51
|
Logeshwaran P, Krishnan K, Naidu R, Megharaj M. Purification and characterization of a novel fenamiphos hydrolysing enzyme from Microbacterium esteraromaticum MM1. CHEMOSPHERE 2020; 252:126549. [PMID: 32229357 DOI: 10.1016/j.chemosphere.2020.126549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 02/22/2020] [Accepted: 03/17/2020] [Indexed: 05/26/2023]
Abstract
Fenamiphos is a neurotoxic organophosphorus pesticide used widely to control pests of crops. Fenamiphos and its toxic oxidation products have been detected in surface and groundwaters. A novel enzyme capable of hydrolysing P-O-C bond of fenamiphos is purified from Microbacterium esteraromaticum MM1 total cellular protein using a combination of methods. The purified fenamiphos hydrolysing enzyme (FHE) was identified as enolase (phosphopyruvate hydratase), a housekeeping enzyme with molecular mass and pI value of 45 kDa and 4.5, respectively. The optimum pH and temperature for the activity of the FHE are 7 and 25 °C, respectively. We studied the influence of metal ions and inhibitors on the enzyme activity. The enzyme was strongly activated by Mg2+ whereas Hg2+ and phenylmethyl sulfonyl fluoride (PMSF) inhibited the enzyme. The kinetic parameters, Km and Vmax for fenamiphos hydrolysis were estimated to be 584.15 ± 16.22 μM and 6.46 ± 0.13 μM min-1, respectively. The FHE was functionally active against its original substrate (2-phosphoglycerate) with Km value of 5.82 ± 1.42 μM and Vmax of 4.2 ± 0.1 μM min-1. This enzyme has great potential for its application in the detoxification of fenamiphos and its warfare homologs. To our knowledge, this is the first report on the purification of fenamiphos hydrolysing enzyme.
Collapse
Affiliation(s)
- Panneerselvan Logeshwaran
- Global Centre for Environmental Remediation (GCER), Faculty of Science, The University of Newcastle, Callaghan, NSW, 2308, Australia; Cooperative Research Centre for Contamination Assessment and Remediation of the Environment (CRCCARE), ATC Building, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Kannan Krishnan
- Global Centre for Environmental Remediation (GCER), Faculty of Science, The University of Newcastle, Callaghan, NSW, 2308, Australia; Cooperative Research Centre for Contamination Assessment and Remediation of the Environment (CRCCARE), ATC Building, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Ravi Naidu
- Global Centre for Environmental Remediation (GCER), Faculty of Science, The University of Newcastle, Callaghan, NSW, 2308, Australia; Cooperative Research Centre for Contamination Assessment and Remediation of the Environment (CRCCARE), ATC Building, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Mallavarapu Megharaj
- Global Centre for Environmental Remediation (GCER), Faculty of Science, The University of Newcastle, Callaghan, NSW, 2308, Australia; Cooperative Research Centre for Contamination Assessment and Remediation of the Environment (CRCCARE), ATC Building, University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
52
|
Warraich AA, Mohammed AR, Perrie Y, Hussain M, Gibson H, Rahman A. Evaluation of anti-biofilm activity of acidic amino acids and synergy with ciprofloxacin on Staphylococcus aureus biofilms. Sci Rep 2020; 10:9021. [PMID: 32488138 PMCID: PMC7265346 DOI: 10.1038/s41598-020-66082-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Acidic amino acids, aspartic acid (Asp) and glutamic acid (Glu) can enhance the solubility of many poorly soluble drugs including ciprofloxacin (Cip). One of the mechanisms of resistance within a biofilm is retardation of drug diffusion due to poor penetration across the matrix. To overcome this challenge, this work set to investigate novel counter ion approach with acidic amino acids, which we hypothesised will disrupt the biofilm matrix as well as simultaneously improve drug effectiveness. The anti-biofilm activity of D-Asp and D-Glu was studied on Staphylococcus aureus biofilms. Synergistic effect of combining D-amino acids with Cip was also investigated as a strategy to overcome anti-microbial resistance in these biofilms. Interestingly at equimolar combinations, D-Asp and D-Glu were able to significantly disperse (at 20 mM and 40 mM) established biofilms and inhibit (at 10 mM, 20 mM and 40 mM) new biofilm formation in the absence of an antibiotic. Moreover, our study confirmed L-amino acids also exhibit anti-biofilm activity. The synergistic effect of acidic amino acids with Cip was observed at lower concentration ranges (<40 mM amino acids and <90.54 µM, respectively), which resulted in 96.89% (inhibition) and 97.60% (dispersal) reduction in CFU with exposure to 40 mM amino acids. Confocal imaging indicated that the amino acids disrupt the honeycomb-like extracellular DNA (eDNA) meshwork whilst also preventing its formation.
Collapse
Affiliation(s)
- Annsar A Warraich
- Aston Pharmacy School, Aston University, Birmingham, B4 7ET, UK
- University of Wolverhampton, WV1 1LY, Wolverhampton, UK
| | | | - Yvonne Perrie
- University of Strathclyde, Glasgow, G1 1XQ, Scotland
| | | | - Hazel Gibson
- University of Wolverhampton, WV1 1LY, Wolverhampton, UK
| | - Ayesha Rahman
- University of Wolverhampton, WV1 1LY, Wolverhampton, UK.
| |
Collapse
|
53
|
Ploplis VA, Castellino FJ. Host Pathways of Hemostasis that Regulate Group A Streptococcus pyogenes Pathogenicity. Curr Drug Targets 2020; 21:193-201. [PMID: 31556853 PMCID: PMC7670306 DOI: 10.2174/1389450120666190926152914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/02/2019] [Accepted: 09/06/2019] [Indexed: 11/22/2022]
Abstract
A hallmark feature of severe Group A Streptococcus pyogenes (GAS) infection is dysregulated hemostasis. Hemostasis is the primary pathway for regulating blood flow through events that contribute towards clot formation and its dissolution. However, a number of studies have identified components of hemostasis in regulating survival and dissemination of GAS. Several proteins have been identified on the surface of GAS and they serve to either facilitate invasion to host distal sites or regulate inflammatory responses to the pathogen. GAS M-protein, a surface-exposed virulence factor, appears to be a major target for interactions with host hemostasis proteins. These interactions mediate biochemical events both on the surface of GAS and in the solution when M-protein is released into the surrounding environment through shedding or regulated proteolytic processes that dictate the fate of this pathogen. A thorough understanding of the mechanisms associated with these interactions could lead to novel approaches for altering the course of GAS pathogenicity.
Collapse
Affiliation(s)
- Victoria A. Ploplis
- University of Notre Dame, W.M. Keck Center for Transgene Research, 230 Raclin-Carmichael Hall, Notre Dame, IN 46556 USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Francis J. Castellino
- University of Notre Dame, W.M. Keck Center for Transgene Research, 230 Raclin-Carmichael Hall, Notre Dame, IN 46556 USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
54
|
Principle and potential applications of the non-classical protein secretory pathway in bacteria. Appl Microbiol Biotechnol 2019; 104:953-965. [PMID: 31853566 DOI: 10.1007/s00253-019-10285-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/22/2019] [Accepted: 10/27/2019] [Indexed: 12/15/2022]
Abstract
In addition to the extracellular proteins secreted by known secretory pathways, a number of cytoplasmic proteins without predicable or known signal sequences or secretory motifs have been found in the extracellular milieu, and were consequently classified as non-classically secreted proteins. Non-classical protein secretion is considered to be a general, conserved cellular phenomenon in both eukaryotes and prokaryotes. There are several research hotspots on the non-classical protein secretory pathway, and the most important two of them are the recognition principle of substrate proteins and possible secretory mechanisms. To date, researchers have made some progress in understanding the characteristics of these proteins. For example, it was discovered that many non-classically secreted proteins exist and are secreted in multimeric form. Some of these proteins prefer to be clustered and exported at the poles and the septum of the cell. The majority of these proteins play different functions when they are in the intra- and extracellular environments, and several of their functions are related to survival and pathogenicity. Furthermore, non-classically secreted proteins can be used as leading proteins to guide a POI (protein of interest) out of the cells, which provides a novel strategy for protein secretion with potential applications in the industry. Summarizing these findings, this review emphasizes the hot spots related to non-classically secreted proteins in bacteria, lists the most important hypotheses on the selection and secretion mechanisms of non-classically secreted proteins, and put forward their potential applications.
Collapse
|
55
|
Tang T, Chen G, Guo A, Xu Y, Zhao L, Wang M, Lu C, Jiang Y, Zhang C. Comparative proteomic and genomic analyses of Brucella abortus biofilm and planktonic cells. Mol Med Rep 2019; 21:731-743. [PMID: 31974592 PMCID: PMC6947884 DOI: 10.3892/mmr.2019.10888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 07/26/2019] [Indexed: 12/24/2022] Open
Abstract
The present study aimed to explore the differences in protein and gene expression of Brucella abortus cultured under biofilm and planktonic conditions. The proteins unique to biofilms and planktonic B. abortus were separated by two-dimensional (2-D) electrophoresis and then identified by matrix-assisted laser desorption/ionization-tandem time of flight-mass spectrometry (MALDI-TOF/TOF-MS). High-throughput sequencing and bioinformatic analyses were performed to identify differentially expressed genes between B. abortus cultured under biofilm and planktonic conditions. The proteins and genes identified by proteomic and genomic analyses were further evaluated via western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analyses. 2-D electrophoresis identified 20 differentially expressed protein spots between biofilms and planktonic cells, which corresponded to 18 individual proteins (12 downregulated and 6 upregulated) after MALDI-TOF/TOF-MS analysis, including elongation factor Tu and enolase. RT-qPCR analysis revealed that all of the 18 genes were downregulated in biofilms compared with planktonic cells. Western blot analysis identified 9 downregulated and 3 upregulated proteins. High-throughput sequencing and bioinformatic analyses identified 14 function and pathway-associated genes (e.g., BAbS19_I14970). RT-qPCR analysis of the 14 genes showed that they were upregulated in biofilm compared with in planktonic state. In conclusion, these differentially expressed genes may play important roles in bacterial defense, colonization, invasion, and virulence.
Collapse
Affiliation(s)
- Taishan Tang
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Guoqiang Chen
- Division of Animal and Plant Quarantine Supervision, Suzhou Entry Exit Inspection and Quarantine Bureau, Suzhou, Jiangsu 215021, P.R. China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Ye Xu
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| | - Linli Zhao
- The Inspection and Quarantine Technology Center, Inner Mongolia Entry Exit Inspection and Quarantine Bureau, Hohhot, Inner Mongolia 010020, P.R. China
| | - Mengrui Wang
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| | - Chengping Lu
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yuan Jiang
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| | - Changyin Zhang
- Animal, Plant and Food Inspection Center, Jiangsu Entry Exit Inspection and Quarantine Bureau, Nanjing, Jiangsu 210001, P.R. China
| |
Collapse
|
56
|
Rópolo AS, Feliziani C, Touz MC. Unusual proteins in Giardia duodenalis and their role in survival. ADVANCES IN PARASITOLOGY 2019; 106:1-50. [PMID: 31630755 DOI: 10.1016/bs.apar.2019.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The capacity of the parasite Giardia duodenalis to perform complex functions with minimal amounts of proteins and organelles has attracted increasing numbers of scientists worldwide, trying to explain how this parasite adapts to internal and external changes to survive. One explanation could be that G. duodenalis evolved from a structurally complex ancestor by reductive evolution, resulting in adaptation to its parasitic lifestyle. Reductive evolution involves the loss of genes, organelles, and functions that commonly occur in many parasites, by which the host renders some structures and functions redundant. However, there is increasing data that Giardia possesses proteins able to perform more than one function. During recent decades, the concept of moonlighting was described for multitasking proteins, which involves only proteins with an extra independent function(s). In this chapter, we provide an overview of unusual proteins in Giardia that present multifunctional properties depending on the location and/or parasite requirement. We also discuss experimental evidence that may allow some giardial proteins to be classified as moonlighting proteins by examining the properties of moonlighting proteins in general. Up to date, Giardia does not seem to require the numerous redundant proteins present in other organisms to accomplish its normal functions, and thus this parasite may be an appropriate model for understanding different aspects of moonlighting proteins, which may be helpful in the design of drug targets.
Collapse
Affiliation(s)
- Andrea S Rópolo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza Feliziani
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María C Touz
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
57
|
Yuan Y, Ayinuola YA, Singh D, Ayinuola O, Mayfield JA, Quek A, Whisstock JC, Law RHP, Lee SW, Ploplis VA, Castellino FJ. Solution structural model of the complex of the binding regions of human plasminogen with its M-protein receptor from Streptococcus pyogenes. J Struct Biol 2019; 208:18-29. [PMID: 31301349 PMCID: PMC6983471 DOI: 10.1016/j.jsb.2019.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/01/2019] [Accepted: 07/09/2019] [Indexed: 11/22/2022]
Abstract
VEK50 is a truncated peptide from a Streptococcal pyogenes surface human plasminogen (hPg) binding M-protein (PAM). VEK50 contains the full A-domain of PAM, which is responsible for its low nanomolar binding to hPg. The interaction of VEK50 with kringle 2, the PAM-binding domain in hPg (K2hPg), has been studied by high-resolution NMR spectroscopy. The data show that each VEK50 monomer in solution contains two tight binding sites for K2hPg, one each in the a1- (RH1; R17H18) and a2- (RH2; R30H31) repeats within the A-domain of VEK50. Two mutant forms of VEK50, viz., VEK50[RH1/AA] (VEK50ΔRH1) and VEK50[RH2/AA] (VEK50ΔRH2), were designed by replacing each RH with AA, thus eliminating one of the K2hPg binding sites within VEK50, and allowing separate study of each binding site. Using 13C- and 15N-labeled peptides, NMR-derived solution structures of VEK50 in its complex with K2hPg were solved. We conclude that the A-domain of PAM can accommodate two molecules of K2hPg docked within a short distance of each other, and the strength of the binding is slightly different for each site. The solution structure of the VEK50/K2hPg, complex, which is a reductionist model of the PAM/hPg complex, provides insights for the binding mechanism of PAM to a host protein, a process that is critical to S. pyogenes virulence.
Collapse
Affiliation(s)
- Yue Yuan
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Yetunde A Ayinuola
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Damini Singh
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Olawole Ayinuola
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jeffrey A Mayfield
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Adam Quek
- Department of Biochemistry and Molecular Biology, Monash University, Clayton 3800 VIC, Australia
| | - James C Whisstock
- Department of Biochemistry and Molecular Biology, Monash University, Clayton 3800 VIC, Australia
| | - Ruby H P Law
- Department of Biochemistry and Molecular Biology, Monash University, Clayton 3800 VIC, Australia
| | - Shaun W Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Victoria A Ploplis
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Francis J Castellino
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
58
|
Ning M, Xiu Y, Yuan M, Bi J, Hou L, Gu W, Wang W, Meng Q. Spiroplasma eriocheiris Invasion Into Macrobrachium rosenbergii Hemocytes Is Mediated by Pathogen Enolase and Host Lipopolysaccharide and β-1, 3-Glucan Binding Protein. Front Immunol 2019; 10:1852. [PMID: 31440244 PMCID: PMC6694788 DOI: 10.3389/fimmu.2019.01852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 07/23/2019] [Indexed: 01/01/2023] Open
Abstract
Spiroplasma eriocheiris is a crustacean pathogen, without a cell wall, that causes enormous economic loss. Macrobrachium rosenbergii hemocytes are the major targets during S. eriocheiris infection. As wall-less bacteria, S. eriocheiris, its membrane protein should interact with host membrane protein directly and firstly when invaded in host cell. In this investigation, six potential hemocyte receptor proteins were identified firstly that mediate interaction between S. eriocheiris and M. rosenbergii. Among these proteins, lipopolysaccharide and β-1, 3-glucan binding protein (MrLGBP) demonstrated to bind to S. eriocheiris using bacterial binding assays and confocal microscopy. Four spiroplasma ligand proteins for MrLGBP were isolated and identified. But, competitive assessment demonstrated that only enolase of S. eriocheiris (SeEnolase) could be a candidate ligand for MrLGBP. Subsequently, the interaction between MrLGBP and SeEnolase was confirmed by co-immunoprecipitation and co-localization in vitro. After the interaction between MrLGBP and SeEnolase was inhibited by antibody neutralization test, the virulence ability of S. eriocheiris was effectively reduced. The quantity of S. eriocheiris decreased in Drosophila S2 cells after overexpression of MrLGBP, compared with the controls. In addition, RNA interference (RNAi) knockdown of MrLGBP made M. rosenbergii more sensitive to S. eriocheiris infection. Further studies found that the immune genes, including MrLGBP and prophenoloxidase (MrproPO), MrRab7A, and Mrintegrin α1 were significantly up-regulated by SeEnolase stimulation. After SeEnolase pre-stimulation, the ability of M. rosenbergii resistance to S. eriocheiris was significantly improved. Collectively, this investigation demonstrated that MrLGBP and pathogen SeEnolase involved in mediating S. eriocheiris invasion into M. rosenbergii hemocytes.
Collapse
Affiliation(s)
- Mingxiao Ning
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yunji Xiu
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China.,Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, China
| | - Meijun Yuan
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jingxiu Bi
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Libo Hou
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wei Gu
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China.,Co-innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, China
| | - Wen Wang
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Qingguo Meng
- Jiangsu Key Laboratory for Aquatic Crustacean Diseases, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, China.,College of Life Sciences, Nanjing Normal University, Nanjing, China.,Co-innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, China
| |
Collapse
|
59
|
Fischetti VA. Surface Proteins on Gram-Positive Bacteria. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0012-2018. [PMID: 31373270 PMCID: PMC6684298 DOI: 10.1128/microbiolspec.gpp3-0012-2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Indexed: 12/14/2022] Open
Abstract
Surface proteins are critical for the survival of gram-positive bacteria both in the environment and to establish an infection. Depending on the organism, their surface proteins are evolutionarily tailored to interact with specific ligands on their target surface, be it inanimate or animate. Most surface molecules on these organisms are covalently anchored to the peptidoglycan through an LPxTG motif found at the C-terminus. These surface molecules are generally modular with multiple binding or enzymatic domains designed for a specific survival function. For example, some molecules will bind serum proteins like fibronectin or fibrinogen in one domain and have a separate function in another domain. In addition, enzymes such as those responsible for the production of ATP may be generally found on some bacterial surfaces, but when or how they are used in the life of these bacteria is currently unknown. While surface proteins are required for pathogenicity but not viability, targeting the expression of these molecules on the bacterial surface would prevent infection but not death of the organism. Given that the number of different surface proteins could be in the range of two to three dozen, each with two or three separate functional domains (with hundreds to thousands of each protein on a given organism), exemplifies the complexity that exists on the bacterial surface. Because of their number, we could not adequately describe the characteristics of all surface proteins in this chapter. However, since the streptococcal M protein was one of the first gram-positive surface protein to be completely sequenced, and perhaps one of the best studied, we will use M protein as a model for surface proteins in general, pointing out differences with other surface molecules when necessary.
Collapse
Affiliation(s)
- Vincent A Fischetti
- Laboratory of Bacterial Pathogenesis and Immunology, Rockefeller University, New York, NY 10065
| |
Collapse
|
60
|
Echeverría-Valencia G, Silva-Miranda M, Ekaza E, Vallecillo AJ, Parada C, Sada-Ovalle I, Altare F, Espitia C. Interaction of mycobacteria with Plasmin(ogen) affects phagocytosis and granuloma development. Tuberculosis (Edinb) 2019; 117:36-44. [DOI: 10.1016/j.tube.2019.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 04/29/2019] [Accepted: 05/29/2019] [Indexed: 11/30/2022]
|
61
|
Didiasova M, Schaefer L, Wygrecka M. When Place Matters: Shuttling of Enolase-1 Across Cellular Compartments. Front Cell Dev Biol 2019; 7:61. [PMID: 31106201 PMCID: PMC6498095 DOI: 10.3389/fcell.2019.00061] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/05/2019] [Indexed: 12/22/2022] Open
Abstract
Enolase is a glycolytic enzyme, which catalyzes the inter-conversion of 2-phosphoglycerate to phosphoenolpyruvate. Altered expression of this enzyme is frequently observed in cancer and accounts for the Warburg effect, an adaptive response of tumor cells to hypoxia. In addition to its catalytic function, ENO-1 exhibits other activities, which strongly depend on its cellular and extracellular localization. For example, the association of ENO-1 with mitochondria membrane was found to be important for the stability of the mitochondrial membrane, and ENO-1 sequestration on the cell surface was crucial for plasmin-mediated pericellular proteolysis. The latter activity of ENO-1 enables many pathogens but also immune and cancer cells to invade the tissue, leading further to infection, inflammation or metastasis formation. The ability of ENO-1 to conduct so many diverse processes is reflected by its contribution to a high number of pathologies, including type 2 diabetes, cardiovascular hypertrophy, fungal and bacterial infections, cancer, systemic lupus erythematosus, hepatic fibrosis, Alzheimer's disease, rheumatoid arthritis, and systemic sclerosis. These unexpected non-catalytic functions of ENO-1 and their contributions to diseases are the subjects of this review.
Collapse
Affiliation(s)
- Miroslava Didiasova
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University Frankfurt, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.,Member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
62
|
Xia X, Qin W, Zhu H, Wang X, Jiang J, Hu J. How Streptococcus suis serotype 2 attempts to avoid attack by host immune defenses. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 52:516-525. [PMID: 30954397 DOI: 10.1016/j.jmii.2019.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 02/20/2019] [Accepted: 03/07/2019] [Indexed: 01/08/2023]
Abstract
Streptococcus suis (S. suis) type 2 (SS2) is an important zoonotic pathogen that causes swine streptococcosis, a widespread infectious disease that occurs in pig production areas worldwide and causes serious economic losses in the pork industry. Hosts recognize pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs) to activate both innate and acquired immune responses. However, S. suis has evolved multiple mechanisms to escape host defenses. Pathogenic proteins, such as enolase, double-component regulatory systems, factor H-combining proteins and other pathogenic and virulence factors, contribute to immune escape by evading host phagocytosis, reactive oxygen species (ROS), complement-mediated immune destruction, etc. SS2 can prevent neutrophil extracellular trap (NET) formation to avoid being trapped by porcine neutrophils and disintegrate host immunoglobulins via IgA1 hydrolases and IgM proteases. Currently, the pathogenesis of arthritis and meningitis caused by SS2 infection remains unclear, and further studies are necessary to elucidate it. Understanding immune evasion mechanisms after SS2 infection is important for developing high-efficiency vaccines and targeted drugs.
Collapse
Affiliation(s)
- Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China; Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China; Postdoctoral Research Station, Henan Agriculture University, Zhengzhou, China
| | - Wanhai Qin
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity, Meibergdreef 9, 1105AZ Amsterdam, Netherlands
| | - Huili Zhu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Xin Wang
- College of Agriculture and Forestry Science, Linyi University, Linyi, China
| | - Jinqing Jiang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China.
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China; Postdoctoral Research Base, Henan Institute of Science and Technology, Xinxiang, China.
| |
Collapse
|
63
|
Malke H. Genetics and Pathogenicity Factors of Group C and G Streptococci. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0002-2017. [PMID: 30873932 PMCID: PMC11590425 DOI: 10.1128/microbiolspec.gpp3-0002-2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Indexed: 12/17/2022] Open
Abstract
Of the eight phylogenetic groups comprising the genus Streptococcus, Lancefield group C and G streptococci (GCS and GGS, resp.) occupy four of them, including the Pyogenic, Anginosus, and Mitis groups, and one Unnamed group so far. These organisms thrive as opportunistic commensals in both humans and animals but may also be associated with clinically serious infections, often resembling those due to their closest genetic relatives, the group A streptoccci (GAS). Advances in molecular genetics, taxonomic approaches and phylogenomic studies have led to the establishment of at least 12 species, several of which being subdivided into subspecies. This review summarizes these advances, citing 264 early and recent references. It focuses on the molecular structure and genetic regulation of clinically important proteins associated with the cell wall, cytoplasmic membrane and extracellular environment. The article also addresses the question of how, based on the current knowledge, basic research and translational medicine might proceed to further advance our understanding of these multifaceted organisms. Particular emphasis in this respect is placed on streptokinase as the protein determining the host specificity of infection and the Rsh-mediated stringent response with its potential for supporting bacterial survival under nutritional stress conditions.
Collapse
Affiliation(s)
- Horst Malke
- Friedrich Schiller University Jena, Faculty of Biology and Pharmacy, D-07743 Jena, Germany, and University of Oklahoma Health Sciences Center, Department of Microbiology and Immunology, Oklahoma City, OK 73190
| |
Collapse
|
64
|
Hammerschmidt S, Rohde M, Preissner KT. Extracellular Matrix Interactions with Gram-Positive Pathogens. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0041-2018. [PMID: 31004421 PMCID: PMC11590433 DOI: 10.1128/microbiolspec.gpp3-0041-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Indexed: 01/10/2023] Open
Abstract
The main strategies used by pathogenic bacteria to infect eukaryotic tissue include their adherence to cells and the extracellular matrix (ECM), the subsequent colonization and invasion as well as the evasion of immune defences. A variety of structurally and functionally characterized adhesins and binding proteins of gram-positive bacteria facilitate these processes by specifically recognizing and interacting with various components of the host ECM, including different collagens, fibronectin and other macromolecules. The ECM affects the cellular physiology of our body and is critical for adhesion, migration, proliferation, and differentiation of many host cell types, but also provides the support for infiltrating pathogens, particularly under conditions of injury and trauma. Moreover, microbial binding to a variety of adhesive components in host tissue fluids leads to structural and/or functional alterations of host proteins and to the activation of cellular mechanisms that influence tissue and cell invasion of pathogens. Since the diverse interactions of gram-positive bacteria with the ECM represent important pathogenicity mechanisms, their characterization not only allows a better understanding of microbial invasion but also provides clues for the design of novel therapeutic strategies to manage infectious diseases.
Collapse
Affiliation(s)
- Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, University of Greifswald, D-17487 Greifswald, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz-Center for Infection Research, D-38124 Braunschweig, Germany
| | - Klaus T Preissner
- Institute for Biochemistry, Medical School, Justus-Liebig-University, D-35392 Giessen, Germany
| |
Collapse
|
65
|
Wang Y, Wang X, Ali F, Li Z, Fu Y, Yang X, Lin W, Lin X. Comparative Extracellular Proteomics of Aeromonas hydrophila Reveals Iron-Regulated Secreted Proteins as Potential Vaccine Candidates. Front Immunol 2019; 10:256. [PMID: 30833947 PMCID: PMC6387970 DOI: 10.3389/fimmu.2019.00256] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/29/2019] [Indexed: 01/07/2023] Open
Abstract
In our previous study, several iron-related outer membrane proteins in Aeromonas hydrophila, a serious pathogen of farmed fish, conferred high immunoprotectivity to fish, and were proposed as potential vaccine candidates. However, the protective efficacy of these extracellular proteins against A. hydrophila remains largely unknown. Here, we identified secreted proteins that were differentially expressed in A. hydrophila LP-2 in response to iron starvation using an iTRAQ-based quantitative proteomics method. We identified 341 proteins, of which 9 were upregulated in response to iron starvation and 24 were downregulated. Many of the differently expressed proteins were associated with protease activity. We confirmed our proteomics results with Western blotting and qPCR. We constructed three mutants by knocking out three genes encoding differentially expressed proteins (Δorf01830, Δorf01609, and Δorf03641). The physiological characteristics of these mutants were investigated. In all these mutant strains, protease activity decreased, and Δorf01609, and Δorf01830 were less virulent in zebrafish. This indicated that the proteins encoded by these genes may play important roles in bacterial infection. We next evaluated the immune response provoked by the six iron-related recombinant proteins (ORF01609, ORF01830, ORF01839, ORF02943, ORF03355, and ORF03641) in zebrafish as well as the immunization efficacy of these proteins. Immunization with these proteins significantly increased the zebrafish immune response. In addition, the relative percent survival (RPS) of the immunized zebrafish was 50-80% when challenged with three virulent A. hydrophila strains, respectively. Thus, these extracellular secreted proteins might be effective vaccine candidates against A. hydrophila infection in fish.
Collapse
Affiliation(s)
- Yuqian Wang
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaoyun Wang
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,Key Laboratory of Crop Ecology and Molecular Physiology, Fujian Agriculture and Forestry University, Fujian Province University, Fuzhou, China
| | - Farman Ali
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,Key Laboratory of Crop Ecology and Molecular Physiology, Fujian Agriculture and Forestry University, Fujian Province University, Fuzhou, China
| | - Zeqi Li
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,Key Laboratory of Crop Ecology and Molecular Physiology, Fujian Agriculture and Forestry University, Fujian Province University, Fuzhou, China
| | - Yuying Fu
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,Key Laboratory of Crop Ecology and Molecular Physiology, Fujian Agriculture and Forestry University, Fujian Province University, Fuzhou, China
| | - Xiaojun Yang
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,Key Laboratory of Crop Ecology and Molecular Physiology, Fujian Agriculture and Forestry University, Fujian Province University, Fuzhou, China
| | - Wenxiong Lin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,Key Laboratory of Crop Ecology and Molecular Physiology, Fujian Agriculture and Forestry University, Fujian Province University, Fuzhou, China
| | - Xiangmin Lin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,Key Laboratory of Crop Ecology and Molecular Physiology, Fujian Agriculture and Forestry University, Fujian Province University, Fuzhou, China
| |
Collapse
|
66
|
Deshmukh SS, Kornblatt MJ, Kornblatt JA. The influence of truncating the carboxy-terminal amino acid residues of streptococcal enolase on its ability to interact with canine plasminogen. PLoS One 2019; 14:e0206338. [PMID: 30653526 PMCID: PMC6336276 DOI: 10.1371/journal.pone.0206338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/16/2018] [Indexed: 11/23/2022] Open
Abstract
The native octameric structure of streptococcal enolase from Streptococcus pyogenes increasingly dissociates as amino acid residues are removed one by one from the carboxy-terminus. These truncations gradually convert native octameric enolase into monomers and oligomers. In this work, we investigated how these truncations influence the interaction between Streptococcal enolase and canine plasminogen. We used dual polarization interferometry (DPI), localized surface plasmon resonance (LSPR), and sedimentation velocity analytical ultracentrifugation (AUC) to study the interaction. The DPI was our first technique, was performed on all the truncations and used one exclusive kind of chip. The LSRP was used to show that the DPI results were not dependent on the type of chip used. The AUC was required to show that our surface results were not the result of selecting a minority population in any given sample; the majority of the protein was responsible for the binding phenomenon we observed. By comparing results from these techniques we identified one detail that is essential for streptococcal enolase to bind plasminogen: In our hands the individual monomers bind plasminogen; dimers, trimers, tetramers may or may not bind, the fully intact, native, octamer does not bind plasminogen. We also evaluated the contribution to the equilibrium constant made by surface binding as well as in solution. On a surface, the association coefficient is about twice that in solution. The difference is probably not significant. Finally, the fully octameric form of the protein that does not contain a hexa-his N-terminal peptide does not bind to a silicon oxynitride surface, does not bind to an Au-nanoparticle surface, does not bind to a surface coated with Ni-NTA nor does it bind to a surface coated with DPgn. The likelihood is great that the enolase species on the surface of Streptococcus pyogenes is an x-mer of the native octamer.
Collapse
Affiliation(s)
| | - M. Judith Kornblatt
- Department of Chemistry and Biochemistry, Concordia University, Montreal Qc, Canada
| | | |
Collapse
|
67
|
Hu FZ, Król JE, Tsai CHS, Eutsey RA, Hiller LN, Sen B, Ahmed A, Hillman T, Buchinsky FJ, Nistico L, Dice B, Longwell M, Horsey E, Ehrlich GD. Deletion of genes involved in the ketogluconate metabolism, Entner-Doudoroff pathway, and glucose dehydrogenase increase local and invasive virulence phenotypes in Streptococcus pneumoniae. PLoS One 2019; 14:e0209688. [PMID: 30620734 PMCID: PMC6324787 DOI: 10.1371/journal.pone.0209688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/10/2018] [Indexed: 11/18/2022] Open
Abstract
Streptococcus pneumoniae displays increased resistance to antibiotic therapy following biofilm formation. A genome-wide search revealed that SP 0320 and SP 0675 (respectively annotated as 5-keto-D-gluconate-5-reductase and glucose dehydrogenase) contain the highest degree of homology to CsgA of Myxococcus xanthus, a signaling factor that promotes cell aggregation and biofilm formation. Single and double SP 0320 and SP 0675 knockout mutants were created in strain BS72; however, no differences were observed in the biofilm-forming phenotypes of mutants compared to the wild type strain. Using the chinchilla model of otitis media and invasive disease, all three mutants exhibited greatly increased virulence compared to the wild type strain (increased pus formation, tympanic membrane rupture, mortality rates). The SP 0320 gene is located in an operon with SP 0317, SP 0318 and SP 0319, which we bioinformatically annotated as being part of the Entner-Doudoroff pathway. Deletion of SP 0317 also resulted in increased mortality in chinchillas; however, mutations in SP 0318 and SP 0319 did not alter the virulence of bacteria compared to the wild type strain. Complementing the SP 0317, SP 0320 and SP 0675 mutant strains reversed the virulence phenotype. We prepared recombinant SP 0317, SP 0318, SP 0320 and SP 0675 proteins and confirmed their functions. These data reveal that disruption of genes involved in the degradation of ketogluconate, the Entner-Doudoroff pathway, and glucose dehydrogenase significantly increase the virulence of bacteria in vivo; two hypothetical models involving virulence triggered by reduced in carbon-flux through the glycolytic pathways are presented.
Collapse
Affiliation(s)
- Fen Z. Hu
- Center for Genomic Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Otolaryngology-Head and Neck Surgery, Drexel University College of Medicine, Philadelphia, PA, United States of America
- * E-mail: (FZH); (GDE)
| | - Jarosław E. Król
- Center for Genomic Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Center for Advanced Microbial Processing, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Chen Hsuan Sherry Tsai
- Center for Genomic Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Rory A. Eutsey
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Luisa N. Hiller
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Bhaswati Sen
- Center for Genomic Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Center for Advanced Microbial Processing, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Azad Ahmed
- Center for Genomic Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Center for Advanced Microbial Processing, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Todd Hillman
- Center of Excellence in Biofilm Research, Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Farrel J. Buchinsky
- Center of Excellence in Biofilm Research, Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Laura Nistico
- Center of Excellence in Biofilm Research, Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Bethany Dice
- Center of Excellence in Biofilm Research, Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Mark Longwell
- Center of Excellence in Biofilm Research, Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Edward Horsey
- Center of Excellence in Biofilm Research, Allegheny Health Network, Pittsburgh, PA, United States of America
| | - Garth D. Ehrlich
- Center for Genomic Sciences, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Otolaryngology-Head and Neck Surgery, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Center for Advanced Microbial Processing, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States of America
- Center of Excellence in Biofilm Research, Allegheny Health Network, Pittsburgh, PA, United States of America
- * E-mail: (FZH); (GDE)
| |
Collapse
|
68
|
Pancholi V. Group A Streptococcus-Mediated Host Cell Signaling. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0021-2018. [PMID: 30767846 PMCID: PMC11590744 DOI: 10.1128/microbiolspec.gpp3-0021-2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Indexed: 12/14/2022] Open
Abstract
In the past decade, the field of the cellular microbiology of group A Streptococcus (S. pyogenes) infection has made tremendous advances and touched upon several important aspects of pathogenesis, including receptor biology, invasive and evasive phenomena, inflammasome activation, strain-specific autophagic bacterial killing, and virulence factor-mediated programmed cell death. The noteworthy aspect of S. pyogenes-mediated cell signaling is the recognition of the role of M protein in a variety of signaling events, starting with the targeting of specific receptors on the cell surface and on through the induction and evasion of NETosis, inflammasome, and autophagy/xenophagy to pyroptosis and apoptosis. Variations in reports on S. pyogenes-mediated signaling events highlight the complex mechanism of pathogenesis and underscore the importance of the host cell and S. pyogenes strain specificity, as well as in vitro/in vivo experimental parameters. The severity of S. pyogenes infection is, therefore, dependent on the virulence gene expression repertoire in the host environment and on host-specific dynamic signaling events in response to infection. Commonly known as an extracellular pathogen, S. pyogenes finds host macrophages as safe havens wherein it survives and even multiplies. The fact that endothelial cells are inherently deficient in autophagic machinery compared to epithelial cells and macrophages underscores the invasive nature of S. pyogenes and its ability to cause severe systemic diseases. S. pyogenes is still one of the top 10 causes of infectious mortality. Understanding the orchestration of dynamic host signaling networks will provide a better understanding of the increasingly complex mechanism of S. pyogenes diseases and novel ways of therapeutically intervening to thwart severe and often fatal infections.
Collapse
Affiliation(s)
- Vijay Pancholi
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH 43210
| |
Collapse
|
69
|
Galloway-Peña J, DebRoy S, Brumlow C, Li X, Tran TT, Horstmann N, Yao H, Chen K, Wang F, Pan BF, Hawke DH, Thompson EJ, Arias CA, Fowler VG, Bhatti MM, Kalia A, Flores AR, Shelburne SA. Hypervirulent group A Streptococcus emergence in an acaspular background is associated with marked remodeling of the bacterial cell surface. PLoS One 2018; 13:e0207897. [PMID: 30517150 PMCID: PMC6281247 DOI: 10.1371/journal.pone.0207897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/06/2018] [Indexed: 12/28/2022] Open
Abstract
Inactivating mutations in the control of virulence two-component regulatory system (covRS) often account for the hypervirulent phenotype in severe, invasive group A streptococcal (GAS) infections. As CovR represses production of the anti-phagocytic hyaluronic acid capsule, high level capsule production is generally considered critical to the hypervirulent phenotype induced by CovRS inactivation. There have recently been large outbreaks of GAS strains lacking capsule, but there are currently no data on the virulence of covRS-mutated, acapsular strains in vivo. We investigated the impact of CovRS inactivation in acapsular serotype M4 strains using a wild-type (M4-SC-1) and a naturally-occurring CovS-inactivated strain (M4-LC-1) that contains an 11bp covS insertion. M4-LC-1 was significantly more virulent in a mouse bacteremia model but caused smaller lesions in a subcutaneous mouse model. Over 10% of the genome showed significantly different transcript levels in M4-LC-1 vs. M4-SC-1 strain. Notably, the Mga regulon and multiple cell surface protein-encoding genes were strongly upregulated-a finding not observed for CovS-inactivated, encapsulated M1 or M3 GAS strains. Consistent with the transcriptomic data, transmission electron microscopy revealed markedly altered cell surface morphology of M4-LC-1 compared to M4-SC-1. Insertional inactivation of covS in M4-SC-1 recapitulated the transcriptome and cell surface morphology. Analysis of the cell surface following CovS-inactivation revealed that the upregulated proteins were part of the Mga regulon. Inactivation of mga in M4-LC-1 reduced transcript levels of multiple cell surface proteins and reversed the cell surface alterations consistent with the effect of CovS inactivation on cell surface composition being mediated by Mga. CovRS-inactivating mutations were detected in 20% of current invasive serotype M4 strains in the United States. Thus, we discovered that hypervirulent M4 GAS strains with covRS mutations can arise in an acapsular background and that such hypervirulence is associated with profound alteration of the cell surface.
Collapse
Affiliation(s)
- Jessica Galloway-Peña
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Sruti DebRoy
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Chelcy Brumlow
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiqi Li
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Truc T. Tran
- Center for Antimicrobial Resistance and Microbial Genomics and Division of Infectious Diseases, UTHealth McGovern Medical School, Houston, Texas, United States of America
| | - Nicola Horstmann
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hui Yao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Fang Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Bih-Fang Pan
- The Proteomics and Metabolomics Facility, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - David H. Hawke
- The Proteomics and Metabolomics Facility, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Erika J. Thompson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Cesar A. Arias
- Center for Antimicrobial Resistance and Microbial Genomics and Division of Infectious Diseases, UTHealth McGovern Medical School, Houston, Texas, United States of America
- Center for Infectious Diseases, UTHealth School of Public Health, Houston, Texas, United States of America
- Molecular Genetics and Antimicrobial Resistance Unit-International Center for Microbial Genomics, Universidad El Bosque, Bogota, Colombia
| | - Vance G. Fowler
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Micah M. Bhatti
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Awdhesh Kalia
- Graduate Program in Diagnostic Genetics, School of Health Professions, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Anthony R. Flores
- Center for Antimicrobial Resistance and Microbial Genomics and Division of Infectious Diseases, UTHealth McGovern Medical School, Houston, Texas, United States of America
- Department of Pediatrics, University of Texas Health Science Center McGovern Medical School, Houston, Texas, United States of America
| | - Samuel A. Shelburne
- Department of Infectious Diseases Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- The Proteomics and Metabolomics Facility, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
70
|
Di Domenico EG, Cavallo I, Bordignon V, D'Agosto G, Pontone M, Trento E, Gallo MT, Prignano G, Pimpinelli F, Toma L, Ensoli F. The Emerging Role of Microbial Biofilm in Lyme Neuroborreliosis. Front Neurol 2018; 9:1048. [PMID: 30559713 PMCID: PMC6287027 DOI: 10.3389/fneur.2018.01048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 11/19/2018] [Indexed: 01/04/2023] Open
Abstract
Lyme borreliosis (LB) is the most common tick-borne disease caused by the spirochete Borrelia burgdorferi in North America and Borrelia afzelii or Borrelia garinii in Europe and Asia, respectively. The infection affects multiple organ systems, including the skin, joints, and the nervous system. Lyme neuroborreliosis (LNB) is the most dangerous manifestation of Lyme disease, occurring in 10-15% of infected individuals. During the course of the infection, bacteria migrate through the host tissues altering the coagulation and fibrinolysis pathways and the immune response, reaching the central nervous system (CNS) within 2 weeks after the bite of an infected tick. The early treatment with oral antimicrobials is effective in the majority of patients with LNB. Nevertheless, persistent forms of LNB are relatively common, despite targeted antibiotic therapy. It has been observed that the antibiotic resistance and the reoccurrence of Lyme disease are associated with biofilm-like aggregates in B. burgdorferi, B. afzelii, and B. garinii, both in vitro and in vivo, allowing Borrelia spp. to resist to adverse environmental conditions. Indeed, the increased tolerance to antibiotics described in the persisting forms of Borrelia spp., is strongly reminiscent of biofilm growing bacteria, suggesting a possible role of biofilm aggregates in the development of the different manifestations of Lyme disease including LNB.
Collapse
Affiliation(s)
- Enea Gino Di Domenico
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Ilaria Cavallo
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Valentina Bordignon
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Giovanna D'Agosto
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Martina Pontone
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Elisabetta Trento
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Maria Teresa Gallo
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Grazia Prignano
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Fulvia Pimpinelli
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Luigi Toma
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Fabrizio Ensoli
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| |
Collapse
|
71
|
Qiu C, Yuan Y, Zajicek J, Liang Z, Balsara RD, Brito-Robionson T, Lee SW, Ploplis VA, Castellino FJ. Contributions of different modules of the plasminogen-binding Streptococcus pyogenes M-protein that mediate its functional dimerization. J Struct Biol 2018; 204:151-164. [PMID: 30071314 PMCID: PMC6544907 DOI: 10.1016/j.jsb.2018.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/26/2018] [Accepted: 07/28/2018] [Indexed: 10/28/2022]
Abstract
Group A Streptococcus pyogenes (GAS) is a causative agent of pharyngeal and dermal infections in humans. A major virulence determinant of GAS is its dimeric signature fibrillar M-protein (M-Prt), which is evolutionarily designed in modules, ranging from a hypervariable extracellular N-terminal region to a progressively more highly conserved C-terminus that is covalently anchored to the cell wall. Of the >250 GAS isolates classified, only the subset of skin-trophic Pattern D strains expresses a specific serotype of M-Prt, PAM, that directly binds to host human plasminogen (hPg) via its extracellular NH2-terminal variable A-domain region. This interaction allows these GAS strains to accumulate components of the host fibrinolytic system on their surfaces to serve extracellular functions. While structure-function studies have been accomplished on M-Prts from Pattern A-C GAS isolates with different direct ligand binding properties compared to PAM, much less is known regarding the structure-function relationships of PAM-type M-Prts, particularly their dimerization determinants. To examine these questions, PAMs from seven GAS strains with sequence variations in the NH2-terminal ligand binding domains, as well as truncated versions of PAM, were designed and studied. The results from bioinformatic and biophysical analyses show that the different domains of PAM are disparately engaged in dimerization. From these data, we propose an experimentally-based model for PAM secondary and quaternary structures that is highly dependent on the conserved helical C-terminal C-D-domains. In addition, while the N-terminal regions of PAMs are variable in sequence, the binding properties of hPg and its activated product, plasmin, to the A-domain, remain intact.
Collapse
Affiliation(s)
- Cunjia Qiu
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Yue Yuan
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Jaroslav Zajicek
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Zhong Liang
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Rashna D Balsara
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Teresa Brito-Robionson
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Shaun W Lee
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Victoria A Ploplis
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Francis J Castellino
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
72
|
Meyburgh CM, Bragg RR, Boucher CE. Detection of virulence factors of South African Lactococcus garvieae isolated from rainbow trout, Oncorhynchus mykiss (Walbaum). Onderstepoort J Vet Res 2018; 85:e1-e9. [PMID: 30326716 PMCID: PMC6324078 DOI: 10.4102/ojvr.v85i1.1568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 07/20/2018] [Accepted: 07/25/2018] [Indexed: 11/25/2022] Open
Abstract
Lactococcus garvieae is a Gram-positive bacterium that causes mortalities in freshwater and marine fish worldwide and therefore results in severe economic losses in the aquaculture industry. Apart from the apparent integral role of the exopolysaccharide (EPS) capsule in pathogenesis, factors associated with virulence of this bacterium are poorly understood. However, recent studies have indicated that the ability of L. garvieae to cause disease does not depend on the presence of the EPS capsule. Lack of knowledge of virulence factors, pathogenesis and serology of L. garvieae is an impediment to the development of effective typing methods and control measures. This study, therefore, aimed to detect the presence of EPS capsules and other putative virulence factors in South African L. garvieae fish pathogenic isolates and a non-virulent isolate, and to identify possible candidates for subunit vaccine development. No indication of the presence of the EPS capsule was detected by negative staining or amplification of the EPS biosynthesis gene cluster in the virulent isolates or the avirulent strain, discrediting the notion that the EPS capsule is the sole determinant of virulence. However, a set of putative virulence factor genes was detected in all isolates, and candidates for subunit vaccine development (enolase, lactate dehydrogenase phosphoenolpyruvate-protein phosphotransferase) were identified by identification of extracellular proteins of virulent strains.
Collapse
Affiliation(s)
- Cornelia M Meyburgh
- Department of Microbial, Biochemical and Food Biotechnology, University of the Free State.
| | | | | |
Collapse
|
73
|
Dobrut A, Brzozowska E, Górska S, Pyclik M, Gamian A, Bulanda M, Majewska E, Brzychczy-Włoch M. Epitopes of Immunoreactive Proteins of Streptococcus Agalactiae: Enolase, Inosine 5'-Monophosphate Dehydrogenase and Molecular Chaperone GroEL. Front Cell Infect Microbiol 2018; 8:349. [PMID: 30333963 PMCID: PMC6176014 DOI: 10.3389/fcimb.2018.00349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/11/2018] [Indexed: 11/13/2022] Open
Abstract
Three Streptococcus agalactiae (group B streptococci, GBS) immunoreactive proteins: enolase (47.4 kDa), inosine 5'-monophosphate dehydrogenase (IMPDH) (53 kDa) and molecular chaperone GroEL (57 kDa) were subjected to investigation. Enolase protein was described in our previous paper, whereas IMPDH and GroEL were presented for the first time. The aim of our paper was to provide mapping of specific epitopes, highly reactive with umbilical cord blood serum. Bioinformatic analyses allowed to select 32 most likely epitopes for enolase, 36 peptides for IMPDH and 41 immunoreactive peptides for molecular chaperone GroEL, which were synthesized by PEPSCAN. Ten peptides: two in enolase, one in IMPDH and seven in molecular chaperone GroEL have been identified as potentially highly selective epitopes that can be used as markers in rapid immunological diagnostic tests or constitute a component of an innovative vaccine against GBS infections.
Collapse
Affiliation(s)
- Anna Dobrut
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Ewa Brzozowska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Sabina Górska
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marcelina Pyclik
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Andrzej Gamian
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Małgorzata Bulanda
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Elzbieta Majewska
- Department of Clinical Obstetrics and Perinatology, University Hospital, Krakow, Poland
| | - Monika Brzychczy-Włoch
- Department of Molecular Medical Microbiology, Chair of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
74
|
An TJ, Benvenuti MA, Mignemi ME, Thomsen IP, Schoenecker JG. Pediatric Musculoskeletal Infection: Hijacking the Acute-Phase Response. JBJS Rev 2018; 4:01874474-201609000-00004. [PMID: 27760072 DOI: 10.2106/jbjs.rvw.15.00099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tissue injury activates the acute-phase response mediated by the liver, which promotes coagulation, immunity, and tissue regeneration. To survive and disseminate, musculoskeletal pathogens express virulence factors that modulate and hijack this response. As the acute-phase reactants required by these pathogens are most abundant in damaged tissue, these infections are predisposed to occur in tissues following traumatic or surgical injury. Staphylococcus aureus expresses the virulence factors coagulase and von Willebrand binding protein to stimulate coagulation and to form a fibrin abscess that protects it from host immune-cell phagocytosis. After the staphylococcal abscess community reaches quorum, which is the colony density that enables cell-to-cell communication and coordinated gene expression, subsequent expression of staphylokinase stimulates activation of fibrinolysis, which ruptures the abscess wall and results in bacterial dissemination. Unlike Staphylococcus aureus, Streptococcus pyogenes expresses streptokinase and other virulence factors to activate fibrinolysis and to rapidly disseminate throughout the body, causing diseases such as necrotizing fasciitis. Understanding the virulence strategies of musculoskeletal pathogens will help to guide clinical diagnosis and decision-making through monitoring of acute-phase markers such as C-reactive protein, erythrocyte sedimentation rate, and fibrinogen.
Collapse
Affiliation(s)
- Thomas J An
- Departments of Orthopaedics (M.E.M. and J.G.S.), Pediatrics (I.P.T. and J.G.S.), Pediatric Infectious Disease (I.P.T.), Pharmacology (J.G.S.), and Pathology (J.G.S.), Vanderbilt University School of Medicine (T.J.A. and M.A.B.), Nashville, Tennessee
| | | | | | | | | |
Collapse
|
75
|
Dutta S, Tewari A, Balaji C, Verma R, Moitra A, Yadav M, Agrawal P, Sahal D, Jarori GK. Strain-transcending neutralization of malaria parasite by antibodies against Plasmodium falciparum enolase. Malar J 2018; 17:304. [PMID: 30126436 PMCID: PMC6102825 DOI: 10.1186/s12936-018-2455-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/13/2018] [Indexed: 11/17/2022] Open
Abstract
Background Plasmodium enolase is a target for the growth neutralizing antibodies. Interestingly, the three invasive stages i.e. sporozoites, merozoites, and ookinetes express this protein on their cell surface. Polyclonal anti-Plasmodium falciparum enolase (Pfeno) antibodies disrupt traversal of ookinete through mosquito mid-gut wall as well as have inhibitory effect on parasite growth at erythrocytic stage. In a recent study, it was observed that immunization with a unique epitope of parasite enolase (EWGWS) could confer partial protection against mouse malaria. Further validation is needed for the protective potential of this unique epitope in otherwise highly conserved enolase. Methods In order to investigate the efficacy of growth inhibitory potential of the epitope of P falciparum enolase, a monoclonal antibody specific to EWGWS is generated. In vitro parasite growth inhibition assays and passive immunization of Plasmodium yoelii (or Plasmodium berghei) infected mice were used to assess the parasite growth neutralizing activity of the antibody. Results Screening a panel of monoclonal antibodies raised against recombinant Pfeno that were specific to EWGWS resulted in isolation of H12E1. This antibody recognized only EWGWS epitope containing enolases. H12E1 strongly inhibited parasite growth in culture. This inhibition was strain transcending. Passive infusion of this antibody in P. yoelii or P. berghei infected mice showed significant reduction in parasitemia as compared to controls (p < 0.001). Surface Plasmon Resonance measurements indicated high affinity binding of H12E1 to P. falciparum enolase (KD ~ 7.6 × 10−9M). Conclusions A monoclonal antibody directed against EWGWS epitope of Pfeno was shown to inhibit the growth of blood stage malarial parasites. This inhibition was species/strain transcending and is likely to arise due to blockade of enolase on the surface of merozoites, functionally implicating Pfeno in invasion related events. Presence of enolase on the cell surface of merozoites and ookinetes could potentially result in inhibition of host cell invasions at erythrocytic and transmission stages in the parasite life cycle. It is suggested that antibodies against EWGWS epitope have the potential to confer dual stage, species and strain transcending protection against malaria. Electronic supplementary material The online version of this article (10.1186/s12936-018-2455-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sneha Dutta
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, India.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Graduate School of Arts and Sciences, Harvard University, Boston, USA
| | - Aneesha Tewari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, India.,Department of Biology, Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology (MIT), Boston, USA
| | - Chinthapalli Balaji
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, India
| | - Reena Verma
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, India
| | - Anasuya Moitra
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, India
| | - Mamta Yadav
- International Center for Genetic Engineering and Biotechnology, Aruna Asif Ali Marg, New Delhi, India
| | - Prakhar Agrawal
- International Center for Genetic Engineering and Biotechnology, Aruna Asif Ali Marg, New Delhi, India
| | - Dinkar Sahal
- International Center for Genetic Engineering and Biotechnology, Aruna Asif Ali Marg, New Delhi, India
| | - Gotam K Jarori
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, India.
| |
Collapse
|
76
|
Mirasol-Meléndez E, Brieba LG, Díaz-Quezada C, López-Hidalgo M, Figueroa-Angulo EE, Ávila-González L, Arroyo-Verástegui R, Benítez-Cardoza CG. Characterization of multiple enolase genes from Trichomonas vaginalis. Potential novel targets for drug and vaccine design. Parasitol Int 2018; 67:444-453. [DOI: 10.1016/j.parint.2018.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 03/10/2018] [Accepted: 04/11/2018] [Indexed: 10/17/2022]
|
77
|
Frick IM, Shannon O, Neumann A, Karlsson C, Wikström M, Björck L. Streptococcal inhibitor of complement (SIC) modulates fibrinolysis and enhances bacterial survival within fibrin clots. J Biol Chem 2018; 293:13578-13591. [PMID: 30002122 PMCID: PMC6120194 DOI: 10.1074/jbc.ra118.001988] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 07/11/2018] [Indexed: 11/06/2022] Open
Abstract
Some strains of the bacterial pathogen Streptococcus pyogenes secrete protein SIC (streptococcal inhibitor of complement), including strains of the clinically relevant M1 serotype. SIC neutralizes the effect of a number of antimicrobial proteins/peptides and interferes with the function of the host complement system. Previous studies have shown that some S. pyogenes proteins bind and modulate coagulation and fibrinolysis factors, raising the possibility that SIC also may interfere with the activity of these factors. Here we show that SIC interacts with both human thrombin and plasminogen, key components of coagulation and fibrinolysis. We found that during clot formation, SIC binds fibrin through its central region and that SIC inhibits fibrinolysis by interacting with plasminogen. Flow cytometry results indicated that SIC and plasminogen bind simultaneously to S. pyogenes bacteria, and fluorescence microscopy revealed co-localization of the two proteins at the bacterial surface. As a consequence, SIC-expressing bacteria entrapped in clots inhibit fibrinolysis, leading to delayed bacterial escape from the clots as compared with mutant bacteria lacking SIC. Moreover, within the clots SIC-expressing bacteria were protected against killing. In an animal model of subcutaneous infection, SIC-expressing bacteria exhibited a delayed systemic spread. These results demonstrate that the bacterial protein SIC interferes with coagulation and fibrinolysis and thereby enhances bacterial survival, a finding that has significant implications for S. pyogenes virulence.
Collapse
Affiliation(s)
- Inga-Maria Frick
- From the Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, SE-22184 Lund, Sweden and
| | - Oonagh Shannon
- From the Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, SE-22184 Lund, Sweden and
| | - Ariane Neumann
- From the Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, SE-22184 Lund, Sweden and
| | - Christofer Karlsson
- From the Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, SE-22184 Lund, Sweden and
| | - Mats Wikström
- the University of Copenhagen, Protein Function and Interactions Group, Novo Nordisk Foundation Center for Protein Research, DK-2200 Copenhagen, Denmark
| | - Lars Björck
- From the Department of Clinical Sciences, Lund, Division of Infection Medicine, Lund University, SE-22184 Lund, Sweden and
| |
Collapse
|
78
|
Jeffery CJ. Protein moonlighting: what is it, and why is it important? Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0523. [PMID: 29203708 DOI: 10.1098/rstb.2016.0523] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2017] [Indexed: 12/23/2022] Open
Abstract
Members of the GroEL/HSP60 protein family have been studied for many years because of their critical roles as ATP-dependent molecular chaperones, so it might come as a surprise that some have important functions in ATP-poor conditions, for example, when secreted outside the cell. At least some members of each of the HSP10, HSP70, HSP90, HSP100 and HSP110 heat shock protein families are also 'moonlighting proteins'. Moonlighting proteins exhibit more than one physiologically relevant biochemical or biophysical function within one polypeptide chain. In this class of multifunctional proteins, the multiple functions are not due to gene fusions or multiple proteolytic fragments. Several hundred moonlighting proteins have been identified, and they include a diverse set of proteins with a large variety of functions. Some participate in multiple biochemical processes by using an active site pocket for catalysis and a different part of the protein's surface to interact with other proteins. Moonlighting proteins play a central role in many diseases, and the development of novel treatments would be aided by more information addressing current questions, for example, how some are targeted to multiple cellular locations and how a single function can be targeted by therapeutics without targeting a function not involved in disease.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.
Collapse
Affiliation(s)
- Constance J Jeffery
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
79
|
Xu J, Huang X, Dong X, Ren Y, Wu M, Shen N, Xie Y, Gu X, Lai W, Jing B, Peng X, Yang G. Serodiagnostic Potential of Alpha-Enolase From Sarcoptes scabiei and Its Possible Role in Host-Mite Interactions. Front Microbiol 2018; 9:1024. [PMID: 29887838 PMCID: PMC5981165 DOI: 10.3389/fmicb.2018.01024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/30/2018] [Indexed: 12/15/2022] Open
Abstract
Infestation of the epidermis with the highly contagious ectoparasite, Sarcoptes scabiei, causes scabies, which is characterized by intense itching, pruritus, and secondary infection. This condition affects humans, livestock, and wildlife worldwide, incurring large economic losses and reducing the quality of human life. In the present study, we cloned the alpha-enolase, a key enzyme in the glycolytic and gluconeogenesis pathways, from S. scabiei var. cuniculi, characterized it and produced soluble recombinant enolase protein (rSsc-eno). We determined the localization of Ssc-eno in isolated mites and mites in lesioned skin. The results showed that native enolase was intensely localized in the tegument of the mouthparts, the entire legs, and the whole mites' body, as well as in the gut and reproduction system. Interestingly, we found that native enolase was widely distributed in mites in lesioned skin, with obvious high protein intensity compared with isolated mites. Building on good immunoreactivity, an indirect enzyme-linked immunosorbent assay (ELISA) based on rSsc-eno showed 92% sensitivity and 95.8% specificity, compared with other indirect ELISA in this study, rSsc-eno based ELISA is better in detecting scabies in rabbits. Besides, this method can detect S. scabiei infection as early as 1 week post infection. Compared with other detection methods, such as traditional microscopic examination and recently published universal conventional PCR, rSsc-eno ELISA was more effective to detect early infection in rabbits. Additionally, in vitro incubation experiments demonstrated the concentration-dependent acaricidal activity of rabbit anti-rSsc-eno sera against larval mites, suggested its potential as a vaccine candidate.
Collapse
Affiliation(s)
- Jing Xu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xing Huang
- Chengdu Agricultural College, Chengdu, China
| | - Xiaowei Dong
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yongjun Ren
- Sichuan Animal Sciences Academy, Chengdu, Sichuan, China.,Animal Breeding and Genetics Key Laboratory of Sichuan Province, Chengdu, China
| | - Maodi Wu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Nengxing Shen
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yue Xie
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaobin Gu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Weiming Lai
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xuerong Peng
- College of Science, Sichuan Agricultural University, Ya'an, China
| | - Guangyou Yang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
80
|
Recombinant Enolase of Trypanosoma cruzi as a Novel Vaccine Candidate against Chagas Disease in a Mouse Model of Acute Infection. J Immunol Res 2018; 2018:8964085. [PMID: 29854848 PMCID: PMC5964559 DOI: 10.1155/2018/8964085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/03/2018] [Indexed: 01/18/2023] Open
Abstract
Trypanosoma cruzi is the protozoan parasite that causes Chagas disease, which is considered by the World Health Organization to be a neglected tropical disease. Two drugs exist for the treatment of Chagas disease, nifurtimox and benznidazole; they are only effective in the acute phase, and a vaccine is currently not available. In this study, we used the recombinant enolase from T. cruzi H8 strain (MHOM/MX/1992/H8 Yucatán) (rTcENO) and its encoding DNA (pBKTcENO) to immunize mice and evaluate their protective effects in an experimental murine model of acute phase infection. Our results showed that mice vaccinated with rTcENO or its encoding DNA were able to generate typical specific antibodies (IgG1, IgG2a, and IgG2b), suggesting that a mixed Th1/Th2 immune response was induced. The parasite burden in the blood was reduced to 69.8% and 71% in mice vaccinated with rTcENO and pBKTcENO, respectively. The group vaccinated with rTcENO achieved 75% survival, in contrast to the group vaccinated with pBKTcENO that showed no survival in comparison to the control groups. Moreover, rTcENO immunization elevated the production of IFN-γ and IL-2 after the parasite challenge, suggesting that the Th1-type immune response was polarized. These results indicated that rTcENO could be used as a vaccine against Chagas disease.
Collapse
|
81
|
Rahi A, Dhiman A, Singh D, Lynn AM, Rehan M, Bhatnagar R. Exploring the interaction between Mycobacterium tuberculosis enolase and human plasminogen using computational methods and experimental techniques. J Cell Biochem 2018; 119:2408-2417. [PMID: 28888036 DOI: 10.1002/jcb.26403] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/29/2017] [Indexed: 12/16/2022]
Abstract
Surface localized microbial enolases' binding with human plasminogen has been increasingly proven to have an important role in initial infection cycle of several human pathogens. Likewise, surface localized Mycobacterium tuberculosis (Mtb) enolase also binds to human plasminogen, and this interaction may entail crucial consequences for granuloma stability. The current study is the first attempt to explore the plasminogen interacting residues of enolase from Mtb. Beginning with the structural modeling of Mtb enolase, the binding pose of Mtb enolase and human plasminogen was predicted using protein-protein docking simulations. The binding pose revealed the interface region with interacting residues and molecular interactions. Next, the interacting residues were refined and ranked by using various criteria. Finally, the selected interacting residues were tested experimentally for their involvement in plasminogen binding. The two consecutive lysine residues, Lys-193 and Lys-194, turned out to be active residues for plasminogen binding. These residues when substituted for alanine along with the most active residue Lys-429, that is, the triple mutant (K193A + K194A + K429A) Mtb enolase, exhibited 40% reduction in plasminogen binding. It is worth noting that Mtb enolase lost nearly half of the plasminogen binding activity with only three simultaneous substitutions, without any significant secondary structure perturbation. Further, the sequence comparison between Mtb and human enolase isoforms suggests the possibility of selective targeting of Mtb enolase to obstruct binding of human plasminogen.
Collapse
Affiliation(s)
- Amit Rahi
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Alisha Dhiman
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Damini Singh
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Andrew M Lynn
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Mohd Rehan
- King, Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
82
|
Kudryavtseva AV, Krasnov GS, Dmitriev AA, Alekseev BY, Kardymon OL, Sadritdinova AF, Fedorova MS, Pokrovsky AV, Melnikova NV, Kaprin AD, Moskalev AA, Snezhkina AV. Mitochondrial dysfunction and oxidative stress in aging and cancer. Oncotarget 2018; 7:44879-44905. [PMID: 27270647 PMCID: PMC5216692 DOI: 10.18632/oncotarget.9821] [Citation(s) in RCA: 374] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/28/2016] [Indexed: 12/16/2022] Open
Abstract
Aging and cancer are the most important issues to research. The population in the world is growing older, and the incidence of cancer increases with age. There is no doubt about the linkage between aging and cancer. However, the molecular mechanisms underlying this association are still unknown. Several lines of evidence suggest that the oxidative stress as a cause and/or consequence of the mitochondrial dysfunction is one of the main drivers of these processes. Increasing ROS levels and products of the oxidative stress, which occur in aging and age-related disorders, were also found in cancer. This review focuses on the similarities between ageing-associated and cancer-associated oxidative stress and mitochondrial dysfunction as their common phenotype.
Collapse
Affiliation(s)
- Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga L Kardymon
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Asiya F Sadritdinova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Maria S Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | | |
Collapse
|
83
|
Jeffery CJ. Keeping good friends close - The surface and secreted proteomes of a probiotic bacterium provide candidate proteins for intestinal attachment and communication with the host. Proteomics 2018; 17. [PMID: 28517912 DOI: 10.1002/pmic.201700112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022]
Abstract
Bacteria use cell surface proteins and secreted proteins to interact with host tissues. Several dozen previously published proteomics studies have identified cell surface proteins for pathogens. In this issue, Celebioglu and Svensson (Proteomics 2017, 17, 1700019) use 2D gel electrophoresis and mass spectrometry to identify secreted and cell surface proteins of a commensual gut bacterium, Lactobacillus acidophilus NCFM. Some of the proteins are known to have functions in the cytoplasm, and their presence on the cell surface suggests they might be moonlighting proteins. In addition, comparisons of proteins used by pathogenic and probiotic species to interact with their hosts could lead to improved treatments of infections and chronic diseases that are associated with an imbalance of pathogenic and probiotic gut bacteria.
Collapse
Affiliation(s)
- Constance J Jeffery
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
84
|
Morozov GI, Porat N, Kushnir T, Najmuldeen H, Adawi A, Chalifa-Caspi V, Benisty R, Ohayon A, Liron O, Azriel S, Malka I, Dotan S, Portnoi M, Piotrowski AA, Kafka D, Hajaj B, Fishilevich T, Shagan M, Tal M, Ellis R, Morrison DA, Mitchell AM, Mitchell TJ, Dagan R, Yesilkaya H, Nebenzahl YM. Flavin Reductase Contributes to Pneumococcal Virulence by Protecting from Oxidative Stress and Mediating Adhesion and Elicits Protection Against Pneumococcal Challenge. Sci Rep 2018; 8:314. [PMID: 29321514 PMCID: PMC5762878 DOI: 10.1038/s41598-017-18645-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/01/2017] [Indexed: 12/26/2022] Open
Abstract
Pneumococcal flavin reductase (FlaR) is known to be cell-wall associated and possess age dependent antigenicity in children. This study aimed at characterizing FlaR and elucidating its involvement in pneumococcal physiology and virulence. Bioinformatic analysis of FlaR sequence identified three-conserved cysteine residues, suggesting a transition metal-binding capacity. Recombinant FlaR (rFlaR) bound Fe2+ and exhibited FAD-dependent NADP-reductase activity, which increased in the presence of cysteine or excess Fe2+ and inhibited by divalent-chelating agents. flaR mutant was highly susceptible to H2O2 compared to its wild type (WT) and complemented strains, suggesting a role for FlaR in pneumococcal oxidative stress resistance. Additionally, flaR mutant demonstrated significantly decreased mice mortality following intraperitoneal infection. Interestingly, lack of FlaR did not affect the extent of phagocytosis by primary mouse peritoneal macrophages but reduced adhesion to A549 cells compared to the WT and complemented strains. Noteworthy are the findings that immunization with rFlaR elicited protection in mice against intraperitoneal lethal challenge and anti-FlaR antisera neutralized bacterial virulence. Taken together, FlaR's roles in pneumococcal physiology and virulence, combined with its lack of significant homology to human proteins, point towards rFlaR as a vaccine candidate.
Collapse
Affiliation(s)
- Giora I Morozov
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nurith Porat
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Tatyana Kushnir
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hastyar Najmuldeen
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, United Kingdom.,Department of Biology, College of Science, University of Sulaimani, Sulaymaniyah, Iraq
| | - Asad Adawi
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vered Chalifa-Caspi
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rachel Benisty
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Ariel Ohayon
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Shalhevet Azriel
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Itai Malka
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | - Andrew A Piotrowski
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Barak Hajaj
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Tali Fishilevich
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Marilou Shagan
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | - Donald A Morrison
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrea M Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Timothy J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ron Dagan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hasan Yesilkaya
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, United Kingdom
| | - Yaffa Mizrachi Nebenzahl
- The Shraga Segal Department of Microbiology and Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
85
|
Delabranche X, Helms J, Meziani F. Immunohaemostasis: a new view on haemostasis during sepsis. Ann Intensive Care 2017; 7:117. [PMID: 29197958 PMCID: PMC5712298 DOI: 10.1186/s13613-017-0339-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022] Open
Abstract
Host infection by a micro-organism triggers systemic inflammation, innate immunity and complement pathways, but also haemostasis activation. The role of thrombin and fibrin generation in host defence is now recognised, and thrombin has become a partner for survival, while it was seen only as one of the "principal suspects" of multiple organ failure and death during septic shock. This review is first focused on pathophysiology. The role of contact activation system, polyphosphates and neutrophil extracellular traps has emerged, offering new potential therapeutic targets. Interestingly, newly recognised host defence peptides (HDPs), derived from thrombin and other "coagulation" factors, are potent inhibitors of bacterial growth. Inhibition of thrombin generation could promote bacterial growth, while HDPs could become novel therapeutic agents against pathogens when resistance to conventional therapies grows. In a second part, we focused on sepsis-induced coagulopathy diagnostic challenge and stratification from "adaptive" haemostasis to "noxious" disseminated intravascular coagulation (DIC) either thrombotic or haemorrhagic. Besides usual coagulation tests, we discussed cellular haemostasis assessment including neutrophil, platelet and endothelial cell activation. Then, we examined therapeutic opportunities to prevent or to reduce "excess" thrombin generation, while preserving "adaptive" haemostasis. The fail of international randomised trials involving anticoagulants during septic shock may modify the hypothesis considering the end of haemostasis as a target to improve survival. On the one hand, patients at low risk of mortality may not be treated to preserve "immunothrombosis" as a defence when, on the other hand, patients at high risk with patent excess thrombin and fibrin generation could benefit from available (antithrombin, soluble thrombomodulin) or ongoing (FXI and FXII inhibitors) therapies. We propose to better assess coagulation response during infection by an improved knowledge of pathophysiology and systematic testing including determination of DIC scores. This is one of the clues to allocate the right treatment for the right patient at the right moment.
Collapse
Affiliation(s)
- Xavier Delabranche
- Université de Strasbourg, Faculté de Médecine & Hôpitaux Universitaires de Strasbourg, Service de Réanimation, Nouvel Hôpital Civil, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Université de Strasbourg, Strasbourg, France
| | - Julie Helms
- Université de Strasbourg, Faculté de Médecine & Hôpitaux Universitaires de Strasbourg, Service de Réanimation, Nouvel Hôpital Civil, Strasbourg, France
- INSERM, EFS Grand Est, BPPS UMR-S 949, Université de Strasbourg, Strasbourg, France
| | - Ferhat Meziani
- Université de Strasbourg, Faculté de Médecine & Hôpitaux Universitaires de Strasbourg, Service de Réanimation, Nouvel Hôpital Civil, Strasbourg, France
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), FMTS, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
86
|
Rox K, Jansen R, Loof TG, Gillen CM, Bernecker S, Walker MJ, Chhatwal GS, Müller R. Linoleic and palmitoleic acid block streptokinase-mediated plasminogen activation and reduce severity of invasive group A streptococcal infection. Sci Rep 2017; 7:11798. [PMID: 28924140 PMCID: PMC5603603 DOI: 10.1038/s41598-017-11276-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/17/2017] [Indexed: 01/06/2023] Open
Abstract
In contrast to mild infections of Group A Streptococcus (GAS) invasive infections of GAS still pose a serious health hazard: GAS disseminates from sterile sites into the blood stream or deep tissues and causes sepsis or necrotizing fasciitis. In this case antibiotics do not provide an effective cure as the bacteria are capable to hide from them very quickly. Therefore, new remedies are urgently needed. Starting from a myxobacterial natural products screening campaign, we identified two fatty acids isolated from myxobacteria, linoleic and palmitoleic acid, specifically blocking streptokinase-mediated activation of plasminogen and thereby preventing streptococci from hijacking the host’s plasminogen/plasmin system. This activity is not inherited by other fatty acids such as oleic acid and is not attributable to the killing of streptococci. Moreover, both fatty acids are superior in their inhibitory properties compared to two clinically used drugs (tranexamic or ε-amino caproic acid) as they show 500–1000 fold lower IC50 values. Using a humanized plasminogen mouse model mimicking the clinical situation of a local GAS infection that becomes systemic, we demonstrate that these fatty acids ameliorate invasive GAS infection significantly. Consequently, linoleic and palmitoleic acid are possible new options to combat GAS invasive diseases.
Collapse
Affiliation(s)
- Katharina Rox
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany.,Department of Medical Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.,Central facility for Microscopy, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.,German Centre for Infection Research (DZIF), Partner Site Braunschweig-Hannover, Hannover, Germany
| | - Rolf Jansen
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.,German Centre for Infection Research (DZIF), Partner Site Braunschweig-Hannover, Hannover, Germany
| | - Torsten G Loof
- Department of Medical Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.,Infection Immunology Research Group, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Christine M Gillen
- School of Chemistry and Molecular Biosciences and Australian Infectious Disease Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Steffen Bernecker
- Department of Microbial Drugs, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.,German Centre for Infection Research (DZIF), Partner Site Braunschweig-Hannover, Hannover, Germany
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences and Australian Infectious Disease Research Centre, The University of Queensland, St. Lucia, Queensland, Australia
| | - Gursharan Singh Chhatwal
- Department of Medical Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany. .,German Centre for Infection Research (DZIF), Partner Site Braunschweig-Hannover, Hannover, Germany.
| |
Collapse
|
87
|
Mahato JP, Rana S, Kumar M, Sarsaiya S. Homology Modeling of Leishmania donovani Enolase and its Molecular Interaction with Novel Inhibitors. J Pharm Bioallied Sci 2017; 9:99-105. [PMID: 28717332 PMCID: PMC5508423 DOI: 10.4103/jpbs.jpbs_241_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction: The treatment of Indian tropical disease such as kala-azar is likely to be troublesome to the clinicians as AmpB- and miltefosine-resistant Leishmania donovani has been reported. The rationale behind designed a novel inhibitors of model of L. donovani enolase and performing a binding study with its inhibitors to gain details of the interaction between protein residues and ligand molecules. Methods and Materials: The L. donovani enolase model consists of two typical domains. The N-terminal one contains three-stranded antiparallel β-sheets, followed by six α-helices. The C-terminal domain composes of eleven-stranded mixed α/β-barrel with connectivity. The first α-helix within the C-terminal domain, H7, and the second β-strand, S7, of the barrel domain was arranged in an antiparallel fashion compared to all other α-helices and β-strands. The root-mean-square deviation between predicted model and template is 0.4 Å. The overall conformation of L. donovani enolase model is similar to those of Trypanosoma cruzi enolase and Streptococcus pneumoniae enolase crystal structures. Result: The key amino acid residues within the docking complex model involved in the interaction between model enolase structure and ligand molecule are Lys70, Asn165, Ala168, Asp17, and Asn213. Conclusion: Our theoretical prediction may lead to the establishment of prophylactic and therapeutic approaches for the treatment of kala-azar. This biomedical informatics analysis will help us to combat future kala-azar.
Collapse
Affiliation(s)
- Jay Prakash Mahato
- Department of Biotechnology, Sri Satya Sai University of Technology and Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Sindhuprava Rana
- Department of Biotechnology, Sri Satya Sai University of Technology and Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Maneesh Kumar
- Department of Biotechnology, College of Commerce, Arts and Science (Magadh University, Bodh Gaya), Patna, Bihar, India
| | - Surendra Sarsaiya
- Department of Biotechnology, College of Commerce, Arts and Science (Magadh University, Bodh Gaya), Patna, Bihar, India
| |
Collapse
|
88
|
Alves LA, Harth-Chu EN, Palma TH, Stipp RN, Mariano FS, Höfling JF, Abranches J, Mattos-Graner RO. The two-component system VicRK regulates functions associated with Streptococcus mutans resistance to complement immunity. Mol Oral Microbiol 2017; 32:419-431. [PMID: 28382721 DOI: 10.1111/omi.12183] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/07/2017] [Accepted: 03/29/2017] [Indexed: 12/16/2022]
Abstract
Streptococcus mutans, a dental caries pathogen, can promote systemic infections upon reaching the bloodstream. The two-component system (TCS) VicRKSm of S. mutans regulates the synthesis of and interaction with sucrose-derived exopolysaccharides (EPS), processes associated with oral and systemic virulence. In this study, we investigated the mechanisms by which VicRKSm affects S. mutans susceptibility to blood-mediated immunity. Compared with parent strain UA159, the vicKSm isogenic mutant (UAvic) showed reduced susceptibility to deposition of C3b of complement, low binding to serum immunoglobulin G (IgG), and low frequency of C3b/IgG-mediated opsonophagocytosis by polymorphonuclear cells in a sucrose-independent way (P<.05). Reverse transcriptase quantitative polymerase chain reaction analysis comparing gene expression in UA159 and UAvic revealed that genes encoding putative peptidases of the complement (pepO and smu.399) were upregulated in UAvic in the presence of serum, although genes encoding murein hydrolases (SmaA and Smu.2146c) or metabolic/surface proteins involved in bacterial interactions with host components (enolase, GAPDH) were mostly affected in a serum-independent way. Among vicKSm -downstream genes (smaA, smu.2146c, lysM, atlA, pepO, smu.399), only pepO and smu.399 were associated with UAvic phenotypes; deletion of both genes in UA159 significantly enhanced levels of C3b deposition and opsonophagocytosis (P<.05). Moreover, consistent with the fibronectin-binding function of PepO orthologues, UAvic showed increased binding to fibronectin. Reduced susceptibility to opsonophagocytosis was insufficient to enhance ex vivo persistence of UAvic in blood, which was associated with growth defects of this mutant under limited nutrient conditions. Our findings revealed that S. mutans employs mechanisms of complement evasion through peptidases, which are controlled by VicRKSm.
Collapse
Affiliation(s)
- Livia A Alves
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Erika N Harth-Chu
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Thais H Palma
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Rafael N Stipp
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Flávia S Mariano
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - José F Höfling
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Jacqueline Abranches
- Department of Oral Biology, College of Dentistry - University of Florida, Gainesville, FL, USA
| | - Renata O Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| |
Collapse
|
89
|
Robb M, Hobbs JK, Woodiga SA, Shapiro-Ward S, Suits MDL, McGregor N, Brumer H, Yesilkaya H, King SJ, Boraston AB. Molecular Characterization of N-glycan Degradation and Transport in Streptococcus pneumoniae and Its Contribution to Virulence. PLoS Pathog 2017; 13:e1006090. [PMID: 28056108 PMCID: PMC5215778 DOI: 10.1371/journal.ppat.1006090] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/27/2016] [Indexed: 11/19/2022] Open
Abstract
The carbohydrate-rich coating of human tissues and cells provide a first point of contact for colonizing and invading bacteria. Commensurate with N-glycosylation being an abundant form of protein glycosylation that has critical functional roles in the host, some host-adapted bacteria possess the machinery to process N-linked glycans. The human pathogen Streptococcus pneumoniae depolymerizes complex N-glycans with enzymes that sequentially trim a complex N-glycan down to the Man3GlcNAc2 core prior to the release of the glycan from the protein by endo-β-N-acetylglucosaminidase (EndoD), which cleaves between the two GlcNAc residues. Here we examine the capacity of S. pneumoniae to process high-mannose N-glycans and transport the products. Through biochemical and structural analyses we demonstrate that S. pneumoniae also possesses an α-(1,2)-mannosidase (SpGH92). This enzyme has the ability to trim the terminal α-(1,2)-linked mannose residues of high-mannose N-glycans to generate Man5GlcNAc2. Through this activity SpGH92 is able to produce a substrate for EndoD, which is not active on high-mannose glycans with α-(1,2)-linked mannose residues. Binding studies and X-ray crystallography show that NgtS, the solute binding protein of an ABC transporter (ABCNG), is able to bind Man5GlcNAc, a product of EndoD activity, with high affinity. Finally, we evaluated the contribution of EndoD and ABCNG to growth of S. pneumoniae on a model N-glycosylated glycoprotein, and the contribution of these enzymes and SpGH92 to virulence in a mouse model. We found that both EndoD and ABCNG contribute to growth of S. pneumoniae, but that only SpGH92 and EndoD contribute to virulence. Therefore, N-glycan processing, but not transport of the released glycan, is required for full virulence in S. pneumoniae. To conclude, we synthesize our findings into a model of N-glycan processing by S. pneumoniae in which both complex and high-mannose N-glycans are targeted, and in which the two arms of this degradation pathway converge at ABCNG. Streptococcus pneumoniae (pneumococcus) is a bacterium that causes extensive morbidity and mortality in humans. Vaccines and antibiotics are effective forms of prevention and treatment, respectively, but present challenges as it is a constant race to vaccinate against the enormous and ever evolving pool of different serotypes of the bacterium while resistance to antibiotics continues to trend upwards. It is thus necessary to better understand the molecular aspects of the host-pneumococcus interaction in order to inform the potential generation of alternative treatment strategies. S. pneumoniae relies on its ability to process the carbohydrates presented on the surface of host cells for full-virulence. In this study, we examine the capability of the bacterium to process high-mannose N-linked sugars, a heretofore unknown ability for S. pneumoniae. The results show that the pneumococcal genome encodes enzymes capable of processing these sugars and that, remarkably, the initiating reaction performed by an enzyme that removes terminal α-(1,2)-linked mannose residues is critical to virulence in a mouse model. This study illuminates an extensive pathway in S. pneumoniae that targets N-linked sugars and is key to the host-pathogen interaction, therefore revealing a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Melissa Robb
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Joanne K. Hobbs
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Shireen A. Woodiga
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Sarah Shapiro-Ward
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Michael D. L. Suits
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Nicholas McGregor
- Michael Smith Laboratories and Department of Chemistry, University of British Columbia, 2185 East Mall, Vancouver, British Columbia, Canada
| | - Harry Brumer
- Michael Smith Laboratories and Department of Chemistry, University of British Columbia, 2185 East Mall, Vancouver, British Columbia, Canada
| | - Hasan Yesilkaya
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, United Kingdom
| | - Samantha J. King
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Alisdair B. Boraston
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
- * E-mail:
| |
Collapse
|
90
|
Jiang H, Chen T, Sun H, Tang Z, Yu J, Lin Z, Ren P, Zhou X, Huang Y, Li X, Yu X. Immune response induced by oral delivery of Bacillus subtilis spores expressing enolase of Clonorchis sinensis in grass carps (Ctenopharyngodon idellus). FISH & SHELLFISH IMMUNOLOGY 2017; 60:318-325. [PMID: 27729275 DOI: 10.1016/j.fsi.2016.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/03/2016] [Accepted: 10/06/2016] [Indexed: 06/06/2023]
Abstract
Clonorchiasis, caused by the consumption of raw or undercooked freshwater fish containing infective metacercariae of Clonorchis sinensisis (C.sinensis), remains a common public health problem. New effective prevention strategies are still urgent to control this food-borne infectious disease. The previous studies suggested Bacillus subtilis (B. subtilis) spores was an ideal vaccines delivery system, and the C.sinensis enolase (CsENO) was a potential vaccine candidate against clonorchiasis. In the current study, we detected CsENO-specific IgM levels by ELISA in sera, intestinal mucus and skin mucus in grass carps (Ctenopharyngodon idella) through oral administration with B. subtilis spores surface expressing CsENO. In addition, immune-related genes expression was also measured by qRT-PCR. Grass carps orally treated with B. subtilis spores or normal forages were used as controls. The results of ELISA manifested that specific IgM levels of grass carps in CsENO group in sera, intestine mucus and skin mucus almost significantly increased from week 4 post the first oral administration when compared to the two control groups. The levels of specific IgM reached its peak in intestine mucus firstly, then in sera, and last in skin mucus. qRT-PCR results showed that 5 immune-related genes expression had different degree of rising trend in CsENO group when compared to the two control groups. Our study demonstrated that orally administrated with B. subtilis spores expressing CsENO induced innate and adaptive immunity, systemic and local mucosal immunity, and humoral and cellular immunity. Our work may pave the way to clarify the exact mechanisms of protective efficacy elicited by B. subtilis spores expressing CsENO and provide new ideas for vaccine development against C. sinensis infection.
Collapse
Affiliation(s)
- Hongye Jiang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Tingjin Chen
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hengchang Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zeli Tang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jinyun Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhipeng Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Pengli Ren
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xinyi Zhou
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yan Huang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xuerong Li
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xinbing Yu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory for Tropical Diseases Control of Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
91
|
ENOblock Does Not Inhibit the Activity of the Glycolytic Enzyme Enolase. PLoS One 2016; 11:e0168739. [PMID: 28030597 PMCID: PMC5193436 DOI: 10.1371/journal.pone.0168739] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 11/20/2016] [Indexed: 11/19/2022] Open
Abstract
Inhibition of glycolysis is of great potential for the treatment of cancer. However, inhibitors of glycolytic enzymes with favorable pharmacological profiles have not been forthcoming. Due to the nature of their active sites, most high-affinity transition-state analogue inhibitors of glycolysis enzymes are highly polar with poor cell permeability. A recent publication reported a novel, non-active site inhibitor of the glycolytic enzyme Enolase, termed ENOblock (N-[2-[2-2-aminoethoxy)ethoxy]ethyl]4-4-cyclohexylmethyl)amino]6-4-fluorophenyl)methyl]amino]1,3,5-triazin-2-yl]amino]benzeneacetamide). This would present a major advance, as this is heterocyclic and fully cell permeable molecule. Here, we present evidence that ENOblock does not inhibit Enolase enzymatic activity in vitro as measured by three different assays, including a novel 31P NMR based method which avoids complications associated with optical interferences in the UV range. Indeed, we note that due to strong UV absorbance, ENOblock interferes with the direct spectrophotometric detection of the product of Enolase, phosphoenolpyruvate. Unlike established Enolase inhibitors, ENOblock does not show selective toxicity to ENO1-deleted glioma cells in culture. While our data do not dispute the biological effects previously attributed to ENOblock, they indicate that such effects must be caused by mechanisms other than direct inhibition of Enolase enzymatic activity.
Collapse
|
92
|
Weng Y, Chen F, Liu Y, Zhao Q, Chen R, Pan X, Liu C, Cheng Z, Jin S, Jin Y, Wu W. Pseudomonas aeruginosa Enolase Influences Bacterial Tolerance to Oxidative Stresses and Virulence. Front Microbiol 2016; 7:1999. [PMID: 28018326 PMCID: PMC5156722 DOI: 10.3389/fmicb.2016.01999] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram negative opportunistic pathogenic bacterium, which causes acute and chronic infections. Upon entering the host, bacteria alter global gene expression to adapt to host environment and avoid clearance by the host. Enolase is a glycolytic enzyme involved in carbon metabolism. It is also a component of RNA degradosome, which is involved in RNA processing and gene regulation. Here, we report that enolase is required for the virulence of P. aeruginosa in a murine acute pneumonia model. Mutation of enolase coding gene (eno) increased bacterial susceptibility to neutrophil mediated killing, which is due to reduced tolerance to oxidative stress. Catalases and alkyl hydroperoxide reductases play a major role in protecting the cell from oxidative damages. In the eno mutant, the expression levels of catalases (KatA and KatB) were similar as those in the wild type strain in the presence of H2O2, however, the expression levels of alkyl hydroperoxide reductases (AhpB and AhpC) were significantly reduced. Overexpression of ahpB but not ahpC in the eno mutant fully restored the bacterial resistance to H2O2 as well as neutrophil mediated killing, and partially restored bacterial virulence in the murine acute pneumonia model. Therefore, we have identified a novel role of enolase in the virulence of P. aeruginosa.
Collapse
Affiliation(s)
- Yuding Weng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Fei Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Yiwei Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Ronghao Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Chang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Shouguang Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai UniversityTianjin, China; Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, GainesvilleFL, USA
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University Tianjin, China
| |
Collapse
|
93
|
Global Analysis and Comparison of the Transcriptomes and Proteomes of Group A Streptococcus Biofilms. mSystems 2016; 1:mSystems00149-16. [PMID: 27933318 PMCID: PMC5141267 DOI: 10.1128/msystems.00149-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/01/2016] [Indexed: 11/20/2022] Open
Abstract
Prokaryotes are thought to regulate their proteomes largely at the level of transcription. However, the results from this first set of global transcriptomic and proteomic analyses of paired microbial samples presented here show that this assumption is false for the majority of genes and their products in S. pyogenes. In addition, the tenuousness of the link between transcription and translation becomes even more pronounced when microbes exist in a biofilm or a stationary planktonic state. Since the transcriptome level does not usually equal the proteome level, the validity attributed to gene expression studies as well as proteomic studies in microbial analyses must be brought into question. Therefore, the results attained by either approach, whether RNA-seq or shotgun proteomics, must be taken in context and evaluated with particular care since they are by no means interchangeable. To gain a better understanding of the genes and proteins involved in group A Streptococcus (GAS; Streptococcus pyogenes) biofilm growth, we analyzed the transcriptome, cellular proteome, and cell wall proteome from biofilms at different stages and compared them to those of plankton-stage GAS. Using high-throughput RNA sequencing (RNA-seq) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) shotgun proteomics, we found distinct expression profiles in the transcriptome and proteome. A total of 46 genes and 41 proteins showed expression across the majority of biofilm time points that was consistently higher or consistently lower than that seen across the majority of planktonic time points. However, there was little overlap between the genes and proteins on these two lists. In line with other studies comparing transcriptomic and proteomic data, the overall correlation between the two data sets was modest. Furthermore, correlation was poorest for biofilm samples. This suggests a high degree of regulation of protein expression by nontranscriptional mechanisms. This report illustrates the benefits and weaknesses of two different approaches to global expression profiling, and it also demonstrates the advantage of using proteomics in conjunction with transcriptomics to gain a more complete picture of global expression within biofilms. In addition, this report provides the fullest characterization of expression patterns in GAS biofilms currently available. IMPORTANCE Prokaryotes are thought to regulate their proteomes largely at the level of transcription. However, the results from this first set of global transcriptomic and proteomic analyses of paired microbial samples presented here show that this assumption is false for the majority of genes and their products in S. pyogenes. In addition, the tenuousness of the link between transcription and translation becomes even more pronounced when microbes exist in a biofilm or a stationary planktonic state. Since the transcriptome level does not usually equal the proteome level, the validity attributed to gene expression studies as well as proteomic studies in microbial analyses must be brought into question. Therefore, the results attained by either approach, whether RNA-seq or shotgun proteomics, must be taken in context and evaluated with particular care since they are by no means interchangeable.
Collapse
|
94
|
Kinnby B, Chávez de Paz LE. Plasminogen coating increases initial adhesion of oral bacteria in vitro. Microb Pathog 2016; 100:10-16. [DOI: 10.1016/j.micpath.2016.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 06/22/2016] [Accepted: 08/02/2016] [Indexed: 11/29/2022]
|
95
|
Hymes JP, Klaenhammer TR. Stuck in the Middle: Fibronectin-Binding Proteins in Gram-Positive Bacteria. Front Microbiol 2016; 7:1504. [PMID: 27713740 PMCID: PMC5031765 DOI: 10.3389/fmicb.2016.01504] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/08/2016] [Indexed: 11/13/2022] Open
Abstract
Fibronectin is a multidomain glycoprotein found ubiquitously in human body fluids and extracellular matrices of a variety of cell types from all human tissues and organs, including intestinal epithelial cells. Fibronectin plays a major role in the regulation of cell migration, tissue repair, and cell adhesion. Importantly, fibronectin also serves as a common target for bacterial adhesins in the gastrointestinal tract. Fibronectin-binding proteins (FnBPs) have been identified and characterized in a wide variety of host-associated bacteria. Single bacterial species can contain multiple, diverse FnBPs. In pathogens, some FnBPs contribute to virulence via host cell attachment, invasion, and interference with signaling pathways. Although FnBPs in commensal and probiotic strains are not sufficient to confer virulence, they are essential for attachment to their ecological niches. Here we describe the interaction between human fibronectin and bacterial adhesins by highlighting the FnBPs of Gram-positive pathogens and commensals. We provide an overview of the occurrence and diversity of FnBPs with a focus on the model pathogenic organisms in which FnBPs are most characterized. Continued investigation of FnBPs is needed to fully understand their divergence and specificity in both pathogens and commensals.
Collapse
Affiliation(s)
- Jeffrey P Hymes
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
| | - Todd R Klaenhammer
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
| |
Collapse
|
96
|
Rahi A, Matta SK, Dhiman A, Garhyan J, Gopalani M, Chandra S, Bhatnagar R. Enolase of Mycobacterium tuberculosis is a surface exposed plasminogen binding protein. Biochim Biophys Acta Gen Subj 2016; 1861:3355-3364. [PMID: 27569900 DOI: 10.1016/j.bbagen.2016.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/12/2016] [Accepted: 08/24/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Enolase, a glycolytic enzyme, has long been studied as an anchorless protein present on the surface of many pathogenic bacteria that aids in tissue remodeling and invasion by binding to host plasminogen. METHODS Anti-Mtb enolase antibodies in human sera were detected using ELISA. Immunoelectron microscopy, immunofluorescence microscopy and flow cytometry were used to show surface localization of Mtb enolase. SPR was used to determine the affinity of enolase-plasminogen interaction. Plasmin formation upon plasminogen binding to enolase and Mtb surface was measured by ELISA. Mice challenge and histopathological studies were undertaken to determine the protective efficacy of enolase immunization. RESULTS Enolase of Mtb is present on its surface and binds human plasminogen with high affinity. There was an average of 2-fold increase in antibody mediated recognition of Mtb enolase in human sera from TB patients with an active disease over control individuals. Substitution of C-terminal lysine to alanine in rEno decreased its binding affinity with human plasminogen by >2-folds. Enolase bound plasminogen showed urokinase mediated conversion into plasmin. Binding of plasminogen to the surface of Mtb and its conversion into fibrinolytic plasmin was significantly reduced in the presence of anti-rEno antibodies. Immunization with rEno also led to a significant decrease in lung CFU counts of mice upon infection with Mtb H37Rv. CONCLUSIONS Mtb enolase is a surface exposed plasminogen binding protein which upon immunization confers significant protection against Mtb challenge. GENERAL SIGNIFICANCE Plasminogen binding has been recognized for Mtb, however, proteins involved have not been characterized. We show here that Mtb enolase is a moonlighting plasminogen binding protein.
Collapse
Affiliation(s)
- Amit Rahi
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Sumit Kumar Matta
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Alisha Dhiman
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Jaishree Garhyan
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Monisha Gopalani
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Subhash Chandra
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Rakesh Bhatnagar
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
97
|
Nitzsche R, Köhler J, Kreikemeyer B, Oehmcke-Hecht S. Streptococcus pyogenes Escapes Killing from Extracellular Histones through Plasminogen Binding and Activation by Streptokinase. J Innate Immun 2016; 8:589-600. [PMID: 27533300 DOI: 10.1159/000448039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/28/2016] [Indexed: 01/05/2023] Open
Abstract
Histones are small basic proteins and highly conserved among eukaryotes. Their main function is binding, packaging and organizing of DNA in the nucleus, but extracellular histones are also potent antimicrobial proteins. Here we found that Streptococcus pyogenes - an important human pathogen - protects itself from histone-killing by the acquisition of plasminogen. Plasminogen, bound to the streptococcal surface, efficiently prevents histone-mediated killing. Moreover, the streptokinase/plasminogen complex degrades all classes of histones and abrogates their antibacterial and hemolytic effects. This novel streptokinase-mediated virulence mechanism may contribute to the escape of S. pyogenes from the human innate immune system.
Collapse
Affiliation(s)
- Ramona Nitzsche
- University Medicine, Institute of Medical Microbiology, Virology and Hygiene, Rostock University, Rostock, Germany
| | | | | | | |
Collapse
|
98
|
Spurbeck RR, Harris PT, Raghunathan K, Arvidson DN, Arvidson CG. A Moonlighting Enolase from Lactobacillus gasseri does not Require Enzymatic Activity to Inhibit Neisseria gonorrhoeae Adherence to Epithelial Cells. Probiotics Antimicrob Proteins 2016; 7:193-202. [PMID: 25917402 DOI: 10.1007/s12602-015-9192-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Enolases are generally thought of as cytoplasmic enzymes involved in glycolysis and gluconeogenesis. However, several bacteria have active forms of enolase associated with the cell surface and these proteins are utilized for functions other than central metabolism. Recently, a surface-associated protein produced by Lactobacillus gasseri ATCC 33323 with homology to enolase was found to inhibit the adherence of the sexually transmitted pathogen, Neisseria gonorrhoeae, to epithelial cells in culture. Here, we show that the protein is an active enolase in vitro. A recombinantly expressed, C-terminal His-tagged version of the protein, His6-Eno3, inhibited gonococcal adherence. Assays utilizing inhibitors of enolase enzymatic activity showed that this inhibitory activity required the substrate-binding site to be in an open conformation; however, the enolase enzymatic activity of the protein was not necessary for inhibition of gonococcal adherence. An L. gasseri strain carrying an insertional mutation in eno3 was viable, indicating that eno3 is not an essential gene in L. gasseri 33323. This observation, along with the results of the enzyme assays, is consistent with reports that this strain encodes more than one enolase. Here we show that the three L. gasseri genes annotated as encoding an enolase are expressed. The L. gasseri eno3 mutant exhibited reduced, but not abolished, inhibition of gonococcal adherence, which supports the hypothesis that L. gasseri inhibition of gonococcal adherence is a multifactorial process.
Collapse
Affiliation(s)
- Rachel R Spurbeck
- The Genetics Program, Michigan State University, East Lansing, MI, USA
| | | | | | | | | |
Collapse
|
99
|
The Fibrinolytic System in the Interstitial Space. Protein Sci 2016. [DOI: 10.1201/9781315374307-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
100
|
Kylväjä R, Ojalehto T, Kainulainen V, Virkola R, Westerlund-Wikström B. Penicillin binding protein 3 of Staphylococcus aureus NCTC 8325-4 binds and activates human plasminogen. BMC Res Notes 2016; 9:389. [PMID: 27488131 PMCID: PMC4972960 DOI: 10.1186/s13104-016-2190-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/28/2016] [Indexed: 11/25/2022] Open
Abstract
Background Staphylococcus aureus is a versatile pathogen expressing a number of virulence-associated adhesive molecules. In a previous study, we generated in a secretion-competent Escherichia coli strain a library of random FLAG-tag positive (FTP) polypeptides of S. aureus. To identify adhesive proteins and gain additional knowledge on putative virulence factors of S. aureus, we here screened the FTP library against human serum proteins. Findings Staphylococcus aureus NCTC 8325-4, origin of the FTP library, adhered to immobilized plasminogen in vitro. In an enzyme-linked immunoassay a C-terminal part of penicillin binding protein 3 (PBP3), included in the FTP library, bound to immobilized plasminogen. We expressed and purified full-length PBP3 and its C-terminal fragments as recombinant proteins. In a time-resolved fluorometry—based assay the PBP3 polypeptides bound to immobilized plasminogen. The polypeptides enhanced formation of plasmin from plasminogen as analyzed by cleavage of a chromogenic plasmin substrate. Conclusions The present findings, although preliminary, demonstrate reliably that S. aureus NCTC 8325-4 adheres to immobilized plasminogen in vitro and that the adhesion may be mediated by a C-terminal fragment of the PBP3 protein. The full length PBP3 and the penicillin binding C-terminal domain of PBP3 expressed as recombinant proteins bound plasminogen and activated plasminogen to plasmin. These phenomena were inhibited by the lysine analogue ε-aminocaproic acid suggesting that the binding is mediated by lysine residues. A detailed molecular description of surface molecules enhancing the virulence of S. aureus will aid in understanding of its pathogenicity and help in design of antibacterial drugs in the future. Electronic supplementary material The online version of this article (doi:10.1186/s13104-016-2190-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Riikka Kylväjä
- General Microbiology, Department of Biosciences, University of Helsinki, P.O.Box 56, FI-00014, University of Helsinki, Helsinki, Finland.,Thermo Fisher Scientific, Ratastie 2, 01620, Vantaa, Finland
| | - Tuomas Ojalehto
- General Microbiology, Department of Biosciences, University of Helsinki, P.O.Box 56, FI-00014, University of Helsinki, Helsinki, Finland.,Orion Diagnostica, Koivu-Mankkaan tie 6, 02200, Espoo, Finland
| | - Veera Kainulainen
- General Microbiology, Department of Biosciences, University of Helsinki, P.O.Box 56, FI-00014, University of Helsinki, Helsinki, Finland.,Pharmacology, Faculty of Medicine, University of Helsinki, P.O.Box 63, FI-00014, University of Helsinki, Helsinki, Finland
| | - Ritva Virkola
- General Microbiology, Department of Biosciences, University of Helsinki, P.O.Box 56, FI-00014, University of Helsinki, Helsinki, Finland
| | - Benita Westerlund-Wikström
- General Microbiology, Department of Biosciences, University of Helsinki, P.O.Box 56, FI-00014, University of Helsinki, Helsinki, Finland.
| |
Collapse
|