1
|
Fritsch KJ, Krüger L, Handtke S, Kohler TP, Ozhiganova A, Jahn K, Wesche J, Greinacher A, Hammerschmidt S. Pneumococcal Neuraminidases Increase Platelet Killing by Pneumolysin. Thromb Haemost 2025; 125:243-254. [PMID: 39029905 DOI: 10.1055/a-2369-8680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
BACKGROUND Platelets prevent extravasation of capillary fluids into the pulmonary interstitial tissue by sealing gaps in inflamed endothelium. This reduces respiratory distress associated with pneumonia. Streptococcus pneumoniae is the leading cause of severe community-acquired pneumonia. Pneumococci produce pneumolysin (PLY), which forms pores in membranes of eukaryotic cells including platelets. Additionally, pneumococci express neuraminidases, which cleave sialic acid residues from eukaryotic glycoproteins. In this study, we investigated the effect of desialylation on PLY binding and pore formation on platelets. MATERIALS AND METHODS We incubated human platelets with purified neuraminidases and PLY, or nonencapsulated S. pneumoniae D39/TIGR4 and isogenic mutants deficient in PLY and/or NanA. We assessed platelet desialylation, PLY binding, and pore formation by flow cytometry. We also analyzed the inhibitory potential of therapeutic immunoglobulin G preparations (IVIG [intravenous immunoglobulin]). RESULTS Wild-type pneumococci cause desialylation of platelet glycoproteins by neuraminidases, which is reduced by 90 to 100% in NanA-deficient mutants. NanC, cleaving only α2,3-linked sialic acid, induced platelet desialylation. PLY binding to platelets then x2doubled (p = 0.0166) and pore formation tripled (p = 0.0373). A neuraminidase cleaving α2,3-, α2,6-, and α2,8-linked sialic acid like NanA was even more efficient. Addition of polyvalent IVIG (5 mg/mL) decreased platelet desialylation induced by NanC up to 90% (p = 0.263) and reduced pore formation >95% (p < 0.0001) when incubated with pneumococci. CONCLUSION Neuraminidases are key virulence factors of pneumococci and desialylate platelet glycoproteins, thereby unmasking PLY-binding sites. This enhances binding of PLY and pore formation showing that pneumococcal neuraminidases and PLY act in concert to kill platelets. However, human polyvalent immunoglobulin G preparations are promising agents for therapeutic intervention during severe pneumococcal pneumonia.
Collapse
Affiliation(s)
- Kristin J Fritsch
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Laura Krüger
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Stefan Handtke
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Rostock, Rostock, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Arina Ozhiganova
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Kristin Jahn
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Jan Wesche
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Andreas Greinacher
- Department of Transfusion Medicine, Institute of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
2
|
Yacoub E, Baby V, Sirand-Pugnet P, Arfi Y, Mardassi H, Blanchard A, Chibani S, Ben Abdelmoumen Mardassi B. A sweeping view of avian mycoplasmas biology drawn from comparative genomic analyses. BMC Genomics 2025; 26:24. [PMID: 39789465 PMCID: PMC11720521 DOI: 10.1186/s12864-024-11201-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Avian mycoplasmas are small bacteria associated with several pathogenic conditions in many wild and poultry bird species. Extensive genomic data are available for many avian mycoplasmas, yet no comparative studies focusing on this group of mycoplasmas have been undertaken so far. RESULTS Here, based on the comparison of forty avian mycoplasma genomes belonging to ten different species, we provide insightful information on the phylogeny, pan/core genome, energetic metabolism, and virulence of these avian pathogens. Analyses disclosed considerable inter- and intra-species genomic variabilities, with genome sizes that can vary by twice as much. Phylogenetic analysis based on concatenated orthologous genes revealed that avian mycoplasmas fell into either Hominis or Pneumoniae groups within the Mollicutes and could split into various clusters. No host co-evolution of avian mycoplasmas can be inferred from the proposed phylogenetic scheme. With 3,237 different gene clusters, the avian mycoplasma group under study proved diverse enough to have an open pan genome. However, a set of 150 gene clusters was found to be shared between all avian mycoplasmas, which is likely encoding essential functions. Comparison of energy metabolism pathways showed that avian mycoplasmas rely on various sources of energy. Superposition between phylogenetic and energy metabolism groups revealed that the glycolytic mycoplasmas belong to two distinct phylogenetic groups (Hominis and Pneumoniae), while all the arginine-utilizing mycoplasmas belong only to Hominis group. This can stand for different evolutionary strategies followed by avian mycoplasmas and further emphasizes the diversity within this group. Virulence determinants survey showed that the involved gene arsenals vary significantly within and between species, and could even be found in species often reported apathogenic. Immunoglobulin-blocking proteins were detected in almost all avian mycoplasmas. Although these systems are not exclusive to this group, they seem to present some particular features making them unique among mycoplasmas. CONCLUSION This comparative genomic study uncovered the significant variable nature of avian mycoplasmas, furthering our knowledge on their biological attributes and evoking new hallmarks.
Collapse
Affiliation(s)
- Elhem Yacoub
- Unit of Mycoplasmas, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Vincent Baby
- Centre de Diagnostic Vétérinaire de L'Université de Montréal (CDVUM), Faculty of Veterinary Medecine, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | | | - Yonathan Arfi
- Univ. Bordeaux, INRAE, UMR BFP, 33882, Villenave d'Ornon, France
| | - Helmi Mardassi
- Unit of Typing and Genetics of Mycobacteria, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Alain Blanchard
- Univ. Bordeaux, INRAE, UMR BFP, 33882, Villenave d'Ornon, France
| | - Salim Chibani
- Unit of Mycoplasmas, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Boutheina Ben Abdelmoumen Mardassi
- Unit of Mycoplasmas, Laboratory of Molecular Microbiology, Vaccinology and Biotechnology Development, Institut Pasteur de Tunis, University Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
3
|
Singh AP, Ahmad S, Raza K, Gautam HK. Computational screening and MM/GBSA-based MD simulation studies reveal the high binding potential of FDA-approved drugs against Cutibacterium acnes sialidase. J Biomol Struct Dyn 2024; 42:6245-6255. [PMID: 37545341 DOI: 10.1080/07391102.2023.2242950] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023]
Abstract
Cutibacterium acnes is an opportunistic pathogen linked with acne vulgaris, affecting 80-90% of teenagers globally. On the leukocyte (WBCs) cell surface, the cell wall anchored sialidase in C. acnes virulence factor, catalysing the sialoconjugates into sialic acids and nutrients for C. acnes resulting in human skin inflammation. The clinical use of antibiotics for acne treatments has severe adverse effects, including microbial dysbiosis and resistance. Therefore, identifying inhibitors for primary virulence factors (Sialidase) was done using molecular docking of 1030 FDA-approved drugs. Initially, based on binding energies (ΔG), Naloxone (ZINC000000389747), Fenoldopam (ZINC000022116608), Labetalol (ZINC000000403010) and Thalitone (ZINC000000057255) were identified that showed high binding energies as -10.2, -10.1, -9.9 and -9.8 kcal/mol, respectively. In 2D analysis, these drugs also showed considerable structural conformer of hydrogen and hydrophobic interactions. Further, a 100 ns MD simulation study found the lowest deviation and fluctuations with various intermolecular interactions to stabilise the complexes. Out of 4, the Naloxone molecule showed robust, steady, and stable RMSD 0.23 ± 0.18 nm. Further, MMGBSA analysis supports MD results and found strong binding energy (ΔG) -29.71 ± 4.97 kcal/mol. In Comparative studies with Neu5Ac2en (native substrate) revealed naloxone has a higher affinity for sialidase. The PCA analysis showed that Naloxone and Thalitone were actively located on the active site, and other compounds were flickered. Our extensive computational and statistical report demonstrates that these FDA drugs can be validated as potential sialidase inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akash Pratap Singh
- Infectious Disease Laboratory, Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad, India
| | - Shaban Ahmad
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Hemant K Gautam
- Infectious Disease Laboratory, Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad, India
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Kruckow KL, Zhao K, Bowdish DME, Orihuela CJ. Acute organ injury and long-term sequelae of severe pneumococcal infections. Pneumonia (Nathan) 2023; 15:5. [PMID: 36870980 PMCID: PMC9985869 DOI: 10.1186/s41479-023-00110-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is a major public health problem, as it is a main cause of otitis media, community-acquired pneumonia, bacteremia, sepsis, and meningitis. Acute episodes of pneumococcal disease have been demonstrated to cause organ damage with lingering negative consequences. Cytotoxic products released by the bacterium, biomechanical and physiological stress resulting from infection, and the corresponding inflammatory response together contribute to organ damage accrued during infection. The collective result of this damage can be acutely life-threatening, but among survivors, it also contributes to the long-lasting sequelae of pneumococcal disease. These include the development of new morbidities or exacerbation of pre-existing conditions such as COPD, heart disease, and neurological impairments. Currently, pneumonia is ranked as the 9th leading cause of death, but this estimate only considers short-term mortality and likely underestimates the true long-term impact of disease. Herein, we review the data that indicates damage incurred during acute pneumococcal infection can result in long-term sequelae which reduces quality of life and life expectancy among pneumococcal disease survivors.
Collapse
Affiliation(s)
- Katherine L Kruckow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kevin Zhao
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Dawn M E Bowdish
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Carlos J Orihuela
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
7
|
Mathew BJ, Gupta P, Naaz T, Rai R, Gupta S, Gupta S, Chaurasiya SK, Purwar S, Biswas D, Vyas AK, Singh AK. Role of Streptococcus pneumoniae extracellular glycosidases in immune evasion. Front Cell Infect Microbiol 2023; 13:1109449. [PMID: 36816580 PMCID: PMC9937060 DOI: 10.3389/fcimb.2023.1109449] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Streptococcus pneumoniae (pneumococcus) typically colonizes the human upper airway asymptomatically but upon reaching other sites of the host body can cause an array of diseases such as pneumonia, bacteremia, otitis media, and meningitis. Be it colonization or progression to disease state, pneumococcus faces multiple challenges posed by host immunity ranging from complement mediated killing to inflammation driven recruitment of bactericidal cells for the containment of the pathogen. Pneumococcus has evolved several mechanisms to evade the host inflicted immune attack. The major pneumococcal virulence factor, the polysaccharide capsule helps protect the bacteria from complement mediated opsonophagocytic killing. Another important group of pneumococcal proteins which help bacteria to establish and thrive in the host environment is surface associated glycosidases. These enzymes can hydrolyze host glycans on glycoproteins, glycolipids, and glycosaminoglycans and consequently help bacteria acquire carbohydrates for growth. Many of these glycosidases directly or indirectly facilitate bacterial adherence and are known to modulate the function of host defense/immune proteins likely by removing glycans and thereby affecting their stability and/or function. Furthermore, these enzymes are known to contribute the formation of biofilms, the bacterial communities inherently resilient to antimicrobials and host immune attack. In this review, we summarize the role of these enzymes in host immune evasion.
Collapse
Affiliation(s)
- Bijina J. Mathew
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | - Priyal Gupta
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Tabassum Naaz
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Rupal Rai
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | - Sudheer Gupta
- Research and Development, 3B Blackbio Biotech India Ltd., Bhopal, India
| | - Sudipti Gupta
- Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Shivendra K. Chaurasiya
- Department of Biological Science and Engineering, Maulana Azad National Institute of Technology, Bhopal, India
| | - Shashank Purwar
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Debasis Biswas
- Department of Microbiology, All India Institute of Medical Sciences, Bhopal, India
| | - Ashish Kumar Vyas
- John C Martin Centre for Liver Research and Innovation, Liver Foundation Sonarpur, Kolkata, India
| | - Anirudh K. Singh
- School of Sciences, SAM Global University, Raisen, India,*Correspondence: Anirudh K. Singh,
| |
Collapse
|
8
|
Klenow L, Elfageih R, Gao J, Wan H, Withers SG, de Gier JW, Daniels R. Influenza virus and pneumococcal neuraminidases enhance catalysis by similar yet distinct sialic acid-binding strategies. J Biol Chem 2023; 299:102891. [PMID: 36634846 PMCID: PMC9929470 DOI: 10.1016/j.jbc.2023.102891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Influenza A viruses and the bacterium Streptococcus pneumoniae (pneumococci) both express neuraminidases that catalyze release of sialic acid residues from oligosaccharides and glycoproteins. Although these respiratory pathogen neuraminidases function in a similar environment, it remains unclear if these enzymes use similar mechanisms for sialic acid cleavage. Here, we compared the enzymatic properties of neuraminidases from two influenza A subtypes (N1 and N2) and the pneumococcal strain TIGR4 (NanA, NanB, and NanC). Insect cell-produced N1 and N2 tetramers exhibited calcium-dependent activities and stabilities that varied with pH. In contrast, E. coli-produced NanA, NanB, and NanC were isolated as calcium insensitive monomers with stabilities that were more resistant to pH changes. Using a synthetic substrate (MUNANA), all neuraminidases showed similar pH optimums (pH 6-7) that were primarily defined by changes in catalytic rate rather than substrate binding affinity. Upon using a multivalent substrate (fetuin sialoglycans), much higher specific activities were observed for pneumococcal neuraminidases that contain an additional lectin domain. In virions, N1 and especially N2 also showed enhanced specific activity toward fetuin that was lost upon the addition of detergent, indicating the sialic acid-binding capacity of neighboring hemagglutinin molecules likely contributes to catalysis of natural multivalent substrates. These results demonstrate that influenza and pneumococcal neuraminidases have evolved similar yet distinct strategies to optimize their catalytic activity.
Collapse
Affiliation(s)
- Laura Klenow
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Rageia Elfageih
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Jin Gao
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hongquan Wan
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Stephen G. Withers
- Department of Chemistry, University of British Columbia, Vancouver, Canada
| | - Jan-Willem de Gier
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Robert Daniels
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA.
| |
Collapse
|
9
|
Escuret V, Terrier O. Co-infection of the respiratory epithelium, scene of complex functional interactions between viral, bacterial, and human neuraminidases. Front Microbiol 2023; 14:1137336. [PMID: 37213507 PMCID: PMC10192862 DOI: 10.3389/fmicb.2023.1137336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/03/2023] [Indexed: 05/23/2023] Open
Abstract
The activity of sialic acids, known to play critical roles in biology and many pathological processes, is finely regulated by a class of enzymes called sialidases, also known as neuraminidases. These are present in mammals and many other biological systems, such as viruses and bacteria. This review focuses on the very particular situation of co-infections of the respiratory epithelium, the scene of complex functional interactions between viral, bacterial, and human neuraminidases. This intrinsically multidisciplinary topic combining structural biology, biochemistry, physiology, and the study of host-pathogen interactions, opens up exciting research perspectives that could lead to a better understanding of the mechanisms underlying virus-bacteria co-infections and their contribution to the aggravation of respiratory pathology, notably in the context of pre-existing pathological contexts. Strategies that mimic or inhibit the activity of the neuraminidases could constitute interesting treatment options for viral and bacterial infections.
Collapse
|
10
|
Vezina B, Judd LM, McDougall FK, Boardman WSJ, Power ML, Hawkey J, Brisse S, Monk JM, Holt KE, Wyres KL. Transmission of Klebsiella strains and plasmids within and between grey-headed flying fox colonies. Environ Microbiol 2022; 24:4425-4436. [PMID: 35590448 PMCID: PMC9790207 DOI: 10.1111/1462-2920.16047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/11/2022] [Indexed: 12/30/2022]
Abstract
The grey-headed flying fox (Pteropus poliocephalus) is an endemic Australian fruit bat, known to carry zoonotic pathogens. We recently showed they harbour bacterial pathogen Klebsiella pneumoniae and closely related species in the K. pneumoniae species complex (KpSC); however, the dynamics of KpSC transmission and gene flow within flying fox colonies are poorly understood. High-resolution genome comparisons of 39 KpSC isolates from grey-headed flying foxes identified five putative strain transmission clusters (four intra- and one inter-colony). The instance of inter-colony strain transmission of K. africana was found between two flying fox populations within flying distance, indicating either direct or indirect transmission through a common food/water source. All 11 plasmids identified within the KpSC isolates showed 73% coverage (mean) and ≥95% identity to human-associated KpSC plasmids, indicating gene flow between human clinical and grey-headed flying fox isolates. Along with strain transmission, inter-species horizontal plasmid transmission between K. pneumoniae and Klebsiella africana was also identified within a flying fox colony. Finally, genome-scale metabolic models were generated to predict and compare substrate usage to previously published KpSC models, from human and environmental sources. These models indicated no distinction on the basis of metabolic capabilities. Instead, metabolic capabilities were consistent with population structure and ST/lineage.
Collapse
Affiliation(s)
- Ben Vezina
- Department of Infectious Diseases, Central Clinical SchoolMonash UniversityMelbourneVic.Australia
| | - Louise M. Judd
- Department of Infectious Diseases, Central Clinical SchoolMonash UniversityMelbourneVic.Australia
| | | | | | - Michelle L. Power
- Department of Biological SciencesMacquarie UniversityNSW2109Australia
| | - Jane Hawkey
- Department of Infectious Diseases, Central Clinical SchoolMonash UniversityMelbourneVic.Australia
| | - Sylvain Brisse
- Institut PasteurUniversité de Paris, Biodiversity and Epidemiology of Bacterial PathogensParisFrance
| | | | - Kathryn E. Holt
- Department of Infectious Diseases, Central Clinical SchoolMonash UniversityMelbourneVic.Australia
- Department of Infection BiologyLondon School of Hygiene and Tropical MedicineLondonUK
| | - Kelly L. Wyres
- Department of Infectious Diseases, Central Clinical SchoolMonash UniversityMelbourneVic.Australia
| |
Collapse
|
11
|
Yu ACY, Volkers G, Jongkees SAK, Worrall LJ, Withers SG, Strynadka NCJ. Crystal structure of the Propionibacterium acnes surface sialidase, a drug target for P. acnes-associated diseases. Glycobiology 2021; 32:162-170. [PMID: 34792586 DOI: 10.1093/glycob/cwab094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/28/2021] [Accepted: 05/19/2021] [Indexed: 11/12/2022] Open
Abstract
Propionibacterium acnes, though generally considered part of the normal flora of human skin, is an opportunistic pathogen associated with acne vulgaris as well as other diseases, including endocarditis, endophthalmitis and prosthetic joint infections. Its virulence potential is also supported by knowledge gained from its sequenced genome. Indeed, a vaccine targeting a putative cell wall-anchored P. acnes sialidase has been shown to suppress cytotoxicity and pro-inflammatory cytokine release induced by the organism, and is proposed as an alternative treatment for P. acnes-associated diseases. Here, we report the crystal structures of the surface sialidase and its complex with the transition-state mimic Neu5Ac2en. Our structural and kinetic analyses, together with insight from a glycan array screen, which probes subtle specificities of the sialidase for α-2,3-sialosides, provide a basis for the structure-based design of novel small-molecule therapeutics against P. acnes infections.
Collapse
Affiliation(s)
- Angel C Y Yu
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, 2350 Health Sciences Mall, V6T 1Z3, Canada
| | - Gesa Volkers
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, 2350 Health Sciences Mall, V6T 1Z3, Canada
| | - Seino A K Jongkees
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, V6T 1Z1, Canada
| | - Liam J Worrall
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, 2350 Health Sciences Mall, V6T 1Z3, Canada
| | - Stephen G Withers
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.,Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia, V6T 1Z1, Canada
| | - Natalie C J Strynadka
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, 2350 Health Sciences Mall, V6T 1Z3, Canada
| |
Collapse
|
12
|
Musikant D, Higa R, Rodríguez CE, Edreira MM, Campetella O, Jawerbaum A, Leguizamón MS. Sialic acid removal by trans-sialidase modulates MMP-2 activity during Trypanosoma cruzi infection. Biochimie 2021; 186:82-93. [PMID: 33891967 PMCID: PMC8187320 DOI: 10.1016/j.biochi.2021.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/21/2021] [Accepted: 04/14/2021] [Indexed: 01/05/2023]
Abstract
Matrix metalloproteinases (MMPs) not only play a relevant role in homeostatic processes but are also involved in several pathological mechanisms associated with infectious diseases. As their clinical relevance in Chagas disease has recently been highlighted, we studied the modulation of circulating MMPs by Trypanosoma cruzi infection. We found that virulent parasites from Discrete Typing Units (DTU) VI induced higher proMMP-2 and MMP-2 activity in blood, whereas both low (DTU I) and high virulence parasites induced a significant decrease in proMMP-9 plasma activity. Moreover, trans-sialidase, a relevant T. cruzi virulence factor, is involved in MMP-2 activity modulation both in vivo and in vitro. It removes α2,3-linked sialyl residues from cell surface glycoconjugates, which then triggers the PKC/MEK/ERK signaling pathway. Additionally, bacterial sialidases specific for this sialyl residue linkage displayed similar MMP modulation profiles and triggered the same signaling pathways. This novel pathogenic mechanism, dependent on sialic acid removal by the neuraminidase activity of trans-sialidase, can be exploited by different pathogens expressing sialidases with similar specificity. Thus, here we present a new pathogen strategy through the regulation of the MMP network.
Collapse
Affiliation(s)
- Daniel Musikant
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Ciudad de Buenos Aires, Argentina
| | - Romina Higa
- Consejo Nacional de Investigaciones Científicas y Técnicas, (CONICET) Godoy Cruz 2290, C1425FQB, Ciudad de Buenos Aires, Argentina; Laboratorio de Reproducción y Metabolismo, CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 C1121ABG, Ciudad de Buenos Aires, Argentina
| | - Cristina E Rodríguez
- Departamento de Microbiología, IMPAM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 C1121ABG, Ciudad de Buenos Aires, Argentina
| | - Martin M Edreira
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, C1428EGA, Ciudad de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, (CONICET) Godoy Cruz 2290, C1425FQB, Ciudad de Buenos Aires, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales IQUIBICEN-CONICET, Universidad de Buenos Aires, Intendente Güiraldes 2160 C1428EGA, Ciudad de Buenos Aires, Argentina
| | - Oscar Campetella
- Consejo Nacional de Investigaciones Científicas y Técnicas, (CONICET) Godoy Cruz 2290, C1425FQB, Ciudad de Buenos Aires, Argentina; Instituto de Investigaciones Biotecnológicas IIBio, Universidad Nacional de San Martín, 25 de Mayo y Francia B1650HMP, San Martín, San Martin, Argentina
| | - Alicia Jawerbaum
- Consejo Nacional de Investigaciones Científicas y Técnicas, (CONICET) Godoy Cruz 2290, C1425FQB, Ciudad de Buenos Aires, Argentina; Laboratorio de Reproducción y Metabolismo, CEFYBO-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155 C1121ABG, Ciudad de Buenos Aires, Argentina
| | - María S Leguizamón
- Consejo Nacional de Investigaciones Científicas y Técnicas, (CONICET) Godoy Cruz 2290, C1425FQB, Ciudad de Buenos Aires, Argentina; Instituto de Investigaciones Biotecnológicas IIBio, Universidad Nacional de San Martín, 25 de Mayo y Francia B1650HMP, San Martín, San Martin, Argentina.
| |
Collapse
|
13
|
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
|
14
|
Tseng YW, Chang CC, Chang YC. Novel Virulence Role of Pneumococcal NanA in Host Inflammation and Cell Death Through the Activation of Inflammasome and the Caspase Pathway. Front Cell Infect Microbiol 2021; 11:613195. [PMID: 33777832 PMCID: PMC7991587 DOI: 10.3389/fcimb.2021.613195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/05/2021] [Indexed: 01/15/2023] Open
Abstract
Streptococcus pneumoniae is one of most deadly Gram-positive bacterium that causes significant mortality and morbidity worldwide. Intense inflammation and cytotoxicity is a hallmark of invasive pneumococcal disease. Pneumococcal NanA has been shown to exaggerate the production of inflammatory cytokines via unmasking of inhibitory Siglec-5 from its sialyl cis-ligands. To further investigate the mechanistic role of NanA and Siglec-5 in pneumococccal diseases, we systemically analyzed genes and signaling pathways differentially regulated in macrophages infected with wild type and NanA-deficient pneumococcus. We found that NanA-mediated desialylation impairs the Siglec-5-TLR-2 interaction and reduces the recruitment of phosphatase SHP-1 to Siglec-5. This dysregulated crosstalk between TLR-2 and inhibitory Siglec-5 exaggerated multiple inflammatory and death signaling pathways and consequently caused excessive inflammation and cytotoxicity in the infected macrophage. Collectively, our results reveal a novel virulence role of NanA in pneumococcal pathogenesis and suggest that targeting NanA activity may ameliorate the pneumococcus-mediated inflammation and cytotoxicity in severe invasive pneumococcal diseases.
Collapse
Affiliation(s)
- Yu-Wen Tseng
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Chi Chang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Chi Chang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
15
|
Staphylococcal Protein A ( spa) Locus Is a Hot Spot for Recombination and Horizontal Gene Transfer in Staphylococcus pseudintermedius. mSphere 2020; 5:5/5/e00666-20. [PMID: 33115833 PMCID: PMC7593597 DOI: 10.1128/msphere.00666-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus pseudintermedius is a major canine pathogen but can also occasionally infect humans. Identification of genetic factors contributing to the virulence and clonal success of multidrug-resistant S. pseudintermedius clones is critical for the development of therapeutics against this pathogen. Here, we characterized the genome sequences of a global collection of 622 S. pseudintermedius isolates. We show that all major clones, besides carrying core virulence genes, which are present in all strains, carry one or more lineage-specific genes. Many of these genes have been acquired from other bacterial species through a horizontal gene transfer mechanism. Importantly, we have discovered that the staphylococcal protein A gene (spa), a widely used marker for molecular typing of S. pseudintermedius strains and a potential vaccine candidate antigen, is deleted in 62% of strains. Furthermore, the spa locus in S. pseudintermedius acts as a reservoir to accumulate lineage-associated genes with adaptive functions. Staphylococcus pseudintermedius is a major canine pathogen but also occasionally colonizes and infects humans. Multidrug-resistant methicillin-resistant S. pseudintermedius (MDR MRSP) strains have emerged globally, making treatment and control of this pathogen challenging. Sequence type 71 (ST71), ST68, and ST45 are the most widespread and successful MDR MRSP clones. The potential genetic factors underlying the clonal success of these and other predominant clones remain unknown. Characterization of the pangenome, lineage-associated accessory genes, and genes acquired through horizontal gene transfer from other bacteria is important for identifying such factors. Here, we analyzed genome sequence data from 622 S. pseudintermedius isolates to investigate the evolution of pathogenicity across lineages. We show that the predominant clones carry one or more lineage-associated virulence genes. The gene encoding staphylococcal protein A (SpA), a key virulence factor involved in immune evasion and a potential vaccine antigen, is deleted in 62% of isolates. Most importantly, we have discovered that the spa locus is a hot spot for recombination and horizontal gene transfer in S. pseudintermedius, where genes related to restriction modification, prophage immunity, mercury resistance, and nucleotide and carbohydrate metabolism have been acquired in different lineages. Our study also establishes that ST45 is composed of two distinct sublineages that differ in their accessory gene content and virulence potential. Collectively, this study reports several previously undetected lineage-associated genetic factors that may have a role in the clonal success of the major MDR MRSP clones. These data provide a framework for future experimental studies on S. pseudintermedius pathogenesis and for developing novel therapeutics against this pathogen. IMPORTANCEStaphylococcus pseudintermedius is a major canine pathogen but can also occasionally infect humans. Identification of genetic factors contributing to the virulence and clonal success of multidrug-resistant S. pseudintermedius clones is critical for the development of therapeutics against this pathogen. Here, we characterized the genome sequences of a global collection of 622 S. pseudintermedius isolates. We show that all major clones, besides carrying core virulence genes, which are present in all strains, carry one or more lineage-specific genes. Many of these genes have been acquired from other bacterial species through a horizontal gene transfer mechanism. Importantly, we have discovered that the staphylococcal protein A gene (spa), a widely used marker for molecular typing of S. pseudintermedius strains and a potential vaccine candidate antigen, is deleted in 62% of strains. Furthermore, the spa locus in S. pseudintermedius acts as a reservoir to accumulate lineage-associated genes with adaptive functions.
Collapse
|
16
|
Zhou J, Sun T, Kang W, Tang D, Feng Q. Pathogenic and antimicrobial resistance genes in Streptococcus oralis strains revealed by comparative genome analysis. Genomics 2020; 112:3783-3793. [PMID: 32334114 DOI: 10.1016/j.ygeno.2020.04.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 02/05/2023]
Abstract
Streptococcus oralis is an early colonizer bacterium in dental plaques and is considered a potential pathogen of infective endocarditis (IE) disease. In this study, we built a complete genome map of Streptococcus oralis strain SOT, Streptococcus oralis strain SOD and Streptococcus infantis strain SO and performed comparative genomic analysis among these three strains. The results showed that there are five genomic islands (GIs) in strain SOT and one CRISPR in strain SOD. Each genome harbors various pathogenic genes related to diseases and drug resistance, while the antibiotic resistance genes in strains SOT and SOD were quite similar but different from those in strain SO. In addition, we identified 17 main virulence factors and capsule-related genes in three strains. These results suggest the pathogenic potential of Streptococcus strains, which lay a foundation for the prevention and treatment of a Streptococcus oralis infection.
Collapse
Affiliation(s)
- Jiannan Zhou
- Department of Human Microbiome, School and Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Tianyong Sun
- Department of Human Microbiome, School and Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Wenyan Kang
- Department of Human Microbiome, School and Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Di Tang
- Department of Human Microbiome, School and Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Qiang Feng
- Department of Human Microbiome, School and Hospital of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China; State Key Laboratory of Microbial Technology, Shandong University,266237 Qingdao, Shandong, China; NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, Shandong, China.
| |
Collapse
|
17
|
Abril AG, Carrera M, Böhme K, Barros-Velázquez J, Rama JLR, Calo-Mata P, Sánchez-Pérez A, Villa TG. Proteomic Characterization of Antibiotic Resistance, and Production of Antimicrobial and Virulence Factors in Streptococcus Species Associated with Bovine Mastitis. Could Enzybiotics Represent Novel Therapeutic Agents Against These Pathogens? Antibiotics (Basel) 2020; 9:antibiotics9060302. [PMID: 32512932 PMCID: PMC7344566 DOI: 10.3390/antibiotics9060302] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/28/2020] [Accepted: 06/03/2020] [Indexed: 01/31/2023] Open
Abstract
Streptococcus spp. are major mastitis pathogens present in dairy products, which produce a variety of virulence factors that are involved in streptococcal pathogenicity. These include neuraminidase, pyrogenic exotoxin, and M protein, and in addition they might produce bacteriocins and antibiotic-resistance proteins. Unjustifiable misuse of antimicrobials has led to an increase in antibiotic-resistant bacteria present in foodstuffs. Identification of the mastitis-causing bacterial strain, as well as determining its antibiotic resistance and sensitivity is crucial for effective therapy. The present work focused on the LC–ESI–MS/MS (liquid chromatography–electrospray ionization tandem mass spectrometry) analysis of tryptic digestion peptides from mastitis-causing Streptococcus spp. isolated from milk. A total of 2706 non-redundant peptides belonging to 2510 proteins was identified and analyzed. Among them, 168 peptides were determined, representing proteins that act as virulence factors, toxins, anti-toxins, provide resistance to antibiotics that are associated with the production of lantibiotic-related compounds, or play a role in the resistance to toxic substances. Protein comparisons with the NCBI database allowed the identification of 134 peptides as specific to Streptococcus spp., while two peptides (EATGNQNISPNLTISNAQLNLEDKNK and DLWC*NM*IIAAK) were found to be species-specific to Streptococcus dysgalactiae. This proteomic repository might be useful for further studies and research work, as well as for the development of new therapeutics for the mastitis-causing Streptococcus strains.
Collapse
Affiliation(s)
- Ana G. Abril
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Campus Sur 15782, Universidad de Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.G.A.); (J.-L.R.R.)
| | - Mónica Carrera
- Marine Research Institute (IIM), Spanish National Research Council (CSIC), Eduardo Cabello 6, 36208 Vigo, Pontevedra, Spain;
| | - Karola Böhme
- Agroalimentary Technological Center of Lugo, Montirón 154, 27002 Lugo, Spain;
| | - Jorge Barros-Velázquez
- Departamento de Química Analítica, Nutrición y Bromatología, Area de Tecnología de los Alimentos, Facultad de Veterinaria, Campus Lugo, Universidad de Santiago de Compostela, 27002 Lugo, Spain; (J.B.-V.); (P.C.-M.)
| | - José-Luis R. Rama
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Campus Sur 15782, Universidad de Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.G.A.); (J.-L.R.R.)
| | - Pilar Calo-Mata
- Departamento de Química Analítica, Nutrición y Bromatología, Area de Tecnología de los Alimentos, Facultad de Veterinaria, Campus Lugo, Universidad de Santiago de Compostela, 27002 Lugo, Spain; (J.B.-V.); (P.C.-M.)
| | - Angeles Sánchez-Pérez
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia;
| | - Tomás G. Villa
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Campus Sur 15782, Universidad de Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.G.A.); (J.-L.R.R.)
- Correspondence:
| |
Collapse
|
18
|
Yan T, Tang X, Sun L, Tian R, Li Z, Liu G. Co infection of respiratory syncytial viruses (RSV) and streptococcus pneumonia modulates pathogenesis and dependent of serotype and phase variant. Microb Pathog 2020; 144:104126. [PMID: 32173494 DOI: 10.1016/j.micpath.2020.104126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 11/17/2022]
Abstract
Streptococcus pneumoniae (pneumococcus) is touted to be the generally found pathogen in patients with respiratory issues and there is an epidemiologic linkage present between Respiratory syncytial virus (RSV). This study aim at investigating the interaction between RSV and two serotypes of S. pneumoniae using a distinct animal model and a well-established colonizing pneumococcal strain. Phase variants phenotype of each strain was determined under oblique light. Co infection model was developed using BALB/c mice housed in a BSL-2 facility. Coinfection experiments were performed and number of bacterial colonies was quantified and phase determination was evaluated. RSV was detected in sample through real-time quantitative PCR. Adherence assays were performed to determine adherence of Spn strains and its knock out ΔNanA to nasopharyngeal carcinoma (NPC) epithelial CNE3 cell line. The biofilm viability was determined and phase composition was counted using plate count. Neuraminidase activity was measured in fluorometircassessed using 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (MUAN) as substrate as described in earlier literature. The GraphPad Software version 5.01 i.e., GraphPad Prism was used to conduct the statistical analysis. The extent of bacterial colonization was increased significantly (p < 0.05), when the mice were co infected. Nasal epithelium remained intact in mock sample with features of a thick mucociliary border. A small percentage of pneumococci exhibit phase variation between opaque phase and transparent phase. The percentage adherent of both phase were not found to be varying significantly within serotype but it was seen that nonpathogenic type 27 was more adherent. Biofilm formation was selectively more for transparent phase from a mixed-phase inoculum. Adherence of both phase variant of S. pneumoniae to nasopharyngeal epithelial cells 2 h post infection expressed as the percentage of adherent bacteria relative to the inoculum. In absence of viral infection, the nasal colonization of the opaque and the transparent variant was increased many folds, which was a significant differences. The extent of nasal colonization by the ΔNanA mutant strain were significantly reduced post-bacterial infection for both type of wild-type (P < 0.05). The findings explore insights into the interactions occurring between S. pneumoniae and RSV during respiratory infections and pneumococcal acquisition, indicate that pneumococcal serotypes have different ability to cause infection as well as co infections and potentially follow an unappreciated mechanism. Much more research work is needed to further understand the minutiae of this interaction within co-infection process.
Collapse
Affiliation(s)
- Tingfei Yan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaoyan Tang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Lei Sun
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Runhua Tian
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Zhiming Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Guangzhen Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China.
| |
Collapse
|
19
|
Jensen CS, Norsigian CJ, Fang X, Nielsen XC, Christensen JJ, Palsson BO, Monk JM. Reconstruction and Validation of a Genome-Scale Metabolic Model of Streptococcus oralis (iCJ415), a Human Commensal and Opportunistic Pathogen. Front Genet 2020; 11:116. [PMID: 32194617 PMCID: PMC7063969 DOI: 10.3389/fgene.2020.00116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/31/2020] [Indexed: 11/22/2022] Open
Abstract
The mitis group of streptococci (MGS) is a member of the healthy human microbiome in the oral cavity and upper respiratory tract. Troublingly, some MGS are able to escape this niche and cause infective endocarditis, a severe and devastating disease. Genome-scale models have been shown to be valuable in investigating metabolism of bacteria. Here we present the first genome-scale model, iCJ415, for Streptococcus oralis SK141. We validated the model using gene essentiality and amino acid auxotrophy data from closely related species. iCJ415 has 71-76% accuracy in predicting gene essentiality and 85% accuracy in predicting amino acid auxotrophy. Further, the phenotype of S. oralis was tested using the Biolog Phenotype microarrays, giving iCJ415 a 82% accuracy in predicting carbon sources. iCJ415 can be used to explore the metabolic differences within the MGS, and to explore the complicated metabolic interactions between different species in the human oral cavity.
Collapse
Affiliation(s)
- Christian S Jensen
- The Regional Department of Clinical Microbiology, Region Zealand, Slagelse, Denmark
| | - Charles J Norsigian
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Xin Fang
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Xiaohui C Nielsen
- The Regional Department of Clinical Microbiology, Region Zealand, Slagelse, Denmark
| | - Jens Jørgen Christensen
- The Regional Department of Clinical Microbiology, Region Zealand, Slagelse, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Jonathan M Monk
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
20
|
Relationship Between Prevalence of Pneumococcal Serotypes and Their Neuraminidases in Carriers, Predictive Facts? ARCHIVES OF PEDIATRIC INFECTIOUS DISEASES 2020. [DOI: 10.5812/pedinfect.14100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
21
|
Briles DE, Paton JC, Mukerji R, Swiatlo E, Crain MJ. Pneumococcal Vaccines. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0028-2018. [PMID: 31858954 PMCID: PMC10921951 DOI: 10.1128/microbiolspec.gpp3-0028-2018] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Indexed: 01/14/2023] Open
Abstract
Streptococcus pneumoniae is a Gram-Positive pathogen that is a major causative agent of pneumonia, otitis media, sepsis and meningitis across the world. The World Health Organization estimates that globally over 500,000 children are killed each year by this pathogen. Vaccines offer the best protection against S. pneumoniae infections. The current polysaccharide conjugate vaccines have been very effective in reducing rates of invasive pneumococcal disease caused by vaccine type strains. However, the effectiveness of these vaccines have been somewhat diminished by the increasing numbers of cases of invasive disease caused by non-vaccine type strains, a phenomenon known as serotype replacement. Since, there are currently at least 98 known serotypes of S. pneumoniae, it may become cumbersome and expensive to add many additional serotypes to the current 13-valent vaccine, to circumvent the effect of serotype replacement. Hence, alternative serotype independent strategies, such as vaccination with highly cross-reactive pneumococcal protein antigens, should continue to be investigated to address this problem. This chapter provides a comprehensive discussion of pneumococcal vaccines past and present, protein antigens that are currently under investigation as vaccine candidates, and other alternatives, such as the pneumococcal whole cell vaccine, that may be successful in reducing current rates of disease caused by S. pneumoniae.
Collapse
Affiliation(s)
- D E Briles
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - J C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, 5005, Australia
| | - R Mukerji
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - E Swiatlo
- Section of Infectious Diseases, Southeast Louisiana Veterans Health Care System, New Orleans, LA
| | - M J Crain
- Department of Pediatrics and Microbiology, University of Alabama at Birmingham
| |
Collapse
|
22
|
Bule P, Chuzel L, Blagova E, Wu L, Gray MA, Henrissat B, Rapp E, Bertozzi CR, Taron CH, Davies GJ. Inverting family GH156 sialidases define an unusual catalytic motif for glycosidase action. Nat Commun 2019; 10:4816. [PMID: 31645552 PMCID: PMC6811678 DOI: 10.1038/s41467-019-12684-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Sialic acids are a family of related sugars that play essential roles in many biological events intimately linked to cellular recognition in both health and disease. Sialidases are therefore orchestrators of cellular biology and important therapeutic targets for viral infection. Here, we sought to define if uncharacterized sialidases would provide distinct paradigms in sialic acid biochemistry. We show that a recently discovered sialidase family, whose first member EnvSia156 was isolated from hot spring metagenomes, defines an unusual structural fold and active centre constellation, not previously described in sialidases. Consistent with an inverting mechanism, EnvSia156 reveals a His/Asp active center in which the His acts as a Brønsted acid and Asp as a Brønsted base in a single-displacement mechanism. A predominantly hydrophobic aglycone site facilitates accommodation of a variety of 2-linked sialosides; a versatility that offers the potential for glycan hydrolysis across a range of biological and technological platforms.
Collapse
Affiliation(s)
- Pedro Bule
- Department of Chemistry, University of York, York, YO10 5DD, UK
| | - Léa Chuzel
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Elena Blagova
- Department of Chemistry, University of York, York, YO10 5DD, UK
| | - Liang Wu
- Department of Chemistry, University of York, York, YO10 5DD, UK
| | - Melissa A Gray
- Department of Chemistry, Stanford University, Stanford, CA, 94305-4404, USA
| | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques (AFMB), Centre National de la Recherche Scientifique (CNRS, UMR7257), Institut National Agronomique (INRA, USC 1408) and Aix-Marseille Université (AMU), 13288 Marseille cedex 9, Marseille, France
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106, Magdeburg, Germany
- glyXera GmbH, Leipziger Strasse 44-ZENIT, Magdeburg, Germany
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, 94305-4404, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305-4404, USA
| | | | - Gideon J Davies
- Department of Chemistry, University of York, York, YO10 5DD, UK.
| |
Collapse
|
23
|
Abstract
Many important interactions between bacterial pathogens and their hosts are highly specific binding events that involve host or pathogen carbohydrate structures (glycans). Glycan interactions can mediate adhesion, invasion and immune evasion and can act as receptors for toxins. Several bacterial pathogens can also enzymatically alter host glycans to reveal binding targets, degrade the host cell glycans or alter the function of host glycoproteins. In recent years, high-throughput screening technologies, such as lectin, glycan and mucin microarrays, have transformed the field by identifying new bacterial-host glycointeractions, which are crucial for colonization, persistence and disease. In this Review, we discuss interactions involving both host and bacterial glycans that have a role in bacterial pathogenesis. We also highlight recent technological advances that have illuminated the glycoscience of microbial pathogenesis.
Collapse
|
24
|
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: 14] [Impact Index Per Article: 2.3] [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
|
25
|
Xiao K, Wang X, Yu H. Comparative studies of catalytic pathways for Streptococcus pneumoniae sialidases NanA, NanB and NanC. Sci Rep 2019; 9:2157. [PMID: 30770840 PMCID: PMC6377674 DOI: 10.1038/s41598-018-38131-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/14/2018] [Indexed: 11/09/2022] Open
Abstract
Streptococcus pneumoniae (S. pneumoniae) is a leading human pathogen, which takes large responsibility for severe otitis media, acute meningitis and septicaemia. It encodes up to three distinct sialidases: NanA, NanB and NanC, which are promising drug targets. Recent experimental studies have shown that these three sialidases might work together up to the ultimate step, where NanA and NanB produce N-acetylneuraminic acid (Neu5Ac) and 2,7-anhydro-Neu5Ac following the functions of sialidase and intramolecular trans-sialidase, whilst NanC carries on a ping-pong mechanism that produces or removes 2-deoxy-2,3-didehydro-Neu5AC. It is intriguing that these sialidases have similar active sites but operate via three distinct reaction pathways. To clarify this issue, herein we present the first systematic computational investigation on the catalytic pathways for S. pneumoniae NanA, NanB and NanC based on combined quantum mechanics/molecular mechanics simulations, and propose the most preferred routes for the three S. pneumoniae sialidases. Our findings support the mechanisms of NanA and NanC that were proposed by previous experimental studies, whereas the role of water in NanB was found to differ slightly from our current understandings. The mechanistic insights obtained from this work are expected to assist in the design of potent inhibitors targeting these key enzymes for therapeutic applications.
Collapse
Affiliation(s)
- Kela Xiao
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2500, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW, 2500, Australia
| | - Xingyong Wang
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2500, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW, 2500, Australia
| | - Haibo Yu
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2500, Australia. .,Molecular Horizons, University of Wollongong, Wollongong, NSW, 2500, Australia. .,Illawarra Health and Medical Research Institute, Wollongong, NSW, 2500, Australia.
| |
Collapse
|
26
|
Abstract
S. pneumoniae is a component of the commensal nasopharyngeal microflora of humans, but from this reservoir, it can progress to localized or invasive disease with a frequency that translates into massive global morbidity and mortality. However, the factors that govern the switch from commensal to pathogen, as well as those that determine disease tropism, are poorly understood. Here we show that capacity to utilize raffinose can determine the nature of the disease caused by a given pneumococcal strain. Moreover, our findings provide an interesting example of convergent evolution, whereby pneumococci belonging to two unrelated serotypes/lineages exhibit SNPs in separate genes affecting raffinose uptake and utilization that correlate with distinct pathogenic profiles in vivo. This further underscores the critical role of differential carbohydrate metabolism in the pathogenesis of localized versus invasive pneumococcal disease. Streptococcus pneumoniae is commonly carried asymptomatically in the human nasopharynx, but it also causes serious and invasive diseases such as pneumonia, bacteremia, and meningitis, as well as less serious but highly prevalent infections such as otitis media. We have previously shown that closely related pneumococci (of the same capsular serotype and multilocus sequence type [ST]) can display distinct pathogenic profiles in mice that correlate with clinical isolation site (e.g., blood versus ear), suggesting stable niche adaptation within a clonal lineage. This has provided an opportunity to identify determinants of disease tropism. Genomic analysis identified 17 and 27 single nucleotide polymorphisms (SNPs) or insertions/deletions in protein coding sequences between blood and ear isolates of serotype 14 ST15 and serotype 3 ST180, respectively. SNPs in raffinose uptake and utilization genes (rafR or rafK) were detected in both serotypes/lineages. Ear isolates were consistently defective in growth in media containing raffinose as the sole carbon source, as well as in expression of raffinose pathway genes aga, rafG, and rafK, relative to their serotype/ST-matched blood isolates. Similar differences were also seen between serotype 23F ST81 blood and ear isolates. Analysis of rafR allelic exchange mutants of the serotype 14 ST15 blood and ear isolates demonstrated that the SNP in rafR was entirely responsible for their distinct in vitro phenotypes and was also the determinant of differential tropism for the lungs versus ear and brain in a mouse intranasal challenge model. These data suggest that the ability of pneumococci to utilize raffinose determines the nature of disease.
Collapse
|
27
|
Sharapova YA, Švedas VK. Molecular Modeling of the Binding of the Allosteric Inhibitor Optactin at a New Binding Site in Neuraminidase A from Streptococcus pneumoniae. ACTA ACUST UNITED AC 2018. [DOI: 10.3103/s0027131418050097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Slack TJ, Li W, Shi D, McArthur JB, Zhao G, Li Y, Xiao A, Khedri Z, Yu H, Liu Y, Chen X. Triazole-linked transition state analogs as selective inhibitors against V. cholerae sialidase. Bioorg Med Chem 2018; 26:5751-5757. [PMID: 30389408 PMCID: PMC6326775 DOI: 10.1016/j.bmc.2018.10.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/15/2018] [Accepted: 10/26/2018] [Indexed: 01/07/2023]
Abstract
Sialidases or neuraminidases are enzymes that catalyze the cleavage of terminal sialic acids from oligosaccharides and glycoconjugates. They play important roles in bacterial and viral infection and have been attractive targets for drug development. Structure-based drug design has led to potent inhibitors against neuraminidases of influenza A viruses that have been used successfully as approved therapeutics. However, selective and effective inhibitors against bacterial and human sialidases are still being actively pursued. Guided by crystal structural analysis, several derivatives of 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (Neu5Ac2en or DANA) were designed and synthesized as triazole-linked transition state analogs. Inhibition studies revealed that glycopeptide analog E-(TriazoleNeu5Ac2en)-AKE and compound (TriazoleNeu5Ac2en)-A were selective inhibitors against Vibrio cholerae sialidase, while glycopeptide analog (TriazoleNeu5Ac2en)-AdE selectively inhibited Vibrio cholerae and A. ureafaciens sialidases.
Collapse
Affiliation(s)
- Teri J. Slack
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA,These authors contributed equality to this work
| | - Wanqing Li
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA,These authors contributed equality to this work
| | - Dashuang Shi
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave, NW, Washington DC 20012, USA
| | - John B. McArthur
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Gengxiang Zhao
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave, NW, Washington DC 20012, USA
| | - Yanhong Li
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - An Xiao
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Zahra Khedri
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Hai Yu
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Yang Liu
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Ave, NW, Washington DC 20012, USA
| | - Xi Chen
- Department of Chemistry, University of California-Davis, One Shields Avenue, Davis, CA 95616, USA,Corresponding author. Tel: +1 530 754 6037; fax: +1 530 752 8995. (X. Chen)
| |
Collapse
|
29
|
The Role of Pneumococcal Virulence Factors in Ocular Infectious Diseases. Interdiscip Perspect Infect Dis 2018; 2018:2525173. [PMID: 30538741 PMCID: PMC6257906 DOI: 10.1155/2018/2525173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/07/2018] [Indexed: 12/24/2022] Open
Abstract
Streptococcus pneumoniae is a gram-positive, facultatively anaerobic pathogen that can cause severe infections such as pneumonia, meningitis, septicemia, and middle ear infections. It is also one of the top pathogens contributing to bacterial keratitis and conjunctivitis. Though two pneumococcal vaccines exist for the prevention of nonocular diseases, they do little to fully prevent ocular infections. This pathogen has several virulence factors that wreak havoc on the conjunctiva, cornea, and intraocular system. Polysaccharide capsule aids in the evasion of host complement system. Pneumolysin (PLY) is a cholesterol-dependent cytolysin that acts as pore-forming toxin. Neuraminidases assist in adherence and colonization by exposing cell surface receptors to the pneumococcus. Zinc metalloproteinases contribute to evasion of the immune system and disease severity. The main purpose of this review is to consolidate the multiple studies that have been conducted on several pneumococcal virulence factors and the role each plays in conjunctivitis, keratitis, and endophthalmitis.
Collapse
|
30
|
Wang J, Song M, Pan J, Shen X, Liu W, Zhang X, Li H, Deng X. Quercetin impairs Streptococcus pneumoniae biofilm formation by inhibiting sortase A activity. J Cell Mol Med 2018; 22:6228-6237. [PMID: 30334338 PMCID: PMC6237587 DOI: 10.1111/jcmm.13910] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/20/2018] [Indexed: 02/03/2023] Open
Abstract
Biofilm formation mediated by sortase A (srtA) is important for bacterial colonisation and resistance to antibiotics. Thus, the inhibitor of SrtA may represent a promising agent for bacterial infection. The structure of Streptococcus pneumoniae D39 srtA has been characterised by crystallisation. Site‐directed mutagenesis was used for the determination of the key residues for the activity of S. pneumoniae D39 srtA. An effective srtA inhibitor, quercetin, and its mechanism was further identified using srtA activity inhibition assay and molecular modelling. In this study, the crystal structure of S. pneumoniae D39 srtA has been solved and shown to contain a unique domain B. Additionally, its transpeptidase activity was evaluated in vitro. Based on the structure, we identified Cys207 as the catalytic residue, with His141 and Arg215 serving as binding sites for the peptide substrate. We found that quercetin can specifically compete with the natural substrate, leading to a significant decrease in the catalytic activity of this enzyme. In cells co‐cultured with this small molecule inhibitor, NanA cannot anchor to the cell wall effectively, and biofilm formation and biomass decrease significantly. Interestingly, when we supplemented cultures with sialic acid, a crucial signal for pneumococcal coloniation and the invasion of the host in the co‐culture system, biofilm loss did not occur. This result indicates that quercetin inhibits biofilm formation by affecting sialic acid production. In conclusion, the inhibition of pneumococcal srtA by the small molecule quercetin offers a novel strategy for pneumococcal preventative therapy.
Collapse
Affiliation(s)
- Jianfeng Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Meng Song
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Juan Pan
- Tianjin International Travel Healthcare Center, Tianjin, China
| | - Xue Shen
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wentao Liu
- Heilongjiang Veterinary Drug and Feed Super Vision Institute, Haerbin, China
| | - Xueke Zhang
- Heilongjiang Veterinary Drug and Feed Super Vision Institute, Haerbin, China
| | - Hongen Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
31
|
Xiong J, Zhang C, Xu D. Catalytic mechanism of type C sialidase from Streptococcus pneumoniae: from covalent intermediate to final product. J Mol Model 2018; 24:297. [PMID: 30259133 DOI: 10.1007/s00894-018-3822-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 09/04/2018] [Indexed: 12/24/2022]
Abstract
Streptococcus pneumoniae is a Gram-positive human pathogenic bacterium, which is the main cause of pneumonia and meningitis in children and the elderly. Three sialidases (or neuraminidases) encoded from Streptococcus pneumoniae could catalyze the cleavage of sialic acid linkages. This mechanism is directly connected with infection, apoptosis, and signaling, and usually considered to be one of the critical virulence factors. Type C neuraminidase (NanC) is unique because its primary product of Neu5Ac2en is considered to be an inhibitor to the other two sialidases. Experimentally, there are two different pathways for the formation mechanism of Neu5Ac2en catalyzed by NanC. In this work, a combined quantum mechanical and molecular mechanical approach was employed in all calculations. Starting from the covalent sialylated intermediate, we first examined the reaction to Neu5Ac2en and found the reaction prefers a direct proton abstraction mechanism rather than the water mediated proton abstraction mechanism. Free energy profiles can confirm that Neu5Ac2en is the major product of NanC. Functional roles of some important residues were also investigated, e.g., D315 acts as the proton acceptor during the formation of Neu5Ac2en, while the general base for the hydrolytic reaction to Neu5Ac. This study can facilitate the understanding of the catalytic mechanism of NanC and has the potential to aid in future inhibitor design studies.
Collapse
Affiliation(s)
- Jing Xiong
- MOE Key Laboratory of Green Chemistry & Technology, College of Chemistry, Sichuan University, Chengdu, Sichuan, 610064, People's Republic of China
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan, 610500, People's Republic of China
| | - Chunchun Zhang
- Analytical&Testing Center, Sichuan University, Chengdu, Sichuan, 610064, People's Republic of China.
| | - Dingguo Xu
- MOE Key Laboratory of Green Chemistry & Technology, College of Chemistry, Sichuan University, Chengdu, Sichuan, 610064, People's Republic of China.
| |
Collapse
|
32
|
Xiao A, Slack TJ, Li Y, Shi D, Yu H, Li W, Liu Y, Chen X. Streptococcus pneumoniae Sialidase SpNanB-Catalyzed One-Pot Multienzyme (OPME) Synthesis of 2,7-Anhydro-Sialic Acids as Selective Sialidase Inhibitors. J Org Chem 2018; 83:10798-10804. [PMID: 30105908 DOI: 10.1021/acs.joc.8b01519] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Streptococcus pneumoniae sialidase SpNanB is an intramolecular trans-sialidase (IT-sialidase) and a virulence factor that is essential for streptococcal infection of the upper and lower respiratory tract. SpNanB catalyzes the formation of 2,7-anhydro- N-acetylneuraminic acid (2,7-anhydro-Neu5Ac), a potential prebiotic that can be used as the sole carbon source of a common human gut commensal anaerobic bacterium. We report here the development of an efficient one-pot multienzyme (OPME) system for synthesizing 2,7-anhydro-Neu5Ac and its derivatives. Based on a crystal structure analysis, an N-cyclohexyl derivative of 2,7-anhydro-neuraminic acid was designed, synthesized, and shown to be a selective inhibitor against SpNanB and another Streptococcus pneumoniae sialidase SpNanC. This study demonstrates a new strategy of synthesizing 2,7-anhydro-sialic acids in a gram scale and the potential application of their derivatives as selective sialidase inhibitors.
Collapse
Affiliation(s)
- An Xiao
- Department of Chemistry , University of California , One Shields Avenue , Davis , California 95616 , United States
| | - Teri J Slack
- Department of Chemistry , University of California , One Shields Avenue , Davis , California 95616 , United States
| | - Yanhong Li
- Department of Chemistry , University of California , One Shields Avenue , Davis , California 95616 , United States
| | - Dashuang Shi
- Children's National Medical Center , 111 Michigan Ave , NW, Washington, DC 20012 , United States
| | - Hai Yu
- Department of Chemistry , University of California , One Shields Avenue , Davis , California 95616 , United States
| | - Wanqing Li
- Department of Chemistry , University of California , One Shields Avenue , Davis , California 95616 , United States
| | - Yang Liu
- Children's National Medical Center , 111 Michigan Ave , NW, Washington, DC 20012 , United States
| | - Xi Chen
- Department of Chemistry , University of California , One Shields Avenue , Davis , California 95616 , United States
| |
Collapse
|
33
|
Miao X, He J, Zhang L, Zhao X, Ge R, He QY, Sun X. A Novel Iron Transporter SPD_1590 in Streptococcus pneumoniae Contributing to Bacterial Virulence Properties. Front Microbiol 2018; 9:1624. [PMID: 30079056 PMCID: PMC6062600 DOI: 10.3389/fmicb.2018.01624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/28/2018] [Indexed: 12/27/2022] Open
Abstract
Streptococcus pneumoniae, a Gram-positive human pathogen, has evolved three main transporters for iron acquisition from the host: PiaABC, PiuABC, and PitABC. Our previous study had shown that the mRNA and protein levels of SPD_1590 are significantly upregulated in the ΔpiuA/ΔpiaA/ΔpitA triple mutant, suggesting that SPD_1590 might be a novel iron transporter in S. pneumoniae. In the present study, using spd1590-knockout, -complemented, and -overexpressing strains and the purified SPD_1590 protein, we show that SPD_1590 can bind hemin, probably supplementing the function of PiuABC, to provide the iron necessary for the bacterium. Furthermore, the results of iTRAQ quantitative proteomics and cell-infection studies demonstrate that, similarly to other metal-ion uptake proteins, SPD_1590 is important for bacterial virulence properties. Overall, these results provide a better understanding of the biology of this clinically important bacterium.
Collapse
Affiliation(s)
- Xinyu Miao
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jiaojiao He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Liang Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xinlu Zhao
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ruiguang Ge
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, College of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xuesong Sun
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
34
|
Houston S, Lithgow KV, Osbak KK, Kenyon CR, Cameron CE. Functional insights from proteome-wide structural modeling of Treponema pallidum subspecies pallidum, the causative agent of syphilis. BMC STRUCTURAL BIOLOGY 2018; 18:7. [PMID: 29769048 PMCID: PMC5956850 DOI: 10.1186/s12900-018-0086-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/27/2018] [Indexed: 12/21/2022]
Abstract
Background Syphilis continues to be a major global health threat with 11 million new infections each year, and a global burden of 36 million cases. The causative agent of syphilis, Treponema pallidum subspecies pallidum, is a highly virulent bacterium, however the molecular mechanisms underlying T. pallidum pathogenesis remain to be definitively identified. This is due to the fact that T. pallidum is currently uncultivatable, inherently fragile and thus difficult to work with, and phylogenetically distinct with no conventional virulence factor homologs found in other pathogens. In fact, approximately 30% of its predicted protein-coding genes have no known orthologs or assigned functions. Here we employed a structural bioinformatics approach using Phyre2-based tertiary structure modeling to improve our understanding of T. pallidum protein function on a proteome-wide scale. Results Phyre2-based tertiary structure modeling generated high-confidence predictions for 80% of the T. pallidum proteome (780/978 predicted proteins). Tertiary structure modeling also inferred the same function as primary structure-based annotations from genome sequencing pipelines for 525/605 proteins (87%), which represents 54% (525/978) of all T. pallidum proteins. Of the 175 T. pallidum proteins modeled with high confidence that were not assigned functions in the previously annotated published proteome, 167 (95%) were able to be assigned predicted functions. Twenty-one of the 175 hypothetical proteins modeled with high confidence were also predicted to exhibit significant structural similarity with proteins experimentally confirmed to be required for virulence in other pathogens. Conclusions Phyre2-based structural modeling is a powerful bioinformatics tool that has provided insight into the potential structure and function of the majority of T. pallidum proteins and helped validate the primary structure-based annotation of more than 50% of all T. pallidum proteins with high confidence. This work represents the first T. pallidum proteome-wide structural modeling study and is one of few studies to apply this approach for the functional annotation of a whole proteome. Electronic supplementary material The online version of this article (10.1186/s12900-018-0086-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simon Houston
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Karen Vivien Lithgow
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | | | - Chris Richard Kenyon
- HIV/STI Unit, Institute of Tropical Medicine, Antwerp, Belgium.,Division of Infectious Diseases and HIV Medicine, University of Cape Town, Cape Town, South Africa
| | - Caroline E Cameron
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
35
|
Sharapova Y, Suplatov D, Švedas V. Neuraminidase A from Streptococcus pneumoniae has a modular organization of catalytic and lectin domains separated by a flexible linker. FEBS J 2018; 285:2428-2445. [PMID: 29704878 DOI: 10.1111/febs.14486] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/01/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022]
Abstract
Neuraminidase A (NanA) of the pathogen Streptococcus pneumoniae cleaves receptors of the human respiratory epithelial surface during bacterial colonization. The full-size structure of NanA that contains one lectin and one catalytic domain within a single polypeptide chain remains unresolved. Both domains are crucial for the microorganism's virulence and considered as promising antimicrobial targets. Methods of bioinformatics and molecular dynamics have been implemented to model NanA's structure and study interaction between the lectin and catalytic domains in three neuraminidases NanA, NanB, and NanC from Streptococcus pneumoniae. A significant difference in spatial organization of these homologous enzymes has been revealed. The lectin and catalytic domains of NanB and NanC form rigid globules stabilized by multiple interdomain interactions, whereas in NanA, the two domains are separated by a 16 amino acids long flexible linker - a characteristic of proteins that require conformational flexibility for their functioning. The biological role of this structural adaptation of NanA as a key virulence enzyme is discussed.
Collapse
Affiliation(s)
- Yana Sharapova
- Faculty of Bioengineering and Bioinformatics, Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Russia
| | - Dmitry Suplatov
- Faculty of Bioengineering and Bioinformatics, Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Russia
| | - Vytas Švedas
- Faculty of Bioengineering and Bioinformatics, Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Russia
| |
Collapse
|
36
|
Ercoli G, Fernandes VE, Chung WY, Wanford JJ, Thomson S, Bayliss CD, Straatman K, Crocker PR, Dennison A, Martinez-Pomares L, Andrew PW, Moxon ER, Oggioni MR. Intracellular replication of Streptococcus pneumoniae inside splenic macrophages serves as a reservoir for septicaemia. Nat Microbiol 2018; 3:600-610. [PMID: 29662129 PMCID: PMC6207342 DOI: 10.1038/s41564-018-0147-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 03/08/2018] [Indexed: 01/21/2023]
Abstract
Bacterial septicaemia is a major cause of mortality, but its pathogenesis remains poorly understood. In experimental pneumococcal murine intravenous infection, an initial reduction of bacteria in the blood is followed hours later by a fatal septicaemia. These events represent a population bottleneck driven by efficient clearance of pneumococci by splenic macrophages and neutrophils, but as we show in this study, accompanied by occasional intracellular replication of bacteria that are taken up by a subset of CD169+ splenic macrophages. In this model, proliferation of these sequestered bacteria provides a reservoir for dissemination of pneumococci into the bloodstream, as demonstrated by its prevention using an anti-CD169 monoclonal antibody treatment. Intracellular replication of pneumococci within CD169+ splenic macrophages was also observed in an ex vivo porcine spleen, where the microanatomy is comparable with humans. We also showed that macrolides, which effectively penetrate macrophages, prevented septicaemia, whereas beta-lactams, with inefficient intracellular penetration, failed to prevent dissemination to the blood. Our findings define a shift in our understanding of the pneumococcus from an exclusively extracellular pathogen to one with an intracellular phase. These findings open the door to the development of treatments that target this early, previously unrecognized intracellular phase of bacterial sepsis.
Collapse
Affiliation(s)
- Giuseppe Ercoli
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Vitor E Fernandes
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Wen Y Chung
- Hepato-Pancreato-Biliary Unit, Leicester General Hospital, University of Hospitals of Leicester, NHS Trust, Leicester, UK
| | - Joseph J Wanford
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Sarah Thomson
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | | | - Kornelis Straatman
- Centre for Core Biotechnology Services, University of Leicester, Leicester, UK
| | - Paul R Crocker
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Ashley Dennison
- Hepato-Pancreato-Biliary Unit, Leicester General Hospital, University of Hospitals of Leicester, NHS Trust, Leicester, UK
| | - Luisa Martinez-Pomares
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, UK
| | - Peter W Andrew
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | | | - Marco R Oggioni
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Nasher F, Förster S, Yildirim EC, Grandgirard D, Leib SL, Heller M, Hathaway LJ. Foreign peptide triggers boost in pneumococcal metabolism and growth. BMC Microbiol 2018; 18:23. [PMID: 29580217 PMCID: PMC5870813 DOI: 10.1186/s12866-018-1167-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 03/15/2018] [Indexed: 01/19/2023] Open
Abstract
Background Nonencapsulated Streptococcus pneumoniae bacteria are successful colonizers of the human nasopharynx and often possess genes aliB-like ORF 1 and 2 in place of capsule genes. AliB-like ORF 2 binds peptide FPPQSV, found in Prevotella species, resulting in enhanced colonization. How this response is mediated is so far unknown. Results Here we show that the peptide increases expression of genes involved in release of host carbohydrates, carbohydrate uptake and carbohydrate metabolism. In particular, the peptide increased expression of 1,5-anhydro-D-fructose reductase, a metabolic enzyme of an alternative starch and glycogen degrading pathway found in many organisms, in both transcriptomic and proteomic data. The peptide enhanced pneumococcal growth giving a competitive advantage to a strain with aliB-like ORF 2, over its mutant lacking the gene. Possession of aliB-like ORF 2 did not affect release of inflammatory cytokine CXCL8 from epithelial cells in culture and the nonencapsulated wild type strain was not able to establish disease or inflammation in an infant rat model of meningitis. Conclusions We propose that AliB-like ORF 2 confers an advantage in colonization by enhancing carbohydrate metabolism resulting in a boost in growth. This may explain the widespread presence of aliB-like ORF 2 in the nonencapsulated pneumococcal population in the human nasopharynx. Electronic supplementary material The online version of this article (10.1186/s12866-018-1167-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fauzy Nasher
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Friedbühlstrasse 51, CH-3001, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sunniva Förster
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Friedbühlstrasse 51, CH-3001, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland.,Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Efe C Yildirim
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Friedbühlstrasse 51, CH-3001, Bern, Switzerland
| | - Denis Grandgirard
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Friedbühlstrasse 51, CH-3001, Bern, Switzerland
| | - Stephen L Leib
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Friedbühlstrasse 51, CH-3001, Bern, Switzerland
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, CH-3010, Bern, Switzerland
| | - Lucy J Hathaway
- Institute for Infectious Diseases, Faculty of Medicine, University of Bern, Friedbühlstrasse 51, CH-3001, Bern, Switzerland.
| |
Collapse
|
40
|
Evidence for a carbohydrate-binding module (CBM) of Tannerella forsythia NanH sialidase, key to interactions at the host–pathogen interface. Biochem J 2018; 475:1159-1176. [DOI: 10.1042/bcj20170592] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/20/2018] [Accepted: 02/26/2018] [Indexed: 12/15/2022]
Abstract
Bacterial sialidases cleave terminal sialic acid from a variety of host glycoproteins, and contribute to survival and growth of many human-dwelling bacterial species, including various pathogens. Tannerella forsythia, an oral, Gram-negative, fastidious anaerobe, is a key organism in periodontal disease and possesses a dedicated sialic acid utilisation and scavenging (nan) operon, including NanH sialidase. Here, we describe biochemical characterisation of recombinant NanH, including its action on host-relevant sialoglycans such as sialyl Lewis A and sialyl Lewis X (SLeA/X), and on human cell-attached sialic acids directly, uncovering that it is a highly active broad specificity sialidase. Furthermore, the N-terminal domain of NanH was hypothesised and proved to be capable of binding to a range of sialoglycans and non-sialylated derivatives with Kd in the micromolar range, as determined by steady-state tryptophan fluorescence spectroscopy, but it has no catalytic activity in isolation from the active site. We consider this domain to represent the founding member of a novel subfamily of carbohydrate-binding module (CBM), involved in glycosidase-ligand binding. In addition, we created a catalytically inactive version of the NanH enzyme (FRIP → YMAP) that retained its ability to bind sialic acid-containing ligands and revealed for the first time that binding activity of a CBM is enhanced by association with the catalytic domain. Finally, we investigated the importance of Lewis-type sialoglycans on T. forsythia–host interactions, showing that nanomolar amounts of SLeA/X were capable of reducing invasion of oral epithelial cells by T. forsythia, suggesting that these are key ligands for bacterial–cellular interactions during periodontal disease.
Collapse
|
41
|
Yang WH, Heithoff DM, Aziz PV, Sperandio M, Nizet V, Mahan MJ, Marth JD. Recurrent infection progressively disables host protection against intestinal inflammation. Science 2018; 358:358/6370/eaao5610. [PMID: 29269445 DOI: 10.1126/science.aao5610] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/13/2017] [Indexed: 12/15/2022]
Abstract
Intestinal inflammation is the central pathological feature of colitis and the inflammatory bowel diseases. These syndromes arise from unidentified environmental factors. We found that recurrent nonlethal gastric infections of Gram-negative Salmonella enterica Typhimurium (ST), a major source of human food poisoning, caused inflammation of murine intestinal tissue, predominantly the colon, which persisted after pathogen clearance and irreversibly escalated in severity with repeated infections. ST progressively disabled a host mechanism of protection by inducing endogenous neuraminidase activity, which accelerated the molecular aging and clearance of intestinal alkaline phosphatase (IAP). Disease was linked to a Toll-like receptor 4 (TLR4)-dependent mechanism of IAP desialylation with accumulation of the IAP substrate and TLR4 ligand, lipopolysaccharide-phosphate. The administration of IAP or the antiviral neuraminidase inhibitor zanamivir was therapeutic by maintaining IAP abundance and function.
Collapse
Affiliation(s)
- Won Ho Yang
- Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Sanford Burnham Prebys Medical Discovery Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Douglas M Heithoff
- Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Peter V Aziz
- Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Sanford Burnham Prebys Medical Discovery Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Markus Sperandio
- Walter-Brendel-Centre for Experimental Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael J Mahan
- Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Jamey D Marth
- Center for Nanomedicine, University of California, Santa Barbara, Santa Barbara, CA 93106, USA. .,Sanford Burnham Prebys Medical Discovery Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| |
Collapse
|
42
|
Bitzan M, Zieg J. Influenza-associated thrombotic microangiopathies. Pediatr Nephrol 2018; 33:2009-2025. [PMID: 28884355 PMCID: PMC6153504 DOI: 10.1007/s00467-017-3783-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/11/2017] [Accepted: 08/08/2017] [Indexed: 12/26/2022]
Abstract
Thrombotic microangiopathy (TMA) refers to phenotypically similar disorders, including hemolytic uremic syndromes (HUS) and thrombotic thrombocytopenic purpura (TTP). This review explores the role of the influenza virus as trigger of HUS or TTP. We conducted a literature survey in PubMed and Google Scholar using HUS, TTP, TMA, and influenza as keywords, and extracted and analyzed reported epidemiological and clinical data. We identified 25 cases of influenza-associated TMA. Five additional cases were linked to influenza vaccination and analyzed separately. Influenza A was found in 83%, 10 out of 25 during the 2009 A(H1N1) pandemic. Two patients had bona fide TTP with ADAMTS13 activity <10%. Median age was 15 years (range 0.5-68 years), two thirds were male. Oligoanuria was documented in 81% and neurological involvement in 40% of patients. Serum C3 was reduced in 5 out of 14 patients (36%); Coombs test was negative in 7 out of 7 and elevated fibrin/fibrinogen degradation products were documented in 6 out of 8 patients. Pathogenic complement gene mutations were found in 7 out of 8 patients tested (C3, MCP, or MCP combined with CFB or clusterin). Twenty out of 24 patients recovered completely, but 3 died (12%). Ten of the surviving patients underwent plasma exchange (PLEX) therapy, 5 plasma infusions. Influenza-mediated HUS or TTP is rare. A sizable proportion of tested patients demonstrated mutations associated with alternative pathway of complement dysregulation that was uncovered by this infection. Further research is warranted targeting the roles of viral neuraminidase, enhanced virus-induced complement activation and/or ADAMTS13 antibodies, and rational treatment approaches.
Collapse
Affiliation(s)
- Martin Bitzan
- Division of Nephrology, The Montreal Children's Hospital, McGill University Health Centre, 1001, boul. Décarie-Room B RC.6651, Montréal, QC, H4A 3J1, Canada.
| | - Jakub Zieg
- 0000 0004 1937 116Xgrid.4491.8Department of Pediatric Nephrology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| |
Collapse
|
43
|
Motib A, Guerreiro A, Al-Bayati F, Piletska E, Manzoor I, Shafeeq S, Kadam A, Kuipers O, Hiller L, Cowen T, Piletsky S, Andrew PW, Yesilkaya H. Modulation of Quorum Sensing in a Gram-Positive Pathogen by Linear Molecularly Imprinted Polymers with Anti-infective Properties. Angew Chem Int Ed Engl 2017; 56:16555-16558. [DOI: 10.1002/anie.201709313] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/01/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Anfal Motib
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| | - Antonio Guerreiro
- MIP Diagnostics Ltd, Fielding Johnson Bldg.; University of Leicester; Leicester LE1 7RH UK
| | - Firas Al-Bayati
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| | - Elena Piletska
- Chemistry Department; University of Leicester; Leicester LE1 7RH UK
| | - Irfan Manzoor
- Molecular Genetics; University of Groningen; Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Sulman Shafeeq
- Molecular Genetics; University of Groningen; Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Anagha Kadam
- Department of Biological Sciences; Carnegie Mellon University; 4400 Fifth Avenue Pittsburgh PA 15213 USA
| | - Oscar Kuipers
- Molecular Genetics; University of Groningen; Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Luisa Hiller
- Department of Biological Sciences; Carnegie Mellon University; 4400 Fifth Avenue Pittsburgh PA 15213 USA
| | - Todd Cowen
- Chemistry Department; University of Leicester; Leicester LE1 7RH UK
| | - Sergey Piletsky
- Chemistry Department; University of Leicester; Leicester LE1 7RH UK
| | - Peter W. Andrew
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| | - Hasan Yesilkaya
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| |
Collapse
|
44
|
Motib A, Guerreiro A, Al-Bayati F, Piletska E, Manzoor I, Shafeeq S, Kadam A, Kuipers O, Hiller L, Cowen T, Piletsky S, Andrew PW, Yesilkaya H. Modulation of Quorum Sensing in a Gram-Positive Pathogen by Linear Molecularly Imprinted Polymers with Anti-infective Properties. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201709313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Anfal Motib
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| | - Antonio Guerreiro
- MIP Diagnostics Ltd, Fielding Johnson Bldg.; University of Leicester; Leicester LE1 7RH UK
| | - Firas Al-Bayati
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| | - Elena Piletska
- Chemistry Department; University of Leicester; Leicester LE1 7RH UK
| | - Irfan Manzoor
- Molecular Genetics; University of Groningen; Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Sulman Shafeeq
- Molecular Genetics; University of Groningen; Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Anagha Kadam
- Department of Biological Sciences; Carnegie Mellon University; 4400 Fifth Avenue Pittsburgh PA 15213 USA
| | - Oscar Kuipers
- Molecular Genetics; University of Groningen; Nijenborgh 7 9747 AG Groningen The Netherlands
| | - Luisa Hiller
- Department of Biological Sciences; Carnegie Mellon University; 4400 Fifth Avenue Pittsburgh PA 15213 USA
| | - Todd Cowen
- Chemistry Department; University of Leicester; Leicester LE1 7RH UK
| | - Sergey Piletsky
- Chemistry Department; University of Leicester; Leicester LE1 7RH UK
| | - Peter W. Andrew
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| | - Hasan Yesilkaya
- Department of Infection, Immunity & Inflammation; University of Leicester; Leicester LE1 9HN UK
| |
Collapse
|
45
|
Rasmussen LH, Højholt K, Dargis R, Christensen JJ, Skovgaard O, Justesen US, Rosenvinge FS, Moser C, Lukjancenko O, Rasmussen S, Nielsen XC. In silico assessment of virulence factors in strains of Streptococcus oralis and Streptococcus mitis isolated from patients with Infective Endocarditis. J Med Microbiol 2017; 66:1316-1323. [PMID: 28874232 DOI: 10.1099/jmm.0.000573] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose. Streptococcus oralis and Streptococcus mitis belong to the Mitis group, which are mostly commensals in the human oral cavity. Even though S. oralis and S. mitis are oral commensals, they can be opportunistic pathogens causing infective endocarditis. A recent taxonomic re-evaluation of the Mitis group has embedded the species Streptococcus tigurinus and Streptococcus dentisani into the species S. oralis as subspecies. In this study, the distribution of virulence factors that contribute to bacterial immune evasion, colonization and adhesion was assessed in clinical strains of S. oralis (subsp. oralis, subsp. tigurinus and subsp. dentisani) and S. mitis. Methodology. Forty clinical S. oralis (subsp. oralis, subsp. dentisani and subsp. tigurinus) and S. mitis genomes were annotated with the pipeline PanFunPro and aligned against the VFDB database for assessment of virulence factors.Results/Key findings. Three homologues of pavA, psaA and lmb, encoding adhesion proteins, were present in all strains. Seven homologues of nanA, nanB, ply, lytA, lytB, lytC and iga, of importance regarding survival in blood and modulation of the human immune system, were variously present in the genomes. Few S. oralis subspecies specific differences were observed. iga homologues were identified in S. oralis subsp. oralis, whereas lytA homologues were identified in S. oralis subsp. oralis and subsp. tigurinus. Conclusion. Differences in the presence of virulence factors among the three S. oralis subspecies were observed. The virulence gene profiles of the 40 S. mitis and S. oralis (subsp. oralis, subsp. dentisani and subsp. tigurinus) contribute with important new knowledge regarding these species and new subspecies.
Collapse
Affiliation(s)
- Louise H Rasmussen
- Department of Clinical Microbiology, Slagelse Hospital, Ingemannsvej 46, 4200 Slagelse, Denmark.,Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Katrine Højholt
- Department of Clinical Microbiology, Slagelse Hospital, Ingemannsvej 46, 4200 Slagelse, Denmark.,Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kemitorvet, Building 208, 2800 Kgs Lyngby, Denmark
| | - Rimtas Dargis
- Department of Clinical Microbiology, Slagelse Hospital, Ingemannsvej 46, 4200 Slagelse, Denmark
| | - Jens Jørgen Christensen
- Department of Clinical Microbiology, Slagelse Hospital, Ingemannsvej 46, 4200 Slagelse, Denmark.,Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Ole Skovgaard
- Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark
| | - Ulrik S Justesen
- Department of Clinical Microbiology, Odense University Hospital, J. B. Winsløws Vej 21, 2, 5000 Odense C, Denmark.,Department of Clinical Microbiology, Rigshospitalet, University Hospital of Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - Flemming S Rosenvinge
- Department of Clinical Microbiology, Vejle Hospital, Kabbeltoft 25, 7100 Vejle, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Rigshospitalet, University Hospital of Copenhagen, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - Oksana Lukjancenko
- National Food Institute, Technical University of Denmark, Søltofts plads, Building 221, 2800 Kgs Lyngby, Denmark
| | - Simon Rasmussen
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kemitorvet, Building 208, 2800 Kgs Lyngby, Denmark
| | - Xiaohui C Nielsen
- Department of Clinical Microbiology, Slagelse Hospital, Ingemannsvej 46, 4200 Slagelse, Denmark
| |
Collapse
|
46
|
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
|
47
|
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]
|
48
|
Li MF, Li XL, Fan KL, Yu YY, Gong J, Geng SY, Liang YF, Huang L, Qiu JH, Tian XH, Wang WT, Zhang XL, Yu QX, Zhang YF, Lin P, Wang LN, Li X, Hou M, Liu LY, Peng J. Platelet desialylation is a novel mechanism and a therapeutic target in thrombocytopenia during sepsis: an open-label, multicenter, randomized controlled trial. J Hematol Oncol 2017; 10:104. [PMID: 28494777 PMCID: PMC5426054 DOI: 10.1186/s13045-017-0476-1] [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: 03/21/2017] [Accepted: 05/02/2017] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Studies in murine models suggested that platelet desialylation was an important mechanism of thrombocytopenia during sepsis. METHODS First, we performed a prospective, multicenter, observational study that enrolled septic patients with or without thrombocytopenia to determine the association between platelet desialylation and thrombocytopenia in patients with sepsis, severe sepsis, and septic shock. Gender- and age-matched healthy adults were selected as normal controls in analysis of the platelet desialylation levels (study I). Next, we conducted an open-label randomized controlled trial (RCT) in which the patients who had severe sepsis with thrombocytopenia (platelet counts ≤50 × 109/L) were randomly assigned to receive antimicrobial therapy alone (control group) or antimicrobial therapy plus oseltamivir (oseltamivir group) in a 1:1 ratio (study II). The primary outcomes were platelet desialylation level at study entry, overall platelet response rate within 14 days post-randomization, and all-cause mortality within 28 days post-randomization. Secondary outcomes included platelet recovery time, the occurrence of bleeding events, and the amount of platelets transfused within 14 days post-randomization. RESULTS The platelet desialylation levels increased significantly in the 127 septic patients with thrombocytopenia compared to the 134 patients without thrombocytopenia. A platelet response was achieved in 45 of the 54 patients in the oseltamivir group (83.3%) compared with 34 of the 52 patients in the control group (65.4%; P = 0.045). The median platelet recovery time was 5 days (interquartile range 4-6) in the oseltamivir group compared with 7 days (interquartile range 5-10) in the control group (P = 0.003). The amount of platelets transfused decreased significantly in the oseltamivir group compared to the control group (P = 0.044). There was no difference in the overall 28-day mortality regardless of whether oseltamivir was used. The Sequential Organ Failure Assessment score and platelet recovery time were independent indicators of oseltamivir therapy. The main reason for all of the mortalities was multiple-organ failure. CONCLUSIONS Thrombocytopenia was associated with increased platelet desialylation in septic patients. The addition of oseltamivir could significantly increase the platelet response rate, shorten platelet recovery time, and reduce platelet transfusion. TRIAL REGISTRATION Chinese Clinical Trial Registry, ChiCTR-IPR-16008542 .
Collapse
Affiliation(s)
- Mei-Feng Li
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China.,Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Xiao-Li Li
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Kai-Liang Fan
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China.,Department of Emergency, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying-Yi Yu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Jing Gong
- Division of Preventive Medicine, Center for Disease Control and Prevention of Yantai Development Zone, Yantai, China
| | - Shu-Ying Geng
- Department of Internal Medicine, Infectious Disease Hospital of Yantai, Yantai, China
| | - Ya-Feng Liang
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Ling Huang
- Intensive Care Unit, Yantaishan Hospital of Yantai, Yantai, China
| | - Ji-Hua Qiu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Xing-Han Tian
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Wen-Ting Wang
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Xiao-Lu Zhang
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Qing-Xia Yu
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Yuan-Feng Zhang
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Peng Lin
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China
| | - Li-Na Wang
- Department of Internal Medicine, Infectious Disease Hospital of Yantai, Yantai, China
| | - Xin Li
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China.,Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Shandong University, Jinan, China
| | - Lu-Yi Liu
- Intensive Care Unit, and Clinical Laboratory, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong, China.
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
49
|
Zhang J, Cui YL, Jiang YM. [Immunoprotective effect of combined pneumococcal endopeptidase O and pneumococcal surface adhesin A vaccines against Streptococcus pneumoniae infection]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:583-589. [PMID: 28506354 PMCID: PMC7389137 DOI: 10.7499/j.issn.1008-8830.2017.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/06/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate the prokaryotic expression of proteins pneumococcal endopeptidase O (PepO) and pneumococcal surface adhesin A (PsaA) in Streptococcus pneumoniae and their immunoprotective effect as vaccine candidate proteins. METHODS Specific primers of target gene fragments were designed, and then PCR amplification was performed to establish recombinant plasmids pET28a(+)-pepO and pET28a(+)-psaA, which were transformed into host cells, Escherichia coli BL21 and DE3, respectively, to induce expression. Highly purified target proteins PepO and PsaA were obtained after purification. Mucosal immunization was performed for BALB/c mice and specific antiserum was prepared. ELISA was used to measure the antibody titer, and Western blot was used to analyze the specificity of the antiserum of target proteins. The mice were randomly divided into negative control group, PepO group, PsaA group, and PepO+PsaA combined immunization group, with 18 mice in each group. The models of different serotypes of Streptococcus pneumoniae infection were established to evaluate the immunoprotective effect of target proteins used alone or in combination. RESULTS The target proteins PepO and PsaA were successfully obtained and Western blot demonstrated that the antiserum of these proteins had good specificity. There was no significant difference in the titers of IgA in saliva and IgG in serum between the PepO group and the combined immunization group (P>0.05); however, these two groups had significantly higher antibody titers than the PsaA group (P<0.05). The PepO, PsaA, and combined immunization groups had significantly higher protection rates for mice infected with Streptococcus pneumoniae D39 and CMCC31436 in the nasal cavity than the negative control group (P<0.05). The PepO and combined immunization groups had a significantly higher protection rate for mice infected with Streptococcus pneumoniae D39 than the PsaA group (P<0.05). The results of colonization experiment showed that compared with the control group, the PepO, PsaA, and combined immunization groups showed a significant reduction in the colonization of Streptococcus pneumoniae (CMCC31693 and CMCC31207) in the nasopharynx and lung (P<0.05). The combined immunization group showed a better effect on reducing the colonization of CMCC31207 in the lung than the PepO and PsaA alone groups. CONCLUSIONS Combined PepO/PsaA vaccines may produce a better protective effect by mucosal immunization compared with the vaccine used alone in mice. The combined vaccines can effectively reduce the colonization of Streptococcus pneumoniae in the nasopharynx and lung. Therefore, such protein vaccines may have a great potential for research and development.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Clinical Laboratory, West China Second University Hospital, Sichuan University/Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Chengdu 610041, China.
| | | | | |
Collapse
|
50
|
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: 53] [Impact Index Per Article: 6.6] [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
|