1
|
Rühling M, Schmelz F, Kempf A, Paprotka K, Fraunholz MJ. Identification of the Staphylococcus aureus endothelial cell surface interactome by proximity labeling. mBio 2025:e0365424. [PMID: 40162795 DOI: 10.1128/mbio.03654-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Virulence strategies of pathogens depend on interaction with host cells. The binding and activation of receptors located on the plasma membrane are crucial for the attachment to or pathogen internalization by host cells. Identifying host cell receptors is often difficult, and hence, the identity of many proteins that play important roles during host-pathogen interaction remains elusive. We developed a novel proximity labeling approach by decorating the opportunistic pathogen Staphylococcus aureus with ascorbate peroxidase 2. Upon addition of hydrogen peroxide, the peroxidase initiates proximity biotinylation of S. aureus host interaction partners, thereby enabling the identification of these proteins. Here, we demonstrate an endothelial cell surface interactome of 305 proteins, including novel S. aureus co-receptors such as neuronal adhesion molecule, protein tyrosine kinase PTK7, melanotransferrin, protein-tyrosine kinase MET, and CD109. Filtering the interactome for validated surface proteins resulted in a list of 89 protein candidates, 53% of which were described to interact with S. aureus or other pathogens. IMPORTANCE Staphylococcus aureus is an opportunistic pathogen that enters host cells such as epithelial or endothelial cells. Intracellular pathogens have been observed in vivo and are thought to serve immune evasion, avoidance of antibiotic treatment, and chronicity of infection. Thus, it is important to understand the mechanisms by which the bacteria are internalized by host cells; however, screening for pathogen-host receptors is difficult. Here, we developed a novel proximity labeling approach, which enabled the identification of several previously unknown host receptors of S. aureus that are engaged during a rapid uptake pathway for the bacteria.
Collapse
Affiliation(s)
- Marcel Rühling
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Fabio Schmelz
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Alicia Kempf
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Kerstin Paprotka
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Martin J Fraunholz
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
2
|
Speziale P, Foster TJ, Arciola CR. The endothelium at the interface between tissues and Staphylococcus aureus in the bloodstream. Clin Microbiol Rev 2025; 38:e0009824. [PMID: 39807893 PMCID: PMC11905367 DOI: 10.1128/cmr.00098-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
SUMMARYStaphylococcus aureus is a major human pathogen. It can cause many types of infections, in particular bacteremia, which frequently leads to infective endocarditis, osteomyelitis, sepsis, and other debilitating diseases. The development of secondary infections is based on the bacterium's ability to associate with endothelial cells lining blood vessels. The success of endothelial colonization and infection by S. aureus relies on its ability to express a wide array of cell wall-anchored and secreted virulence factors. Establishment of endothelial infection by the pathogen is a multistep process involving adhesion, invasion, extravasation, and dissemination of the bacterium into surrounding tissues. The process is dependent on the type of endothelium in different organs (tissues) and pathogenetic potential of the individual strains. In this review, we report an update on the organization of the endothelium in the vessels, the structure and function of the virulence factors of S. aureus, and the several aspects of bacteria-endothelial cell interactions. After these sections, we will discuss recent advances in understanding the specific mechanisms of infections that develop in the heart, bone and joints, lung, and brain. Finally, we describe how neutrophils bind to endothelial cells, migrate to the site of infection to kill bacteria in the tissues, and how staphylococci counteract neutrophils' actions. Knowledge of the molecular details of S. aureus-endothelial cell interactions will promote the development of new therapeutic strategies and tools to combat this formidable pathogen.
Collapse
Affiliation(s)
- Pietro Speziale
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Pavia, Italy
| | - Timothy J Foster
- Department of Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Carla Renata Arciola
- Laboratory of Pathology of Implant Infections, Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
3
|
Smith OER, Bharat TAM. Architectural dissection of adhesive bacterial cell surface appendages from a "molecular machines" viewpoint. J Bacteriol 2024; 206:e0029024. [PMID: 39499080 PMCID: PMC7616799 DOI: 10.1128/jb.00290-24] [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] [Indexed: 11/07/2024] Open
Abstract
The ability of bacteria to interact with and respond to their environment is crucial to their lifestyle and survival. Bacterial cells routinely need to engage with extracellular target molecules, in locations spatially separated from their cell surface. Engagement with distant targets allows bacteria to adhere to abiotic surfaces and host cells, sense harmful or friendly molecules in their vicinity, as well as establish symbiotic interactions with neighboring cells in multicellular communities such as biofilms. Binding to extracellular molecules also facilitates transmission of information back to the originating cell, allowing the cell to respond appropriately to external stimuli, which is critical throughout the bacterial life cycle. This requirement of bacteria to bind to spatially separated targets is fulfilled by a myriad of specialized cell surface molecules, which often have an extended, filamentous arrangement. In this review, we compare and contrast such molecules from diverse bacteria, which fulfil a range of binding functions critical for the cell. Our comparison shows that even though these extended molecules have vastly different sequence, biochemical and functional characteristics, they share common architectural principles that underpin bacterial adhesion in a variety of contexts. In this light, we can consider different bacterial adhesins under one umbrella, specifically from the point of view of a modular molecular machine, with each part fulfilling a distinct architectural role. Such a treatise provides an opportunity to discover fundamental molecular principles governing surface sensing, bacterial adhesion, and biofilm formation.
Collapse
Affiliation(s)
- Olivia E. R. Smith
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Tanmay A. M. Bharat
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
4
|
Tricou LP, Mouton W, Cara A, Trouillet-Assant S, Bouvard D, Laurent F, Diot A, Josse J. Staphylococcus aureus can use an alternative pathway to be internalized by osteoblasts in absence of β1 integrins. Sci Rep 2024; 14:28643. [PMID: 39562631 PMCID: PMC11576967 DOI: 10.1038/s41598-024-78754-z] [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: 09/12/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Staphylococcus aureus main internalization mechanism in osteoblasts relies on a tripartite interaction between bacterial fibronectin-binding proteins, extracellular matrix soluble fibronectin, and osteoblasts' β1 integrins. Caveolins, and particularly caveolin-1, have been shown to limit the plasma membrane microdomain mobility, and consequently reduce the uptake of S. aureus in keratinocytes. In this study, we aimed to deepen our understanding of the molecular mechanisms underlying S. aureus internalization in osteoblasts. Mechanistically, S. aureus internalization requires endosomal recycling of β1 integrins as well as downstream effectors such as Src, Rac1, and PAK1. Surprisingly, in β1 integrin deficient osteoblasts, S. aureus internalization is restored when Caveolin-1 is absent and requires αvβ3/5 integrins as backup fibronectin receptors. Altogether, our data support that β1 integrins regulate the level of detergent-resistant membrane at the plasma membrane in a an endosomal and Caveolin-1 dependent manner.
Collapse
Affiliation(s)
- Léo-Paul Tricou
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- ISPB Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
- Department of Orthopaedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - William Mouton
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Andréa Cara
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Sophie Trouillet-Assant
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- Joint Research Unit Civils Hospices of Lyon-bioMérieux, Hospices Civils de Lyon, Lyon Sud Hospital, Pierre-Bénite, France
| | - Daniel Bouvard
- Centre de Recherche en Biologie Cellulaire de Montpellier (CRBM), CNRS UMR 5237, Université de Montpellier, Montpellier, France
| | - Frédéric Laurent
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
- ISPB Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
- Centre Interrégional de Référence pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Alan Diot
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Josse
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, CNRS, UMR5308, ENS de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, Lyon, France.
- ISPB Faculté de Pharmacie, Université Claude Bernard Lyon 1, Lyon, France.
- Centre Interrégional de Référence pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
5
|
Ke S, Kil H, Roggy C, Shields T, Quinn Z, Quinn AP, Small JM, Towne FD, Brooks AE, Brooks BD. Potential Therapeutic Targets for Combination Antibody Therapy Against Staphylococcus aureus Infections. Antibiotics (Basel) 2024; 13:1046. [PMID: 39596740 PMCID: PMC11591076 DOI: 10.3390/antibiotics13111046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Despite the significant advances in antibiotic treatments and therapeutics, Staphylococcus aureus (S. aureus) remains a formidable pathogen, primarily due to its rapid acquisition of antibiotic resistance. Known for its array of virulence factors, including surface proteins that promote adhesion to host tissues, enzymes that break down host barriers, and toxins that contribute to immune evasion and tissue destruction, S. aureus poses a serious health threat. Both the Centers for Disease Control and Prevention (CDC) and the World Health Organization (WHO) classify S. aureus as an ESKAPE pathogen, recognizing it as a critical threat to global health. The increasing prevalence of drug-resistant S. aureus underscores the need for new therapeutic strategies. This review discusses a promising approach that combines monoclonal antibodies targeting multiple S. aureus epitopes, offering synergistic efficacy in treating infections. Such strategies aim to reduce the capacity of the pathogen to develop resistance, presenting a potent adjunct or alternative to conventional antibiotic treatments.
Collapse
Affiliation(s)
- Sharon Ke
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Hyein Kil
- Department of Surgery, Virtua Health, Camden, NJ 08103, USA
| | - Conner Roggy
- Department of Orthopaedic Surgery, Community Memorial Healthcare, Ventura, CA 93003, USA
| | - Ty Shields
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Zachary Quinn
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Alyssa P. Quinn
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - James M. Small
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Francina D. Towne
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Amanda E. Brooks
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Benjamin D. Brooks
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| |
Collapse
|
6
|
Ma X, Ma J, Liu J, Hao H, Hou H, Zhang G. Inhibitory Effect of Phenethyl Isothiocyanate on the Adhesion and Biofilm Formation of Staphylococcus aureus and Application on Beef. Foods 2024; 13:3362. [PMID: 39517145 PMCID: PMC11544944 DOI: 10.3390/foods13213362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
This study aimed to explore the mechanism by which phenethyl isothiocyanate (PEITC) inhibited the adhesion and biofilm formation of Staphylococcus aureus (S. aureus). PEITC exhibited antimicrobial efficacy against S. aureus, demonstrating a minimum inhibition concentration (MIC) of 1 mmol/L. PEITC exerted its antibacterial effect by disrupting cell membrane integrity, and it decreased total adenosine triphosphate (ATP) production after 1 and 4 h treatment. PEITC at 0.5 mmol/L increased the level of intracellular reactive oxygen species (ROS) by 26.39% compared to control. The mature biofilm of S. aureus was destroyed by 86.4% after treatment with PEITC for 24 h. Adhesion tests revealed that PEITC at 0.5 mmol/L reduced 44.51% of the S. aureus that adhered to NCM460 cells. Furthermore, at the genetic level, PEITC significantly downregulated the related genes by 31.26% to 97.04%, including agrB, agrD, isdA, ebh, luxS, fnbA, and icaR. Moreover, PEITC markedly inhibited S. aureus proliferation in beef preserved at temperatures of 25 and 4 °C, respectively. In summary, the present study suggests that PEITC effectively inhibits the adhesion and biofilm formation of S. aureus by affecting the relevant genes of S. aureus and holds promise for microbial management in meat products.
Collapse
Affiliation(s)
- Xiaojing Ma
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.M.)
| | - Jinle Ma
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.M.)
| | - Jianan Liu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.M.)
| | - Hongshun Hao
- Department of Inorganic Nonmetallic Material Engineering, Dalian Polytechnic University, Dalian 116034, China
| | - Hongman Hou
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.M.)
- Liaoning Key Lab for Aquatic Processing Quality and Safety, Dalian 116034, China
| | - Gongliang Zhang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; (X.M.)
- Liaoning Key Lab for Aquatic Processing Quality and Safety, Dalian 116034, China
| |
Collapse
|
7
|
Goldmann O, Lang JC, Rohde M, May T, Molinari G, Medina E. Alpha-hemolysin promotes internalization of Staphylococcus aureus into human lung epithelial cells via caveolin-1- and cholesterol-rich lipid rafts. Cell Mol Life Sci 2024; 81:435. [PMID: 39412594 PMCID: PMC11488825 DOI: 10.1007/s00018-024-05472-0] [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: 02/01/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024]
Abstract
Staphylococcus aureus is a pathogen associated with severe respiratory infections. The ability of S. aureus to internalize into lung epithelial cells complicates the treatment of respiratory infections caused by this bacterium. In the intracellular environment, S. aureus can avoid elimination by the immune system and the action of circulating antibiotics. Consequently, interfering with S. aureus internalization may represent a promising adjunctive therapeutic strategy to enhance the efficacy of conventional treatments. Here, we investigated the host-pathogen molecular interactions involved in S. aureus internalization into human lung epithelial cells. Lipid raft-mediated endocytosis was identified as the main entry mechanism. Thus, bacterial internalization was significantly reduced after the disruption of lipid rafts with methyl-β-cyclodextrin. Confocal microscopy confirmed the colocalization of S. aureus with lipid raft markers such as ganglioside GM1 and caveolin-1. Adhesion of S. aureus to α5β1 integrin on lung epithelial cells via fibronectin-binding proteins (FnBPs) was a prerequisite for bacterial internalization. A mutant S. aureus strain deficient in the expression of alpha-hemolysin (Hla) was significantly impaired in its capacity to enter lung epithelial cells despite retaining its capacity to adhere. This suggests a direct involvement of Hla in the bacterial internalization process. Among the receptors for Hla located in lipid rafts, caveolin-1 was essential for S. aureus internalization, whereas ADAM10 was dispensable for this process. In conclusion, this study supports a significant role of lipid rafts in S. aureus internalization into human lung epithelial cells and highlights the interaction between bacterial Hla and host caveolin-1 as crucial for the internalization process.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Julia C Lang
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, 171 77, Sweden
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Tobias May
- InSCREENeX GmbH, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Gabriella Molinari
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany.
| |
Collapse
|
8
|
Guo M, Li Y, Tang J, Wang Q, Wang Q, Zhou H, Lin H, Ma Z, Fan H. Glaesserella parasuis serotype 4 exploits fibronectin via RlpA for tracheal colonization following porcine circovirus type 2 infection. PLoS Pathog 2024; 20:e1012513. [PMID: 39264911 PMCID: PMC11392263 DOI: 10.1371/journal.ppat.1012513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
Porcine circovirus type 2 (PCV2) often causes disease through coinfection with other bacterial pathogens, including Glaesserella parasuis (G. parasuis), which causes high morbidity and mortality, but the role played by PCV2 and bacterial and host factors contributing to this process have not been defined. Bacterial attachment is assumed to occur via specific receptor-ligand interactions between adhesins on the bacterial cell and host proteins adsorbed to the implant surface. Mass spectrometry (MS) analysis of PCV2-infected swine tracheal epithelial cells (STEC) revealed that the expression of Extracellular matrix protein (ECM) Fibronectin (Fn) increased significantly on the infected cells surface. Importantly, efficient G. parasuis serotype 4 (GPS4) adherence to STECs was imparted by interactions with Fn. Furthermore, abrogation of adherence was gained by genetic knockout of Fn, Fn and Integrin β1 antibody blocking. Fn is frequently exploited as a receptor for bacterial pathogens. To explore the GPS4 adhesin that interacts with Fn, recombinant Fn N-terminal type I and type II domains were incubated with GPS4, and the interacting proteins were pulled down for MS analysis. Here, we show that rare lipoprotein A (RlpA) directly interacts with host Fibronectin mediating GPS4 adhesion. Finally, we found that PCV2-induced Fibronectin expression and adherence of GPS4 were prevented significantly by TGF-β signaling pathway inhibitor SB431542. Our data suggest the RlpA-Fn interaction to be a potentially promising novel therapeutic target to combat PCV2 and GPS4 coinfection.
Collapse
Affiliation(s)
- Mengru Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuhui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jinsheng Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qing Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Qiancheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huixing Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhe Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- College of Animal Science, Anhui Science and Technology University, Fengyang, China
| |
Collapse
|
9
|
Moulick S, Roy DN. Bioflavonoid Baicalein Modulates Tetracycline Resistance by Inhibiting Efflux Pump in Staphylococcus aureus. Microb Drug Resist 2024; 30:363-371. [PMID: 39133125 DOI: 10.1089/mdr.2024.0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024] Open
Abstract
The rise in antibiotic resistance among bacterial pathogens, particularly Staphylococcus aureus, has become a critical global health issue, necessitating the search for novel antimicrobial agents. S. aureus uses various mechanisms to resist antibiotics, including the activation of efflux pumps, biofilm formation, and enzymatic modification of drugs. This study explores the potential of baicalein, a bioflavonoid from Scutellaria baicalensis, in modulating tetracycline resistance in S. aureus by inhibiting efflux pumps. The synergistic action of baicalein and tetracycline was evaluated through various assays. The minimum inhibitory concentration (MIC) of baicalein and tetracycline against S. aureus was 256 and 1.0 μg/mL, respectively. Baicalein at 64 μg/mL reduced the MIC of tetracycline by eightfold, indicating a synergistic effect (fractional inhibitory concentration index: 0.375). Time-kill kinetics demonstrated a 1.0 log CFU/mL reduction in bacterial count after 24 hours with the combination treatment. The ethidium bromide accumulation assay showed that baicalein mediated significant inhibition of efflux pumps, with a dose-dependent increase in fluorescence. In addition, baicalein inhibited DNA synthesis by 73% alone and 92% in combination with tetracycline. It also markedly reduced biofilm formation and the invasiveness of S. aureus into HeLa cells by 52% at 64 μg/mL. These findings suggest that baicalein enhances tetracycline efficacy and could be a promising adjunct therapy to combat multidrug-resistant S. aureus infections.
Collapse
Affiliation(s)
- Soumitra Moulick
- TCG Lifesciences Private Limited, Kolkata, India
- Department of Biotechnology, National Institute of Technology Raipur, Chhattisgarh, India
| | - Dijendra Nath Roy
- Department of Biotechnology, National Institute of Technology Raipur, Chhattisgarh, India
| |
Collapse
|
10
|
Rühling M, Kersting L, Wagner F, Schumacher F, Wigger D, Helmerich DA, Pfeuffer T, Elflein R, Kappe C, Sauer M, Arenz C, Kleuser B, Rudel T, Fraunholz M, Seibel J. Trifunctional sphingomyelin derivatives enable nanoscale resolution of sphingomyelin turnover in physiological and infection processes via expansion microscopy. Nat Commun 2024; 15:7456. [PMID: 39198435 PMCID: PMC11358447 DOI: 10.1038/s41467-024-51874-w] [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: 03/12/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Sphingomyelin is a key molecule of sphingolipid metabolism, and its enzymatic breakdown is associated with various infectious diseases. Here, we introduce trifunctional sphingomyelin derivatives that enable the visualization of sphingomyelin distribution and sphingomyelinase activity in infection processes. We demonstrate this by determining the activity of a bacterial sphingomyelinase on the plasma membrane of host cells using a combination of Förster resonance energy transfer and expansion microscopy. We further use our trifunctional sphingomyelin probes to visualize their metabolic state during infections with Chlamydia trachomatis and thereby show that chlamydial inclusions primarily contain the cleaved forms of the molecules. Using expansion microscopy, we observe that the proportion of metabolized molecules increases during maturation from reticulate to elementary bodies, indicating different membrane compositions between the two chlamydial developmental forms. Expansion microscopy of trifunctional sphingomyelins thus provides a powerful microscopy tool to analyze sphingomyelin metabolism in cells at nanoscale resolution.
Collapse
Affiliation(s)
- Marcel Rühling
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Louise Kersting
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Fabienne Wagner
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | | | - Dominik Wigger
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Dominic A Helmerich
- Chair of Biotechnology & Biophysics, Biocenter, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Tom Pfeuffer
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Robin Elflein
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Christian Kappe
- Institute of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str 2, Berlin, Germany
| | - Markus Sauer
- Chair of Biotechnology & Biophysics, Biocenter, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Christoph Arenz
- Institute of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str 2, Berlin, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Thomas Rudel
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Martin Fraunholz
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany.
| |
Collapse
|
11
|
Yu D, Lu Z, Chong Y. Integrins as a bridge between bacteria and cells: key targets for therapeutic wound healing. BURNS & TRAUMA 2024; 12:tkae022. [PMID: 39015251 PMCID: PMC11250365 DOI: 10.1093/burnst/tkae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 12/17/2023] [Accepted: 04/22/2024] [Indexed: 07/18/2024]
Abstract
Integrins are heterodimers composed of α and β subunits that are bonded through non-covalent interactions. Integrins mediate the dynamic connection between extracellular adhesion molecules and the intracellular actin cytoskeleton. Integrins are present in various tissues and organs where these heterodimers participate in diverse physiological and pathological responses at the molecular level in living organisms. Wound healing is a crucial process in the recovery from traumatic diseases and comprises three overlapping phases: inflammation, proliferation and remodeling. Integrins are regulated during the entire wound healing process to enhance processes such as inflammation, angiogenesis and re-epithelialization. Prolonged inflammation may result in failure of wound healing, leading to conditions such as chronic wounds. Bacterial colonization of a wound is one of the primary causes of chronic wounds. Integrins facilitate the infectious effects of bacteria on the host organism, leading to chronic inflammation, bacterial colonization, and ultimately, the failure of wound healing. The present study investigated the role of integrins as bridges for bacteria-cell interactions during wound healing, evaluated the role of integrins as nodes for bacterial inhibition during chronic wound formation, and discussed the challenges and prospects of using integrins as therapeutic targets in wound healing.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| |
Collapse
|
12
|
Ji X, Zhu W, Lu H, Wu Z, Chen H, Lin C, Zeng Z, You C, Li L. Antibiotic Resistance Profiles and MLST Typing of Staphylococcus Aureus Clone Associated with Skin and Soft Tissue Infections in a Hospital of China. Infect Drug Resist 2024; 17:2555-2566. [PMID: 38933775 PMCID: PMC11199319 DOI: 10.2147/idr.s465951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Objective To analyze the antibiotic resistance profile, virulence genes, and molecular typing of Staphylococcus aureus (S. aureus) strains isolated in skin and soft tissue infections at the First Affiliated Hospital, Gannan Medical University, to better understand the molecular epidemiological characteristics of S. aureus. Methods In 2023, 65 S. aureus strains were isolated from patients with skin and soft tissue infections. Strain identification and susceptibility tests were performed using VITEK 2 and gram-positive bacteria identification cards. DNA was extracted using a DNA extraction kit, and all genes were amplified using polymerase chain reaction. Multilocus sequence typing (MLST) was used for molecular typing. Results In this study, of the 65 S. aureus strains were tested for their susceptibility to 16 antibiotics, the highest resistance rate to penicillin G was 95.4%. None of the staphylococcal isolates showed resistance to ceftaroline, daptomycin, linezolid, tigecycline, teicoplanin, or vancomycin. fnbA was the most prevalent virulence gene (100%) in S. aureus strains isolated in skin and soft tissue infections, followed by arcA (98.5%). Statistical analyses showed that the resistance rates of methicillin-resistant S. aureus isolates to various antibiotics were significantly higher than those of methicillin-susceptible S. aureus isolates. Fifty sequence types (STs), including 44 new ones, were identified by MLST. Conclusion In this study, the high resistance rate to penicillin G and the high carrying rate of virulence gene fnbA and arcA of S.aureus were determine, and 44 new STs were identified, which may be associated with the geographical location of southern Jiangxi and local trends in antibiotic use. The study of the clonal lineage and evolutionary relationships of S. aureus in these regions may help in understanding the molecular epidemiology and provide the experimental basis for pathogenic bacteria prevention and treatment.
Collapse
Affiliation(s)
- Xiaojuan Ji
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| | - Wenjie Zhu
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| | - Hongfei Lu
- Clinical Laboratory, First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Zhiwei Wu
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| | - Huaqing Chen
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| | - Chunli Lin
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| | - Zhaolin Zeng
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| | - Cong You
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| | - Longnian Li
- Department of Dermatology, First Affiliated Hospital of Gannan Medical University, Joint Organization of Jiangxi Clinical Medicine Research Center for Dermatology, Ganzhou, People’s Republic of China
| |
Collapse
|
13
|
Iibushi J, Nozawa T, Toh H, Nakagawa I. ATG9B regulates bacterial internalization via actin rearrangement. iScience 2024; 27:109623. [PMID: 38706859 PMCID: PMC11066431 DOI: 10.1016/j.isci.2024.109623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 05/07/2024] Open
Abstract
Invasive bacterial pathogens are internalized by host cells through endocytosis, which is regulated by a cascade of actin rearrangement signals triggered by host cell receptors or bacterial proteins delivered into host cells. However, the molecular mechanisms that mediate actin rearrangement to promote bacterial invasion are not fully understood. Here, we show that the autophagy-related (ATG) protein ATG9B regulates the internalization of various bacteria by controlling actin rearrangement. ATG knockout screening and knockdown experiments in HeLa cells identified ATG9B as a critical factor for bacterial internalization. In particular, cells with ATG9B knockdown exhibited an accumulation of actin filaments and phosphorylated LIM kinase and cofilin, suggesting that ATG9B is involved in actin depolymerization. Furthermore, the kinase activity of Unc-51-like autophagy-activating kinase 1 was found to regulate ATG9B localization and actin remodeling. These findings revealed a newly discovered function of ATG proteins in bacterial infection rather than autophagy-mediated immunity.
Collapse
Affiliation(s)
- Junpei Iibushi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Hirotaka Toh
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku 606-8501, Kyoto, Japan
| |
Collapse
|
14
|
Shi Y, Muenzner P, Schanz-Jurinka S, Hauck CR. The phosphatidylinositol-5' phosphatase synaptojanin1 limits integrin-mediated invasion of Staphylococcus aureus. Microbiol Spectr 2024; 12:e0200623. [PMID: 38358281 PMCID: PMC10986543 DOI: 10.1128/spectrum.02006-23] [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: 05/12/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
The gram-positive bacterium Staphylococcus aureus can invade non-professional phagocytic cells by associating with the plasma protein fibronectin to exploit host cell integrins. Integrin-mediated internalization of these pathogens is facilitated by the local production of phosphatidylinositol-4,5-bisphosphate (PI-4,5-P2) via an integrin-associated isoform of phosphatidylinositol-5' kinase. In this study, we addressed the role of PI-4,5-P2-directed phosphatases on internalization of S. aureus. ShRNA-mediated knockdown of individual phosphoinositide 5-phosphatases revealed that synaptojanin1 (SYNJ1) is counteracting invasion of S. aureus into mammalian cells. Indeed, shRNA-mediated depletion as well as genetic deletion of synaptojanin1 via CRISPR/Cas9 resulted in a gain-of-function phenotype with regard to integrin-mediated uptake. Surprisingly, the surface level of integrins was slightly downregulated in Synj1-KO cells. Nevertheless, these cells showed enhanced local accumulation of PI-4,5-P2 and exhibited increased internalization of S. aureus. While the phosphorylation level of the integrin-associated protein tyrosine kinase FAK was unaltered, the integrin-binding and -activating protein talin was enriched in the vicinity of S. aureus in synaptojanin1 knockout cells. Scanning electron microscopy revealed enlarged membrane invaginations in the absence of synaptojanin1 explaining the increased capability of these cells to internalize integrin-bound microorganisms. Importantly, the enhanced uptake by Synj1-KO cells and the exaggerated morphological features were rescued by the re-expression of the wild-type enzyme but not phosphatase inactive mutants. Accordingly, synaptojanin1 activity limits integrin-mediated invasion of S. aureus, corroborating the important role of PI-4,5-P2 during this process.IMPORTANCEStaphylococcus aureus, an important bacterial pathogen, can invade non-professional phagocytes by capturing host fibronectin and engaging integrin α5β1. Understanding how S. aureus exploits this cell adhesion receptor for efficient cell entry can also shed light on the physiological regulation of integrins by endocytosis. Previous studies have found that a specific membrane lipid, phosphatidylinositol-4,5-bisphosphate (PIP2), supports the internalization process. Here, we extend these findings and report that the local levels of PIP2 are controlled by the activity of the PIP2-directed lipid phosphatase Synaptojanin1. By dephosphorylating PIP2 at bacteria-host cell attachment sites, Synaptojanin1 counteracts the integrin-mediated uptake of the microorganisms. Therefore, our study not only generates new insight into subversion of cellular receptors by pathogenic bacteria but also highlights the role of host cell proteins acting as restriction factors for bacterial invasion at the plasma membrane.
Collapse
Affiliation(s)
- Yong Shi
- Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Petra Muenzner
- Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany
| | | | - Christof R. Hauck
- Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| |
Collapse
|
15
|
Mohammad S, Karim MR, Iqbal S, Lee JH, Mathiyalagan R, Kim YJ, Yang DU, Yang DC. Atopic dermatitis: Pathophysiology, microbiota, and metabolome - A comprehensive review. Microbiol Res 2024; 281:127595. [PMID: 38218095 DOI: 10.1016/j.micres.2023.127595] [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: 07/17/2023] [Revised: 10/11/2023] [Accepted: 12/24/2023] [Indexed: 01/15/2024]
Abstract
Atopic dermatitis (AD) is a prevalent inflammatory skin condition that commonly occurs in children. Genetics, environment, and defects in the skin barrier are only a few of the factors that influence how the disease develops. As human microbiota research has advanced, more scientific evidence has shown the critical involvement of the gut and skin bacteria in the pathogenesis of atopic dermatitis. Microbiome dysbiosis, defined by changed diversity and composition, as well as the development of pathobionts, has been identified as a potential cause for recurring episodes of atopic dermatitis. Gut dysbiosis causes "leaky gut syndrome" by disrupting the epithelial lining of the gut, which allows bacteria and other endotoxins to enter the bloodstream and cause inflammation. The same is true for the disruption of cutaneous homeostasis caused by skin dysbiosis, which enables bacteria and other pathogens to reach deeper skin layers or even systemic circulation, resulting in inflammation. Furthermore, it is now recognized that the gut and skin microbiota releases both beneficial and toxic metabolites. Here, this review covers a range of topics related to AD, including its pathophysiology, the microbiota-AD connection, commonly used treatments, and the significance of metabolomics in AD prevention, treatment, and management, recognizing its potential in providing valuable insights into the disease.
Collapse
Affiliation(s)
- Shahnawaz Mohammad
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Md Rezaul Karim
- Department of Biopharmaceutical Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Safia Iqbal
- Department of Biopharmaceutical Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Microbiology, Varendra Institute of Biosciences, Affiliated by Rajshahi University, Natore, Rajshahi 6400, Bangladesh
| | - Jung Hyeok Lee
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Yeon Ju Kim
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Dong Uk Yang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| | - Deok Chun Yang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea; Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.
| |
Collapse
|
16
|
van den Biggelaar RHGA, Walburg KV, van den Eeden SJF, van Doorn CLR, Meiler E, de Ries AS, Meijer AH, Ottenhoff THM, Saris A. Identification of kinase modulators as host-directed therapeutics against intracellular methicillin-resistant Staphylococcus aureus. Front Cell Infect Microbiol 2024; 14:1367938. [PMID: 38590439 PMCID: PMC10999543 DOI: 10.3389/fcimb.2024.1367938] [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: 01/09/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
The increasing prevalence of antimicrobial-resistant Staphylococcus aureus strains, especially methicillin-resistant S. aureus (MRSA), poses a threat to successful antibiotic treatment. Unsuccessful attempts to develop a vaccine and rising resistance to last-resort antibiotics urge the need for alternative treatments. Host-directed therapy (HDT) targeting critical intracellular stages of S. aureus emerges as a promising alternative, potentially acting synergistically with antibiotics and reducing the risk of de novo drug resistance. We assessed 201 ATP-competitive kinase inhibitors from Published Kinase Inhibitor Sets (PKIS1 and PKIS2) against intracellular MRSA. Seventeen hit compounds were identified, of which the two most effective and well-tolerated hit compounds (i.e., GW633459A and GW296115X) were selected for further analysis. The compounds did not affect planktonic bacterial cultures, while they were active in a range of human cell lines of cervical, skin, lung, breast and monocyte origin, confirming their host-directed mechanisms. GW633459A, structurally related to lapatinib, exhibited an HDT effect on intracellular MRSA independently of its known human epidermal growth factor receptor (EGFR)/(HER) kinase family targets. GW296115X activated adenosine monophosphate-activated protein kinase (AMPK), thereby enhancing bacterial degradation via autophagy. Finally, GW296115X not only reduced MRSA growth in human cells but also improved the survival rates of MRSA-infected zebrafish embryos, highlighting its potential as HDT.
Collapse
Affiliation(s)
- Robin H G A van den Biggelaar
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Kimberley V Walburg
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Susan J F van den Eeden
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Cassandra L R van Doorn
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Eugenia Meiler
- Global Health Medicines R&D, GlaxoSmithKline, Tres Cantos, Spain
| | - Alex S de Ries
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | | - Tom H M Ottenhoff
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Anno Saris
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
17
|
Yu D, Lu Z, Nie F, Chong Y. Integrins regulation of wound healing processes: insights for chronic skin wound therapeutics. Front Cell Infect Microbiol 2024; 14:1324441. [PMID: 38505290 PMCID: PMC10949986 DOI: 10.3389/fcimb.2024.1324441] [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: 10/19/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024] Open
Abstract
Integrins are heterodimers composed of non-covalently associated alpha and beta subunits that mediate the dynamic linkage between extracellular adhesion molecules and the intracellular actin cytoskeleton. Integrins are present in various tissues and organs and are involved in different physiological and pathological molecular responses in vivo. Wound healing is an important process in the recovery from traumatic diseases and consists of three overlapping phases: inflammation, proliferation, and remodeling. Integrin regulation acts throughout the wound healing process to promote wound healing. Prolonged inflammation may lead to failure of wound healing, such as wound chronicity. One of the main causes of chronic wound formation is bacterial colonization of the wound. In this review, we review the role of integrins in the regulation of wound healing processes such as angiogenesis and re-epithelialization, as well as the role of integrins in mediating bacterial infections during wound chronicity, and the challenges and prospects of integrins as therapeutic targets for infected wound healing.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fengsong Nie
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
18
|
Numata T, Iwamoto K, Matsunae K, Miyake R, Suehiro M, Yanagida N, Kan T, Takahagi S, Hide M, Tanaka A. A Staphylococcus epidermidis strain inhibits the uptake of Staphylococcus aureus derived from atopic dermatitis skin into the keratinocytes. J Dermatol Sci 2024; 113:113-120. [PMID: 38395669 DOI: 10.1016/j.jdermsci.2024.01.006] [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: 07/06/2023] [Revised: 11/20/2023] [Accepted: 01/24/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Various bacterial species form a microbiome in the skin. In the past, dead Staphylococcus aureus derived from atopic dermatitis (AD) are taken up by keratinocytes; however, whether live S. aureus can be taken up by keratinocytes is unknown. OBJECTIVE This study aimed to examine whether live AD strains of S. aureus internalize into the keratinocytes and how the internalization changes under conditions in which other bacterial species including S. epidermidis are present. METHODS HaCaT cells were cultured with live S. aureus and S. epidermidis (live or heat-treated) or their culture supernatants. After coculture, the change in the amount of S. aureus in the cytoplasm of HaCaT cells was analyzed using, a high-throughput imaging system, Opera Phenix™. RESULTS Live S. aureus were taken up in the cytoplasm of HaCaT cells. Coculturing live S. aureus with live S. epidermidis or the culture supernatants decreased the abundance of S. aureus in the cytoplasm. The heat-treated culture supernatants of live S. epidermidis or culture supernatants of other S. strains did not decrease the abundance of S. aureus in the cytoplasm. CONCLUSION Live S. aureus was internalized into the cytoplasm of HaCaT cells as does heat-treated S. aureus. In addition, the heat-sensitive substances secreted by coculture with S. epidermidis and keratinocytes inhibited the uptake of S. aureus by keratinocytes.
Collapse
Affiliation(s)
- Tomofumi Numata
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Kazumasa Iwamoto
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Kyouka Matsunae
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Ryu Miyake
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Masataka Suehiro
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Nozomi Yanagida
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Takanobu Kan
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Shunsuke Takahagi
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
19
|
Liu K, Tong J, Liu X, Liang D, Ren F, Jiang N, Hao Z, Li S, Wang Q. The Discovery of Novel Agents against Staphylococcus aureus by Targeting Sortase A: A Combination of Virtual Screening and Experimental Validation. Pharmaceuticals (Basel) 2023; 17:58. [PMID: 38256891 PMCID: PMC11100315 DOI: 10.3390/ph17010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 01/24/2024] Open
Abstract
Staphylococcus aureus (S. aureus), commonly known as "superbugs", is a highly pathogenic bacterium that poses a serious threat to human health. There is an urgent need to replace traditional antibiotics with novel drugs to combat S. aureus. Sortase A (SrtA) is a crucial transpeptidase involved in the adhesion process of S. aureus. The reduction in virulence and prevention of S. aureus infections have made it a significant target for antimicrobial drugs. In this study, we combined virtual screening with experimental validation to identify potential drug candidates from a drug library. Three hits, referred to as Naldemedine, Telmisartan, and Azilsartan, were identified based on docking binding energy and the ratio of occupied functional sites of SrtA. The stability analysis manifests that Naldemedine and Telmisartan have a higher binding affinity to the hydrophobic pockets. Specifically, Telmisartan forms stable hydrogen bonds with SrtA, resulting in the highest binding energy. Our experiments prove that the efficiency of adhesion and invasion by S. aureus can be decreased without significantly affecting bacterial growth. Our work identifies Telmisartan as the most promising candidate for inhibiting SrtA, which can help combat S. aureus infection.
Collapse
Affiliation(s)
- Kang Liu
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (K.L.); (J.T.); (D.L.); (F.R.); (N.J.); (Z.H.)
| | - Jiangbo Tong
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (K.L.); (J.T.); (D.L.); (F.R.); (N.J.); (Z.H.)
| | - Xu Liu
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, China;
| | - Dan Liang
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (K.L.); (J.T.); (D.L.); (F.R.); (N.J.); (Z.H.)
| | - Fangzhe Ren
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (K.L.); (J.T.); (D.L.); (F.R.); (N.J.); (Z.H.)
| | - Nan Jiang
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (K.L.); (J.T.); (D.L.); (F.R.); (N.J.); (Z.H.)
| | - Zhenyu Hao
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (K.L.); (J.T.); (D.L.); (F.R.); (N.J.); (Z.H.)
| | - Shixin Li
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China; (K.L.); (J.T.); (D.L.); (F.R.); (N.J.); (Z.H.)
| | - Qiang Wang
- Department of the Heart and Great Vessels, Affiliated Hospital of Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
20
|
Robertin S, Brokatzky D, Lobato-Márquez D, Mostowy S. Regulation of integrin α5β1-mediated Staphylococcus aureus cellular invasion by the septin cytoskeleton. Eur J Cell Biol 2023; 102:151359. [PMID: 37683588 DOI: 10.1016/j.ejcb.2023.151359] [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: 04/05/2023] [Revised: 08/10/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Staphylococcus aureus, a Gram-positive bacterial pathogen, is an urgent health threat causing a wide range of clinical infections. Originally viewed as a strict extracellular pathogen, accumulating evidence has revealed S. aureus to be a facultative intracellular pathogen subverting host cell signalling to support invasion. The majority of clinical isolates produce fibronectin-binding proteins A and B (FnBPA and FnBPB) to interact with host integrin α5β1, a key component of focal adhesions. S. aureus binding of integrin α5β1 promotes its clustering on the host cell surface, triggering activation of focal adhesion kinase (FAK) and cytoskeleton rearrangements to promote bacterial invasion into non-phagocytic cells. Here, we discover that septins, a component of the cytoskeleton that assembles on membranes, are recruited as collar-like structures with actin to S. aureus invasion sites engaging integrin α5β1. To investigate septin recruitment to the plasma membrane in a bacteria-free system, we used FnBPA-coated latex beads and showed that septins are recruited upon activation of integrin α5β1. SEPT2 depletion reduced S. aureus invasion, but increased surface expression of integrin α5 and adhesion of S. aureus to host cells. Consistent with this, SEPT2 depletion increased cellular protein levels of integrin α5 and β1 subunits, as well as FAK. Collectively, these results provide insights into regulation of integrin α5β1 and invasion of S. aureus by the septin cytoskeleton.
Collapse
Affiliation(s)
- Stevens Robertin
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Damián Lobato-Márquez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|
21
|
Zhu F, Ma S, Wen H, Rao M, Zhang P, Peng W, Cui Y, Yang H, Tan C, Chen J, Pan P. Development of a novel circular mRNA vaccine of six protein combinations against Staphylococcus aureus. J Biomol Struct Dyn 2023; 41:10525-10545. [PMID: 36533395 DOI: 10.1080/07391102.2022.2154846] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
Staphylococcus aureus is an extraordinarily versatile pathogen, which is currently the most common cause of nosocomial and community infections. Considering that increased antibiotic resistance may hasten the spread of S. aureus, developing an effective vaccine can possibly aid in its control. The RNA vaccine coding immunodominance epitopes from bacteria provide a potential method to induce T and B cell immune responses by translating them into cells. Furthermore, using bioinformatics to create circular RNA vaccines can ensure that the translation of the vaccine is potent and durable. In this study, 7 cytotoxic T lymphocyte (CTL) epitopes, 4 helper T lymphocyte (HTL) epitopes, and 15 B cell epitopes from 6 proteins that are closely associated with the S. aureus virulence and invasion and critical to natural immune responses were mapped. To verify their interactions, all epitopes were docked with the corresponding MHC alleles. The final vaccine was composed of 26 epitopes and the adjuvant β-defencin, and a disulfide bond was also introduced to improve its stability. After the prediction of structure and characteristics, the developed vaccine was docked with TLR2 and TLR4, which induce immunological responses in S. aureus infection. According to the molecular dynamic simulation, the vaccine might interact strongly with TLRs. Meanwhile, it performed well in immunological simulation and population coverage prediction. Finally, the vaccine was converted into a circular RNA using a series of helper sequences to aid in vaccine circulation translation. Hopefully, this proposed structure will be proven to serve a viable vaccine against S. aureus.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fei Zhu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Shiyang Ma
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Haicheng Wen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Mingjun Rao
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Peipei Zhang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Wenzhong Peng
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Yanhui Cui
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Hang Yang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Caixia Tan
- Department of Infection Control Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Chen
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| | - Pinhua Pan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, P.R. China
| |
Collapse
|
22
|
Green LR, Issa R, Albaldi F, Urwin L, Thompson R, Khalid H, Turner CE, Ciani B, Partridge LJ, Monk PN. CD9 co-operation with syndecan-1 is required for a major staphylococcal adhesion pathway. mBio 2023; 14:e0148223. [PMID: 37486132 PMCID: PMC10470606 DOI: 10.1128/mbio.01482-23] [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: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
Epithelial colonization is a critical first step in bacterial pathogenesis. Staphylococcus aureus can utilize several host factors to associate with cells, including α5β1 integrin and heparan sulfate proteoglycans, such as the syndecans. Here, we demonstrate that a partner protein of both integrins and syndecans, the host membrane adapter protein tetraspanin CD9, is essential for syndecan-mediated staphylococcal adhesion. Fibronectin is also essential in this process, while integrins are only critical for post-adhesion entry into human epithelial cells. Treatment of epithelial cells with CD9-derived peptide or heparin caused significant reductions in staphylococcal adherence, dependent on both CD9 and syndecan-1. Exogenous fibronectin caused a CD9-dependent increase in staphylococcal adhesion, whereas blockade of β1 integrins did not affect adhesion but did reduce the subsequent internalization of adhered bacteria. CD9 disruption or deletion increased β1 integrin-mediated internalization, suggesting that CD9 coordinates sequential staphylococcal adhesion and internalization. CD9 controls staphylococcal adhesion through syndecan-1, using a mechanism that likely requires CD9-mediated syndecan organization to correctly display fibronectin at the host cell surface. We propose that CD9-derived peptides or heparin analogs could be developed as anti-adhesion treatments to inhibit the initial stages of staphylococcal pathogenesis. IMPORTANCE Staphylococcus aureus infection is a significant cause of disease and morbidity. Staphylococci utilize multiple adhesion pathways to associate with epithelial cells, including interactions with proteoglycans or β1 integrins through a fibronectin bridge. Interference with another host protein, tetraspanin CD9, halves staphylococcal adherence to epithelial cells, although CD9 does not interact directly with bacteria. Here, we define the role of CD9 in staphylococcal adherence and uptake, observing that CD9 coordinates syndecan-1, fibronectin, and β1 integrins to allow efficient staphylococcal infection. Two treatments that disrupt this action are effective and may provide an alternative to antibiotics. We provide insights into the mechanisms that underlie staphylococcal infection of host cells, linking two known adhesion pathways together through CD9 for the first time.
Collapse
Affiliation(s)
- Luke R. Green
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Rahaf Issa
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Fawzyah Albaldi
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Lucy Urwin
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Ruth Thompson
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Henna Khalid
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Claire E. Turner
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Barbara Ciani
- Department of Chemistry, University of Sheffield, Sheffield, United Kingdom
| | - Lynda J. Partridge
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Peter N. Monk
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| |
Collapse
|
23
|
Motta C, Pellegrini A, Camaione S, Geoghegan J, Speziale P, Barbieri G, Pietrocola G. von Willebrand factor-binding protein (vWbp)-activated factor XIII and transglutaminase 2 (TG2) promote cross-linking between FnBPA from Staphylococcus aureus and fibrinogen. Sci Rep 2023; 13:11683. [PMID: 37468579 DOI: 10.1038/s41598-023-38972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/18/2023] [Indexed: 07/21/2023] Open
Abstract
The secreted von Willebrand factor-binding protein (vWbp) from Staphylococcus aureus interacts with the coagulation factors prothrombin and fibrinogen (Fbg), leading to the non-proteolytic transglutaminase activation of Factor XIII (FXIII). In this study we found that vWbp-activated FXIII catalyses the incorporation of amino-donor dansylcadaverine into region A of fibronectin-binding protein A (FnBPA). Incubation of Fbg with recombinant region A of S. aureus Fbg-binding proteins FnBPA, FnBPB, ClfA or ClfB in presence of vWbp-activated FXIII resulted in the formation of high molecular heteropolymers with FnBPA only, suggesting a specificity of the cross-linking reaction between fibrin(ogen) and the staphylococcal surface. As previously observed, cross-linking sites were mapped to the α-chain and the N1 subdomain of fibrin(ogen) and region A of FnBPA, respectively. Comparable results were obtained when tissue tranglutaminase-2 (TG2) was tested for cross-linking of FnBPA and Fbg. Of note, FnBPA-mediated covalent cross-linking promoted by vWbp-activated FXIII was also observed when bacteria were allowed to attach to fibrin(ogen). Together these findings suggest a novel pathogenetic mechanism by which the transglutaminase action of FXIII and/or TG2 contributes to entrapment and persistence of S. aureus in blood and host tissues.
Collapse
Affiliation(s)
- Chiara Motta
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Stefano Camaione
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Joan Geoghegan
- Institute of Microbiology and Infection, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Pietro Speziale
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | | |
Collapse
|
24
|
Leidecker M, Bertling A, Hussain M, Bischoff M, Eble JA, Fender AC, Jurk K, Rumpf C, Herrmann M, Kehrel BE, Niemann S. Protein Disulfide Isomerase and Extracellular Adherence Protein Cooperatively Potentiate Staphylococcal Invasion into Endothelial Cells. Microbiol Spectr 2023; 11:e0388622. [PMID: 36995240 PMCID: PMC10269700 DOI: 10.1128/spectrum.03886-22] [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: 09/26/2022] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
Invasion of host cells is an important feature of Staphylococcus aureus. The main internalization pathway involves binding of the bacteria to host cells, e.g., endothelial cells, via a fibronectin (Fn) bridge between S. aureus Fn binding proteins and α5β1-integrin, followed by phagocytosis. The secreted extracellular adherence protein (Eap) has been shown to promote this cellular uptake pathway of not only S. aureus, but also of bacteria otherwise poorly taken up by host cells, such as Staphylococcus carnosus. The exact mechanisms are still unknown. Previously, we demonstrated that Eap induces platelet activation by stimulation of the protein disulfide isomerase (PDI), a catalyst of thiol-disulfide exchange reactions. Here, we show that Eap promotes PDI activity on the surface of endothelial cells, and that this contributes critically to Eap-driven staphylococcal invasion. PDI-stimulated β1-integrin activation followed by increased Fn binding to host cells likely accounts for the Eap-enhanced uptake of S. aureus into non-professional phagocytes. Additionally, Eap supports the binding of S. carnosus to Fn-α5β1 integrin, thereby allowing its uptake into endothelial cells. To our knowledge, this is the first demonstration that PDI is crucial for the uptake of bacteria into host cells. We describe a hitherto unknown function of Eap-the promotion of an enzymatic activity with subsequent enhancement of bacterial uptake-and thus broaden mechanistic insights into its importance as a driver of bacterial pathogenicity. IMPORTANCE Staphylococcus aureus can invade and persist in non-professional phagocytes, thereby escaping host defense mechanisms and antibiotic treatment. The intracellular lifestyle of S. aureus contributes to the development of infection, e.g., in infective endocarditis or chronic osteomyelitis. The extracellular adherence protein secreted by S. aureus promotes its own internalization as well as that of bacteria that are otherwise poorly taken up by host cells, such as Staphylococcus carnosus. In our study, we demonstrate that staphylococcal uptake by endothelial cells requires catalytic disulfide exchange activity by the cell-surface protein disulfide isomerase, and that this critical enzymatic function is enhanced by Eap. The therapeutic application of PDI inhibitors has previously been investigated in the context of thrombosis and hypercoagulability. Our results add another intriguing possibility: therapeutically targeting PDI, i.e., as a candidate approach to modulate the initiation and/or course of S. aureus infectious diseases.
Collapse
Affiliation(s)
- Marleen Leidecker
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Anne Bertling
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
| | - Muzaffar Hussain
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg, Germany
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Anke C. Fender
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
- Institute of Pharmacology, University Hospital Essen, Essen, Germany
| | - Kerstin Jurk
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christine Rumpf
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Beate E. Kehrel
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| |
Collapse
|
25
|
Wang C, Paiva TO, Motta C, Speziale P, Pietrocola G, Dufrêne YF. Catch Bond-Mediated Adhesion Drives Staphylococcus aureus Host Cell Invasion. NANO LETTERS 2023. [PMID: 37267288 DOI: 10.1021/acs.nanolett.3c01387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Various viruses and pathogenic bacteria interact with annexin A2 to invade mammalian cells. Here, we show that Staphylococcus aureus engages in extremely strong catch bonds for host cell invasion. By means of single-molecule atomic force microscopy, we find that bacterial surface-located clumping factors bind annexin A2 with extraordinary strength, indicating that these bonds are extremely resilient to mechanical tension. By determining the lifetimes of the complexes under increasing mechanical stress, we demonstrate that the adhesins form catch bonds with their ligand that are capable to sustain forces of 1500-1700 pN. The force-dependent adhesion mechanism identified here provides a molecular framework to explain how S. aureus pathogens tightly attach to host cells during invasion and shows promise for the design of new therapeutics against intracellular S. aureus.
Collapse
Affiliation(s)
- Can Wang
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Telmo O Paiva
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Chiara Motta
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Pietro Speziale
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Giampiero Pietrocola
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Viale Taramelli 3/b, 27100 Pavia, Italy
| | - Yves F Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
26
|
Fu Y, Shi Y, Wang L, Zhao Y, Wang R, Li K, Zhang S, Zha X, Wang W, Zhao X, Yang W. All-Natural Immunomodulatory Bioadhesive Hydrogel Promotes Angiogenesis and Diabetic Wound Healing by Regulating Macrophage Heterogeneity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206771. [PMID: 36862027 PMCID: PMC10161050 DOI: 10.1002/advs.202206771] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/10/2023] [Indexed: 05/06/2023]
Abstract
Macrophages are highly heterogeneous and exhibit a diversity of functions and phenotypes. They can be divided into pro-inflammatory macrophages (M1) and anti-inflammatory macrophages (M2). Diabetic wounds are characterized by a prolonged inflammatory phase and difficulty in healing due to the accumulation of pro-inflammatory (M1) macrophages in the wound. Therefore, hydrogel dressings with macrophage heterogeneity regulation function hold great promise in promoting diabetic wound healing in clinical applications. However, the precise conversion of pro-inflammatory M1 to anti-inflammatory M2 macrophages by simple and biosafe approaches is still a great challenge. Here, an all-natural hydrogel with the ability to regulate macrophage heterogeneity is developed to promote angiogenesis and diabetic wound healing. The protocatechuic aldehyde hybridized collagen-based all-natural hydrogel exhibits good bioadhesive and antibacterial properties as well as reactive oxygen species scavenging ability. More importantly, the hydrogel is able to convert M1 macrophages into M2 macrophages without the need for any additional ingredients or external intervention. This simple and safe immunomodulatory approach shows great application potential for shortening the inflammatory phase of diabetic wound repair and accelerating wound healing.
Collapse
Affiliation(s)
- Ya‐Jun Fu
- College of Polymer Science and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Yi‐Feng Shi
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Li‐Ya Wang
- Department of NephrologyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Yi‐Fan Zhao
- Department of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengdu610032P. R. China
| | - Rao‐Kaijuan Wang
- Department of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengdu610032P. R. China
| | - Kai Li
- Department of Thoracic OncologyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Shu‐Ting Zhang
- College of Polymer Science and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Xiang‐Jun Zha
- Laboratory of Liver TransplantationWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Wei Wang
- Department of NeurosurgeryWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Xing Zhao
- Department of NephrologyWest China HospitalSichuan UniversityChengdu610041P. R. China
| | - Wei Yang
- College of Polymer Science and EngineeringSichuan UniversityChengdu610065P. R. China
| |
Collapse
|
27
|
Mbanga J, Kodzai NP, Oosthuysen WF. Antibiotic resistance, pathotypes, and pathogen-host interactions in Escherichia coli from hospital wastewater in Bulawayo, Zimbabwe. PLoS One 2023; 18:e0282273. [PMID: 36862713 PMCID: PMC9980749 DOI: 10.1371/journal.pone.0282273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/10/2023] [Indexed: 03/03/2023] Open
Abstract
This study aimed to characterise E. coli strains isolated from hospital wastewater effluent in Bulawayo, Zimbabwe, using both molecular and cytological approaches. Wastewater samples were aseptically collected from the sewerage mains of a major public referral hospital in Bulawayo province weekly for one month. A total of 94 isolates were isolated and confirmed as E. coli through biotyping and PCR targeting the uidA housekeeping gene. A total of 7 genes (eagg, eaeA, stx, flicH7, ipaH, lt, and st genes) coding for virulence in diarrheagenic E. coli were targeted. Antibiotic susceptibility of E. coli was determined against a panel of 12 antibiotics through the disk diffusion assay. The infectivity status of the observed pathotypes was investigated using HeLa cells through adherence, invasion, and intracellular assay. None of the 94 isolates tested positive for the ipaH and flicH7genes. However, 48 (53.3%) isolates were enterotoxigenic E. coli (ETEC) (lt gene positive), 2 (2.13%) isolates were enteroaggregative E. coli (EAEC) (eagg gene), and 1 (1.06%) isolate was enterohaemorrhagic E. coli (EHEC) (stx and eaeA). A high level of sensitivity was observed in E. coli against ertapenem (98.9%), and Azithromycin (75.5%). The highest resistance was against ampicillin (92.6%) and sulphamethoxazole-trimethoprim (90.4%). Seventy-nine (84%) E. coli isolates exhibited multidrug resistance. The infectivity study results indicated that environmentally isolated pathotypes were as infective as the clinically isolated pathotypes for all three parameters. No adherent cells were observed using ETEC, and no cells were observed in the intracellular survival assay using EAEC. This study revealed that hospital wastewater is a hotspot for pathogenic E. coli and that the environmentally isolated pathotypes maintained their ability to colonise and infect mammalian cells.
Collapse
Affiliation(s)
- Joshua Mbanga
- Department of Applied Biology and Biochemistry, National University of Science and Technology, Bulawayo, Zimbabwe
- * E-mail:
| | - Nokukhanya P. Kodzai
- Department of Applied Biology and Biochemistry, National University of Science and Technology, Bulawayo, Zimbabwe
| | | |
Collapse
|
28
|
Goncheva MI, Gibson RM, Shouldice AC, Dikeakos JD, Heinrichs DE. The Staphylococcus aureus protein IsdA increases SARS CoV-2 replication by modulating JAK-STAT signaling. iScience 2023; 26:105975. [PMID: 36687318 PMCID: PMC9838083 DOI: 10.1016/j.isci.2023.105975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/28/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (CoV-2) pandemic has affected millions globally. A significant complication of CoV-2 infection is secondary bacterial co-infection, as seen in approximately 25% of severe cases. The most common organism isolated during co-infection is Staphylococcus aureus. Here, we describe the development of an in vitro co-infection model where both viral and bacterial replication kinetics may be examined. We demonstrate CoV-2 infection does not alter bacterial interactions with host epithelial cells. In contrast, S. aureus enhances CoV-2 replication by 10- to 15-fold. We identify this pro-viral activity is due to the S. aureus iron-regulated surface determinant A (IsdA) protein and demonstrate IsdA modifies host transcription. We find that IsdA alters Janus Kinase - Signal Transducer and Activator of Transcription (JAK-STAT) signaling, by affecting JAK2-STAT3 levels, ultimately leading to increased viral replication. These findings provide key insight into the molecular interactions between host cells, CoV-2 and S. aureus during co-infection.
Collapse
Affiliation(s)
- Mariya I. Goncheva
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada,Corresponding author
| | - Richard M. Gibson
- ImPaKT Laboratory, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Ainslie C. Shouldice
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - David E. Heinrichs
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada,Corresponding author
| |
Collapse
|
29
|
Staphylococcus aureus induces mitophagy to promote its survival within bovine mammary epithelial cells. Vet Microbiol 2023; 280:109697. [PMID: 36827937 DOI: 10.1016/j.vetmic.2023.109697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023]
Abstract
Mitophagy occurs in a variety of pathogenic infections. However, the role of mitophagy in the intracellular survival of Staphylococcus aureus (S.aureus) within bovine mammary epithelial cells (BMECs) and which molecules specifically mediate the induction of mitophagy remains unclear. Therefore, this study aims to investigate the role and mechanism of mitophagy in the intracellular survival of S.aureus. Here, we reported that S.aureus induced complete mitophagy to promote its survival within BMECs. The further mechanistic study showed that S. aureus induced mitophagy by activating the p38-PINK1-Parkin signaling pathway. These findings expand our knowledge of the intracellular survival mechanism of S.aureus in the host and provide a desirable therapeutic strategy against S.aureus and other intracellular infections.
Collapse
|
30
|
Lyon LM, Doran KS, Horswill AR. Staphylococcus aureus Fibronectin-Binding Proteins Contribute to Colonization of the Female Reproductive Tract. Infect Immun 2023; 91:e0046022. [PMID: 36511703 PMCID: PMC9872658 DOI: 10.1128/iai.00460-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an opportunistic pathogen and frequent colonizer of human skin and mucosal membranes, including the vagina, with vaginal colonization reaching nearly 25% in some pregnant populations. MRSA vaginal colonization can lead to aerobic vaginitis (AV), and during pregnancy, bacterial ascension into the upper reproductive tract can lead to adverse birth outcomes. USA300, the most prominent MRSA lineage to colonize pregnant individuals, is a robust biofilm former and causative agent of invasive infections; however, little is known about how it colonizes and ascends in the female reproductive tract (FRT). Our previous studies showed that a MRSA mutant of seven fibrinogen-binding adhesins was deficient in FRT epithelial attachment and colonization. Using both monolayer and multilayer air-liquid interface cell culture models, we determine that one class of these adhesins, the fibronectin binding proteins (FnBPA and FnBPB), are critical for association with human vaginal epithelial cells (hVECs) and hVEC invasion through interactions with α5β1 integrin. We observe that both FnBPs are important for biofilm formation as single and double fnbAB mutants exhibit reduced biofilm formation on hVECs. Using heterologous expression of fnbA and fnbB in Staphylococcus carnosus, FnBPs are also found to be sufficient for hVEC cellular association, invasion, and biofilm formation. In addition, we found that an ΔfnbAB mutant displays attenuated ascension in our murine vaginal colonization model. Better understanding of MRSA FRT colonization and ascension can ultimately inform treatment strategies to limit MRSA vaginal burden or prevent ascension, especially during pregnancy and in those prone to AV.
Collapse
Affiliation(s)
- Laurie M. Lyon
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| | - Kelly S. Doran
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
- Department of Veterans Affairs, Eastern Colorado Healthcare System, Aurora, Colorado, USA
| |
Collapse
|
31
|
Rodrigues Lopes I, Alcantara LM, Silva RJ, Josse J, Vega EP, Cabrerizo AM, Bonhomme M, Lopez D, Laurent F, Vandenesch F, Mano M, Eulalio A. Microscopy-based phenotypic profiling of infection by Staphylococcus aureus clinical isolates reveals intracellular lifestyle as a prevalent feature. Nat Commun 2022; 13:7174. [PMID: 36418309 PMCID: PMC9684519 DOI: 10.1038/s41467-022-34790-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022] Open
Abstract
Staphylococcus aureus is increasingly recognized as a facultative intracellular pathogen, although the significance and pervasiveness of its intracellular lifestyle remain controversial. Here, we applied fluorescence microscopy-based infection assays and automated image analysis to profile the interaction of 191 S. aureus isolates from patients with bone/joint infections, bacteremia, and infective endocarditis, with four host cell types, at five times post-infection. This multiparametric analysis revealed that almost all isolates are internalized and that a large fraction replicate and persist within host cells, presenting distinct infection profiles in non-professional vs. professional phagocytes. Phenotypic clustering highlighted interesting sub-groups, including one comprising isolates exhibiting high intracellular replication and inducing delayed host death in vitro and in vivo. These isolates are deficient for the cysteine protease staphopain A. This study establishes S. aureus intracellular lifestyle as a prevalent feature of infection, with potential implications for the effective treatment of staphylococcal infections.
Collapse
Affiliation(s)
- Ines Rodrigues Lopes
- grid.8051.c0000 0000 9511 4342RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Laura Maria Alcantara
- grid.8051.c0000 0000 9511 4342RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ricardo Jorge Silva
- grid.8051.c0000 0000 9511 4342Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Jerome Josse
- grid.15140.310000 0001 2175 9188Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Elena Pedrero Vega
- grid.4711.30000 0001 2183 4846National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Ana Marina Cabrerizo
- grid.4711.30000 0001 2183 4846National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Melanie Bonhomme
- grid.15140.310000 0001 2175 9188Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Daniel Lopez
- grid.4711.30000 0001 2183 4846National Centre for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Frederic Laurent
- grid.15140.310000 0001 2175 9188Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France ,grid.413852.90000 0001 2163 3825Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Francois Vandenesch
- grid.15140.310000 0001 2175 9188Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France ,grid.413852.90000 0001 2163 3825Centre National de Référence des Staphylocoques, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Miguel Mano
- grid.8051.c0000 0000 9511 4342Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Department of Life Sciences, University of Coimbra, Coimbra, Portugal ,grid.13097.3c0000 0001 2322 6764British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London, United Kingdom
| | - Ana Eulalio
- grid.8051.c0000 0000 9511 4342RNA & Infection Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,grid.7311.40000000123236065Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal ,grid.7445.20000 0001 2113 8111Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
32
|
Granata V, Possetti V, Parente R, Bottazzi B, Inforzato A, Sobacchi C. The osteoblast secretome in Staphylococcus aureus osteomyelitis. Front Immunol 2022; 13:1048505. [PMID: 36483565 PMCID: PMC9723341 DOI: 10.3389/fimmu.2022.1048505] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
Osteomyelitis (OM) is an infectious disease of the bone predominantly caused by the opportunistic bacterium Staphylococcus aureus (S. aureus). Typically established upon hematogenous spread of the pathogen to the musculoskeletal system or contamination of the bone after fracture or surgery, osteomyelitis has a complex pathogenesis with a critical involvement of both osteal and immune components. Colonization of the bone by S. aureus is traditionally proposed to induce functional inhibition and/or apoptosis of osteoblasts, alteration of the RANKL/OPG ratio in the bone microenvironment and activation of osteoclasts; all together, these events locally subvert tissue homeostasis causing pathological bone loss. However, this paradigm has been challenged in recent years, in fact osteoblasts are emerging as active players in the induction and orientation of the immune reaction that mounts in the bone during an infection. The interaction with immune cells has been mostly ascribed to osteoblast-derived soluble mediators that add on and synergize with those contributed by professional immune cells. In this respect, several preclinical and clinical observations indicate that osteomyelitis is accompanied by alterations in the local and (sometimes) systemic levels of both pro-inflammatory (e.g., IL-6, IL-1α, TNF-α, IL-1β) and anti-inflammatory (e.g., TGF-β1) cytokines. Here we revisit the role of osteoblasts in bacterial OM, with a focus on their secretome and its crosstalk with cellular and molecular components of the bone microenvironment and immune system.
Collapse
Affiliation(s)
- Valentina Granata
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Milan Unit, National Research Council - Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy
| | - Valentina Possetti
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | | | - Antonio Inforzato
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Cristina Sobacchi
- IRCCS Humanitas Research Hospital, Rozzano, Italy,Milan Unit, National Research Council - Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy,*Correspondence: Cristina Sobacchi,
| |
Collapse
|
33
|
Cegarra C, Cameron B, Chaves C, Dabdoubi T, Do TM, Genêt B, Roudières V, Shi Y, Tchepikoff P, Lesuisse D. An innovative strategy to identify new targets for delivering antibodies to the brain has led to the exploration of the integrin family. PLoS One 2022; 17:e0274667. [PMID: 36108060 PMCID: PMC9477330 DOI: 10.1371/journal.pone.0274667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Increasing brain exposure of biotherapeutics is key to success in central nervous system disease drug discovery. Accessing the brain parenchyma is especially difficult for large polar molecules such as biotherapeutics and antibodies because of the blood-brain barrier. We investigated a new immunization strategy to identify novel receptors mediating transcytosis across the blood-brain barrier.
Method
We immunized mice with primary non-human primate brain microvascular endothelial cells to obtain antibodies. These antibodies were screened for their capacity to bind and to be internalized by primary non-human primate brain microvascular endothelial cells and Human Cerebral Microvascular Endothelial Cell clone D3. They were further evaluated for their transcytosis capabilities in three in vitro blood-brain barrier models. In parallel, their targets were identified by two different methods and their pattern of binding to human tissue was investigated using immunohistochemistry.
Results
12 antibodies with unique sequence and internalization capacities were selected amongst more than six hundred. Aside from one antibody targeting Activated Leukocyte Cell Adhesion Molecule and one targeting Striatin3, most of the other antibodies recognized β1 integrin and its heterodimers. The antibody with the best transcytosis capabilities in all blood-brain barrier in vitro models and with the best binding capacity was an anti-αnβ1 integrin. In comparison, commercial anti-integrin antibodies performed poorly in transcytosis assays, emphasizing the originality of the antibodies derived here. Immunohistochemistry studies showed specific vascular staining on human and non-human primate tissues.
Conclusions
This transcytotic behavior has not previously been reported for anti-integrin antibodies. Further studies should be undertaken to validate this new mechanism in vivo and to evaluate its potential in brain delivery.
Collapse
Affiliation(s)
- Céline Cegarra
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
- * E-mail:
| | | | - Catarina Chaves
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | | | - Tuan-Minh Do
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Bruno Genêt
- Integrated Drug Discovery, Sanofi, Vitry-Sur-Seine, France
| | - Valérie Roudières
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| | - Yi Shi
- Histology, Translational Sciences, Sanofi, Vitry-Sur-Seine, France
| | | | - Dominique Lesuisse
- Rare and Neurologic Diseases Research Therapeutic Area, Sanofi, Chilly Mazarin, France
| |
Collapse
|
34
|
Miyake R, Iwamoto K, Sakai N, Matsunae K, Aziz F, Sugai M, Takahagi S, Tanaka A, Hide M. Uptake of Staphylococcus aureus by keratinocytes is reduced by interferon-fibronectin pathway and filaggrin expression. J Dermatol 2022; 49:1148-1157. [PMID: 35983802 DOI: 10.1111/1346-8138.16546] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/16/2022] [Accepted: 07/28/2022] [Indexed: 11/27/2022]
Abstract
Staphylococcus aureus (S. aureus) is frequently detected in the skin of patients with atopic dermatitis (AD). AD skin-derived strains of S. aureus (AD strain) are selectively internalized into keratinocytes (HaCaT cells) compared to standard strains. However, the mechanism of AD strain internalization by keratinocytes and effect of the skin environment on internalization remain unclear. HaCaT cells were exposed to heat-killed AD or standard strains of fluorescently labeled S. aureus, with or without interferon (IFN)-γ, interleukin (IL)-4, and IL-13 cytokines, for 24 h. Filaggrin and fibronectin expression in HaCaT cells was knocked down using small interfering RNA. The amount of internalized S. aureus was evaluated using a cell imaging system. The effects of INF-γ, IL-4, and S. aureus exposure on mRNA expression in HaCaT cells were analyzed using single-cell RNA sequencing. AD strains adhered to HaCaT cells in approximately 15 min and were increasingly internalized for up to 3 h (2361 ± 467 spots/100 cells, mean ± SD), whereas the standard strain was not (991 ± 71 spots/100 cells). In the presence of IFN-γ, both the number of internalized strains and fibronectin expression significantly decreased compared to in the control, whereas Th2 cytokines had no significant effects. The number of internalized AD strains was significantly higher in filaggrin knockdown and lower in fibronectin knockdown HaCaT cells compared to in the control. RNA sequencing revealed that IFN-γ decreased both fibronectin and filaggrin expression. Keratinocyte internalization of the AD strain may be predominantly mediated by the INF-γ-fibronectin pathway and partially regulated by filaggrin expression.
Collapse
Affiliation(s)
- Ryu Miyake
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Kazumasa Iwamoto
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences Hiroshima University, Hiroshima, Japan
| | - Kyoka Matsunae
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Fatkhanuddin Aziz
- Department of Bioresources Technology and Veterinary, Vocational, College Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Motoyuki Sugai
- Antimicrobial Resistance Research Center National Institute of Infectious Diseases, Tokyo, Japan
| | - Shunsuke Takahagi
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences Hiroshima University, Hiroshima, Japan.,Department of Dermatology Hiroshima Citizens Hospital, Hiroshima, Japan
| |
Collapse
|
35
|
Khateb H, Sørensen RS, Cramer K, Eklund AS, Kjems J, Meyer RL, Jungmann R, Sutherland DS. The Role of Nanoscale Distribution of Fibronectin in the Adhesion of Staphylococcus aureus Studied by Protein Patterning and DNA-PAINT. ACS NANO 2022; 16:10392-10403. [PMID: 35801826 PMCID: PMC9330902 DOI: 10.1021/acsnano.2c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Staphylococcus aureus is a widespread and highly virulent pathogen that can cause superficial and invasive infections. Interactions between S. aureus surface receptors and the extracellular matrix protein fibronectin mediate the bacterial invasion of host cells and is implicated in the colonization of medical implant surfaces. In this study, we investigate the role of distribution of both fibronectin and cellular receptors on the adhesion of S. aureus to interfaces as a model for primary adhesion at tissue interfaces or biomaterials. We present fibronectin in patches of systematically varied size (100-1000 nm) in a background of protein and bacteria rejecting chemistry based on PLL-g-PEG and studied S. aureus adhesion under flow. We developed a single molecule imaging assay for localizing fibronectin binding receptors on the surface of S. aureus via the super-resolution DNA points accumulation for imaging in nanoscale topography (DNA-PAINT) technique. Our results indicate that S. aureus adhesion to fibronectin biointerfaces is regulated by the size of available ligand patterns, with an adhesion threshold of 300 nm and larger. DNA-PAINT was used to visualize fibronectin binding receptor organization in situ at ∼7 nm localization precision and with a surface density of 38-46 μm-2, revealing that the engagement of two or more receptors is required for strong S. aureus adhesion to fibronectin biointerfaces.
Collapse
Affiliation(s)
- Heba Khateb
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| | - Rasmus S. Sørensen
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| | - Kimberly Cramer
- Max
Planck Institute of Biochemistry, Martinsried 82152, Germany
| | | | - Jorgen Kjems
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
- Department
of Molecular Biology and Genetics Aarhus
University Aarhus
C 8000, Denmark
| | - Rikke L. Meyer
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| | - Ralf Jungmann
- Max
Planck Institute of Biochemistry, Martinsried 82152, Germany
- Faculty
of Physics and Center for Nanoscience, Ludwig
Maximilian University, Munich 80539, Germany
| | - Duncan S. Sutherland
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University Aarhus C 8000, Denmark
| |
Collapse
|
36
|
Kinney KJ, Stach JM, Kulhankova K, Brown M, Salgado-Pabón W. Vegetation Formation in Staphylococcus Aureus Endocarditis Inversely Correlates With RNAIII and sarA Expression in Invasive Clonal Complex 5 Isolates. Front Cell Infect Microbiol 2022; 12:925914. [PMID: 35860377 PMCID: PMC9289551 DOI: 10.3389/fcimb.2022.925914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/14/2022] [Indexed: 01/29/2023] Open
Abstract
Infective endocarditis (IE) is one of the most feared and lethal diseases caused by Staphylococcus aureus. Once established, the infection is fast-progressing and tissue destructive. S. aureus of the clonal complex 5 (CC5) commonly cause IE yet are severely understudied. IE results from bacterial colonization and formation of tissue biofilms (known as vegetations) on injured or inflamed cardiac endothelium. S. aureus IE is promoted by adhesins, coagulases, and superantigens, with the exotoxins and exoenzymes likely contributing to tissue destruction and dissemination. Expression of the large repertoire of virulence factors required for IE and sequelae is controlled by complex regulatory networks. We investigated the temporal expression of the global regulators agr (RNAIII), rot, sarS, sarA, sigB, and mgrA in 8 invasive CC5 isolates and established intrinsic expression patterns associated with IE outcomes. We show that vegetation formation, as tested in the rabbit model of IE, inversely correlates with RNAIII and sarA expression during growth in Todd-Hewitt broth (TH). Large vegetations with severe sequelae arise from strains with high-level expression of colonization factors but slower transition towards expression of the exotoxins. Overall, strains proficient in vegetation formation, a hallmark of IE, exhibit lower expression of RNAIII and sarA. Simultaneous high expression of RNAIII, sarA, sigB, and mgrA is the one phenotype assessed in this study that fails to promote IE. Thus, RNAIII and sarA expression that provides for rheostat control of colonization and virulence genes, rather than an on and off switch, promote both vegetation formation and lethal sepsis.
Collapse
Affiliation(s)
- Kyle J. Kinney
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Jessica M. Stach
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Katarina Kulhankova
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Matthew Brown
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Wilmara Salgado-Pabón
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
37
|
Wang X, Liu Y. Offense and Defense in Granulomatous Inflammation Disease. Front Cell Infect Microbiol 2022; 12:797749. [PMID: 35846773 PMCID: PMC9277142 DOI: 10.3389/fcimb.2022.797749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Granulomatous inflammation (GI) diseases are a group of chronic inflammation disorders characterized by focal collections of multinucleated giant cells, epithelioid cells and macrophages, with or without necrosis. GI diseases are closely related to microbes, especially virulent intracellular bacterial infections are important factors in the progression of these diseases. They employ a range of strategies to survive the stresses imposed upon them and persist in host cells, becoming the initiator of the fighting. Microbe-host communication is essential to maintain functions of a healthy host, so defense capacity of hosts is another influence factor, which is thought to combine to determine the result of the fighting. With the development of gene research technology, many human genetic loci were identified to be involved in GI diseases susceptibility, providing more insights into and knowledge about GI diseases. The current review aims to provide an update on the most recent progress in the identification and characterization of bacteria in GI diseases in a variety of organ systems and clinical conditions, and examine the invasion and escape mechanisms of pathogens that have been demonstrated in previous studies, we also review the existing data on the predictive factors of the host, mainly on genetic findings. These strategies may improve our understanding of the mechanisms underlying GI diseases, and open new avenues for the study of the associated conditions in the future.
Collapse
Affiliation(s)
- Xinwen Wang
- Shaanxi Clinical Research Center for Oral Diseases, National Clinical Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Department of Oral Medicine, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| | - Yuan Liu
- Shaanxi International Joint Research Center for Oral Diseases, State Key Laboratory of Military Stomatology, Department of Histology and Pathology, School of Stomatology, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
38
|
Mechanisms and Implications of Bacterial Invasion across the Human Skin Barrier. Microbiol Spectr 2022; 10:e0274421. [PMID: 35532353 PMCID: PMC9241919 DOI: 10.1128/spectrum.02744-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atopic dermatitis (AD) is associated with a deficiency of skin lipids, increased populations of Staphylococcus aureus in the microbiome, and structural defects in the stratum corneum (SC), the outermost layer of human skin. However, the pathogenesis of AD is ambiguous, as it is unclear whether observed changes are the result of AD or contribute to the pathogenesis of the disease. Previous studies have shown that S. aureus is capable of permeating across isolated human SC tissue when lipids are depleted to levels consistent with AD conditions. In this study, we expand upon this discovery to determine the mechanisms and implications of bacterial penetration into the SC barrier. Specifically, we establish if bacteria are permeating intercellularly or employing a combination of both inter- and intracellular travel. The mechanical implications of bacterial invasion, lipid depletion, and media immersion are also evaluated using a newly developed, physiologically relevant, temperature-controlled drip chamber. Results reveal for the first time that S. aureus can be internalized by corneocytes, indicating transcellular movement through the tissue during permeation, consistent with previous theoretical models. S. aureus also degrades the mechanical integrity of human SC, particularly when the tissue is partially depleted of lipids. These observed mechanical changes are likely the cause of broken or ruptured tissue seen as exudative lesions in AD flares. This work further highlights the necessity of lipids in skin microbial barrier function. IMPORTANCE Millions of people suffer from the chronic inflammatory skin disease atopic dermatitis (AD), whose symptoms are associated with a deficiency of skin lipids that exhibit antimicrobial functions and increased populations of the opportunistic pathogen Staphylococcus aureus. However, the pathogenesis of AD is ambiguous, and it remains unclear if these observed changes are merely the result of AD or contribute to the pathogenesis of the disease. In this article, we demonstrate the necessity of skin lipids in preventing S. aureus from penetrating the outermost barrier of human skin, thereby causing a degradation in tissue integrity. This bacterial permeation into the viable epidermis could act as an inflammatory trigger of the disease. When coupled with delipidated AD tissue conditions, bacterial permeation can also explain increased tissue fragility, potentially causing lesion formation in AD patients that results in further enhancing bacterial permeability across the stratum corneum and the development of chronic conditions.
Collapse
|
39
|
Fibronectin binding protein B binds to loricrin and promotes corneocyte adhesion by Staphylococcus aureus. Nat Commun 2022; 13:2517. [PMID: 35523796 PMCID: PMC9076634 DOI: 10.1038/s41467-022-30271-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 04/19/2022] [Indexed: 12/23/2022] Open
Abstract
Colonisation of humans by Staphylococcus aureus is a major risk factor for infection, yet the bacterial and host factors involved are not fully understood. The first step during skin colonisation is adhesion of the bacteria to corneocytes in the stratum corneum where the cornified envelope protein loricrin is the main ligand for S. aureus. Here we report a novel loricrin-binding protein of S. aureus, the cell wall-anchored fibronectin binding protein B (FnBPB). Single-molecule force spectroscopy revealed both weak and ultra-strong (2 nN) binding of FnBPB to loricrin and that mechanical stress enhanced the strength of these bonds. Treatment with a peptide derived from fibrinogen decreased the frequency of strong interactions, suggesting that both ligands bind to overlapping sites within FnBPB. Finally, we show that FnBPB promotes adhesion to human corneocytes by binding strongly to loricrin, highlighting the relevance of this interaction to skin colonisation. The first step during skin colonization by is its adhesion to corneocytes. Da Costa et al. show that the cell wall-anchored fibronectin binding protein B (FnBPB) of S. aureus binds to loricrin. Applying single cell force spectroscopy, they demonstrate that this interaction promotes adhesion of S. aureus to human corneocytes.
Collapse
|
40
|
Diversity and pathogenesis of Staphylococcus aureus from bovine mastitis: current understanding and future perspectives. BMC Vet Res 2022; 18:115. [PMID: 35331225 PMCID: PMC8944054 DOI: 10.1186/s12917-022-03197-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/03/2022] [Indexed: 11/10/2022] Open
Abstract
Staphylococcus aureus is a leading cause of bovine mastitis worldwide. Despite some improved understanding of disease pathogenesis, progress towards new methods for the control of intramammary infections (IMI) has been limited, particularly in the field of vaccination. Although herd management programs have helped to reduce the number of clinical cases, S. aureus mastitis remains a major disease burden. This review summarizes the past 16 years of research on bovine S. aureus population genetics, and molecular pathogenesis that have been conducted worldwide. We describe the diversity of S. aureus associated with bovine mastitis and the geographical distribution of S. aureus clones in different continents. We also describe studies investigating the evolution of bovine S. aureus and the importance of host-adaptation in its emergence as a mastitis pathogen. The available information on the prevalence of virulence determinants and their functional relevance during the pathogenesis of bovine mastitis are also discussed. Although traits such as biofilm formation and innate immune evasion are critical for the persistence of bacteria, the current understanding of the key host-pathogen interactions that determine the outcome of S. aureus IMI is very limited. We suggest that greater investment in research into the genetic and molecular basis of bovine S. aureus pathogenesis is essential for the identification of novel therapeutic and vaccine targets.
Collapse
|
41
|
Ngo QV, Faass L, Sähr A, Hildebrand D, Eigenbrod T, Heeg K, Nurjadi D. Inflammatory Response Against Staphylococcus aureus via Intracellular Sensing of Nucleic Acids in Keratinocytes. Front Immunol 2022; 13:828626. [PMID: 35281009 PMCID: PMC8907419 DOI: 10.3389/fimmu.2022.828626] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/07/2022] [Indexed: 12/02/2022] Open
Abstract
Staphylococcus aureus is one of the clinically most relevant pathogens causing infections. Humans are often exposed to S. aureus. In approximately one-third of the healthy population it can be found on the skin either for long or short periods as colonizing "commensals", without inducing infections or an inflammatory immune response. While tolerating S. aureus seems to be limited to certain individuals and time periods in most cases, Staphylococcus epidermidis is tolerated permanently on the skin of almost all individuals without activating overwhelming skin inflammation. To investigate this, we co-cultured a keratinocyte cell line (HaCaT) with viable S. aureus or S. epidermidis to study the differences in the immune activation. S. aureus activated keratinocytes depicted by a profound IL-6 and IL-8 response, whereas S. epidermidis did not. Our data indicate that internalization of S. aureus and the subsequent intracellular sensing of bacterial nucleic acid may be essential for initiating inflammatory response in keratinocytes. Internalized dsRNA activates IL-6 and IL-8 release, but not TNF-α or IFNs by human keratinocytes. This is a non-specific effect of dsRNA, which can be induced using Poly(I:C), as well as RNA from S. aureus and S. epidermidis. However, only viable S. aureus were able to induce this response as these bacteria and not S. epidermidis were actively internalized by HaCaT. The stimulatory effect of S. aureus seems to be independent of the TLR3, -7 and -8 pathways.
Collapse
Affiliation(s)
- Quang Vinh Ngo
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Larissa Faass
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Max von Pettenkofer Institute, Chair for Medical Microbiology and Hygiene, Ludwig Maximilians University Munich, Munich, Germany
| | - Aline Sähr
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Dagmar Hildebrand
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Tatjana Eigenbrod
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Klaus Heeg
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
42
|
Suehiro Y, Nomura R, Matayoshi S, Otsugu M, Iwashita N, Nakano K. Evaluation of the collagen-binding properties and virulence of killed Streptococcus mutans in a silkworm model. Sci Rep 2022; 12:2800. [PMID: 35181690 PMCID: PMC8857238 DOI: 10.1038/s41598-022-06345-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 01/18/2022] [Indexed: 11/10/2022] Open
Abstract
Streptococcus mutans, a major pathogen of dental caries, is also known as a causative agent of cardiovascular disease. A 120 kDa collagen-binding protein (Cnm) of S. mutans is an important contributor to the pathogenicity of cardiovascular disease. Although dead bacteria have been detected in cardiovascular specimens by molecular biological methods, the pathogenicity of the bacteria remains unknown. Here, we analyzed the pathogenicity of killed S. mutans by focusing on collagen-binding ability and the effects on silkworms. In live S. mutans, Cnm-positive S. mutans had high collagen-binding activity, while Cnm-negative S. mutans had no such activity. After treatment with killed Cnm-positive S. mutans, amoxicillin-treated bacteria still had collagen-binding ability, while lysozyme-treated bacteria lost this ability. When live and amoxicillin-treated S. mutans strains were administered to silkworms, the survival rates of the silkworms were reduced; this reduction was more pronounced in Cnm-positive S. mutans infection than in Cnm-negative S. mutans infection. However, the administration of any of the lysozyme-treated bacteria did not reduce the survival rate of the silkworms. These results suggest that amoxicillin-killed Cnm-positive S. mutans strains maintain collagen-binding properties and pathogenicity in the silkworm model, and are possibly associated with pathogenicity in cardiovascular diseases.
Collapse
Affiliation(s)
- Yuto Suehiro
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, 1-8 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Ryota Nomura
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, 1-8 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Saaya Matayoshi
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, 1-8 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Masatoshi Otsugu
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, 1-8 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Naoki Iwashita
- Laboratory of Veterinary Pharmacology, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, 1-8 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
43
|
Schwarz C, Buchholz R, Jawad M, Hoesker V, Terwesten-Solé C, Karst U, Linsen L, Vogl T, Hoerr V, Wildgruber M, Faber C. Fingerprints of Element Concentrations in Infective Endocarditis Obtained by Mass Spectrometric Imaging and t-Distributed Stochastic Neighbor Embedding. ACS Infect Dis 2022; 8:360-372. [PMID: 35045258 DOI: 10.1021/acsinfecdis.1c00485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Staphylococcus aureus-induced infective endocarditis (IE) is a life-threatening disease. Differences in virulence between distinct S. aureus strains, which are partly based on the molecular mechanisms during bacterial adhesion, are not fully understood. Yet, distinct molecular or elemental patterns, occurring during specific steps in the adhesion process, may help to identify novel targets for accelerated diagnosis or improved treatment. Here, we use laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) of post-mortem tissue slices of an established mouse model of IE to obtain fingerprints of element distributions in infected aortic valve tissue. Three S. aureus strains with different virulence due to deficiency in distinct adhesion molecules (fibronectin-binding protein A and staphylococcal protein A) were used to assess strain-specific patterns. Data analysis was performed by t-distributed stochastic neighbor embedding (t-SNE) of mass spectrometry imaging data, using manual reference tissue classification in histological specimens. This procedure allowed for obtaining distinct element patterns in infected tissue for all three bacterial strains and for comparing those to patterns observed in healthy mice or after sterile inflammation of the valve. In tissue from infected mice, increased concentrations of calcium, zinc, and magnesium were observed compared to noninfected mice. Between S. aureus strains, pronounced variations were observed for manganese. The presented approach is sensitive for detection of S. aureus infection. For strain-specific tissue characterization, however, further improvements such as establishing a database with elemental fingerprints may be required.
Collapse
Affiliation(s)
- Christian Schwarz
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University Hospital Münster, 48149 Münster, Germany
| | - Rebecca Buchholz
- Institute of Inorganic and Analytical Chemistry, University of Münster, 48149 Münster, Germany
| | - Muhammad Jawad
- Institute of Computer Science, University of Münster, 48149 Münster, Germany
| | - Vanessa Hoesker
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University Hospital Münster, 48149 Münster, Germany
| | | | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, 48149 Münster, Germany
| | - Lars Linsen
- Institute of Computer Science, University of Münster, 48149 Münster, Germany
| | - Thomas Vogl
- Institute of Immunology, University Hospital Münster, 48149 Münster, Germany
| | - Verena Hoerr
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University Hospital Münster, 48149 Münster, Germany
| | - Moritz Wildgruber
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University Hospital Münster, 48149 Münster, Germany
- Department for Radiology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Cornelius Faber
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University Hospital Münster, 48149 Münster, Germany
| |
Collapse
|
44
|
Ma D, Brothers KM, Maher PL, Phillips NJ, Simonetti D, Pasculle AW, Richardson AR, Cooper VS, Urish KL. Staphylococcus aureus genotype variation among and within periprosthetic joint infections. J Orthop Res 2022; 40:420-428. [PMID: 33713379 PMCID: PMC8435540 DOI: 10.1002/jor.25031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/29/2021] [Accepted: 03/10/2021] [Indexed: 02/04/2023]
Abstract
Staphylococcus aureus is a common organism in orthopedic infections, but little is known about the genetic diversity of strains during an infectious process. Using periprosthetic joint infection (PJI) as a model, a prospective study was designed to quantify genetic variation among S. aureus strains both among and within patients. Whole genome sequencing and multilocus sequence typing was performed to genotype these two populations at high resolution. In nasal cultures, 78% of strains were of clonal complexes CC5, CC8, and CC30. In PJI cultures, only 63% could be classified in these common clonal complexes. The PJI cultures had a larger proportion of atypical strains, and these atypical strains were associated with poor host status and compromised immune conditions. Mutations in genes involved in fibronectin binding (ebh, fnbA, clfA, and clfB) systematically distinguished later PJI isolates from the first PJI isolate from each patient. Repeated mutations in S. aureus genes associated with extracellular matrix binding were identified, suggesting adaptive, parallel evolution of S. aureus during the development of PJI.
Collapse
Affiliation(s)
- Dongzhu Ma
- Arthritis and Arthroplasty Design Group; Department of Orthopaedic Surgery; University of Pittsburgh; Pittsburgh, Pennsylvania, USA
| | - Kimberly M. Brothers
- Arthritis and Arthroplasty Design Group; Department of Orthopaedic Surgery; University of Pittsburgh; Pittsburgh, Pennsylvania, USA
| | - Patrick L. Maher
- Arthritis and Arthroplasty Design Group; Department of Orthopaedic Surgery; University of Pittsburgh; Pittsburgh, Pennsylvania, USA
| | - Nathan J. Phillips
- Department of Microbiology and Molecular Genetics, and Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Simonetti
- Clinical Microbiology Laboratory; University of Pittsburgh Medical Center; Pittsburgh, Pennsylvania, USA
| | - A. William Pasculle
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony R. Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vaughn S. Cooper
- Department of Microbiology and Molecular Genetics, and Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kenneth L. Urish
- Arthritis and Arthroplasty Design Group; Department of Orthopaedic Surgery; University of Pittsburgh; Pittsburgh, Pennsylvania, USA
| |
Collapse
|
45
|
Pleiotropic Effects of Statins: New Therapeutic Approaches to Chronic, Recurrent Infection by Staphylococcus aureus. Pharmaceutics 2021; 13:pharmaceutics13122047. [PMID: 34959329 PMCID: PMC8706520 DOI: 10.3390/pharmaceutics13122047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 01/01/2023] Open
Abstract
An emergent approach to bacterial infection is the use of host rather than bacterial-directed strategies. This approach has the potential to improve efficacy in especially challenging infection settings, including chronic, recurrent infection due to intracellular pathogens. For nearly two decades, the pleiotropic effects of statin drugs have been examined for therapeutic usefulness beyond the treatment of hypercholesterolemia. Interest originated after retrospective studies reported decreases in the risk of death due to bacteremia or sepsis for those on a statin regimen. Although subsequent clinical trials have yielded mixed results and earlier findings have been questioned for biased study design, in vitro and in vivo studies have provided clear evidence of protective mechanisms that include immunomodulatory effects and the inhibition of host cell invasion. Ultimately, the benefits of statins in an infection setting appear to require attention to the underlying host response and to the timing of the dosage. From this examination of statin efficacy, additional novel host-directed strategies may produce adjunctive therapeutic approaches for the treatment of infection where traditional antimicrobial therapy continues to yield poor outcomes. This review focuses on the opportunistic pathogen, Staphylococcus aureus, as a proof of principle in examining the promise and limitations of statins in recalcitrant infection.
Collapse
|
46
|
Casillas-Ituarte NN, Staats AM, Lower BH, Stoodley P, Lower SK. Host blood proteins as bridging ligand in bacterial aggregation as well as anchor point for adhesion in the molecular pathogenesis of Staphylococcus aureus infections. Micron 2021; 150:103137. [PMID: 34392091 PMCID: PMC8484042 DOI: 10.1016/j.micron.2021.103137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/09/2021] [Accepted: 08/03/2021] [Indexed: 11/20/2022]
Abstract
Fibronectin (Fn) and fibrinogen (Fg) are major host proteins present in the extracellular matrix, blood, and coatings on indwelling medical devices. The ability of Staphylococcus aureus to cause infections in humans depends on favorable interactions with these host ligands. Closely related bacterial adhesins, fibronectin-binding proteins A and B (FnBPA, FnBPB) were evaluated for two key steps in pathogenesis: clumping and adhesion. Experiments utilized optical spectrophotometry, flow cytometry, and atomic force microscopy to probe FnBPA/B alone or in combination in seven different strains of S. aureus and Lactococcus lactis, a Gram-positive surrogate that naturally lacks adhesins to mammalian ligands. In the absence of soluble ligands, both FnBPA and FnBPB were capable of interacting with adjacent FnBPs from neighboring bacteria to mediate clumping. In the presence of soluble host ligands, clumping was enhanced particularly under shear stress and with Fn present in the media. FnBPB exhibited greater ability to clump compared to FnBPA. The strength of adhesion was similar for immobilized Fn to FnBPA and FnBPB. These findings suggest that these two distinct but closely related bacterial adhesins, have different functional capabilities to interact with host ligands in different settings (e.g., soluble vs. immobilized). Survival and persistence of S. aureus in a human host may depend on complementary roles of FnBPA and FnBPB as they interact with different conformations of Fn or Fg (compact in solution vs. extended on a surface) present in different physiological spaces.
Collapse
Affiliation(s)
- Nadia N Casillas-Ituarte
- School of Earth Sciences, The Ohio State University, Columbus, OH, 43210, USA; School of Environment and Natural Resources, The Ohio State University, Columbus, OH, 43210, USA.
| | - Amelia M Staats
- Departments of Microbiology and Microbial Infection and Immunity, The Ohio State University, 43210, Columbus, OH, USA
| | - Brian H Lower
- School of Environment and Natural Resources, The Ohio State University, Columbus, OH, 43210, USA
| | - Paul Stoodley
- Departments of Microbiology and Microbial Infection and Immunity, The Ohio State University, 43210, Columbus, OH, USA; Department of Orthopaedics, The Ohio State University, Columbus, OH, 43210, USA
| | - Steven K Lower
- School of Earth Sciences, The Ohio State University, Columbus, OH, 43210, USA; School of Environment and Natural Resources, The Ohio State University, Columbus, OH, 43210, USA; Departments of Microbiology and Microbial Infection and Immunity, The Ohio State University, 43210, Columbus, OH, USA
| |
Collapse
|
47
|
Ginsenoside 20(S)-Rh2 promotes cellular pharmacokinetics and intracellular antibacterial activity of levofloxacin against Staphylococcus aureus through drug efflux inhibition and subcellular stabilization. Acta Pharmacol Sin 2021; 42:1930-1941. [PMID: 34462563 PMCID: PMC8564512 DOI: 10.1038/s41401-021-00751-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
Intracellular Staphylococcus aureus (S. aureus) often causes clinical failure and relapse after antibiotic treatment. We previously found that 20(S)-ginsenoside Rh2 [20(S)-Rh2] enhanced the therapeutic effect of quinolones in a mouse model of peritonitis, which we attributed to the increased concentrations of quinolones within bacteria. In this study, we investigated the enhancing effect of 20(S)-Rh2 on levofloxacin (LVF) from a perspective of intracellular bacteria. In S. aureus 25923-infected mice, coadministration of LVF (1.5 mg/kg, i.v.) and 20(S)-Rh2 (25, 50 mg/kg, i.g.) markedly increased the survival rate, and decreased intracellular bacteria counts accompanied by increased accumulation of LVF in peritoneal macrophages. In addition, 20(S)-Rh2 (1, 5, 10 μM) dose-dependently increased the uptake and accumulation of LVF in peritoneal macrophages from infected mice without drug treatment. In a model of S. aureus 25923-infected THP-1 macrophages, we showed that 20(S)-Rh2 (1, 5, 10 μM) dose-dependently enhanced the intracellular antibacterial activity of LVF. At the cellular level, 20(S)-Rh2 increased the intracellular accumulation of LVF by inhibiting P-gp and BCRP. PK-PD modeling revealed that 20(S)-Rh2 altered the properties of the cell but not LVF. At the subcellular level, 20(S)-Rh2 did not increase the distribution of LVF in lysosomes but exhibited a stronger sensitizing effect in acidic environments. Molecular dynamics (MD) simulations showed that 20(S)-Rh2 improved the stability of the DNA gyrase-LVF complex in lysosome-like acidic conditions. In conclusion, 20(S)-Rh2 promotes the cellular pharmacokinetics and intracellular antibacterial activities of LVF against S. aureus through efflux transporter inhibition and subcellular stabilization, which is beneficial for infection treatment.
Collapse
|
48
|
Laumay F, Benchetrit H, Corvaglia AR, van der Mee-Marquet N, François P. The Staphylococcus aureus CC398 Lineage: An Evolution Driven by the Acquisition of Prophages and Other Mobile Genetic Elements. Genes (Basel) 2021; 12:genes12111752. [PMID: 34828356 PMCID: PMC8623586 DOI: 10.3390/genes12111752] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/31/2022] Open
Abstract
Among clinically relevant lineages of Staphylococcus aureus, the lineage or clonal complex 398 (CC398) is of particular interest. Strains from this lineage were only described as livestock colonizers until 2007. Progressively, cases of infection were reported in humans in contact with farm animals, and now, CC398 isolates are increasingly identified as the cause of severe infections even in patients without any contact with animals. These observations suggest that CC398 isolates have spread not only in the community but also in the hospital setting. In addition, several recent studies have reported that CC398 strains are evolving towards increased virulence and antibiotic resistance. Identification of the origin and emergence of this clonal complex could probably benefit future large-scale studies that aim to detect sources of contamination and infection. Current evidence indicates that the evolution of CC398 strains towards these phenotypes has been driven by the acquisition of prophages and other mobile genetic elements. In this short review, we summarize the main knowledge of this major lineage of S. aureus that has become predominant in the human clinic worldwide within a single decade.
Collapse
Affiliation(s)
- Floriane Laumay
- Genomic Research Laboratory, Service of Infectious Diseases, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (F.L.); (A.-R.C.)
- Institut des Agents Infectieux, Centre de Biologie du Nord, Hospices Civils de Lyon, F-69003 Lyon, France
| | - Hugo Benchetrit
- UFR de Chimie et de Biologie, Faculté des Sciences, Université Grenoble Alpes, 38000 Grenoble, France;
| | - Anna-Rita Corvaglia
- Genomic Research Laboratory, Service of Infectious Diseases, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (F.L.); (A.-R.C.)
- Geneva Centre for Emerging Viral Diseases, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland
| | | | - Patrice François
- Genomic Research Laboratory, Service of Infectious Diseases, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland; (F.L.); (A.-R.C.)
- Correspondence:
| |
Collapse
|
49
|
Niemann S, Nguyen MT, Eble JA, Chasan AI, Mrakovcic M, Böttcher RT, Preissner KT, Roßlenbroich S, Peters G, Herrmann M. More Is Not Always Better-the Double-Headed Role of Fibronectin in Staphylococcus aureus Host Cell Invasion. mBio 2021; 12:e0106221. [PMID: 34663090 PMCID: PMC8524341 DOI: 10.1128/mbio.01062-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/17/2021] [Indexed: 12/30/2022] Open
Abstract
While Staphylococcus aureus has classically been considered an extracellular pathogen, these bacteria are also capable of being taken up by host cells, including nonprofessional phagocytes such as endothelial cells, epithelial cells, or osteoblasts. The intracellular S. aureus lifestyle contributes to infection development. The predominant recognition and internalization pathway appears to be the binding of the bacteria via a fibronectin bridge to the α5β1-integrin on the host cell membrane, followed by phagocytosis. Although osteoblasts showed high expression of α5β1-integrin and fibronectin, and bacteria adhered to osteoblasts to a high proportion, here we demonstrate by internalization assays and immunofluorescence microscopy that S. aureus was less engulfed in osteoblasts than in epithelial cells. The addition of exogenous fibronectin during the infection of cells with S. aureus resulted in an increased uptake by epithelial cells but not by osteoblasts. This contrasts with the previous conception of the uptake mechanism, where high expression of integrin and fibronectin would promote the bacterial uptake into host cells. Extracellular fibronectin surrounding osteoblasts, but not epithelial cells, is organized in a fibrillary network. The inhibition of fibril formation, the short interfering RNA-mediated reduction of fibronectin expression, and the disruption of the fibronectin-fibril meshwork all resulted in a significant increase in S. aureus uptake by osteoblasts. Thus, the network of fibronectin fibrils appears to strongly reduce the uptake of S. aureus into a given host cell, indicating that the supramolecular structure of fibronectin determines the capacity of particular host cells to internalize the pathogen. IMPORTANCE Traditionally, Staphylococcus aureus has been considered an extracellular pathogen. However, among other factors, the frequent failure of antimicrobial therapy and the ability of the pathogen to cause recurrent disease have established the concept of eukaryotic invasion of the pathogen, thereby evading the host's immune system. In the current model of host cell invasion, bacteria initially bind to α5β1 integrin on the host cell side via a fibronectin bridge, which eventually leads to phagocytosis of S. aureus by host cells. However, in this study, we demonstrate that not the crude amount but the supramolecular structure of fibronectin molecules deposited on the eukaryotic cell surface plays an essential role in bacterial uptake by host cells. Our findings explain the large differences of S. aureus uptake efficacy in different host cell types as well as in vivo differences between courses of bacterial infections and the localization of bacteria in different clinical settings.
Collapse
Affiliation(s)
- Silke Niemann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Minh-Thu Nguyen
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Achmet I. Chasan
- Institute of Immunology, University of Münster, Münster, Germany
| | - Maria Mrakovcic
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
- Institute of Infectiology, University of Münster, Münster, Germany
| | - Ralph T. Böttcher
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, Martinsried, Germany
| | - Klaus T. Preissner
- Kerckhoff-Herzforschungsinstitut, Department of Cardiology, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Steffen Roßlenbroich
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Münster, Münster, Germany
| | - Georg Peters
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
50
|
Ying YT, Ren WJ, Tan X, Yang J, Liu R, Du AF. Annexin A2-Mediated Internalization of Staphylococcus aureus into Bovine Mammary Epithelial Cells Requires Its Interaction with Clumping Factor B. Microorganisms 2021; 9:2090. [PMID: 34683411 PMCID: PMC8538401 DOI: 10.3390/microorganisms9102090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 09/26/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of contagious mastitis in dairy cattle. Internalization of S. aureus by bovine mammary gland epithelial cells is thought to be responsible for persistent and chronic intramammary infection, but the underlying mechanisms are not fully understood. METHODS In the present study, we evaluated the role of Annexin A2 (AnxA2), a membrane-binding protein, in S. aureus invasion into bovine mammary epithelial cell line (MAC-T). In vitro binding assays were performed to co-immunoprecipitate the binding proteins of AnxA2 in the lysates of S. aureus. RESULTS AnxA2 mediated the internalization but not adherence of S. aureus. Engagement of AnxA2 stimulated an integrin-linked protein kinase (ILK)/p38 MAPK cascade to induce S. aureus invasion. One of the AnxA2-precipitated proteins was identified as S. aureus clumping factor B (ClfB) through use of mass spectrometry. Direct binding of ClfB to AnxA2 was further confirmed by using a pull-down assay. Pre-incubation with recombinant ClfB protein enhanced S. aureus internalization, an effect that was specially blocked by anti-AnxA2 antibody. CONCLUSION Our results demonstrate that binding of ClfB to AnxA2 has a function in promoting S. aureus internalization. Targeting the interaction of ClfB and AnxA2 may confer protection against S. aureus mastitis.
Collapse
Affiliation(s)
- Yi-Tian Ying
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Wei-Jia Ren
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Xun Tan
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Jing Yang
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Rui Liu
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
| | - Ai-Fang Du
- Department of Veterinary Medicine, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China; (Y.-T.Y.); (W.-J.R.); (J.Y.); (R.L.); (A.-F.D.)
- Veterinary Medical Center, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| |
Collapse
|