Engineered Bacterial Outer Membrane Vesicles with Enhanced Functionality

https://doi.org/10.1016/j.jmb.2008.03.076Get rights and content

Abstract

We have engineered bacterial outer membrane vesicles (OMVs) with dramatically enhanced functionality by fusing several heterologous proteins to the vesicle-associated toxin ClyA of Escherichia coli. Similar to native unfused ClyA, chimeric ClyA fusion proteins were found localized in bacterial OMVs and retained activity of the fusion partners, demonstrating for the first time that ClyA can be used to co-localize fully functional heterologous proteins directly in bacterial OMVs. For instance, fusions of ClyA to the enzymes β-lactamase and organophosphorus hydrolase resulted in synthetic OMVs that were capable of hydrolyzing β-lactam antibiotics and paraoxon, respectively. Similarly, expression of an anti-digoxin single-chain Fv antibody fragment fused to the C terminus of ClyA resulted in designer “immuno-MVs” that could bind tightly and specifically to the antibody's cognate antigen. Finally, OMVs displaying green fluorescent protein fused to the C terminus of ClyA were highly fluorescent and, as a result of this new functionality, could be easily tracked during vesicle interaction with human epithelial cells. We expect that the relative plasticity exhibited by ClyA as a fusion partner should prove useful for: (i) further mechanistic studies to identify the vesiculation machinery that regulates OMV secretion and to map the intracellular routing of ClyA-containing OMVs during invasion of host cells; and (ii) biotechnology applications such as surface display of proteins and delivery of biologics.

Introduction

Extracellular secretion of virulence factors is a strategy utilized by invading bacteria to establish a colonization niche, communicate with host cells, and modulate host defense and response. With few exceptions, bacterial protein secretion systems are characterized by the membrane translocation of a single protein or else small protein complexes.1, 2, 3, 4, 5 Recently, however, the production and release of outer membrane vesicles (OMVs) has been demonstrated as a novel secretion mechanism for the transmission of a diverse group of proteins and lipids to mammalian cells.6 OMVs are small proteoliposomes with an average diameter of 50–200 nm that are constitutively released from the outer membrane of pathogenic and non-pathogenic species of Gram-negative bacteria during growth.7 Biochemical analysis has demonstrated that OMVs are composed of outer membrane proteins, lipopolysaccharide, phospholipids and soluble periplasmic proteins,8, 9 the latter of which become trapped in the vesicle lumen during release from the cell surface. OMVs are largely devoid of inner membrane and cytoplasm components, although several studies indicate that chromosomal, phage and plasmid DNA can infiltrate OMVs as a means of OMV-mediated transfer of genetic information between bacteria.10, 11, 12, 13

An intriguing yet poorly understood phenomenon pertaining to OMVs is the observation that certain membrane and/or soluble periplasmic proteins are enriched in vesicles while others are preferentially excluded. The majority of these enriched proteins happen to be virulence factors including, for example, Escherichia coli cytolysin A (ClyA),14 enterotoxigenic E. coli heat labile enterotoxin (LT),8 and Actinobacillus actinomycetemcomitans leukotoxin,15 whereas proteins that are excluded from OMVs include numerous unidentified outer membrane (OM) proteins15 as well as E. coli DsbA.14 The preferential exclusion of proteins raises the interesting possibility that a sorting mechanism exists in the bacterial periplasm for discriminatory loading of a highly specific subset of proteins into OMVs.14, 16 Moreover, the observation that certain virulence factors are enriched in vesicles suggests that OMVs may have a key role in bacterial pathogenesis by mediating transmission of active virulence factors and other bacterial envelope components to host cells. Indeed, numerous vesicle-associated virulence factors (e.g., adhesins, immunomodulatory compounds, proteases and toxins) have been shown to induce cytotoxicity, confer vesicle binding to and invasion of host cells, and modulate the host immune response.8, 17, 18, 19, 20

To date, one of the best studied vesicle-associated virulence factors is the 34 kDa cytotoxin ClyA (also called HlyE or SheA) found in pathogenic and non-pathogenic E. coli strains,14, 21 and in Salmonella enterica serovars Typhi and Paratyphi A.22 Structural studies indicate that the water-soluble form of ClyA is a bundle of four major α-helices, with a small surface-exposed hydrophobic β-hairpin at the “head” end of the structure, and the N and C termini at the “tail” end,23 while lipid-associated ClyA forms an oligomeric pore complex composed of either eight or 13 ClyA subunits.24, 25 Expression of the clyA gene is silenced in non-pathogenic E. coli K-12 laboratory strains by the nucleoid protein H-NS,26 but is derepressed in H-NS-deficient E. coli, thereby inducing cytotoxicity towards cultured mammalian cells.27 More recent evidence indicates that ClyA is exported from E. coli in OMVs and retains a cytolytically active, oligomeric conformation in the vesicles.14 However, the route by which ClyA crosses the bacterial inner membrane and assembles in OMVs remains a mystery, as it carries no canonical signal peptide,21 nor is it N-terminally processed.28 Also undetermined is the role that ClyA has in vesicle-mediated interactions with mammalian cells. Thus, in the present study, we sought to engineer synthetic membrane vesicles (s-MVs) with non-native functions that could be used for a wide range of applications, including the analysis of the complete ClyA translocation process. Specifically, we have programmed s-MVs with enhanced functionality by creating chimeras between heterologous proteins such as green fluorescent protein (GFP) or β-lactamase (Bla) and ClyA. Using these engineered vesicles, we have determined that ClyA is capable of co-localizing a variety of structurally diverse fusion partners to the surface of E. coli and their released vesicles, but only when the periplasmic disulfide bond-forming machinery was present. Importantly, these cell- and OMV-associated proteins retained their biological activity, suggesting that the functionality of natural OMVs can be expanded easily via the expression of ClyA chimeras.

Section snippets

GFP co-localizes in outer membrane vesicles when fused to ClyA

Previous studies demonstrated that genetic fusions between E. coli ClyA and reporter proteins such as Bla and GFP were translocated across the cytoplasmic membrane,29, 30 and that localization was independent of the position (N or C terminus) of ClyA in the fusion protein.29 Separately, Wai and co-workers demonstrated that ClyA was exported from laboratory strains of E. coli cells via OMVs composed of outer membrane and periplasm.31 These same authors reported that ClyA was significantly

Discussion

This work describes the development and characterization of engineered synthetic membrane vesicles (s-MVs) created by genetic fusion of a recombinant polypeptide with the E. coli cytotoxin ClyA. In general, it was observed that most recombinant polypeptide fusions co-localized with ClyA to the bacterial cell surface and into OMVs. Specifically, we demonstrated that direct fusion of Bla, OPH, GFP and anti-digoxin scFv to the C terminus of ClyA resulted in functional display of each protein on

Bacterial strains, plasmids and growth conditions

The bacterial strains and plasmids used in this study are described in Table 2. Strain JCA was made by introducing the dsbA∷Kan allele into JC8031 cells by P1 vir transduction using DHA as the donor. Plasmid pClyA was constructed by ligating the PCR-amplified clyA gene into pBAD18-Cm between SacI and XbaI sites. Insertion of DNA encoding either the gfpmut2 gene,51, 52 or a His6 sequence between XbaI and HindIII sites resulted in plasmids pGFP-ClyA and pClyA-His6, respectively. Plasmid pGFP-ClyA

Acknowledgements

We thank Kyung Hun Yoon (Cornell University) and Sang Hun Lee, Tai Hyun Park and Sung June Kim (Seoul National University) for their assistance with the SPR studies. We thank Harris Bernstein, Wilfred Chen, George Georgiou, Koreaki Ito, Roland Lloubes, Virginia Miller, Sun Nyunt Wai and Joel Weiner for their generous gifts of strains, plasmids and antibodies. This material is based upon work supported by the Ladies Auxiliary to the Veterans of Foreign Wars Cancer Research Fellowship (to

References (68)

  • del CastilloF.J. et al.

    Secretion of the Escherichia coli K-12 SheA hemolysin is independent of its cytolytic activity

    FEMS Microbiol. Lett.

    (2001)
  • KestyN.C. et al.

    Incorporation of heterologous outer membrane and periplasmic proteins into Escherichia coli outer membrane vesicles

    J. Biol. Chem.

    (2004)
  • AtkinsA. et al.

    Structure-function relationships of a novel bacterial toxin, hemolysin E. The role of alpha G

    J. Biol. Chem.

    (2000)
  • DeLisaM.P. et al.

    Genetic analysis of the twin arginine translocator secretion pathway in bacteria

    J. Biol. Chem.

    (2002)
  • ChoiS.H. et al.

    Enhanced performance of a surface plasmon resonance immunosensor for detecting Ab-GAD antibody based on the modified self-assembled monolayers

    Biosens. Bioelectron.

    (2005)
  • LeeJ.W. et al.

    Characterization of a self-assembled monolayer of thiol on a gold surface and the fabrication of a biosensor chip based on surface plasmon resonance for detecting anti-GAD antibody

    Biosens. Bioelectron.

    (2005)
  • QiH.Y. et al.

    DnaK promotes the selective export of outer membrane protein precursors in SecA-deficient Escherichia coli

    J. Biol. Chem.

    (2002)
  • GalanJ.E. et al.

    Type III secretion machines: bacterial devices for protein delivery into host cells

    Science

    (1999)
  • KuehnM.J. et al.

    Bacterial outer membrane vesicles and the host-pathogen interaction

    Genes Dev.

    (2005)
  • BeveridgeT.J.

    Structures of gram-negative cell walls and their derived membrane vesicles

    J. Bacteriol.

    (1999)
  • McBroomA. et al.

    Outer membrane vesicles

  • DorwardD.W. et al.

    Export and intercellular transfer of DNA via membrane blebs of Neisseria gonorrhoeae

    J. Bacteriol.

    (1989)
  • KollingG.L. et al.

    Export of virulence genes and Shiga toxin by membrane vesicles of Escherichia coli O157:H7

    Appl. Environ. Microbiol.

    (1999)
  • YaronS. et al.

    Vesicle-mediated transfer of virulence genes from Escherichia coli O157:H7 to other enteric bacteria

    Appl. Environ. Microbiol.

    (2000)
  • RenelliM. et al.

    DNA-containing membrane vesicles of Pseudomonas aeruginosa PAO1 and their genetic transformation potential

    Microbiology

    (2004)
  • McBroomA.J. et al.

    Release of outer membrane vesicles by Gram-negative bacteria is a novel envelope stress response

    Mol. Microbiol.

    (2007)
  • FioccaR. et al.

    Release of Helicobacter pylori vacuolating cytotoxin by both a specific secretion pathway and budding of outer membrane vesicles. Uptake of released toxin and vesicles by gastric epithelium

    J. Pathol.

    (1999)
  • KadurugamuwaJ.L. et al.

    Delivery of the non-membrane-permeative antibiotic gentamicin into mammalian cells by using Shigella flexneri membrane vesicles

    Antimicrob. Agents Chemother.

    (1998)
  • KestyN.C. et al.

    Enterotoxigenic Escherichia coli vesicles target toxin delivery into mammalian cells

    EMBO J.

    (2004)
  • del CastilloF.J. et al.

    The Escherichia coli K-12 sheA gene encodes a 34-kDa secreted haemolysin

    Mol. Microbiol.

    (1997)
  • OscarssonJ. et al.

    Characterization of a pore-forming cytotoxin expressed by Salmonella enterica serovars typhi and paratyphi A

    Infect. Immun.

    (2002)
  • EiflerN. et al.

    Cytotoxin ClyA from Escherichia coli assembles to a 13-meric pore independent of its redox-state

    EMBO J.

    (2006)
  • WestermarkM. et al.

    Silencing and activation of ClyA cytotoxin expression in Escherichia coli

    J. Bacteriol.

    (2000)
  • Gomez-GomezJ.M. et al.

    Hns mutant unveils the presence of a latent haemolytic activity in Escherichia coli K-12

    Mol. Microbiol.

    (1996)
  • Cited by (136)

    • Bacterial extracellular vesicle applications in cancer immunotherapy

      2023, Bioactive Materials
      Citation Excerpt :

      For instance, bacterial hemolysin toxin ClyA (SheA/HlyE) is reported to be enriched in OMV. Genetic fusion with ClyA can translocate heterologous proteins to OMV [67]. The capability of the periplasmic disulfide bond and its machinery in the OMVs facilitate ClyA to fuse with diverse fusion moieties [67].

    • Bacterial membrane vesicles for vaccine applications

      2022, Advanced Drug Delivery Reviews
      Citation Excerpt :

      Another method of introducing epitopes is to recombinantly fuse protein sequences to native proteins that are naturally expressed on the surface of BMVs [22]. This concept was demonstrated in E. coli by genetically fusing proteins to cytolysin A (ClyA), which is naturally found on the surface of the bacterial BMVs [41]. Taking advantage of this process, functional enzymes, fluorescent proteins, and antibodies were successfully expressed.

    View all citing articles on Scopus

    J.-Y. K. and A. M. D. contributed equally to this work.

    View full text