1
|
De Chiara S, De Simone Carone L, Cirella R, Andretta E, Silipo A, Molinaro A, Mercogliano M, Di Lorenzo F. Beyond the Toll-Like Receptor 4. Structure-Dependent Lipopolysaccharide Recognition Systems: How far are we? ChemMedChem 2025; 20:e202400780. [PMID: 39752323 PMCID: PMC11911305 DOI: 10.1002/cmdc.202400780] [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: 09/30/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
With an enormous potential in immunology and vaccinology, lipopolysaccharides (LPSs) are among the most extensively studied bacteria-derived molecules. LPS centered studies are countless, and their results reverberate in all areas of the life sciences, including chemistry, biology, genetics, biophysics, and medicine. Most of these research activities are focused on the LPS-induced immune response activation by means of Myeloid Differentiation protein-2/Toll Like Receptor 4 (MD-2/TLR4) complex, which currently is the most largely explored LPS sensing pathway. However, the enormous structural variability of LPS allows interactions with numerous other receptors involved in a wide range of equally important immunological scenarios. In this review, we explore these additional LPS recognition systems, which operate within interconnected signaling cascades, highlighting their role in maintaining physiological homeostasis and their involvement in the development of severe human diseases. Understanding these pathways, their interconnections, and the crosstalk between them and TLR4/MD-2 is essential for guiding the development of pharmacologically active molecules that could specifically modulate the inflammatory response, paving the way to new strategies for combating immune-mediated diseases and resistant infections.
Collapse
Affiliation(s)
- Stefania De Chiara
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Luca De Simone Carone
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Roberta Cirella
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Emanuela Andretta
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Alba Silipo
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
- CEINGE, Istituto di Biotecnologie avanzateVia Gaetano Salvatore, 48680131NaplesItaly
| | - Antonio Molinaro
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
- CEINGE, Istituto di Biotecnologie avanzateVia Gaetano Salvatore, 48680131NaplesItaly
- Department of ChemistrySchool of ScienceOsaka University1-1 Osaka University MachikaneyamaToyonakaOsaka560-0043Japan
| | - Marcello Mercogliano
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
| | - Flaviana Di Lorenzo
- Department of chemical sciencesUniversity of Naples Federico IIvia Cinthia 480126NaplesItaly
- CEINGE, Istituto di Biotecnologie avanzateVia Gaetano Salvatore, 48680131NaplesItaly
| |
Collapse
|
2
|
Burzava AL, Zuber A, Hayles A, Morel J, Bright R, Wood J, Palms D, Barker D, Brown T, Vasilev K. Platelet interaction and performance of antibacterial bioinspired nanostructures passivated with human plasma. Mater Today Bio 2024; 29:101236. [PMID: 39399241 PMCID: PMC11467677 DOI: 10.1016/j.mtbio.2024.101236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/02/2024] [Accepted: 09/07/2024] [Indexed: 10/15/2024] Open
Abstract
The ever-increasing ageing of the world population is demanding superior orthopedic devices. Issues such as implant infection, poor osseointegration, or chronic inflammation remain problematic to the lifespan and long-term efficacy of implants. Fabrication of materials with bioinspired nanostructures is one emerging antibacterial strategy to prevent implant infection, however their interactions with blood components, and whether they retain their bactericidal properties in an environment displaying a complex protein corona, remains largely unexplored. In the present study, titanium alloy, commercially pure and plasma-sprayed titania were hydrothermally etched, passivated with human native plasma to develop a protein corona, and then incubated with either Staphylococcus aureus, Pseudomonas aeruginosa or human platelets. Surface analysis was first used to characterize the topography, chemical composition or crystallinity of each material. Fluorescence staining and SEM were performed to evaluate the nanostructure bactericidal properties, as well as to study platelet attachment and morphology. Composition of platelet supernatant was studied using ELISA and flow cytometry. Overall, our study showed that the bioinspired nanostructured surfaces displayed both impressive antibacterial properties in a complex environment, and a superior blood biocompatibility profile in terms of platelet activation (particularly for titanium alloy). Additionally, the amount of pro-inflammatory cytokines released by platelets was found to be no different to that found in native plasma (background levels) and, in some cases, presented a more pro-healing profile with an increased secretion of factors such as TGF-β, PDGF-BB or BMP-2. The nanostructured surfaces performed equally, or better, than hydroxyapatite-coated titanium which is one of the current gold standards in orthopedics. Although further in vivo studies are required to validate these results, such bioinspired nanostructured surfaces certainly show promise to be safely applied to medical device surfaces used in orthopedics and other areas.
Collapse
Affiliation(s)
- Anouck L.S. Burzava
- STEM, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
- Laboratoire Softmat, Université de Toulouse, CNRS, UMR 5623, Université Toulouse III – Paul Sabatier, 31062, Toulouse, France
| | - Agnieszka Zuber
- STEM, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
| | - Andrew Hayles
- STEM, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, 5042, South Australia, Australia
| | - James Morel
- School of Chemical Engineering, UNSW Sydney, New South Wales, 2052, Australia
| | - Richard Bright
- STEM, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Jonathan Wood
- STEM, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
| | - Dennis Palms
- STEM, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Dan Barker
- Corin Australia, Pymble, New South Wales, 2073, Australia
| | - Toby Brown
- Corin Australia, Pymble, New South Wales, 2073, Australia
| | - Krasimir Vasilev
- STEM, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, 5042, South Australia, Australia
| |
Collapse
|
3
|
Paulikat AD, Schwudke D, Hammerschmidt S, Voß F. Lipidation of pneumococcal proteins enables activation of human antigen-presenting cells and initiation of an adaptive immune response. Front Immunol 2024; 15:1392316. [PMID: 38711516 PMCID: PMC11070533 DOI: 10.3389/fimmu.2024.1392316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Streptococcus pneumoniae remains a significant global threat, with existing vaccines having important limitations such as restricted serotype coverage and high manufacturing costs. Pneumococcal lipoproteins are emerging as promising vaccine candidates due to their surface exposure and conservation across various serotypes. While prior studies have explored their potential in mice, data in a human context and insights into the impact of the lipid moiety remain limited. In the present study, we examined the immunogenicity of two pneumococcal lipoproteins, DacB and MetQ, both in lipidated and non-lipidated versions, by stimulation of primary human immune cells. Immune responses were assessed by the expression of common surface markers for activation and maturation as well as cytokines released into the supernatant. Our findings indicate that in the case of MetQ lipidation was crucial for activation of human antigen-presenting cells such as dendritic cells and macrophages, while non-lipidated DacB demonstrated an intrinsic potential to induce an innate immune response. Nevertheless, immune responses to both proteins were enhanced by lipidation. Interestingly, following stimulation of dendritic cells with DacB, LipDacB and LipMetQ, cytokine levels of IL-6 and IL-23 were significantly increased, which are implicated in triggering potentially important Th17 cell responses. Furthermore, LipDacB and LipMetQ were able to induce proliferation of CD4+ T cells indicating their potential to induce an adaptive immune response. These findings contribute valuable insights into the immunogenic properties of pneumococcal lipoproteins, emphasizing their potential role in vaccine development against pneumococcal infections.
Collapse
Affiliation(s)
- Antje D. Paulikat
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Dominik Schwudke
- Division of Bioanalytical Chemistry, Research Center Borstel - Leibniz Lung Center, Borstel, Germany
- German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- German Center for Lung Research (DZL), Airway Research Center North (ARCN), Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Franziska Voß
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| |
Collapse
|
4
|
Laverde D, Armiento S, Molinaro A, Huebner J, De Castro C, Romero-Saavedra F. Identification of a capsular polysaccharide from Enterococcus faecium U0317 using a targeted approach to discover immunogenic carbohydrates for vaccine development. Carbohydr Polym 2024; 330:121731. [PMID: 38368077 DOI: 10.1016/j.carbpol.2023.121731] [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: 11/03/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 02/19/2024]
Abstract
Enterococcus faecium, a gram-positive opportunistic pathogen, has become a major concern for nosocomial infections due to its resistance to several antibiotics, including vancomycin. Finding novel alternatives for treatment prevention, such as vaccines, is therefore crucial. In this study, we used various techniques to discover a novel capsular polysaccharide. Firstly, we identified an encapsulated E. faecium strain by evaluating the opsonophagocytic activity of fifteen strains with antibodies targeting the well-known lipoteichoic acid antigen. This activity was attributed to an unknown polysaccharide. We then prepared a crude cell wall glycopolymer and fractionated it, guided by immunodot-blot analysis. The most immunoreactive fractions were used for opsonophagocytic inhibition assays. The fraction containing the inhibitory polysaccharide underwent structural characterization using NMR and chemical analyses. The elucidated structure presents a branched repeating unit, with the linear part being: →)-β-d-Gal-(1 → 4)-β-d-Glc-(1 → 4)-β-d-Gal-(1 → 4)-β-d-GlcNAc-(1→, further decorated with a terminal α-d-Glc and a d-phosphoglycerol moiety, attached to O-2 and O-3 of the 4-linked Gal unit, respectively. This polysaccharide was conjugated to BSA and the synthetic glycoprotein used to immunize mice. The resulting sera exhibited good opsonic activity, suggesting its potential as a vaccine antigen. In conclusion, our effector-function-based approach successfully identified an immunogenic capsular polysaccharide with promising applications in immunotherapy.
Collapse
Affiliation(s)
- Diana Laverde
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Samantha Armiento
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Santangelo, Napoli, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Santangelo, Napoli, Italy
| | - Johannes Huebner
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Cristina De Castro
- Department of Chemical Sciences, University of Napoli Federico II, Complesso Universitario Monte Santangelo, Napoli, Italy
| | - Felipe Romero-Saavedra
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
5
|
Klabunde B, Wesener A, Bertrams W, Beinborn I, Paczia N, Surmann K, Blankenburg S, Wilhelm J, Serrania J, Knoops K, Elsayed EM, Laakmann K, Jung AL, Kirschbaum A, Hammerschmidt S, Alshaar B, Gisch N, Abu Mraheil M, Becker A, Völker U, Vollmeister E, Benedikter BJ, Schmeck B. NAD + metabolism is a key modulator of bacterial respiratory epithelial infections. Nat Commun 2023; 14:5818. [PMID: 37783679 PMCID: PMC10545792 DOI: 10.1038/s41467-023-41372-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/30/2023] [Indexed: 10/04/2023] Open
Abstract
Lower respiratory tract infections caused by Streptococcus pneumoniae (Spn) are a leading cause of death globally. Here we investigate the bronchial epithelial cellular response to Spn infection on a transcriptomic, proteomic and metabolic level. We found the NAD+ salvage pathway to be dysregulated upon infection in a cell line model, primary human lung tissue and in vivo in rodents, leading to a reduced production of NAD+. Knockdown of NAD+ salvage enzymes (NAMPT, NMNAT1) increased bacterial replication. NAD+ treatment of Spn inhibited its growth while growth of other respiratory pathogens improved. Boosting NAD+ production increased NAD+ levels in immortalized and primary cells and decreased bacterial replication upon infection. NAD+ treatment of Spn dysregulated the bacterial metabolism and reduced intrabacterial ATP. Enhancing the bacterial ATP metabolism abolished the antibacterial effect of NAD+. Thus, we identified the NAD+ salvage pathway as an antibacterial pathway in Spn infections, predicting an antibacterial mechanism of NAD+.
Collapse
Affiliation(s)
- Björn Klabunde
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - André Wesener
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Wilhelm Bertrams
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Isabell Beinborn
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Nicole Paczia
- Core Facility for Metabolomics and Small Molecule Mass Spectrometry, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Kristin Surmann
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Sascha Blankenburg
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jochen Wilhelm
- Institute for Lung Health (ILH), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-Universität Giessen, German Center for Lung Research (DZL), Giessen, Germany
| | - Javier Serrania
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-Universität Marburg, Marburg, Germany
| | - Kèvin Knoops
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Eslam M Elsayed
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-Universität Marburg, Marburg, Germany
- Department of Biology, Philipps-Universität Marburg, Marburg, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Katrin Laakmann
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Anna Lena Jung
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
- Core Facility Flow Cytometry - Bacterial Vesicles, Philipps-Universität Marburg, Marburg, Germany
| | - Andreas Kirschbaum
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Gießen and Marburg (UKGM), Marburg, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Belal Alshaar
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Mobarak Abu Mraheil
- Institute for Medical Microbiology, Justus-Liebig Universität Giessen, Giessen, Germany
| | - Anke Becker
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-Universität Marburg, Marburg, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Evelyn Vollmeister
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany
| | - Birke J Benedikter
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany.
- University Eye Clinic Maastricht, Maastricht University Medical Center (MUMC+), School for Mental Health and Neuroscience, Maastricht University, P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands.
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-Universität Marburg, Marburg, Germany.
- Institute for Lung Health (ILH), Giessen, Germany.
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-Universität Marburg, Marburg, Germany.
- Core Facility Flow Cytometry - Bacterial Vesicles, Philipps-Universität Marburg, Marburg, Germany.
- Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Center Marburg, Philipps-Universität Marburg, Marburg, Germany.
- Member of the German Center for Infectious Disease Research (DZIF), Marburg, Germany.
| |
Collapse
|
6
|
Vilhena C, Du S, Battista M, Westermann M, Kohler T, Hammerschmidt S, Zipfel PF. The choline-binding proteins PspA, PspC, and LytA of Streptococcus pneumoniae and their interaction with human endothelial and red blood cells. Infect Immun 2023; 91:e0015423. [PMID: 37551971 PMCID: PMC10501214 DOI: 10.1128/iai.00154-23] [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: 04/26/2023] [Accepted: 06/23/2023] [Indexed: 08/09/2023] Open
Abstract
Streptococcus pneumoniae is a Gram-positive opportunistic pathogen that can colonize the upper respiratory tract. It is a leading cause of a wide range of infectious diseases, including community-acquired pneumonia and meningitis. Pneumococcal infections cause 1-2 million deaths per year, most of which occur in developing countries. Here, we focused on three choline-binding proteins (CBPs), i.e., PspC, PspA, and LytA. These pneumococcal proteins have different surface-exposed regions but share related choline-binding anchors. These surface-exposed pneumococcal proteins are in direct contact with host cells and have diverse functions. We explored the role of the three CBPs on adhesion and pathogenicity in a human host by performing relevant imaging and functional analyses, such as electron microscopy, confocal laser scanning microscopy, and functional quantitative assays, targeting biofilm formation and the hemolytic capacity of S. pneumoniae. In vitro biofilm formation assays and electron microscopy experiments were used to examine the ability of knockout mutant strains lacking the lytA, pspC, or pspA genes to adhere to surfaces. We found that LytA plays an important role in robust synthesis of the biofilm matrix. PspA and PspC appeared crucial for the hemolytic effects of S. pneumoniae on human red blood cells. Furthermore, all knockout mutants caused less damage to endothelial cells than wild-type bacteria, highlighting the significance of each CPB for the overall pathogenicity of S. pneumoniae. Hence, in addition to their structural function within the cell wall of S. pneumoniae, each of these three surface-exposed CBPs controls or mediates multiple steps during bacterial pathogenesis.
Collapse
Affiliation(s)
- Cláudia Vilhena
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Shanshan Du
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Miriana Battista
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Martin Westermann
- Centre for Electron Microscopy, Jena University Hospital, Friedrich-Schiller-University of Jena, Jena, Germany
| | - Thomas Kohler
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Peter F. Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
7
|
Tabary M, Gheware A, Peñaloza HF, Lee JS. The matricellular protein thrombospondin-1 in lung inflammation and injury. Am J Physiol Cell Physiol 2022; 323:C857-C865. [PMID: 35912991 PMCID: PMC9467471 DOI: 10.1152/ajpcell.00182.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/22/2022]
Abstract
Matricellular proteins comprise a diverse group of molecular entities secreted into the extracellular space. They interact with the extracellular matrix (ECM), integrins, and other cell-surface receptors, and can alter matrix strength, cell attachment to the matrix, and cell-cell adhesion. A founding member of this group is thrombospondin-1 (TSP-1), a high molecular-mass homotrimeric glycoprotein. Given the importance of the matrix and ECM remodeling in the lung following injury, TSP-1 has been implicated in a number of lung pathologies. This review examines the role of TSP-1 as a damage controller in the context of lung inflammation, injury resolution, and repair in noninfectious and infectious models. This review also discusses the potential role of TSP-1 in human diseases as it relates to lung inflammation and injury.
Collapse
Affiliation(s)
- Mohammadreza Tabary
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Atish Gheware
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hernán F Peñaloza
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Janet S Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
8
|
Oehmcke-Hecht S, Maletzki C, Surabhi S, Siemens N, Khaimov V, John LM, Peter SM, Hammerschmidt S, Kreikemeyer B. Procoagulant Activity of Blood and Microvesicles Is Disturbed by Pneumococcal Pneumolysin, Which Interacts with Coagulation Factors. J Innate Immun 2022; 15:136-152. [PMID: 35843205 PMCID: PMC10643893 DOI: 10.1159/000525479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/06/2022] [Indexed: 11/17/2023] Open
Abstract
The coagulation and contact systems are parts of the innate immune system as they prevent bleeding and dissemination of pathogens and also contribute to microbial killing by inflammatory reactions and the release of antimicrobial peptides. Here, we investigated the influence of Streptococcus pneumoniae on the coagulation and contact system. S. pneumoniae (pneumococci), but no other investigated streptococcal species, impairs coagulation of blood by autolysis and release of pneumolysin. Defective blood coagulation results from the lysis of tissue factor-producing mononuclear cells and their procoagulant microvesicles, which are the main trigger for blood coagulation during sepsis. In addition, pneumolysin binds coagulation and contact system factors, but this does not result in activation. Thus, pneumococci modulate activation of the coagulation system by releasing pneumolysin, which could potentiate lung injury during pneumonia.
Collapse
Affiliation(s)
- Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, Rostock, Germany
| | - Claudia Maletzki
- Department of Medicine, Clinic III-Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Surabhi Surabhi
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Nikolai Siemens
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Valeria Khaimov
- Institute for ImplantTechnology and Biomaterials e.V., Rostock, Germany
| | - Lisa Marie John
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, Rostock, Germany
| | - Sina Mariella Peter
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, Rostock, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
9
|
Age-Dependent Control of Collagen-Dependent Platelet Responses by Thrombospondin-1-Comparative Analysis of Platelets from Neonates, Children, Adolescents, and Adults. Int J Mol Sci 2021; 22:ijms22094883. [PMID: 34063076 PMCID: PMC8124951 DOI: 10.3390/ijms22094883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023] Open
Abstract
Platelet function is developmentally regulated. Healthy neonates do not spontaneously bleed, but their platelets are hypo-reactive to several agonists. The mechanisms underlying immature platelet function in neonates are incompletely understood. This critical issue remains challenging for the establishment of age-specific reference ranges. In this study, we evaluated platelet reactivity of five pediatric age categories, ranging from healthy full-term neonates up to adolescents (11–18 years) in comparison to healthy adults (>18 years) by flow cytometry. We confirmed that platelet hypo-reactivity detected by fibrinogen binding, P-selectin, and CD63 surface expression was most pronounced in neonates compared to other pediatric age groups. However, maturation of platelet responsiveness varied with age, agonist, and activation marker. In contrast to TRAP and ADP, collagen-induced platelet activation was nearly absent in neonates. Granule secretion markedly remained impaired at least up to 10 years of age compared to adults. We show for the first time that neonatal platelets are deficient in thrombospondin-1, and exogenous platelet-derived thrombospondin-1 allows platelet responsiveness to collagen. Platelets from all pediatric age groups normally responded to the C-terminal thrombospondin-1 peptide RFYVVMWK. Thus, thrombospondin-1 deficiency of neonatal platelets might contribute to the relatively impaired response to collagen, and platelet-derived thrombospondin-1 may control distinct collagen-induced platelet responses.
Collapse
|
10
|
Hirschmann S, Gómez-Mejia A, Mäder U, Karsunke J, Driesch D, Rohde M, Häussler S, Burchhardt G, Hammerschmidt S. The Two-Component System 09 Regulates Pneumococcal Carbohydrate Metabolism and Capsule Expression. Microorganisms 2021; 9:microorganisms9030468. [PMID: 33668344 PMCID: PMC7996280 DOI: 10.3390/microorganisms9030468] [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: 01/01/2021] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 02/05/2023] Open
Abstract
Streptococcus pneumoniae two-component regulatory systems (TCSs) are important systems that perceive and respond to various host environmental stimuli. In this study, we have explored the role of TCS09 on gene expression and phenotypic alterations in S. pneumoniae D39. Our comparative transcriptomic analyses identified 67 differently expressed genes in total. Among those, agaR and the aga operon involved in galactose metabolism showed the highest changes. Intriguingly, the encapsulated and nonencapsulated hk09-mutants showed significant growth defects under nutrient-defined conditions, in particular with galactose as a carbon source. Phenotypic analyses revealed alterations in the morphology of the nonencapsulated hk09- and tcs09-mutants, whereas the encapsulated hk09- and tcs09-mutants produced higher amounts of capsule. Interestingly, the encapsulated D39∆hk09 showed only the opaque colony morphology, while the D39∆rr09- and D39∆tcs09-mutants had a higher proportion of transparent variants. The phenotypic variations of D39ΔcpsΔhk09 and D39ΔcpsΔtcs09 are in accordance with their higher numbers of outer membrane vesicles, higher sensitivity against Triton X-100 induced autolysis, and lower resistance against oxidative stress. In conclusion, these results indicate the importance of TCS09 for pneumococcal metabolic fitness and resistance against oxidative stress by regulating the carbohydrate metabolism and thereby, most likely indirectly, the cell wall integrity and amount of capsular polysaccharide.
Collapse
Affiliation(s)
- Stephanie Hirschmann
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (J.K.); (G.B.)
| | - Alejandro Gómez-Mejia
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (J.K.); (G.B.)
| | - Ulrike Mäder
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Julia Karsunke
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (J.K.); (G.B.)
| | | | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| | - Susanne Häussler
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| | - Gerhard Burchhardt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (J.K.); (G.B.)
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (J.K.); (G.B.)
- Correspondence:
| |
Collapse
|
11
|
Ali MQ, Kohler TP, Burchhardt G, Wüst A, Henck N, Bolsmann R, Voß F, Hammerschmidt S. Extracellular Pneumococcal Serine Proteases Affect Nasopharyngeal Colonization. Front Cell Infect Microbiol 2021; 10:613467. [PMID: 33659218 PMCID: PMC7917122 DOI: 10.3389/fcimb.2020.613467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/30/2020] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pneumoniae has evolved versatile strategies to colonize the nasopharynx of humans. Colonization is facilitated by direct interactions with host cell receptors or via binding to components of the extracellular matrix. In addition, pneumococci hijack host-derived extracellular proteases such as the serine protease plasmin(ogen) for ECM and mucus degradation as well as colonization. S. pneumoniae expresses strain-dependent up to four serine proteases. In this study, we assessed the role of secreted or cell-bound serine proteases HtrA, PrtA, SFP, and CbpG, in adherence assays and in a mouse colonization model. We hypothesized that the redundancy of serine proteases compensates for the deficiency of a single enzyme. Therefore, double and triple mutants were generated in serotype 19F strain EF3030 and serotype 4 strain TIGR4. Strain EF3030 produces only three serine proteases and lacks the SFP encoding gene. In adherence studies using Detroit-562 epithelial cells, we demonstrated that both TIGR4Δcps and 19F mutants without serine proteases or expressing only CbpG, HtrA, or PrtA have a reduced ability to adhere to Detroit-562 cells. Consistent with these results, we show that the mutants of strain 19F, which preferentially colonizes mice, abrogate nasopharyngeal colonization in CD-1 mice after intranasal infection. The bacterial load in the nasopharynx was monitored for 14 days. Importantly, mutants showed significantly lower bacterial numbers in the nasopharynx two days after infection. Similarly, we detected a significantly reduced pneumococcal colonization on days 3, 7, and 14 post-inoculations. To assess the impact of pneumococcal serine proteases on acute infection, we infected mice intranasally with bioluminescent and invasive TIGR4 or isogenic triple mutants expressing only CbpG, HtrA, PrtA, or SFP. We imaged the acute lung infection in real-time and determined the survival of the mice. The TIGR4lux mutant expressing only PrtA showed a significant attenuation and was less virulent in the acute pneumonia model. In conclusion, our results showed that pneumococcal serine proteases contributed significantly to pneumococcal colonization but played only a minor role in pneumonia and invasive diseases. Because colonization is a prerequisite for invasive diseases and transmission, these enzymes could be promising candidates for the development of antimicrobials to reduce pneumococcal transmission.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| |
Collapse
|
12
|
Wolff M, Handtke S, Palankar R, Wesche J, Kohler TP, Kohler C, Gruel Y, Hammerschmidt S, Greinacher A. Activated platelets kill Staphylococcus aureus, but not Streptococcus pneumoniae-The role of FcγRIIa and platelet factor 4/heparinantibodies. J Thromb Haemost 2020; 18:1459-1468. [PMID: 32237268 DOI: 10.1111/jth.14814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/16/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Heparin induced thrombocytopenia (HIT) is likely a misdirected bacterial host defense mechanism. Platelet factor 4 (PF4) binds to polyanions on bacterial surfaces exposing neo-epitopes to which HIT antibodies bind. Platelets are activated by the resulting immune complexes via FcγRIIA, release bactericidal substances, and kill Gram-negative Escherichia coli. OBJECTIVES To assess the role of PF4, anti-PF4/H antibodies and FcγRIIa in killing of Gram-positive bacteria by platelets. METHODS Binding of PF4 to protein-A deficient Staphylococcus aureus (SA113Δspa) and non-encapsulated Streptococcus pneumoniae (D39Δcps) and its conformational change were assessed by flow cytometry using monoclonal (KKO,5B9) and patient derived anti-PF4/H antibodies. Killing of bacteria was quantified by counting colony forming units (cfu) after incubation with platelets or platelet releasate. Using flow cytometry, platelet activation (CD62P-expression, PAC-1 binding) and phosphatidylserine (PS)-exposure were analyzed. RESULTS Monoclonal and patient-derived anti-PF4/H antibodies bound in the presence of PF4 to both S. aureus and S. pneumoniae (1.6-fold increased fluorescence signal for human anti-PF4/H antibodies to 24.0-fold increase for KKO). Staphylococcus aureus (5.5 × 104 cfu/mL) was efficiently killed by platelets (2.7 × 104 cfu/mL) or their releasate (2.9 × 104 cfu/mL). Killing was not further enhanced by PF4 or anti-PF4/H antibodies. Blocking FcγRIIa had no impact on killing of S. aureus by platelets. In contrast, S. pneumoniae was not killed by platelets or releasate. Instead, after incubation with pneumococci platelets were unresponsive to TRAP-6 stimulation and exposed high levels of PS. CONCLUSIONS Anti-PF4/H antibodies seem to have only a minor role for direct killing of Gram-positive bacteria by platelets. Staphylococcus aureus is killed by platelets or platelet releasate. In contrast, S. pneumoniae affects platelet viability.
Collapse
Affiliation(s)
- Martina Wolff
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Stefan Handtke
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Raghavendra Palankar
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Jan Wesche
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Christian Kohler
- Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Yves Gruel
- Département d'Hématologie-Hémostase, Hôpital Universitaire de Tours, Tours, France
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Andreas Greinacher
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
13
|
Hentschker C, Maaß S, Junker S, Hecker M, Hammerschmidt S, Otto A, Becher D. Comprehensive Spectral Library from the Pathogenic Bacterium Streptococcus pneumoniae with Focus on Phosphoproteins. J Proteome Res 2020; 19:1435-1446. [DOI: 10.1021/acs.jproteome.9b00615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Christian Hentschker
- Department of Microbial Proteomics, Institute of Microbiology; University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Sandra Maaß
- Department of Microbial Proteomics, Institute of Microbiology; University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Sabryna Junker
- Department of Microbial Proteomics, Institute of Microbiology; University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Michael Hecker
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology; University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Andreas Otto
- Department of Microbial Proteomics, Institute of Microbiology; University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| | - Dörte Becher
- Department of Microbial Proteomics, Institute of Microbiology; University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany
| |
Collapse
|
14
|
Jun JI, Lau LF. CCN1 is an opsonin for bacterial clearance and a direct activator of Toll-like receptor signaling. Nat Commun 2020; 11:1242. [PMID: 32144270 PMCID: PMC7060279 DOI: 10.1038/s41467-020-15075-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 02/17/2020] [Indexed: 12/30/2022] Open
Abstract
Expression of the matricellular protein CCN1 (CYR61) is associated with inflammation and is required for successful wound repair. Here, we show that CCN1 binds bacterial pathogen-associated molecular patterns including peptidoglycans of Gram-positive bacteria and lipopolysaccharides of Gram-negative bacteria. CCN1 opsonizes methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa and accelerates their removal by phagocytosis and increased production of bactericidal reactive oxygen species in macrophages through the engagement of integrin αvβ3. Mice with myeloid-specific Ccn1 deletion and knock-in mice expressing CCN1 unable to bind αvβ3 are more susceptible to infection by S. aureus or P. aeruginosa, resulting in increased mortality and organ colonization. Furthermore, CCN1 binds directly to TLR2 and TLR4 to activate MyD88-dependent signaling, cytokine expression and neutrophil mobilization. CCN1 is therefore a pattern recognition receptor that opsonizes bacteria for clearance and functions as a damage-associated molecular pattern to activate inflammatory responses, activities that contribute to wound healing and tissue repair. CCN1 is a matricellular protein with a variety of functions, including an effect on wound healing and an association with inflammation. Here, the authors identify a possible mechanism by showing that CCN1 mediates the clearance of bacterial infections in mice and activates TLR signalling.
Collapse
Affiliation(s)
- Joon-Il Jun
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, IL, 60607, USA
| | - Lester F Lau
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, IL, 60607, USA.
| |
Collapse
|
15
|
Devraj G, Guérit S, Seele J, Spitzer D, Macas J, Khel MI, Heidemann R, Braczynski AK, Ballhorn W, Günther S, Ogunshola OO, Mittelbronn M, Ködel U, Monoranu CM, Plate KH, Hammerschmidt S, Nau R, Devraj K, Kempf VAJ. HIF-1α is involved in blood-brain barrier dysfunction and paracellular migration of bacteria in pneumococcal meningitis. Acta Neuropathol 2020; 140:183-208. [PMID: 32529267 PMCID: PMC7360668 DOI: 10.1007/s00401-020-02174-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
Bacterial meningitis is a deadly disease most commonly caused by Streptococcus pneumoniae, leading to severe neurological sequelae including cerebral edema, seizures, stroke, and mortality when untreated. Meningitis is initiated by the transfer of S. pneumoniae from blood to the brain across the blood-cerebrospinal fluid barrier or the blood-brain barrier (BBB). The underlying mechanisms are still poorly understood. Current treatment strategies include adjuvant dexamethasone for inflammation and cerebral edema, followed by antibiotics. The success of dexamethasone is however inconclusive, necessitating new therapies for controlling edema, the primary reason for neurological complications. Since we have previously shown a general activation of hypoxia inducible factor (HIF-1α) in bacterial infections, we hypothesized that HIF-1α, via induction of vascular endothelial growth factor (VEGF) is involved in transmigration of pathogens across the BBB. In human, murine meningitis brain samples, HIF-1α activation was observed by immunohistochemistry. S. pneumoniae infection in brain endothelial cells (EC) resulted in in vitro upregulation of HIF-1α/VEGF (Western blotting/qRT-PCR) associated with increased paracellular permeability (fluorometry, impedance measurements). This was supported by bacterial localization at cell-cell junctions in vitro and in vivo in brain ECs from mouse and humans (confocal, super-resolution, electron microscopy, live-cell imaging). Hematogenously infected mice showed increased permeability, S. pneumoniae deposition in the brain, along with upregulation of genes in the HIF-1α/VEGF pathway (RNA sequencing of brain microvessels). Inhibition of HIF-1α with echinomycin, siRNA in bEnd5 cells or using primary brain ECs from HIF-1α knock-out mice revealed reduced endothelial permeability and transmigration of S. pneumoniae. Therapeutic rescue using the HIF-1α inhibitor echinomycin resulted in increased survival and improvement of BBB function in S. pneumoniae-infected mice. We thus demonstrate paracellular migration of bacteria across BBB and a critical role for HIF-1α/VEGF therein and hence propose targeting this pathway to prevent BBB dysfunction and ensuing brain damage in infections.
Collapse
Affiliation(s)
- Gayatri Devraj
- Institute for Medical Microbiology and Infection Control, Goethe University, Frankfurt am Main, Germany
| | - Sylvaine Guérit
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany
| | - Jana Seele
- Institute of Neuropathology, University Medical Center, Göttingen, Germany ,Department of Geriatrics, Evangelisches Krankenhaus, Göttingen-Weende, Germany
| | - Daniel Spitzer
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany ,Department of Neurology, Goethe University, Frankfurt am Main, Germany
| | - Jadranka Macas
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany
| | - Maryam I. Khel
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany
| | - Roxana Heidemann
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Anne K. Braczynski
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany ,Department of Neurology, Technische Hochschule University Hospital, Aachen, Germany
| | - Wibke Ballhorn
- Institute for Medical Microbiology and Infection Control, Goethe University, Frankfurt am Main, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Michel Mittelbronn
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany ,Luxembourg Centre of Neuropathology (LCNP), Luxembourg, Luxembourg ,Laboratoire National de Santé (LNS), Dudelange, Luxembourg ,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg, Luxembourg ,NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
| | - Uwe Ködel
- Department of Neurology, Ludwig-Maximilians University, Munich, Germany
| | - Camelia M. Monoranu
- Department of Neuropathology, Institute of Pathology, Julius Maximilians University, Würzburg, Germany
| | - Karl H. Plate
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany ,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Roland Nau
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | - Kavi Devraj
- Edinger Institute/Neurological Institute, Goethe University, Frankfurt am Main, Germany. .,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany.
| | - Volkhard A. J. Kempf
- Institute for Medical Microbiology and Infection Control, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
16
|
Hammerschmidt S, Rohde M, Preissner KT. Extracellular Matrix Interactions with Gram-Positive Pathogens. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0041-2018. [PMID: 31004421 PMCID: PMC11590433 DOI: 10.1128/microbiolspec.gpp3-0041-2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Indexed: 01/10/2023] Open
Abstract
The main strategies used by pathogenic bacteria to infect eukaryotic tissue include their adherence to cells and the extracellular matrix (ECM), the subsequent colonization and invasion as well as the evasion of immune defences. A variety of structurally and functionally characterized adhesins and binding proteins of gram-positive bacteria facilitate these processes by specifically recognizing and interacting with various components of the host ECM, including different collagens, fibronectin and other macromolecules. The ECM affects the cellular physiology of our body and is critical for adhesion, migration, proliferation, and differentiation of many host cell types, but also provides the support for infiltrating pathogens, particularly under conditions of injury and trauma. Moreover, microbial binding to a variety of adhesive components in host tissue fluids leads to structural and/or functional alterations of host proteins and to the activation of cellular mechanisms that influence tissue and cell invasion of pathogens. Since the diverse interactions of gram-positive bacteria with the ECM represent important pathogenicity mechanisms, their characterization not only allows a better understanding of microbial invasion but also provides clues for the design of novel therapeutic strategies to manage infectious diseases.
Collapse
Affiliation(s)
- Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, University of Greifswald, D-17487 Greifswald, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz-Center for Infection Research, D-38124 Braunschweig, Germany
| | - Klaus T Preissner
- Institute for Biochemistry, Medical School, Justus-Liebig-University, D-35392 Giessen, Germany
| |
Collapse
|
17
|
Binsker U, Kohler TP, Hammerschmidt S. Contribution of Human Thrombospondin-1 to the Pathogenesis of Gram-Positive Bacteria. J Innate Immun 2019; 11:303-315. [PMID: 30814475 PMCID: PMC6738282 DOI: 10.1159/000496033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
A successful colonization of different compartments of the human host requires multifactorial contacts between bacterial surface proteins and host factors. Extracellular matrix proteins and matricellular proteins such as thrombospondin-1 play a pivotal role as adhesive substrates to ensure a strong interaction with pathobionts like the Gram-positive Streptococcus pneumoniae and Staphylococcus aureus. The human glycoprotein thrombospondin-1 is a component of the extracellular matrix and is highly abundant in the bloodstream during bacteremia. Human platelets secrete thrombospondin-1, which is then acquired by invading pathogens to facilitate colonization and immune evasion. Gram-positive bacteria express a broad spectrum of surface-exposed proteins, some of which also recognize thrombospondin-1. This review highlights the importance of thrombospondin-1 as an adhesion substrate to facilitate colonization, and we summarize the variety of thrombospondin-1-binding proteins of S. pneumoniae and S. aureus.
Collapse
Affiliation(s)
- Ulrike Binsker
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Greifswald, Germany
- Department of Microbiology, NYU Langone Health, Alexandria Center for the Life Sciences, New York City, New York, USA
| | - Thomas P Kohler
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Greifswald, Germany
| | - Sven Hammerschmidt
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Greifswald, Germany,
| |
Collapse
|
18
|
Bülow S, Zeller L, Werner M, Toelge M, Holzinger J, Entzian C, Schubert T, Waldow F, Gisch N, Hammerschmidt S, Gessner A. Bactericidal/Permeability-Increasing Protein Is an Enhancer of Bacterial Lipoprotein Recognition. Front Immunol 2018; 9:2768. [PMID: 30581431 PMCID: PMC6293271 DOI: 10.3389/fimmu.2018.02768] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
Adequate perception of immunologically important pathogen-associated molecular patterns like lipopolysaccharide and bacterial lipoproteins is essential for efficient innate and adaptive immune responses. In the context of Gram-negative infection, bactericidal/permeability-increasing protein (BPI) neutralizes endotoxic activity of lipopolysaccharides, and thus prohibits hyperactivation. So far, no immunological function of BPI has been described in Gram-positive infections. Here, we show a significant elevation of BPI in Gram-positive meningitis and, surprisingly, a positive correlation between BPI and pro-inflammatory markers like TNFα. To clarify the underlying mechanisms, we identify BPI ligands of Gram-positive origin, specifically bacterial lipopeptides and lipoteichoic acids, and determine essential structural motifs for this interaction. Importantly, the interaction of BPI with these newly defined ligands significantly enhances the immune response in peripheral blood mononuclear cells (PBMCs) mediated by Gram-positive bacteria, and thereby ensures their sensitive perception. In conclusion, we define BPI as an immune enhancing pattern recognition molecule in Gram-positive infections.
Collapse
Affiliation(s)
- Sigrid Bülow
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Lisa Zeller
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Maren Werner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Martina Toelge
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Jonas Holzinger
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | | | | | - Franziska Waldow
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomcis of Microbes, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
19
|
Uliano-Silva M, Dondero F, Dan Otto T, Costa I, Lima NCB, Americo JA, Mazzoni CJ, Prosdocimi F, Rebelo MDF. A hybrid-hierarchical genome assembly strategy to sequence the invasive golden mussel, Limnoperna fortunei. Gigascience 2018; 7:4750781. [PMID: 29267857 PMCID: PMC5836269 DOI: 10.1093/gigascience/gix128] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/11/2017] [Indexed: 11/13/2022] Open
Abstract
Background For more than 25 years, the golden mussel, Limnoperna fortunei, has aggressively invaded South American freshwaters, having travelled more than 5000 km upstream across 5 countries. Along the way, the golden mussel has outcompeted native species and economically harmed aquaculture, hydroelectric powers, and ship transit. We have sequenced the complete genome of the golden mussel to understand the molecular basis of its invasiveness and search for ways to control it. Findings We assembled the 1.6-Gb genome into 20 548 scaffolds with an N50 length of 312 Kb using a hybrid and hierarchical assembly strategy from short and long DNA reads and transcriptomes. A total of 60 717 coding genes were inferred from a customized transcriptome-trained AUGUSTUS run. We also compared predicted protein sets with those of complete molluscan genomes, revealing an exacerbation of protein-binding domains in L. fortunei. Conclusions We built one of the best bivalve genome assemblies available using a cost-effective approach using Illumina paired-end, mate-paired, and PacBio long reads. We expect that the continuous and careful annotation of L. fortunei's genome will contribute to the investigation of bivalve genetics, evolution, and invasiveness, as well as to the development of biotechnological tools for aquatic pest control.
Collapse
Affiliation(s)
- Marcela Uliano-Silva
- Carlos Chagas Filho Biophysics Institute (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Evolutionary Genetics, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany.,Berlin Center for Genomics in Biodiversity Research, Berlin, Germany
| | - Francesco Dondero
- Department of Science and Technological Innovation (DiSIT), Università del Piemonte Orientale Amedeo Avogadro, Vercelli-Novara-Alessandria, Italy
| | - Thomas Dan Otto
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK.,Centre of Immunobiology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Igor Costa
- Leopoldo de Meis Biomedical Biochemistry Institute (IBqM), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nicholas Costa Barroso Lima
- Leopoldo de Meis Biomedical Biochemistry Institute (IBqM), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Bioinformatics Laboratory (LabInfo) of the National Laboratory for Scientific Computing, Petrópolis, Rio de Janeiro, Brazil
| | - Juliana Alves Americo
- Carlos Chagas Filho Biophysics Institute (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila Junqueira Mazzoni
- Department of Evolutionary Genetics, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany.,Berlin Center for Genomics in Biodiversity Research, Berlin, Germany
| | - Francisco Prosdocimi
- Leopoldo de Meis Biomedical Biochemistry Institute (IBqM), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauro de Freitas Rebelo
- Carlos Chagas Filho Biophysics Institute (IBCCF), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Voß F, Kohler TP, Meyer T, Abdullah MR, van Opzeeland FJ, Saleh M, Michalik S, van Selm S, Schmidt F, de Jonge MI, Hammerschmidt S. Intranasal Vaccination With Lipoproteins Confers Protection Against Pneumococcal Colonisation. Front Immunol 2018; 9:2405. [PMID: 30405609 PMCID: PMC6202950 DOI: 10.3389/fimmu.2018.02405] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/28/2018] [Indexed: 12/29/2022] Open
Abstract
Streptococcus pneumoniae is endowed with a variety of surface-exposed proteins representing putative vaccine candidates. Lipoproteins are covalently anchored to the cell membrane and highly conserved among pneumococcal serotypes. Here, we evaluated these lipoproteins for their immunogenicity and protective potential against pneumococcal colonisation. A multiplex-based immunoproteomics approach revealed the immunogenicity of selected lipoproteins. High antibody titres were measured in sera from mice immunised with the lipoproteins MetQ, PnrA, PsaA, and DacB. An analysis of convalescent patient sera confirmed the immunogenicity of these lipoproteins. Examining the surface localisation and accessibility of the lipoproteins using flow cytometry indicated that PnrA and DacB were highly abundant on the surface of the bacteria. Mice were immunised intranasally with PnrA, DacB, and MetQ using cholera toxin subunit B (CTB) as an adjuvant, followed by an intranasal challenge with S. pneumoniae D39. PnrA protected the mice from pneumococcal colonisation. For the immunisation with DacB and MetQ, a trend in reducing the bacterial load could be observed, although this effect was not statistically significant. The reduction in bacterial colonisation was correlated with the increased production of antigen-specific IL-17A in the nasal cavity. Immunisation induced high systemic IgG levels with a predominance for the IgG1 isotype, except for DacB, where IgG levels were substantially lower compared to MetQ and PnrA. Our results indicate that lipoproteins are interesting targets for future vaccine strategies as they are highly conserved, abundant, and immunogenic.
Collapse
Affiliation(s)
- Franziska Voß
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Tanja Meyer
- Department of Functional Genomics, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Mohammed R Abdullah
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Fred J van Opzeeland
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Malek Saleh
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Stephan Michalik
- Department of Functional Genomics, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Saskia van Selm
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank Schmidt
- Department of Functional Genomics, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,ZIK-FunGene, Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Marien I de Jonge
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomics of Microbes, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
21
|
Solanki V, Tiwari M, Tiwari V. Host-bacteria interaction and adhesin study for development of therapeutics. Int J Biol Macromol 2018; 112:54-64. [PMID: 29414732 DOI: 10.1016/j.ijbiomac.2018.01.151] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/15/2022]
Abstract
Host-pathogen interaction is one of the most important areas of study to understand the adhesion of the pathogen to the host organisms. To adhere on the host cell surface, bacteria assemble the diverse adhesive structures on its surface, which play a foremost role in targeting to the host cell. We have highlighted different bacterial adhesins which are either protein mediated or glycan mediated. The present article listed examples of different bacterial adhesin proteins involved in the interactions with their host, types and subtypes of the fimbriae and non-fimbriae bacterial adhesins. Different bacterial surface adhesin subunits interact with host via different host surface biomolecules. We have also discussed the interactome of some of the pathogens with their host. Therefore, the present study will help researchers to have a detailed understanding of different interacting bacterial adhesins and henceforth, develop new therapies, adhesin specific antibodies and vaccines, which can effectively control pathogenicity of the pathogens.
Collapse
Affiliation(s)
- Vandana Solanki
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer 305817, India.
| |
Collapse
|
22
|
Kanwal S, Jensch I, Palm GJ, Brönstrup M, Rohde M, Kohler TP, Somplatzki D, Tegge W, Jenkinson HF, Hammerschmidt S. Mapping the recognition domains of pneumococcal fibronectin-binding proteins PavA and PavB demonstrates a common pattern of molecular interactions with fibronectin type III repeats. Mol Microbiol 2017; 105:839-859. [PMID: 28657670 DOI: 10.1111/mmi.13740] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2017] [Indexed: 11/29/2022]
Abstract
Colonization of mucosal respiratory surfaces is a prerequisite for the human pathobiont Streptococcus pneumoniae (the pneumococcus) to cause severe invasive infections. The arsenal of pneumococcal adhesins interacts with a multitude of extracellular matrix proteins. A paradigm for pneumococci is their interaction with the adhesive glycoprotein fibronectin, which facilitates bacterial adherence to host cells. Here, we deciphered the molecular interaction between fibronectin and pneumococcal fibronectin-binding proteins (FnBPs) PavA and PavB respectively. We show in adherence and binding studies that the pneumococcal interaction with fibronectin is a non-human specific trait. PavA and PavB target at least 13 out of 15 type III fibronectin domains as demonstrated in ligand overlay assays, surface plasmon resonance studies and SPOT peptide arrays. Strikingly, both pneumococcal FnBPs recognize similar peptides in targeted type III repeats. Structural comparisons revealed that the targeted type III repeat epitopes cluster on the inner strands of both β-sheets forming the fibronectin domains. Importantly, synthetic peptides of FnIII1 , FnIII5 or FnIII15 bind directly to FnBPs PavA and PavB respectively. In conclusion, our study suggests a common pattern of molecular interactions between pneumococcal FnBPs and fibronectin. The specific epitopes recognized in this study can potentially be tested as antimicrobial targets in further scientific endeavours.
Collapse
Affiliation(s)
- Sajida Kanwal
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, D-17487, Germany
| | - Inga Jensch
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, D-17487, Germany
| | - Gottfried J Palm
- Department of Structural Biology, Institute for Biochemistry, University of Greifswald, Greifswald, D-17487, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research and German Centre for Infection Research (DZIF), Braunschweig, D-38124, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, ZEIM, Helmholtz Centre for Infection Research, Braunschweig, D-38124, Germany
| | - Thomas P Kohler
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, D-17487, Germany
| | - Daniela Somplatzki
- Research Center for Infectious Diseases, University of Würzburg, Würzburg, D-97070, Germany
| | - Werner Tegge
- Department of Chemical Biology, Helmholtz Centre for Infection Research and German Centre for Infection Research (DZIF), Braunschweig, D-38124, Germany
| | - Howard F Jenkinson
- Department of Oral and Dental Science, University of Bristol, Bristol, UK
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, D-17487, Germany.,Research Center for Infectious Diseases, University of Würzburg, Würzburg, D-97070, Germany
| |
Collapse
|
23
|
Binsker U, Kohler TP, Krauel K, Kohler S, Habermeyer J, Schwertz H, Hammerschmidt S. Serotype 3 pneumococci sequester platelet-derived human thrombospondin-1 via the adhesin and immune evasion protein Hic. J Biol Chem 2017; 292:5770-5783. [PMID: 28209711 DOI: 10.1074/jbc.m116.760504] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/15/2017] [Indexed: 11/06/2022] Open
Abstract
Streptococcus pneumoniae serotype 3 strains emerge frequently within clinical isolates of invasive diseases. Bacterial invasion into deeper tissues is associated with colonization and immune evasion mechanisms. Thus, pneumococci express a versatile repertoire of surface proteins sequestering and interacting specifically with components of the human extracellular matrix and serum. Hic, a PspC-like pneumococcal surface protein, possesses vitronectin and factor H binding activity. Here, we show that heterologously expressed Hic domains interact, similar to the classical PspC molecule, with human matricellular thrombospondin-1 (hTSP-1). Binding studies with isolated human thrombospondin-1 and various Hic domains suggest that the interaction between hTSP-1 and Hic differs from binding to vitronectin and factor H. Binding of Hic to hTSP-1 is inhibited by heparin and chondroitin sulfate A, indicating binding to the N-terminal globular domain or type I repeats of hTSP-1. Competitive inhibition experiments with other pneumococcal hTSP-1 adhesins demonstrated that PspC and PspC-like Hic recognize similar domains, whereas PavB and Hic can bind simultaneously to hTSP-1. In conclusion, Hic binds specifically hTSP-1; however, truncation in the N-terminal part of Hic decreases the binding activity, suggesting that the full length of the α-helical regions of Hic is required for an optimal interaction.
Collapse
Affiliation(s)
- Ulrike Binsker
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, D-17487 Greifswald, Germany and
| | - Thomas P Kohler
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, D-17487 Greifswald, Germany and
| | - Krystin Krauel
- the Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, D-17489 Greifswald, Germany
| | - Sylvia Kohler
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, D-17487 Greifswald, Germany and
| | - Johanna Habermeyer
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, D-17487 Greifswald, Germany and
| | - Hansjörg Schwertz
- the Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, D-17489 Greifswald, Germany
| | - Sven Hammerschmidt
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, D-17487 Greifswald, Germany and
| |
Collapse
|
24
|
Gutiérrez-Fernández J, Saleh M, Alcorlo M, Gómez-Mejía A, Pantoja-Uceda D, Treviño MA, Voß F, Abdullah MR, Galán-Bartual S, Seinen J, Sánchez-Murcia PA, Gago F, Bruix M, Hammerschmidt S, Hermoso JA. Modular Architecture and Unique Teichoic Acid Recognition Features of Choline-Binding Protein L (CbpL) Contributing to Pneumococcal Pathogenesis. Sci Rep 2016; 6:38094. [PMID: 27917891 PMCID: PMC5137146 DOI: 10.1038/srep38094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 11/04/2016] [Indexed: 12/27/2022] Open
Abstract
The human pathogen Streptococcus pneumoniae is decorated with a special class of surface-proteins known as choline-binding proteins (CBPs) attached to phosphorylcholine (PCho) moieties from cell-wall teichoic acids. By a combination of X-ray crystallography, NMR, molecular dynamics techniques and in vivo virulence and phagocytosis studies, we provide structural information of choline-binding protein L (CbpL) and demonstrate its impact on pneumococcal pathogenesis and immune evasion. CbpL is a very elongated three-module protein composed of (i) an Excalibur Ca2+-binding domain -reported in this work for the very first time-, (ii) an unprecedented anchorage module showing alternate disposition of canonical and non-canonical choline-binding sites that allows vine-like binding of fully-PCho-substituted teichoic acids (with two choline moieties per unit), and (iii) a Ltp_Lipoprotein domain. Our structural and infection assays indicate an important role of the whole multimodular protein allowing both to locate CbpL at specific places on the cell wall and to interact with host components in order to facilitate pneumococcal lung infection and transmigration from nasopharynx to the lungs and blood. CbpL implication in both resistance against killing by phagocytes and pneumococcal pathogenesis further postulate this surface-protein as relevant among the pathogenic arsenal of the pneumococcus.
Collapse
Affiliation(s)
- Javier Gutiérrez-Fernández
- Department of Crystallography and Structural Biology, "Rocasolano" Institute of Physical-Chemistry, CSIC, Serrano 119, E-28006-Madrid, Spain
| | - Malek Saleh
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, D-17487 Greifswald, Germany
| | - Martín Alcorlo
- Department of Crystallography and Structural Biology, "Rocasolano" Institute of Physical-Chemistry, CSIC, Serrano 119, E-28006-Madrid, Spain
| | - Alejandro Gómez-Mejía
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, D-17487 Greifswald, Germany
| | - David Pantoja-Uceda
- Department of Biological Physical Chemistry. "Rocasolano" Institute of Physical-Chemistry, CSIC, Serrano 119, E-28006-Madrid, Spain
| | - Miguel A Treviño
- Department of Biological Physical Chemistry. "Rocasolano" Institute of Physical-Chemistry, CSIC, Serrano 119, E-28006-Madrid, Spain
| | - Franziska Voß
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, D-17487 Greifswald, Germany
| | - Mohammed R Abdullah
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, D-17487 Greifswald, Germany
| | - Sergio Galán-Bartual
- Department of Crystallography and Structural Biology, "Rocasolano" Institute of Physical-Chemistry, CSIC, Serrano 119, E-28006-Madrid, Spain
| | - Jolien Seinen
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, D-17487 Greifswald, Germany
| | - Pedro A Sánchez-Murcia
- Department of Biomedical Sciences, Unidad Asociada al IQM-CSIC, Universidad de Alcalá, E-28871 Alcalá de Henares, Madrid, Spain
| | - Federico Gago
- Department of Biomedical Sciences, Unidad Asociada al IQM-CSIC, Universidad de Alcalá, E-28871 Alcalá de Henares, Madrid, Spain
| | - Marta Bruix
- Department of Biological Physical Chemistry. "Rocasolano" Institute of Physical-Chemistry, CSIC, Serrano 119, E-28006-Madrid, Spain
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, D-17487 Greifswald, Germany
| | - Juan A Hermoso
- Department of Crystallography and Structural Biology, "Rocasolano" Institute of Physical-Chemistry, CSIC, Serrano 119, E-28006-Madrid, Spain
| |
Collapse
|
25
|
Host-derived extracellular RNA promotes adhesion of Streptococcus pneumoniae to endothelial and epithelial cells. Sci Rep 2016; 6:37758. [PMID: 27892961 PMCID: PMC5125276 DOI: 10.1038/srep37758] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/31/2016] [Indexed: 12/27/2022] Open
Abstract
Streptococcus pneumoniae is the most frequent cause of community-acquired pneumonia. The infection process involves bacterial cell surface receptors, which interact with host extracellular matrix components to facilitate colonization and dissemination of bacteria. Here, we investigated the role of host-derived extracellular RNA (eRNA) in the process of pneumococcal alveolar epithelial cell infection. Our study demonstrates that eRNA dose-dependently increased S. pneumoniae invasion of alveolar epithelial cells. Extracellular enolase (Eno), a plasminogen (Plg) receptor, was identified as a novel eRNA-binding protein on S. pneumoniae surface, and six Eno eRNA-binding sites including a C-terminal 15 amino acid motif containing lysine residue 434 were characterized. Although the substitution of lysine 434 for glycine (K434G) markedly diminished the binding of eRNA to Eno, the adherence to and internalization into alveolar epithelial cells of S. pneumoniae strain carrying the C-terminal lysine deletion and the mutation of internal Plg-binding motif were only marginally impaired. Accordingly, using a mass spectrometric approach, we identified seven novel eRNA-binding proteins in pneumococcal cell wall. Given the high number of eRNA-interacting proteins on pneumococci, treatment with RNase1 completely inhibited eRNA-mediated pneumococcal alveolar epithelial cell infection. Our data support further efforts to employ RNAse1 as an antimicrobial agent to combat pneumococcal infectious diseases.
Collapse
|
26
|
Manning J, Dunne EM, Wescombe PA, Hale JDF, Mulholland EK, Tagg JR, Robins-Browne RM, Satzke C. Investigation of Streptococcus salivarius-mediated inhibition of pneumococcal adherence to pharyngeal epithelial cells. BMC Microbiol 2016; 16:225. [PMID: 27681377 PMCID: PMC5041332 DOI: 10.1186/s12866-016-0843-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 09/20/2016] [Indexed: 02/04/2023] Open
Abstract
Background Pneumococcal adherence to the nasopharyngeal epithelium is a critical step in colonisation and disease. The probiotic bacterium, Streptococcus salivarius, can inhibit pneumococcal adherence to epithelial cells in vitro. We investigated the mechanism(s) of inhibition using a human pharyngeal epithelial cell line (Detroit 562) following pre-administration of two different strains of S. salivarius. Results Whilst the bacteriocin-encoding megaplasmids of S. salivarius strains K12 and M18 were essential to prevent pneumococcal growth on solid media, they were not required to inhibit pneumococcal adherence. Experiments testing S. salivarius K12 and two pneumococcal isolates (serotypes 19F and 6A) showed that inhibition of 19F may involve S. salivarius-mediated blocking of pneumococcal binding sites: a negative correlation was observed between adherence of K12 and 19F, and no inhibition occurred when K12 was prevented from contacting epithelial cells. K12-mediated inhibition of adherence by 6A may involve additional mechanisms, since no correlation was observed between adherence of K12 and 6A, and K12 could inhibit 6A adherence in the absence of cell contact. Conclusions These results suggest that S. salivarius employs several mechanisms, including blocking pneumococcal binding sites, to reduce pneumococcal adherence to pharyngeal epithelial cells. These findings extend our understanding of how probiotics may inhibit pneumococcal adherence and could assist with the development of novel strategies to prevent pneumococcal colonisation in the future. Electronic supplementary material The online version of this article (doi:10.1186/s12866-016-0843-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jayne Manning
- Pneumococcal Research, Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Eileen M Dunne
- Pneumococcal Research, Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia
| | | | | | - E Kim Mulholland
- Pneumococcal Research, Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,London School of Hygiene and Tropical Medicine, London, UK
| | - John R Tagg
- Blis Technologies Ltd, Dunedin, New Zealand.,Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Roy M Robins-Browne
- Infectious Diseases and Microbiology, Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Catherine Satzke
- Pneumococcal Research, Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia. .,Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
27
|
Peterson RA, Gueniche A, Adam de Beaumais S, Breton L, Dalko-Csiba M, Packer NH. Sweating the small stuff: Glycoproteins in human sweat and their unexplored potential for microbial adhesion. Glycobiology 2015; 26:218-29. [DOI: 10.1093/glycob/cwv102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/02/2015] [Indexed: 12/19/2022] Open
|
28
|
Genome-Wide Association Study of Staphylococcus aureus Carriage in a Community-Based Sample of Mexican-Americans in Starr County, Texas. PLoS One 2015; 10:e0142130. [PMID: 26569114 PMCID: PMC4646511 DOI: 10.1371/journal.pone.0142130] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/16/2015] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is the number one cause of hospital-acquired infections. Understanding host pathogen interactions is paramount to the development of more effective treatment and prevention strategies. Therefore, whole exome sequence and chip-based genotype data were used to conduct rare variant and genome-wide association analyses in a Mexican-American cohort from Starr County, Texas to identify genes and variants associated with S. aureus nasal carriage. Unlike most studies of S. aureus that are based on hospitalized populations, this study used a representative community sample. Two nasal swabs were collected from participants (n = 858) 11–17 days apart between October 2009 and December 2013, screened for the presence of S. aureus, and then classified as either persistent, intermittent, or non-carriers. The chip-based and exome sequence-based single variant association analyses identified 1 genome-wide significant region (KAT2B) for intermittent and 11 regions suggestively associated with persistent or intermittent S. aureus carriage. We also report top findings from gene-based burden analyses of rare functional variation. Notably, we observed marked differences between signals associated with persistent and intermittent carriage. In single variant analyses of persistent carriage, 7 of 9 genes in suggestively associated regions and all 5 top gene-based findings are associated with cell growth or tight junction integrity or are structural constituents of the cytoskeleton, suggesting that variation in genes associated with persistent carriage impact cellular integrity and morphology.
Collapse
|
29
|
Pulmonary immunostimulation with MALP-2 in influenza virus-infected mice increases survival after pneumococcal superinfection. Infect Immun 2015; 83:4617-29. [PMID: 26371127 DOI: 10.1128/iai.00948-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/08/2015] [Indexed: 02/06/2023] Open
Abstract
Pulmonary infection with influenza virus is frequently complicated by bacterial superinfection, with Streptococcus pneumoniae being the most prevalent causal pathogen and hence often associated with high morbidity and mortality rates. Local immunosuppression due to pulmonary influenza virus infection has been identified as a major cause of the pathogenesis of secondary bacterial lung infection. Thus, specific local stimulation of the pulmonary innate immune system in subjects with influenza virus infection might improve the host defense against secondary bacterial pathogens. In the present study, we examined the effect of pulmonary immunostimulation with Toll-like receptor 2 (TLR-2)-stimulating macrophage-activating lipopeptide 2 (MALP-2) in influenza A virus (IAV)-infected mice on the course of subsequent pneumococcal superinfection. Female C57BL/6N mice infected with IAV were treated with MALP-2 on day 5 and challenged with S. pneumoniae on day 6. Intratracheal MALP-2 application increased proinflammatory cytokine and chemokine release and enhanced the recruitment of leukocytes, mainly neutrophils, into the alveolar space of IAV-infected mice, without detectable systemic side effects. Local pulmonary instillation of MALP-2 in IAV-infected mice 24 h before transnasal pneumococcal infection considerably reduced the bacterial number in the lung tissue without inducing exaggerated inflammation. The pulmonary viral load was not altered by MALP-2. Clinically, MALP-2 treatment of IAV-infected mice increased survival rates and reduced hypothermia and body weight loss after pneumococcal superinfection compared to those of untreated coinfected mice. In conclusion, local immunostimulation with MALP-2 in influenza virus-infected mice improved pulmonary bacterial elimination and increased survival after subsequent pneumococcal superinfection.
Collapse
|
30
|
Terzulli M, Ruiz LC, Kugadas A, Masli S, Gadjeva M. TSP-1 Deficiency Alters Ocular Microbiota: Implications for Sjögren's Syndrome Pathogenesis. J Ocul Pharmacol Ther 2015; 31:413-8. [PMID: 26352162 PMCID: PMC4575514 DOI: 10.1089/jop.2015.0017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/19/2015] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The potential role of commensals as triggering factors that promote inflammation in dry eye disease has not been explored. The objective of this study was to evaluate whether ocular microbiota changes with the onset of dry eye disease in thrombospondin-1-deficient (TSP-1(-/-)) mice, a strain that develops Sjögren's syndrome-like disease. METHODS Conjunctival swabs were collected from TSP-1(-/-) and C57BL/6 mice and analyzed for bacterial presence. Opsonophagocytosis of the bacterial conjunctival isolates derived from the aged TSP-1(-/-) mice by neutrophils derived from either TSP-1(-/-) or C57BL/6 bone marrow was evaluated. The bactericidal activities of TSP-1-derived peptide were examined. RESULTS We found that in TSP-1(-/-) mice, the conjunctival colonization with Staphylococcus aureus and coagulase negative staphylococci sp (CNS) species was significantly increased with aging and preceded that of the wild-type C57BL/6 control mice. This correlated with increased neutrophil infiltration into the conjunctiva of the TSP-1(-/-) mice, suggesting that TSP-1 plays a significant role in regulating immunity to commensals. Accordingly, the TSP-1(-/-) PMNs opsonophagocytozed the ocular commensals less efficiently than the TSP-1-sufficient neutrophils. Furthermore, a TSP-1-derived peptide, 4N1K, exhibited significant antimicrobial activity when compared to a control peptide against commensal sp. CONCLUSION These studies illustrate that alterations in the commensal frequency occur in the early stages of development of Sjögren's-like pathology and suggest that interventions that limit commensal outgrowth such as the use of TSP-1-derived peptides could be used for treatment during the early stages of the disease to reduce the commensal burden and ensuing inflammation.
Collapse
Affiliation(s)
- Marielle Terzulli
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Abirami Kugadas
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sharmila Masli
- Department of Ophthalmology, Boston University, School of Medicine, Boston, MA
| | - Mihaela Gadjeva
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
31
|
Gonzalez-Aspajo G, Belkhelfa H, Haddioui-Hbabi L, Bourdy G, Deharo E. Sacha Inchi Oil (Plukenetia volubilis L.), effect on adherence of Staphylococus aureus to human skin explant and keratinocytes in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2015; 171:330-334. [PMID: 26087228 DOI: 10.1016/j.jep.2015.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plukenetia volubilis L. (Euphorbiaceae) is a domesticated vine distributed from the high-altitude Andean rain forest to the lowlands of the Peruvian Amazon. Oil from the cold-pressed seeds, sold under the commercial name of Sacha Inchi Oil (SIO) is actually much in favour because it contains a high percentage of omega 3 and omega 6, and is hence used as a dietary supplement. SIO is also used traditionally for skin care, in order to maintain skin softness, and for the treatment of wounds, insect bites and skin infections, in a tropical context where the skin is frequently damaged. AIMS OF THE STUDY This study was designed in order to verify whether the traditional use of SIO for skin care would have any impact on Staphylococcus aureus growth and skin adherence, as S. aureus is involved in many skin pathologies (impetigo, folliculitis, furuncles and subcutaneous abscesses) being one if the main pathogens that can be found on the skin. Therefore, our objective was to assess SIO bactericidal activity and interference with adherence to human skin explants and the keratinocyte cell line. Cytotoxicity on that cells was also determined. The activity of SIO was compared to coconut oil (CocO), which is widely used for skin care but has different unsaturated fatty acids contents. MATERIALS AND METHODS Laboratory testing with certified oil, determined antibacterial activity against radio labelled S. aureus. Cytotoxic effects were measured with XTT on keratinocyte cells and with neutral red on human skin explants; phenol was used as cytotoxic control. Adherence assays were carried out by mixing H3-labelled S. aureus bacteria with keratinocyte cells and human skin explants, incubated with oils 2h before (to determine the inhibition of adherence, assimilated to a preventive effect) or 2h after the contact of the biological material with S. aureus (to assess the detachment of the bacteria, assimilated to a curative effect). Residual radioactivity measured after washings made it possible to determine the adherence intensity. Bactericidal effect was determined by colony counting on trypticase soy agar. RESULTS Laboratory assays showed that SIO and CocO, tested undiluted, were not cytotoxic on keratinocytes nor human explants and were not bactericidal neither. SIO was more active as antiadherent (preventive) than CocO on keratinocytes. There was no significant difference between detachment effects (curative) of both oils on keratinocytes but SIO was almost 5 times more active on the detachment of S. aureus from human skin explants. CONCLUSION From that study it can be concluded that the use of SIO on dermal cells is safe and efficient in the inhibition of S. aureus adherence. Our results tend to support the traditional use of undiluted SIO in skin care.
Collapse
Affiliation(s)
- German Gonzalez-Aspajo
- Institut de Recherche pour le Développement (IRD), UMR 152 Pharma-DEV, F-31062 Toulouse cedex 09, France; Université de Toulouse 3, UMR 152 Pharma-DEV, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse cedex 09, France
| | - Haouaria Belkhelfa
- Fonderephar, Université Toulouse 3, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse cedex 09, France
| | - Laïla Haddioui-Hbabi
- Fonderephar, Université Toulouse 3, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse cedex 09, France
| | - Geneviève Bourdy
- Institut de Recherche pour le Développement (IRD), UMR 152 Pharma-DEV, F-31062 Toulouse cedex 09, France; Université de Toulouse 3, UMR 152 Pharma-DEV, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse cedex 09, France
| | - Eric Deharo
- Institut de Recherche pour le Développement (IRD), UMR 152 Pharma-DEV, F-31062 Toulouse cedex 09, France; Université de Toulouse 3, UMR 152 Pharma-DEV, Faculté des Sciences Pharmaceutiques, F-31062 Toulouse cedex 09, France.
| |
Collapse
|
32
|
Binsker U, Kohler TP, Krauel K, Kohler S, Schwertz H, Hammerschmidt S. Pneumococcal Adhesins PavB and PspC Are Important for the Interplay with Human Thrombospondin-1. J Biol Chem 2015; 290:14542-55. [PMID: 25897078 DOI: 10.1074/jbc.m114.623876] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Indexed: 11/06/2022] Open
Abstract
The human matricellular glycoprotein thrombospondin-1 (hTSP-1) is released by activated platelets and mediates adhesion of Gram-positive bacteria to various host cells. In staphylococci, the adhesins extracellular adherence protein (Eap) and autolysin (Atl), both surface-exposed proteins containing repeating structures, were shown to be involved in the acquisition of hTSP-1 to the bacterial surface. The interaction partner(s) on the pneumococcal surface was hitherto unknown. Here, we demonstrate for the first time that pneumococcal adherence and virulence factor B (PavB) and pneumococcal surface protein C (PspC) are key players for the interaction of Streptococcus pneumoniae with matricellular hTSP-1. PavB and PspC are pneumococcal surface-exposed adhesins and virulence factors exhibiting repetitive sequences in their core structure. Heterologously expressed fragments of PavB and PspC containing repetitive structures exhibit hTSP-1 binding activity as shown by ELISA and surface plasmon resonance studies. Binding of hTSP-1 is charge-dependent and inhibited by heparin. Importantly, the deficiency in PavB and PspC reduces the recruitment of soluble hTSP-1 by pneumococci and decreases hTSP-1-mediated pneumococcal adherence to human epithelial cells. Platelet activation assays suggested that PavB and PspC are not involved in the activation of purified human platelets by pneumococci. In conclusion, this study indicates a pivotal role of PavB and PspC for pneumococcal recruitment of soluble hTSP-1 to the bacterial surface and binding of pneumococci to host cell-bound hTSP-1 during adhesion.
Collapse
Affiliation(s)
- Ulrike Binsker
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany and
| | - Thomas P Kohler
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany and
| | - Krystin Krauel
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, D-17489 Greifswald, Germany
| | - Sylvia Kohler
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany and
| | - Hansjörg Schwertz
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, D-17489 Greifswald, Germany
| | - Sven Hammerschmidt
- From the Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany and
| |
Collapse
|
33
|
Mohan S, Hertweck C, Dudda A, Hammerschmidt S, Skerka C, Hallström T, Zipfel PF. Tuf of Streptococcus pneumoniae is a surface displayed human complement regulator binding protein. Mol Immunol 2014; 62:249-64. [PMID: 25046156 DOI: 10.1016/j.molimm.2014.06.029] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/21/2014] [Accepted: 06/23/2014] [Indexed: 11/15/2022]
Abstract
Streptococcus pneumoniae is a Gram-positive bacterium, causing acute sinusitis, otitis media, and severe diseases such as pneumonia, bacteraemia, meningitis and sepsis. Here we identify elongation factor Tu (Tuf) as a new Factor H binding protein of S. pneumoniae. The surface protein PspC which also binds a series of other human immune inhibitors, was the first identified pneumococcal Factor H binding protein of S. pneumoniae. Pneumococcal Tuf, a 55 kDa pneumococcal moonlighting protein which is displayed on the surface of pneumococci, is also located in the cytoplasm and is detected in the culture supernatant. Tuf binds the human complement inhibitors Factor H, FHL-1, CFHR1 and also the proenzyme plasminogen. Factor H and FHL-1 bound to Tuf, retain their complement regulatory activities. Similarly, plasminogen bound to Tuf was accessible for the activator uPA and activated plasmin cleaved the synthetic chromogenic substrate S-2251 as well as the natural substrates fibrinogen and the complement proteins C3 and C3b. Taken together, Tuf of S. pneumoniae is a new multi-functional bacterial virulence factor that helps the pathogen in complement escape and likely also in ECM degradation.
Collapse
Affiliation(s)
- Sarbani Mohan
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Christian Hertweck
- Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Antje Dudda
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University, Greifswald, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Teresia Hallström
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany; Faculty of Biology, Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
34
|
Kohler S, Hallström T, Singh B, Riesbeck K, Spartà G, Zipfel PF, Hammerschmidt S. Binding of vitronectin and Factor H to Hic contributes to immune evasion of Streptococcus pneumoniae serotype 3. Thromb Haemost 2014; 113:125-42. [PMID: 25181963 DOI: 10.1160/th14-06-0561] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 07/24/2014] [Indexed: 11/05/2022]
Abstract
Streptococcus pneumoniae serotype 3 strains are highly resistant to opsonophagocytosis due to recruitment of the complement inhibitor Factor H via Hic, a member of the pneumococcal surface protein C (PspC) family. In this study, we demonstrated that Hic also interacts with vitronectin, a fluid-phase regulator involved in haemostasis, angiogenesis, and the terminal complement cascade as well as a component of the extracellular matrix. Blocking of Hic by specific antiserum or genetic deletion significantly reduced pneumococcal binding to soluble and immobilised vitronectin and to Factor H, respectively. In parallel, ectopic expression of Hic on the surface of Lactococcus lactis conferred binding to soluble and immobilised vitronectin as well as Factor H. Molecular analyses with truncated Hic fragments narrowed down the vitronectin-binding site to the central core of Hic (aa 151-201). This vitronectin-binding region is separate from that of Factor H, which binds to the N-terminus of Hic (aa 38-92). Binding of pneumococcal Hic was localised to the C-terminal heparin-binding domain (HBD3) of vitronectin. However, an N-terminal region to HBD3 was further involved in Hic-binding to immobilised vitronectin. Finally, vitronectin bound to Hic was functionally active and inhibited formation of the terminal complement complex. In conclusion, Hic interacts with vitronectin and simultaneously with Factor H, and both human proteins may contribute to colonisation and invasive disease caused by serotype 3 pneumococci.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sven Hammerschmidt
- Prof. Dr. Sven Hammerschmidt, Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University Greifswald, Friedrich-Ludwig-Jahn-Strasse 15A, 17487 Greifswald, Germany, Tel.: +49 3834 864161, Fax: +49 3834 864172, E-mail:
| |
Collapse
|
35
|
Tie L, Chen LY, Chen DD, Xie HH, Channon KM, Chen AF. GTP cyclohydrolase I prevents diabetic-impaired endothelial progenitor cells and wound healing by suppressing oxidative stress/thrombospondin-1. Am J Physiol Endocrinol Metab 2014; 306:E1120-31. [PMID: 24644242 DOI: 10.1152/ajpendo.00696.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cell (EPC) dysfunction is a key contributor to diabetic refractory wounds. Endothelial nitric oxide synthase (eNOS), which critically regulates the mobilization and function of EPCs, is uncoupled in diabetes due to decreased cofactor tetrahydrobiopterin (BH4). We tested whether GTP cyclohydrolase I (GTPCH I), the rate-limiting enzyme of BH4 synthesis, preserves EPC function in type 1 diabetic mice. Type 1 diabetes was induced in wild-type (WT) and GTPCH I transgenic (Tg-GCH) mice by intraperitoneal injection of streptozotocin (STZ). EPCs were isolated from the peripheral blood and bone marrow of WT, Tg-GCH, and GTPCH I-deficient hph-1 mice. The number of EPCs was significantly lower in STZ-WT mice and hph-1 mice and was rescued in STZ Tg-GCH mice. Furthermore, GTPCH I overexpression improved impaired diabetic EPC migration and tube formation. EPCs from WT, Tg-GCH, and STZ-Tg-GCH mice were administered to diabetic excisional wounds and accelerated wound healing significantly, with a concomitant augmentation of angiogenesis. Flow cytometry measurements showed that intracellular nitric oxide (NO) levels were reduced significantly in STZ-WT and hph-1 mice, paralleled by increased superoxide anion levels; both were rescued in STZ-Tg-GCH mice. Western blot analysis revealed that thrombospondin-1 (TSP-1) was significantly upregulated in the EPCs of STZ-WT mice and hph-1 mice and suppressed in STZ-treated Tg-GCH mice. Our results demonstrate that the GTPCH I/BH4 pathway is critical to preserve EPC quantity, function, and regenerative capacity during wound healing in type 1 diabetic mice at least partly through the attenuation of superoxide and TSP-1 levels and augmentation of NO level.
Collapse
Affiliation(s)
- Lu Tie
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lu-Yuan Chen
- Department of Cardiology, Guangdong General Hospital, Guangzhou, China; and
| | - Dan-Dan Chen
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - He-Hui Xie
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Keith M Channon
- Department of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Alex F Chen
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China;
| |
Collapse
|
36
|
Agarwal V, Sroka M, Fulde M, Bergmann S, Riesbeck K, Blom AM. Binding of Streptococcus pneumoniae endopeptidase O (PepO) to complement component C1q modulates the complement attack and promotes host cell adherence. J Biol Chem 2014; 289:15833-44. [PMID: 24739385 DOI: 10.1074/jbc.m113.530212] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Gram-positive species Streptococcus pneumoniae is a human pathogen causing severe local and life-threatening invasive diseases associated with high mortality rates and death. We demonstrated recently that pneumococcal endopeptidase O (PepO) is a ubiquitously expressed, multifunctional plasminogen and fibronectin-binding protein facilitating host cell invasion and evasion of innate immunity. In this study, we found that PepO interacts directly with the complement C1q protein, thereby attenuating the classical complement pathway and facilitating pneumococcal complement escape. PepO binds both free C1q and C1 complex in a dose-dependent manner based on ionic interactions. Our results indicate that recombinant PepO specifically inhibits the classical pathway of complement activation in both hemolytic and complement deposition assays. This inhibition is due to direct interaction of PepO with C1q, leading to a strong activation of the classical complement pathway, and results in consumption of complement components. In addition, PepO binds the classical complement pathway inhibitor C4BP, thereby regulating downstream complement activation. Importantly, pneumococcal surface-exposed PepO-C1q interaction mediates bacterial adherence to host epithelial cells. Taken together, PepO facilitates C1q-mediated bacterial adherence, whereas its localized release consumes complement as a result of its activation following binding of C1q, thus representing an additional mechanism of human complement escape by this versatile pathogen.
Collapse
Affiliation(s)
| | | | - Marcus Fulde
- the Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625 Hannover, Germany, and
| | - Simone Bergmann
- the Institute of Microbiology, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Kristian Riesbeck
- Medical Microbiology, Department of Laboratory Medicine Malmö, Lund University, 20502 Malmö, Sweden
| | - Anna M Blom
- From the Divisions of Medical Protein Chemistry and
| |
Collapse
|
37
|
Pribyl T, Moche M, Dreisbach A, Bijlsma JJE, Saleh M, Abdullah MR, Hecker M, van Dijl JM, Becher D, Hammerschmidt S. Influence of impaired lipoprotein biogenesis on surface and exoproteome of Streptococcus pneumoniae. J Proteome Res 2014; 13:650-67. [PMID: 24387739 DOI: 10.1021/pr400768v] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Surface proteins are important for the fitness and virulence of the Gram-positive pathogen Streptococcus pneumoniae. They are crucial for interaction of the pathogen with its human host during infection. Therefore, the analysis of the pneumococcal surface proteome is an important task that requires powerful tools. In this study, two different methods, an optimized biotinylation approach and shaving with trypsin beads, were applied to study the pneumococcal surface proteome and to identify surface-exposed protein domains, respectively. The identification of nearly 95% of the predicted lipoproteins and 75% of the predicted sortase substrates reflects the high coverage of the two classical surface protein classes accomplished in this study. Furthermore, the biotinylation approach was applied to study the impact of an impaired lipoprotein maturation pathway on the cell envelope proteome and exoproteome. Loss of the lipoprotein diacylglyceryl transferase Lgt leads to striking changes in the lipoprotein distribution. Many lipoproteins disappear from the surface proteome and accumulate in the exoproteome. Further insights into lipoprotein processing in pneumococci are provided by immunoblot analyses of bacterial lysates and corresponding supernatant fractions. Taken together, the first comprehensive overview of the pneumococcal surface and exoproteome is presented, and a model for lipoprotein processing in S. pneumoniae is proposed.
Collapse
Affiliation(s)
- Thomas Pribyl
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald , Friedrich-Ludwig-Jahn-Str. 15a, Greifswald D-17487, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kohler TP, Gisch N, Binsker U, Schlag M, Darm K, Völker U, Zähringer U, Hammerschmidt S. Repeating structures of the major staphylococcal autolysin are essential for the interaction with human thrombospondin 1 and vitronectin. J Biol Chem 2013; 289:4070-82. [PMID: 24371140 DOI: 10.1074/jbc.m113.521229] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Human thrombospondin 1 (hTSP-1) is a matricellular glycoprotein facilitating bacterial adherence to and invasion into eukaryotic cells. However, the bacterial adhesin(s) remain elusive. In this study, we show a dose-dependent binding of soluble hTSP-1 to Gram-positive but not Gram-negative bacteria. Diminished binding of soluble hTSP-1 to proteolytically pretreated staphylococci suggested a proteinaceous nature of potential bacterial adhesin(s) for hTSP-1. A combination of separation of staphylococcal surface proteins by two-dimensional gel electrophoresis with a ligand overlay assay with hTSP-1 and identification of the target protein by mass spectrometry revealed the major staphylococcal autolysin Atl as a bacterial binding protein for hTSP-1. Binding experiments with heterologously expressed repeats of the AtlE amidase from Staphylococcus epidermidis suggest that the repeating sequences (R1ab-R2ab) of the N-acetyl-muramoyl-L-alanine amidase of Atl are essential for binding of hTSP-1. Atl has also been identified previously as a staphylococcal vitronectin (Vn)-binding protein. Similar to the interaction with hTSP-1, the R1ab-R2ab repeats of Atl are shown here to be crucial for the interaction of Atl with the complement inhibition and matrix protein Vn. Competition assays with hTSP-1 and Vn revealed the R1ab-R2ab repeats of AtlE as the common binding domain for both host proteins. Furthermore, Vn competes with hTSP-1 for binding to Atl repeats and vice versa. In conclusion, this study identifies the Atl repeats as bacterial adhesive structures interacting with the human glycoproteins hTSP-1 and Vn. Finally, this study provides insight into the molecular interplay between hTSP-1 and Vn, respectively, and a bacterial autolysin.
Collapse
|
39
|
Towhid ST, Nega M, Schmidt EM, Schmid E, Albrecht T, Münzer P, Borst O, Götz F, Lang F. Stimulation of platelet apoptosis by peptidoglycan from Staphylococcus aureus 113. Apoptosis 2013; 17:998-1008. [PMID: 22752708 DOI: 10.1007/s10495-012-0718-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Peptidoglycan (PGN), a component of bacterial cell wall and belonging to "Microbe-Associated Molecular Patterns" (MAMP) triggers host reactions contributing to the pathophysiology of infectious disease. Host cell responses to PGN exposure include apoptosis. Bacterial infections may result in activation of blood platelets and thrombocytopenia. The present study explored, whether HPLC-purified fractions of PGNs from Staphylococcus aureus 113 triggers apoptosis of platelets. To this end platelets were exposed to PGN fractions and annexin-V binding determined to depict cell membrane scrambling, DiOC6 fluorescence to estimate depolarization of mitochondrial potential, Fluo-3AM staining for intracellular Ca(2+) activity ([Ca(2+)](i)) and immunofluorescence to quantify protein abundance of active caspase-3. As a result, a 30 min exposure to monomeric fraction (mPGN) (≥50 ng/ml) was followed by annexin-V binding, paralleled by increase of [Ca(2+)](i), mitochondrial depolarization, caspase-3 activation and integrin α(IIb)β(3) upregulation. The annexin-V binding was significantly blunted by anti-TLR-2 antibodies, in absence of extracellular Ca(2+), and by pancaspase inhibitor zVAD-FMK (1 μM). In conclusion, PGN triggers apoptosis of platelets in activation-dependent manner, characterized by mitochondrial depolarization, caspase-3 activation and cell membrane scrambling.
Collapse
Affiliation(s)
- Syeda T Towhid
- Department of Physiology, University of Tübingen, Gmelinstr. 5, 72076, Tübingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Saleh M, Bartual SG, Abdullah MR, Jensch I, Asmat TM, Petruschka L, Pribyl T, Gellert M, Lillig CH, Antelmann H, Hermoso JA, Hammerschmidt S. Molecular architecture of Streptococcus pneumoniae surface thioredoxin-fold lipoproteins crucial for extracellular oxidative stress resistance and maintenance of virulence. EMBO Mol Med 2013; 5:1852-70. [PMID: 24136784 PMCID: PMC3914529 DOI: 10.1002/emmm.201202435] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 08/15/2013] [Accepted: 09/10/2013] [Indexed: 01/27/2023] Open
Abstract
The respiratory pathogen Streptococcus pneumoniae has evolved efficient mechanisms to resist oxidative stress conditions and to displace other bacteria in the nasopharynx. Here we characterize at physiological, functional and structural levels two novel surface-exposed thioredoxin-family lipoproteins, Etrx1 and Etrx2. The impact of both Etrx proteins and their redox partner methionine sulfoxide reductase SpMsrAB2 on pneumococcal pathogenesis was assessed in mouse virulence studies and phagocytosis assays. The results demonstrate that loss of function of either both Etrx proteins or SpMsrAB2 dramatically attenuated pneumococcal virulence in the acute mouse pneumonia model and that Etrx proteins compensate each other. The deficiency of Etrx proteins or SpMsrAB2 further enhanced bacterial uptake by macrophages, and accelerated pneumococcal killing by H2O2 or free methionine sulfoxides (MetSO). Moreover, the absence of both Etrx redox pathways provokes an accumulation of oxidized SpMsrAB2 in vivo. Taken together our results reveal insights into the role of two extracellular electron pathways required for reduction of SpMsrAB2 and surface-exposed MetSO. Identification of this system and its target proteins paves the way for the design of novel antimicrobials.
Collapse
Affiliation(s)
- Malek Saleh
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Greifswald, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Agarwal V, Ahl J, Riesbeck K, Blom AM. An alternative role of C1q in bacterial infections: facilitating Streptococcus pneumoniae adherence and invasion of host cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:4235-45. [PMID: 24038089 DOI: 10.4049/jimmunol.1300279] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Streptococcus pneumoniae (pneumococcus) is a major human pathogen, which evolved numerous successful strategies to colonize the host. In this study, we report a novel mechanism of pneumococcal-host interaction, whereby pneumococci use a host complement protein C1q, primarily involved in the host-defense mechanism, for colonization and subsequent dissemination. Using cell-culture infection assays and confocal microscopy, we observed that pneumococcal surface-bound C1q significantly enhanced pneumococcal adherence to and invasion of host epithelial and endothelial cells. Flow cytometry demonstrated a direct, Ab-independent binding of purified C1q to various clinical isolates of pneumococci. This interaction was seemingly capsule serotype independent and mediated by the bacterial surface-exposed proteins, as pretreatment of pneumococci with pronase E but not sodium periodate significantly reduced C1q binding. Moreover, similar binding was observed using C1 complex as the source of C1q. Furthermore, our data show that C1q bound to the pneumococcal surface through the globular heads and with the host cell-surface receptor(s)/glycosaminoglycans via its N-terminal collagen-like stalk, as the presence of C1q N-terminal fragment and low m.w. heparin but not the C-terminal globular heads blocked C1q-mediated pneumococcal adherence to host cells. Taken together, we demonstrate for the first time, to our knowledge, a unique function of complement protein C1q, as a molecular bridge between pneumococci and the host, which promotes bacterial cellular adherence and invasion. Nevertheless, in some conditions, this mechanism could be also beneficial for the host as it may result in uptake and clearance of the bacteria.
Collapse
Affiliation(s)
- Vaibhav Agarwal
- Medical Protein Chemistry, Department of Laboratory Medicine Malmö, Lund University, SE-205 02 Malmö, Sweden
| | | | | | | |
Collapse
|
42
|
The interaction between bacterial enolase and plasminogen promotes adherence of Streptococcus pneumoniae to epithelial and endothelial cells. Int J Med Microbiol 2013; 303:452-62. [PMID: 23906818 DOI: 10.1016/j.ijmm.2013.06.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/24/2013] [Accepted: 06/02/2013] [Indexed: 11/21/2022] Open
Abstract
Binding and conversion of the plasma protein plasminogen is an important pathogenesis mechanism of the human pathogen Streptococcus pneumoniae. Once converted into plasmin, the proteolytic activity of this major fibrinolysis component promotes degradation of extracellular matrix and the dissolution of fibrin clots. Here, we present the exploitation of plasminogen-binding as a further pivotal strategy of pneumococci facilitating adherence to eukaryotic cells. Flow cytometric measurements demonstrated the immobilization of plasminogen on host cell surfaces of human alveolar type II pneumocytes (A549), nasopharyngeal epithelium (Detroit 562) and brain-derived endothelial cells (HBMEC). These host-derived cells were employed in cell culture infection analyses followed by confocal microscopy to monitor the plasminogen-mediated adherence. Results of these studies revealed that host cell-bound plasminogen promotes pneumococcal adherence to human epithelial and endothelial cells in dose-dependent manner, whereas pneumococcal internalization was not enhanced. As an opposed effect pneumococcal-bound plasminogen reduced attachment to the epithelial and endothelial cells, and increased the interaction with neutrophil granulocytes. Moreover, the surface-displayed enolase, which serves as major pneumococcal plasminogen receptor, was identified as a key factor for plasminogen-mediated bacterial attachment in infection analyses with S. pneumoniae enolase mutants.
Collapse
|
43
|
Voss S, Hallström T, Saleh M, Burchhardt G, Pribyl T, Singh B, Riesbeck K, Zipfel PF, Hammerschmidt S. The choline-binding protein PspC of Streptococcus pneumoniae interacts with the C-terminal heparin-binding domain of vitronectin. J Biol Chem 2013; 288:15614-27. [PMID: 23603906 DOI: 10.1074/jbc.m112.443507] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Adherence of Streptococcus pneumoniae is directly mediated by interactions of adhesins with eukaryotic cellular receptors or indirectly by exploiting matrix and serum proteins as molecular bridges. Pneumococci engage vitronectin, the human adhesive glycoprotein and complement inhibitor, to facilitate attachment to epithelial cells of the mucosal cavity, thereby modulating host cell signaling. In this study, we identified PspC as a vitronectin-binding protein interacting with the C-terminal heparin-binding domain of vitronectin. PspC is a multifunctional surface-exposed choline-binding protein displaying various adhesive properties. Vitronectin binding required the R domains in the mature PspC protein, which are also essential for the interaction with the ectodomain of the polymeric immunoglobulin receptor and secretory IgA. Consequently, secretory IgA competitively inhibited binding of vitronectin to purified PspC and to PspC-expressing pneumococci. In contrast, Factor H, which binds to the N-terminal part of mature PspC molecules, did not interfere with the PspC-vitronectin interaction. Using a series of vitronectin peptides, the C-terminal heparin-binding domain was shown to be essential for the interaction of soluble vitronectin with PspC. Binding experiments with immobilized vitronectin suggested a region N-terminal to the identified heparin-binding domain as an additional binding region for PspC, suggesting that soluble, immobilized, as well as cellularly bound vitronectin possesses different conformations. Finally, vitronectin bound to PspC was functionally active and inhibited the deposition of the terminal complement complex. In conclusion, this study identifies and characterizes (on the molecular level) the interaction between the pneumococcal adhesin PspC and the human glycoprotein vitronectin.
Collapse
Affiliation(s)
- Sylvia Voss
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, D-17487 Greifswald, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gisch N, Kohler T, Ulmer AJ, Müthing J, Pribyl T, Fischer K, Lindner B, Hammerschmidt S, Zähringer U. Structural reevaluation of Streptococcus pneumoniae Lipoteichoic acid and new insights into its immunostimulatory potency. J Biol Chem 2013; 288:15654-67. [PMID: 23603911 DOI: 10.1074/jbc.m112.446963] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Streptococcus pneumoniae is a Gram-positive human pathogen with a complex lipoteichoic acid (pnLTA) structure. Because the current structural model for pnLTA shows substantial inconsistencies, we reinvestigated purified and, more importantly, O-deacylated pnLTA, which is most suitable for NMR spectroscopy and electrospray ionization-MS spectrometry. We analyzed pnLTA of nonencapsulated pneumococcal strains D39Δcps and TIGR4Δcps, respectively. The data obtained allowed us to (re)define (i) the position and linkage of the repeating unit, (ii) the putative α-GalpNAc substitution at the ribitiol 5-phosphate (Rib-ol-5-P), and (iii) the length of (i.e. the number of repeating units in) the pnLTA chain. We here also describe for the first time that the terminal sugar residues in the pnLTA (Forssman disaccharide; α-D-GalpNAc-(1→3)-β-D-GalpNAc-(1→)), responsible for the cross-reactivity with anti-Forssman antigen antibodies, can be heterogeneous with respect to its degree of phosphorylcholine substitution in both O-6-positions. To assess the proinflammatory potency of pnLTA, we generated a (lipopeptide-free) Δlgt mutant of strain D39Δcps, isolated its pnLTA, and showed that it is capable of inducing IL-6 release in human mononuclear cells, independent of TLR2 activation. This finding was quite in contrast to LTA of the Staphylococcus aureus SA113Δlgt mutant, which did not activate human mononuclear cells in our experiments. Remarkably, this is also contrary to various other reports showing a proinflammatory potency of S. aureus LTA. Taken together, our study refines the structure of pnLTA and indicates that pneumococcal and S. aureus LTAs differ not only in their structure but also in their bioactivity.
Collapse
Affiliation(s)
- Nicolas Gisch
- Division of Immunochemistry, Leibniz-Center for Medicine and Biosciences, Borstel, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Agarwal V, Kuchipudi A, Fulde M, Riesbeck K, Bergmann S, Blom AM. Streptococcus pneumoniae endopeptidase O (PepO) is a multifunctional plasminogen- and fibronectin-binding protein, facilitating evasion of innate immunity and invasion of host cells. J Biol Chem 2013; 288:6849-63. [PMID: 23341464 DOI: 10.1074/jbc.m112.405530] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Streptococcus pneumoniae infections remain a major cause of morbidity and mortality worldwide. Therefore a detailed understanding and characterization of the mechanism of host cell colonization and dissemination is critical to gain control over this versatile pathogen. Here we identified a novel 72-kDa pneumococcal protein endopeptidase O (PepO), as a plasminogen- and fibronectin-binding protein. Using a collection of clinical isolates, representing different serotypes, we found PepO to be ubiquitously present both at the gene and protein level. In addition, PepO protein was secreted in a growth phase-dependent manner to the culture supernatants of the pneumococcal isolates. Recombinant PepO bound human plasminogen and fibronectin in a dose-dependent manner and plasminogen did not compete with fibronectin for binding PepO. PepO bound plasminogen via lysine residues and the interaction was influenced by ionic strength. Moreover, upon activation of PepO-bound plasminogen by urokinase-type plasminogen activator, generated plasmin cleaved complement protein C3b thus assisting in complement control. Furthermore, direct binding assays demonstrated the interaction of PepO with epithelial and endothelial cells that in turn blocked pneumococcal adherence. Moreover, a pepO-mutant strain showed impaired adherence to and invasion of host cells compared with their isogenic wild-type strains. Taken together, the results demonstrated that PepO is a ubiquitously expressed plasminogen- and fibronectin-binding protein, which plays role in pneumococcal invasion of host cells and aids in immune evasion.
Collapse
Affiliation(s)
- Vaibhav Agarwal
- Department of Laboratory Medicine, Medical Protein Chemistry, Lund University, Malmö S-205 02, Sweden
| | | | | | | | | | | |
Collapse
|
46
|
Konar M, Sachin O, Priya A, Ghosh S. Identification of key proteins of cultured human intestinal cells involved in interaction with enteroaggregativeEscherichia coli. ACTA ACUST UNITED AC 2012; 66:177-90. [DOI: 10.1111/j.1574-695x.2012.00998.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 05/15/2012] [Accepted: 05/24/2012] [Indexed: 11/28/2022]
|
47
|
Platelet factor 4 binding to lipid A of Gram-negative bacteria exposes PF4/heparin-like epitopes. Blood 2012; 120:3345-52. [DOI: 10.1182/blood-2012-06-434985] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AbstractThe positively charged chemokine platelet factor 4 (PF4) forms immunogenic complexes with heparin and other polyanions. Resulting antibodies can induce the adverse drug effect heparin-induced thrombocytopenia. PF4 also binds to bacteria, thereby exposing the same neoantigen(s) as with heparin. In this study, we identified the negatively charged lipopolysaccharide (LPS) as the PF4 binding structure on Gram-negative bacteria. We demonstrate by flow cytometry that mutant bacteria with progressively truncated LPS structures show increasingly enhanced PF4 binding activity. PF4 bound strongest to mutants lacking the O-antigen and core structure of LPS, but still exposing lipid A on their surfaces. Strikingly, PF4 bound more efficiently to bisphosphorylated lipid A than to monophosphorylated lipid A, suggesting that phosphate residues of lipid A mediate PF4 binding. Interactions of PF4 with Gram-negative bacteria, where only the lipid A part of LPS is exposed, induce epitopes on PF4 resembling those on PF4/heparin complexes as shown by binding of human anti-PF4/heparin antibodies. As both the lipid A on the surface of Gram-negative bacteria and the amino acids of PF4 contributing to polyanion binding are highly conserved, our results further support the hypothesis that neoepitope formation on PF4 after binding to bacteria is an ancient host defense mechanism.
Collapse
|
48
|
Horn M, Bertling A, Brodde MF, Müller A, Roth J, Van Aken H, Jurk K, Heilmann C, Peters G, Kehrel BE. Human neutrophil alpha-defensins induce formation of fibrinogen and thrombospondin-1 amyloid-like structures and activate platelets via glycoprotein IIb/IIIa. J Thromb Haemost 2012; 10:647-61. [PMID: 22268819 DOI: 10.1111/j.1538-7836.2012.04640.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Human neutrophil α-defensins (HNPs) are important constituents of the innate immune system. Beyond their antimicrobial properties, HNPs also have pro-inflammatory features. While HNPs in plasma from healthy individuals are barely detectable, their level is strongly elevated in septic plasma and plasma from patients with acute coronary syndromes. OBJECTIVES As thrombosis and inflammation are intertwined processes and activation of human polymorphonuclear leukocytes (PMNL) and subsequent degranulation is associated with full activation of surrounding platelets, we studied the effect of HNPs on platelet function. METHODS The effect of HNPs on platelet activation parameters and apoptosis was investigated via aggregometry, flow cytometry, confocal microscopy and the ELISA technique. RESULTS It was found that HNPs activate platelets in pathophysiologically relevant doses, inducing fibrinogen and thrombospondin-1 binding, aggregation, granule secretion, sCD40L shedding, and procoagulant activity. HNPs bound directly to the platelet membrane, induced membrane pore formation, microparticle formation, mitochondrial membrane depolarization and caspase-3-activity. Confocal microscopy revealed the HNP-induced formation of polymeric fibrinogen and thrombospondin-1 amyloid-like structures, which bound microorganisms. Platelets adhered to these structures and formed aggregates. Blocking of glycoprotein IIb/IIIa (GPIIb/IIIa) markedly inhibited HNP-induced platelet activation. In addition, heparin, heparinoid, serpins and α(2)-macroglobulin, which all bind to HNPs, blocked HNP-1-induced platelet activation in contrast to direct thrombin inhibitors such as hirudin. CONCLUSIONS HNPs activate platelets and induce platelet apoptosis by formation of amyloid-like proteins. As these structures entrapped bacteria and fungi, they might reflect an additional function of HNPs in host defense. The described mechanism links again thrombosis and infection.
Collapse
Affiliation(s)
- M Horn
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University of Muenster, Muenster, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Asmat TM, Klingbeil K, Jensch I, Burchhardt G, Hammerschmidt S. Heterologous expression of pneumococcal virulence factor PspC on the surface of Lactococcus lactis confers adhesive properties. MICROBIOLOGY-SGM 2012; 158:771-780. [PMID: 22222496 DOI: 10.1099/mic.0.053603-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lactococcus lactis is a non-pathogenic bacterium that is used in the food industry but is also used as a heterologous host to reveal protein functions of pathogenic bacteria. The adhesin PspC from Streptococcus pneumoniae is a choline-binding protein that is non-covalently anchored to the bacterial cell wall. To assess the exclusive impact of pneumococcal surface protein C (PspC) on the interplay with its host we generated recombinant L. lactis producing a nisin-inducible and covalently anchored variant of PspC on the lactococcal cell surface. A translational fusion of the 5'-end of pspC3.4 with the 3'-end of hic (pspC11.4) was designed to decorate the surface of L. lactis with a chimeric PspC. The PspC3.4 part comprises the first 281 aa residues of PspC3.4, while the Hic sequence consists of the proline-rich and sortase-anchored domain. The results demonstrated that PspC is sufficient for adhesion and subsequent invasion of host epithelial cells expressing the human polymeric Ig receptor (hpIgR). Moreover, invasion via hpIgR was even more pronounced when the chimeric PspC was produced by lactococci compared with pneumococci. This study shows also for the first time that PspC plays no significant role during phagocytosis by macrophages. In contrast, recruitment of Factor H via the PspC chimer has a dramatic effect on phagocytosis of recombinant but not wild-type lactococci, as Factor H interacts specifically with the amino-terminal part of PspC and mediates the contact with phagocytes. Furthermore, L. lactis expressing PspC increased intracellular calcium levels in pIgR-expressing epithelial cells, thus resembling the effect of pneumococci, which induced release of Ca(2+) from intracellular stores via the PspC-pIgR mechanism. In conclusion, expression of the chimeric PspC confers adhesive properties to L. lactis and indicates the potential of L. lactis as a suitable host to study the impact of individual bacterial factors on their capacity to interfere with the host and manipulate eukaryotic epithelial cells.
Collapse
Affiliation(s)
- Tauseef M Asmat
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany
| | - Katharina Klingbeil
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany
| | - Inga Jensch
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany
| | - Gerhard Burchhardt
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany
| |
Collapse
|
50
|
Härtel T, Eylert E, Schulz C, Petruschka L, Gierok P, Grubmüller S, Lalk M, Eisenreich W, Hammerschmidt S. Characterization of central carbon metabolism of Streptococcus pneumoniae by isotopologue profiling. J Biol Chem 2011; 287:4260-74. [PMID: 22167202 DOI: 10.1074/jbc.m111.304311] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The metabolism of Streptococcus pneumoniae was studied by isotopologue profiling after bacterial cultivation in chemically defined medium supplemented with [U-(13)C(6)]- or [1,2-(13)C(2)]glucose. GC/MS analysis of protein-derived amino acids showed lack of (13)C label in amino acids that were also essential for pneumococcal growth. Ala, Ser, Asp, and Thr displayed high (13)C enrichments, whereas Phe, Tyr, and Gly were only slightly labeled. The analysis of the labeling patterns showed formation of triose phosphate and pyruvate via the Embden-Meyerhof-Parnas pathway. The labeling patterns of Asp and Thr suggested formation of oxaloacetate exclusively via the phosphoenolpyruvate carboxylase reaction. Apparently, α-ketoglutarate was generated from unlabeled glutamate via the aspartate transaminase reaction. A fraction of Phe and Tyr obtained label via the chorismate route from erythrose 4-phosphate, generated via the pentose phosphate pathway, and phosphoenolpyruvate. Strikingly, the data revealed no significant flux from phosphoglycerate to Ser and Gly but showed formation of Ser via the reverse reaction, namely by hydroxymethylation of Gly. The essential Gly was acquired from the medium, and the biosynthesis pathway was confirmed in experiments using [U-(13)C(2)]glycine as a tracer. The hydroxymethyl group in Ser originated from formate, which was generated by the pyruvate formate-lyase. Highly similar isotopologue profiles were observed in corresponding experiments with pneumococcal mutants deficient in PavA, CodY, and glucose-6-phosphate dehydrogenase pointing to the robustness of the core metabolic network used by these facultative pathogenic bacteria. In conclusion, this study demonstrates the dual utilization of carbohydrates and amino acids under in vitro conditions and identifies the unconventional de novo biosynthesis of serine by pneumococci.
Collapse
Affiliation(s)
- Tobias Härtel
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Strasse 15a, D-17487 Greifswald, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|