1
|
Chen N, Jiang D, Liu Y, Zhang Z, Zhou Y, Zhu Z. Preparation of Escherichia coli ghost of anchoring bovine Pasteurella multocida OmpH and its immunoprotective effect. BMC Vet Res 2023; 19:192. [PMID: 37803295 PMCID: PMC10557220 DOI: 10.1186/s12917-023-03743-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 09/15/2023] [Indexed: 10/08/2023] Open
Abstract
Pasteurella multocida is a pathogen that can infect humans and animals. A ghost is an empty bacterial body devoid of cytoplasm and nucleic acids that can be efficiently presented by antigen-presenting cells. To study a novel ghost vector vaccine with cross-immune protection, we used bacteriophage PhiX174 RF1 and Pasteurella multocida standard strain CVCC393 as templates to amplify the split genes E and OmpH to construct a bidirectional expression vector E'-OmpH-pET28a-ci857-E. This is proposed to prepare a ghost Escherichia coli (engineered bacteria) capable of attaching and producing Pasteurella multocida OmpH on the inner membrane of Escherichia coli (BL21). The aim is to assess the antibody levels and the effectiveness of immune protection by conducting a mouse immunoprotective test. The bidirectional expression vector E'-OmpH-pET28a-ci857-E was successfully constructed. After induction by IPTG, identification by SDS-PAGE, western blot, ghost culture and transmission electron microscope detection, it was proven that the Escherichia coli ghost anchored to Pasteurella multocida OmpH was successfully prepared. The immunoprotective test in mice showed that the antibody levels of Pasteurella multocida inactivated vaccine, OmpH, ghost (aluminum glue adjuvant) and ghost (Freund's adjuvant) on day 9 after immunization were significantly different from those of the PBS control group (P < 0.01). The immune protection rates were 100%, 80%, 75%, and 65%, respectively, and the PBS negative control was 0%, which proved that they all had specific immune protection effects. Therefore, this study lays the foundation for the further study of ghosts as carriers of novel vaccine-presenting proteins.
Collapse
Affiliation(s)
- Nannan Chen
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Dongjun Jiang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, 272067, China
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272067, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing, 163319, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, 163319, China
| | - Zecai Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing, 163319, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, 163319, China
| | - Yulong Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing, 163319, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, 163319, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Daqing, 163319, China.
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, 163319, China.
| |
Collapse
|
2
|
Schuster B, Sleytr UB. S-Layer Ultrafiltration Membranes. MEMBRANES 2021; 11:275. [PMID: 33918014 PMCID: PMC8068369 DOI: 10.3390/membranes11040275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/30/2021] [Accepted: 04/03/2021] [Indexed: 11/29/2022]
Abstract
Monomolecular arrays of protein subunits forming surface layers (S-layers) are the most common outermost cell envelope components of prokaryotic organisms (bacteria and archaea). Since S-layers are periodic structures, they exhibit identical physicochemical properties for each constituent molecular unit down to the sub-nanometer level. Pores passing through S-layers show identical size and morphology and are in the range of ultrafiltration membranes. The functional groups on the surface and in the pores of the S-layer protein lattice are accessible for chemical modifications and for binding functional molecules in very precise fashion. S-layer ultrafiltration membranes (SUMs) can be produced by depositing S-layer fragments as a coherent (multi)layer on microfiltration membranes. After inter- and intramolecular crosslinking of the composite structure, the chemical and thermal resistance of these membranes was shown to be comparable to polyamide membranes. Chemical modification and/or specific binding of differently sized molecules allow the tuning of the surface properties and molecular sieving characteristics of SUMs. SUMs can be utilized as matrices for the controlled immobilization of functional biomolecules (e.g., ligands, enzymes, antibodies, and antigens) as required for many applications (e.g., biosensors, diagnostics, enzyme- and affinity-membranes). Finally, SUM represent unique supporting structures for stabilizing functional lipid membranes at meso- and macroscopic scale.
Collapse
Affiliation(s)
- Bernhard Schuster
- Institute for Synthetic Bioarchitectures, Department of NanoBiotechnology, BOKU—University of Natural Resources and Life Sciences, Vienna, Muthgasse 11, 1190 Vienna, Austria
| | - Uwe B. Sleytr
- Institute for Synthetic Bioarchitectures, Department of NanoBiotechnology, BOKU—University of Natural Resources and Life Sciences, Vienna, Muthgasse 11, 1190 Vienna, Austria
| |
Collapse
|
3
|
Mann VR, Manea F, Borys NJ, Ajo-Franklin CM, Cohen BE. Controlled and Stable Patterning of Diverse Inorganic Nanocrystals on Crystalline Two-Dimensional Protein Arrays. Biochemistry 2021; 60:1063-1074. [PMID: 33691067 PMCID: PMC8162747 DOI: 10.1021/acs.biochem.1c00032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Controlled patterning of nanoparticles on bioassemblies enables synthesis of complex materials for applications in optics, nanoelectronics, and sensing. Biomolecular self-assembly offers molecular control for engineering patterned nanomaterials, but current approaches have been limited in their ability to combine high nanoparticle coverage with generality that enables incorporation of multiple nanoparticle types. Here, we synthesize photonic materials on crystalline two-dimensional (2D) protein sheets using orthogonal bioconjugation reactions, organizing quantum dots (QDs), gold nanoparticles (AuNPs), and upconverting nanoparticles along the surface-layer (S-layer) protein SbsB from the extremophile Geobacillus stearothermophilus. We use electron and optical microscopy to show that isopeptide bond-forming SpyCatcher and SnoopCatcher systems enable the simultaneous and controlled conjugation of multiple types of nanoparticles (NPs) at high densities along the SbsB sheets. These NP conjugation reactions are orthogonal to each other and to Au-thiol bond formation, allowing tailorable nanoparticle combinations at sufficient labeling efficiencies to permit optical interactions between nanoparticles. Fluorescence lifetime imaging of SbsB sheets conjugated to QDs and AuNPs at distinct attachment sites shows spatially heterogeneous QD emission, with shorter radiative decays and brighter fluorescence arising from plasmonic enhancement at short interparticle distances. This specific, stable, and efficient conjugation of NPs to 2D protein sheets enables the exploration of interactions between pairs of nanoparticles at defined distances for the engineering of protein-based photonic nanomaterials.
Collapse
Affiliation(s)
- Victor R. Mann
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Francesca Manea
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
- Current address: Perfect Day Foods, Berkeley, CA 94608
| | - Nicholas J. Borys
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
- Current address: Department of Physics, Montana State University, Bozeman, MT 59717
| | - Caroline M. Ajo-Franklin
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
- Division of Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
- Synthetic Biology Institute, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
- Department of BioSciences, Rice University, Houston, TX 77005
| | - Bruce E. Cohen
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
- Division of Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| |
Collapse
|
4
|
Schuster B. S-Layer Protein-Based Biosensors. BIOSENSORS 2018; 8:E40. [PMID: 29641511 PMCID: PMC6023001 DOI: 10.3390/bios8020040] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 01/14/2023]
Abstract
The present paper highlights the application of bacterial surface (S-) layer proteins as versatile components for the fabrication of biosensors. One technologically relevant feature of S-layer proteins is their ability to self-assemble on many surfaces and interfaces to form a crystalline two-dimensional (2D) protein lattice. The S-layer lattice on the surface of a biosensor becomes part of the interface architecture linking the bioreceptor to the transducer interface, which may cause signal amplification. The S-layer lattice as ultrathin, highly porous structure with functional groups in a well-defined special distribution and orientation and an overall anti-fouling characteristics can significantly raise the limit in terms of variety and the ease of bioreceptor immobilization, compactness of bioreceptor molecule arrangement, sensitivity, specificity, and detection limit for many types of biosensors. The present paper discusses and summarizes examples for the successful implementation of S-layer lattices on biosensor surfaces in order to give a comprehensive overview on the application potential of these bioinspired S-layer protein-based biosensors.
Collapse
Affiliation(s)
- Bernhard Schuster
- Institute for Synthetic Bioarchitectures, Department of NanoBiotechnology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 11, 1190 Vienna, Austria.
| |
Collapse
|
5
|
Moudgil A, Kalyani N, Sinsinbar G, Das S, Mishra P. S-Layer Protein for Resistive Switching and Flexible Nonvolatile Memory Device. ACS APPLIED MATERIALS & INTERFACES 2018; 10:4866-4873. [PMID: 29308639 DOI: 10.1021/acsami.7b15062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this work, a flexible resistive switching memory device consisting of S-layer protein (Slp) is demonstrated for the first time. This novel device (Al/Slp/indium tin oxide/polyethylene terephthalte) based on a simple and easy fabrication method is capable of bistable switching to low resistive state (LRS) and high resistive state (HRS). This device exhibits bistable memory behavior with stability and a long retention time (>4 × 103 s), being stable up to a 500 cycle endurance test and with significant HRS/LRS ratio. The device possesses consistent switching performance for more than 100 times bending, corresponding to desired applicability for biocompatible wearable electronics. The memory mechanism is attributed to a trapping/de-trapping process in S-layer protein. These promising results of the flexible memory device could find a way in the wearable storage applications like smart bands and sports equipments' sensors.
Collapse
Affiliation(s)
- Akshay Moudgil
- Centre for Applied Research in Electronics and ‡Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi , Hauz Khas, New Delhi 110016, India
| | - Neeti Kalyani
- Centre for Applied Research in Electronics and ‡Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi , Hauz Khas, New Delhi 110016, India
| | - Gaurav Sinsinbar
- Centre for Applied Research in Electronics and ‡Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi , Hauz Khas, New Delhi 110016, India
| | - Samaresh Das
- Centre for Applied Research in Electronics and ‡Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi , Hauz Khas, New Delhi 110016, India
| | - Prashant Mishra
- Centre for Applied Research in Electronics and ‡Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi , Hauz Khas, New Delhi 110016, India
| |
Collapse
|
6
|
Alsuwyeh AA, Alanazi F, Shakeel F, Salem-Bekhit MM, Haq N. Estimation of Anti-neoplastic Drug Doxorubicin in Bacterial Ghost Matrix by New “Environmentally Benign” RP-HPLC Method: A Step Towards Sustainable Development of Pharmaceutical Industry. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2018; 43:181-190. [DOI: 10.1007/s13369-017-2664-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
7
|
Youssof AME, Salem-Bekhit MM, Shakeel F, Alanazi FK, Haq N. Analysis of anti-neoplastic drug in bacterial ghost matrix, w/o/w double nanoemulsion and w/o nanoemulsion by a validated 'green' liquid chromatographic method. Talanta 2016; 154:292-298. [PMID: 27154677 DOI: 10.1016/j.talanta.2016.03.086] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 11/30/2022]
Abstract
The objective of the present investigation was to develop and validate a 'green' reversed phase high-performance liquid chromatography (RP-HPLC) method for rapid analysis of a cytotoxic drug 5-fluorouracil (5-FU) in bulk drug, marketed injection, water-in-oil (w/o) nanoemulsion, double water-in-oil-in-water (w/o/w) nanoemulsion and bacterial ghost (BG) matrix. The chromatography study was carried out at room temperature (25±1°C) using an HPLC system with the help of ultraviolet (UV)-visible detector. The chromatographic performance was achieved with a Nucleodur 150mm×4.6mm RP C8 column filled with 5µm filler as a static phase. The mobile phase consisted of ethyl acetate: methanol (7:3% v/v) which was delivered at a flow rate of 1.0mLmin(-1) and the drug was detected in UV mode at 254nm. The developed method was validated in terms of linearity (r(2)=0.998), accuracy (98.19-102.09%), precision (% RSD=0.58-1.17), robustness (% RSD=0.12-0.53) and sensitivity with satisfactory results. The efficiency of the method was demonstrated by the assay of the drug in marketed injection, w/o nanoemulsion, w/o/w nanoemulsion and BG with satisfactory results. The successful resolution of the drug along with its degradation products clearly established the stability-indicating nature of the proposed method. Overall, these results suggested that the proposed analytical method could be effectively applied to the routine analysis of 5-FU in bulk drug, various pharmaceutical dosage forms and BG.
Collapse
Affiliation(s)
- Abdullah M E Youssof
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mounir M Salem-Bekhit
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Faiyaz Shakeel
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Center of Excellence in Biotechnology Research, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nazrul Haq
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; Center of Excellence in Biotechnology Research, King Saud University, P.O. Box 2460, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
8
|
Nguyen-Mau SM, Oh SY, Schneewind DI, Missiakas D, Schneewind O. Bacillus anthracis SlaQ Promotes S-Layer Protein Assembly. J Bacteriol 2015; 197:3216-27. [PMID: 26216847 PMCID: PMC4560277 DOI: 10.1128/jb.00492-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/22/2015] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Bacillus anthracis vegetative forms assemble an S-layer comprised of two S-layer proteins, Sap and EA1. A hallmark of S-layer proteins are their C-terminal crystallization domains, which assemble into a crystalline lattice once these polypeptides are deposited on the bacterial surface via association between their N-terminal S-layer homology domains and the secondary cell wall polysaccharide. Here we show that slaQ, encoding a small cytoplasmic protein conserved among pathogenic bacilli elaborating S-layers, is required for the efficient secretion and assembly of Sap and EA1. S-layer protein precursors cosediment with SlaQ, and SlaQ appears to facilitate Sap assembly. Purified SlaQ polymerizes and when mixed with purified Sap promotes the in vitro formation of tubular S-layer structures. A model is discussed whereby SlaQ, in conjunction with S-layer secretion factors SecA2 and SlaP, promotes localized secretion and S-layer assembly in B. anthracis. IMPORTANCE S-layer proteins are endowed with the propensity for self-assembly into crystalline arrays. Factors promoting S-layer protein assembly have heretofore not been reported. We identified Bacillus anthracis SlaQ, a small cytoplasmic protein that facilitates S-layer protein assembly in vivo and in vitro.
Collapse
Affiliation(s)
- Sao-Mai Nguyen-Mau
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - So-Young Oh
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Daphne I Schneewind
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Dominique Missiakas
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Olaf Schneewind
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
9
|
Schuster B, Sleytr UB. Biomimetic interfaces based on S-layer proteins, lipid membranes and functional biomolecules. J R Soc Interface 2014; 11:20140232. [PMID: 24812051 PMCID: PMC4032536 DOI: 10.1098/rsif.2014.0232] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/15/2014] [Indexed: 12/20/2022] Open
Abstract
Designing and utilization of biomimetic membrane systems generated by bottom-up processes is a rapidly growing scientific and engineering field. Elucidation of the supramolecular construction principle of archaeal cell envelopes composed of S-layer stabilized lipid membranes led to new strategies for generating highly stable functional lipid membranes at meso- and macroscopic scale. In this review, we provide a state-of-the-art survey of how S-layer proteins, lipids and polymers may be used as basic building blocks for the assembly of S-layer-supported lipid membranes. These biomimetic membrane systems are distinguished by a nanopatterned fluidity, enhanced stability and longevity and, thus, provide a dedicated reconstitution matrix for membrane-active peptides and transmembrane proteins. Exciting areas in the (lab-on-a-) biochip technology are combining composite S-layer membrane systems involving specific membrane functions with the silicon world. Thus, it might become possible to create artificial noses or tongues, where many receptor proteins have to be exposed and read out simultaneously. Moreover, S-layer-coated liposomes and emulsomes copying virus envelopes constitute promising nanoformulations for the production of novel targeting, delivery, encapsulation and imaging systems.
Collapse
Affiliation(s)
- Bernhard Schuster
- Department of NanoBiotechnology, University of Natural Resources and Life Sciences, Institute for Synthetic Bioarchitectures, Muthgasse 11, 1190 Vienna, Austria
| | - Uwe B. Sleytr
- Department of NanoBiotechnology, University of Natural Resources and Life Sciences, Institute for Biophysics, Muthgasse 11, 1190 Vienna, Austria
| |
Collapse
|
10
|
Baneyx F, Matthaei JF. Self-assembled two-dimensional protein arrays in bionanotechnology: from S-layers to designed lattices. Curr Opin Biotechnol 2013; 28:39-45. [PMID: 24832073 DOI: 10.1016/j.copbio.2013.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 10/30/2013] [Accepted: 11/01/2013] [Indexed: 02/04/2023]
Abstract
Although the crystalline S-layer arrays that form the exoskeleton of many archaea and bacteria have been studied for decades, a long-awaited crystal structure coupled with a growing understanding of the S-layer assembly process are injecting new excitement in the field. The trend is amplified by computational strategies that allow for in silico design of protein building blocks capable of self-assembling into 2D lattices and other prescribed quaternary structures. We review these and other recent developments toward achieving unparalleled control over the geometry, chemistry and function of protein-based 2D objects from the nanoscale to the mesoscale.
Collapse
Affiliation(s)
- François Baneyx
- Department of Chemical Engineering, University of Washington, Box 351750, Seattle, WA 98195-1750, USA.
| | - James F Matthaei
- Department of Chemical Engineering, University of Washington, Box 351750, Seattle, WA 98195-1750, USA
| |
Collapse
|
11
|
Khmelenina VN, Suzina NE, Trotsenko YA. Surface layers of methanotrophic bacteria. Microbiology (Reading) 2013. [DOI: 10.1134/s0026261713050068] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
12
|
Baranova E, Fronzes R, Garcia-Pino A, Van Gerven N, Papapostolou D, Péhau-Arnaudet G, Pardon E, Steyaert J, Howorka S, Remaut H. SbsB structure and lattice reconstruction unveil Ca2+ triggered S-layer assembly. Nature 2012; 487:119-22. [PMID: 22722836 DOI: 10.1038/nature11155] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 04/19/2012] [Indexed: 11/09/2022]
Abstract
S-layers are regular two-dimensional semipermeable protein layers that constitute a major cell-wall component in archaea and many bacteria. The nanoscale repeat structure of the S-layer lattices and their self-assembly from S-layer proteins (SLPs) have sparked interest in their use as patterning and display scaffolds for a range of nano-biotechnological applications. Despite their biological abundance and the technological interest in them, structural information about SLPs is limited to truncated and assembly-negative proteins. Here we report the X-ray structure of the SbsB SLP of Geobacillus stearothermophilus PV72/p2 by the use of nanobody-aided crystallization. SbsB consists of a seven-domain protein, formed by an amino-terminal cell-wall attachment domain and six consecutive immunoglobulin-like domains, that organize into a φ-shaped disk-like monomeric crystallization unit stabilized by interdomain Ca(2+) ion coordination. A Ca(2+)-dependent switch to the condensed SbsB quaternary structure pre-positions intermolecular contact zones and renders the protein competent for S-layer assembly. On the basis of crystal packing, chemical crosslinking data and cryo-electron microscopy projections, we present a model for the molecular organization of this SLP into a porous protein sheet inside the S-layer. The SbsB lattice represents a previously undescribed structural model for protein assemblies and may advance our understanding of SLP physiology and self-assembly, as well as the rational design of engineered higher-order structures for biotechnology.
Collapse
Affiliation(s)
- Ekaterina Baranova
- Structural and Molecular Microbiology, VIB Department of Structural Biology, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Langemann T, Koller VJ, Muhammad A, Kudela P, Mayr UB, Lubitz W. The Bacterial Ghost platform system: production and applications. Bioeng Bugs 2012; 1:326-36. [PMID: 21326832 DOI: 10.4161/bbug.1.5.12540] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 06/01/2010] [Accepted: 06/01/2010] [Indexed: 12/23/2022] Open
Abstract
The Bacterial Ghost (BG) platform technology is an innovative system for vaccine, drug or active substance delivery and for technical applications in white biotechnology. BGs are cell envelopes derived from Gram-negative bacteria. BGs are devoid of all cytoplasmic content but have a preserved cellular morphology including all cell surface structures. Using BGs as delivery vehicles for subunit or DNA-vaccines the particle structure and surface properties of BGs are targeting the carrier itself to primary antigen-presenting cells. Furthermore, BGs exhibit intrinsic adjuvant properties and trigger an enhanced humoral and cellular immune response to the target antigen. Multiple antigens of the native BG envelope and recombinant protein or DNA antigens can be combined in a single type of BG. Antigens can be presented on the inner or outer membrane of the BG as well as in the periplasm that is sealed during BG formation. Drugs or supplements can also be loaded to the internal lumen or periplasmic space of the carrier. BGs are produced by batch fermentation with subsequent product recovery and purification via tangential flow filtration. For safety reasons all residual bacterial DNA is inactivated during the BG production process by the use of staphylococcal nuclease A and/or the treatment with β-propiolactone. After purification BGs can be stored long-term at ambient room temperature as lyophilized product. The production cycle from the inoculation of the pre-culture to the purified BG concentrate ready for lyophilization does not take longer than a day and thus meets modern criteria of rapid vaccine production rather than keeping large stocks of vaccines. The broad spectrum of possible applications in combination with the comparably low production costs make the BG platform technology a safe and sophisticated product for the targeted delivery of vaccines and active agents as well as carrier of immobilized enzymes for applications in white biotechnology.
Collapse
|
14
|
Horejs C, Ristl R, Tscheliessnig R, Sleytr UB, Pum D. Single-molecule force spectroscopy reveals the individual mechanical unfolding pathways of a surface layer protein. J Biol Chem 2011; 286:27416-24. [PMID: 21690085 PMCID: PMC3149335 DOI: 10.1074/jbc.m111.251322] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 06/15/2011] [Indexed: 12/14/2022] Open
Abstract
Surface layers (S-layers) represent an almost universal feature of archaeal cell envelopes and are probably the most abundant bacterial cell proteins. S-layers are monomolecular crystalline structures of single protein or glycoprotein monomers that completely cover the cell surface during all stages of the cell growth cycle, thereby performing their intrinsic function under a constant intra- and intermolecular mechanical stress. In gram-positive bacteria, the individual S-layer proteins are anchored by a specific binding mechanism to polysaccharides (secondary cell wall polymers) that are linked to the underlying peptidoglycan layer. In this work, atomic force microscopy-based single-molecule force spectroscopy and a polyprotein approach are used to study the individual mechanical unfolding pathways of an S-layer protein. We uncover complex unfolding pathways involving the consecutive unfolding of structural intermediates, where a mechanical stability of 87 pN is revealed. Different initial extensibilities allow the hypothesis that S-layer proteins adapt highly stable, mechanically resilient conformations that are not extensible under the presence of a pulling force. Interestingly, a change of the unfolding pathway is observed when individual S-layer proteins interact with secondary cell wall polymers, which is a direct signature of a conformational change induced by the ligand. Moreover, the mechanical stability increases up to 110 pN. This work demonstrates that single-molecule force spectroscopy offers a powerful tool to detect subtle changes in the structure of an individual protein upon binding of a ligand and constitutes the first conformational study of surface layer proteins at the single-molecule level.
Collapse
Affiliation(s)
| | - Robin Ristl
- From the Department for Nanobiotechnology and
| | - Rupert Tscheliessnig
- the Austrian Centre of Industrial Biotechnology, c/o Institute for Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
| | | | - Dietmar Pum
- From the Department for Nanobiotechnology and
| |
Collapse
|
15
|
Horejs C, Mitra MK, Pum D, Sleytr UB, Muthukumar M. Monte Carlo study of the molecular mechanisms of surface-layer protein self-assembly. J Chem Phys 2011; 134:125103. [PMID: 21456703 DOI: 10.1063/1.3565457] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The molecular mechanisms guiding the self-assembly of proteins into functional or pathogenic large-scale structures can be only understood by studying the correlation between the structural details of the monomer and the eventual mesoscopic morphologies. Among the myriad structural details of protein monomers and their manifestations in the self-assembled morphologies, we seek to identify the most crucial set of structural features necessary for the spontaneous selection of desired morphologies. Using a combination of the structural information and a Monte Carlo method with a coarse-grained model, we have studied the functional protein self-assembly into S(surface)-layers, which constitute the crystallized outer most cell envelope of a great variety of bacterial cells. We discover that only few and mainly hydrophobic amino acids, located on the surface of the monomer, are responsible for the formation of a highly ordered anisotropic protein lattice. The coarse-grained model presented here reproduces accurately many experimentally observed features including the pore formation, chemical description of the pore structure, location of specific amino acid residues at the protein-protein interfaces, and surface accessibility of specific amino acid residues. In addition to elucidating the molecular mechanisms and explaining experimental findings in the S-layer assembly, the present work offers a tool, which is chemical enough to capture details of primary sequences and coarse-grained enough to explore morphological structures with thousands of protein monomers, to promulgate design rules for spontaneous formation of specific protein assemblies.
Collapse
Affiliation(s)
- Christine Horejs
- Department for Nanobiotechnology, University of Natural Resources and Applied Life Sciences, 1190 Vienna, Austria
| | | | | | | | | |
Collapse
|
16
|
Horejs C, Gollner H, Pum D, Sleytr UB, Peterlik H, Jungbauer A, Tscheliessnig R. Atomistic structure of monomolecular surface layer self-assemblies: toward functionalized nanostructures. ACS NANO 2011; 5:2288-2297. [PMID: 21375257 DOI: 10.1021/nn1035729] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The concept of self-assembly is one of the most promising strategies for the creation of defined nanostructures and therefore became an essential part of nanotechnology for the controlled bottom-up design of nanoscale structures. Surface layers (S-layers), which represent the cell envelope of a great variety of prokaryotic cells, show outstanding self-assembly features in vitro and have been successfully used as the basic matrix for molecular construction kits. Here we present the three-dimensional structure of an S-layer lattice based on tetrameric unit cells, which will help to facilitate the directed binding of various molecules on the S-layer lattice, thereby creating functional nanoarrays for applications in nanobiotechnology. Our work demonstrates the successful combination of computer simulations, electron microscopy (TEM), and small-angle X-ray scattering (SAXS) as a tool for the investigation of the structure of self-assembling or aggregating proteins, which cannot be determined by X-ray crystallography. To the best of our knowledge, this is the first structural model at an amino acid level of an S-layer unit cell that exhibits p4 lattice symmetry.
Collapse
Affiliation(s)
- Christine Horejs
- Department for Nanobiotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
17
|
Horejs C, Pum D, Sleytr UB, Peterlik H, Jungbauer A, Tscheliessnig R. Surface layer protein characterization by small angle x-ray scattering and a fractal mean force concept: from protein structure to nanodisk assemblies. J Chem Phys 2011; 133:175102. [PMID: 21054069 DOI: 10.1063/1.3489682] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Surface layers (S-layers) are the most commonly observed cell surface structure of prokaryotic organisms. They are made up of proteins that spontaneously self-assemble into functional crystalline lattices in solution, on various solid surfaces, and interfaces. While classical experimental techniques failed to recover a complete structural model of an unmodified S-layer protein, small angle x-ray scattering (SAXS) provides an opportunity to study the structure of S-layer monomers in solution and of self-assembled two-dimensional sheets. For the protein under investigation we recently suggested an atomistic structural model by the use of molecular dynamics simulations. This structural model is now refined on the basis of SAXS data together with a fractal assembly approach. Here we show that a nondiluted critical system of proteins, which crystallize into monomolecular structures, might be analyzed by SAXS if protein-protein interactions are taken into account by relating a fractal local density distribution to a fractal local mean potential, which has to fulfill the Poisson equation. The present work demonstrates an important step into the elucidation of the structure of S-layers and offers a tool to analyze the structure of self-assembling systems in solution by means of SAXS and computer simulations.
Collapse
Affiliation(s)
- Christine Horejs
- Department for Nanobiotechnology, University of Natural Resources and Applied Life Sciences, 1090 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
18
|
Sleytr UB, Schuster B, Egelseer EM, Pum D, Horejs CM, Tscheliessnig R, Ilk N. Nanobiotechnology with S-layer proteins as building blocks. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 103:277-352. [PMID: 21999999 DOI: 10.1016/b978-0-12-415906-8.00003-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
One of the key challenges in nanobiotechnology is the utilization of self- assembly systems, wherein molecules spontaneously associate into reproducible aggregates and supramolecular structures. In this contribution, we describe the basic principles of crystalline bacterial surface layers (S-layers) and their use as patterning elements. The broad application potential of S-layers in nanobiotechnology is based on the specific intrinsic features of the monomolecular arrays composed of identical protein or glycoprotein subunits. Most important, physicochemical properties and functional groups on the protein lattice are arranged in well-defined positions and orientations. Many applications of S-layers depend on the capability of isolated subunits to recrystallize into monomolecular arrays in suspension or on suitable surfaces (e.g., polymers, metals, silicon wafers) or interfaces (e.g., lipid films, liposomes, emulsomes). S-layers also represent a unique structural basis and patterning element for generating more complex supramolecular structures involving all major classes of biological molecules (e.g., proteins, lipids, glycans, nucleic acids, or combinations of these). Thus, S-layers fulfill key requirements as building blocks for the production of new supramolecular materials and nanoscale devices as required in molecular nanotechnology, nanobiotechnology, biomimetics, and synthetic biology.
Collapse
Affiliation(s)
- Uwe B Sleytr
- Department of NanoBiotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
19
|
The Structure of Bacterial S-Layer Proteins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 103:73-130. [DOI: 10.1016/b978-0-12-415906-8.00004-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
20
|
Badelt-Lichtblau H, Kainz B, Völlenkle C, Egelseer EM, Sleytr UB, Pum D, Ilk N. Genetic engineering of the S-layer protein SbpA of Lysinibacillus sphaericus CCM 2177 for the generation of functionalized nanoarrays. Bioconjug Chem 2010; 20:895-903. [PMID: 19402706 DOI: 10.1021/bc800445r] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The mesophilic organism Lysinibacillus sphaericus CCM 2177 produces the surface (S)-layer protein SbpA, which after secretion completely covers the cell surface with a crystalline array exhibiting square lattice symmetry. Because of its excellent in vitro recrystallization properties on solid supports, SbpA represents a suitable candidate for genetically engineering to create a versatile self-assembly system for the development of a molecular construction kit for nanobiotechnological applications. The first goal of this study was to investigate the surface location of 3 different C-terminal amino acid positions within the S-layer lattice formed by SbpA. Therefore, three derivatives of SbpA were constructed, in which 90, 173, or 200 C-terminal amino acids were deleted, and the sequence encoding the short affinity tag Strep-tag II as well as a single cysteine residue were fused to their C-terminal end. Recrystallization studies of the rSbpA/STII/Cys fusion proteins indicated that C-terminal truncation and functionalization of the S-layer protein did not interfere with the self-assembly capability. Fluorescent labeling demonstrated that the orientation of the crystalline rSbpA(31-1178)/STII/Cys lattice on solid supports was the same, like the orientation of wild-type S-layer protein SbpA on the bacterial cell. In soluble and recrystallized rSbpA/STII/Cys fusion proteins, Strep-tag II was used for prescreening of the surface accessibility, whereas the thiol group of the end-standing cysteine residue was exploited for site-directed chemical linkage of differently sized preactivated macromolecules via heterobifunctional cross-linkers. Finally, functionalized two-dimensional S-layer lattices formed by rSbpA(31-1178)/STII/Cys exhibiting highly accessible cysteine residues in a well-defined arrangement on the surface were utilized for the template-assisted patterning of gold nanoparticles.
Collapse
Affiliation(s)
- Helga Badelt-Lichtblau
- Center for NanoBiotechnology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
21
|
Cui X, Duckworth JA, Lubitz P, Molinia FC, Haller C, Lubitz W, Cowan PE. Humoral immune responses in brushtail possums (Trichosurus vulpecula) induced by bacterial ghosts expressing possum zona pellucida 3 protein. Vaccine 2010; 28:4268-74. [DOI: 10.1016/j.vaccine.2010.04.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2006] [Revised: 04/12/2010] [Accepted: 04/15/2010] [Indexed: 10/19/2022]
|
22
|
Kinns H, Badelt-Lichtblau H, Egelseer EM, Sleytr UB, Howorka S. Identifying assembly-inhibiting and assembly-tolerant sites in the SbsB S-layer protein from Geobacillus stearothermophilus. J Mol Biol 2009; 395:742-53. [PMID: 19836402 DOI: 10.1016/j.jmb.2009.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2009] [Revised: 10/07/2009] [Accepted: 10/08/2009] [Indexed: 10/20/2022]
Abstract
Surface layer (S-layer) proteins self-assemble into two-dimensional crystalline lattices that cover the cell wall of all archaea and many bacteria. We have generated assembly-negative protein variants of high solubility that will facilitate high-resolution structure determination. Assembly-negative versions of the S-layer protein SbsB from Geobacillus stearothermophilus PV72/p2 were obtained using an insertion mutagenesis screen. The haemagglutinin epitope tag was inserted at 23 amino acid positions known to be located on the monomer protein surface from a previous cysteine accessibility screen. Limited proteolysis, circular dichroism, and fluorescence were used to probe whether the epitope insertion affected the secondary and tertiary structures of the monomer, while electron microscopy and size-exclusion chromatography were employed to examine proteins' ability to self-assemble. The screen not only identified assembly-compromised mutants with native fold but also yielded correctly folded, self-assembling mutants suitable for displaying epitopes for biomedical and biophysical applications, as well as cryo-electron microscopy imaging. Our study marks an important step in the analysis of the S-layer structure. In addition, the approach of concerted insertion and cysteine mutagenesis can likely be applied for other supramolecular assemblies.
Collapse
Affiliation(s)
- Helen Kinns
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | | | | | | | | |
Collapse
|
23
|
Surface location of individual residues of SlpA provides insight into the Lactobacillus brevis S-layer. J Bacteriol 2009; 191:3339-49. [PMID: 19304849 DOI: 10.1128/jb.01782-08] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial surface layer (S-layer) proteins are excellent candidates for in vivo and in vitro nanobiotechnological applications because of their ability to self-assemble into two-dimensional lattices that form the outermost layer of many Eubacteria and most Archaea species. Despite this potential, knowledge about their molecular architecture is limited. In this study, we investigated SlpA, the S-layer protein of the potentially probiotic bacterium Lactobacillus brevis ATCC 8287 by cysteine-scanning mutagenesis and chemical modification. We generated a series of 46 mutant proteins by replacing single amino acids with cysteine, which is not present in the wild-type protein. Most of the replaced amino acids were located in the self-assembly domain (residues 179 to 435) that likely faces the outer surface of the lattice. As revealed by electron microscopy, all the mutant proteins were able to form self-assembly products identical to that of the wild type, proving that this replacement does not dramatically alter the protein conformation. The surface accessibility of the sulfhydryl groups introduced was studied with two maleimide-containing marker molecules, TMM(PEG)(12) (molecular weight [MW], 2,360) and AlexaFluor488-maleimide (MW = 720), using both monomeric proteins in solution and proteins allowed to self-assemble on cell wall fragments. Using the acquired data and available domain information, we assigned the mutated residues into four groups according to their location in the protein monomer and lattice structure: outer surface of the lattice (9 residues), inner surface of the lattice (9), protein interior (12), and protein-protein interface/pore regions (16). This information is essential, e.g., in the development of therapeutic and other health-related applications of Lactobacillus S-layers.
Collapse
|
24
|
Anyatonwu G, Joseph SK. Surface accessibility and conformational changes in the N-terminal domain of type I inositol trisphosphate receptors: studies using cysteine substitution mutagenesis. J Biol Chem 2009; 284:8093-102. [PMID: 19141613 DOI: 10.1074/jbc.m806932200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To identify surface-accessible residues and monitor conformational changes of the type I inositol 1,4,5-trisphosphate receptor protein in membranes, we have introduced 10 cysteine substitutions into the N-terminal ligand-binding domain. The reactivity of these mutants with progressively larger maleimide-polyethylene glycol derivatives (MPEG) was measured using a gel shift assay of tryptic fragments. The results indicate that the mutations fall into four categories as follows: sites that are highly accessible based on reactivity with the largest 20-kDa MPEG (S2C); sites that are moderately accessible based on reactivity only with 5-kDa MPEG (S6C, S7C, A189C, and S277C); sites whose accessibility is markedly enhanced by Ca(2+) (S171C, S277C, and A575C); and sites that are inaccessible irrespective of incubation conditions (S217C, A245C, and S436C). The stimulation of accessibility induced by Ca(2+) at the S277C site occurred with an EC(50) of 0.8 mum and was mimicked by Sr(2+) but not Ba(2+). Inositol 1,4,5-trisphosphate alone did not affect reactivity of any of the mutants in the presence or absence of Ca(2+). The data are interpreted using crystal structures and EM reconstructions of the receptor. Our data identify N-terminal regions of the protein that become exposed upon Ca(2+) binding and suggest possible orientations of the suppressor and ligand-binding domains that have implications for the mechanism of gating of the channel.
Collapse
Affiliation(s)
- Georgia Anyatonwu
- Department of Pathology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
25
|
Tschiggerl H, Casey JL, Parisi K, Foley M, Sleytr UB. Display of a peptide mimotope on a crystalline bacterial cell surface layer (S-layer) lattice for diagnosis of Epstein-Barr virus infection. Bioconjug Chem 2008; 19:860-5. [PMID: 18376854 DOI: 10.1021/bc7003523] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fusion proteins based on the crystalline bacterial cell surface layer (S-layer) proteins SbpA from Bacillus sphaericus CCM 2177 and SbsB from Geobacillus stearothermophilus PV72/p2 and a peptide mimotope F1 that mimics an immunodominant epitope of Epstein-Barr virus (EBV) were designed and overexpressed in Escherichia coli. Constructs were designed such that the peptide mimotope was presented either at the C-terminus (SbpA/F1) or at the N-terminus (SbsB/F1) of the respective S-layer proteins. The resulting S-layer fusion proteins, SbpA/F1 and SbsB/F1, fully retained the intrinsic self-assembly capability of the S-layer moiety into monomolecular lattices. As determined by immunodot assays and ELISAs using monoclonal antibodies, the F1 mimotope was well-presented on the outer surface of the S-layer lattices and accessible for antibody binding. Further comparison of the two S-layer fusion proteins showed that the S-layer fusion protein SbpA/F1 had a higher antibody binding capacity than SbsB/F1 in aqueous solution and in immune sera, illustrating the importance of epitope orientation on the performance of solid-phase immunoassays. To assess the diagnostic values of S-layer mimotope fusion protein SbpA/F1, we screened a panel of 83 individual EBV IgM-positive, EBV negative, and potential cross-reactive sera for their reactivities. This resulted in 98.2% specificity and 89.3% sensitivity, and furthermore no cross-reactivity with related viral disease states including rheumatoid factor was observed. This study shows the potential of S-layer fusion proteins as a matrix for site-directed immobilization of small ligands in solid-phase immunoassays using EBV diagnostics as a model system.
Collapse
Affiliation(s)
- Helga Tschiggerl
- Center for NanoBiotechnology, University of Natural Resources and Applied Life Sciences, Gregor-Mendel-Strasse 33, A-1180 Vienna, Austria
| | | | | | | | | |
Collapse
|
26
|
Kinns H, Howorka S. The surface location of individual residues in a bacterial S-layer protein. J Mol Biol 2008; 377:589-604. [PMID: 18262545 DOI: 10.1016/j.jmb.2008.01.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 12/22/2007] [Accepted: 01/08/2008] [Indexed: 10/22/2022]
Abstract
Bacterial surface layer (S-layer) proteins self-assemble into large two-dimensional crystalline lattices that form the outermost cell-wall component of all archaea and many eubacteria. Despite being a large class of self-assembling proteins, little is known about their molecular architecture. We investigated the S-layer protein SbsB from Geobacillus stearothermophilus PV72/p2 to identify residues located at the subunit-subunit interface and to determine the S-layer's topology. Twenty-three single cysteine mutants, which were previously mapped to the surface of the SbsB monomer, were subjected to a cross-linking screen using the photoactivatable, sulfhydryl-reactive reagent N-[4-(p-azidosalicylamido)butyl]-3'-(2'-pyridyldithio)propionamide. Gel electrophoretic analysis on the formation of cross-linked dimers indicated that 8 out of the 23 residues were located at the interface. In combination with surface accessibility data for the assembled protein, 10 residues were assigned to positions at the inner, cell-wall-facing lattice surface, while 5 residues were mapped to the outer, ambient-exposed lattice surface. In addition, the cross-linking screen identified six positions of intramolecular cross-linking within the assembled protein but not in the monomeric S-layer protein. Most likely, these intramolecular cross-links result from conformational changes upon self-assembly. The results are an important step toward the further structural elucidation of the S-layer protein via, for example, X-ray crystallography and cryo-electron microscopy. Our approach of identifying the surface location of residues is relevant to other planar supramolecular protein assemblies.
Collapse
Affiliation(s)
- Helen Kinns
- Department of Chemistry, University College London, Christopher Ingold Building, 20 Gordon Street, London WC1H 0AJ, England, UK
| | | |
Collapse
|
27
|
Sleytr UB, Huber C, Ilk N, Pum D, Schuster B, Egelseer EM. S-layers as a tool kit for nanobiotechnological applications. FEMS Microbiol Lett 2007; 267:131-44. [PMID: 17328112 DOI: 10.1111/j.1574-6968.2006.00573.x] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Crystalline bacterial cell surface layers (S-layers) have been identified in a great number of different species of bacteria and represent an almost universal feature of archaea. Isolated native S-layer proteins and S-layer fusion proteins incorporating functional sequences self-assemble into monomolecular crystalline arrays in suspension, on a great variety of solid substrates and on various lipid structures including planar membranes and liposomes. S-layers have proven to be particularly suited as building blocks and patterning elements in a biomolecular construction kit involving all major classes of biological molecules (proteins, lipids, glycans, nucleic acids and combinations of them) enabling innovative approaches for the controlled 'bottom-up' assembly of functional supramolecular structures and devices. Here, we review the basic principles of S-layer proteins and the application potential of S-layers in nanobiotechnology and biomimetics including life and nonlife sciences.
Collapse
Affiliation(s)
- Uwe B Sleytr
- Center for NanoBiotechnology, University of Natural Resources and Applied Life Sciences Vienna, Gregor Mendel Strasse 33, A-1180 Vienna, Austria.
| | | | | | | | | | | |
Collapse
|
28
|
Howorka S. Creating regular arrays of nanoparticles with self-assembling protein building blocks. ACTA ACUST UNITED AC 2007. [DOI: 10.1039/b701221g] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
29
|
Sleytr UB, Egelseer EM, Ilk N, Pum D, Schuster B. S-Layers as a basic building block in a molecular construction kit. FEBS J 2006; 274:323-34. [PMID: 17181542 DOI: 10.1111/j.1742-4658.2006.05606.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Crystalline arrays of protein or glycoprotein subunits forming surface layers (S-layers) are the most common outermost envelope components of prokaryotic organisms (archaea and bacteria). The wealth of information on the structure, chemistry, genetics, morphogenesis, and function of S-layers has revealed a broad application potential. As S-layers are periodic structures, they exhibit identical physicochemical properties for each molecular unit down to the subnanometer level and possess pores of identical size and morphology. Many applications of S-layers in nanobiotechnology depend on the ability of isolated subunits to recrystallize into monomolecular lattices in suspension or on suitable surfaces and interfaces. S-Layer lattices can be exploited as scaffolding and patterning elements for generating more complex supramolecular assemblies and structures, as required for life and nonlife science applications.
Collapse
Affiliation(s)
- Uwe B Sleytr
- Center for NanoBiotechnology, University of Natural Resources and Applied Life Sciences Vienna, Austria.
| | | | | | | | | |
Collapse
|
30
|
Steffens S, Oldendorf J, Haufe G, Galla HJ. Organized collapse structures in mixtures of chiral ethyl 2-azido-4-fluoro-3-hydroxystearates. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2006; 22:1428-35. [PMID: 16460057 DOI: 10.1021/la0521589] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Monolayers of enantiomeric compounds as well as diastereomeric mixtures and racemic/diastereomeric mixtures of ethyl 2-azido-4-fluoro-3-hydroxystearates have been investigated using surface pressure-area isotherms and Brewster angle microscopy. All monolayers collapse out of the liquid-expanded phase, forming 3D collapse structures which were visualized with scanning force microscopy. The enantiomeric compound and the diastereomeric mixtures form unique fiber-like network structures with heights between 20 and 40 nm. Interestingly, the shape of the enantiomeric fiber structures is straight, whereas the diastereomeric mixtures exhibit curved fibers of different sizes. The racemic mixture however forms circular 10 nm high and 20-50 microm broad structures. The shape of unconventional collapse structures could be changed by using distinct ratios of diastereomeric or racemic/diastereomeric mixed compounds.
Collapse
Affiliation(s)
- Silke Steffens
- Institut für Biochemie der Universität Münster, Wilhelm-Klemm Strasse 2, Münster, Germany
| | | | | | | |
Collapse
|
31
|
Abstract
Gene fusion technology is a key tool in facilitating gene function studies. Hybrid molecules in which all the components are joined precisely, without the presence of intervening and unwanted extraneous sequences, enable accurate studies of molecules and the characterization of individual components. This article reviews situations in which seamlessly fused genes and proteins are required or desired and describes molecular approaches that are available for generating these hybrid molecules.
Collapse
Affiliation(s)
- Quinn Lu
- Gene Expression and Protein Biochemistry, Discovery Research, GlaxoSmithKline, Mail Code: UE0548, 709 Swedeland Road, King of Prussia, PA 19406, USA.
| |
Collapse
|
32
|
Candela T, Mignot T, Hagnerelle X, Haustant M, Fouet A. Genetic analysis of Bacillus anthracis Sap S-layer protein crystallization domain. Microbiology (Reading) 2005; 151:1485-1490. [PMID: 15870458 DOI: 10.1099/mic.0.27832-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bacillus anthracis, the aetiological agent of anthrax, synthesizes two surface-layer (S-layer) proteins. S-layers are two-dimensional crystalline arrays that completely cover bacteria. In rich medium, the B. anthracis S-layer consists of Sap during the exponential growth phase. Sap is a modular protein composed of an SLH (S-layer homology)-anchoring domain followed by a putative crystallization domain (Sapc). A projection map of the two-dimensional Sap array has been established on deflated bacteria. In this work, the authors used two approaches to investigate whether Sapc is the crystallization domain. The purified Sapc polypeptide (604 aa) was sufficient to form a crystalline structure, as illustrated by electron microscopy. Consistent with this result, the entire Sapc domain promoted auto-interaction in a bacterial two-hybrid screen developed for the present study. The screen was derived from a system that takes advantage of the Bordetella pertussis cyclase subdomain structure to enable one to identify peptides that interact. A screening strategy was then employed to study Sapc subdomains that mediate interaction. A random library, derived from the Sapc domain, was constructed and screened. The selected polypeptides interacting with the complete Sapc were all larger (155 aa and above) than the mean size of the randomly cloned peptides (approx. 60 residues). This result suggests that, in contrast with observations for other interactions studied with this two-hybrid system, large fragments were required to ensure efficient interaction. It was noteworthy that only one polypeptide, which spanned aa 148–358, was able to interact with less than the complete Sapc, in fact, with itself.
Collapse
Affiliation(s)
- Thomas Candela
- Unité Toxines et Pathogénie Bactérienne (CNRS, URA 2172), Institut Pasteur, 28 rue du Dr Roux, 75724 Paris cedex 15, France
| | - Tâm Mignot
- Unité Toxines et Pathogénie Bactérienne (CNRS, URA 2172), Institut Pasteur, 28 rue du Dr Roux, 75724 Paris cedex 15, France
| | - Xavier Hagnerelle
- Unité de Biochimie Structurale (CNRS, URA 2185), Institut Pasteur, 28 rue du Dr Roux, 75724 Paris cedex 15, France
| | - Michel Haustant
- Unité Toxines et Pathogénie Bactérienne (CNRS, URA 2172), Institut Pasteur, 28 rue du Dr Roux, 75724 Paris cedex 15, France
| | - Agnès Fouet
- Unité Toxines et Pathogénie Bactérienne (CNRS, URA 2172), Institut Pasteur, 28 rue du Dr Roux, 75724 Paris cedex 15, France
| |
Collapse
|
33
|
Mader C, Huber C, Moll D, Sleytr UB, Sára M. Interaction of the crystalline bacterial cell surface layer protein SbsB and the secondary cell wall polymer of Geobacillus stearothermophilus PV72 assessed by real-time surface plasmon resonance biosensor technology. J Bacteriol 2004; 186:1758-68. [PMID: 14996807 PMCID: PMC355981 DOI: 10.1128/jb.186.6.1758-1768.2004] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2003] [Accepted: 12/05/2003] [Indexed: 11/20/2022] Open
Abstract
The interaction between S-layer protein SbsB and the secondary cell wall polymer (SCWP) of Geobacillus stearothermophilus PV72/p2 was investigated by real-time surface plasmon resonance biosensor technology. The SCWP is an acidic polysaccharide that contains N-acetylglucosamine, N-acetylmannosamine, and pyruvic acid. For interaction studies, recombinant SbsB (rSbsB) and two truncated forms consisting of either the S-layer-like homology (SLH) domain (3SLH) or the residual part of SbsB were used. Independent of the setup, the data showed that the SLH domain was exclusively responsible for SCWP binding. The interaction was found to be highly specific, since neither the peptidoglycan nor SCWPs from other organisms nor other polysaccharides were recognized. Data analysis from that setup in which 3SLH was immobilized on a sensor chip and SCWP represented the soluble analyte was done in accordance with a model that describes binding of a bivalent analyte to a fixed ligand in terms of an overall affinity for all binding sites. The measured data revealed the presence of at least two binding sites on a single SCWP molecule with a distance of about 14 nm and an overall Kd of 7.7 x 10(-7) M. Analysis of data from the inverted setup in which the SCWP was immobilized on a sensor chip was done in accordance with an extension of the heterogeneous-ligand model, which indicated the existence of three binding sites with low (Kd = 2.6 x 10(-5) M), medium (Kd = 6.1 x 10(-8) M), and high (Kd = 6.7 x 10(-11) M) affinities. Since in this setup 3SLH was the soluble analyte and the presence of small amounts of oligomers in even monomeric protein solutions cannot be excluded, the high-affinity binding site may result from avidity effects caused by binding of at least dimeric 3SLH. Solution competition assays performed with both setups confirmed the specificity of the protein-carbohydrate interaction investigated.
Collapse
Affiliation(s)
- Christoph Mader
- Center for Ultrastructure Research and Ludwig Boltzmann Institute for Molecular Nanotechnology, University of Natural Resources and Applied Life Sciences Vienna, Austria.
| | | | | | | | | |
Collapse
|
34
|
Riedmann EM, Kyd JM, Smith AM, Gomez-Gallego S, Jalava K, Cripps AW, Lubitz W. Construction of recombinant S-layer proteins (rSbsA) and their expression in bacterial ghosts--a delivery system for the nontypeable Haemophilus influenzae antigen Omp26. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2003; 37:185-92. [PMID: 12832124 DOI: 10.1016/s0928-8244(03)00070-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This study has investigated the feasibility of a combination of recombinant surface layer (S-layer) proteins and empty bacterial cell envelopes (ghosts) to deliver candidate antigens for a vaccine against nontypeable Haemophilus influenzae (NTHi) infections. The S-layer gene sbsA from Bacillus stearothermophilus PV72 was used for the construction of fusion proteins. Fusion of maltose binding protein (MBP) to the N-terminus of SbsA allowed expression of the S-layer in the periplasm of Escherichia coli. The outer membrane protein (Omp) 26 of NTHi was inserted into the N-terminal and C-terminal regions of SbsA. The presence of the fused antigen Omp26 was demonstrated by Western blot experiments using anti-Omp26 antisera. Electron microscopy showed that the recombinant SbsA maintained the ability to self-assemble into sheet-like and cylindrical structures. Recombinant E. coli cell envelopes (ghosts) were produced by the expression of SbsA/Omp26 fusion proteins prior to gene E-mediated lysis. Intraperitoneal immunization with these recombinant bacterial ghosts induced an Omp26-specific antibody response in BALB/c mice. These results demonstrate that the NTHi antigen, Omp26, was expressed in the S-layer self-assembly product and this construct was immunogenic for Omp26 when administered to mice in bacterial cell envelopes.
Collapse
Affiliation(s)
- Eva M Riedmann
- Institute of Microbiology and Genetics, Vienna Biocentre, University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
35
|
Smit E, Jager D, Martinez B, Tielen FJ, Pouwels PH. Structural and functional analysis of the S-layer protein crystallisation domain of Lactobacillus acidophilus ATCC 4356: evidence for protein-protein interaction of two subdomains. J Mol Biol 2002; 324:953-64. [PMID: 12470951 DOI: 10.1016/s0022-2836(02)01135-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The structure of the crystallisation domain, SAN, of the S(A)-protein of Lactobacillus acidophilus ATCC 4356 was analysed by insertion and deletion mutagenesis, and by proteolytic treatment. Mutant S(A)-protein synthesised in Escherichia coli with 7-13 amino acid insertions near the N terminus or within regions of sequence variation in SAN (amino acid position 7, 45, 114, 125, 193), or in the cell wall-binding domain (position 345) could form crystalline sheets, whereas insertions in conserved regions or in regions with predicted secondary structure elements (positions 30, 67, 88 and 156) destroyed this capacity. FACscan analysis of L.acidophilus synthesising three crystallising and one non-crystallising S(A)-protein c-myc (19 amino acid residues) insertion mutant was performed with c-myc antibodies. Fluorescence was most pronounced for insertions at positions 125 and 156, less for position 45 and severely reduced for position 7. By cytometric flow sorting a transformant harbouring the mutant S(A)-protein gene (position 125) was isolated that showed an increased fluorescense signal. Immunofluorescence microscopy suggested that the transformant synthesized mutant S(A)-protein only. PCR analysis of the transformant grown in the absence of selection pressure indicated that the mutant allele was stably integrated in the chromosome. Proteolytic treatment of S(A)-protein indicated that only sites near the middle of SAN are susceptible, although potential cleavage sites are present through the entire molecule. Expression in E.coli of DNA sequences encoding the two halves of SAN yielded peptides that could oligomerize. Our results indicate that SAN consists of a approximately 12kDa N and a approximately 18kDa C-terminal subdomain linked by a surface exposed loop. The capacity of S(A)-protein of L.acidophilus to present epitopes, up to approximately 19 amino acid residues in length, at the bacterial surface in a genetically stable form, makes the system, in principle, suitable for application as an oral delivery vehicle.
Collapse
Affiliation(s)
- Egbert Smit
- Department of Applied Microbiology and Gene Technology, TNO Nutrition and Food Research Institute, Utrechtseweg 48, 3700 AJ, Zeist, The Netherlands
| | | | | | | | | |
Collapse
|
36
|
Antikainen J, Anton L, Sillanpää J, Korhonen TK. Domains in the S-layer protein CbsA of Lactobacillus crispatus involved in adherence to collagens, laminin and lipoteichoic acids and in self-assembly. Mol Microbiol 2002; 46:381-94. [PMID: 12406216 DOI: 10.1046/j.1365-2958.2002.03180.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The protein regions in the S-layer protein CbsA of Lactobacillus crispatus JCM 5810, needed for binding to collagens and laminin, anchoring to bacterial cell wall, as well as self-assembly, were mapped by deletion analysis of His-tagged peptides isolated from Escherichia coli and by heterologous expression on Lactobacillus casei. Mature CbsA is 410 amino acids long, and stepwise genetic truncation at both termini revealed that the region 32-271 carries the infor-mation for self-assembly of CbsA into a periodic structure. The lactobacillar S-layer proteins exhibit sequence variation in their assembly domain, but the border regions 30-34 and 269-274 in CbsA are conserved in valine-rich short sequences. Short deletions or substitutions at these regions affected the morphology of His-CbsA polymers, which varied from sheet-like to cylindrical tubular polymers, and further truncation beyond the DNA encoding residues 32 and 271 leads to a non-periodic aggregation. The self-assembly of the truncated peptides, as seen by electron microscopy, was correlated with their behaviour in a cross-linking study. The shorter peptides not forming a regular polymer were observed by the cross-linking study and mass spectrometry to form dimers, trimers and tetramers, whereas the other peptides were cross-linked to large multimers only. Binding of solubilized type I and IV collagens was observed with the His-CbsA peptides 1-274 and 31-287, but not with the smaller peptides regardless of their ability to form regular polymers. Strain JCM 5810 also adheres to immobilized laminin and, in order to analyse the possible laminin binding by CbsA, cbsA and its fragments were expressed on the surface of L. casei. Expression of the CbsA peptides 1-274, 1-287, 28-287 and 31-287 on L. casei conferred adhesiveness to both laminin and collagen immobilized on glass as well as to laminin- and collagen-containing regions in chicken colon and ileum. The C-terminal peptides 251-410 and 288-410 bound to L. crispatus JCM 5810 cells from which the S-layer had been depleted by chemical extraction, whereas no binding was seen with the His-CbsA peptides 1-250 or 1-269 or to cells with an intact S-layer. The His-CbsA peptides 251-410 and 288-410 bound to teichoic acids of several bacterial species. The results show that CbsA is an adhesive complex with an N-terminal assembly domain exhibiting affinity for pericellular tissue components and a cationic C-terminal domain binding to negatively charged cell wall components.
Collapse
Affiliation(s)
- Jenni Antikainen
- Department of Biosciences, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
37
|
Weygand M, Kjaer K, Howes PB, Wetzer B, Pum D, Sleytr UB, Lösche M. Structural Reorganization of Phospholipid Headgroups upon Recrystallization of an S-Layer Lattice. J Phys Chem B 2002. [DOI: 10.1021/jp0146418] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Markus Weygand
- Institute of Experimental Physics I, Leipzig University, Linnéstr. 5, D-04103 Leipzig, Germany, Materials Research Department, Risø National Laboratory, DK-4000 Roskilde, Denmark, Department of Physics and Astronomy, University of Leicester, Leicester, LE1 7RH, U.K., and Center for Ultrastructure Research and Ludwig-Boltzmann-Institute for Molecular Nanotechnology, University for Agricultural Sciences, A-1180 Vienna, Austria
| | - Kristian Kjaer
- Institute of Experimental Physics I, Leipzig University, Linnéstr. 5, D-04103 Leipzig, Germany, Materials Research Department, Risø National Laboratory, DK-4000 Roskilde, Denmark, Department of Physics and Astronomy, University of Leicester, Leicester, LE1 7RH, U.K., and Center for Ultrastructure Research and Ludwig-Boltzmann-Institute for Molecular Nanotechnology, University for Agricultural Sciences, A-1180 Vienna, Austria
| | - Paul B. Howes
- Institute of Experimental Physics I, Leipzig University, Linnéstr. 5, D-04103 Leipzig, Germany, Materials Research Department, Risø National Laboratory, DK-4000 Roskilde, Denmark, Department of Physics and Astronomy, University of Leicester, Leicester, LE1 7RH, U.K., and Center for Ultrastructure Research and Ludwig-Boltzmann-Institute for Molecular Nanotechnology, University for Agricultural Sciences, A-1180 Vienna, Austria
| | - Barbara Wetzer
- Institute of Experimental Physics I, Leipzig University, Linnéstr. 5, D-04103 Leipzig, Germany, Materials Research Department, Risø National Laboratory, DK-4000 Roskilde, Denmark, Department of Physics and Astronomy, University of Leicester, Leicester, LE1 7RH, U.K., and Center for Ultrastructure Research and Ludwig-Boltzmann-Institute for Molecular Nanotechnology, University for Agricultural Sciences, A-1180 Vienna, Austria
| | - Dietmar Pum
- Institute of Experimental Physics I, Leipzig University, Linnéstr. 5, D-04103 Leipzig, Germany, Materials Research Department, Risø National Laboratory, DK-4000 Roskilde, Denmark, Department of Physics and Astronomy, University of Leicester, Leicester, LE1 7RH, U.K., and Center for Ultrastructure Research and Ludwig-Boltzmann-Institute for Molecular Nanotechnology, University for Agricultural Sciences, A-1180 Vienna, Austria
| | - Uwe B. Sleytr
- Institute of Experimental Physics I, Leipzig University, Linnéstr. 5, D-04103 Leipzig, Germany, Materials Research Department, Risø National Laboratory, DK-4000 Roskilde, Denmark, Department of Physics and Astronomy, University of Leicester, Leicester, LE1 7RH, U.K., and Center for Ultrastructure Research and Ludwig-Boltzmann-Institute for Molecular Nanotechnology, University for Agricultural Sciences, A-1180 Vienna, Austria
| | - Mathias Lösche
- Institute of Experimental Physics I, Leipzig University, Linnéstr. 5, D-04103 Leipzig, Germany, Materials Research Department, Risø National Laboratory, DK-4000 Roskilde, Denmark, Department of Physics and Astronomy, University of Leicester, Leicester, LE1 7RH, U.K., and Center for Ultrastructure Research and Ludwig-Boltzmann-Institute for Molecular Nanotechnology, University for Agricultural Sciences, A-1180 Vienna, Austria
| |
Collapse
|
38
|
Breitwieser A, Egelseer EM, Moll D, Ilk N, Hotzy C, Bohle B, Ebner C, Sleytr UB, Sára M. A recombinant bacterial cell surface (S-layer)-major birch pollen allergen-fusion protein (rSbsC/Bet v1) maintains the ability to self-assemble into regularly structured monomolecular lattices and the functionality of the allergen. Protein Eng Des Sel 2002; 15:243-9. [PMID: 11932495 DOI: 10.1093/protein/15.3.243] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The mature crystalline bacterial cell surface (S-layer) protein SbsC of Bacillus stearothermophilus ATCC 12980 comprises amino acids 31-1099 and assembles into an oblique lattice type. As the deletion of up to 179 C-terminal amino acids did not interfere with the self-assembly properties of SbsC, the sequence encoding the major birch pollen allergen (Bet v1) was fused to the sequence encoding the truncated form rSbsC(31-920). The S-layer fusion protein, termed rSbsC/Bet v1, maintained the ability to self-assemble into flat sheets and open-ended cylinders. The presence and the functionality of the fused Bet v1 sequence was proved by blot experiments using BIP1, a monoclonal antibody against Bet v1 and Bet v1-specific IgE-containing serum samples from birch pollen allergic patients. The location and accessibility of the allergen moiety on the outer surface of the S-layer lattice were demonstrated by immunogold labeling of the rSbsC/Bet v1 monolayer, which was obtained by oriented recrystallization of the S-layer fusion protein on native cell wall sacculi. Thereby, the specific interactions between the N-terminal part of SbsC and a distinct type of secondary cell wall polymer were exploited. This is the first S-layer fusion protein described that had retained the specific properties of the S-layer protein moiety in addition to those of the fused functional peptide sequence.
Collapse
Affiliation(s)
- Andreas Breitwieser
- Center for Ultrastructure Research and Ludwig Boltzmann-Institute for Molecular Nanotechnology, University of Agricultural Sciences, Gregor Mendelstrasse 33, 1180 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Each subunit of a voltage-gated potassium channel (Kv) contains six putative transmembrane segments, S1-S6, and a cytosolic N-terminal recognition domain, T1. Although it is well-established that Kv channels are tetrameric structures, the protein-protein, protein-lipid, and protein-aqueous interfaces are not precisely mapped. The topological accessibility of specific amino acids may help to identify these border residues. Toward this end, a variant of the substituted-cysteine-accessibility method that relies on mass-labeling of accessible SH groups with a large SH reagent, methoxy-polyethylene glycol maleimide, and gel shift assay has been used. Pegylation of full-length Kv1.3, as well as Kv1.3 fragments, integrated into microsomal membranes, allows topological characterization of the 12 native cysteines (C1-C12), as well as cysteines engineered into a T1-T1 interface. Cysteines engineered into the T1-T1 interface had lower rates of pegylation than cytosolic-facing cysteines, namely, C5 in the T1 domain and C10-C12 in the C terminus.
Collapse
Affiliation(s)
- J Lu
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6085, USA
| | | |
Collapse
|
40
|
Howorka S, Movileanu L, Braha O, Bayley H. Kinetics of duplex formation for individual DNA strands within a single protein nanopore. Proc Natl Acad Sci U S A 2001; 98:12996-3001. [PMID: 11606775 PMCID: PMC60813 DOI: 10.1073/pnas.231434698] [Citation(s) in RCA: 167] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A single oligonucleotide was covalently attached to a genetically engineered subunit of the heptameric protein pore, alpha-hemolysin, to allow DNA duplex formation inside the pore lumen. Single-channel current recording was used to study the properties of the modified pore. On addition of an oligonucleotide 8 bases in length and with a sequence complementary to the tethered DNA strand, current blockades with durations of hundreds of milliseconds occurred, representing hybridization events of individual oligonucleotides to the tethered DNA strand. Kinetic constants for DNA duplex formation at the single molecule level were derived and found to be consistent with established literature values for macroscopic duplex formation. The resultant equilibrium constant for duplex formation in the nanopore was found to be close to the experimentally derived constant for duplex formation in solution. A good agreement between the equilibrium constants for duplex formation in the nanopore and in solution was also found for two other oligonucleotide pairs. In addition, the nanopore recordings revealed details of the kinetics difficult to obtain by conventional methods, like surface plasmon resonance, which measure ensemble properties. By investigating the temperature dependence of DNA duplex formation at the single molecule level, the standard enthalpy and entropy of the interaction could be obtained.
Collapse
Affiliation(s)
- S Howorka
- Department of Medical Biochemistry and Genetics, The Texas A&M University System Health Science Center, 440 Reynolds Medical Building, College Station, TX 77843-1114, USA.
| | | | | | | |
Collapse
|