1
|
Healy CM, Pham EA, Dye KJ, Rouchon CN, McMillan B, Frank KL. The adjacent ATP-binding protein-encoding genes of the Enterococcus faecalis phosphate-specific transport ( pst) locus have non-overlapping cellular functions. J Bacteriol 2025; 207:e0003325. [PMID: 40227050 PMCID: PMC12096839 DOI: 10.1128/jb.00033-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
The widely conserved pst-phoU operon encodes a low-velocity, high-affinity, ATP-dependent importer for inorganic phosphate (Pi). The pstB gene encodes the ATPase that powers the import of Pi into the cell. In some Firmicutes, including the gastrointestinal commensal and opportunistic pathogen Enterococcus faecalis, the pst-phoU locus contains adjacent pstB genes. In this work, we compared the functionality of E. faecalis pstB1 and pstB2. E. faecalis pstB1 and pstB2 share sequence similarities with verified PstB ATPases from Escherichia coli and Streptococcus pneumoniae and only share ~60% amino acid identity with each other. Deletion of pstB1 was associated with a growth defect in low Pi-containing chemically defined medium (CDM), reduced Pi uptake, and a moderate increase in alkaline phosphatase (AP) activity. Deletion of pstB2 fully inhibited growth in CDM regardless of inorganic phosphorus source but did not hinder growth in rich, undefined medium. The ΔpstB2 mutant also exhibited a significant increase in AP activity that was associated with extracellular Pi accumulation. Overexpression of pstB2 in the pstB1 mutant was sufficient to restore growth in low-Pi CDM, Pi uptake, and AP activity, but this was not recapitulated with overexpression of pstB1 in the ΔpstB2 mutant. Deletion of either pstB paralog increased expression of the tandem paralog, and overexpression of pstB2 in ΔpstB2 reduced pstB1 expression. These results suggest that the E. faecalis pstB2-encoded ATPase is required for Pi import, while the pstB1-encoded ATPase has an accessory role in Pi import that can be duplicated by the presence of excess PstB2. IMPORTANCE Phosphate is critical for all microbial life. In many bacteria, inorganic phosphate (Pi) is imported by the high-affinity, low-velocity Pst-PhoU system. The pstB gene encodes the ATPase that powers Pi import. The pst-phoU operon in many Firmicutes, including the human commensal and opportunistic pathogen Enterococcus faecalis, contains adjacent pstB genes, pstB1 and pstB2. No studies on the relative biological contributions of tandem pstB paralogs in any microbe have been published. This genetic study indicates that E. faecalis pstB1 and pstB2 do not have equivalent functions. The pstB2 gene encodes an ATPase that is required for Pi import, while the ATPase encoded by pstB1 has an accessory role in Pi import that can be duplicated by the presence of excess PstB2.
Collapse
Affiliation(s)
- Christopher M. Healy
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Evelyn A. Pham
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Keane J. Dye
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Candace N. Rouchon
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Biko McMillan
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Kristi L. Frank
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
2
|
Bhardwaj RG, Khalaf ME, Karched M. Secretome analysis and virulence assessment in Abiotrophia defectiva. J Oral Microbiol 2024; 16:2307067. [PMID: 38352067 PMCID: PMC10863525 DOI: 10.1080/20002297.2024.2307067] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024] Open
Abstract
Background Abiotrophia defectiva, although infrequently occurring, is a notable cause of culture-negative infective endocarditis with limited research on its virulence. Associated with oral infections such as dental caries, exploring its secretome may provide insights into virulence mechanisms. Our study aimed to analyze and characterize the secretome of A. defectiva strain CCUG 27639. Methods Secretome of A. defectiva was prepared from broth cultures and subjected to mass spectrometry and proteomics for protein identification. Inflammatory potential of the secretome was assessed by ELISA. Results Eighty-four proteins were identified, with diverse subcellular localizations predicted by PSORTb. Notably, 20 were cytoplasmic, 12 cytoplasmic membrane, 5 extracellular, and 9 cell wall-anchored proteins. Bioinformatics tools revealed 54 proteins secreted via the 'Sec' pathway and 8 via a non-classical pathway. Moonlighting functions were found in 23 proteins, with over 20 exhibiting potential virulence properties, including peroxiredoxin and oligopeptide ABC transporter substrate-binding protein. Gene Ontology and KEGG analyses categorized protein sequences in various pathways. STRING analysis revealed functional protein association networks. Cytokine profiling demonstrated significant proinflammatory cytokine release (IL-8, IL-1β, and CCL5) from human PBMCs. Conclusions Our study provides a comprehensive understanding of A. defectiva's secretome, laying the foundation for insights into its pathogenicity.
Collapse
Affiliation(s)
- Radhika G Bhardwaj
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences College of Dentistry, Kuwait University, Safat, Kuwait
| | - Mai E Khalaf
- Department of General Dental Practice, College of Dentistry, Kuwait University, Safat, Kuwait
| | - Maribasappa Karched
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences College of Dentistry, Kuwait University, Safat, Kuwait
| |
Collapse
|
3
|
Wang F, Wang Z, Tang J. The interactions of Candida albicans with gut bacteria: a new strategy to prevent and treat invasive intestinal candidiasis. Gut Pathog 2023; 15:30. [PMID: 37370138 DOI: 10.1186/s13099-023-00559-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The gut microbiota plays an important role in human health, as it can affect host immunity and susceptibility to infectious diseases. Invasive intestinal candidiasis is strongly associated with gut microbiota homeostasis. However, the nature of the interaction between Candida albicans and gut bacteria remains unclear. OBJECTIVE This review aimed to determine the nature of interaction and the effects of gut bacteria on C. albicans so as to comprehend an approach to reducing intestinal invasive infection by C. albicans. METHODS This review examined 11 common gut bacteria's interactions with C. albicans, including Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Enterococcus faecalis, Staphylococcus aureus, Salmonella spp., Helicobacter pylori, Lactobacillus spp., Bacteroides spp., Clostridium difficile, and Streptococcus spp. RESULTS Most of the studied bacteria demonstrated both synergistic and antagonistic effects with C. albicans, and just a few bacteria such as P. aeruginosa, Salmonella spp., and Lactobacillus spp. demonstrated only antagonism against C. albicans. CONCLUSIONS Based on the nature of interactions reported so far by the literature between gut bacteria and C. albicans, it is expected to provide new ideas for the prevention and treatment of invasive intestinal candidiasis.
Collapse
Affiliation(s)
- Fei Wang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200240, China
| | - Zetian Wang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200240, China.
| | - Jianguo Tang
- Department of Trauma-Emergency & Critical Care Medicine, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200240, China.
| |
Collapse
|
4
|
Liu Y, Tang Y, Ren S, Chen L. Antibacterial Components and Modes of the Methanol-Phase Extract from Commelina communis Linn. PLANTS (BASEL, SWITZERLAND) 2023; 12:890. [PMID: 36840240 PMCID: PMC9966474 DOI: 10.3390/plants12040890] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Infectious diseases caused by pathogenic bacteria severely threaten human health. Traditional Chinese herbs are potential sources of new or alternative medicine. In this study, we analyzed for the first time antibacterial substances in the methanol-phase extract from a traditional Chinese herb-Commelina communis Linn-which showed an inhibition rate of 58.33% against 24 species of common pathogenic bacteria. The extract was further purified using preparative high-performance liquid chromatography (Prep-HPLC), which generated four single fragments (Fragments 1 to 4). The results revealed that Fragment 1 significantly increased bacterial cell surface hydrophobicity and membrane permeability and decreased membrane fluidity, showing disruptive effects on cell integrity of Gram-positive and Gram-negative bacteria, such as Bacillus cereus, Enterococcus faecalis, Staphylococcus aureus, and Salmonella enterica subsp., compared to the control groups (p < 0.05). In sum, 65 compounds with known functions in Fragment 1 were identified using liquid chromatography and mass spectrometry (LC-MS), of which quercetin-3-o-glucuronide was predominant (19.35%). Comparative transcriptomic analysis revealed multiple altered metabolic pathways mediated by Fragment 1, such as inhibited ABC transporters, ribosome, citrate cycle and oxidative phosphorylation, and upregulated nitrogen metabolism and purine metabolism, thereby resulting in the repressed bacterial growth and even death (p < 0.05). Overall, the results of this study demonstrate that Fragment 1 from C. communis Linn is a promising candidate against common pathogenic bacteria.
Collapse
Affiliation(s)
- Yue Liu
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Shanghai 201306, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Yingping Tang
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Shanghai 201306, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Shunlin Ren
- Department of Internal Medicine, Virginia Commonwealth University/McGuire VA Medical Centre, Richmond, VA 23298, USA
| | - Lanming Chen
- Key Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Shanghai 201306, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
5
|
Kalfopoulou E, Huebner J. Advances and Prospects in Vaccine Development against Enterococci. Cells 2020; 9:cells9112397. [PMID: 33147722 PMCID: PMC7692742 DOI: 10.3390/cells9112397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/26/2022] Open
Abstract
Enterococci are the second most common Gram-positive pathogen responsible for nosocomial infections. Due to the limited number of new antibiotics that reach the medical practice and the resistance of enterococci to the current antibiotic options, passive and active immunotherapies have emerged as a potential prevention and/or treatment strategy against this opportunistic pathogen. In this review, we explore the pathogenicity of these bacteria and their interaction with the host immune response. We provide an overview of the capsular polysaccharides and surface-associated proteins that have been described as potential antigens in anti-enterococcal vaccine formulations. In addition, we describe the current status in vaccine development against enterococci and address the importance and the current advances toward the development of well-defined vaccines with broad coverage against enterococci.
Collapse
Affiliation(s)
- Ermioni Kalfopoulou
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, 81675 Munich, Germany;
| | - Johannes Huebner
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, 80337 Munich, Germany
- Correspondence: ; Tel.: +49-89-44005-7970
| |
Collapse
|
6
|
Scheuplein NJ, Bzdyl NM, Kibble EA, Lohr T, Holzgrabe U, Sarkar-Tyson M. Targeting Protein Folding: A Novel Approach for the Treatment of Pathogenic Bacteria. J Med Chem 2020; 63:13355-13388. [PMID: 32786507 DOI: 10.1021/acs.jmedchem.0c00911] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Infectious diseases are a major cause of morbidity and mortality worldwide, exacerbated by increasing antibiotic resistance in many bacterial species. The development of drugs with new modes of action is essential. A leading strategy is antivirulence, with the aim to target bacterial proteins that are important in disease causation and progression but do not affect growth, resulting in reduced selective pressure for resistance. Immunophilins, a superfamily of peptidyl-prolyl cis-trans isomerase (PPIase) enzymes have been shown to be important for virulence in a broad-spectrum of pathogenic bacteria. This Perspective will provide an overview of the recent advances made in understanding the role of each immunophilin family, cyclophilins, FK506 binding proteins (FKBPs), and parvulins in bacteria. Inhibitor design and medicinal chemistry strategies for development of novel drugs against bacterial FKBPs will be discussed. Furthermore, drugs against human cyclophilins and parvulins will be reviewed in their current indication as antiviral and anticancer therapies.
Collapse
Affiliation(s)
- Nicolas J Scheuplein
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Nicole M Bzdyl
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, 6009 Perth, Australia
| | - Emily A Kibble
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, 6009 Perth, Australia.,School of Veterinary and Life Sciences, Murdoch University, 6150 Murdoch, Australia
| | - Theresa Lohr
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Mitali Sarkar-Tyson
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, 6009 Perth, Australia
| |
Collapse
|
7
|
Dautin N, Argentini M, Mohiman N, Labarre C, Cornu D, Sago L, Chami M, Dietrich C, de Sousa d'Auria C, Houssin C, Masi M, Salmeron C, Bayan N. Role of the unique, non-essential phosphatidylglycerol::prolipoprotein diacylglyceryl transferase (Lgt) in Corynebacterium glutamicum. MICROBIOLOGY-SGM 2020; 166:759-776. [PMID: 32490790 DOI: 10.1099/mic.0.000937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bacterial lipoproteins are secreted proteins that are post-translationally lipidated. Following synthesis, preprolipoproteins are transported through the cytoplasmic membrane via the Sec or Tat translocon. As they exit the transport machinery, they are recognized by a phosphatidylglycerol::prolipoprotein diacylglyceryl transferase (Lgt), which converts them to prolipoproteins by adding a diacylglyceryl group to the sulfhydryl side chain of the invariant Cys+1 residue. Lipoprotein signal peptidase (LspA or signal peptidase II) subsequently cleaves the signal peptide, liberating the α-amino group of Cys+1, which can eventually be further modified. Here, we identified the lgt and lspA genes from Corynebacterium glutamicum and found that they are unique but not essential. We found that Lgt is necessary for the acylation and membrane anchoring of two model lipoproteins expressed in this species: MusE, a C. glutamicum maltose-binding lipoprotein, and LppX, a Mycobacterium tuberculosis lipoprotein. However, Lgt is not required for these proteins' signal peptide cleavage, or for LppX glycosylation. Taken together, these data show that in C. glutamicum the association of some lipoproteins with membranes through the covalent attachment of a lipid moiety is not essential for further post-translational modification.
Collapse
Affiliation(s)
- Nathalie Dautin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France.,Present address: Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, CNRS, Institut de Biologie Physico-Chimique, F-75005 Paris, France
| | - Manuela Argentini
- Present address: Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France.,Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Niloofar Mohiman
- Present address: Curakliniken, Erikslustvägen 22, 217 73 Malmö, Sweden.,Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Cécile Labarre
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - David Cornu
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Laila Sago
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Mohamed Chami
- CBioEM lab, Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Christiane Dietrich
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Célia de Sousa d'Auria
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Christine Houssin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Muriel Masi
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Christophe Salmeron
- Present address: Observatoire Océanologique de Banyuls Sur Mer, FR 3724-Laboratoire Arago - Sorbonne Université / CNRS, France.,Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| | - Nicolas Bayan
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, 91198, Gif-sur-Yvette cedex, France
| |
Collapse
|
8
|
Sterling AJ, Snelling WJ, Naughton PJ, Ternan NG, Dooley JSG. Competent but complex communication: The phenomena of pheromone-responsive plasmids. PLoS Pathog 2020; 16:e1008310. [PMID: 32240270 PMCID: PMC7117660 DOI: 10.1371/journal.ppat.1008310] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Enterococci are robust gram-positive bacteria that are found in a variety of surroundings and that cause a significant number of healthcare-associated infections. The genus possesses a high-efficiency pheromone-responsive plasmid (PRP) transfer system for genetic exchange that allows antimicrobial-resistance determinants to spread within bacterial populations. The pCF10 plasmid system is the best characterised, and although other PRP systems are structurally similar, they lack exact functional homologues of pCF10-encoded genes. In this review, we provide an overview of the enterococcal PRP systems, incorporating functional details for the less-well-defined systems. We catalogue the virulence-associated elements of the PRPs that have been identified to date, and we argue that this reinforces the requirement for elucidation of the less studied systems.
Collapse
Affiliation(s)
- Amy J. Sterling
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, Londonderry, Northern Ireland
- * E-mail:
| | - William J. Snelling
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, Londonderry, Northern Ireland
| | - Patrick J. Naughton
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, Londonderry, Northern Ireland
| | - Nigel G. Ternan
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, Londonderry, Northern Ireland
| | - James S. G. Dooley
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, Londonderry, Northern Ireland
| |
Collapse
|
9
|
Suryaletha K, Narendrakumar L, John J, Radhakrishnan MP, George S, Thomas S. Decoding the proteomic changes involved in the biofilm formation of Enterococcus faecalis SK460 to elucidate potential biofilm determinants. BMC Microbiol 2019; 19:146. [PMID: 31253082 PMCID: PMC6599329 DOI: 10.1186/s12866-019-1527-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/20/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Enterococcus faecalis is a major clinically relevant nosocomial bacterial pathogen frequently isolated from polymicrobial infections. The biofilm forming ability of E. faecalis attributes a key role in its virulence and drug resistance. Biofilm cells are phenotypically and metabolically different from their planktonic counterparts and many aspects involved in E. faecalis biofilm formation are yet to be elucidated. The strain E. faecalis SK460 used in the present study is esp (Enterococcal surface protein) and fsr (two-component signal transduction system) negative non-gelatinase producing strong biofilm former isolated from a chronic diabetic foot ulcer patient. We executed a label-free quantitative proteomic approach to elucidate the differential protein expression pattern at planktonic and biofilm stages of SK460 to come up with potential determinants associated with Enterococcal biofilm formation. RESULTS The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of proteomic data revealed that biofilm cells expressed higher levels of proteins which are associated with glycolysis, amino acid biosynthesis, biosynthesis of secondary metabolites, microbial metabolism in diverse environments and stress response factors. Besides these basic survival pathways, LuxS-mediated quorum sensing, arginine metabolism, rhamnose biosynthesis, pheromone and adhesion associated proteins were found to be upregulated during the biofilm transit from planktonic stages. The selected subsets were validated by quantitative real-time PCR. In silico functional interaction analysis revealed that the genes involved in upregulated pathways pose a close molecular interaction thereby coordinating the regulatory network to thrive as a biofilm community. CONCLUSIONS The present study describes the first report of the quantitative proteome analysis of an esp and fsr negative non gelatinase producing E. faecalis. Proteome analysis evidenced enhanced expression of glycolytic pathways, stress response factors, LuxS quorum signaling system, rhamnopolysaccharide synthesis and pheromone associated proteins in biofilm phenotype. We also pointed out the relevance of LuxS quorum sensing and pheromone associated proteins in the biofilm development of E. faecalis which lacks the Fsr quorum signaling system. These validated biofilm determinants can act as potential inhibiting targets in Enterococcal infections.
Collapse
Affiliation(s)
- Karthika Suryaletha
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India
| | - Lekshmi Narendrakumar
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India
| | - Joby John
- Department of Surgery, Government Medical College Hospital, Trivandrum, Kerala, 695011, India
| | - Megha Periyappilly Radhakrishnan
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India
| | - Sanil George
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, 695014, India
| | - Sabu Thomas
- Cholera and Biofilm Research Laboratory, Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, (National Institute under the Department of Biotechnology, Government of India), Trivandrum, Kerala, 695014, India.
| |
Collapse
|
10
|
Cirrincione S, Neumann B, Zühlke D, Riedel K, Pessione E. Detailed Soluble Proteome Analyses of a Dairy-Isolated Enterococcus faecalis: A Possible Approach to Assess Food Safety and Potential Probiotic Value. Front Nutr 2019; 6:71. [PMID: 31157229 PMCID: PMC6533484 DOI: 10.3389/fnut.2019.00071] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
Abstract
Enterococci are common inhabitants of the gastrointestinal tracts of humans and animals and thanks to their capability to tolerate different environmental conditions and their high rates of gene transfer, they are able to colonize various ecological niches, as food matrices. Enterococcus faecalis bacteria are defined as controversial microorganisms. From one side they are used as food starters, bio-control agents and probiotics to improve human or animal health. From the other side, in the last two decades enterococci have emerged as important nosocomial pathogens, because bearing high-level of resistance to antibiotics and several putative virulence factors. In this study, the soluble proteome quantitation data (LC-MS/MS) of the food-isolated strain E. faecalis D27 (dairy-isolate) was compared with the soluble proteome quantitation data of the pathogenic E. faecalis UW3114 (urinary tract infection isolate) and with the one of the health promoting strain E. faecalis Symbioflor1, respectively. The comparison of cytosolic protein expression profiles highlighted statistically significant changes in the abundance of proteins mainly involved in specific metabolic pathways, nutrient transport, stress response, and cell wall modulation. Moreover, especially in the dairy isolate and the clinical isolate, several proteins with potential pathogenic implications were found, such as serine proteases, von Willebrand factor, serine hydrolase with beta lactamase activity, efflux transporter, and proteins involved in horizontal gene transfer. The analysis of the extracellular proteome provided interesting results concerning proteins involved in bacterial communication, such as pheromones and conjugative elements and also proteins able to interact with human components. The phenotypic characterization evaluating (i) biofilm formation (ii) hemolytic activity on blood agar plates (iii) protease activity (iv) gelatinase (v) antibiotic resistance pattern, enabled us to elucidate the risks associated with the poor characterized foodborne E. faecalis D27.
Collapse
Affiliation(s)
- Simona Cirrincione
- Department of Life Sciences and Systems Biology, Univerity of Torino, Turin, Italy
| | - Bernd Neumann
- Department for Microbial Physiology and Molecular Biology, University of Greifswald, Greifswald, Germany
| | - Daniela Zühlke
- Department for Microbial Physiology and Molecular Biology, University of Greifswald, Greifswald, Germany
| | - Katharina Riedel
- Department for Microbial Physiology and Molecular Biology, University of Greifswald, Greifswald, Germany
| | - Enrica Pessione
- Department of Life Sciences and Systems Biology, Univerity of Torino, Turin, Italy
| |
Collapse
|
11
|
Cathro P, McCarthy P, Hoffmann P, Zilm P. Isolation and identification of Enterococcus faecalis membrane proteins using membrane shaving, 1D SDS/PAGE, and mass spectrometry. FEBS Open Bio 2016; 6:586-93. [PMID: 27419061 PMCID: PMC4887974 DOI: 10.1002/2211-5463.12075] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/22/2016] [Accepted: 04/11/2016] [Indexed: 11/24/2022] Open
Abstract
Enterococcus faecalis is a significant nosocomial pathogen, which is able to survive in diverse environments and resist killing with antimicrobial therapies. The expression of cell membrane proteins play an important role in how bacteria respond to environmental stress. As such, the capacity to identify and study membrane protein expression is critical to our understanding of how specific proteins influence bacterial survival. Here, we describe a combined approach to identify membrane proteins of E. faecalis ATCC V583 using membranes fractionated by either 1D SDS/PAGE or membrane shaving, coupled with LC‐ESI mass spectrometry. We identified 222 membrane‐associated proteins, which represent approximately 24% of the predicted membrane‐associated proteome: 170 were isolated using 1D SDS/PAGE and 68 with membrane shaving, with 36 proteins being common to both the techniques. Of the proteins identified by membrane shaving, 97% were membrane‐associated with the majority being integral membrane proteins (89%). Most of the proteins identified with known physiology are involved with transportation across the membrane. The combined 1D SDS/PAGE and membrane shaving approach has produced the greatest number of membrane proteins identified from E. faecalis to date. These protocols will aid future researchers investigating changes in the membrane proteome of E. faecalis by improving our understanding of how E. faecalis adapts and responds to its environment.
Collapse
Affiliation(s)
- Peter Cathro
- Oral Microbiology Laboratory School of Dentistry The University of Adelaide South Australia Australia
| | - Peter McCarthy
- Neurovascular Research Laboratory Centre for Cancer Biology University of South Australia Adelaide South Australia Australia
| | - Peter Hoffmann
- Adelaide Proteomics Centre The University of Adelaide South Australia Australia
| | - Peter Zilm
- Oral Microbiology Laboratory School of Dentistry The University of Adelaide South Australia Australia
| |
Collapse
|
12
|
Sabatino R, Di Cesare A, Pasquaroli S, Vignaroli C, Citterio B, Amiri M, Rossi L, Magnani M, Mauro A, Biavasco F. Adherence and intracellular survival within human macrophages of Enterococcus faecalis isolates from coastal marine sediment. Microbes Infect 2015; 17:660-4. [DOI: 10.1016/j.micinf.2015.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 11/25/2022]
|
13
|
Romero-Saavedra F, Laverde D, Budin-Verneuil A, Muller C, Bernay B, Benachour A, Hartke A, Huebner J. Characterization of Two Metal Binding Lipoproteins as Vaccine Candidates for Enterococcal Infections. PLoS One 2015; 10:e0136625. [PMID: 26322633 PMCID: PMC4556446 DOI: 10.1371/journal.pone.0136625] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 08/05/2015] [Indexed: 12/19/2022] Open
Abstract
Background Enterococcus faecium and faecalis are Gram-positive opportunistic pathogens that have become leading causes of nosocomial infections over the last decades. Especially multidrug resistant enterococci have become a challenging clinical problem worldwide. Therefore, new treatment options are needed and the identification of alternative targets for vaccine development has emerged as a feasible alternative to fight the infections caused by these pathogens. Results We extrapolate the transcriptomic data from a mice peritonitis infection model in E. faecalis to identify putative up-regulated surface proteins under infection conditions in E. faecium. After the bionformatic analyses two metal binding lipoproteins were identified to have a high homology (>72%) between the two species, the manganese ABC transporter substrate-binding lipoprotein (PsaAfm,) and the zinc ABC transporter substrate-binding lipoprotein (AdcAfm). These candidate lipoproteins were overexpressed in Escherichia coli and purified. The recombinant proteins were used to produce rabbit polyclonal antibodies that were able to induce specific opsonic antibodies that mediated killing of the homologous strain E. faecium E155 as well as clinical strains E. faecium E1162, Enterococcus faecalis 12030, type 2 and type 5. Mice were passively immunized with the antibodies raised against recombinant lipoproteins, showing significant reduction of colony counts in mice livers after the bacterial challenge and demonstrating the efficacy of these metal binding lipoproteins as promising vaccine candidates to treat infections caused by these enterococcal pathogens. Conclusion Overall, our results demonstrate that these two metal binding lipoproteins elicited specific, opsonic and protective antibodies, with an extensive cross-reactivity and serotype-independent coverage among these two important nocosomial pathogens. Pointing these two protein antigens as promising immunogens, that can be used as single components or as carrier proteins together with polysaccharide antigens in vaccine development against enterococcal infections.
Collapse
Affiliation(s)
- Felipe Romero-Saavedra
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Freiburg, Germany
- EA4655 U2RM Stress/Virulence, University of Caen Lower-Normandy, Caen, France
| | - Diana Laverde
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Freiburg, Germany
- EA4655 U2RM Stress/Virulence, University of Caen Lower-Normandy, Caen, France
| | | | - Cécile Muller
- EA4655 U2RM Stress/Virulence, University of Caen Lower-Normandy, Caen, France
| | - Benoit Bernay
- Proteogen platform SFR ICORE 4206, University of Caen Lower-Normandy, Caen, France
| | - Abdellah Benachour
- EA4655 U2RM Stress/Virulence, University of Caen Lower-Normandy, Caen, France
| | - Axel Hartke
- EA4655 U2RM Stress/Virulence, University of Caen Lower-Normandy, Caen, France
| | - Johannes Huebner
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Freiburg, Germany
- Division of Pediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
- * E-mail:
| |
Collapse
|
14
|
Bitoun JP, Wen ZT. Transcription factor Rex in regulation of pathophysiology in oral pathogens. Mol Oral Microbiol 2015; 31:115-24. [PMID: 26172563 DOI: 10.1111/omi.12114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2015] [Indexed: 11/29/2022]
Abstract
The NAD(+) and NADH-sensing transcriptional regulator Rex is widely conserved across gram-positive bacteria. Rex monitors cellular redox poise and controls the expression of genes/operons involved in diverse pathways including alternative fermentation, oxidative stress responses, and biofilm formation. The oral cavity undergoes frequent and drastic fluctuations in nutrient availability, pH, temperature, oxygen tension, saliva, and shear forces. The oral streptococci are major colonizers of oral mucosa and tooth surfaces and include commensals as well as opportunistic pathogens, including the primary etiological agent of dental caries, Streptococcus mutans. Current understanding of the Rex regulon in oral bacteria is mostly based on studies in S. mutans and endodontic pathogen Enterococcus faecalis. Indeed, other oral bacteria encode homologs of the Rex protein and much is to be gleaned from more in-depth studies. Our current understanding has Rex positioned at the interface of oxygen and energy metabolism. In biofilms, heterogeneous oxygen tension influences the ratio of intracellular NADH and NAD(+) , which is finely tuned through glycolysis and fermentation. In S. mutans, Rex regulates the expression of glycolytic enzyme NAD(+) -dependent glyceraldehyde 3-phosphate dehydrogenase, and NADH-dependent fermentation enzymes/complexes lactate dehydrogenase, pyruvate dehydrogenase, alcohol-acetaldehyde dehydrogenase, and fumarate reductase. In addition, Rex controls the expression of NADH oxidase, a major enzyme used to eliminate oxidative stress and regenerate NAD(+) . Here, we summarize recent studies carried out on the Rex regulators in S. mutans and E. faecalis. This research has important implications for understanding how Rex monitors redox balance and optimizes fermentation pathways for survival and subsequent pathogenicity.
Collapse
Affiliation(s)
- J P Bitoun
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Z T Wen
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, USA.,Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.,Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
15
|
Theilacker C, Diederich AK, Otto A, Sava IG, Wobser D, Bao Y, Hese K, Broszat M, Henneke P, Becher D, Huebner J. Enterococcus faecalis Glycolipids Modulate Lipoprotein-Content of the Bacterial Cell Membrane and Host Immune Response. PLoS One 2015; 10:e0132949. [PMID: 26172831 PMCID: PMC4501811 DOI: 10.1371/journal.pone.0132949] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 06/21/2015] [Indexed: 12/25/2022] Open
Abstract
In this study, we investigated the impact of the cell membrane composition of E. faecalis on its recognition by the host immune system. To this end, we employed an E. faecalis deletion mutant (ΔbgsA) that does not synthesize the major cell membrane glycolipid diglycosyl-diacylglycerol (DGlcDAG). Proteomic analysis revealed that 13 of a total of 21 upregulated surface-associated proteins of E. faecalis ΔbgsA were lipoproteins. This led to a total lipoprotein content in the cell membrane of 35.8% in ΔbgsA compared to only 9.4% in wild-type bacteria. Increased lipoprotein content strongly affected the recognition of ΔbgsA by mouse macrophages in vitro with an increased stimulation of TNF-α production by heat-fixed bacteria and secreted antigens. Inactivation of the prolipoprotein diacylglycerol transferase (lgt) in ΔbgsA abrogated TNF-α induction by a ΔbgsA_lgt double mutant indicating that lipoproteins mediate increased activation of mouse macrophages by ΔbgsA. Heat-fixed ΔbgsA bacteria, culture supernatant, or cell membrane lipid extract activated transfected HEK cells in a TLR2-dependent fashion; the same was not true of wild-type bacteria. In mice infected intraperitoneally with a sublethal dose of E. faecalis we observed a 70% greater mortality in mice infected with ΔbgsA compared with wild-type-infected mice. Increased mortality due to ΔbgsA infection was associated with elevated plasma levels of the inflammatory cytokines TNF-α, IL-6 and MIP-2. In summary, our results provide evidence that an E. faecalis mutant lacking its major bilayer forming glycolipid DGlcDAG upregulates lipoprotein expression leading to increased activation of the host innate immune system and virulence in vivo.
Collapse
Affiliation(s)
- Christian Theilacker
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- * E-mail:
| | - Ann-Kristin Diederich
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- Division of Pediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
- Department of Microbiology, Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Andreas Otto
- Institute for Microbiology, Department of Microbial Physiology, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Irina G. Sava
- Research Center for Nutrition and Food Science, Technical University Munich, Freising, Germany
| | - Dominique Wobser
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Yinyin Bao
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Katrin Hese
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
| | - Melanie Broszat
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Center for Chronic Immunodeficiency (CCI), University Medical Center Freiburg, Freiburg, Germany
- Center for Paediatrics and Adolescent Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Dörte Becher
- Institute for Microbiology, Department of Microbial Physiology, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Johannes Huebner
- Center for Infectious Disease and Travel Medicine, University Medical Center Freiburg, Freiburg, Germany
- Division of Pediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
16
|
Ocvirk S, Sava IG, Lengfelder I, Lagkouvardos I, Steck N, Roh JH, Tchaptchet S, Bao Y, Hansen JJ, Huebner J, Carroll IM, Murray BE, Sartor RB, Haller D. Surface-Associated Lipoproteins Link Enterococcus faecalis Virulence to Colitogenic Activity in IL-10-Deficient Mice Independent of Their Expression Levels. PLoS Pathog 2015; 11:e1004911. [PMID: 26067254 PMCID: PMC4466351 DOI: 10.1371/journal.ppat.1004911] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/24/2015] [Indexed: 12/22/2022] Open
Abstract
The commensal Enterococcus faecalis is among the most common causes of nosocomial infections. Recent findings regarding increased abundance of enterococci in the intestinal microbiota of patients with inflammatory bowel diseases and induction of colitis in IL-10-deficient (IL-10-/-) mice put a new perspective on the contribution of E. faecalis to chronic intestinal inflammation. Based on the expression of virulence-related genes in the inflammatory milieu of IL-10-/- mice using RNA-sequencing analysis, we characterized the colitogenic role of two bacterial structures that substantially impact on E. faecalis virulence by different mechanisms: the enterococcal polysaccharide antigen and cell surface-associated lipoproteins. Germ-free wild type and IL-10-/- mice were monoassociated with E. faecalis wild type OG1RF or the respective isogenic mutants for 16 weeks. Intestinal tissue and mesenteric lymph nodes (MLN) were collected to characterize tissue pathology, loss of intestinal barrier function, bacterial adhesion to intestinal epithelium and immune cell activation. Bone marrow-derived dendritic cells (BMDC) were stimulated with bacterial lysates and E. faecalis virulence was additionally investigated in three invertebrate models. Colitogenic activity of wild type E. faecalis (OG1RF score: 7.2±1.2) in monoassociated IL-10-/- mice was partially impaired in E. faecalis lacking enterococcal polysaccharide antigen (ΔepaB score: 4.7±2.3; p<0.05) and was almost completely abrogated in E. faecalis deficient for lipoproteins (Δlgt score: 2.3±2.3; p<0.0001). Consistently both E. faecalis mutants showed significantly impaired virulence in Galleria mellonella and Caenorhabditis elegans. Loss of E-cadherin in the epithelium was shown for all bacterial strains in inflamed IL-10-/- but not wild type mice. Inactivation of epaB in E. faecalis reduced microcolony and biofilm formation in vitro, altered bacterial adhesion to intestinal epithelium of germ-free Manduca sexta larvae and impaired penetration into the colonic mucus layer of IL-10-/- mice. Lipoprotein-deficient E. faecalis exhibited an impaired TLR2-mediated activation of BMDCs in vitro despite their ability to fully reactivate MLN cells as well as MLN-derived colitogenic T cells ex vivo. E. faecalis virulence factors accounting for bacterial adhesion to mucosal surfaces as well as intestinal barrier disruption partially contribute to colitogenic activity of E. faecalis. Beyond their well-known role in infections, cell surface-associated lipoproteins are essential structures for colitogenic activity of E. faecalis by mediating innate immune cell activation. Enterococcus faecalis is a commensal of the human intestinal core microbiota harboring several putative virulence factors, which highlight its role as opportunistic pathogen. This dualistic character is supported by recent evidence linking Enterococcus spp. to the pathogenesis of inflammatory bowel diseases (IBD). Although several studies suggest a crucial role for opportunistic pathogens in IBD pathogenesis targeting genetically susceptible individuals, the dynamic relationship between disease-relevant host compartments and specific bacterial structures able to trigger intestinal inflammation remain unclear. Here, we report that cell surface-associated lipoproteins and the enterococcal polysaccharide antigen, which are relevant for E. faecalis virulence in invertebrate infection models, but whose expression is minimally affected by the intestinal inflammatory milieu, exhibit colitogenic activity in a mouse model susceptible for chronic colitis. Bacterial lipoproteins trigger innate immune cell activation and are a critical prerequisite for E. faecalis-induced colitis. The enterococcal polysaccharide antigen mediates bacterial mucus penetration and adhesion to mucosal surfaces, promotes the formation of biofilm and contributes to E. faecalis colitogenic activity. Using E. faecalis as a model organism, we demonstrate that colitogenic activity of opportunistic pathogens can be assigned to specific bacterial structures, a finding that may help to identify the most essential steps in IBD-related microbe-host interactions.
Collapse
Affiliation(s)
- Soeren Ocvirk
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Irina G. Sava
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Isabella Lengfelder
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Ilias Lagkouvardos
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Natalie Steck
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
| | - Jung H. Roh
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical School, Houston, Texas, United States of America
| | - Sandrine Tchaptchet
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Yinyin Bao
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Jonathan J. Hansen
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Johannes Huebner
- Division of Infectious Diseases, Department of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Ian M. Carroll
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Barbara E. Murray
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical School, Houston, Texas, United States of America
| | - R. Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Dirk Haller
- Technische Universität München, Chair of Nutrition and Immunology, ZIEL–Research Center for Nutrition and Food Sciences, Freising-Weihenstephan, Germany
- * E-mail:
| |
Collapse
|
17
|
Arntzen MØ, Karlskås IL, Skaugen M, Eijsink VGH, Mathiesen G. Proteomic Investigation of the Response of Enterococcus faecalis V583 when Cultivated in Urine. PLoS One 2015; 10:e0126694. [PMID: 25915650 PMCID: PMC4411035 DOI: 10.1371/journal.pone.0126694] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 04/07/2015] [Indexed: 11/19/2022] Open
Abstract
Enterococcus faecalis is a robust bacterium, which is able to survive in and adapt to hostile environments such as the urinary tract and bladder. In this label-free quantitative proteomic study based on MaxQuant LFQ algorithms, we identified 127 proteins present in the secretome of the clinical vancomycin-resistant isolate E. faecalis V583 and we compared proteins secreted in the initial phase of cultivation in urine with the secretome during cultivation in standard laboratory medium, 2xYT. Of the 54 identified proteins predicted to be secreted, six were exclusively found after cultivation in urine including the virulence factor EfaA ("endocarditis specific antigen") and its homologue EF0577 ("adhesion lipoprotein"). These two proteins are both involved in manganese transport, known to be an important determinant of colonization and infection, and may additionally function as adhesins. Other detected urine-specific proteins are involved in peptide transport (EF0063 and EF3106) and protease inhibition (EF3054). In addition, we found an uncharacterized protein (EF0764), which had not previously been linked to the adaptation of V583 to a urine environment, and which is unique to E. faecalis. Proteins found in both environments included a histone-like protein, EF1550, that was up-regulated during cultivation in urine and that has a homologue in streptococci (HlpA) known to be involved in bacterial adhesion to host cells. Up-regulated secreted proteins included autolysins. These results from secretome analyses are largely compatible with previously published data from transcriptomics studies. All in all, the present data indicate that transport, in particular metal transport, adhesion, cell wall remodelling and the unknown function carried out by the unique EF0764 are important for enterococcal adaptation to the urine environment. These results provide a basis for a more targeted exploration of novel proteins involved in the adaptability and pathogenicity of E. faecalis.
Collapse
Affiliation(s)
- Magnus Øverlie Arntzen
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, N-1432 Ås, Norway
- * E-mail:
| | - Ingrid Lea Karlskås
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, N-1432 Ås, Norway
| | - Morten Skaugen
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, N-1432 Ås, Norway
| | - Vincent G. H. Eijsink
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, N-1432 Ås, Norway
| | - Geir Mathiesen
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, N-1432 Ås, Norway
| |
Collapse
|
18
|
A genomic virulence reference map of Enterococcus faecalis reveals an important contribution of phage03-like elements in nosocomial genetic lineages to pathogenicity in a Caenorhabditis elegans infection model. Infect Immun 2015; 83:2156-67. [PMID: 25776747 DOI: 10.1128/iai.02801-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/07/2015] [Indexed: 02/07/2023] Open
Abstract
In the present study, the commensal and pathogenic host-microbe interaction of Enterococcus faecalis was explored using a Caenorhabditis elegans model system. The virulence of 28 E. faecalis isolates representing 24 multilocus sequence types (MLSTs), including human commensal and clinical isolates as well as isolates from animals and of insect origin, was investigated using C. elegans strain glp-4 (bn2ts); sek-1 (km4). This revealed that 6 E. faecalis isolates behaved in a commensal manner with no nematocidal effect, while the remaining strains showed a time to 50% lethality ranging from 47 to 120 h. Principal component analysis showed that the difference in nematocidal activity explained 94% of the variance in the data. Assessment of known virulence traits revealed that gelatinase and cytolysin production accounted for 40.8% and 36.5% of the observed pathogenicity, respectively. However, coproduction of gelatinase and cytolysin did not increase virulence additively, accounting for 50.6% of the pathogenicity and therefore indicating a significant (26.7%) saturation effect. We employed a comparative genomic analysis approach using the 28 isolates comprising a collection of 82,356 annotated coding sequences (CDS) to identify 2,325 patterns of presence or absence among the investigated strains. Univariate statistical analysis of variance (ANOVA) established that individual patterns positively correlated (n = 61) with virulence. The patterns were investigated to identify potential new virulence traits, among which we found five patterns consisting of the phage03-like gene clusters. Strains harboring phage03 showed, on average, 17% higher killing of C. elegans (P = 4.4e(-6)). The phage03 gene cluster was also present in gelatinase-and-cytolysin-negative strain E. faecalis JH2-2. Deletion of this phage element from the JH2-2 clinical strain rendered the mutant apathogenic in C. elegans, and a similar mutant of the nosocomial V583 isolate showed significantly attenuated virulence. Bioinformatics investigation indicated that, unlike other E. faecalis virulence traits, phage03-like elements were found at a higher frequency among nosocomial isolates. In conclusion, our report provides a valuable virulence map that explains enhancement in E. faecalis virulence and contributes to a deeper comprehension of the genetic mechanism leading to the transition from commensalism to a pathogenic lifestyle.
Collapse
|
19
|
Saxena S, Khan N, Dehinwal R, Kumar A, Sehgal D. Conserved surface accessible nucleoside ABC transporter component SP0845 is essential for pneumococcal virulence and confers protection in vivo. PLoS One 2015; 10:e0118154. [PMID: 25689507 PMCID: PMC4331430 DOI: 10.1371/journal.pone.0118154] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 01/05/2015] [Indexed: 11/30/2022] Open
Abstract
Streptococcus pneumoniae is a leading cause of bacterial pneumonia, sepsis and meningitis. Surface accessible proteins of S. pneumoniae are being explored for the development of a protein-based vaccine in order to overcome the limitations of existing polysaccharide-based pneumococcal vaccines. To identify a potential vaccine candidate, we resolved surface-associated proteins of S. pneumoniae TIGR4 strain using two-dimensional gel electrophoresis followed by immunoblotting with antisera generated against whole heat-killed TIGR4. Ten immunoreactive spots were identified by mass spectrometric analysis that included a putative lipoprotein SP0845. Analysis of the inferred amino acid sequence of sp0845 homologues from 36 pneumococcal strains indicated that SP0845 was highly conserved (>98% identity) and showed less than 11% identity with any human protein. Our bioinformatic and functional analyses demonstrated that SP0845 is the substrate-binding protein of an ATP-binding cassette (ABC) transporter that is involved in nucleoside uptake with cytidine, uridine, guanosine and inosine as the preferred substrates. Deletion of the gene encoding SP0845 renders pneumococci avirulent suggesting that it is essential for virulence. Immunoblot analysis suggested that SP0845 is expressed in in vitro grown pneumococci and during mice infection. Immunofluorescence microscopy and flow cytometry data indicated that SP0845 is surface exposed in encapsulated strains and accessible to antibodies. Subcutaneous immunization with recombinant SP0845 induced high titer antibodies in mice. Hyperimmune sera raised against SP0845 promoted killing of encapsulated pneumococcal strains in a blood bactericidal assay. Immunization with SP0845 protected mice from intraperitoneal challenge with heterologous pneumococcal serotypes. Based on its surface accessibility, role in virulence and ability to elicit protective immunity, we propose that SP0845 may be a potential candidate for a protein-based pneumococcal vaccine.
Collapse
Affiliation(s)
- Sneha Saxena
- Molecular Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Naeem Khan
- Molecular Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Ruchika Dehinwal
- Molecular Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Ajay Kumar
- Molecular Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Devinder Sehgal
- Molecular Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
- * E-mail:
| |
Collapse
|
20
|
Frank KL, Colomer-Winter C, Grindle SM, Lemos JA, Schlievert PM, Dunny GM. Transcriptome analysis of Enterococcus faecalis during mammalian infection shows cells undergo adaptation and exist in a stringent response state. PLoS One 2014; 9:e115839. [PMID: 25545155 PMCID: PMC4278851 DOI: 10.1371/journal.pone.0115839] [Citation(s) in RCA: 28] [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: 07/21/2014] [Accepted: 12/01/2014] [Indexed: 11/30/2022] Open
Abstract
As both a commensal and a major cause of healthcare-associated infections in humans, Enterococcus faecalis is a remarkably adaptable organism. We investigated how E. faecalis adapts in a mammalian host as a pathogen by characterizing changes in the transcriptome during infection in a rabbit model of subdermal abscess formation using transcriptional microarrays. The microarray experiments detected 222 and 291 differentially regulated genes in E. faecalis OG1RF at two and eight hours after subdermal chamber inoculation, respectively. The profile of significantly regulated genes at two hours post-inoculation included genes involved in stress response, metabolism, nutrient acquisition, and cell surface components, suggesting genome-wide adaptation to growth in an altered environment. At eight hours post-inoculation, 88% of the differentially expressed genes were down-regulated and matched a transcriptional profile consistent with a (p)ppGpp-mediated stringent response. Subsequent subdermal abscess infections with E. faecalis mutants lacking the (p)ppGpp synthetase/hydrolase RSH, the small synthetase RelQ, or both enzymes, suggest that intracellular (p)ppGpp levels, but not stringent response activation, influence persistence in the model. The ability of cells to synthesize (p)ppGpp was also found to be important for growth in human serum and whole blood. The data presented in this report provide the first genome-wide insights on E. faecalis in vivo gene expression and regulation measured by transcriptional profiling during infection in a mammalian host and show that (p)ppGpp levels affect viability of E. faecalis in multiple conditions relevant to mammalian infection. The subdermal abscess model can serve as a novel experimental system for studying the E. faecalis stringent response in the context of the mammalian immune system.
Collapse
Affiliation(s)
- Kristi L. Frank
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Cristina Colomer-Winter
- Center for Oral Biology and Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Suzanne M. Grindle
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - José A. Lemos
- Center for Oral Biology and Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Patrick M. Schlievert
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Gary M. Dunny
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| |
Collapse
|
21
|
Lacroix T, Loux V, Gendrault A, Hoebeke M, Gibrat JF. Insyght: navigating amongst abundant homologues, syntenies and gene functional annotations in bacteria, it's that symbol! Nucleic Acids Res 2014; 42:gku867. [PMID: 25249626 PMCID: PMC4245967 DOI: 10.1093/nar/gku867] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 08/28/2014] [Accepted: 09/10/2014] [Indexed: 11/14/2022] Open
Abstract
High-throughput techniques have considerably increased the potential of comparative genomics whilst simultaneously posing many new challenges. One of those challenges involves efficiently mining the large amount of data produced and exploring the landscape of both conserved and idiosyncratic genomic regions across multiple genomes. Domains of application of these analyses are diverse: identification of evolutionary events, inference of gene functions, detection of niche-specific genes or phylogenetic profiling. Insyght is a comparative genomic visualization tool that combines three complementary displays: (i) a table for thoroughly browsing amongst homologues, (ii) a comparator of orthologue functional annotations and (iii) a genomic organization view designed to improve the legibility of rearrangements and distinctive loci. The latter display combines symbolic and proportional graphical paradigms. Synchronized navigation across multiple species and interoperability between the views are core features of Insyght. A gene filter mechanism is provided that helps the user to build a biologically relevant gene set according to multiple criteria such as presence/absence of homologues and/or various annotations. We illustrate the use of Insyght with scenarios. Currently, only Bacteria and Archaea are supported. A public instance is available at http://genome.jouy.inra.fr/Insyght. The tool is freely downloadable for private data set analysis.
Collapse
Affiliation(s)
- Thomas Lacroix
- INRA, UR 1077 Mathématique Informatique et Génome, 78352 Jouy-en-Josas, France
| | - Valentin Loux
- INRA, UR 1077 Mathématique Informatique et Génome, 78352 Jouy-en-Josas, France
| | - Annie Gendrault
- INRA, UR 1077 Mathématique Informatique et Génome, 78352 Jouy-en-Josas, France
| | - Mark Hoebeke
- CNRS, UPMC, FR2424, ABiMS, Station Biologique, 29680 Roscoff, France
| | | |
Collapse
|
22
|
Wu C, Liang B, Gong Y, Zhang L, Zou Y, Ge J. Streptococcus acidominimus causing invasive disease in humans: a case series. J Med Case Rep 2014; 8:57. [PMID: 24529345 PMCID: PMC3927825 DOI: 10.1186/1752-1947-8-57] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 12/09/2013] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Streptococcus acidominimus is a member of the viridans group streptococci and is rarely pathogenic in humans, making it difficult to assess its epidemiologic and clinical significance. CASE PRESENTATION We report the cases of five Han Chinese patients with invasive diseases caused by S. acidominimus over a one-year time frame. Three of the patients developed continuous fever after surgery, consisting of a successful elective laparoscopic cholecystectomy (case 1), a laparoscopic esophageal resection and gastroesophageal anastomosis (case 2), and a liver transplant in a patient with liver cancer (case 3). For these three patients, cultures of the purulent drainage material grew S. acidominimus. Case 4 concerns a 52-year-old man who developed sepsis 48 hours after hospitalization for hepatitis, liver cirrhosis and hepatitis-related glomerulonephritis. Case 5 concerns a 55-year-old woman receiving regular hemodialysis who had low-grade fever for one month. For these two patients, blood cultures grew S. acidominimus. An antimicrobial susceptibility test revealed that S. acidominimus was resistant to clindamycin and, to some degree, beta-lactam or macrolides. The S. acidominimus from the patient on hemodialysis was resistant to multiple antibiotics. CONCLUSION S. acidominimus is an ever-increasing cause of disease, especially in patients who are critically ill. It is showing increased resistance to antimicrobial agents, so in patients with viridans group streptococci infections, it is necessary to identify the species to improve the clinical management of S. acidominimus.
Collapse
Affiliation(s)
| | | | | | | | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China.
| | | |
Collapse
|
23
|
Hendrickx APA, van Schaik W, Willems RJL. The cell wall architecture of Enterococcus faecium: from resistance to pathogenesis. Future Microbiol 2014; 8:993-1010. [PMID: 23902146 DOI: 10.2217/fmb.13.66] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The cell wall of Gram-positive bacteria functions as a surface organelle that continuously interacts with its environment through a plethora of cell wall-associated molecules. Enterococcus faecium is a normal inhabitant of the GI tract of mammals, but has recently become an important etiological agent of hospital-acquired infections in debilitated patients. Insights into the assembly and function of enterococcal cell wall components and their interactions with the host during colonization and infection are essential to explain the worldwide emergence of E. faecium as an important multiantibiotic-resistant nosocomial pathogen. Understanding the biochemistry of cell wall biogenesis and principles of antibiotic resistance at the molecular level may open up new frontiers in research on enterococci, particularly for the development of novel antimicrobial strategies. In this article, we outline the current knowledge on the most important antimicrobial resistance mechanisms that involve peptidoglycan synthesis and the role of cell wall constituents, including lipoteichoic acid, wall teichoic acid, capsular polysaccharides, LPxTG cell wall-anchored surface proteins, WxL-type surface proteins and pili, in the pathogenesis of E. faecium.
Collapse
Affiliation(s)
- Antoni P A Hendrickx
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | | | |
Collapse
|
24
|
Kelesidis T. The zoonotic potential of daptomycin non-susceptible enterococci. Zoonoses Public Health 2013; 62:1-6. [PMID: 24274811 DOI: 10.1111/zph.12091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Indexed: 11/30/2022]
Abstract
Daptomycin non-susceptible Enterococcus (DNSE) is an emerging clinical problem. Little is known about how de novo DNSE infections develop or the risk factors associated with them. Determining risk factors associated with de novo DNSE infections will aid in understanding the mechanisms of daptomycin non-susceptibility. Humans in contact with animals worldwide are at risk of carriage of multidrug-resistant bacteria. Herein, I review the scientific evidence that supports the hypothesis that transport of daptomycin non-susceptibility genes between animals and humans may be a possible mechanism for development of de novo daptomycin non-susceptibility in enterococci.
Collapse
Affiliation(s)
- T Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
25
|
Aktan Y, Tan S, Icgen B. Characterization of lead-resistant river isolate Enterococcus faecalis and assessment of its multiple metal and antibiotic resistance. ENVIRONMENTAL MONITORING AND ASSESSMENT 2013; 185:5285-93. [PMID: 23079796 DOI: 10.1007/s10661-012-2945-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 10/08/2012] [Indexed: 05/25/2023]
Abstract
Contamination of surface waters has a direct impact on the public health of entire communities. Microorganisms inhabiting contaminated surface waters have developed mechanisms of coping with a variety of toxic metals and drugs. Investigations were carried out to isolate and identify lead-resistant bacteria from the river Kızılırmak along the city of Kırıkkale, Turkey. Of the 33 lead-resistant isolates, one isolate with a minimal inhibitory concentration of 1,200 mg L(-1) was isolated and identified as Enterococcus faecalis by using biochemical tests and 16S rRNA sequencing. Lead-resistant E. faecalis isolate was found out to be resistant to other heavy metals like aluminum, lithium, barium, chromium, iron, silver, tin, nickel, zinc, and strontium and to drugs like amikacin, aztreonam, and gentamicin. E. faecalis harbored four plasmids with the molecular sizes of 1.58, 3.06, 22.76, and 28.95 kb. Plasmid profile analyses of cured derivatives revealed that the lead resistance ability of E. faecalis was still existing despite the elimination of all the plasmids. Moreover, the antibiotic resistance pattern of the cured derivatives did not demonstrate any change from the parental strain. Our findings indicated that the lead resistance genes of E. faecalis were located on the chromosomal DNA rather than the plasmid.
Collapse
Affiliation(s)
- Yasin Aktan
- Department of Biology, Kırıkkale University, 71450 Kırıkkale, Turkey
| | | | | |
Collapse
|
26
|
Nakayama H, Kurokawa K, Lee BL. Lipoproteins in bacteria: structures and biosynthetic pathways. FEBS J 2012; 279:4247-68. [PMID: 23094979 DOI: 10.1111/febs.12041] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/02/2012] [Accepted: 10/19/2012] [Indexed: 11/29/2022]
Abstract
Bacterial lipoproteins are characterized by the presence of a conserved N-terminal lipid-modified cysteine residue that allows the hydrophilic protein to anchor onto bacterial cell membranes. These proteins play important roles in a wide variety of bacterial physiological processes, including virulence, and induce innate immune reactions by functioning as ligands of the mammalian Toll-like receptor 2. We review recent advances in our understanding of bacterial lipoprotein structure, biosynthesis and structure-function relationships between bacterial lipoproteins and Toll-like receptor 2. Notably, 40 years after the first report of the triacyl structure of Braun's lipoprotein in Escherichia coli, recent intensive MS-based analyses have led to the discovery of three new lipidated structures of lipoproteins in monoderm bacteria: the lyso, N-acetyl and peptidyl forms. Moreover, the bacterial lipoprotein structure is considered to be constant in each bacterium; however, lipoprotein structures in Staphylococcus aureus vary between the diacyl and triacyl forms depending on the environmental conditions. Thus, the lipidation state of bacterial lipoproteins, particularly in monoderm bacteria, is more complex than previously assumed.
Collapse
Affiliation(s)
- Hiroshi Nakayama
- Biomolecular Characterization Team, RIKEN Advanced Science Institute, Wako, Saitama, Japan.
| | | | | |
Collapse
|
27
|
A phylum level analysis reveals lipoprotein biosynthesis to be a fundamental property of bacteria. Protein Cell 2012; 3:163-70. [PMID: 22410786 DOI: 10.1007/s13238-012-2023-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Bacterial lipoproteins are proteins that are post-translationally modified with a diacylglyceride at an N-terminal cysteine, which serves to tether these proteins to the outer face of the plasma membrane or to the outer membrane. This paper reviews recent insights into the enzymology of bacterial lipoprotein biosynthesis and localization. Moreover, we use bioinformatic analyses of bacterial lipoprotein signal peptide features and of the key biosynthetic enzymes to consider the distribution of lipoprotein biosynthesis at the phylum level. These analyses support the important conclusion that lipoprotein biosynthesis is a fundamental pathway utilized across the domain bacteria. Moreover, with the exception of a small number of sequences likely to derive from endosymbiont genomes, the enzymes of bacterial lipoprotein biosynthesis appear unique to bacteria, making this pathway an attractive target for the development of novel antimicrobials. Whilst lipoproteins with comparable signal peptide features are encoded in the genomes of Archaea, it is clear that these lipoproteins have a distinctive biosynthetic pathway that has yet to be characterized.
Collapse
|
28
|
Involvement of peptidylprolyl cis/trans isomerases in Enterococcus faecalis virulence. Infect Immun 2012; 80:1728-35. [PMID: 22331431 DOI: 10.1128/iai.06251-11] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Peptidylprolyl cis/trans isomerases (PPIases) are enzymes involved in protein folding. Analysis of the genome sequence of Enterococcus faecalis V583 allowed for identification of 3 PPIases carrying genes. ef2898 encodes an intracellular PPIase which was not shown to be important for the E. faecalis stress response or virulence. The other two PPIases, the parvulin family rotamase EF0685 and the cyclophilin family member EF1534, are expected to be surface-exposed proteins. They were shown to be important for virulence and resistance to NaCl. A Δef0685 Δef1534 mutant was also more resistant to oxidative stress, was able to grow under a high manganese concentration, and showed altered resistance to ampicillin and quinolone antibiotics.
Collapse
|
29
|
Hanson BR, Neely MN. Coordinate regulation of Gram-positive cell surface components. Curr Opin Microbiol 2012; 15:204-10. [PMID: 22236805 DOI: 10.1016/j.mib.2011.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/16/2011] [Accepted: 12/22/2011] [Indexed: 01/07/2023]
Abstract
The cell surface of Gram-positive pathogens represents a complex association of glycopolymers that control cell division, homeostasis, immune evasion, tissue invasion, and resistance to antimicrobials. These glycopolymers include the peptidoglycan cell wall, wall-teichoic acids, lipoteichoic acids, and capsular polysaccharide. Disruption of individual factors often results in pleiotropic effects, making it difficult to discern regulation and function. In this review we collate recent work describing these pleiotropic phenotypes, and propose that this is due to coordinated regulation of biosynthesis or modification of these cell surface components. A better understanding of the regulatory networks that control the relative prevalence of each factor on the cell surface or their modulated functions may help facilitate the identification of new targets for antimicrobial therapy.
Collapse
Affiliation(s)
- Brett R Hanson
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | |
Collapse
|
30
|
Reffuveille F, Serror P, Chevalier S, Budin-Verneuil A, Ladjouzi R, Bernay B, Auffray Y, Rincé A. The prolipoprotein diacylglyceryl transferase (Lgt) of Enterococcus faecalis contributes to virulence. MICROBIOLOGY-SGM 2011; 158:816-825. [PMID: 22135097 DOI: 10.1099/mic.0.055319-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Enterococcus faecalis is an opportunistic pathogen responsible for nosocomial infections. Lipoproteins in Gram-positive bacteria are translocated across the plasma membrane and anchored by the fatty acid group. They perform critical roles, with some described as virulence determinants. The aim of this study was to explore the roles of E. faecalis lipoproteins in the stress response and virulence. We constructed a mutant affected in the predicted prolipoprotein diacylglyceryl transferase gene lgt, and examined the role of Lgt in membrane anchoring, growth, the stress response and virulence. Inactivation of lgt enhanced growth in a high concentration of Mn(2+) or under oxidative stress in vitro, and significantly decreased virulence.
Collapse
Affiliation(s)
- Fany Reffuveille
- USC INRA 2017, Microbiologie de l'Environnement, EA956, Université de Caen, France
| | - Pascale Serror
- AgroParisTech, UMR Micalis, F-78350 Jouy-en-Josas, France
- INRA, UMR1319 Micalis, F-78350 Jouy-en-Josas, France
| | - Sylvie Chevalier
- Laboratoire de Microbiologie du Froid, Signaux et Micro-Environnement, UPRES EA4312, Université de Rouen, France
| | | | - Rabia Ladjouzi
- USC INRA 2017, Microbiologie de l'Environnement, EA956, Université de Caen, France
| | - Benoit Bernay
- Plateforme Proteogen IFR ICORE 146, Université de Caen, France
| | - Yanick Auffray
- USC INRA 2017, Microbiologie de l'Environnement, EA956, Université de Caen, France
| | - Alain Rincé
- USC INRA 2017, Microbiologie de l'Environnement, EA956, Université de Caen, France
| |
Collapse
|