Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biotechnol Adv ; 72: 108336, 2024.
Article in English | MEDLINE | ID: mdl-38432422

ABSTRACT

The population dynamics of the human microbiome have been associated with inflammatory bowel disease, cancer, obesity, autoimmune diseases, and many other human disease states. An emerging paradigm in treatment is the administration of live engineered organisms, also called next-generation probiotics. However, the efficacy of these microbial therapies can be limited by the organism's overall performance in the harsh and nutrient-limited environment of the gut. In this review, we summarize the current state of the art use of bacterial and yeast strains as probiotics, highlight the recent development of genetic tools for engineering new therapeutic functions in these organisms, and report on the latest therapeutic applications of engineered probiotics, including recent clinical trials. We also discuss the supplementation of prebiotics as a method of manipulating the microbiome and improving the overall performance of engineered live biotherapeutics.


Subject(s)
Microbiota , Probiotics , Humans , Probiotics/therapeutic use , Prebiotics
2.
Int J Mol Sci ; 25(1)2023 Dec 30.
Article in English | MEDLINE | ID: mdl-38203712

ABSTRACT

Gut microbiota plays a crucial role in inflammatory bowel diseases (IBD) and can potentially prevent IBD through microbial-derived metabolites, making it a promising therapeutic avenue. Recent evidence suggests that despite an unclear underlying mechanism, red cabbage juice (RCJ) alleviates Dextran Sodium Sulfate (DSS)-induced colitis in mice. Thus, the study aims to unravel the molecular mechanism by which RCJ modulates the gut microbiota to alleviate DSS-induced colitis in mice. Using C57BL/6J mice, we evaluated RCJ's protective role in DSS-induced colitis through two cycles of 3% DSS. Mice were daily gavaged with PBS or RCJ until the endpoint, and gut microbiota composition was analyzed via shotgun metagenomics. RCJ treatment significantly improved body weight (p ≤ 0.001), survival in mice (p < 0.001) and reduced disease activity index (DAI) scores. Further, RCJ improved colonic barrier integrity by enhancing the expression of protective colonic mucins (p < 0.001) and tight junction proteins (p ≤ 0.01) in RCJ + DSS-treated mice compared to the DSS group. Shotgun metagenomic analysis revealed an enrichment of short-chain fatty acids (SCFAs)-producing bacteria (p < 0.05), leading to increased Peroxisome Proliferator-Activated Receptor Gamma (PPAR-γ) activation (p ≤ 0.001). This, in turn, resulted in repression of the nuclear factor κB (NFκB) signaling pathway, causing decreased production of inflammatory cytokines and chemokines. Our study demonstrates colitis remission in a DSS-induced mouse model, showcasing RCJ as a potential modulator for gut microbiota and metabolites, with promising implications for IBD prevention and treatment.


Subject(s)
Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Animals , Mice , Mice, Inbred C57BL , Colitis/chemically induced , Homeostasis
3.
Int J Biol Macromol ; 164: 548-556, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32693143

ABSTRACT

Cytokines are key factors affecting the fate of intestinal stem cells (ISCs) and effective reagents to manipulate ISCs for research purpose. Tumor necrosis factor alpha (TNFα) is a cytokine produced primarily by monocytes and macrophages. It can induce apoptotic cell death and inflammation, and to inhibit tumorigenesis and viral replication. Additionally, TNFα has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). It is therefore important to identify the mechanism by which individual cytokines affect particular cell types. For this purpose, we used both conventional (CONV) and altered Schaedler flora (ASF) C3H/HeN mice to elucidate the effect of different microbial populations (complex versus defined) on growth of miniguts derived from two different intestinal environments. Furthermore, we studied the effects of different concentrations of TNFα extracted from the lymph and spleen on the growth and viability of ISCs recovered from mice bearing the ASF or CONV microbiota. The effect of TNFα on miniguts growth depends not only on the source and concentration, but also on the intestinal microenvironment from which the ISCs were derived. The findings suggest that TNFα influences the proliferation of miniguts derived from ISCs and, therefore, modulates mucosal homeostasis of the host.


Subject(s)
Intestines/microbiology , Lymph/immunology , Organoids/growth & development , Spleen/immunology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cellular Microenvironment/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Intestines/cytology , Intestines/drug effects , Mice , Organoids/drug effects , Organoids/microbiology , Primary Cell Culture , Stem Cells/cytology , Stem Cells/drug effects
4.
Metab Eng ; 61: 120-130, 2020 09.
Article in English | MEDLINE | ID: mdl-32474056

ABSTRACT

Adaptive laboratory evolution is often used to improve the performance of microbial cell factories. Reverse engineering of evolved strains enables learning and subsequent incorporation of novel design strategies via the design-build-test-learn cycle. Here, we reverse engineer a strain of Escherichia coli previously evolved for increased tolerance of octanoic acid (C8), an attractive biorenewable chemical, resulting in increased C8 production, increased butanol tolerance, and altered membrane properties. Here, evolution was determined to have occurred first through the restoration of WaaG activity, involved in the production of lipopolysaccharides, then an amino acid change in RpoC, a subunit of RNA polymerase, and finally mutation of the BasS-BasR two component system. All three mutations were required in order to reproduce the increased growth rate in the presence of 20 mM C8 and increased cell surface hydrophobicity; the WaaG and RpoC mutations both contributed to increased C8 titers, with the RpoC mutation appearing to be the major driver of this effect. Each of these mutations contributed to changes in the cell membrane. Increased membrane integrity and rigidity and decreased abundance of extracellular polymeric substances can be attributed to the restoration of WaaG. The increase in average lipid tail length can be attributed to the RpoCH419P mutation, which also confers tolerance to other industrially-relevant inhibitors, such as furfural, vanillin and n-butanol. The RpoCH419P mutation may impact binding or function of the stringent response alarmone ppGpp to RpoC site 1. Each of these mutations provides novel strategies for engineering microbial robustness, particularly at the level of the microbial cell membrane.


Subject(s)
Caprylates/metabolism , DNA-Directed RNA Polymerases , Escherichia coli Proteins , Escherichia coli , Glucosyltransferases , Metabolic Engineering , Mutation, Missense , Amino Acid Substitution , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism
5.
Int J Mol Sci ; 21(10)2020 May 20.
Article in English | MEDLINE | ID: mdl-32443851

ABSTRACT

The short-chain fatty acid butyrate plays critical roles in human gut health, affecting immunomodulation, cell differentiation, and apoptosis, while also serving as the preferred carbon source for colon cells. In this work, we have engineered a model probiotic organism, Escherichia coli Nissle 1917 (EcN, serotype O6:K5:H1), to produce butyrate from genomic loci up to approximately 1 g/L (11 mM). Then, for real-time monitoring of butyrate production in cultures, we developed a high-throughput biosensor that responds to intracellular butyrate concentrations, with green fluorescent protein as the reporter. This work provides a foundation for studies of butyrate for therapeutic applications.


Subject(s)
Biosensing Techniques/methods , Butyrates/metabolism , Escherichia coli Proteins/metabolism , Probiotics/metabolism , Butyrates/analysis , Escherichia coli , Escherichia coli Proteins/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Recombinant Proteins
6.
Curr Opin Biotechnol ; 61: 181-188, 2020 02.
Article in English | MEDLINE | ID: mdl-31986469

ABSTRACT

Lactic acid bacteria play a vital role as starter cultures in the fermentation of a variety of foods, often altering the flavor and texture in addition to aiding in preservation. Their importance to the industry has made them targets for metabolic engineering to improve relevant phenotypes and several methods of genome engineering in these organisms have been established. While the efficiency of these techniques is variable, in recent years the ability to select for successful recombinants has markedly increased the throughput of genome engineering. This review summarizes recent advances in genome engineering in lactic acid bacteria and applications that will be enabled by high-throughput techniques.


Subject(s)
Lactobacillales/genetics , Fermentation , Food Microbiology , Metabolic Engineering , Taste
7.
Fungal Genet Biol ; 137: 103333, 2020 04.
Article in English | MEDLINE | ID: mdl-31923554

ABSTRACT

The presence of commensal fungal species in the human gut indicates that organisms from this kingdom have the potential to benefit the host as well. Saccharomyces boulardii, a yeast strain isolated about a hundred years ago, is the most well-characterized probiotic yeast. Though for the most part it genetically resembles Saccharomyces cerevisiae, specific phenotypic differences make it better suited for the gut microenvironment such as better acid and heat tolerance. Several studies using animal hosts suggest that S. boulardii can be used as a biotherapeutic in humans. Clinical trials indicate that it can alleviate symptoms from gastrointestinal (GI) tract infections to some extent, but further trials are needed to understand the full therapeutic potential of S. boulardii. Improvement on probiotic function using engineered yeast is an attractive future direction, though genome modification tools for use in S. boulardii have been limited until recently. However, some tools available for S. cerevisiae should be applicable for S. boulardii as well. In this review, we summarize the observed probiotic effect of this yeast and the state of the art for genome engineering tools that could help enhance its probiotic properties.


Subject(s)
Probiotics/metabolism , Probiotics/therapeutic use , Saccharomyces boulardii/metabolism , Animals , Humans , Saccharomyces/genetics , Saccharomyces/metabolism , Saccharomyces boulardii/genetics , Yeasts/genetics , Yeasts/metabolism
8.
J Ind Microbiol Biotechnol ; 46(9-10): 1445-1459, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31201649

ABSTRACT

The human gut is an ecosystem comprising trillions of microbes interacting with the host. The composition of the microbiota and their interactions play roles in different biological processes and in the development of human diseases. Close relationships between dietary modifications, microbiota composition and health status have been established. This review focuses on prebiotics, or compounds which selectively encourage the growth of beneficial bacteria, their mechanisms of action and benefits to human hosts. We also review advances in synthesis technology for human milk oligosaccharides, part of one of the most well-characterized prebiotic-probiotic relationships. Current and future research in this area points to greater use of prebiotics as tools to manipulate the microbial and metabolic diversity of the gut for the benefit of human health.


Subject(s)
Gastrointestinal Microbiome , Metabolome , Animals , Bacteria/metabolism , Humans , Oligosaccharides/metabolism , Prebiotics , Probiotics
9.
Metab Eng Commun ; 9: e00088, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31008057

ABSTRACT

Asparagine-linked (N-linked) glycosylation is the most common protein modification in eukaryotes, affecting over two-thirds of the proteome. Glycosylation is also critical to the pharmacokinetic activity and immunogenicity of many therapeutic proteins currently produced in complex eukaryotic hosts. The discovery of a protein glycosylation pathway in the pathogen Campylobacter jejuni and its subsequent transfer into laboratory strains of Escherichia coli has spurred great interest in glycoprotein production in prokaryotes. However, prokaryotic glycoprotein production has several drawbacks, including insufficient availability of non-native glycan precursors. To address this limitation, we used a constraint-based model of E. coli metabolism in combination with heuristic optimization to design gene knockout strains that overproduced glycan precursors. First, we incorporated reactions associated with C. jejuni glycan assembly into a genome-scale model of E. coli metabolism. We then identified gene knockout strains that coupled optimal growth to glycan synthesis. Simulations suggested that these growth-coupled glycan overproducing strains had metabolic imbalances that rerouted flux toward glycan precursor synthesis. We then validated the model-identified knockout strains experimentally by measuring glycan expression using a flow cytometric-based assay involving fluorescent labeling of cell surface-displayed glycans. Overall, this study demonstrates the promising role that metabolic modeling can play in optimizing the performance of a next-generation microbial glycosylation platform.

10.
J Vis Exp ; (146)2019 04 13.
Article in English | MEDLINE | ID: mdl-31033942

ABSTRACT

Human milk oligosaccharides (HMOs) are complex carbohydrate components of human breast milk that exhibit plentiful benefits on infant health. However, optimization of their biotechnological synthesis is limited by the relatively low throughput of detection and quantification of monosaccharide and linkages. Conventional techniques of glycan analysis include chromatographic/mass-spectrometric methods with throughput on the order of hundreds of samples per day without automation. We demonstrate here, a genetically encoded bacterial biosensor for the high-throughput, linkage-specific detection and quantification of the fucosylated HMO structures, 2'-fucosyllactose and 3-fucosyllactose, which we achieved via heterologous expression of fucosidases. As the presence of lactose in milk or in biotechnological processes could lead to false positives, we also demonstrate the reduction of signal from lactose using different strategies. Due to the high throughput of this technique, many reaction conditions or bioreactor parameters could be assayed in parallel in a matter of hours, allowing for the optimization of HMO manufacturing.


Subject(s)
Bioreactors , Biotechnology/methods , Genetic Engineering , Milk, Human/metabolism , Trisaccharides/metabolism , Glycosylation , Humans , Oligosaccharides/metabolism , alpha-L-Fucosidase/genetics
11.
Eng Life Sci ; 19(6): 478-485, 2019 Jun.
Article in English | MEDLINE | ID: mdl-32625025

ABSTRACT

E. coli Nissle 1917 (EcN) has long been used as an over-the-counter probiotic and has shown potential to be used as a live biotherapeutic. It contains two stably replicating cryptic plasmids, pMUT1, and pMUT2, the function of which is unclear but the presence of which may increase the metabolic burden on the cell, particularly in the context of added recombinant plasmids. In this work, we present a clustered regularly interspaced short palindromic repeats-Cas9-based method of curing cryptic plasmids, producing strains cured of one or both plasmids. We then assayed heterologous protein production from three different recombinant plasmids in wild-type and cured EcN derivatives and found that production of reporter proteins was not significantly different across strains. In addition, we replaced pMUT2 with an engineered version containing an inserted antibiotic resistance reporter gene and demonstrated that the engineered plasmid was stable over 90 generations without selection. These findings have broad implications for the curing of cryptic plasmids and for stable heterologous expression of proteins in this host. Specifically, curing of cryptic plasmids may not be necessary for optimal heterologous expression in this host.

12.
Cell Chem Biol ; 25(10): 1292-1303.e4, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30017916

ABSTRACT

Human milk oligosaccharides (HMOs) are important prebiotic complex carbohydrates with demonstrated beneficial effects on the microbiota of neonates. However, optimization of their biotechnological synthesis is limited by the relatively low throughput of monosaccharide and linkage analysis. To enable high-throughput screening of HMO structures, we constructed a whole-cell biosensor that uses heterologous expression of glycosidases to generate linkage-specific, quantitative fluorescent readout for a range of HMOs at detection limits down to 20 µM in approximately 6 hr. We also demonstrate the use of this system for orthogonal control of growth rate or protein expression of particular strains in mixed populations. This work enables rapid non-chromatographic linkage analysis and lays the groundwork for the application of directed evolution to biosynthesis of complex carbohydrates as well as the prebiotic manipulation of population dynamics in natural and engineered microbial communities.


Subject(s)
Biosensing Techniques/methods , Escherichia coli/metabolism , Milk, Human/chemistry , Oligosaccharides/analysis , Acetylglucosamine/analysis , Acetylglucosamine/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Humans , Lactose/analogs & derivatives , Lactose/analysis , Lactose/metabolism , Milk, Human/metabolism , Oligosaccharides/metabolism , Sialic Acids/analysis , Sialic Acids/metabolism
13.
Appl Environ Microbiol ; 84(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30030228

ABSTRACT

Fermentative production of many attractive biorenewable fuels and chemicals is limited by product toxicity in the form of damage to the microbial cell membrane. Metabolic engineering of the production organism can help mitigate this problem, but there is a need for identification and prioritization of the most effective engineering targets. Here, we use a set of previously characterized environmental Escherichia coli isolates with high tolerance and production of octanoic acid, a model membrane-damaging biorenewable product, as a case study for identifying and prioritizing membrane engineering strategies. This characterization identified differences in the membrane lipid composition, fluidity, integrity, and cell surface hydrophobicity from those of the lab strain MG1655. Consistent with previous publications, decreased membrane fluidity was associated with increased fatty acid production ability. Maintenance of high membrane integrity or longer membrane lipids seemed to be of less importance than fluidity. Cell surface hydrophobicity was also directly associated with fatty acid production titers, with the strength of this association demonstrated by plasmid-based expression of the multiple stress resistance outer membrane protein BhsA. This expression of bhsA was effective in altering hydrophobicity, but the direction and magnitude of the change differed between strains. Thus, additional strategies are needed to reliably engineer cell surface hydrophobicity. This work demonstrates the ability of environmental microbiological studies to impact the metabolic engineering design-build-test-learn cycle and possibly increase the economic viability of fermentative bioprocesses.IMPORTANCE The production of bulk fuels and chemicals in a bio-based fermentation process requires high product titers. This is often difficult to achieve, because many of the target molecules damage the membrane of the microbial cell factory. Engineering the composition of the membrane in order to decrease its vulnerability to this damage has proven to be an effective strategy for improving bioproduction, but additional strategies and engineering targets are needed. Here, we studied a small set of environmental Escherichia coli isolates that have higher production titers of octanoic acid, a model biorenewable chemical, than those of the lab strain MG1655. We found that membrane fluidity and cell surface hydrophobicity are strongly associated with improved octanoic acid production. Fewer genetic modification strategies have been demonstrated for tuning hydrophobicity relative to fluidity, leading to the conclusion that there is a need for expanding hydrophobicity engineering strategies in E. coli.


Subject(s)
Caprylates/metabolism , Cell Membrane/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Cell Membrane/chemistry , Cell Membrane/genetics , Environmental Microbiology , Escherichia coli/chemistry , Escherichia coli/isolation & purification , Fatty Acids/metabolism , Hydrophobic and Hydrophilic Interactions , Membrane Fluidity , Membrane Lipids/metabolism , Metabolic Engineering
14.
PLoS One ; 11(1): e0146916, 2016.
Article in English | MEDLINE | ID: mdl-26771672

ABSTRACT

Chemical genomics expands our understanding of microbial tolerance to inhibitory chemicals, but its scope is often limited by the throughput of genome-scale library construction and genotype-phenotype mapping. Here we report a method for rapid, parallel, and deep characterization of the response to antibiotics in Escherichia coli using a barcoded genome-scale library, next-generation sequencing, and streamlined bioinformatics software. The method provides quantitative growth data (over 200,000 measurements) and identifies contributing antimicrobial resistance and susceptibility alleles. Using multivariate analysis, we also find that subtle differences in the population responses resonate across multiple levels of functional hierarchy. Finally, we use machine learning to identify a unique allelic and proteomic fingerprint for each antibiotic. The method can be broadly applied to tolerance for any chemical from toxic metabolites to next-generation biofuels and antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/genetics , Genome, Bacterial/genetics , Alleles , Chromosome Mapping , Drug Resistance, Microbial/genetics , Escherichia coli/drug effects , Genomic Library
15.
Nat Commun ; 6: 8072, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26311203

ABSTRACT

Current methods for producing immunoglobulin G (IgG) antibodies in engineered cells often require refolding steps or secretion across one or more biological membranes. Here, we describe a robust expression platform for biosynthesis of full-length IgG antibodies in the Escherichia coli cytoplasm. Synthetic heavy and light chains, both lacking canonical export signals, are expressed in specially engineered E. coli strains that permit formation of stable disulfide bonds within the cytoplasm. IgGs with clinically relevant antigen- and effector-binding activities are readily produced in the E. coli cytoplasm by grafting antigen-specific variable heavy and light domains into a cytoplasmically stable framework and remodelling the fragment crystallizable domain with amino-acid substitutions that promote binding to Fcγ receptors. The resulting cytoplasmic IgGs­named 'cyclonals'­effectively bypass the potentially rate-limiting steps of membrane translocation and glycosylation.


Subject(s)
Antibody Formation/genetics , Cytoplasm/metabolism , Escherichia coli/genetics , Immunoglobulin G/biosynthesis , Organisms, Genetically Modified/genetics , Antibodies , Bacteriophages/genetics , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Glycosylation , Plasmids/genetics , Protein Transport , Surface Plasmon Resonance
16.
Nat Biotechnol ; 33(6): 631-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25798935

ABSTRACT

Multiplexed genome engineering approaches can be used to generate targeted genetic diversity in cell populations on laboratory timescales, but methods to track mutations and link them to phenotypes have been lacking. We present an approach for tracking combinatorial engineered libraries (TRACE) through the simultaneous mapping of millions of combinatorially engineered genomes at single-cell resolution. Distal genomic sites are assembled into individual DNA constructs that are compatible with next-generation sequencing strategies. We used TRACE to map growth selection dynamics for Escherichia coli combinatorial libraries created by recursive multiplex recombineering at a depth 10(4)-fold greater than before. TRACE was used to identify genotype-to-phenotype correlations and to map the evolutionary trajectory of two individual combinatorial mutants in E. coli. Combinatorial mutations in the human ES2 ovarian carcinoma cell line were also assessed with TRACE. TRACE completes the combinatorial engineering cycle and enables more sophisticated approaches to genome engineering in both bacteria and eukaryotic cells than are currently possible.


Subject(s)
Escherichia coli/genetics , Genetic Engineering , Genetic Variation , Mutation/genetics , Genetic Association Studies , Genome, Bacterial , Genomics , High-Throughput Nucleotide Sequencing , Humans , Single-Cell Analysis
17.
Biotechnol J ; 8(12): 1445-51, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23894044

ABSTRACT

Predicting the structural consequences of site-specific glycosylation remains a major challenge due in part to the lack of convenient experimental tools for rapidly determining how glycosylation influences protein folding. To address this shortcoming, we developed a genetic selection that directly links the in vivo folding of asparagine-linked (N-linked) glycoproteins with antibiotic resistance. Using this assay, we identified three known or putative glycoproteins from Campylobacter jejuni (Peb3, CjaA, and Cj0610c) whose folding was significantly affected by N-glycosylation. We also used the genetic selection to isolate a glycoengineered variant of the Escherichia coli colicin E7 immunity protein (Im7) whose intracellular folding and stability were enhanced as a result of N-glycosylation. In addition to monitoring the effect of glycan attachment on protein folding in living cells, this strategy could easily be extended for optimizing protein folding in vivo and engineering glycosylation enzymes, pathways, and hosts for optimal performance.


Subject(s)
Asparagine/metabolism , Genetic Engineering/methods , Glycoproteins , Protein Folding , Protein Stability , Asparagine/chemistry , Asparagine/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation
18.
Methods Mol Biol ; 985: 223-46, 2013.
Article in English | MEDLINE | ID: mdl-23417807

ABSTRACT

Recent advances in homologous recombination in Escherichia coli have enabled improved genome engineering by multiplex recombineering. In this chapter, we present trackable multiplex recombineering (TRMR), a method for gene-trait mapping which creates simulated knockdown and overexpression mutants for virtually all genes in the E. coli genome. The method combines oligonucleotide synthesis with multiplex recombineering to create two libraries comprising of over 8,000 E. coli strains in total that can be selected for traits of interest via high-throughput screening or selection. DNA barcodes included in the recombineering cassette allow for rapid characterization of a naïve or selected population via DNA microarray analysis. Important considerations for oligonucleotide design, DNA library construction, recombineering, strain characterization, and selection are discussed.


Subject(s)
Escherichia coli/genetics , Genetic Engineering/methods , Base Sequence , Cloning, Molecular/methods , DNA Barcoding, Taxonomic , Escherichia coli Proteins/genetics , Gene Knockdown Techniques , Genome, Bacterial , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phenotype , Recombinant Proteins/genetics , Transcriptome
19.
Protein Sci ; 19(10): 2006-13, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20669235

ABSTRACT

We have developed a filamentous phage display system for the detection of asparagine-linked glycoproteins in Escherichia coli that carry a plasmid encoding the protein glycosylation locus (pgl) from Campylobacter jejuni. In our assay, fusion of target glycoproteins to the minor phage coat protein g3p results in the display of glycans on phage. The glyco-epitope displayed on phage is the product of biosynthetic enzymes encoded by the C. jejuni pgl pathway and minimally requires three essential factors: a pathway for oligosaccharide biosynthesis, a functional oligosaccharyltransferase, and an acceptor protein with a D/E-X(1)-N-X(2)-S/T motif. Glycosylated phages could be recovered by lectin chromatography with enrichment factors as high as 2 × 10(5) per round of panning and these enriched phages retained their infectivity after panning. Using this assay, we show that desired glyco-phenotypes can be reliably selected by panning phage-displayed glycoprotein libraries on lectins that are specific for the glycan. For instance, we used our phage selection to identify permissible residues in the -2 position of the bacterial consensus acceptor site sequence. Taken together, our results demonstrate that a genotype-phenotype link can be established between the phage-associated glyco-epitope and the phagemid-encoded genes for any of the three essential components of the glycosylation process. Thus, we anticipate that our phage display system can be used to isolate interesting variants in any step of the glycosylation process, thereby making it an invaluable tool for genetic analysis of protein glycosylation and for glycoengineering in E. coli cells.


Subject(s)
Glycoproteins/genetics , Inovirus/genetics , Peptide Library , Viral Proteins/genetics , Asparagine/genetics , Asparagine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Campylobacter jejuni/genetics , Campylobacter jejuni/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Glycoproteins/metabolism , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Inovirus/metabolism , Mutation , Oligosaccharides/metabolism , Plasmids/genetics , Polysaccharides/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Proteins/metabolism
20.
Protein Sci ; 19(5): 1079-90, 2010 May.
Article in English | MEDLINE | ID: mdl-20440843

ABSTRACT

An array of genetic screens and selections has been developed for reporting protein folding and solubility in the cytoplasm of living cells. However, there are currently no analogous folding assays for the bacterial periplasm, despite the significance of this compartment for the expression of recombinant proteins, especially those requiring important posttranslational modifications (e.g., disulfide bond formation). Here, we describe an engineered genetic selection for monitoring protein folding in the periplasmic compartment of Escherichia coli cells. In this approach, target proteins are sandwiched between an N-terminal signal recognition particle (SRP)-dependent signal peptide and a C-terminal selectable marker, TEM-1 beta-lactamase. The resulting chimeras are localized to the periplasmic space via the cotranslational SRP pathway. Using a panel of native and heterologous proteins, we demonstrate that the folding efficiency of various target proteins correlates directly with in vivo beta-lactamase activity and thus resistance to ampicillin. We also show that this reporter is useful for the discovery of extrinsic periplasmic factors (e.g., chaperones) that affect protein folding and for obtaining folding-enhanced proteins via directed evolution. Collectively, these data demonstrate that our periplasmic folding reporter is a powerful tool for screening and engineering protein folding in a manner that does not require any structural or functional information about the target protein.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli/genetics , Periplasm/metabolism , Protein Engineering/methods , Amino Acid Sequence , Ampicillin Resistance/genetics , Blotting, Western , Colony Count, Microbial , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Biological , Molecular Sequence Data , Oxidation-Reduction , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Folding , Protein Multimerization , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism , Solubility , beta-Lactamases/genetics , beta-Lactamases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL