Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Metab Eng ; 53: 59-68, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30772453

RESUMO

Recombineering-based redesign of bacterial genomes by adding, removing or editing large segments of genomic DNA is emerging as a powerful technique for expanding the range of functions that an organism can perform. Here, we describe a glyco-recoding strategy whereby major non-essential polysaccharide gene clusters in K-12 Escherichia coli are replaced with orthogonal glycosylation components for both biosynthesis of heterologous glycan structures and site-specific glycan conjugation to target proteins. Specifically, the native enterobacterial common antigen (ECA) and O-polysaccharide (O-PS) antigen loci were systematically replaced with ∼9-10 kbp of synthetic DNA encoding Campylobacter jejuni enzymes required for asparagine-linked (N-linked) protein glycosylation. Compared to E. coli cells carrying the same glycosylation machinery on extrachromosomal plasmids, glyco-recoded strains attached glycans to acceptor protein targets with equal or greater efficiency while exhibiting markedly better growth phenotypes and higher glycoprotein titers. Overall, our results define a convenient and reliable framework for bacterial glycome editing that provides a more stable route for chemical diversification of proteins in vivo and effectively expands the bacterial glycoengineering toolkit.


Assuntos
Proteínas de Bactérias , Campylobacter jejuni/genética , Escherichia coli , Edição de Genes , Família Multigênica , Polissacarídeos Bacterianos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Polissacarídeos Bacterianos/biossíntese , Polissacarídeos Bacterianos/genética
2.
Metab Eng ; 47: 488-495, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29702274

RESUMO

A synthetic pathway for production of the eukaryotic trimannosyl chitobiose glycan (mannose3-N-acetylglucosamine2, Man3GlcNAc2) and its transfer to specific asparagine residues in target proteins was previously engineered in Escherichia coli, providing this simple microbe with the ability to perform a complex post-translational protein modification. Here, we leveraged a flow cytometric fluorescence-based assay to improve Man3GlcNAc2 glycan biosynthesis in E. coli cells. Specifically, pathway improvements were identified, including reducing pathway enzyme expression levels and overexpressing nucleotide sugar biosynthesis genes, which enhanced production of lipid-linked Man3GlcNAc2 by nearly 50-fold to 13.9 µg/L. In turn, cells producing higher levels of the Man3GlcNAc2 substrate yielded up to 10 times more glycosylated acceptor protein (to ~ 14 mg/L) than their non-optimized counterparts. These results demonstrate the use of flow cytometry screening as a powerful tool for interrogating the surfaces of glyco-engineered bacteria and identifying meaningful improvements in glycan biosynthesis. We anticipate this approach will enable further optimization of bacterial glycan biosynthesis pathways using new strain engineering tools from metabolic engineering and synthetic biology.


Assuntos
Escherichia coli , Citometria de Fluxo , Glucagon , Microrganismos Geneticamente Modificados , Proteínas Recombinantes de Fusão , Escherichia coli/citologia , Escherichia coli/genética , Escherichia coli/metabolismo , Engenharia Genética , Glucagon/genética , Glicosilação , Humanos , Microrganismos Geneticamente Modificados/citologia , Microrganismos Geneticamente Modificados/genética , Microrganismos Geneticamente Modificados/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética
3.
Glycobiology ; 26(4): 398-409, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26610891

RESUMO

Bacterial N-linking oligosaccharyl transferases (OTase enzymes) transfer lipid-linked glycans to selected proteins in the periplasm and were first described in the intestinal pathogen Campylobacter jejuni, a member of the ε-proteobacteria-subdivision of bacteria. More recently, orthologues from other ε-proteobacterial Campylobacter and Helicobacter species and a δ-proteobacterium, Desulfovibrio desulfuricans, have been described, suggesting that these two subdivisions of bacteria may be a source of further N-linked protein glycosylation systems. Whole-genome sequencing of both ε- and δ-proteobacteria from deep-sea vent habitats, a rich source of species from these subdivisions, revealed putative ORFs encoding OTase enzymes and associated adjacent glycosyltransferases similar to the C. jejuni N-linked glycosylation locus. We expressed putative OTase ORFs from the deep-sea vent species Nitratiruptor tergarcus, Sulfurovum lithotrophicum and Deferribacter desulfuricans in Escherichia coli and showed that they were able to functionally complement the C. jejuni OTase, CjPglB. The enzymes were shown to possess relaxed glycan specificity, transferring diverse glycan structures and demonstrated different glycosylation sequon specificities. Additionally, a permissive D. desulfuricans acceptor protein was identified, and we provide evidence that the N-linked glycan synthesized by N. tergarcus and S. lithotrophicum contains an acetylated sugar at the reducing end. This work demonstrates that deep-sea vent bacteria encode functional N-glycosylation machineries and are a potential source of biotechnologically important OTase enzymes.


Assuntos
Hexosiltransferases/genética , Proteínas de Membrana/genética , Polissacarídeos/metabolismo , Proteobactérias/genética , Escherichia coli/genética , Genoma Bacteriano , Glicosilação , Hexosiltransferases/biossíntese , Hexosiltransferases/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Oceanos e Mares , Polissacarídeos/biossíntese , Proteobactérias/enzimologia , Especificidade por Substrato
4.
Adv Biochem Eng Biotechnol ; 175: 167-200, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-30099598

RESUMO

Bacteria have garnered increased interest in recent years as a platform for the biosynthesis of a variety of glycomolecules such as soluble oligosaccharides, surface-exposed carbohydrates, and glycoproteins. The ability to engineer commonly used laboratory species such as Escherichia coli to efficiently synthesize non-native sugar structures by recombinant expression of enzymes from various carbohydrate biosynthesis pathways has allowed for the facile generation of important products such as conjugate vaccines, glycosylated outer membrane vesicles, and a variety of other research reagents for studying and understanding the role of glycans in living systems. This chapter highlights some of the key discoveries and technologies for equipping bacteria with the requisite biosynthetic machinery to generate such products. As the bacterial glyco-toolbox continues to grow, these technologies are expected to expand the range of glycomolecules produced recombinantly in bacterial systems, thereby opening up this platform to an even larger number of applications.


Assuntos
Glicoproteínas , Polissacarídeos , Bactérias/genética , Bactérias/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilação
5.
Nat Commun ; 9(1): 3396, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30127449

RESUMO

The original version of this Article contained an error in Figure 2, wherein the bottom right western blot panel in Figure 2a was blank. This has now been corrected in both the PDF and HTML versions of the Article.

6.
Nat Commun ; 9(1): 2686, 2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-30002445

RESUMO

The emerging discipline of bacterial glycoengineering has made it possible to produce designer glycans and glycoconjugates for use as vaccines and therapeutics. Unfortunately, cell-based production of homogeneous glycoproteins remains a significant challenge due to cell viability constraints and the inability to control glycosylation components at precise ratios in vivo. To address these challenges, we describe a novel cell-free glycoprotein synthesis (CFGpS) technology that seamlessly integrates protein biosynthesis with asparagine-linked protein glycosylation. This technology leverages a glyco-optimized Escherichia coli strain to source cell extracts that are selectively enriched with glycosylation components, including oligosaccharyltransferases (OSTs) and lipid-linked oligosaccharides (LLOs). The resulting extracts enable a one-pot reaction scheme for efficient and site-specific glycosylation of target proteins. The CFGpS platform is highly modular, allowing the use of multiple distinct OSTs and structurally diverse LLOs. As such, we anticipate CFGpS will facilitate fundamental understanding in glycoscience and make possible applications in on demand biomanufacturing of glycoproteins.


Assuntos
Escherichia coli/genética , Glicoproteínas/genética , Biossíntese de Proteínas/genética , Transcrição Gênica/genética , Biotecnologia/métodos , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Glicosilação , Hexosiltransferases/genética , Hexosiltransferases/metabolismo , Lipopolissacarídeos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Reprodutibilidade dos Testes
7.
Open Biol ; 6(4): 150243, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27110302

RESUMO

Currently, Streptococcus pneumoniae is responsible for over 14 million cases of pneumonia worldwide annually, and over 1 million deaths, the majority of them children. The major determinant for pathogenesis is a polysaccharide capsule that is variable and is used to distinguish strains based on their serotype. The capsule forms the basis of the pneumococcal polysaccharide vaccine (PPV23) that contains purified capsular polysaccharide from 23 serotypes, and the pneumococcal conjugate vaccine (PCV13), containing 13 common serotypes conjugated to CRM197 (mutant diphtheria toxin). Purified capsule from S. pneumoniae is required for pneumococcal conjugate vaccine production, and costs can be prohibitively high, limiting accessibility of the vaccine in low-income countries. In this study, we demonstrate the recombinant expression of the capsule-encoding locus from four different serotypes of S. pneumoniae within Escherichia coli. Furthermore, we attempt to identify the minimum set of genes necessary to reliably and efficiently express these capsules heterologously. These E. coli strains could be used to produce a supply of S. pneumoniae serotype-specific capsules without the need to culture pathogenic bacteria. Additionally, these strains could be applied to synthetic glycobiological applications: recombinant vaccine production using E. coli outer membrane vesicles or coupling to proteins using protein glycan coupling technology.


Assuntos
Cápsulas Bacterianas/metabolismo , Escherichia coli/metabolismo , Polissacarídeos Bacterianos/metabolismo , Recombinação Genética/genética , Streptococcus pneumoniae/metabolismo , Vias Biossintéticas/genética , Elementos de DNA Transponíveis/genética , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Genes Bacterianos , Loci Gênicos , Immunoblotting , Lipopolissacarídeos/metabolismo , Mutação/genética , Sorotipagem , Streptococcus pneumoniae/genética
8.
J Med Microbiol ; 61(Pt 7): 919-926, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22516134

RESUMO

The discovery of the Campylobacter jejuni N-linked glycosylation system combined with its functional expression in Escherichia coli marked the dawn of a new era in glycoengineering. The process, termed protein glycan coupling technology (PGCT), has, in particular, been applied to the development of glycoconjugate vaccines. In this review, we highlight recent technical developments in this area, including the first structural determination of the coupling enzyme PglB, the use of glycotags for optimal glycan attachment and the possible applications of other glycosylation systems and how these may improve and extend PGCT.


Assuntos
Vacinas Bacterianas/química , Vacinas Bacterianas/imunologia , Biotecnologia/métodos , Glicoconjugados/química , Glicoconjugados/imunologia , Tecnologia Farmacêutica/métodos , Campylobacter jejuni/enzimologia , Escherichia coli/genética , Humanos , Processamento de Proteína Pós-Traducional , Vacinas Conjugadas/química , Vacinas Conjugadas/imunologia , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA