on the array. The methods described here could provide a more efficient strategy to produce oligonucleotide arrays.


Subject(s)
Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Oligonucleotides/chemical synthesis , Computer Simulation
SELECTION OF CITATIONS
SEARCH DETAIL