1
|
Montgomery MT, Ortigoza M, Loomis C, Weiser JN. Neuraminidase-mediated enhancement of Streptococcus pneumoniae colonization is associated with altered mucus characteristics and distribution. mBio 2024:e0257924. [PMID: 39660923 DOI: 10.1128/mbio.02579-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
Upon entry into the upper respiratory tract (URT), Streptococcus pneumoniae (Spn) upregulates neuraminidases (NA) that cleave sialic acid (SA) from host glycans. Because sialylation is thought to contribute to the physical properties that determine mucus function, we posited that Spn directly alters host mucus through NA activity. By directly imaging the colonized URT, we demonstrated NA-mediated alterations to the characteristics and distribution of mucus along the respiratory epithelium, where colonizing bacteria are found. Mucus exposed to NA showed increased localization within goblet cells and lining the glycocalyx. By contrast, NA-naïve mucus was more likely to be observed sloughing away from the epithelial surface. We also visualized Spn in the URT and observed that NA promoted efficient bacterial localization to the firm mucus layer overlying the glycocalyx, whereas NA-deficient Spn was associated more with loose mucus. By facilitating tighter association with the glycocalyx, NA promoted increased Spn colonization density. The magnitude of the NA-mediated effect on colonization was widened during late colonization by increased evasion of host-mediated clearance mechanisms. Thus, Spn-encoded NAs directly modify the host environment by desialylating mucus, which allows close interaction with mucus at the epithelium, and this is associated with enhanced bacterial colonization. IMPORTANCE Although severe illness and death caused by Spn result from secondary invasive diseases including pneumonia, sepsis, and meningitis, stable colonization of the upper respiratory tract (URT) is a prerequisite to invasive disease. Therefore, understanding host-Spn dynamics during asymptomatic colonization of the URT is warranted with respect to the pathogenesis of Spn disease. In this study, we found that Spn NA activity directly alters mucus characteristics that result in increased density and duration of URT colonization. Therefore, targeting Spn NA activity during URT colonization may be a viable strategy to mitigate Spn infection.
Collapse
Affiliation(s)
- Matthew T Montgomery
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
| | - Mila Ortigoza
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, New York University School of Medicine, New York, New York, USA
| | - Cynthia Loomis
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Jeffrey N Weiser
- Department of Microbiology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
2
|
Yang S, Chen J, Fu J, Huang J, Li T, Yao Z, Ye X. Disease-Associated Streptococcus pneumoniae Genetic Variation. Emerg Infect Dis 2024; 30:39-49. [PMID: 38146979 PMCID: PMC10756394 DOI: 10.3201/eid3001.221927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023] Open
Abstract
Streptococcus pneumoniae is an opportunistic pathogen that causes substantial illness and death among children worldwide. The genetic backgrounds of pneumococci that cause infection versus asymptomatic carriage vary substantially. To determine the evolutionary mechanisms of opportunistic pathogenicity, we conducted a genomic surveillance study in China. We collected 783 S. pneumoniae isolates from infected and asymptomatic children. By using a 2-stage genomewide association study process, we compared genomic differences between infection and carriage isolates to address genomic variation associated with pathogenicity. We identified 8 consensus k-mers associated with adherence, antimicrobial resistance, and immune modulation, which were unevenly distributed in the infection isolates. Classification accuracy of the best k-mer predictor for S. pneumoniae infection was good, giving a simple target for predicting pathogenic isolates. Our findings suggest that S. pneumoniae pathogenicity is complex and multifactorial, and we provide genetic evidence for precise targeted interventions.
Collapse
|
3
|
Egorova A, Richter M, Khrenova M, Dietrich E, Tsedilin A, Kazakova E, Lepioshkin A, Jahn B, Chernyshev V, Schmidtke M, Makarov V. Pyrrolo[2,3- e]indazole as a novel chemotype for both influenza A virus and pneumococcal neuraminidase inhibitors. RSC Adv 2023; 13:18253-18261. [PMID: 37350858 PMCID: PMC10282731 DOI: 10.1039/d3ra02895j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023] Open
Abstract
Influenza infections are often exacerbated by secondary bacterial infections, primarily caused by Streptococcus pneumoniae. Both respiratory pathogens have neuraminidases that support infection. Therefore, we hypothesized that dual inhibitors of viral and bacterial neuraminidases might be an advantageous strategy for treating seasonal and pandemic influenza pneumonia complicated by bacterial infections. By screening our in-house chemical library, we discovered a new chemotype that may be of interest for a further campaign to find small molecules against influenza. Our exploration of the pyrrolo[2,3-e]indazole space led to the identification of two hit compounds, 6h and 12. These molecules were well-tolerated by MDCK cells and inhibited the replication of H3N2 and H1N1 influenza A virus strains. Moreover, both compounds suppress viral and pneumococcal neuraminidases indicating their dual activity. Given its antiviral activity, pyrrolo[2,3-e]indazole has been identified as a promising scaffold for the development of novel neuraminidase inhibitors that are active against influenza A virus and S. pneumoniae.
Collapse
Affiliation(s)
- Anna Egorova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS) 33-2 Leninsky Prospect 119071 Moscow Russia
| | - Martina Richter
- Department of Medical Microbiology, Section of Experimental Virology, Jena University Hospital Hans-Knöll-Straße 2 07745 Jena Germany
| | - Maria Khrenova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS) 33-2 Leninsky Prospect 119071 Moscow Russia
- Chemistry Department, Lomonosov Moscow State University 1-3 Leninskie Gory 119991 Moscow Russia
| | - Elisabeth Dietrich
- Department of Medical Microbiology, Section of Experimental Virology, Jena University Hospital Hans-Knöll-Straße 2 07745 Jena Germany
| | - Andrey Tsedilin
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS) 33-2 Leninsky Prospect 119071 Moscow Russia
| | - Elena Kazakova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS) 33-2 Leninsky Prospect 119071 Moscow Russia
| | - Alexander Lepioshkin
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS) 33-2 Leninsky Prospect 119071 Moscow Russia
| | - Birgit Jahn
- Department of Medical Microbiology, Section of Experimental Virology, Jena University Hospital Hans-Knöll-Straße 2 07745 Jena Germany
| | - Vladimir Chernyshev
- Chemistry Department, Lomonosov Moscow State University 1-3 Leninskie Gory 119991 Moscow Russia
- Frumkin Institute of Physical Chemistry and Electrochemistry of the Russian Academy of Sciences 31-4 Leninsky Prospect 119071 Moscow Russia
| | - Michaela Schmidtke
- Department of Medical Microbiology, Section of Experimental Virology, Jena University Hospital Hans-Knöll-Straße 2 07745 Jena Germany
| | - Vadim Makarov
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS) 33-2 Leninsky Prospect 119071 Moscow Russia
| |
Collapse
|
4
|
Feng C, Cross AS, Vasta GR. Galectin-1 mediates interactions between polymorphonuclear leukocytes and vascular endothelial cells, and promotes their extravasation during lipopolysaccharide-induced acute lung injury. Mol Immunol 2023; 156:127-135. [PMID: 36921487 PMCID: PMC10154945 DOI: 10.1016/j.molimm.2023.02.011] [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: 10/17/2022] [Revised: 01/29/2023] [Accepted: 02/26/2023] [Indexed: 03/14/2023]
Abstract
The lung airway epithelial surface is heavily covered with sialic acids as the terminal carbohydrate on most cell surface glycoconjugates and can be removed by microbial neuraminidases or endogenous sialidases. By desialylating the lung epithelial surface, neuraminidase acts as an important virulence factor in many mucosal pathogens, such as influenza and S. pneumoniae. Desialylation exposes the subterminal galactosyl moieties - the binding glycotopes for galectins, a family of carbohydrate-recognition proteins playing important roles in various aspects of immune responses. Galectin-1 and galectin-3 have been extensively studied in their roles related to host immune responses, but some questions about their role(s) in leukocyte recruitment during lung bacterial infection remain unanswered. In this study, we found that both galectin-1 and galectin-3 bind to polymorphonuclear leukocytes (PMNs) and enhance the interaction of endothelial intercellular adhesion molecule-1 (ICAM-1) with PMNs, which is further increased by PMN desialylation. In addition, we observed that in vitro galectin-1 mediates the binding of PMNs, particularly desialylated PMNs, onto the endothelial cells. Finally, in a murine model for LPS-mediated acute lung injury, we observed that galectin-1 modulates PMN infiltration to the lung without altering the expression of chemoattractant cytokines. We conclude that galectins, particularly galectin-1, may function as adhesion molecules that mediate PMN-endothelial cell interactions, and modulate PMN infiltration during acute lung injury.
Collapse
Affiliation(s)
- Chiguang Feng
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA; Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Alan S Cross
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gerardo R Vasta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA; Institute of Marine and Environmental Technology, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
5
|
Loop 422–437 in NanA from Streptococcus pneumoniae plays the role of an active site lid and is associated with allosteric regulation. Comput Biol Med 2022; 144:105290. [DOI: 10.1016/j.compbiomed.2022.105290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/20/2022] [Accepted: 02/01/2022] [Indexed: 11/03/2022]
|
6
|
Bazmi RR, Panichayupakaranant P. Synergistic interactions between artocarpin-rich extract, lawsone methyl ether and ampicillin on anti-MRSA and their antibiofilm formation. Lett Appl Microbiol 2022; 74:777-786. [PMID: 35100449 DOI: 10.1111/lam.13662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/05/2023]
Abstract
Artocarpin-rich extract (ARE) was prepared using a green technology and standardized to contain 49.6% w/w artocarpin, while lawsone methyl ether was prepared using a green semi-synthesis. ARE, LME and ampicillin exhibited weak anti-MRSA activity with the MICs of 31.2-62.5 µg/mL. Based on the checkerboard assay, the synergistic interaction between ARE (0.03 µg/mL) and LME (0.49 µg/mL) against four MRSA isolates were observed with the fractional inhibitory concentration index (FICI) value of 0.008, while those of ARE (1.95-7.81 µg/mL) and ampicillin (0.49 µg/mL) as well as LME (0.49-1.95 µg/mL) and ampicillin (0.49 µg/mL) were 0.016-0.257. The time kill confirmed the synergistic interactions against MRSA with different degrees. The combination of ARE and LME as well as its combinations with ampicillin altered the membrane permeability of MRSA, which led to release of the intracellular materials. In addition, each compound inhibited the biofilm formation of standard MRSA (DMST 20654) and the clinical isolate (MRSA 1096). These findings suggested that cocktails containing ARE and LME might be used to overcome problems associated with MRSA. Additionally, the results implied that combination of either ARE or LME with available conventional antibiotic agents might be effective in countering these perilous pathogens.
Collapse
Affiliation(s)
- Rizwan Rashid Bazmi
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai 90112, Thailand.,Faculty of Pharmaceutical Sciences, Govt. College University Faisalabad, Pakistan
| | - Pharkphoom Panichayupakaranant
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai 90112, Thailand.,Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai 90112, Thailand
| |
Collapse
|
7
|
Indraratna AD, Everest-Dass A, Skropeta D, Sanderson-Smith M. OUP accepted manuscript. FEMS Microbiol Rev 2022; 46:6519265. [PMID: 35104861 PMCID: PMC9075583 DOI: 10.1093/femsre/fuac001] [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: 07/26/2021] [Revised: 11/29/2021] [Accepted: 01/25/2022] [Indexed: 11/12/2022] Open
Abstract
Host carbohydrates, or glycans, have been implicated in the pathogenesis of many bacterial infections. Group A Streptococcus (GAS) is a Gram-positive bacterium that readily colonises the skin and oropharynx, and is a significant cause of mortality in humans. While the glycointeractions orchestrated by many other pathogens are increasingly well-described, the understanding of the role of human glycans in GAS disease remains incomplete. Although basic investigation into the mechanisms of GAS disease is ongoing, several glycointeractions have been identified and are examined herein. The majority of research in this context has focussed on bacterial adherence, however, glycointeractions have also been implicated in carbohydrate metabolism; evasion of host immunity; biofilm adaptations; and toxin-mediated haemolysis. The involvement of human glycans in these diverse avenues of pathogenesis highlights the clinical value of understanding glycointeractions in combatting GAS disease.
Collapse
Affiliation(s)
- Anuk D Indraratna
- Illawarra Health and Medical Research Institute, Northfields Ave, Keiraville New South Wales 2522, Australia
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia
| | - Arun Everest-Dass
- Institute for Glycomics, Griffith University, Gold Coast Campus, Parklands Drive, Southport, Queensland, 4215, Australia
| | - Danielle Skropeta
- Illawarra Health and Medical Research Institute, Northfields Ave, Keiraville New South Wales 2522, Australia
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia
| | - Martina Sanderson-Smith
- Corresponding author: Illawarra Health and Medical Research Institute, Bld 32, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia. Tel: +61 2 42981935; E-mail:
| |
Collapse
|
8
|
Hammond AJ, Binsker U, Aggarwal SD, Ortigoza MB, Loomis C, Weiser JN. Neuraminidase B controls neuraminidase A-dependent mucus production and evasion. PLoS Pathog 2021; 17:e1009158. [PMID: 33819312 PMCID: PMC8049478 DOI: 10.1371/journal.ppat.1009158] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/15/2021] [Accepted: 03/01/2021] [Indexed: 11/20/2022] Open
Abstract
Binding of Streptococcus pneumoniae (Spn) to nasal mucus leads to entrapment and clearance via mucociliary activity during colonization. To identify Spn factors allowing for evasion of mucus binding, we used a solid-phase adherence assay with immobilized mucus of human and murine origin. Spn bound large mucus particles through interactions with carbohydrate moieties. Mutants lacking neuraminidase A (nanA) or neuraminidase B (nanB) showed increased mucus binding that correlated with diminished removal of terminal sialic acid residues on bound mucus. The non-additive activity of the two enzymes raised the question why Spn expresses two neuraminidases and suggested they function in the same pathway. Transcriptional analysis demonstrated expression of nanA depends on the enzymatic function of NanB. As transcription of nanA is increased in the presence of sialic acid, our findings suggest that sialic acid liberated from host glycoconjugates by the secreted enzyme NanB induces the expression of the cell-associated enzyme NanA. The absence of detectable mucus desialylation in the nanA mutant, in which NanB is still expressed, suggests that NanA is responsible for the bulk of the modification of host glycoconjugates. Thus, our studies describe a functional role for NanB in sialic acid sensing in the host. The contribution of the neuraminidases in vivo was then assessed in a murine model of colonization. Although mucus-binding mutants showed an early advantage, this was only observed in a competitive infection, suggesting a complex role of neuraminidases. Histologic examination of the upper respiratory tract demonstrated that Spn stimulates mucus production in a neuraminidase-dependent manner. Thus, an increase production of mucus containing secretions appears to be balanced, in vivo, by decreased mucus binding. We postulate that through the combined activity of its neuraminidases, Spn evades mucus binding and mucociliary clearance, which is needed to counter neuraminidase-mediated stimulation of mucus secretions.
Collapse
Affiliation(s)
- Alexandria J. Hammond
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Ulrike Binsker
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Surya D. Aggarwal
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Mila Brum Ortigoza
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, New York University School of Medicine, New York, New York, United States of America
| | - Cynthia Loomis
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Jeffrey N. Weiser
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| |
Collapse
|
9
|
Sanchez-Rosario Y, Johnson MDL. Media Matters, Examining Historical and Modern Streptococcus pneumoniae Growth Media and the Experiments They Affect. Front Cell Infect Microbiol 2021; 11:613623. [PMID: 33834003 PMCID: PMC8021847 DOI: 10.3389/fcimb.2021.613623] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
While some bacteria can thrive for generations in minerals and salts, many require lavish nutrition and specific chemicals to survive to the point where they can be observed and researched. Although researchers once boiled and rendered animal flesh and bones to obtain a media that facilitated bacterial growth, we now have a plethora of formulations and manufacturers to provide dehydrated flavors of historical, modified, and modern media. The purpose of media has evolved from simple isolation to more measured study. However, in some instances, media formulated to aid the metabolic, nutritional, or physical properties of microbes may not be best suited for studying pathogen behavior or resilience as a function of host interactions. While there have been comparative studies on handfuls of these media in Streptococcus pneumoniae, this review focuses on describing both the historical and modern composition of common complex (Todd Hewitt and M17), semi-defined (Adams and Roe), and defined pneumococcal media (RPMI and Van de Rijn and Kessler), key components discovered/needed for cultivation/growth enhancement, and effects these different media have on bacterial phenotypes and experimental outcomes. While many researchers find the best conditions to grow and experiment on their bacteria of choice, the reasons for some researchers to use a specific medium is at best, not discussed, and at worst, arbitrary. As such, the goal of this review is to highlight the differences in pneumococcal media to encourage investigators to challenge their decisions on why they use a given medium, discuss the recipe, and explain their reasoning.
Collapse
Affiliation(s)
| | - Michael D L Johnson
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States.,BIO5 Institute, University of Arizona, Tucson, AZ, United States.,Valley Fever Center for Excellence, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
10
|
Hansen AL, Reily C, Novak J, Renfrow MB. Immunoglobulin A Glycosylation and Its Role in Disease. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:433-477. [PMID: 34687019 DOI: 10.1007/978-3-030-76912-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Human IgA is comprised of two subclasses, IgA1 and IgA2. Monomeric IgA (mIgA), polymeric IgA (pIgA), and secretory IgA (SIgA) are the main molecular forms of IgA. The production of IgA rivals all other immunoglobulin isotypes. The large quantities of IgA reflect the fundamental roles it plays in immune defense, protecting vulnerable mucosal surfaces against invading pathogens. SIgA dominates mucosal surfaces, whereas IgA in circulation is predominately monomeric. All forms of IgA are glycosylated, and the glycans significantly influence its various roles, including antigen binding and the antibody effector functions, mediated by the Fab and Fc portions, respectively. In contrast to its protective role, the aberrant glycosylation of IgA1 has been implicated in the pathogenesis of autoimmune diseases, such as IgA nephropathy (IgAN) and IgA vasculitis with nephritis (IgAVN). Furthermore, detailed characterization of IgA glycosylation, including its diverse range of heterogeneity, is of emerging interest. We provide an overview of the glycosylation observed for each subclass and molecular form of IgA as well as the range of heterogeneity for each site of glycosylation. In many ways, the role of IgA glycosylation is in its early stages of being elucidated. This chapter provides an overview of the current knowledge and research directions.
Collapse
Affiliation(s)
- Alyssa L Hansen
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Colin Reily
- Departments of Medicine and Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Matthew B Renfrow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
11
|
Sharapova Y, Švedas V, Suplatov D. Catalytic and lectin domains in neuraminidase A from Streptococcus pneumoniae are capable of an intermolecular assembly: Implications for biofilm formation. FEBS J 2020; 288:3217-3230. [PMID: 33108702 DOI: 10.1111/febs.15610] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 01/14/2023]
Abstract
Neuraminidase A from Streptococcus pneumoniae (NanA) is a cell wall-bound modular enzyme containing one lectin and one catalytic domain. Unlike homologous NanB and NanC expressed by the same bacterium, the two domains within one NanA molecule do not form a stable interaction and are spatially separated by a 16-amino acid-long flexible linker. In this work, the ability of NanA to form intermolecular assemblies was characterized using the methods of molecular modeling and bioinformatic analysis based on crystallographic data and by bringing together previously published experimental data. It was concluded that two catalytic domains, as well as one catalytic and one lectin domain, originating from two cell wall-bound NanA molecules, can interact through a previously uncharacterized interdomain interface to form complexes stabilized by a network of intermolecular hydrogen bonds and salt bridges. Supercomputer modeling strongly indicated that artocarpin, an earlier experimentally discovered inhibitor of the pneumococcal biofilm formation, is able to bind to a site located in the catalytic domain of one NanA entity and prevent its interaction with the lectin or catalytic domain of another NanA entity, thus directly precluding the generation of intermolecular assemblies. The revealed structural adaptation is discussed as one plausible mechanism of noncatalytic participation of this potentially key pathogenicity enzyme in pneumococcal biofilm formation.
Collapse
Affiliation(s)
- Yana Sharapova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vytas Švedas
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry Suplatov
- Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
12
|
Biharee A, Sharma A, Kumar A, Jaitak V. Antimicrobial flavonoids as a potential substitute for overcoming antimicrobial resistance. Fitoterapia 2020; 146:104720. [PMID: 32910994 DOI: 10.1016/j.fitote.2020.104720] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Infectious diseases are the leading cause of death in 21st century due to antimicrobial resistance and scarcity of new molecules to undertake rising infections. There could be a multiple reasons behind antimicrobial resistance whether it is increased drug metabolism or bacterial endotoxins. The demand of effective medication is increasing day by day to treat microbial infections and combat antimicrobial resistance. In recent years most of the synthetic antimicrobials developed resistance so natural products could provide better options to fulfill this demand. There has been increasing interest in the research on flavonoids because various flavonoids were found to be effective against pathogenic microorganisms. OBJECTIVE The objective of this article will be to explore antimicrobial activity of flavonoids with special focus on their possible mechanism of action. METHODS The article reviewed recent literature related to flavonoids with antimicrobial activity, which were isolated from various sources and the compounds showing fairly good activity against tested microbial species were discussed. RESULTS By throughout literature review it has been found that flavonoids show antimicrobial effect by inhibiting virulence factors, efflux pump, biofilm formation, membrane disruption, cell envelop synthesis, nucleic acid synthesis, and bacterial motility inhibition. CONCLUSION Most of the antimicrobial drugs available now a days are ineffective due to development of resistance to them. Flavonoids have the potential to overcome this emerging crisis as this class of natural products showed the antimicrobial activity by different mechanisms than those of conventional drugs, so flavonoid could be an effective treatment of pathogenic infections.
Collapse
Affiliation(s)
- Avadh Biharee
- Laboratory of Natural Products, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151001, India
| | - Aditi Sharma
- Laboratory of Natural Products, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151001, India
| | - Amit Kumar
- Laboratory of Natural Products, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151001, India
| | - Vikas Jaitak
- Laboratory of Natural Products, Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab 151001, India..
| |
Collapse
|
13
|
Epidemiological and genetic characterization of Clostridium butyricum cultured from neonatal cases of necrotizing enterocolitis in China. Infect Control Hosp Epidemiol 2020; 41:900-907. [PMID: 32539870 PMCID: PMC7511950 DOI: 10.1017/ice.2019.289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective: Laboratory-based characterization and traceback of Clostridium butyricum isolates linked to outbreak cases of neonatal necrotizing enterocolitis (NEC) in a hospital in China. Methods: In total, 37 samples were collected during the NEC outbreak. Classical bacteriological methods were applied to isolate and identify Clostridium spp. Meanwhile, 24 samples collected after an outbreak were similarly tested. All Clostridium isolates were identified to species level as either C. butyricum or C. sporogenes. These isolates were subsequently subtyped using pulsed-field gel electrophoresis (PFGE). Genomic DNA was purified from 2 representative C. butyricum isolates and sequenced to completion. Results: Of 37 samples collected during the NEC outbreak, 17 (45.95%) were positive for Clostridium spp. One species, C. butyricum, was cultured from 10 samples. Another species cultured from 2 other samples was identified as C. sporogenes. Both of these species were cocultured from 5 samples. Pulsotyping showed that the 15 C. butyricum and the 7 C. sporogenes isolates produced indistinguishable DNA profiles. No NEC cases were reported after disinfection following the outbreak, and all samples collected after the outbreak were negative for Clostridium spp. Whole-genome sequencing (WGS) indicated that sialidase, hemolysin, and enterotoxin virulence factors were located on the chromosomes of 2 C. butyricum isolates. Conclusions: The outbreak of NEC was epidemiologically linked to C. butyricum contamination within the hospital. This is the first report of an NEC outbreak associated with C. butyricum infection in China.
Collapse
|
14
|
Liang J, Mantelos A, Toh ZQ, Tortorella SM, Ververis K, Vongsvivut J, Bambery KR, Licciardi PV, Hung A, Karagiannis TC. Investigation of potential anti-pneumococcal effects of l-sulforaphane and metabolites: Insights from synchrotron-FTIR microspectroscopy and molecular docking studies. J Mol Graph Model 2020; 97:107568. [PMID: 32097886 DOI: 10.1016/j.jmgm.2020.107568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/10/2020] [Indexed: 01/06/2023]
Abstract
Streptococcus pneumoniae infection can lead to pneumococcal disease, a major cause of mortality in children under the age of five years. In low- and middle-income country settings where pneumococcal disease burden is high, vaccine use is low and widespread antibiotic use has led to increased rates of multi-drug resistant pneumococci. l-sulforaphane (LSF), derived from broccoli and other cruciferous vegetables, has established anti-inflammatory, antioxidant, and anti-microbial properties. Hence, we sought to investigate the potential role of LSF against pneumococcal infection. Using a combination of in vitro and computational methods, the results showed that LSF and relevant metabolites had a potential to reduce pneumococcal adherence through modulation of host receptors, regulation of inflammation, or through direct modification of bacterial factors. Treatment with LSF and metabolites reduced pneumococcal adherence to respiratory epithelial cells. Synchrotron-Fourier transform infrared microspectroscopy (S-FTIR) revealed biochemical changes in protein and lipid profiles of lung epithelial cells following treatment with LSF or metabolites. Molecular docking studies of 116 pneumococcal and 89 host factors revealed a potent effect for the metabolite LSF-glutathione (GSH). A comprehensive list of factors involved in interactions between S. pneumoniae and host cells was compiled to construct a bacterium and host interaction network. Network analysis revealed plasminogen, fibronectin, and RrgA as key factors involved in pneumococcal-host interactions. Therefore, we propose that these constitute critical targets for direct inhibition by LSF and/or metabolites, which may disrupt pneumococcal-host adherence. Overall, our findings further enhance understanding of the potential role of LSF to modulate pneumococcal-host dynamics.
Collapse
Affiliation(s)
- Julia Liang
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia; School of Science, RMIT University, VIC, 3001, Australia
| | - Anita Mantelos
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia; Murdoch Children's Research Institute, Melbourne, Parkville, VIC, 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Zheng Quan Toh
- Murdoch Children's Research Institute, Melbourne, Parkville, VIC, 3052, Australia
| | - Stephanie M Tortorella
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Katherine Ververis
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | | | - Keith R Bambery
- ANSTO Australian Synchrotron, 800 Blackburn Road, Clayton, VIC, 3168, Australia
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Melbourne, Parkville, VIC, 3052, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Andrew Hung
- School of Science, RMIT University, VIC, 3001, Australia
| | - Tom C Karagiannis
- Epigenomic Medicine, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
15
|
Wang YH. Sialidases From Clostridium perfringens and Their Inhibitors. Front Cell Infect Microbiol 2020; 9:462. [PMID: 31998664 PMCID: PMC6966327 DOI: 10.3389/fcimb.2019.00462] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/16/2019] [Indexed: 12/27/2022] Open
Abstract
Clostridium perfringens is an important human and animal pathogen that is the primary causative agent of necrotizing enteritis and enterotoxemia in many types of animals; it causes traumatic gas gangrene in humans and animals and is associated with cases of food poisoning in humans. C. perfringens produces a variety of toxins as well as many enzymes, including three sialidases, NanH, NanI, and NanJ. Sialidases could be important virulence factors that promote the pathogenesis of C. perfringens. Among them, NanI promotes the colonization of C. perfringens in the intestinal tract and enhances the cytotoxic activity and association of several major C. perfringens toxins with host cells. In recent years, studies on the structure and functions of sialidases have yielded interesting results, and the functions of sialic acid and sialidases in bacterial pathogenesis have become a hot research topic. An in-depth understanding and additional studies of sialidases will further elucidate mechanisms of C. perfringens pathogenesis and could promote the development and clinical applications of sialidase inhibitors. This article reviews the structural characteristics, expression regulation, roles of sialidases in C. perfringens pathogenesis, and effects of their inhibitors.
Collapse
Affiliation(s)
- Yan-Hua Wang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
16
|
Brissac T, Orihuela CJ. In Vitro Adhesion, Invasion, and Transcytosis of Streptococcus pneumoniae with Host Cells. Methods Mol Biol 2019; 1968:137-146. [PMID: 30929212 DOI: 10.1007/978-1-4939-9199-0_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Physical interactions of bacteria with host cells are often a principal aspect of bacterial pathogenesis. In the case of Streptococcus pneumoniae (Spn), which does not produce a secreted toxin, adhesion to and/or invasion of host cells is necessary for colonization of the nasopharynx and subsequently to cause opportunistic disease in its human host. Knowledge of how pneumococci interact with host cells thereby helps to explain its biology and may identify potential targets for intervention. One of the simplest, yet powerful, assays that can be leveraged to dissect the molecular basis of this vital host-pathogen interaction is the in vitro adhesion and invasion assay. Among many key results, this assay has been used to discover the bacterial and host determinants involved in bacterial attachment, identify host signaling networks required for uptake of the bacteria into an endosome, and the characterization of the intracellular trafficking machinery that is subverted by Spn during development of bacteremia and meningitis. These assays have also been used to characterize the epithelial, endothelial, and/or immune cell response to these bacteria, and to learn how pneumococci disperse from an established biofilm to a planktonic phenotype to colonize another niche and/or transmit. Herein, we will review this protocol, highlighting how simple changes in the bacterial strain or host cell line can elucidate the underlying molecular mechanisms for Spn virulence.
Collapse
Affiliation(s)
- Terry Brissac
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carlos J Orihuela
- Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
17
|
Janesch P, Rouha H, Badarau A, Stulik L, Mirkina I, Caccamo M, Havlicek K, Maierhofer B, Weber S, Groß K, Steinhäuser J, Zerbs M, Varga C, Dolezilkova I, Maier S, Zauner G, Nielson N, Power CA, Nagy E. Assessing the function of pneumococcal neuraminidases NanA, NanB and NanC in in vitro and in vivo lung infection models using monoclonal antibodies. Virulence 2019; 9:1521-1538. [PMID: 30289054 PMCID: PMC6177239 DOI: 10.1080/21505594.2018.1520545] [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] [Indexed: 02/03/2023] Open
Abstract
Streptococcus pneumoniae isolates express up to three neuraminidases (sialidases), NanA, NanB and NanC, all of which cleave the terminal sialic acid of glycan-structures that decorate host cell surfaces. Most research has focused on the role of NanA with limited investigations evaluating the roles of all three neuraminidases in host-pathogen interactions. We generated two highly potent monoclonal antibodies (mAbs), one that blocks the enzymatic activity of NanA and one cross-neutralizing NanB and NanC. Total neuraminidase activity of clinical S. pneumoniae isolates could be inhibited by this mAb combination in enzymatic assays. To detect desialylation of cell surfaces by pneumococcal neuraminidases, primary human tracheal/bronchial mucocilial epithelial tissues were infected with S. pneumoniae and stained with peanut lectin. Simultaneous targeting of the neuraminidases was required to prevent desialylation, suggesting that inhibition of NanA alone is not sufficient to preserve terminal lung glycans. Importantly, we also found that all three neuraminidases increased the interaction of S. pneumoniae with human airway epithelial cells. Lectin-staining of lung tissues of mice pre-treated with mAbs before intranasal challenge with S. pneumoniae confirmed that both anti-NanA and anti-NanBC mAbs were required to effectively block desialylation of the respiratory epithelium in vivo. Despite this, no effect on survival, reduction in pulmonary bacterial load, or significant changes in cytokine responses were observed. This suggests that neuraminidases have no pivotal role in this murine pneumonia model that is induced by high bacterial challenge inocula and does not progress from colonization as it happens in the human host.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Karin Groß
- a Arsanis Biosciences , Vienna , Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Robinson LS, Schwebke J, Lewis WG, Lewis AL. Identification and characterization of NanH2 and NanH3, enzymes responsible for sialidase activity in the vaginal bacterium Gardnerella vaginalis. J Biol Chem 2019; 294:5230-5245. [PMID: 30723162 DOI: 10.1074/jbc.ra118.006221] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/29/2019] [Indexed: 11/06/2022] Open
Abstract
Gardnerella vaginalis is abundant in bacterial vaginosis (BV), a condition associated with adverse reproductive health. Sialidase activity is a diagnostic feature of BV and is produced by a subset of G. vaginalis strains. Although its genetic basis has not been formally identified, sialidase activity is presumed to derive from the sialidase A gene, named here nanH1 In this study, BLAST searches predicted two additional G. vaginalis sialidases, NanH2 and NanH3. When expressed in Escherichia coli, NanH2 and NanH3 both displayed broad abilities to cleave sialic acids from α2-3- and α2-6-linked N- and O-linked sialoglycans, including relevant mucosal substrates. In contrast, recombinant NanH1 had limited activity against synthetic and mucosal substrates under the conditions tested. Recombinant NanH2 was much more effective than NanH3 in cleaving sialic acids bearing a 9-O-acetyl ester. Similarly, G. vaginalis strains encoding NanH2 cleaved and foraged significantly more Neu5,9Ac2 than strains encoding only NanH3. Among a collection of 34 G. vaginalis isolates, nanH2, nanH3, or both were present in all 15 sialidase-positive strains but absent from all 19 sialidase-negative isolates, including 16 strains that were nanH1-positive. We conclude that NanH2 and NanH3 are the primary sources of sialidase activity in G. vaginalis and that these two enzymes can account for the previously described substrate breadth cleaved by sialidases in human vaginal specimens of women with BV. Finally, PCRs of nanH2 or nanH3 from human vaginal specimens had 81% sensitivity and 78% specificity in distinguishing between Lactobacillus dominance and BV, as determined by Nugent scoring.
Collapse
Affiliation(s)
- Lloyd S Robinson
- From the Departments of Molecular Microbiology and.,Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri 63110 and
| | - Jane Schwebke
- the Division of Infectious Diseases, University of Alabama, Birmingham, Alabama 35294
| | - Warren G Lewis
- From the Departments of Molecular Microbiology and.,Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri 63110 and
| | - Amanda L Lewis
- From the Departments of Molecular Microbiology and .,Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri 63110 and.,Obstetrics and Gynecology and
| |
Collapse
|
19
|
NanI Sialidase Is an Important Contributor to Clostridium perfringens Type F Strain F4969 Intestinal Colonization in Mice. Infect Immun 2018; 86:IAI.00462-18. [PMID: 30297524 DOI: 10.1128/iai.00462-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/28/2018] [Indexed: 12/19/2022] Open
Abstract
Clostridium perfringens type F (formerly enterotoxigenic C. perfringens type A) strains produce an enterotoxin (CPE) to cause acute cases of food poisoning and chronic nonfoodborne human gastrointestinal diseases (NFD), e.g., antibiotic-associated diarrhea (AAD). NFD strains also produce NanI sialidase, an extracellular enzyme that releases sialic acids from sialyated host macromolecules. Recent in vitro studies suggested that NanI may contribute to NFD strain intestinal colonization by enhancing the adherence of such strains to intestinal cells and promoting their bacterial growth using generated sialic acid as an energy source. The current study tested this hypothesis by developing a mouse intestinal colonization model involving clindamycin pretreatment to produce conditions mimicking those during AAD. In this model, the type F NFD strain F4969 persisted for at least 4 days in the small intestine, cecum, and colon. When clindamycin-pretreated mice were challenged by oral gavage with equivalent numbers of F4969 bacteria or its isogenic nanI null mutant, significantly lower numbers of the nanI mutant were recovered from all intestinal segments, and it was completely cleared from the small intestine by day 4. Complementation of the mutant to restore NanI production also promoted colonization. When the same nanI null mutant strain was coinoculated into the mouse model together with a nanI-producing strain, the numbers of this mutant were restored to wild-type F4969 levels in all intestinal segments. This result suggests that sialidases produced by other bacteria might also provide some support for C. perfringens intestinal colonization. Collectively, these in vivo findings identify NanI to be the first known significant contributor to chronic intestinal colonization by NFD strains.
Collapse
|
20
|
A recombinant conjugated pneumococcal vaccine that protects against murine infections with a similar efficacy to Prevnar-13. NPJ Vaccines 2018; 3:53. [PMID: 30393571 PMCID: PMC6208403 DOI: 10.1038/s41541-018-0090-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/08/2018] [Indexed: 11/09/2022] Open
Abstract
The pneumococcal conjugate vaccine (PCV) strongly protects against vaccine serotypes, but the rapid expansion of non-vaccine serotype disease and the vaccine's high expense has reduced its overall impact. We have developed Protein Glycan Coupling Technology (PGCT) as a flexible methodology for making low-cost polysaccharide/protein glycoconjugates recombinantly in Escherichia coli. We have used PGCT to make a recombinant PCV containing serotype 4 capsular polysaccharide linked to the Streptococcus pneumoniae proteins NanA, PiuA, and Sp0148. The introduction of the Campylobacter jejuni UDP-glucose 4-epimerase gene GalE (gne) into E. coli improved the yield of the resulting glycoprotein. PGCT glycoconjugate vaccination generated strong antibody responses in mice to both the capsule and the carrier protein antigens, with the PiuA/capsule glycoconjugate inducing similar anti-capsular antibody responses as the commercial PCV Prevnar-13. Antibody responses to PGCT glycoconjugates opsonised S. pneumoniae and Streptococcus mitis expressing the serotype 4 capsule and promoted neutrophil phagocytosis of S. pneumoniae to a similar level as antisera generated by vaccination with Prevnar-13. Vaccination with the PGCT glycoconjugates protected mice against meningitis and septicaemia with the same efficacy as vaccination with Prevnar-13. In addition, vaccination with the protein antigen components from PGCT glycoconjugates alone provided partial protection against septicaemia and colonisation. These data demonstrate that a vaccine made by PGCT is as effective as Prevnar-13, identifies PiuA as a carrier protein for glycoconjugate vaccines, and demonstrates that linking capsular antigen to S. pneumoniae protein antigens has additional protective benefits that could provide a degree of serotype-independent immunity.
Collapse
|
21
|
Desialylation of Platelets by Pneumococcal Neuraminidase A Induces ADP-Dependent Platelet Hyperreactivity. Infect Immun 2018; 86:IAI.00213-18. [PMID: 30037798 DOI: 10.1128/iai.00213-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/13/2018] [Indexed: 11/20/2022] Open
Abstract
Platelets are increasingly recognized to play a role in the complications of Streptococcus pneumoniae infections. S. pneumoniae expresses neuraminidases, which may alter glycans on the platelet surface. In the present study, we investigated the capability of pneumococcal neuraminidase A (NanA) to remove sialic acid (desialylation) from the platelet surface, the consequences for the platelet activation status and reactivity, and the ability of neuraminidase inhibitors to prevent these effects. Our results show that soluble NanA induces platelet desialylation. Whereas desialylation itself did not induce platelet activation (P-selectin expression and platelet fibrinogen binding), platelets became hyperreactive to ex vivo stimulation by ADP and cross-linked collagen-related peptide (CRP-XL). Platelet aggregation with leukocytes also increased. These processes were dependent on the ADP pathway, as inhibitors of the pathway (apyrase and ticagrelor) abrogated platelet hyperreactivity. Inhibition of NanA-induced platelet desialylation by neuraminidase inhibitors (e.g., oseltamivir acid) also prevented the platelet effects of NanA. Collectively, our findings show that soluble NanA can desialylate platelets, leading to platelet hyperreactivity, which can be prevented by neuraminidase inhibitors.
Collapse
|
22
|
Janapatla RP, Chen CL, Hsu MH, Liao WT, Chiu CH. Immunization with pneumococcal neuraminidases NanA, NanB and NanC to generate neutralizing antibodies and to increase survival in mice. J Med Microbiol 2018; 67:709-723. [PMID: 29557769 DOI: 10.1099/jmm.0.000724] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose. Pneumococcal virulence protein-based vaccines can provide serotype-independent protection against pneumococcal infections. Many studies, including clinical observational studies on Thomsen-Friedenrich antigen exposure and haemolytic uremic syndrome, defined the role of neuraminidases NanA, NanB and NanC in host-pneumococcus interaction. Since neuraminidases are major virulence proteins, they are potential targets for both vaccines and small molecule inhibitors. Here we explored the utility of three neuraminidases as protein vaccine antigens to generate neutralizing antibodies and to increase survival following pneumococcal infections.Methodology. Rabbits and mice were immunized subcutaneously with enzymatically active recombinant NanA, NanB and NanC as individual or a combination of the three neuraminidases. Antisera titres were determined by ELISA. Neuraminidase activity inhibition by antiserum was tested by peanut lectin and flow cytometry. Clinical isolates with serotype 3, 6B, 14, 15B, 19A and 23F were used to infect immunized mice by tail vein injection.Results/Key findings. Presence of high levels of IgG antibodies in antisera against NanA, NanB and NanC indicates that all of the three neuraminidases are immunogenic vaccine antigens. To generate potent NanA neutralizing antibodies, both lectin and catalytic domains are essential, whereas for NanB and NanC a single lectin domain is sufficient. Immunization with triple neuraminidases increased the survival of mice when intravenously challenged with clinical isolates of serotype 3 (40 %), 6B (60 %), 15B (60 %), 19A (40 %) and 23F (30 %).Conclusion. We recommend the inclusion of three pneumococcal neuraminidases in future protein vaccine formulations to prevent invasive pneumococcal infection caused by various serotypes.
Collapse
Affiliation(s)
| | - Chyi-Liang Chen
- Molecular Infectious Diseases Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Mei-Hua Hsu
- Molecular Infectious Diseases Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Wan-Ting Liao
- Molecular Infectious Diseases Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Cheng-Hsun Chiu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, ROC.,Molecular Infectious Diseases Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| |
Collapse
|
23
|
Hobbs JK, Pluvinage B, Boraston AB. Glycan-metabolizing enzymes in microbe-host interactions: the Streptococcus pneumoniae paradigm. FEBS Lett 2018; 592:3865-3897. [PMID: 29608212 DOI: 10.1002/1873-3468.13045] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/31/2022]
Abstract
Streptococcus pneumoniae is a frequent colonizer of the upper airways; however, it is also an accomplished pathogen capable of causing life-threatening diseases. To colonize and cause invasive disease, this bacterium relies on a complex array of factors to mediate the host-bacterium interaction. The respiratory tract is rich in functionally important glycoconjugates that display a vast range of glycans, and, thus, a key component of the pneumococcus-host interaction involves an arsenal of bacterial carbohydrate-active enzymes to depolymerize these glycans and carbohydrate transporters to import the products. Through the destruction of host glycans, the glycan-specific metabolic machinery deployed by S. pneumoniae plays a variety of roles in the host-pathogen interaction. Here, we review the processing and metabolism of the major host-derived glycans, including N- and O-linked glycans, Lewis and blood group antigens, proteoglycans, and glycogen, as well as some dietary glycans. We discuss the role of these metabolic pathways in the S. pneumoniae-host interaction, speculate on the potential of key enzymes within these pathways as therapeutic targets, and relate S. pneumoniae as a model system to glycan processing in other microbial pathogens.
Collapse
Affiliation(s)
- Joanne K Hobbs
- Department of Biochemistry and Microbiology, University of Victoria, British Columbia, Canada
| | - Benjamin Pluvinage
- Department of Biochemistry and Microbiology, University of Victoria, British Columbia, Canada
| | - Alisdair B Boraston
- Department of Biochemistry and Microbiology, University of Victoria, British Columbia, Canada
| |
Collapse
|
24
|
Gratz N, Loh LN, Mann B, Gao G, Carter R, Rosch J, Tuomanen EI. Pneumococcal neuraminidase activates TGF-β signalling. MICROBIOLOGY-SGM 2017; 163:1198-1207. [PMID: 28749326 PMCID: PMC5817201 DOI: 10.1099/mic.0.000511] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuraminidase A (NanA) is an important virulence factor that is anchored to the pneumococcal cell wall and cleaves sialic acid on host substrates. We noted that a secreted allele of NanA was over-represented in invasive pneumococcal isolates and promoted the development of meningitis when swapped into the genome of non-meningitis isolates replacing cell wall-anchored NanA. Both forms of recombinant NanA directly activated transforming growth factor (TGF)-β, increased SMAD signalling and promoted loss of endothelial tight junction ZO-1. However, in assays using whole bacteria, only the cell-bound NanA decreased expression of ZO-1 and showed NanA dependence of bacterial invasion of endothelial cells. We conclude that NanA secretion versus retention on the cell surface does not influence neurotropism of clinical isolates. However, we describe a new NanA-TGF-β signalling axis that leads to decreased blood-brain barrier integrity and enhances bacterial invasion.
Collapse
Affiliation(s)
- Nina Gratz
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Lip Nam Loh
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Beth Mann
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Geli Gao
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Robert Carter
- Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jason Rosch
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Elaine I. Tuomanen
- Departments of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- *Correspondence: Elaine I. Tuomanen,
| |
Collapse
|
25
|
Park JY, Hwan Lim S, Ram Kim B, Jae Jeong H, Kwon HJ, Song GY, Bae Ryu Y, Song Lee W. Sialidase inhibitory activity of diarylnonanoid and neolignan compounds extracted from the seeds of Myristica fragrans. Bioorg Med Chem Lett 2017; 27:3060-3064. [DOI: 10.1016/j.bmcl.2017.05.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/11/2017] [Accepted: 05/17/2017] [Indexed: 11/15/2022]
|
26
|
Song M, Teng Z, Li M, Niu X, Wang J, Deng X. Epigallocatechin gallate inhibits Streptococcus pneumoniae virulence by simultaneously targeting pneumolysin and sortase A. J Cell Mol Med 2017; 21:2586-2598. [PMID: 28402019 PMCID: PMC5618700 DOI: 10.1111/jcmm.13179] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/24/2017] [Indexed: 01/11/2023] Open
Abstract
Streptococcus pneumoniae (pneumococcus), the causative agent of several human diseases, possesses numerous virulence factors associated with pneumococcal infection and pathogenesis. Pneumolysin (PLY), an important virulence factor, is a member of the cholesterol-dependent cytolysin family and has cytolytic activity. Sortase A (SrtA), another crucial pneumococcal virulence determinate, contributes greatly to the anchoring of many virulence-associated surface proteins to the cell wall. In this study, epigallocatechin gallate (EGCG), a natural compound with little known antipneumococcal activity, was shown to directly inhibit PLY-mediated haemolysis and cytolysis by blocking the oligomerization of PLY and simultaneously reduce the peptidase activity of SrtA. The biofilm formation, production of neuraminidase A (NanA, the pneumococcal surface protein anchored by SrtA), and bacterial adhesion to human epithelial cells (Hep2) were inhibited effectively when S. pneumoniae D39 was cocultured with EGCG. The results from molecular dynamics simulations and mutational analysis confirmed the interaction of EGCG with PLY and SrtA, and EGCG binds to Glu277, Tyr358, and Arg359 in PLY and Thr169, Lys171, and Phe239 in SrtA. In vivo studies further demonstrated that EGCG protected mice against S. pneumoniae pneumonia. Our results imply that EGCG is an effective inhibitor of both PLY and SrtA and that an antivirulence strategy that directly targets PLY and SrtA using EGCG is a promising therapeutic option for S. pneumoniae pneumonia.
Collapse
Affiliation(s)
- Meng Song
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zihao Teng
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Meng Li
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaodi Niu
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China
| |
Collapse
|
27
|
Pneumococcal Neuraminidase A (NanA) Promotes Biofilm Formation and Synergizes with Influenza A Virus in Nasal Colonization and Middle Ear Infection. Infect Immun 2017; 85:IAI.01044-16. [PMID: 28096183 DOI: 10.1128/iai.01044-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/10/2017] [Indexed: 01/08/2023] Open
Abstract
Even in the vaccine era, Streptococcus pneumoniae (the pneumococcus) remains a leading cause of otitis media, a significant public health burden, in large part because of the high prevalence of nasal colonization with the pneumococcus in children. The primary pneumococcal neuraminidase, NanA, which is a sialidase that catalyzes the cleavage of terminal sialic acids from host glycoconjugates, is involved in both of these processes. Coinfection with influenza A virus, which also expresses a neuraminidase, exacerbates nasal colonization and disease by S. pneumoniae, in part via the synergistic contributions of the viral neuraminidase. The specific role of its pneumococcal counterpart, NanA, in this interaction, however, is less well understood. We demonstrate in a mouse model that NanA-deficient pneumococci are impaired in their ability to cause both nasal colonization and middle ear infection. Coinfection with neuraminidase-expressing influenza virus and S. pneumoniae potentiates both colonization and infection but not to wild-type levels, suggesting an intrinsic role of NanA. Using in vitro models, we show that while NanA contributes to both epithelial adherence and biofilm viability, its effect on the latter is actually independent of its sialidase activity. These data indicate that NanA contributes both enzymatically and nonenzymatically to pneumococcal pathogenesis and, as such, suggest that it is not a redundant bystander during coinfection with influenza A virus. Rather, its expression is required for the full synergism between these two pathogens.
Collapse
|
28
|
Deacetylation of sialic acid by esterases potentiates pneumococcal neuraminidase activity for mucin utilization, colonization and virulence. PLoS Pathog 2017; 13:e1006263. [PMID: 28257499 PMCID: PMC5352144 DOI: 10.1371/journal.ppat.1006263] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 03/15/2017] [Accepted: 02/27/2017] [Indexed: 01/19/2023] Open
Abstract
Pneumococcal neuraminidase is a key enzyme for sequential deglycosylation of host glycans, and plays an important role in host survival, colonization, and pathogenesis of infections caused by Streptococcus pneumoniae. One of the factors that can affect the activity of neuraminidase is the amount and position of acetylation present in its substrate sialic acid. We hypothesised that pneumococcal esterases potentiate neuraminidase activity by removing acetylation from sialic acid, and that will have a major effect on pneumococcal survival on mucin, colonization, and virulence. These hypotheses were tested using isogenic mutants and recombinant esterases in microbiological, biochemical and in vivo assays. We found that pneumococcal esterase activity is encoded by at least four genes, SPD_0534 (EstA) was found to be responsible for the main esterase activity, and the pneumococcal esterases are specific for short acyl chains. Assay of esterase activity by using natural substrates showed that both the Axe and EstA esterases could use acetylated xylan and Bovine Sub-maxillary Mucin (BSM), a highly acetylated substrate, but only EstA was active against tributyrin (triglyceride). Incubation of BSM with either Axe or EstA led to the acetate release in a time and concentration dependent manner, and pre-treatment of BSM with either enzyme increased sialic acid release on subsequent exposure to neuraminidase A. qRT-PCR results showed that the expression level of estA and axe increased when exposed to BSM and in respiratory tissues. Mutation of estA alone or in combination with nanA (codes for neuraminidase A), or the replacement of its putative serine active site to alanine, reduced the pneumococcal ability to utilise BSM as a sole carbon source, sialic acid release, colonization, and virulence in a mouse model of pneumococcal pneumonia.
Collapse
|
29
|
Andre GO, Converso TR, Politano WR, Ferraz LFC, Ribeiro ML, Leite LCC, Darrieux M. Role of Streptococcus pneumoniae Proteins in Evasion of Complement-Mediated Immunity. Front Microbiol 2017; 8:224. [PMID: 28265264 PMCID: PMC5316553 DOI: 10.3389/fmicb.2017.00224] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 01/31/2017] [Indexed: 12/14/2022] Open
Abstract
The complement system plays a central role in immune defense against Streptococcus pneumoniae. In order to evade complement attack, pneumococci have evolved a number of mechanisms that limit complement mediated opsonization and subsequent phagocytosis. This review focuses on the strategies employed by pneumococci to circumvent complement mediated immunity, both in vitro and in vivo. At last, since many of the proteins involved in interactions with complement components are vaccine candidates in different stages of validation, we explore the use of these antigens alone or in combination, as potential vaccine approaches that aim at elimination or drastic reduction in the ability of this bacterium to evade complement.
Collapse
Affiliation(s)
- Greiciely O Andre
- Laboratório de Biologia Celular e Molecular de Microrganismos, Universidade São Francisco Bragança Paulista, Brazil
| | - Thiago R Converso
- Centro de Biotecnologia, Instituto ButantanSão Paulo, Brazil; Programa de Pós-graduação Interunidades em Biotecnologia, Universidade de São PauloSão Paulo, Brazil
| | - Walter R Politano
- Laboratório de Biologia Celular e Molecular de Microrganismos, Universidade São Francisco Bragança Paulista, Brazil
| | - Lucio F C Ferraz
- Laboratório de Biologia Celular e Molecular de Microrganismos, Universidade São Francisco Bragança Paulista, Brazil
| | - Marcelo L Ribeiro
- Laboratório de Farmacologia, Universidade São Francisco Bragança Paulista, Brazil
| | | | - Michelle Darrieux
- Laboratório de Biologia Celular e Molecular de Microrganismos, Universidade São Francisco Bragança Paulista, Brazil
| |
Collapse
|
30
|
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
|
31
|
Li J, Uzal FA, McClane BA. Clostridium perfringens Sialidases: Potential Contributors to Intestinal Pathogenesis and Therapeutic Targets. Toxins (Basel) 2016; 8:E341. [PMID: 27869757 PMCID: PMC5127137 DOI: 10.3390/toxins8110341] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 11/10/2016] [Accepted: 11/13/2016] [Indexed: 02/06/2023] Open
Abstract
Clostridium perfringens is a major cause of histotoxic and intestinal infections of humans and other animals. This Gram-positive anaerobic bacterium can produce up to three sialidases named NanH, NanI, and NanJ. The role of sialidases in histotoxic infections, such as gas gangrene (clostridial myonecrosis), remains equivocal. However, recent in vitro studies suggest that NanI may contribute to intestinal virulence by upregulating production of some toxins associated with intestinal infection, increasing the binding and activity of some of those toxins, and enhancing adherence of C. perfringens to intestinal cells. Possible contributions of NanI to intestinal colonization are further supported by observations that the C. perfringens strains causing acute food poisoning in humans often lack the nanI gene, while other C. perfringens strains causing chronic intestinal infections in humans usually carry a nanI gene. Certain sialidase inhibitors have been shown to block NanI activity and reduce C. perfringens adherence to cultured enterocyte-like cells, opening the possibility that sialidase inhibitors could be useful therapeutics against C. perfringens intestinal infections. These initial in vitro observations should be tested for their in vivo significance using animal models of intestinal infections.
Collapse
Affiliation(s)
- Jihong Li
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Room 420, Bridgeside Point II Building, 450 Technology Drive, Pittsburgh, PA 15219, USA.
| | - Francisco A Uzal
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, CA 92408, USA.
| | - Bruce A McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Room 420, Bridgeside Point II Building, 450 Technology Drive, Pittsburgh, PA 15219, USA.
| |
Collapse
|
32
|
In vivo screen of genetically conserved Streptococcus pneumoniae proteins for protective immunogenicity. Vaccine 2016; 34:6292-6300. [PMID: 27816374 DOI: 10.1016/j.vaccine.2016.10.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/22/2016] [Accepted: 10/22/2016] [Indexed: 11/21/2022]
Abstract
We evaluated 52 different E. coli expressed pneumococcal proteins as immunogens in a BALB/c mouse model of S. pneumoniae lung infection. Proteins were selected based on genetic conservation across disease-causing serotypes and bioinformatic prediction of antibody binding to the target antigen. Seven proteins induced protective responses, in terms of reduced lung burdens of the serotype 3 pneumococci. Three of the protective proteins were histidine triad protein family members (PhtB, PhtD and PhtE). Four other proteins, all bearing LPXTG linkage domains, also had activity in this model (PrtA, NanA, PavB and Eng). PrtA, NanA and Eng were also protective in a CBA/N mouse model of lethal pneumococcal infection. Despite data inferring widespread genomic conservation, flow-cytometer based antisera binding studies confirmed variable levels of antigen expression across a panel of pneumococcal serotypes. Finally, BALB/c mice were immunized and intranasally challenged with a viulent serotype 8 strain, to help understand the breadth of protection. Those mouse studies reaffirmed the effectiveness of the histidine triad protein grouping and a single LPXTG protein, PrtA.
Collapse
|
33
|
Blanchette KA, Shenoy AT, Milner J, Gilley RP, McClure E, Hinojosa CA, Kumar N, Daugherty SC, Tallon LJ, Ott S, King SJ, Ferreira DM, Gordon SB, Tettelin H, Orihuela CJ. Neuraminidase A-Exposed Galactose Promotes Streptococcus pneumoniae Biofilm Formation during Colonization. Infect Immun 2016; 84:2922-32. [PMID: 27481242 PMCID: PMC5038079 DOI: 10.1128/iai.00277-16] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/21/2016] [Indexed: 01/26/2023] Open
Abstract
Streptococcus pneumoniae is an opportunistic pathogen that colonizes the nasopharynx. Herein we show that carbon availability is distinct between the nasopharynx and bloodstream of adult humans: glucose is absent from the nasopharynx, whereas galactose is abundant. We demonstrate that pneumococcal neuraminidase A (NanA), which cleaves terminal sialic acid residues from host glycoproteins, exposed galactose on the surface of septal epithelial cells, thereby increasing its availability during colonization. We observed that S. pneumoniae mutants deficient in NanA and β-galactosidase A (BgaA) failed to form biofilms in vivo despite normal biofilm-forming abilities in vitro Subsequently, we observed that glucose, sucrose, and fructose were inhibitory for biofilm formation, whereas galactose, lactose, and low concentrations of sialic acid were permissive. Together these findings suggested that the genes involved in biofilm formation were under some form of carbon catabolite repression (CCR), a regulatory network in which genes involved in the uptake and metabolism of less-preferred sugars are silenced during growth with preferred sugars. Supporting this notion, we observed that a mutant deficient in pyruvate oxidase, which converts pyruvate to acetyl-phosphate under non-CCR-inducing growth conditions, was unable to form biofilms. Subsequent comparative transcriptome sequencing (RNA-seq) analyses of planktonic and biofilm-grown pneumococci showed that metabolic pathways involving the conversion of pyruvate to acetyl-phosphate and subsequently leading to fatty acid biosynthesis were consistently upregulated during diverse biofilm growth conditions. We conclude that carbon availability in the nasopharynx impacts pneumococcal biofilm formation in vivo Additionally, biofilm formation involves metabolic pathways not previously appreciated to play an important role.
Collapse
Affiliation(s)
- Krystle A Blanchette
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Anukul T Shenoy
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeffrey Milner
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ryan P Gilley
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Erin McClure
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cecilia A Hinojosa
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Nikhil Kumar
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sean C Daugherty
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Luke J Tallon
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sandra Ott
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Samantha J King
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Daniela M Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Stephen B Gordon
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Carlos J Orihuela
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA Department of Microbiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
34
|
A Multi-Serotype Approach Clarifies the Catabolite Control Protein A Regulon in the Major Human Pathogen Group A Streptococcus. Sci Rep 2016; 6:32442. [PMID: 27580596 PMCID: PMC5007534 DOI: 10.1038/srep32442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/04/2016] [Indexed: 12/20/2022] Open
Abstract
Catabolite control protein A (CcpA) is a highly conserved, master regulator of carbon source utilization in gram-positive bacteria, but the CcpA regulon remains ill-defined. In this study we aimed to clarify the CcpA regulon by determining the impact of CcpA-inactivation on the virulence and transcriptome of three distinct serotypes of the major human pathogen Group A Streptococcus (GAS). CcpA-inactivation significantly decreased GAS virulence in a broad array of animal challenge models consistent with the idea that CcpA is critical to gram-positive bacterial pathogenesis. Via comparative transcriptomics, we established that the GAS CcpA core regulon is enriched for highly conserved CcpA binding motifs (i.e. cre sites). Conversely, strain-specific differences in the CcpA transcriptome seems to consist primarily of affected secondary networks. Refinement of cre site composition via analysis of the core regulon facilitated development of a modified cre consensus that shows promise for improved prediction of CcpA targets in other medically relevant gram-positive pathogens.
Collapse
|
35
|
Silva LN, Zimmer KR, Macedo AJ, Trentin DS. Plant Natural Products Targeting Bacterial Virulence Factors. Chem Rev 2016; 116:9162-236. [PMID: 27437994 DOI: 10.1021/acs.chemrev.6b00184] [Citation(s) in RCA: 279] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Decreased antimicrobial efficiency has become a global public health issue. The paucity of new antibacterial drugs is evident, and the arsenal against infectious diseases needs to be improved urgently. The selection of plants as a source of prototype compounds is appropriate, since plant species naturally produce a wide range of secondary metabolites that act as a chemical line of defense against microorganisms in the environment. Although traditional approaches to combat microbial infections remain effective, targeting microbial virulence rather than survival seems to be an exciting strategy, since the modulation of virulence factors might lead to a milder evolutionary pressure for the development of resistance. Additionally, anti-infective chemotherapies may be successfully achieved by combining antivirulence and conventional antimicrobials, extending the lifespan of these drugs. This review presents an updated discussion of natural compounds isolated from plants with chemically characterized structures and activity against the major bacterial virulence factors: quorum sensing, bacterial biofilms, bacterial motility, bacterial toxins, bacterial pigments, bacterial enzymes, and bacterial surfactants. Moreover, a critical analysis of the most promising virulence factors is presented, highlighting their potential as targets to attenuate bacterial virulence. The ongoing progress in the field of antivirulence therapy may therefore help to translate this promising concept into real intervention strategies in clinical areas.
Collapse
Affiliation(s)
- Laura Nunes Silva
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| | - Karine Rigon Zimmer
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre , Porto Alegre, Rio Grande do Sul 90050-170, Brazil
| | - Alexandre José Macedo
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil.,Instituto Nacional do Semiárido , Campina Grande, Paraı́ba 58429-970, Brazil
| | - Danielle Silva Trentin
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Rio Grande do Sul 91501-970, Brazil
| |
Collapse
|
36
|
Xu Z, von Grafenstein S, Walther E, Fuchs JE, Liedl KR, Sauerbrei A, Schmidtke M. Sequence diversity of NanA manifests in distinct enzyme kinetics and inhibitor susceptibility. Sci Rep 2016; 6:25169. [PMID: 27125351 PMCID: PMC4850393 DOI: 10.1038/srep25169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 04/11/2016] [Indexed: 01/20/2023] Open
Abstract
Streptococcus pneumoniae is the leading pathogen causing bacterial pneumonia and meningitis. Its surface-associated virulence factor neuraminidase A (NanA) promotes the bacterial colonization by removing the terminal sialyl residues from glycoconjugates on eukaryotic cell surface. The predominant role of NanA in the pathogenesis of pneumococci renders it an attractive target for therapeutic intervention. Despite the highly conserved activity of NanA, our alignment of the 11 NanAs revealed the evolutionary diversity of this enzyme. The amino acid substitutions we identified, particularly those in the lectin domain and in the insertion domain next to the catalytic centre triggered our special interest. We synthesised the representative NanAs and the mutagenized derivatives from E. coli for enzyme kinetics study and neuraminidase inhibitor susceptibility test. Via molecular docking we got a deeper insight into the differences between the two major variants of NanA and their influence on the ligand-target interactions. In addition, our molecular dynamics simulations revealed a prominent intrinsic flexibility of the linker between the active site and the insertion domain, which influences the inhibitor binding. Our findings for the first time associated the primary sequence diversity of NanA with the biochemical properties of the enzyme and with the inhibitory efficiency of neuraminidase inhibitors.
Collapse
Affiliation(s)
- Zhongli Xu
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Susanne von Grafenstein
- University of Innsbruck, Institute for General, Inorganic and Theoretical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Innrain 80/82, 6020 Innsbruck, Austria
| | - Elisabeth Walther
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Julian E Fuchs
- University of Innsbruck, Institute for General, Inorganic and Theoretical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Innrain 80/82, 6020 Innsbruck, Austria
| | - Klaus R Liedl
- University of Innsbruck, Institute for General, Inorganic and Theoretical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Innrain 80/82, 6020 Innsbruck, Austria
| | - Andreas Sauerbrei
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Michaela Schmidtke
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| |
Collapse
|
37
|
Vandevelde NM, Tulkens PM, Van Bambeke F. Modulating antibiotic activity towards respiratory bacterial pathogens by co-medications: a multi-target approach. Drug Discov Today 2016; 21:1114-29. [PMID: 27094105 DOI: 10.1016/j.drudis.2016.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/17/2016] [Accepted: 04/05/2016] [Indexed: 01/01/2023]
Abstract
Non-antibiotic drugs can modulate bacterial physiology and/or antibiotic activity, opening perspectives for innovative therapeutic strategies. Focusing on respiratory pathogens and considering in vitro, in vivo, and clinical data, here we examine the effect of these drugs on the expression of resistance mechanisms, biofilm formation, and intracellular survival, as well as their influence on the activity of antibiotics on bacteria. Beyond the description of the effects observed, we also comment on concentrations that are active and discuss the mechanisms of drug-drug or drug-target interactions. This discussion should be helpful in defining useful targets for adjuvant therapy and establishing the corresponding pharmacophores for further drug fine-tuning.
Collapse
Affiliation(s)
- Nathalie M Vandevelde
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Paul M Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
38
|
Abstract
Sialic acids, or the more broad term nonulosonic acids, comprise a family of nine-carbon keto-sugars ubiquitous on mammalian mucous membranes as terminal modifications of mucin glycoproteins. Sialic acids have a limited distribution among bacteria, and the ability to catabolize sialic acids is mainly confined to pathogenic and commensal species. This ability to utilize sialic acid as a carbon source is correlated with bacterial virulence, especially, in the sialic acid rich environment of the oral cavity, respiratory, intestinal, and urogenital tracts. This chapter discusses the distribution of sialic acid catabolizers among the sequenced bacterial genomes and examines the studies that have linked sialic acid catabolism with increased in vivo fitness in a number of species using several animal models. This chapter presents the most recent findings in sialobiology with a focus on sialic acid catabolism, which demonstrates an important relationship between the catabolism of sialic acid and bacterial pathogenesis.
Collapse
|
39
|
Janapatla RP, Hsu MH, Liao WT, Chien KY, Lee HY, Chiu CH. Low Serum Fetuin-A as a Biomarker to Predict Pneumococcal Necrotizing Pneumonia and Hemolytic Uremic Syndrome in Children. Medicine (Baltimore) 2016; 95:e3221. [PMID: 27043691 PMCID: PMC4998552 DOI: 10.1097/md.0000000000003221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Streptococcus pneumoniae, a neuraminidase-producing pathogen, can cause invasive pneumococcal disease (IPD) with or without hemolytic uremic syndrome (HUS) in humans. We aimed to identify serum sialoglycoproteins that are targeted by neuraminidases in severe pneumococcal infection. We hypothesized that serum sialoglycoprotein such as fetuin-A can serve as a biomarker to predict IPD or HUS. We constructed serum sialoglycoprotein profiles before and after pneumococcal neuraminidase treatment using liquid chromatography-tandem mass spectrometry (LC-MS/MS), a proteomic approach. An observational study was conducted using clinical data and serum samples from pediatric patients with pneumococcal infection to verify the predictive role of fetuin-A in IPD. Serum fetuin-A levels were determined by enzyme-linked immunosorbent assay. The most abundant serum sialoglycoproteins identified by LC-MS/MS after neuraminidase treatment and peanut lectin capture were immunoglobulins, apolipoproteins, fibrinogens, keratins, complement system proteins, and fetuin-A. Serum fetuin-A levels in the HUS patients were significantly lower (207 ± 80 mg/L, P < 0.001) than in patients with lobar pneumonia (610 ± 190 mg/L) as well as the healthy controls (630 ± 250 mg/L). In comparing HUS with necrotizing pneumonia and lobar pneumonia, the ROC area under the curve was 0.842; a cutoff value of 298 mg/L yielded sensitivity of 92.9% (95% CI: 68.5-98.7%) and specificity of 71.9% (95% CI: 54.6-84.4%). This observational study with validation cohorts of patients with HUS, complicated pneumonia, and lobar pneumonia demonstrates the high performance of low serum fetuin-A levels as a biomarker to predict severe IPD and HUS in children.
Collapse
Affiliation(s)
- Rajendra Prasad Janapatla
- From the Molecular Infectious Disease Research Center (RPJ, MHH, W-TL, H-YL, C-HC), Chang Gung Memorial Hospital; Graduate Institute of Biomedical Sciences (K-YC, C-HC); and Division of Pediatric Infectious Diseases (H-YL, C-HC), Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
40
|
Bittaye M, Cash P. Streptococcus pneumoniae proteomics: determinants of pathogenesis and vaccine development. Expert Rev Proteomics 2015; 12:607-21. [PMID: 26524107 DOI: 10.1586/14789450.2015.1108844] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Streptococcus pneumoniae is a major pathogen that is responsible for a variety of invasive diseases. The bacteria gain entry initially by establishing a carriage state in the nasopharynx from where they migrate to other sites in the body. The worldwide distribution of the bacteria and the severity of the diseases have led to a significant level of interest in the development of vaccines against the bacteria. Current vaccines, based on the bacterial polysaccharide, have a number of limitations including poor immunogenicity and limited effectiveness against all pneumococcal serotypes. There are many challenges in developing vaccines that will be effective against the diverse range of isolates and serotypes for this highly variable bacterial pathogen. This review considers how proteomic technologies have extended our understanding of the pathogenic mechanisms of nasopharyngeal colonization and disease development as well as the critical areas in developing protein-based vaccines.
Collapse
Affiliation(s)
- Mustapha Bittaye
- a Division of Applied Medicine , University of Aberdeen , Aberdeen , Scotland
| | - Phil Cash
- a Division of Applied Medicine , University of Aberdeen , Aberdeen , Scotland
| |
Collapse
|
41
|
Walters KA, D'Agnillo F, Sheng ZM, Kindrachuk J, Schwartzman LM, Kuestner RE, Chertow DS, Golding BT, Taubenberger JK, Kash JC. 1918 pandemic influenza virus and Streptococcus pneumoniae co-infection results in activation of coagulation and widespread pulmonary thrombosis in mice and humans. J Pathol 2015; 238:85-97. [PMID: 26383585 DOI: 10.1002/path.4638] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/25/2015] [Accepted: 09/07/2015] [Indexed: 12/15/2022]
Abstract
To study bacterial co-infection following 1918 H1N1 influenza virus infection, mice were inoculated with the 1918 influenza virus, followed by Streptococcus pneumoniae (SP) 72 h later. Co-infected mice exhibited markedly more severe disease, shortened survival time and more severe lung pathology, including widespread thrombi. Transcriptional profiling revealed activation of coagulation only in co-infected mice, consistent with the extensive thrombogenesis observed. Immunohistochemistry showed extensive expression of tissue factor (F3) and prominent deposition of neutrophil elastase on endothelial and epithelial cells in co-infected mice. Lung sections of SP-positive 1918 autopsy cases showed extensive thrombi and prominent staining for F3 in alveolar macrophages, monocytes, neutrophils, endothelial and epithelial cells, in contrast to co-infection-positive 2009 pandemic H1N1 autopsy cases. This study reveals that a distinctive feature of 1918 influenza virus and SP co-infection in mice and humans is extensive expression of tissue factor and activation of the extrinsic coagulation pathway leading to widespread pulmonary thrombosis.
Collapse
Affiliation(s)
| | - Felice D'Agnillo
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, USA
| | - Zong-Mei Sheng
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - Jason Kindrachuk
- Critical Care Medicine Department, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Louis M Schwartzman
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | | | - Daniel S Chertow
- Critical Care Medicine Department, National Institutes of Health (NIH), Bethesda, MD, USA.,Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - Basil T Golding
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Center for Biologics Evaluation and Research, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, USA
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - John C Kash
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
42
|
Owen CD, Lukacik P, Potter JA, Sleator O, Taylor GL, Walsh MA. Streptococcus pneumoniae NanC: STRUCTURAL INSIGHTS INTO THE SPECIFICITY AND MECHANISM OF A SIALIDASE THAT PRODUCES A SIALIDASE INHIBITOR. J Biol Chem 2015; 290:27736-48. [PMID: 26370075 PMCID: PMC4646021 DOI: 10.1074/jbc.m115.673632] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Indexed: 12/26/2022] Open
Abstract
Streptococcus pneumoniae is an important human pathogen that causes a range of disease states. Sialidases are important bacterial virulence factors. There are three pneumococcal sialidases: NanA, NanB, and NanC. NanC is an unusual sialidase in that its primary reaction product is 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (Neu5Ac2en, also known as DANA), a nonspecific hydrolytic sialidase inhibitor. The production of Neu5Ac2en from α2–3-linked sialosides by the catalytic domain is confirmed within a crystal structure. A covalent complex with 3-fluoro-β-N-acetylneuraminic acid is also presented, suggesting a common mechanism with other sialidases up to the final step of product formation. A conformation change in an active site hydrophobic loop on ligand binding constricts the entrance to the active site. In addition, the distance between the catalytic acid/base (Asp-315) and the ligand anomeric carbon is unusually short. These features facilitate a novel sialidase reaction in which the final step of product formation is direct abstraction of the C3 proton by the active site aspartic acid, forming Neu5Ac2en. NanC also possesses a carbohydrate-binding module, which is shown to bind α2–3- and α2–6-linked sialosides, as well as N-acetylneuraminic acid, which is captured in the crystal structure following hydration of Neu5Ac2en by NanC. Overall, the pneumococcal sialidases show remarkable mechanistic diversity while maintaining a common structural scaffold.
Collapse
Affiliation(s)
- C David Owen
- From the Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, Fife KY16 9ST, United Kingdom
| | - Petra Lukacik
- Diamond Light Source and Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 0FA, United Kingdom, and
| | - Jane A Potter
- From the Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, Fife KY16 9ST, United Kingdom
| | - Olivia Sleator
- the Medical Research Council France, c/o European Synchrotron Radiation Facility, BP 220, 38043 Grenoble, France
| | - Garry L Taylor
- From the Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, Fife KY16 9ST, United Kingdom,
| | - Martin A Walsh
- Diamond Light Source and the Medical Research Council France, c/o European Synchrotron Radiation Facility, BP 220, 38043 Grenoble, France
| |
Collapse
|
43
|
Paixão L, Oliveira J, Veríssimo A, Vinga S, Lourenço EC, Ventura MR, Kjos M, Veening JW, Fernandes VE, Andrew PW, Yesilkaya H, Neves AR. Host glycan sugar-specific pathways in Streptococcus pneumoniae: galactose as a key sugar in colonisation and infection [corrected]. PLoS One 2015; 10:e0121042. [PMID: 25826206 PMCID: PMC4380338 DOI: 10.1371/journal.pone.0121042] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/12/2015] [Indexed: 01/13/2023] Open
Abstract
The human pathogen Streptococcus pneumoniae is a strictly fermentative organism that relies on glycolytic metabolism to obtain energy. In the human nasopharynx S. pneumoniae encounters glycoconjugates composed of a variety of monosaccharides, which can potentially be used as nutrients once depolymerized by glycosidases. Therefore, it is reasonable to hypothesise that the pneumococcus would rely on these glycan-derived sugars to grow. Here, we identified the sugar-specific catabolic pathways used by S. pneumoniae during growth on mucin. Transcriptome analysis of cells grown on mucin showed specific upregulation of genes likely to be involved in deglycosylation, transport and catabolism of galactose, mannose and N acetylglucosamine. In contrast to growth on mannose and N-acetylglucosamine, S. pneumoniae grown on galactose re-route their metabolic pathway from homolactic fermentation to a truly mixed acid fermentation regime. By measuring intracellular metabolites, enzymatic activities and mutant analysis, we provide an accurate map of the biochemical pathways for galactose, mannose and N-acetylglucosamine catabolism in S. pneumoniae. Intranasal mouse infection models of pneumococcal colonisation and disease showed that only mutants in galactose catabolic genes were attenuated. Our data pinpoint galactose as a key nutrient for growth in the respiratory tract and highlights the importance of central carbon metabolism for pneumococcal pathogenesis.
Collapse
Affiliation(s)
- Laura Paixão
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Joana Oliveira
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - André Veríssimo
- Centre for Intelligent Systems, LAETA, IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Susana Vinga
- Centre for Intelligent Systems, LAETA, IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Eva C. Lourenço
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - M. Rita Ventura
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Morten Kjos
- Molecular Genetics Group, Groningen Biomolecular Sciences and Biotechnology Institute, Centre for Synthetic Biology, University of Groningen, Groningen, The Netherlands
| | - Jan-Willem Veening
- Molecular Genetics Group, Groningen Biomolecular Sciences and Biotechnology Institute, Centre for Synthetic Biology, University of Groningen, Groningen, The Netherlands
| | - Vitor E. Fernandes
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, United Kingdom
| | - Peter W. Andrew
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, United Kingdom
| | - Hasan Yesilkaya
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, United Kingdom
| | - Ana Rute Neves
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
- * E-mail:
| |
Collapse
|
44
|
Vandevelde NM, Tulkens PM, Muccioli GG, Van Bambeke F. Modulation of the activity of moxifloxacin and solithromycin in an in vitro pharmacodynamic model of Streptococcus pneumoniae naive and induced biofilms. J Antimicrob Chemother 2015; 70:1713-26. [PMID: 25712316 DOI: 10.1093/jac/dkv032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/21/2015] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Bacterial biofilms developing in the bronchial tree of patients experiencing acute exacerbations of chronic bronchitis (AECBs) are suggested to cause relapses and recurrences of the disease because the matrix barrier impairs antibiotic access to the offending organisms. We examined whether bronchodilators could modulate pneumococcal biofilm development and antibiotic action using an in vitro model. METHODS Streptococcus pneumoniae strains from patients hospitalized for AECBs and two reference strains (ATCC 49619 and R6) were screened for biofilm formation (multi-well plates; 2-11 days of growth). Ipratropium and salbutamol (alone or in combination) were added at concentrations of 1.45 and 7.25 mg/L, respectively (mimicking those in the bronchial tree), and their effects were measured on biofilm formation and modulation of the activity of antibiotics [full antibiotic concentration-dependent effects (pharmacodynamic model)] with a focus on moxifloxacin and solithromycin. Bacterial viability and biomass were measured by the reduction of resazurin and crystal violet staining, respectively. Release of sialic acid (from biofilm) and neuraminidase activity were measured using enzymatic and HPLC-MS detection of sialic acid. RESULTS All clinical isolates produced biofilms, but with fast disassembly if from patients who had received muscarinic antagonists. Ipratropium caused: (i) reduced biomass formation and faster biofilm disassembly with free sialic acid release; and (ii) a marked improvement of antibiotic activity (bacterial killing and biomass reduction). Salbutamol stimulated neuraminidase activity associated with improved antibiotic killing activity (reversed by zanamivir) but modest biomass reduction. CONCLUSIONS Ipratropium and, to a lesser extent, salbutamol may cooperate with antibiotics for bacterial clearance and disassembly of pneumococcal biofilms.
Collapse
Affiliation(s)
- Nathalie M Vandevelde
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Paul M Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
45
|
Szilágyi Á, Györke Z, Bereczki C, Kelen K, Tóth-Heyn P, Tulassay T, Reusz GS, Szabó AJ, Prohászka Z. The use of a rapid fluorogenic neuraminidase assay to differentiate acute Streptococcus pneumoniae-associated hemolytic uremic syndrome (HUS) from other forms of HUS. ACTA ACUST UNITED AC 2015; 53:e117-9. [DOI: 10.1515/cclm-2014-0400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 09/01/2014] [Indexed: 12/25/2022]
|
46
|
Walther E, Richter M, Xu Z, Kramer C, von Grafenstein S, Kirchmair J, Grienke U, Rollinger JM, Liedl KR, Slevogt H, Sauerbrei A, Saluz HP, Pfister W, Schmidtke M. Antipneumococcal activity of neuraminidase inhibiting artocarpin. Int J Med Microbiol 2014; 305:289-97. [PMID: 25592264 DOI: 10.1016/j.ijmm.2014.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 12/01/2014] [Accepted: 12/14/2014] [Indexed: 02/06/2023] Open
Abstract
Streptococcus (S.) pneumoniae is a major cause of secondary bacterial pneumonia during influenza epidemics. Neuraminidase (NA) is a virulence factor of both pneumococci and influenza viruses. Bacterial neuraminidases (NAs) are structurally related to viral NA and susceptible to oseltamivir, an inhibitor designed to target viral NA. This prompted us to evaluate the antipneumococcal potential of two NA inhibiting natural compounds, the diarylheptanoid katsumadain A and the isoprenylated flavone artocarpin. Chemiluminescence, fluorescence-, and hemagglutination-based enzyme assays were applied to determine the inhibitory efficiency (IC(50) value) of the tested compounds towards pneumococcal NAs. The mechanism of inhibition was studied via enzyme kinetics with recombinant NanA NA. Unlike oseltamivir, which competes with the natural substrate of NA, artocarpin exhibits a mixed-type inhibition with a Ki value of 9.70 μM. Remarkably, artocarpin was the only NA inhibitor (NAI) for which an inhibitory effect on pneumococcal growth (MIC: 0.99-5.75 μM) and biofilm formation (MBIC: 1.15-2.97 μM) was observable. In addition, we discovered that the bactericidal effect of artocarpin can reduce the viability of pneumococci by a factor of >1000, without obvious harm to lung epithelial cells. This renders artocarpin a promising natural product for further investigations.
Collapse
Affiliation(s)
- E Walther
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - M Richter
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - Z Xu
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - C Kramer
- University of Innsbruck, Institute for General, Inorganic and Theoretical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Innrain 80/82, 6020 Innsbruck, Austria
| | - S von Grafenstein
- University of Innsbruck, Institute for General, Inorganic and Theoretical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Innrain 80/82, 6020 Innsbruck, Austria
| | - J Kirchmair
- University of Hamburg, Center for Bioinformatics, Bundesstraße 43, 20146 Hamburg, Germany
| | - U Grienke
- University of Innsbruck, Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), Innrain 80/82, 6020 Innsbruck, Austria; University of Vienna, Department of Pharmacognosy, Althanstraße 14, 1090 Vienna, Austria
| | - J M Rollinger
- University of Vienna, Department of Pharmacognosy, Althanstraße 14, 1090 Vienna, Austria
| | - K R Liedl
- University of Innsbruck, Institute for General, Inorganic and Theoretical Chemistry and Center for Molecular Biosciences Innsbruck (CMBI), Innrain 80/82, 6020 Innsbruck, Austria
| | - H Slevogt
- Jena University Hospital, ZIK Septomics, Albert-Einstein-Straße 10, 07745 Jena, Germany
| | - A Sauerbrei
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany
| | - H P Saluz
- Leibniz Institute for Natural Product Research and Infection Biology, Beutenbergstraße 11a, 07745 Jena, Germany
| | - W Pfister
- Jena University Hospital, Department of Medical Microbiology, Erlanger Allee 101, 07747 Jena, Germany
| | - M Schmidtke
- Jena University Hospital, Department of Virology and Antiviral Therapy, Hans-Knöll-Straße 2, 07745 Jena, Germany.
| |
Collapse
|
47
|
Jack AA, Daniels DE, Jepson MA, Vickerman MM, Lamont RJ, Jenkinson HF, Nobbs AH. Streptococcus gordonii comCDE (competence) operon modulates biofilm formation with Candida albicans. MICROBIOLOGY-SGM 2014; 161:411-421. [PMID: 25505189 DOI: 10.1099/mic.0.000010] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Candida albicans is a pleiomorphic fungus that forms mixed species biofilms with Streptococcus gordonii, an early colonizer of oral cavity surfaces. Activation of quorum sensing (QS; intercellular signalling) promotes monospecies biofilm development by these micro-organisms, but the role of QS in mixed species communities is not understood. The comCDE genes in S. gordonii encode a sensor-regulator system (ComDE), which is activated by the comC gene product (CSP, competence stimulating peptide) and modulates expression of QS-regulated genes. Dual species biofilms of S. gordonii ΔcomCDE or ΔcomC mutants with C. albicans showed increased biomass compared to biofilms of S. gordonii DL1 wild-type with C. albicans. The ΔcomCDE mutant dual species biofilms in particular contained more extracellular DNA (eDNA), and could be dispersed with DNase I or protease treatment. Exogenous CSP complemented the S. gordonii ΔcomC transformation deficiency, as well as the ΔcomC-C. albicans biofilm phenotype. Purified CSP did not affect C. albicans hyphal filament formation but inhibited monospecies biofilm formation by C. albicans. The results suggest that the S. gordonii comCDE QS-system modulates the production of eDNA and the incorporation of C. albicans into dual species biofilms.
Collapse
Affiliation(s)
- Alison A Jack
- School of Oral and Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Debbie E Daniels
- School of Biochemistry, University of Bristol, University Walk, Bristol BS8 1TD, UK.,School of Oral and Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Mark A Jepson
- School of Biochemistry, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - M Margaret Vickerman
- Department of Oral Biology, University at Buffalo, 223 Foster Hall, Buffalo, NY 14214, USA
| | - Richard J Lamont
- Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, 501 South Preston Street, Louisville, KY 40202, USA
| | - Howard F Jenkinson
- School of Oral and Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Angela H Nobbs
- School of Oral and Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| |
Collapse
|
48
|
Feldman C, Anderson R. Review: Current and new generation pneumococcal vaccines. J Infect 2014; 69:309-25. [DOI: 10.1016/j.jinf.2014.06.006] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/16/2014] [Indexed: 12/22/2022]
|
49
|
Singh AK, Pluvinage B, Higgins MA, Dalia AB, Woodiga SA, Flynn M, Lloyd AR, Weiser JN, Stubbs KA, Boraston AB, King SJ. Unravelling the multiple functions of the architecturally intricate Streptococcus pneumoniae β-galactosidase, BgaA. PLoS Pathog 2014; 10:e1004364. [PMID: 25210925 PMCID: PMC4161441 DOI: 10.1371/journal.ppat.1004364] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/28/2014] [Indexed: 11/18/2022] Open
Abstract
Bacterial cell-surface proteins play integral roles in host-pathogen interactions. These proteins are often architecturally and functionally sophisticated and yet few studies of such proteins involved in host-pathogen interactions have defined the domains or modules required for specific functions. Streptococcus pneumoniae (pneumococcus), an opportunistic pathogen that is a leading cause of community acquired pneumonia, otitis media and bacteremia, is decorated with many complex surface proteins. These include β-galactosidase BgaA, which is specific for terminal galactose residues β-1-4 linked to glucose or N-acetylglucosamine and known to play a role in pneumococcal growth, resistance to opsonophagocytic killing, and adherence. This study defines the domains and modules of BgaA that are required for these distinct contributions to pneumococcal pathogenesis. Inhibitors of β-galactosidase activity reduced pneumococcal growth and increased opsonophagocytic killing in a BgaA dependent manner, indicating these functions require BgaA enzymatic activity. In contrast, inhibitors increased pneumococcal adherence suggesting that BgaA bound a substrate of the enzyme through a distinct module or domain. Extensive biochemical, structural and cell based studies revealed two newly identified non-enzymatic carbohydrate-binding modules (CBMs) mediate adherence to the host cell surface displayed lactose or N-acetyllactosamine. This finding is important to pneumococcal biology as it is the first adhesin-carbohydrate receptor pair identified, supporting the widely held belief that initial pneumococcal attachment is to a glycoconjugate. Perhaps more importantly, this is the first demonstration that a CBM within a carbohydrate-active enzyme can mediate adherence to host cells and thus this study identifies a new class of carbohydrate-binding adhesins and extends the paradigm of CBM function. As other bacterial species express surface-associated carbohydrate-active enzymes containing CBMs these findings have broad implications for bacterial adherence. Together, these data illustrate that comprehending the architectural sophistication of surface-attached proteins can increase our understanding of the different mechanisms by which these proteins can contribute to bacterial pathogenesis.
Collapse
Affiliation(s)
- Anirudh K. Singh
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Benjamin Pluvinage
- Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Melanie A. Higgins
- Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Ankur B. Dalia
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Shireen A. Woodiga
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Matthew Flynn
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
| | - Audrey R. Lloyd
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
| | - Jeffrey N. Weiser
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Keith A. Stubbs
- School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Alisdair B. Boraston
- Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
- * E-mail: (ABB); (SJK)
| | - Samantha J. King
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (ABB); (SJK)
| |
Collapse
|
50
|
Identification and characterization of a novel secreted glycosidase with multiple glycosidase activities in Streptococcus intermedius. J Bacteriol 2014; 196:2817-26. [PMID: 24858187 DOI: 10.1128/jb.01727-14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus intermedius is a known human pathogen and belongs to the anginosus group (S. anginosus, S. intermedius, and S. constellatus) of streptococci (AGS). We found a large open reading frame (6,708 bp) in the lac operon, and bioinformatic analysis suggested that this gene encodes a novel glycosidase that can exhibit β-d-galactosidase and N-acetyl-β-d-hexosaminidase activities. We, therefore, named this protein "multisubstrate glycosidase A" (MsgA). To test whether MsgA has these glycosidase activities, the msgA gene was disrupted in S. intermedius. The msgA-deficient mutant no longer showed cell- and supernatant-associated β-d-galactosidase, β-d-fucosidase, N-acetyl-β-d-glucosaminidase, and N-acetyl-β-d-galactosaminidase activities, and all phenotypes were complemented in trans with a recombinant plasmid carrying msgA. Purified MsgA had all four of these glycosidase activities and exhibited the lowest Km with 4-methylumbelliferyl-linked N-acetyl-β-d-glucosaminide and the highest kcat with 4-methylumbelliferyl-linked β-d-galactopyranoside. In addition, the purified LacZ domain of MsgA had β-d-galactosidase and β-d-fucosidase activities, and the GH20 domain exhibited both N-acetyl-β-d-glucosaminidase and N-acetyl-β-d-galactosaminidase activities. The β-d-galactosidase and β-d-fucosidase activities of MsgA are thermolabile, and the optimal temperature of the reaction was 40°C, whereas almost all enzymatic activities disappeared at 49°C. The optimal temperatures for the N-acetyl-β-d-glucosaminidase and N-acetyl-β-d-galactosaminidase activities were 58 and 55°C, respectively. The requirement of sialidase treatment to remove sialic acid residues of the glycan branch end for glycan degradation by MsgA on human α1-antitrypsin indicates that MsgA has exoglycosidase activities. MsgA and sialidase might have an important function in the production and utilization of monosaccharides from oligosaccharides, such as glycans for survival in a normal habitat and for pathogenicity of S. intermedius.
Collapse
|