1
|
Douzi B. Surface Plasmon Resonance: A Sensitive Tool to Study Protein-Protein Interactions. Methods Mol Biol 2024; 2715:363-382. [PMID: 37930540 DOI: 10.1007/978-1-0716-3445-5_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Surface plasmon resonance (SPR) is one of the most commonly used techniques to study protein-protein interactions. The main advantage of SPR is the ability of measuring binding affinities and association/dissociation kinetics of complexes in real time, in a label-free environment, and using relatively small quantities of materials. The method is based on the immobilization of one of the binding partners, called the "ligand," on a dedicated sensor surface. Immobilization is followed by the injection of the other partner, called the "analyte," over the surface containing the ligand. The binding is monitored by following changes in the refractive index of the medium close to the sensor surface upon injection of the analyte. During the last 15 years, SPR has been intensively used in the study of bacterial secretion systems due to its ability of detecting highly dynamic complexes, which are difficult to investigate by other techniques. This chapter will guide users in setting up SPR experiments in order to identify protein complexes and to assess their binding affinity and/or kinetics. It will include detailed protocols for (i) immobilization of proteins with the amine coupling capture method, (ii) analyte-binding analysis, (iii) affinity/kinetics measurements, and (iv) data analysis.
Collapse
|
2
|
Li Y, Santos-Moreno J, Francetic O. The periplasmic coiled coil formed by the assembly platform proteins PulL and PulM is critical for function of the Klebsiella type II secretion system. Res Microbiol 2023; 174:104075. [PMID: 37141929 DOI: 10.1016/j.resmic.2023.104075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Bacteria use type II secretion systems (T2SS) to secrete to their surface folded proteins that confer diverse functions, from nutrient acquisition to virulence. In the Klebsiella species, T2SS-mediated secretion of pullulanase (PulA) requires assembly of a dynamic filament called the endopilus. The inner membrane assembly platform (AP) subcomplex is essential for endopilus assembly and PulA secretion. AP components PulL and PulM interact with each other through their C-terminal globular domains and transmembrane segments. Here, we investigated the roles of their periplasmic helices, predicted to form a coiled coil, in assembly and function of the PulL-PulM complex. PulL and PulM variants lacking these periplasmic helices were defective for interaction in the bacterial two-hybrid (BACTH) assay. Their functions in PulA secretion and assembly of PulG subunits into endopilus filaments were strongly reduced. Interestingly, deleting the cytoplasmic peptide of PulM nearly abolished the function of variant PulMΔN and its interaction with PulG, but not with PulL, in the BACTH assay. Nevertheless, PulL was specifically proteolyzed in the presence of the PulMΔN variant, suggesting that PulM N-terminal peptide stabilizes PulL in the cytoplasm. We discuss the implications of these results for the T2S endopilus and type IV pilus assembly mechanisms.
Collapse
Affiliation(s)
- Yuanyuan Li
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Biochemistry of Macromolecular Interactions Unit, F-75015 Paris, France.
| | - Javier Santos-Moreno
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Biochemistry of Macromolecular Interactions Unit, F-75015 Paris, France.
| | - Olivera Francetic
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Biochemistry of Macromolecular Interactions Unit, F-75015 Paris, France.
| |
Collapse
|
3
|
Gerbaud G, Barbat B, Tribout M, Etienne E, Belle V, Douzi B, Voulhoux R, Bonucci A. Refining the Dynamic Network of T2SS Endopilus Tip Heterocomplex Combining cw-EPR and Nitroxide-Gd III Distance Measurements. Chembiochem 2023; 24:e202300099. [PMID: 36999435 DOI: 10.1002/cbic.202300099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/30/2023] [Indexed: 04/01/2023]
Abstract
The type 2 secretion system (T2SS) is a bacterial nanomachine composed of an inner membrane assembly platform, an outer membrane pore and a dynamic endopilus. T2SS endopili are organized into a homo-multimeric body formed by the major pilin capped by a heterocomplex of four minor pilins. The first model of the T2SS endopilus was recently released, even if structural dynamics insights are still required to decipher the role of each protein in the full tetrameric complex. Here, we applied continuous-wave and pulse EPR spectroscopy using nitroxide-gadolinium orthogonal labelling strategies to investigate the hetero-oligomeric assembly of the minor pilins. Overall, our data are in line with the endopilus model even if they evidenced conformational flexibility and alternative orientations at local scale of specific regions of minor pilins. The integration of different labelling strategies and EPR experiments demonstrates the pertinence of this approach to investigate protein-protein interactions in such multiprotein heterocomplexes.
Collapse
Affiliation(s)
- Guillaume Gerbaud
- BIP-Bioénérgetique et Ingénierie es Protéines, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
| | - Brice Barbat
- LCB-Laboratoire de Chimie Bactérienne, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
| | - Mathilde Tribout
- LCB-Laboratoire de Chimie Bactérienne, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
| | - Emilien Etienne
- BIP-Bioénérgetique et Ingénierie es Protéines, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
| | - Valérie Belle
- BIP-Bioénérgetique et Ingénierie es Protéines, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
| | - Badreddine Douzi
- LCB-Laboratoire de Chimie Bactérienne, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
- Present address: INRAE, DynAMic, Université de Lorraine, 54000, Nancy, France
| | - Romé Voulhoux
- LCB-Laboratoire de Chimie Bactérienne, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
| | - Alessio Bonucci
- BIP-Bioénérgetique et Ingénierie es Protéines, IMM, Aix Marseille Université, CNRS, 13009, Marseille, France
| |
Collapse
|
4
|
Rozano L, Mukuka YM, Hane JK, Mancera RL. Ab Initio Modelling of the Structure of ToxA-like and MAX Fungal Effector Proteins. Int J Mol Sci 2023; 24:ijms24076262. [PMID: 37047233 PMCID: PMC10094246 DOI: 10.3390/ijms24076262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/09/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Pathogenic fungal diseases in crops are mediated by the release of effector proteins that facilitate infection. Characterising the structure of these fungal effectors is vital to understanding their virulence mechanisms and interactions with their hosts, which is crucial in the breeding of plant cultivars for disease resistance. Several effectors have been identified and validated experimentally; however, their lack of sequence conservation often impedes the identification and prediction of their structure using sequence similarity approaches. Structural similarity has, nonetheless, been observed within fungal effector protein families, creating interest in validating the use of computational methods to predict their tertiary structure from their sequence. We used Rosetta ab initio modelling to predict the structures of members of the ToxA-like and MAX effector families for which experimental structures are known to validate this method. An optimised approach was then used to predict the structures of phenotypically validated effectors lacking known structures. Rosetta was found to successfully predict the structure of fungal effectors in the ToxA-like and MAX families, as well as phenotypically validated but structurally unconfirmed effector sequences. Interestingly, potential new effector structural families were identified on the basis of comparisons with structural homologues and the identification of associated protein domains.
Collapse
|
5
|
Characterization of a glycan-binding complex of minor pilins completes the analysis of Streptococcus sanguinis type 4 pili subunits. Proc Natl Acad Sci U S A 2023; 120:e2216237120. [PMID: 36626560 PMCID: PMC9934059 DOI: 10.1073/pnas.2216237120] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Type 4 filaments (T4F)-of which type 4 pili (T4P) are the archetype-are a superfamily of nanomachines nearly ubiquitous in prokaryotes. T4F are polymers of one major pilin, which also contain minor pilins whose roles are often poorly understood. Here, we complete the structure/function analysis of the full set of T4P pilins in the opportunistic bacterial pathogen Streptococcus sanguinis. We determined the structure of the minor pilin PilA, which is unexpectedly similar to one of the subunits of a tip-located complex of four minor pilins, widely conserved in T4F. We found that PilA interacts and dramatically stabilizes the minor pilin PilC. We determined the structure of PilC, showing that it is a modular pilin with a lectin module binding a subset of glycans prevalent in the human glycome, the host of S. sanguinis. Altogether, our findings support a model whereby the minor pilins in S. sanguinis T4P form a tip-located complex promoting adhesion to various host receptors. This has general implications for T4F.
Collapse
|
6
|
Filloux A. Bacterial protein secretion systems: Game of types. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35536734 DOI: 10.1099/mic.0.001193] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein trafficking across the bacterial envelope is a process that contributes to the organisation and integrity of the cell. It is the foundation for establishing contact and exchange between the environment and the cytosol. It helps cells to communicate with one another, whether they establish symbiotic or competitive behaviours. It is instrumental for pathogenesis and for bacteria to subvert the host immune response. Understanding the formation of envelope conduits and the manifold strategies employed for moving macromolecules across these channels is a fascinating playground. The diversity of the nanomachines involved in this process logically resulted in an attempt to classify them, which is where the protein secretion system types emerged. As our knowledge grew, so did the number of types, and their rightful nomenclature started to be questioned. While this may seem a semantic or philosophical issue, it also reflects scientific rigour when it comes to assimilating findings into textbooks and science history. Here I give an overview on bacterial protein secretion systems, their history, their nomenclature and why it can be misleading for newcomers in the field. Note that I do not try to suggest a new nomenclature. Instead, I explore the reasons why naming could have escaped our control and I try to reiterate basic concepts that underlie protein trafficking cross membranes.
Collapse
Affiliation(s)
- Alain Filloux
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
7
|
The molecular basis of FimT-mediated DNA uptake during bacterial natural transformation. Nat Commun 2022; 13:1065. [PMID: 35246533 PMCID: PMC8897410 DOI: 10.1038/s41467-022-28690-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/08/2022] [Indexed: 01/05/2023] Open
Abstract
Naturally competent bacteria encode sophisticated protein machinery for the uptake and translocation of exogenous DNA into the cell. If this DNA is integrated into the bacterial genome, the bacterium is said to be naturally transformed. Most competent bacterial species utilise type IV pili for the initial DNA uptake step. These proteinaceous cell-surface structures are composed of thousands of pilus subunits (pilins), designated as major or minor according to their relative abundance in the pilus. Here, we show that the minor pilin FimT plays an important role in the natural transformation of Legionella pneumophila. We use NMR spectroscopy, in vitro DNA binding assays and in vivo transformation assays to understand the molecular basis of FimT's role in this process. FimT binds to DNA via an electropositive patch, rich in arginines, several of which are well-conserved and located in a conformationally flexible C-terminal tail. FimT orthologues from other Gammaproteobacteria share the ability to bind to DNA. Our results suggest that FimT plays an important role in DNA uptake in a wide range of competent species.
Collapse
|
8
|
Xu Y, Yang L, Wang Y, Zhu Z, Yan J, Qin S, Chen L. Prophage-encoded gene VpaChn25_0734 amplifies ecological persistence of Vibrio parahaemolyticus CHN25. Curr Genet 2022; 68:267-287. [PMID: 35064802 PMCID: PMC8783578 DOI: 10.1007/s00294-022-01229-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/03/2021] [Accepted: 12/31/2021] [Indexed: 12/19/2022]
Abstract
Vibrio parahaemolyticus is a waterborne pathogen that can cause acute gastroenteritis, wound infection, and septicemia in humans. The molecular basis of its pathogenicity is not yet fully understood. Phages are found most abundantly in aquatic environments and play a critical role in horizontal gene transfer. Nevertheless, current literature on biological roles of prophage-encoded genes remaining in V. parahaemolyticus is rare. In this study, we characterized one such gene VpaChn25_0734 (543-bp) in V. parahaemolyticus CHN25 genome. A deletion mutant ΔVpaChn25_0734 (543-bp) was obtained by homologous recombination, and a revertant ΔVpaChn25_0734-com (543-bp) was also constructed. The ΔVpaChn25_0734 (543-bp) mutant was defective in growth and swimming mobility particularly at lower temperatures and/or pH 7.0–8.5. Cell surface hydrophobicity and biofilm formation were significantly decreased in the ΔVpaChn25_0734 (543-bp) mutant (p < 0.05). Based on the in vitro Caco-2 cell model, the deletion of VpaChn25_0734 (543-bp) gene significantly reduced the cytotoxicity of V. parahaemolyticus CHN25 to human intestinal epithelial cells (p < 0.05). Comparative secretomic and transcriptomic analyses revealed a slightly increased extracellular proteins, and thirteen significantly changed metabolic pathways in the ΔVpaChn25_0734 (543-bp) mutant, showing down-regulated carbon source transport and utilization, biofilm formation, and type II secretion system (p < 0.05), consistent with the observed defective phenotypes. Taken, the prophage-encoded gene VpaChn25_0734 (543-bp) enhanced V. parahaemolyticus CHN25 fitness for survival in the environment and the host. The results in this study facilitate better understanding of pathogenesis and genome evolution of V. parahaemolyticus, the leading sea foodborne pathogen worldwide.
Collapse
Affiliation(s)
- Yingwei Xu
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of the People's Republic of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Lianzhi Yang
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of the People's Republic of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Yaping Wang
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of the People's Republic of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Centre, Richmond, VA, USA
| | - Zhuoying Zhu
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of the People's Republic of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Jizhou Yan
- College of Fishers and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Si Qin
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Lanming Chen
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs of the People's Republic of China, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
9
|
Oliveira V, Aschtgen MS, van Erp A, Henriques-Normark B, Muschiol S. The Role of Minor Pilins in Assembly and Function of the Competence Pilus of Streptococcus pneumoniae. Front Cell Infect Microbiol 2022; 11:808601. [PMID: 35004361 PMCID: PMC8727766 DOI: 10.3389/fcimb.2021.808601] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/30/2021] [Indexed: 12/03/2022] Open
Abstract
The remarkable genomic plasticity of Streptococcus pneumoniae largely depends on its ability to undergo natural genetic transformation. To take up extracellular DNA, S. pneumoniae assembles competence pili composed of the major pilin ComGC. In addition to ComGC, four minor pilins ComGD, E, F, and G are expressed during bacterial competence, but their role in pilus biogenesis and transformation is unknown. Here, using a combination of protein-protein interaction assays we show that all four proteins can directly interact with each other. Pneumococcal ComGG stabilizes the minor pilin ComGD and ComGF and can interact with and stabilize the major pilin ComGC, thus, deletion of ComGG abolishes competence pilus assembly. We further demonstrate that minor pilins are present in sheared pili fractions and find ComGF to be incorporated along the competence pilus by immunofluorescence and electron microscopy. Finally, mutants of the invariant Glu5 residue (E5), ComGDE5A or ComGEE5A, but not ComGFE5A, were severely impaired in pilus formation and function. Together, our results suggest that ComGG, lacking E5, is essential for competence pilus assembly and function, and plays a central role in connecting the pneumococcal minor pilins to ComGC.
Collapse
Affiliation(s)
- Vitor Oliveira
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Anke van Erp
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Muschiol
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Structural interactions define assembly adapter function of a type II secretion system pseudopilin. Structure 2021; 29:1116-1127.e8. [PMID: 34139172 DOI: 10.1016/j.str.2021.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/15/2021] [Accepted: 05/28/2021] [Indexed: 01/13/2023]
Abstract
The type IV filament superfamily comprises widespread membrane-associated polymers in prokaryotes. The type II secretion system (T2SS), a virulence pathway in many pathogens, belongs to this superfamily. A knowledge gap in understanding of the T2SS is the molecular role of a small "pseudopilin" protein. Using multiple biophysical techniques, we have deciphered how this missing component of the Xcp T2SS architecture is structurally integrated, and thereby unlocked its function. We demonstrate that low-abundance XcpH is the adapter that bridges a trimeric initiating tip complex, XcpIJK, with a periplasmic filament of XcpG subunits. Each pseudopilin protein caps an XcpG protofilament in an overall pseudopilus compatible with dimensions of the periplasm and the outer membrane-spanning secretin through which substrates pass. Unexpectedly, to fulfill its adapter function, the XcpH N-terminal helix must be unwound, a property shared with XcpG subunits. We provide an experimentally validated three-dimensional structural model of a complete type IV filament.
Collapse
|
11
|
Llanos Salinas SP, Castillo Sánchez LO, Castañeda Miranda G, Rodríguez Reyes EA, Ordoñez López L, Mena Bañuelos R, Alcaraz Sosa LE, Núñez Carrera MG, José Manuel RO, Carmona Gasca CA, Matsunaga J, Haake DA, Candanosa Aranda IE, de la Peña-Moctezuma A. GspD, The Type II Secretion System Secretin of Leptospira, Protects Hamsters against Lethal Infection with a Virulent L. interrogans Isolate. Vaccines (Basel) 2020; 8:vaccines8040759. [PMID: 33327369 PMCID: PMC7768463 DOI: 10.3390/vaccines8040759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/28/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022] Open
Abstract
The wide variety of pathogenic Leptospira serovars and the weak protection offered by the available vaccines encourage the search for protective immunogens against leptospirosis. We found that the secretin GspD of the type II secretion system (T2S) of Leptospira interrogans serovar Canicola was highly conserved amongst pathogenic serovars and was expressed in vivo during infection, as shown by immunohistochemistry. Convalescent sera of hamsters, dogs, and cows showed the presence of IgG antibodies, recognizing a recombinant version of this protein expressed in Escherichia coli (rGspDLC) in Western blot assays. In a pilot vaccination study, a group of eight hamsters was immunized on days zero and 14 with 50 µg of rGspDLC mixed with Freund’s incomplete adjuvant (FIA). On day 28 of the study, 1,000 LD50 (Lethal Dose 50%) of a virulent strain of Leptospira interrogans serovar Canicola (LOCaS46) were inoculated by an intraoral submucosal route (IOSM). Seventy-five percent protection against disease (p = 0.017573, Fisher’s exact test) and 50% protection against infection were observed in this group of vaccinated hamsters. In contrast, 85% of non-vaccinated hamsters died six to nine days after the challenge. These results suggest the potential usefulness of the T2S secretin GspD of Leptospira as a protective recombinant vaccine against leptospirosis.
Collapse
Affiliation(s)
- Samantha Paulina Llanos Salinas
- Teaching, Research and Extension Center for Animal Production in High Plateau, School of Veterinary Medicine and Zootechnics, National Autonomous University of Mexico, Queretaro 76795, Mexico; (S.P.L.S.); (G.C.M.); (I.E.C.A.)
| | - Luz Olivia Castillo Sánchez
- Unidad Académica de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nayarit, Tepic 63155, Mexico; (L.O.C.S.); (C.A.C.G.)
| | - Giselle Castañeda Miranda
- Teaching, Research and Extension Center for Animal Production in High Plateau, School of Veterinary Medicine and Zootechnics, National Autonomous University of Mexico, Queretaro 76795, Mexico; (S.P.L.S.); (G.C.M.); (I.E.C.A.)
| | | | - Liliana Ordoñez López
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán 04510, Mexico; (L.O.L.); (R.M.B.); (R.O.J.M.)
| | - Rodrigo Mena Bañuelos
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán 04510, Mexico; (L.O.L.); (R.M.B.); (R.O.J.M.)
| | - Luz Elena Alcaraz Sosa
- Departamento de Producción Agrícola y Animal, Universidad Autónoma Metropolitana, Tlalpan 14387, Mexico;
| | - María Guadalupe Núñez Carrera
- Facultad de Medicina Veterinaria y Zootecnia, Benemérita Universidad Autónoma de Puebla, Centro Histórico 72000, Mexico;
| | - Ramírez Ortega José Manuel
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán 04510, Mexico; (L.O.L.); (R.M.B.); (R.O.J.M.)
| | - Carlos Alfredo Carmona Gasca
- Unidad Académica de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Nayarit, Tepic 63155, Mexico; (L.O.C.S.); (C.A.C.G.)
| | - James Matsunaga
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA; (J.M.); (D.A.H.)
| | - David A. Haake
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA; (J.M.); (D.A.H.)
| | - Irma Eugenia Candanosa Aranda
- Teaching, Research and Extension Center for Animal Production in High Plateau, School of Veterinary Medicine and Zootechnics, National Autonomous University of Mexico, Queretaro 76795, Mexico; (S.P.L.S.); (G.C.M.); (I.E.C.A.)
| | - Alejandro de la Peña-Moctezuma
- Teaching, Research and Extension Center for Animal Production in High Plateau, School of Veterinary Medicine and Zootechnics, National Autonomous University of Mexico, Queretaro 76795, Mexico; (S.P.L.S.); (G.C.M.); (I.E.C.A.)
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Coyoacán 04510, Mexico; (L.O.L.); (R.M.B.); (R.O.J.M.)
- Correspondence: ; Tel.:+52-414-291-8100
| |
Collapse
|
12
|
Treuner-Lange A, Chang YW, Glatter T, Herfurth M, Lindow S, Chreifi G, Jensen GJ, Søgaard-Andersen L. PilY1 and minor pilins form a complex priming the type IVa pilus in Myxococcus xanthus. Nat Commun 2020; 11:5054. [PMID: 33028835 PMCID: PMC7541494 DOI: 10.1038/s41467-020-18803-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 09/14/2020] [Indexed: 01/05/2023] Open
Abstract
Type IVa pili are ubiquitous and versatile bacterial cell surface filaments that undergo cycles of extension, adhesion and retraction powered by the cell-envelope spanning type IVa pilus machine (T4aPM). The overall architecture of the T4aPM and the location of 10 conserved core proteins within this architecture have been elucidated. Here, using genetics, cell biology, proteomics and cryo-electron tomography, we demonstrate that the PilY1 protein and four minor pilins, which are widely conserved in T4aP systems, are essential for pilus extension in Myxococcus xanthus and form a complex that is an integral part of the T4aPM. Moreover, these proteins are part of the extended pilus. Our data support a model whereby the PilY1/minor pilin complex functions as a priming complex in T4aPM for pilus extension, a tip complex in the extended pilus for adhesion, and a cork for terminating retraction to maintain a priming complex for the next round of extension.
Collapse
Affiliation(s)
- Anke Treuner-Lange
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Str. 10, 35043, Marburg, Germany
| | - Yi-Wei Chang
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Boulevard, Pasadena, CA, 91125, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 422 Curie Boulevard, Philadelphia, PA, 19104, USA
| | - Timo Glatter
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Str. 10, 35043, Marburg, Germany
| | - Marco Herfurth
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Str. 10, 35043, Marburg, Germany
| | - Steffi Lindow
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Str. 10, 35043, Marburg, Germany
| | - Georges Chreifi
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Boulevard, Pasadena, CA, 91125, USA
| | - Grant J Jensen
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Boulevard, Pasadena, CA, 91125, USA
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Lotte Søgaard-Andersen
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Str. 10, 35043, Marburg, Germany.
| |
Collapse
|
13
|
Zhang Y, Wang S, Jia Z. In Situ Proteolysis Condition-Induced Crystallization of the XcpVWX Complex in Different Lattices. Int J Mol Sci 2020; 21:ijms21010308. [PMID: 31906428 PMCID: PMC6981927 DOI: 10.3390/ijms21010308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 12/28/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022] Open
Abstract
Although prevalent in the determination of protein structures; crystallography always has the bottleneck of obtaining high-quality protein crystals for characterizing a wide range of proteins; especially large protein complexes. Stable fragments or domains of proteins are more readily to crystallize; which prompts the use of in situ proteolysis to remove flexible or unstable structures for improving crystallization and crystal quality. In this work; we investigated the effects of in situ proteolysis by chymotrypsin on the crystallization of the XcpVWX complex from the Type II secretion system of Pseudomonas aeruginosa. Different proteolysis conditions were found to result in two distinct lattices in the same crystallization solution. With a shorter chymotrypsin digestion at a lower concentration; the crystals exhibited a P3 hexagonal lattice that accommodates three complex molecules in one asymmetric unit. By contrast; a longer digestion with chymotrypsin of a 10-fold higher concentration facilitated the formation of a compact P212121 orthorhombic lattice with only one complex molecule in each asymmetric unit. The molecules in the hexagonal lattice have shown high atomic displacement parameter values compared with the ones in the orthorhombic lattice. Taken together; our results clearly demonstrate that different proteolysis conditions can result in the generation of distinct lattices in the same crystallization solution; which can be exploited in order to obtain different crystal forms of a better quality
Collapse
Affiliation(s)
- Yichen Zhang
- Department of Biomedical and Molecular Sciences, Queen’s University, 18 Stuart Street, Kingston, ON K7L 3N6, Canada;
| | - Shu Wang
- College of Chemistry, Beijing Normal University, 19 Xinjiekou Outer Street, Beijing 100875, China;
| | - Zongchao Jia
- Department of Biomedical and Molecular Sciences, Queen’s University, 18 Stuart Street, Kingston, ON K7L 3N6, Canada;
- Correspondence: ; Tel.: +86-1-613-533-6277
| |
Collapse
|
14
|
Ghosal D, Kim KW, Zheng H, Kaplan M, Truchan HK, Lopez AE, McIntire IE, Vogel JP, Cianciotto NP, Jensen GJ. In vivo structure of the Legionella type II secretion system by electron cryotomography. Nat Microbiol 2019; 4:2101-2108. [PMID: 31754273 PMCID: PMC6879910 DOI: 10.1038/s41564-019-0603-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
Abstract
The type II secretion system (T2SS) is a multiprotein envelope-spanning assembly that translocates a wide range of virulence factors, enzymes and effectors through the outer membrane of many Gram-negative bacteria1-3. Here, using electron cryotomography and subtomogram averaging methods, we reveal the in vivo structure of an intact T2SS imaged within the human pathogen Legionella pneumophila. Although the T2SS has only limited sequence and component homology with the evolutionarily related type IV pilus (T4P) system4,5, we show that their overall architectures are remarkably similar. Despite similarities, there are also differences, including, for example, that the T2SS-ATPase complex is usually present but disengaged from the inner membrane, the T2SS has a much longer periplasmic vestibule and it has a short-lived flexible pseudopilus. Placing atomic models of the components into our electron cryotomography map produced a complete architectural model of the intact T2SS that provides insights into the structure and function of its components, its position within the cell envelope and the interactions between its different subcomplexes.
Collapse
Affiliation(s)
- Debnath Ghosal
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Ki Woo Kim
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- School of Ecology and Environmental System, Kyungpook National University, Sangju, Korea
| | - Huaixin Zheng
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Mohammed Kaplan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Hilary K Truchan
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alberto E Lopez
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ian E McIntire
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joseph P Vogel
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Nicholas P Cianciotto
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Grant J Jensen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Howard Hughes Medical Institute, Pasadena, CA, USA.
| |
Collapse
|
15
|
Abstract
The type II secretion system (T2SS) delivers toxins and a range of hydrolytic enzymes, including proteases, lipases, and carbohydrate-active enzymes, to the cell surface or extracellular space of Gram-negative bacteria. Its contribution to survival of both extracellular and intracellular pathogens as well as environmental species of proteobacteria is evident. This dynamic, multicomponent machinery spans the entire cell envelope and consists of a cytoplasmic ATPase, several inner membrane proteins, a periplasmic pseudopilus, and a secretin pore embedded in the outer membrane. Despite the trans-envelope configuration of the T2S nanomachine, proteins to be secreted engage with the system first once they enter the periplasmic compartment via the Sec or TAT export system. Thus, the T2SS is specifically dedicated to their outer membrane translocation. The many sequence and structural similarities between the T2SS and type IV pili suggest a common origin and argue for a pilus-mediated mechanism of secretion. This minireview describes the structures, functions, and interactions of the individual T2SS components and the general architecture of the assembled T2SS machinery and briefly summarizes the transport and function of a growing list of T2SS exoproteins. Recent advances in cryo-electron microscopy, which have led to an increased understanding of the structure-function relationship of the secretin channel and the pseudopilus, are emphasized.
Collapse
|
16
|
White RC, Cianciotto NP. Assessing the impact, genomics and evolution of type II secretion across a large, medically important genus: the Legionella type II secretion paradigm. Microb Genom 2019; 5. [PMID: 31166887 PMCID: PMC6617341 DOI: 10.1099/mgen.0.000273] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The type II secretion system (T2SS) plays a major role in promoting bacterial survival in the environment and in human hosts. One of the best characterized T2SS is that of Legionella pneumophila, the agent of Legionnaires’ disease. Secreting at least 25 proteins, including degradative enzymes, eukaryotic-like proteins and novel effectors, this T2SS contributes to the ability of L. pneumophila to grow at low temperatures, infect amoebal and macrophage hosts, damage lung tissue, evade the immune system, and undergo sliding motility. The genes encoding the T2SS are conserved across the genus Legionella, which includes 62 species and >30 pathogens in addition to L. pneumophila. The vast majority of effectors associated with L. pneumophila are shared by a large number of Legionella species, hinting at a critical role for them in the ecology of Legionella as a whole. However, no other species has the same repertoire as L. pneumophila, with, as a general rule, phylogenetically more closely related species sharing similar sets of effectors. T2SS effectors that are involved in infection of a eukaryotic host(s) are more prevalent throughout Legionella, indicating that they are under stronger selective pressure. The Legionella T2SS apparatus is closest to that of Aquicella (another parasite of amoebae), and a significant number of L. pneumophila effectors have their closest homologues in Aquicella. Thus, the T2SS of L. pneumophila probably originated within the order Legionellales, with some of its effectors having arisen within that Aquicella-like progenitor, while other effectors derived from the amoebal host, mimiviruses, fungi and less closely related bacteria.
Collapse
Affiliation(s)
- Richard C White
- 1 Department of Microbiology and Immunology, Northwestern University Medical School, Chicago, IL 60611, USA
| | - Nicholas P Cianciotto
- 1 Department of Microbiology and Immunology, Northwestern University Medical School, Chicago, IL 60611, USA
| |
Collapse
|
17
|
Zhang Y, Faucher F, Zhang W, Wang S, Neville N, Poole K, Zheng J, Jia Z. Structure-guided disruption of the pseudopilus tip complex inhibits the Type II secretion in Pseudomonas aeruginosa. PLoS Pathog 2018; 14:e1007343. [PMID: 30346996 PMCID: PMC6211770 DOI: 10.1371/journal.ppat.1007343] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/01/2018] [Accepted: 09/19/2018] [Indexed: 01/22/2023] Open
Abstract
Pseudomonas aeruginosa utilizes the Type II secretion system (T2SS) to translocate a wide range of large, structured protein virulence factors through the periplasm to the extracellular environment for infection. In the T2SS, five pseudopilins assemble into the pseudopilus that acts as a piston to extrude exoproteins out of cells. Through structure determination of the pseudopilin complexes of XcpVWX and XcpVW and function analysis, we have confirmed that two minor pseudopilins, XcpV and XcpW, constitute a core complex indispensable to the pseudopilus tip. The absence of either XcpV or -W resulted in the non-functional T2SS. Our small-angle X-ray scattering experiment for the first time revealed the architecture of the entire pseudopilus tip and established the working model. Based on the interaction interface of complexes, we have developed inhibitory peptides. The structure-based peptides not only disrupted of the XcpVW core complex and the entire pseudopilus tip in vitro but also inhibited the T2SS in vivo. More importantly, these peptides effectively reduced the virulence of P. aeruginosa towards Caenorhabditis elegans. The Type II secretion system has been characterized as an important virulence factor translocation machine that secrets various toxic proteins from the periplasm into the extracellular milieu used by a wide spectrum of Gram-negative bacteria. Through the characterization of the structure of the pseudopilus tip complex by protein crystallography and small-angle X-ray scattering, we have identified a critical interaction interface in the core binary complex formed by two minor pseudopilins, XcpV and–W, in Pseudomonas aeruginosa. Based on the interaction interface, two inhibitory peptides were developed, which showed potency of disrupting the entire pseudopilus tip complex and further inhibited the Type II secretion system. When applied to Caenorhabditis elegans, these peptides prevent the killing of worms by the P. aeruginosa. Our work has represented the first successful research on the inhibition of the Type II secretion system based on the structure of the pseudopilus tip complex.
Collapse
Affiliation(s)
- Yichen Zhang
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Frédérick Faucher
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Wenwen Zhang
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Shu Wang
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Nolan Neville
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Keith Poole
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Zongchao Jia
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- * E-mail:
| |
Collapse
|
18
|
Michel-Souzy S, Douzi B, Cadoret F, Raynaud C, Quinton L, Ball G, Voulhoux R. Direct interactions between the secreted effector and the T2SS components GspL and GspM reveal a new effector-sensing step during type 2 secretion. J Biol Chem 2018; 293:19441-19450. [PMID: 30337370 DOI: 10.1074/jbc.ra117.001127] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
In many Gram-negative bacteria, the type 2 secretion system (T2SS) plays an important role in virulence because of its capacity to deliver a large amount of fully folded protein effectors to the extracellular milieu. Despite our knowledge of most T2SS components, the mechanisms underlying effector recruitment and secretion by the T2SS remain enigmatic. Using complementary biophysical and biochemical approaches, we identified here two direct interactions between the secreted effector CbpD and two components, XcpYL and XcpZM, of the T2SS assembly platform (AP) in the opportunistic pathogen Pseudomonas aeruginosa Competition experiments indicated that CbpD binding to XcpYL is XcpZM-dependent, suggesting sequential recruitment of the effector by the periplasmic domains of these AP components. Using a bacterial two-hybrid system, we then tested the influence of the effector on the AP protein-protein interaction network. Our findings revealed that the presence of the effector modifies the AP interactome and, in particular, induces XcpZM homodimerization and increases the affinity between XcpYL and XcpZM The observed direct relationship between effector binding and T2SS dynamics suggests an additional synchronizing step during the type 2 secretion process, where the activation of the AP of the T2SS nanomachine is triggered by effector binding.
Collapse
Affiliation(s)
- Sandra Michel-Souzy
- From the CNRS, Aix Marseille Université, Institut de Microbiologie de la Méditerranée (IMM), Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM)/UMR7255, 13009 Marseille, France
| | - Badreddine Douzi
- From the CNRS, Aix Marseille Université, Institut de Microbiologie de la Méditerranée (IMM), Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM)/UMR7255, 13009 Marseille, France.,CNRS, Aix Marseille Université, IMM, Laboratoire de Chimie Bactérienne (LCB)/UMR7283, 13009 Marseille, France, and
| | - Frédéric Cadoret
- From the CNRS, Aix Marseille Université, Institut de Microbiologie de la Méditerranée (IMM), Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM)/UMR7255, 13009 Marseille, France
| | - Claire Raynaud
- From the CNRS, Aix Marseille Université, Institut de Microbiologie de la Méditerranée (IMM), Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM)/UMR7255, 13009 Marseille, France.,CNRS, Aix Marseille Université, IMM, Laboratoire de Chimie Bactérienne (LCB)/UMR7283, 13009 Marseille, France, and
| | - Loïc Quinton
- Laboratory of Mass Spectrometry-MolSys, Department of Chemistry, University of Liège, B4000 Liège, Belgium
| | - Geneviève Ball
- From the CNRS, Aix Marseille Université, Institut de Microbiologie de la Méditerranée (IMM), Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM)/UMR7255, 13009 Marseille, France.,CNRS, Aix Marseille Université, IMM, Laboratoire de Chimie Bactérienne (LCB)/UMR7283, 13009 Marseille, France, and
| | - Romé Voulhoux
- From the CNRS, Aix Marseille Université, Institut de Microbiologie de la Méditerranée (IMM), Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM)/UMR7255, 13009 Marseille, France, .,CNRS, Aix Marseille Université, IMM, Laboratoire de Chimie Bactérienne (LCB)/UMR7283, 13009 Marseille, France, and
| |
Collapse
|
19
|
Gu S, Shevchik VE, Shaw R, Pickersgill RW, Garnett JA. The role of intrinsic disorder and dynamics in the assembly and function of the type II secretion system. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:1255-1266. [PMID: 28733198 DOI: 10.1016/j.bbapap.2017.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/02/2017] [Accepted: 07/17/2017] [Indexed: 10/19/2022]
Abstract
Many Gram-negative commensal and pathogenic bacteria use a type II secretion system (T2SS) to transport proteins out of the cell. These exported proteins or substrates play a major role in toxin delivery, maintaining biofilms, replication in the host and subversion of host immune responses to infection. We review the current structural and functional work on this system and argue that intrinsically disordered regions and protein dynamics are central for assembly, exo-protein recognition, and secretion competence of the T2SS. The central role of intrinsic disorder-order transitions in these processes may be a particular feature of type II secretion.
Collapse
Affiliation(s)
- Shuang Gu
- Queen Mary University of London, School of Biological and Chemical Sciences, London E1 4NS, United Kingdom
| | - Vladimir E Shevchik
- Université de Lyon, F-69003, Université Lyon 1, Lyon, F-69622, INSA-Lyon, Villeurbanne F-69621, CNRS, UMR5240, Microbiologie Adaptation et Pathogénie, Lyon F-69622, France
| | - Rosie Shaw
- Queen Mary University of London, School of Biological and Chemical Sciences, London E1 4NS, United Kingdom
| | - Richard W Pickersgill
- Queen Mary University of London, School of Biological and Chemical Sciences, London E1 4NS, United Kingdom.
| | - James A Garnett
- Queen Mary University of London, School of Biological and Chemical Sciences, London E1 4NS, United Kingdom.
| |
Collapse
|
20
|
Thomassin JL, Santos Moreno J, Guilvout I, Tran Van Nhieu G, Francetic O. The trans-envelope architecture and function of the type 2 secretion system: new insights raising new questions. Mol Microbiol 2017; 105:211-226. [DOI: 10.1111/mmi.13704] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Jenny-Lee Thomassin
- Department of structural biology and chemistry, Biochemistry of Macromolecular Interactions Unit; Institut Pasteur; 28 rue du Dr Roux 75724 Paris Cedex 15 France
- Centre National de la Recherche Scientifique (CNRS); ERL6002 75724 Paris France
| | - Javier Santos Moreno
- Université Paris Diderot (Paris 7) Sorbonne Paris Cité; Paris France
- Laboratory of Intercellular Communication and Microbial Infections; CIRB, Collège de France; 11 Place Marcelin Berthelot 75005 Paris France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050; 75005 Paris France
- Centre National de la Recherche Scientifique (CNRS), UMR7241; 75005 Paris France
- MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres; 75005 Paris France
| | - Ingrid Guilvout
- Department of structural biology and chemistry, Biochemistry of Macromolecular Interactions Unit; Institut Pasteur; 28 rue du Dr Roux 75724 Paris Cedex 15 France
- Centre National de la Recherche Scientifique (CNRS); ERL6002 75724 Paris France
| | - Guy Tran Van Nhieu
- Laboratory of Intercellular Communication and Microbial Infections; CIRB, Collège de France; 11 Place Marcelin Berthelot 75005 Paris France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050; 75005 Paris France
- Centre National de la Recherche Scientifique (CNRS), UMR7241; 75005 Paris France
- MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres; 75005 Paris France
| | - Olivera Francetic
- Department of structural biology and chemistry, Biochemistry of Macromolecular Interactions Unit; Institut Pasteur; 28 rue du Dr Roux 75724 Paris Cedex 15 France
- Centre National de la Recherche Scientifique (CNRS); ERL6002 75724 Paris France
| |
Collapse
|
21
|
Abstract
Surface plasmon resonance (SPR) is one of the most commonly used techniques to study protein-protein interactions. The main advantage of SPR is it gives on the ability to measure the binding affinities and association/dissociation kinetics of complexes in real time, in a label-free environment, and using relatively small quantities of materials. The method is based on the immobilization of one of the binding partners, called the ligand, on a dedicated sensor surface. Immobilization is followed by the injection of the other partner, called the analyte, over the surface containing the ligand. The binding is monitored by subsequent changes in the refractive index of the medium close to the sensor surface upon injection of the analyte. During the last 10 years, SPR has been intensively used in the study of secretion systems because of its ability to detect highly dynamic complexes that are difficult to investigate using other techniques. This chapter will guide users in the setup of SPR experiments in order to identify protein complexes and to assess their binding affinity or kinetics. It will include detailed protocols for (i) the immobilization of proteins with the amine coupling capture method, (ii) analyte-binding analysis, (iii) affinity/kinetic measurements, and (iv) data analysis.Secretion systems are multiprotein complexes allowing the transport of a large number of effectors from the inside to the outside of bacterial cells. The assembly of these supramolecular machineries is ensured by the formation of protein complexes with extremely different times of stability, from transitory to stable interactions. To understand the function of these machineries as well as their modes of association, it is important to study their building blocks by identifying the different interacting partners and assessing their relative affinities and association/dissociation kinetics. For that purpose, scientists combine genetic, biochemical, and biophysical tools. During the last decade, the use of surface plasmon resonance (SPR) in the study of secretion systems has increased spectacularly [1-12]. This in vitro approach is the method of choice to study such dynamic systems owing to its ability to detect both weak and strong interactions ranging from the millimolar to the nanomolar range [13, 14]. SPR can be used as a primary tool to screen interacting partners or as a validation tool for interactions previously identified by other methods (e.g., bacterial two-hybrid, co-immunoprecipitation, chemical crosslinking). The determination of the affinity or kinetics of an interaction, as can be done by SPR, is fundamental to understanding the nature of binding at the cellular level.
Collapse
Affiliation(s)
- Badreddine Douzi
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM, UMR 7255), Institut de Microbiologie de la Méditerranée (IMM), Aix-Marseille Université-Centre National de la Recherche Scientifique (CNRS), 31 Chemin Joseph Aiguier, 13402, Marseille Cedex 20, France.
| |
Collapse
|
22
|
Ng D, Harn T, Altindal T, Kolappan S, Marles JM, Lala R, Spielman I, Gao Y, Hauke CA, Kovacikova G, Verjee Z, Taylor RK, Biais N, Craig L. The Vibrio cholerae Minor Pilin TcpB Initiates Assembly and Retraction of the Toxin-Coregulated Pilus. PLoS Pathog 2016; 12:e1006109. [PMID: 27992883 PMCID: PMC5207764 DOI: 10.1371/journal.ppat.1006109] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/03/2017] [Accepted: 12/02/2016] [Indexed: 01/03/2023] Open
Abstract
Type IV pilus (T4P) systems are complex molecular machines that polymerize major pilin proteins into thin filaments displayed on bacterial surfaces. Pilus functions require rapid extension and depolymerization of the pilus, powered by the assembly and retraction ATPases, respectively. A set of low abundance minor pilins influences pilus dynamics by unknown mechanisms. The Vibrio cholerae toxin-coregulated pilus (TCP) is among the simplest of the T4P systems, having a single minor pilin TcpB and lacking a retraction ATPase. Here we show that TcpB, like its homolog CofB, initiates pilus assembly. TcpB co-localizes with the pili but at extremely low levels, equivalent to one subunit per pilus. We used a micropillars assay to demonstrate that TCP are retractile despite the absence of a retraction ATPase, and that retraction relies on TcpB, as a V. cholerae tcpB Glu5Val mutant is fully piliated but does not induce micropillars movements. This mutant is impaired in TCP-mediated autoagglutination and TcpF secretion, consistent with retraction being required for these functions. We propose that TcpB initiates pilus retraction by incorporating into the growing pilus in a Glu5-dependent manner, which stalls assembly and triggers processive disassembly. These results provide a framework for understanding filament dynamics in more complex T4P systems and the closely related Type II secretion system. Bacterial pathogens utilize a number of highly complex and sophisticated molecular systems to colonize their hosts and alter them, creating customized niches in which to reproduce. One such system is the Type IV pilus system, made up of dozens of proteins that form a macromolecular machine to polymerize small pilin proteins into long thin filaments that are displayed on the bacterial surface. These pili have a remarkable array of functions that rely on their ability to (i) adhere to many substrates, including host cell surfaces, pili from nearby bacteria, DNA and bacterial viruses (bacteriophage), and (ii) to depolymerize or retract, which pulls the bacteria along mucosal surfaces, pulls them close together in protective aggregates, and can even draw in substrates like DNA and bacteriophage for nutrition and genetic variation. For most Type IV pilus systems, retraction is an energy-driven process facilitated by a retraction ATPase. We show here that in the simplest of the Type IV pilus systems, the Vibrio cholerae toxin-coregulated pilus, a pilin-like protein initiates pilus retraction by what appears to be mechanical rather than enzymatic means. Our results provide a framework for understanding more complex Type IV pili and the related Type II secretion systems, which represent targets for novel highly specific antibiotics.
Collapse
Affiliation(s)
- Dixon Ng
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Tony Harn
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Tuba Altindal
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Subramania Kolappan
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jarrad M. Marles
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Rajan Lala
- Biology Department, Brooklyn College, City University of New York, Brooklyn, New York, United States of America
| | - Ingrid Spielman
- Biology Department, Brooklyn College, City University of New York, Brooklyn, New York, United States of America
| | - Yang Gao
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Caitlyn A. Hauke
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Gabriela Kovacikova
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Zia Verjee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Ronald K. Taylor
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Nicolas Biais
- Biology Department, Brooklyn College, City University of New York, Brooklyn, New York, United States of America
- Graduate Center, City University of New York, Brooklyn, New York, United States of America
- * E-mail: (LC); (NB)
| | - Lisa Craig
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- * E-mail: (LC); (NB)
| |
Collapse
|
23
|
Nivaskumar M, Santos-Moreno J, Malosse C, Nadeau N, Chamot-Rooke J, Tran Van Nhieu G, Francetic O. Pseudopilin residue E5 is essential for recruitment by the type 2 secretion system assembly platform. Mol Microbiol 2016; 101:924-41. [DOI: 10.1111/mmi.13432] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Mangayarkarasi Nivaskumar
- Department of Microbiology, Laboratory of Macromolecular Systems and Signalling; Institut Pasteur, CNRS ERL3526; 25 rue du Dr Roux 75724 Paris, Cedex 15 France
- Université Paris Diderot (Paris 7) Sorbonne Paris Cité
| | - Javier Santos-Moreno
- Université Paris Diderot (Paris 7) Sorbonne Paris Cité
- Laboratory of Intercellular Communication and Microbial Infections; CIRB, Collège de France; Paris France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050; France
- Centre National de la Recherche Scientifique (CNRS), UMR7241; France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre; France
| | - Christian Malosse
- Structural Mass spectrometry and Proteomics unit; CNRS UMR3528, Institut Pasteur; Paris France
| | - Nathalie Nadeau
- Department of Microbiology, Laboratory of Macromolecular Systems and Signalling; Institut Pasteur, CNRS ERL3526; 25 rue du Dr Roux 75724 Paris, Cedex 15 France
| | - Julia Chamot-Rooke
- Structural Mass spectrometry and Proteomics unit; CNRS UMR3528, Institut Pasteur; Paris France
| | - Guy Tran Van Nhieu
- Laboratory of Intercellular Communication and Microbial Infections; CIRB, Collège de France; Paris France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050; France
- Centre National de la Recherche Scientifique (CNRS), UMR7241; France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre; France
| | - Olivera Francetic
- Department of Microbiology, Laboratory of Macromolecular Systems and Signalling; Institut Pasteur, CNRS ERL3526; 25 rue du Dr Roux 75724 Paris, Cedex 15 France
| |
Collapse
|
24
|
Leighton TL, Buensuceso RNC, Howell PL, Burrows LL. Biogenesis of Pseudomonas aeruginosa type IV pili and regulation of their function. Environ Microbiol 2015; 17:4148-63. [PMID: 25808785 DOI: 10.1111/1462-2920.12849] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 03/13/2015] [Accepted: 03/14/2015] [Indexed: 12/27/2022]
Abstract
Type IV pili (T4P) are bacterial virulence factors involved in a wide variety of functions including deoxyribonucleic acid uptake, surface attachment, biofilm formation and twitching motility. While T4P are common surface appendages, the systems that assemble them and the regulation of their function differ between species. Pseudomonas aeruginosa, Neisseria spp. and Myxococcus xanthus are common model systems used to study T4P biology. This review focuses on recent advances in P. aeruginosa T4P structural biology, and the regulatory pathways controlling T4P biogenesis and function.
Collapse
Affiliation(s)
- Tiffany L Leighton
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Ryan N C Buensuceso
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - P Lynne Howell
- Program in Molecular Structure & Function, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lori L Burrows
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
25
|
Affiliation(s)
- Alain Filloux
- Alain Filloux, MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; E-mail:
| |
Collapse
|
26
|
Berry JL, Pelicic V. Exceptionally widespread nanomachines composed of type IV pilins: the prokaryotic Swiss Army knives. FEMS Microbiol Rev 2014; 39:134-54. [PMID: 25793961 PMCID: PMC4471445 DOI: 10.1093/femsre/fuu001] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Prokaryotes have engineered sophisticated surface nanomachines that have allowed them to colonize Earth and thrive even in extreme environments. Filamentous machineries composed of type IV pilins, which are associated with an amazing array of properties ranging from motility to electric conductance, are arguably the most widespread since distinctive proteins dedicated to their biogenesis are found in most known species of prokaryotes. Several decades of investigations, starting with type IV pili and then a variety of related systems both in bacteria and archaea, have outlined common molecular and structural bases for these nanomachines. Using type IV pili as a paradigm, we will highlight in this review common aspects and key biological differences of this group of filamentous structures. Using type IV pili as a paradigm, we review common genetic, structural and mechanistic features (many) as well as differences (few) of the exceptionally widespread and functionally versatile prokaryotic nano-machines composed of type IV pilins.
Collapse
Affiliation(s)
- Jamie-Lee Berry
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK
| | - Vladimir Pelicic
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
27
|
Nguyen Y, Sugiman-Marangos S, Harvey H, Bell SD, Charlton CL, Junop MS, Burrows LL. Pseudomonas aeruginosa minor pilins prime type IVa pilus assembly and promote surface display of the PilY1 adhesin. J Biol Chem 2014; 290:601-11. [PMID: 25389296 DOI: 10.1074/jbc.m114.616904] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Type IV pili (T4P) contain hundreds of major subunits, but minor subunits are also required for assembly and function. Here we show that Pseudomonas aeruginosa minor pilins prime pilus assembly and traffic the pilus-associated adhesin and anti-retraction protein, PilY1, to the cell surface. PilV, PilW, and PilX require PilY1 for inclusion in surface pili and vice versa, suggestive of complex formation. PilE requires PilVWXY1 for inclusion, suggesting that it binds a novel interface created by two or more components. FimU is incorporated independently of the others and is proposed to couple the putative minor pilin-PilY1 complex to the major subunit. The production of small amounts of T4P by a mutant lacking the minor pilin operon was traced to expression of minor pseudopilins from the P. aeruginosa type II secretion (T2S) system, showing that under retraction-deficient conditions, T2S minor subunits can prime T4P assembly. Deletion of all minor subunits abrogated pilus assembly. In a strain lacking the minor pseudopilins, PilVWXY1 and either FimU or PilE comprised the minimal set of components required for pilus assembly. Supporting functional conservation of T2S and T4P minor components, our 1.4 Å crystal structure of FimU revealed striking architectural similarity to its T2S ortholog GspH, despite minimal sequence identity. We propose that PilVWXY1 form a priming complex for assembly and that PilE and FimU together stably couple the complex to the major subunit. Trafficking of the anti-retraction factor PilY1 to the cell surface allows for production of pili of sufficient length to support adherence and motility.
Collapse
Affiliation(s)
- Ylan Nguyen
- From the Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Seiji Sugiman-Marangos
- From the Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Hanjeong Harvey
- From the Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Stephanie D Bell
- From the Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Carmen L Charlton
- the Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada, the Provincial Laboratory for Public Health, Edmonton, Alberta T6G 2J2, Canada, and
| | - Murray S Junop
- the Department of Biochemistry, Western University, London, Ontario N6A 3K7, Canada
| | - Lori L Burrows
- From the Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8N 3Z5, Canada,
| |
Collapse
|
28
|
Type II secretion system: A magic beanstalk or a protein escalator. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1568-77. [DOI: 10.1016/j.bbamcr.2013.12.020] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/13/2013] [Accepted: 12/23/2013] [Indexed: 12/12/2022]
|
29
|
The 1.59Å resolution structure of the minor pseudopilin EpsH of Vibrio cholerae reveals a long flexible loop. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:406-15. [DOI: 10.1016/j.bbapap.2013.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/01/2013] [Accepted: 11/24/2013] [Indexed: 01/25/2023]
|
30
|
Mann JM, Carabetta VJ, Cristea IM, Dubnau D. Complex formation and processing of the minor transformation pilins of Bacillus subtilis. Mol Microbiol 2013; 90:1201-15. [PMID: 24164455 PMCID: PMC5687075 DOI: 10.1111/mmi.12425] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2013] [Indexed: 01/06/2023]
Abstract
Transformation in most bacteria is dependent on orthologues of Type 2 secretion and Type 4 pilus system proteins. In each system, pilin proteins (major and minor) are required to make the pilus structure and are essential to the process, although the precise roles of the minor pilins remain unclear. We have explored protein-protein interactions among the competence minor pilins of Bacillus subtilis through in vitro binding studies, immunopurification and mass spectrometry. We demonstrate that the minor pilins directly interact, and the minor pilin ComGG interacts with most of the known proteins required for transformation. We find that ComGG requires other ComG proteins for its stabilization and for processing by the pre-pilin peptidase. These observations, C-terminal mutations in ComGG that prevent processing and the inaccessibility of pre-ComGG to externally added protease suggest a model in which pre-ComGG must be associated with other minor pilins for processing to take place. We propose that ComGG does not become a transmembrane protein until after processing. These behaviours contrast with that of pre-ComGC, the major pilin, which is accessible to externally added protease and requires only the peptidase to be processed. The roles of the pilins and of the pilus in transformation are discussed.
Collapse
Affiliation(s)
- Jessica M. Mann
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 07103, USA
| | - Valerie J. Carabetta
- Public Health Research Institute, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 07103, USA
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - David Dubnau
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 07103, USA
- Public Health Research Institute, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 07103, USA
| |
Collapse
|
31
|
Lallemand M, Login FH, Guschinskaya N, Pineau C, Effantin G, Robert X, Shevchik VE. Dynamic interplay between the periplasmic and transmembrane domains of GspL and GspM in the type II secretion system. PLoS One 2013; 8:e79562. [PMID: 24223969 PMCID: PMC3815138 DOI: 10.1371/journal.pone.0079562] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/24/2013] [Indexed: 11/29/2022] Open
Abstract
The type II secretion system (T2SS) is a multiprotein nanomachine that transports folded proteins across the outer membrane of gram-negative bacteria. The molecular mechanisms that govern the secretion process remain poorly understood. The inner membrane components GspC, GspL and GspM possess a single transmembrane segment (TMS) and a large periplasmic region and they are thought to form a platform of unknown function. Here, using two-hybrid and pull-down assays we performed a systematic mapping of the GspC/GspL/GspM interaction regions in the plant pathogen Dickeya dadantii. We found that the TMS of these components interact with each other, implying a complex interaction network within the inner membrane. We also showed that the periplasmic, ferredoxin-like, domains of GspL and GspM drive homo- and heterodimerizations of these proteins. Disulfide bonding analyses revealed that the respective domain interfaces include the equivalent secondary-structure elements, suggesting alternating interactions of the periplasmic domains, L/L and M/M versus L/M. Finally, we found that displacements of the periplasmic GspM domain mediate coordinated shifts or rotations of the cognate TMS. These data suggest a plausible mechanism for signal transmission between the periplasmic and the cytoplasmic portions of the T2SS machine.
Collapse
Affiliation(s)
- Mathilde Lallemand
- INSA-Lyon, Villeurbanne, France
- CNRS, UMR5240, Microbiologie Adaptation et Pathogénie, Lyon, France
| | - Frédéric H. Login
- Université Lyon 1, Lyon, France
- CNRS, UMR5240, Microbiologie Adaptation et Pathogénie, Lyon, France
| | - Natalia Guschinskaya
- Université Lyon 1, Lyon, France
- CNRS, UMR5240, Microbiologie Adaptation et Pathogénie, Lyon, France
| | - Camille Pineau
- INSA-Lyon, Villeurbanne, France
- CNRS, UMR5240, Microbiologie Adaptation et Pathogénie, Lyon, France
| | | | - Xavier Robert
- Université Lyon 1, Lyon, France
- Laboratory for Biocrystallography and Structural Biology of Therapeutic Targets, Molecular and Structural Bases of Infectious Diseases, CNRS UMR5086, Lyon, France
| | - Vladimir E. Shevchik
- Université Lyon 1, Lyon, France
- INSA-Lyon, Villeurbanne, France
- CNRS, UMR5240, Microbiologie Adaptation et Pathogénie, Lyon, France
- * E-mail:
| |
Collapse
|
32
|
The type II secretion system – a dynamic fiber assembly nanomachine. Res Microbiol 2013; 164:545-55. [DOI: 10.1016/j.resmic.2013.03.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 02/28/2013] [Indexed: 11/17/2022]
|
33
|
Abstract
The type II secretion system is utilized by many Gram-negative bacteria to export folded proteins to the surface and/or the extracellular environment of the cell. Although the function of the system is to move proteins from the periplasm to the outside of the cell, it is a large trans-envelope structure composed of more than a dozen different proteins present in multiple copies, including peripheral, integral inner membrane and integral outer membrane proteins plus a pseudopilus stretching between them. The establishment of this structure as an integral component of the entire envelope including the peptidoglycan layer between the two membranes requires assembly. Many of the participants and processes involved in this assembly have now been established, while other aspects remain to be discovered or more fully understood.
Collapse
Affiliation(s)
- S Peter Howard
- Department of Microbiology and Immunology, University of Saskatchewan, Health Sciences Building, Room 2D01, 107 Wiggins Road, Saskatoon, Saskatchewan, Canada S7N 5E5.
| |
Collapse
|
34
|
Affiliation(s)
- Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| |
Collapse
|
35
|
Cisneros DA, Pehau-Arnaudet G, Francetic O. Heterologous assembly of type IV pili by a type II secretion system reveals the role of minor pilins in assembly initiation. Mol Microbiol 2012; 86:805-18. [PMID: 23006128 DOI: 10.1111/mmi.12033] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2012] [Indexed: 12/16/2022]
Abstract
In Gram-negative bacteria, type IV pilus assembly (T4PS) and type II secretion (T2SS) systems polymerize inner membrane proteins called major pilins or pseudopilins respectively, into thin filaments. Four minor pilins are required in both systems for efficient fibre assembly. Escherichia coli K-12 has a set of T4PS assembly genes that are silent under standard growth conditions. We studied the heterologous assembly of the E. coli type IV pilin PpdD by the Klebsiella oxytoca T2SS called the Pul system. PpdD pilus assembly in this context depended on the expression of the K. oxytoca minor pseudopilin genes pulHIJK or of the E. coli minor pilin genes ppdAB-ygdB-ppdC. The E. coli minor pilins restored assembly of the major pseudopilin PulG in a pulHIJK mutant, but not the secretion of the T2SS substrate pullulanase. Thus, minor pilins and minor pseudopilins are functionally interchangeable in initiating major pilin assembly, further extending the fundamental similarities between the two systems. The data suggest that, in both systems, minor pilins activate the assembly machinery through a common self-assembly mechanism. When produced together, PulG and PpdD assembled into distinct homopolymers, establishing major pilins as key determinants of pilus elongation and structure.
Collapse
Affiliation(s)
- David A Cisneros
- Molecular Genetics Unit, Department of Microbiology, Institut Pasteur, 75015, Paris, France
| | | | | |
Collapse
|
36
|
Burrows LL. Prime time for minor subunits of the type II secretion and type IV pilus systems. Mol Microbiol 2012; 86:765-9. [PMID: 22998166 DOI: 10.1111/mmi.12034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2012] [Indexed: 10/27/2022]
Abstract
The type II secretion system (T2SS) exports folded proteins from the periplasms of Gram-negative bacteria. The type IV pilus system (T4PS) is a multifunctional machine used for adherence, motility and DNA transfer in bacteria and archaea. Partial sequence identity between the two systems suggests that they are related and might function via a similar mechanism, the dynamic assembly and disassembly of pseudopilus (T2SS) or pilus (T4PS) filaments. The major subunit in each system is thought to form the bulk of the (pseudo)pilus, while minor (low-abundance) subunits have proposed roles in assembly initiation, antagonism of disassembly, or modulation of (pseudo)pilus functional properties. In this issue, Cisneros et al. () extend their previous finding that pseudopilus assembly is primed by the minor pseudopilins, showing that the same proteins can initiate assembly of Escherichia coli T4P. Similarly, they show that the E. coli minor pilins prime the polymerization of T2S pseudopili, although unlike genuine pseudopili, the chimeric filaments did not support secretion. This work reinforces the notion of a common assembly mechanism for the T2S and T4P systems.
Collapse
Affiliation(s)
- Lori L Burrows
- Department of Biochemistry and Biomedical Sciences, the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, Ontario, Canada, L8S4K1.
| |
Collapse
|
37
|
Douzi B, Filloux A, Voulhoux R. On the path to uncover the bacterial type II secretion system. Philos Trans R Soc Lond B Biol Sci 2012; 367:1059-72. [PMID: 22411978 DOI: 10.1098/rstb.2011.0204] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gram-negative bacteria have evolved several secretory pathways to release enzymes or toxins into the surrounding environment or into the target cells. The type II secretion system (T2SS) is conserved in Gram-negative bacteria and involves a set of 12 to 16 different proteins. Components of the T2SS are located in both the inner and outer membranes where they assemble into a supramolecular complex spanning the bacterial envelope, also called the secreton. The T2SS substrates transiently go through the periplasm before they are translocated across the outer membrane and exposed to the extracellular milieu. The T2SS is unique in its ability to promote secretion of large and sometimes multimeric proteins that are folded in the periplasm. The present review describes recently identified protein-protein interactions together with structural and functional advances in the field that have contributed to improve our understanding on how the type II secretion apparatus assembles and on the role played by individual proteins of this highly sophisticated system.
Collapse
Affiliation(s)
- Badreddine Douzi
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (CNRS-LISM-UPR 9027), Aix-Marseille Universités, Institut de Microbiologie de la Méditerranée, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | | | | |
Collapse
|
38
|
The type II secretion system: biogenesis, molecular architecture and mechanism. Nat Rev Microbiol 2012; 10:336-51. [PMID: 22466878 DOI: 10.1038/nrmicro2762] [Citation(s) in RCA: 347] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Many gram-negative bacteria use the sophisticated type II secretion system (T2SS) to translocate a wide range of proteins from the periplasm across the outer membrane. The inner-membrane platform of the T2SS is the nexus of the system and orchestrates the secretion process through its interactions with the periplasmic filamentous pseudopilus, the dodecameric outer-membrane complex and a cytoplasmic secretion ATPase. Here, recent structural and biochemical information is reviewed to describe our current knowledge of the biogenesis and architecture of the T2SS and its mechanism of action.
Collapse
|
39
|
McLaughlin LS, Haft RJF, Forest KT. Structural insights into the Type II secretion nanomachine. Curr Opin Struct Biol 2012; 22:208-16. [PMID: 22425326 DOI: 10.1016/j.sbi.2012.02.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 02/16/2012] [Accepted: 02/23/2012] [Indexed: 01/31/2023]
Abstract
The Type II secretion nanomachine transports folded proteins across the outer membrane of Gram-negative bacteria. Recent X-ray crystallography, electron microscopy, and molecular modeling studies provide structural insights into three functionally and spatially connected units of this nanomachine: the cytoplasmic and inner membrane energy-harvesting complex, the periplasmic helical pseudopilus, and the outer membrane secretin. Key advances include cryo-EM reconstruction of the secretin and demonstration that it interacts with both secreted substrates and a crucial transmembrane clamp protein, plus a biochemical and structural explanation of the role of low-abundance pseudopilins in initiating pseudopilus growth. Combining structures and protein interactions, we synthesize a 3D view of the complete complex consistent with a stepwise pathway in which secretin oligomerization defines sites of nanomachine biogenesis.
Collapse
|
40
|
Minor pseudopilin self-assembly primes type II secretion pseudopilus elongation. EMBO J 2011; 31:1041-53. [PMID: 22157749 DOI: 10.1038/emboj.2011.454] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 11/11/2011] [Indexed: 12/23/2022] Open
Abstract
In Gram-negative bacteria, type II secretion systems (T2SS) assemble inner membrane proteins of the major pseudopilin PulG (GspG) family into periplasmic filaments, which could drive protein secretion in a piston-like manner. Three minor pseudopilins PulI, PulJ and PulK are essential for protein secretion in the Klebsiella oxytoca T2SS, but their molecular function is unknown. Here, we demonstrate that together these proteins prime pseudopilus assembly, without actively controlling its length or secretin channel opening. Using molecular dynamics, bacterial two-hybrid assays, cysteine crosslinking and functional analysis, we show that PulI and PulJ nucleate filament assembly by forming a staggered complex in the plasma membrane. Binding of PulK to this complex results in its partial extraction from the membrane and in a 1-nm shift between their transmembrane segments, equivalent to the major pseudopilin register in the assembled PulG filament. This promotes fully efficient pseudopilus assembly and protein secretion. Therefore, we propose that PulI, PulJ and PulK self-assembly is thermodynamically coupled to the initiation of pseudopilus assembly, possibly setting the assembly machinery in motion.
Collapse
|
41
|
Douzi B, Ball G, Cambillau C, Tegoni M, Voulhoux R. Deciphering the Xcp Pseudomonas aeruginosa type II secretion machinery through multiple interactions with substrates. J Biol Chem 2011; 286:40792-801. [PMID: 21949187 DOI: 10.1074/jbc.m111.294843] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The type II secretion system enables gram-negative bacteria to secrete exoproteins into the extracellular milieu. We performed biophysical and biochemical experiments to identify systematic interactions between Pseudomonas aeruginosa Xcp type II secretion system components and their substrates. We observed that three Xcp components, XcpP(C), the secretin XcpQ(D), and the pseudopilus tip, directly and specifically interact with secreted exoproteins. We established that XcpP(C), in addition to its interaction with the substrate, likely shields the entire periplasmic portion of the secreton. It can therefore be considered as the recruiter of the machinery. Moreover, the direct interaction observed between the substrate and the pseudopilus tip validates the piston model hypothesis, in which the pseudopilus pushes the substrate through the secretin pore during the secretion process. All together, our results allowed us to propose a model of the different consecutive steps followed by the substrate during the type II secretion process.
Collapse
Affiliation(s)
- Badreddine Douzi
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM-UPR9027), Institut de Microbiologie de la Méditerranée, CNRS, Aix-Marseille Université, 31 Chemin Joseph Aiguier, 13402 Marseille cedex 20, France
| | | | | | | | | |
Collapse
|
42
|
Filloux A. Protein Secretion Systems in Pseudomonas aeruginosa: An Essay on Diversity, Evolution, and Function. Front Microbiol 2011; 2:155. [PMID: 21811488 PMCID: PMC3140646 DOI: 10.3389/fmicb.2011.00155] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/01/2011] [Indexed: 12/25/2022] Open
Abstract
Protein secretion systems are molecular nanomachines used by Gram-negative bacteria to thrive within their environment. They are used to release enzymes that hydrolyze complex carbon sources into usable compounds, or to release proteins that capture essential ions such as iron. They are also used to colonize and survive within eukaryotic hosts, causing acute or chronic infections, subverting the host cell response and escaping the immune system. In this article, the opportunistic human pathogen Pseudomonas aeruginosa is used as a model to review the diversity of secretion systems that bacteria have evolved to achieve these goals. This diversity may result from a progressive transformation of cell envelope complexes that initially may not have been dedicated to secretion. The striking similarities between secretion systems and type IV pili, flagella, bacteriophage tail, or efflux pumps is a nice illustration of this evolution. Differences are also needed since various secretion configurations call for diversity. For example, some proteins are released in the extracellular medium while others are directly injected into the cytosol of eukaryotic cells. Some proteins are folded before being released and transit into the periplasm. Other proteins cross the whole cell envelope at once in an unfolded state. However, the secretion system requires conserved basic elements or features. For example, there is a need for an energy source or for an outer membrane channel. The structure of this review is thus quite unconventional. Instead of listing secretion types one after each other, it presents a melting pot of concepts indicating that secretion types are in constant evolution and use basic principles. In other words, emergence of new secretion systems could be predicted the way Mendeleïev had anticipated characteristics of yet unknown elements.
Collapse
Affiliation(s)
- Alain Filloux
- Division of Cell and Molecular Biology, Centre for Molecular Microbiology and Infection, Imperial College London London, UK
| |
Collapse
|
43
|
Durand E, Alphonse S, Brochier-Armanet C, Ball G, Douzi B, Filloux A, Bernard C, Voulhoux R. The assembly mode of the pseudopilus: a hallmark to distinguish a novel secretion system subtype. J Biol Chem 2011; 286:24407-16. [PMID: 21586577 DOI: 10.1074/jbc.m111.234278] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In gram-negative bacteria, type II secretion systems assemble a piston-like structure, called pseudopilus, which expels exoproteins out of the cell. The pseudopilus is constituted by a major pseudopilin that when overproduced multimerizes into a long cell surface structure named hyper-pseudopilus. Pseudomonas aeruginosa possesses two type II secretion systems, Xcp and Hxc. Although major pseudopilins are exchangeable among type II secretion systems, we show that XcpT and HxcT are not. We demonstrate that HxcT does not form a hyper-pseudopilus and is different in amino acid sequence and multimerization properties. Using structure-based mutagenesis, we observe that five mutations are sufficient to revert HxcT into a functional XcpT-like protein, which also becomes capable of forming a hyper-pseudopilus. Phylogenetic and experimental analysis showed that the whole Hxc system was acquired by P. aeruginosa PAO1 and other Pseudomonas species through horizontal gene transfer. We thus identified a new type II secretion subfamily, of which the P. aeruginosa Hxc system is the archetype. This finding demonstrates how similar bacterial machineries evolve toward distinct mechanisms that may contribute specific functions.
Collapse
Affiliation(s)
- Eric Durand
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM-UPR9027), CNRS, Université de la Méditerranée, Institut de Microbiologie de la Méditerranée, 31 Chemin Joseph Aiguier, 13402 Marseille Cedex 20, France
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Reichow SL, Korotkov KV, Gonen M, Sun J, Delarosa JR, Hol WGJ, Gonen T. The binding of cholera toxin to the periplasmic vestibule of the type II secretion channel. Channels (Austin) 2011; 5:215-8. [PMID: 21406971 DOI: 10.4161/chan.5.3.15268] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The type II secretion system (T2SS) is a large macromolecular complex spanning the inner and outer membranes of many gram-negative bacteria. The T2SS is responsible for the secretion of virulence factors such as cholera toxin (CT) and heat-labile enterotoxin (LT) from Vibrio cholerae and enterotoxigenic Escherichia coli, respectively. CT and LT are closely related AB5 heterohexamers, composed of one A subunit and a B-pentamer. Both CT and LT are translocated, as folded protein complexes, from the periplasm across the outer membrane through the type II secretion channel, the secretin GspD. We recently published the 19 Å structure of the V. cholerae secretin (VcGspD) in its closed state and showed by SPR measurements that the periplasmic domain of GspD interacts with the B-pentamer complex. Here we extend these studies by characterizing the binding of the cholera toxin B-pentamer to VcGspD using electron microscopy of negatively stained preparations. Our studies indicate that the pentamer is captured within the large periplasmic vestibule of VcGspD. These new results agree well with our previously published studies and are in accord with a piston-driven type II secretion mechanism.
Collapse
Affiliation(s)
- Steve L Reichow
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Gray MD, Bagdasarian M, Hol WGJ, Sandkvist M. In vivo cross-linking of EpsG to EpsL suggests a role for EpsL as an ATPase-pseudopilin coupling protein in the Type II secretion system of Vibrio cholerae. Mol Microbiol 2011; 79:786-98. [PMID: 21255118 DOI: 10.1111/j.1365-2958.2010.07487.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The type II secretion system is a multi-protein complex that spans the cell envelope of Gram-negative bacteria and promotes the secretion of proteins, including several virulence factors. This system is homologous to the type IV pilus biogenesis machinery and contains five proteins, EpsG-K, termed the pseudopilins that are structurally homologous to the type IV pilins. The major pseudopilin EpsG has been proposed to form a pilus-like structure in an energy-dependent process that requires the ATPase, EpsE. A key remaining question is how the membrane-bound EpsG interacts with the cytoplasmic ATPase, and if this is a direct or indirect interaction. Previous studies have established an interaction between the bitopic inner membrane protein EpsL and EpsE; therefore, in this study we used in vivo cross-linking to test the hypothesis that EpsG interacts with EpsL. Our findings suggest that EpsL may function as a scaffold to link EpsG and EpsE and thereby transduce the energy generated by ATP hydrolysis to support secretion. The recent discovery of structural homology between EpsL and a protein in the type IV pilus system implies that this interaction may be conserved and represent an important functional interaction for both the type II secretion and type IV pilus systems.
Collapse
Affiliation(s)
- Miranda D Gray
- University of Michigan Medical School, Department of Microbiology and Immunology, Ann Arbor, MI 48109, USA
| | | | | | | |
Collapse
|
46
|
Franz LP, Douzi B, Durand E, Dyer DH, Voulhoux R, Forest KT. Structure of the minor pseudopilin XcpW from the Pseudomonas aeruginosa type II secretion system. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2011; 67:124-30. [PMID: 21245534 DOI: 10.1107/s0907444910051954] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 12/11/2010] [Indexed: 11/11/2022]
Abstract
Pseudomonas aeruginosa utilizes the type II secretion machinery to transport virulence factors through the outer membrane into the extracellular space. Five proteins in the type II secretion system share sequence homology with pilin subunits of type IV pili and are called the pseudopilins. The major pseudopilin XcpT(G) assembles into an intraperiplasmic pilus and is thought to act in a piston-like manner to push substrates through an outer membrane secretin. The other four minor pseudopilins, XcpU(H), XcpV(I), XcpW(J) and XcpX(K), play less well defined roles in pseudopilus formation. It was recently discovered that these four minor pseudopilins form a quaternary complex that is presumed to initiate the formation of the pseudopilus and to localize to its tip. Here, the structure of XcpW(J) was refined to 1.85 Å resolution. The structure revealed the type IVa pilin fold with an embellished variable antiparallel β-sheet as also found in the XcpW(J) homologue enterotoxigenic Escherichia coli GspJ(W) and the XcpU(H) homologue Vibrio cholerae EpsU(H). It is proposed that the exposed surface of this sheet may cradle the long N-terminal α1 helix of another pseudopilin. The final 31 amino acids of the XcpW(J) structure are instrinsically disordered. Deletion of this unstructured region of XcpW(J) did not prevent type II secretion in vivo.
Collapse
Affiliation(s)
- Laura P Franz
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
47
|
Mba Medie F, Vincentelli R, Drancourt M, Henrissat B. Mycobacterium tuberculosis Rv1090 and Rv1987 encode functional β-glucan-targeting proteins. Protein Expr Purif 2010; 75:172-6. [PMID: 20826214 DOI: 10.1016/j.pep.2010.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 08/27/2010] [Accepted: 08/30/2010] [Indexed: 01/25/2023]
Abstract
Mycobacterium tuberculosis is a facultative intracellular pathogen, and the ability of this bacterium to survive and to grow inside macrophages is central to its virulence. Multiple strategies are employed by M. tuberculosis to ensure survival in macrophages, including secretion of several proteins, which are good candidates to be virulence factors, drug targets for disease intervention, and vaccine antigens. However, some M. tuberculosis secreted proteins do not appear to play any role in the growth or survival of the bacterium in its mammalian host. Among these proteins are three putative cellulose-targeting proteins encoded by the genes Rv0062, Rv1090, and Rv1987. It has been previously shown that Rv0062 encodes an active cellulase. Here we report that Rv1090 and Rv1987 also encode functional proteins. Rv1090 is able to hydrolyze barley β-glucan while Rv1987 displays cellulose-binding activity on filter paper and on microcrystalline cellulose (Avicel). Collectively, these observations point toward a unique unknown relationship between M. tuberculosis and a cellulose-containing host. We hypothesize that amoeba could be such hosts.
Collapse
Affiliation(s)
- Felix Mba Medie
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UMR CNRS 6236, IRD 198, IFR 48, Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | | | | | | |
Collapse
|
48
|
Abstract
The type II secretion system (T2SS) is used by Escherichia coli and other gram-negative bacteria to translocate many proteins, including toxins and proteases, across the outer membrane of the cell and into the extracellular space. Depending on the bacterial species, between 12 and 15 genes have been identified that make up a T2SS operon. T2SSs are widespread among gram-negative bacteria, and most E. coli appear to possess one or two complete T2SS operons. Once expressed, the multiple protein components that form the T2S system are localized in both the inner and outer membranes, where they assemble into an apparatus that spans the cell envelope. This apparatus supports the secretion of numerous virulence factors; and therefore secretion via this pathway is regarded in many organisms as a major virulence mechanism. Here, we review several of the known E. coli T2S substrates that have proven to be critical for the survival and pathogenicity of these bacteria. Recent structural and biochemical information is also reviewed that has improved our current understanding of how the T2S apparatus functions; also reviewed is the role that individual proteins play in this complex system.
Collapse
|
49
|
Giltner CL, Rana N, Lunardo MN, Hussain AQ, Burrows LL. Evolutionary and functional diversity of the Pseudomonas type IVa pilin island. Environ Microbiol 2010; 13:250-264. [PMID: 20738375 DOI: 10.1111/j.1462-2920.2010.02327.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In Pseudomonas aeruginosa, most proteins involved in type IVa pilus (T4aP) biogenesis are highly conserved except for the major pilin PilA and the minor pilins involved in pilus assembly. Here we show that each of the five major pilin alleles is associated with a specific set of minor pilins, and unrelated strains with the same major pilin type have identical minor pilin genes. The sequences of the minor pilin genes of strains with group III and V pilins are identical, suggesting that these groups diverged recently through further evolution of the major pilin cluster. Both gene clusters are localized on a single 'pilin island' containing putative tRNA recombinational hotspots, and a similar organization of pilin genes was identified in other Pseudomonas species. To address the biological significance of group-specific differences, cross-complementation studies using group II (PAO1) and group III (PA14) minor pilins were performed. Heterologous minor pilins complemented twitching motility to various extents except in the case of PilX, which was non-functional in non-native backgrounds. A recombinant PA14 strain expressing the PAO1 minor pilins regained motility only upon co-introduction of the PA14 pilX gene. Comparison of PilX and PilQ secretin sequences from group II, III and V genomes revealed discrete regions of sequence that co-varied between groups. Our data suggest that changes in PilX sequence have led to compensatory changes in the PilQ secretin monomer such that heterologous PilX proteins are no longer able to promote opening of the secretin to allow pili to appear on the cell surface.
Collapse
Affiliation(s)
- Carmen L Giltner
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada
| | - Navpreet Rana
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada
| | - Michael N Lunardo
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada
| | - Anne Q Hussain
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada
| | - Lori L Burrows
- Department of Biochemistry and Biomedical Sciences and the Michael G. DeGroote Institute for Infectious Diseases Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
50
|
Detailed structural and assembly model of the type II secretion pilus from sparse data. Proc Natl Acad Sci U S A 2010; 107:13081-6. [PMID: 20616068 DOI: 10.1073/pnas.1001703107] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Many gram-negative bacteria secrete specific proteins via the type II secretion systems (T2SS). These complex machineries share with the related archaeal flagella and type IV pilus (T4P) biogenesis systems the ability to assemble thin, flexible filaments composed of small, initially inner membrane-localized proteins called "pilins." In the T2SS from Klebsiella oxytoca, periplasmic pseudopili that are essential for pullulanase (PulA) secretion extend beyond the bacterial surface and form pili when the major pilin PulG is overproduced. Here, we describe the detailed, experimentally validated structure of the PulG pilus generated from crystallographic and electron microscopy data by a molecular modeling approach. Two intermolecular salt bridges crucial for function were demonstrated using single and complementary charge inversions. Double-cysteine substitutions in the transmembrane segment of PulG led to position-specific cross-linking of protomers in assembled pili. These biochemical data provided information on residue distances in the filament that were used to derive a refined model of the T2SS pilus at pseudoatomic resolution. PulG is organized as a right-handed helix of subunits, consistent with protomer organization in gonococcal T4P. The conserved character of residues involved in key hydrophobic and electrostatic interactions within the major pseudopilin family supports the general relevance of this model for T2SS pseudopilus structure.
Collapse
|